Created
March 20, 2018 13:58
-
-
Save plasmid02/48d1fb293c0d394ae650922cdaa62302 to your computer and use it in GitHub Desktop.
BibTex library of manually curated list of scientific papers reporting (or potentially) reporting Staphylococcus aureus genome sequencing. Please let me know ([email protected]) about missing articles.
This file contains bidirectional Unicode text that may be interpreted or compiled differently than what appears below. To review, open the file in an editor that reveals hidden Unicode characters.
Learn more about bidirectional Unicode characters
% Generated by Paperpile. Check out http://paperpile.com for more information. | |
% BibTeX export options can be customized via Settings -> BibTeX. | |
@ARTICLE{Uhlemann2014-jn, | |
title = "Molecular tracing of the emergence, diversification, and | |
transmission of S. aureus sequence type 8 in a New York community", | |
author = "Uhlemann, Anne-Catrin and Dordel, Janina and Knox, Justin R and | |
Raven, Kathy E and Parkhill, Julian and Holden, Matthew T G and | |
Peacock, Sharon J and Lowy, Franklin D", | |
abstract = "During the last 2 decades, community-associated | |
methicillin-resistant Staphylococcus aureus (CA-MRSA) strains | |
have dramatically increased the global burden of S. aureus | |
infections. The pandemic sequence type (ST)8/pulsed-field gel | |
type USA300 is the dominant CA-MRSA clone in the United States, | |
but its evolutionary history and basis for biological success are | |
incompletely understood. Here, we use whole-genome sequencing of | |
387 ST8 isolates drawn from an epidemiological network of CA-MRSA | |
infections and colonizations in northern Manhattan to explore | |
short-term evolution and transmission patterns. Phylogenetic | |
analysis predicted that USA300 diverged from a most common recent | |
ancestor around 1993. We found evidence for multiple | |
introductions of USA300 and reconstructed the phylogeographic | |
spread of isolates across neighborhoods. Using pair-wise | |
single-nucleotide polymorphism distances as a measure of genetic | |
relatedness between isolates, we observed that most USA300 | |
isolates had become endemic in households, indicating their | |
critical role as reservoirs for transmission and diversification. | |
Using the maximum single-nucleotide polymorphism variability of | |
isolates from within households as a threshold, we identified | |
several possible transmission networks beyond households. Our | |
study also revealed the evolution of a fluoroquinolone-resistant | |
subpopulation in the mid-1990s and its subsequent expansion at a | |
time of high-frequency outpatient antibiotic use. This | |
high-resolution phylogenetic analysis of ST8 has documented the | |
genomic changes associated with USA300 evolution and how some of | |
its recent evolution has been shaped by antibiotic use. By | |
integrating whole-genome sequencing with detailed epidemiological | |
analyses, our study provides an important framework for | |
delineating the full diversity and spread of USA300 and other | |
emerging pathogens in large urban community populations.", | |
journal = "Proc. Natl. Acad. Sci. U. S. A.", | |
month = apr, | |
year = 2014, | |
keywords = "Staphylococcus;evolution;colonization;Staph\_evolutionary\_history;antibiotcs-evolution-fitness;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Boyle-Vavra2015-pc, | |
title = "{USA300} and {USA500} Clonal Lineages of Staphylococcus aureus | |
Do Not Produce a Capsular Polysaccharide Due to Conserved | |
Mutations in the cap5 Locus", | |
author = "Boyle-Vavra, Susan and Li, Xue and Alam, Md Tauqeer and Read, | |
Timothy D and Sieth, Julia and Cywes-Bentley, Colette and | |
Dobbins, Ginette and David, Michael Z and Kumar, Neha and Eells, | |
Samantha J and Miller, Loren G and Boxrud, David J and Chambers, | |
Henry F and Lynfield, Ruth and Lee, Jean C and Daum, Robert S", | |
abstract = "UNLABELLED: The surface capsular polysaccharide (CP) is a | |
virulence factor that has been used as an antigen in several | |
successful vaccines against bacterial pathogens. A vaccine has | |
not yet been licensed against Staphylococcus aureus, although | |
two multicomponent vaccines that contain CP antigens are in | |
clinical trials. In this study, we evaluated CP production in | |
USA300 methicillin-resistant S. aureus (MRSA) isolates that have | |
become the predominant community-associated MRSA clones in the | |
United States. We found that all 167 USA300 MRSA and 50 USA300 | |
methicillin-susceptible S. aureus (MSSA) isolates were CP | |
negative (CP(-)). Moreover, all 16 USA500 isolates, which have | |
been postulated to be the progenitor lineage of USA300, were | |
also CP(-). Whole-genome sequence analysis of 146 CP(-) USA300 | |
MRSA isolates revealed they all carry a cap5 locus with 4 | |
conserved mutations compared with strain Newman. Genetic | |
complementation experiments revealed that three of these | |
mutations (in the cap5 promoter, cap5D nucleotide 994, and cap5E | |
nucleotide 223) ablated CP production in USA300 and that Cap5E75 | |
Asp, located in the coenzyme-binding domain, is essential for | |
capsule production. All but three USA300 MSSA isolates had the | |
same four cap5 mutations found in USA300 MRSA isolates. Most | |
isolates with a USA500 pulsotype carried three of these four | |
USA300-specific mutations, suggesting the fourth mutation | |
occurred in the USA300 lineage. Phylogenetic analysis of the cap | |
loci of our USA300 isolates as well as publicly available | |
genomes from 41 other sequence types revealed that the | |
USA300-specific cap5 mutations arose sequentially in S. aureus | |
in a common ancestor of USA300 and USA500 isolates. IMPORTANCE: | |
The USA300 MRSA clone emerged as a community-associated pathogen | |
in the United States nearly 20 years ago. Since then, it has | |
rapidly disseminated and now causes health care-associated | |
infections. This study shows that the CP-negative (CP(-)) | |
phenotype has persisted among USA300 isolates and is a universal | |
and characteristic trait of this highly successful MRSA lineage. | |
It is important to note that a vaccine consisting solely of CP | |
antigens would not likely demonstrate high efficacy in the U.S. | |
population, where about half of MRSA isolates comprise USA300. | |
Moreover, conversion of a USA300 strain to a CP-positive (CP(+)) | |
phenotype is unlikely in vivo or in vitro since it would require | |
the reversion of 3 mutations. We have also established that | |
USA300 MSSA isolates and USA500 isolates are CP(-) and provide | |
new insight into the evolution of the USA300 and USA500 | |
lineages.", | |
journal = "MBio", | |
publisher = "American Society for Microbiology", | |
volume = 6, | |
number = 2, | |
pages = "e02585--14", | |
month = apr, | |
year = 2015, | |
keywords = "Staphylococcus;My | |
Publications;My\_Peer\_reviewed;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers" | |
} | |
@ARTICLE{Alam2015-oc, | |
title = "Transmission and Microevolution of {USA300} {MRSA} in {U.S}. | |
Households: Evidence from {Whole-Genome} Sequencing", | |
author = "Alam, Md Tauqeer and Read, Timothy D and Petit, Robert A and | |
Boyle-Vavra, Susan and Miller, Loren G and Eells, Samantha J and | |
Daum, Robert S and David, Michael Z", | |
abstract = "UNLABELLED: Methicillin-resistant Staphylococcus aureus (MRSA) | |
USA300 is a successful S. aureus clone in the United States and | |
a common cause of skin and soft tissue infections (SSTIs). We | |
performed whole-genome sequencing (WGS) of 146 USA300 MRSA | |
isolates from SSTIs and colonization cultures obtained from an | |
investigation conducted from 2008 to 2010 in Chicago and Los | |
Angeles households that included an index case with an S. aureus | |
SSTI. Identifying unique single nucleotide polymorphisms (SNPs) | |
and analyzing whole-genome phylogeny, we characterized isolates | |
to understand transmission dynamics, genetic relatedness, and | |
microevolution of USA300 MRSA within the households. We also | |
compared the 146 USA300 MRSA isolates from our study with the | |
previously published genome sequences of the USA300 MRSA | |
isolates from San Diego (n = 35) and New York City (n = 277). We | |
found little genetic variation within the USA300 MRSA household | |
isolates from Los Angeles (mean number of SNPs $\pm$ standard | |
deviation, 17.6 $\pm$ 35; $\pi$ nucleotide diversity, 3.1 | |
$\times$ 10(-5)) or from Chicago (mean number of SNPs $\pm$ | |
standard deviation, 12 $\pm$ 19; $\pi$ nucleotide diversity, 3.1 | |
$\times$ 10(-5)). The isolates within a household clustered into | |
closely related monophyletic groups, suggesting the introduction | |
into and transmission within each household of a single common | |
USA300 ancestral strain. From a Bayesian evolutionary | |
reconstruction, we inferred that USA300 persisted within | |
households for 2.33 to 8.35 years prior to sampling. We also | |
noted that fluoroquinolone-resistant USA300 clones emerged | |
around 1995 and were more widespread in Los Angeles and New York | |
City than in Chicago. Our findings strongly suggest that unique | |
USA300 MRSA isolates are transmitted within households that | |
contain an individual with an SSTI. Decolonization of household | |
members may be a critical component of prevention programs to | |
control USA300 MRSA spread in the United States. IMPORTANCE: | |
USA300, a virulent and easily transmissible strain of | |
methicillin-resistant Staphylococcus aureus (MRSA), is the | |
predominant community-associated MRSA clone in the United | |
States. It most commonly causes skin infections but also causes | |
necrotizing pneumonia and endocarditis. Strategies to limit the | |
spread of MRSA in the community can only be effective if we | |
understand the most common sources of transmission and the | |
microevolutionary processes that provide a fitness advantage to | |
MRSA. We performed a whole-genome sequence comparison of 146 | |
USA300 MRSA isolates from Chicago and Los Angeles. We show that | |
households represent a frequent site of transmission and a | |
long-term reservoir of USA300 strains; individuals within | |
households transmit the same USA300 strain among themselves. Our | |
study also reveals that a large proportion of the USA300 | |
isolates sequenced are resistant to fluoroquinolone antibiotics. | |
The significance of this study is that if households serve as | |
long-term reservoirs of USA300, household MRSA eradication | |
programs may result in a uniquely effective control method.", | |
journal = "MBio", | |
publisher = "American Society for Microbiology", | |
volume = 6, | |
number = 2, | |
pages = "e00054--15", | |
month = mar, | |
year = 2015, | |
keywords = "Staphylococcus;My Publications;bacterial comparative | |
genomics;antibiotcs-evolution-fitness;My\_Peer\_reviewed;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Glaser2016-qq, | |
title = "Demography and Intercontinental Spread of the {USA300} | |
{Community-Acquired} {Methicillin-Resistant} Staphylococcus | |
aureus Lineage", | |
author = "Glaser, Philippe and Martins-Sim{\~o}es, Patr{\'\i}cia and | |
Villain, Adrien and Barbier, Maxime and Tristan, Anne and | |
Bouchier, Christiane and Ma, Laurence and Bes, Michele and | |
Laurent, Frederic and Guillemot, Didier and Wirth, Thierry and | |
Vandenesch, Fran{\c c}ois", | |
abstract = "Community-acquired methicillin-resistant Staphylococcus aureus | |
(CA-MRSA) was recognized worldwide during the 1990s; in less than | |
a decade, several genetically distinct CA-MRSA lineages carrying | |
Panton-Valentine leukocidin genes have emerged on every | |
continent. Most notably, in the United States, the sequence type | |
18-IV (ST8-IV) clone known as USA300 has become highly prevalent, | |
outcompeting methicillin-susceptible S. aureus (MSSA) and other | |
MRSA strains in both community and hospital settings. CA-MRSA | |
bacteria are much less prevalent in Europe, where the European | |
ST80-IV European CA-MRSA clone, USA300 CA-MRSA strains, and other | |
lineages, such as ST22-IV, coexist. The question that arises is | |
whether the USA300 CA-MRSA present in Europe (i) was imported | |
once or on very few occasions, followed by a broad geographic | |
spread, anticipating an increased prevalence in the future, or | |
(ii) derived from multiple importations with limited spreading | |
success. In the present study, we applied whole-genome sequencing | |
to a collection of French USA300 CA-MRSA strains responsible for | |
sporadic cases and micro-outbreaks over the past decade and | |
United States ST8 MSSA and MRSA isolates. Genome-wide | |
phylogenetic analysis demonstrated that the population structure | |
of the French isolates is the product of multiple introductions | |
dating back to the onset of the USA300 CA-MRSA clone in North | |
America. Coalescent-based demography of the USA300 lineage shows | |
that a strong expansion occurred during the 1990s concomitant | |
with the acquisition of the arginine catabolic mobile element and | |
antibiotic resistance, followed by a sharp decline initiated | |
around 2008, reminiscent of the rise-and-fall pattern previously | |
observed in the ST80 lineage. A future expansion of the USA300 | |
lineage in Europe is therefore very unlikely.IMPORTANCE To trace | |
the origin, evolution, and dissemination pattern of the USA300 | |
CA-MRSA clone in France, we sequenced a collection of strains of | |
this lineage from cases reported in France in the last decade and | |
compared them with 431 ST8 strains from the United States. We | |
determined that the French CA-MRSA USA300 sporadic and | |
micro-outbreak isolates resulted from multiple independent | |
introductions of the USA300 North American lineage. At a global | |
level, in the transition from an MSSA lineage to a successful | |
CA-MRSA clone, it first became resistant to multiple antibiotics | |
and acquired the arginine catabolic mobile element and | |
subsequently acquired resistance to fluoroquinolones, and these | |
two steps were associated with a dramatic demographic expansion. | |
This expansion was followed by the current stabilization and | |
expected decline of this lineage. These findings highlight the | |
significance of horizontal gene acquisitions and point mutations | |
in the success of such disseminated clones and illustrate their | |
cyclic and sporadic life cycle.", | |
journal = "MBio", | |
volume = 7, | |
number = 1, | |
month = mar, | |
year = 2016, | |
keywords = " | |
Staphylococcus;evolution;demography;Staph\_evolutionary\_history;bacterial | |
comparative | |
genomics;Staph\_evolutionary\_history/USA300\_evolution;antibiotcs-evolution-fitness;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Read-lab-shared;Papers\_for\_Michelle;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers" | |
} | |
@ARTICLE{Toleman2016-qd, | |
title = "Systematic surveillance detects multiple silent introductions and | |
household transmission of methicillin-resistant Staphylococcus | |
aureus {USA300} in the east of England", | |
author = "Toleman, Michelle S and Reuter, Sandra and Coll, Francesc and | |
Harrison, Ewan M and Blane, Beth and Brown, Nicholas M and | |
T{\"o}r{\"o}k, M Est{\'e}e and Parkhill, Julian and Peacock, | |
Sharon J", | |
abstract = "Background. The spread of USA300 methicillin-resistant | |
Staphylococcus aureus (MRSA) across the United States (US) | |
resulted in an epidemic of infections. In Europe, only sporadic | |
cases or small clusters of USA300 infections are described and | |
its prevalence in England is unknown. We conducted prospective | |
surveillance for USA300 in the east of England.Methods. We | |
undertook a 12-month prospective observational cohort study of | |
all individuals with MRSA isolated from community and hospital | |
samples submitted to a microbiology laboratory. At least one MRSA | |
isolate from each individual was whole-genome sequenced. USA300 | |
was identified based on sequence analysis, and phylogenetic | |
comparisons were made between these and USA300 genomes from the | |
US.Results. Between April 2012-April 2013, we sequenced 2,283 | |
MRSA isolates (carriage screens and clinical samples) from 1,465 | |
individuals. USA300 was isolated from 24 (1.6\%) cases. Ten cases | |
(42\%) had skin and soft tissue infection and two cases had | |
invasive disease. Phylogenetic analyses identified multiple | |
introductions and household transmission of USA300.Conclusions. | |
Use of a diagnostic laboratory as a sentinel for surveillance has | |
identified repeated introductions of USA300 into the east of | |
England in 2012-2013, with evidence for limited transmission. Our | |
results show how systematic surveillance could provide an | |
early-warning of strain emergence and dissemination.", | |
journal = "J. Infect. Dis.", | |
month = apr, | |
year = 2016, | |
keywords = "Staphylococcus;bacterial comparative | |
genomics;Staph\_evolutionary\_history;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Ge2017-du, | |
title = "{MRSA} and multidrug-resistant Staphylococcus aureus in {U.S}. | |
retail meats, 2010-2011", | |
author = "Ge, Beilei and Mukherjee, Sampa and Hsu, Chih-Hao and Davis, | |
Johnnie A and Tran, Thu Thuy T and Yang, Qianru and Abbott, Jason | |
W and Ayers, Sherry L and Young, Shenia R and Crarey, Emily T and | |
Womack, Niketta A and Zhao, Shaohua and McDermott, Patrick F", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) has been | |
detected in retail meats, although large-scale studies are | |
scarce. We conducted a one-year survey in 2010-2011 within the | |
framework of the National Antimicrobial Resistance Monitoring | |
System. Among 3520 retail meats collected from eight U.S. states, | |
982 (27.9\%) contained S. aureus and 66 (1.9\%) were positive for | |
MRSA. Approximately 10.4\% (107/1032) of S. aureus isolates, | |
including 37.2\% (29/78) of MRSA, were multidrug-resistant | |
(MDRSA). Turkey had the highest MRSA prevalence (3.5\%), followed | |
by pork (1.9\%), beef (1.7\%), and chicken (0.3\%). Whole-genome | |
sequencing was performed for all 66 non-redundant MRSA. Among | |
five multilocus sequence types identified, ST8 (72.7\%) and ST5 | |
(22.7\%) were most common and livestock-associated MRSA ST398 was | |
assigned to one pork isolate. Eleven spa types were represented, | |
predominately t008 (43.9\%) and t2031 (22.7\%). All four types of | |
meats harbored t008, whereas t2031 was recovered from turkey | |
only. The majority of MRSA (84.8\%) possessed SCCmec IV and | |
62.1\% harbored Panton-Valentine leukocidin. Pulsed-field gel | |
electrophoresis showed that all ST8 MRSA belonged to the | |
predominant human epidemic clone USA300, and others included | |
USA100 and USA200. We conclude that a diverse MRSA population was | |
present in U.S. retail meats, albeit at low prevalence.", | |
journal = "Food Microbiol.", | |
volume = 62, | |
pages = "289--297", | |
month = apr, | |
year = 2017, | |
keywords = "LA-MRSA; MDRSA; MRSA; Retail meat; ST398; | |
USA300;Staphylococcus;colonization;Staph\_evolutionary\_history;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Popovich2017-mo, | |
title = "Genomic and Epidemiological Evidence for Community Origins of | |
{Hospital-Onset} {Methicillin-Resistant} Staphylococcus aureus | |
Bloodstream Infections", | |
author = "Popovich, Kyle J and Snitkin, Evan S and Hota, Bala and Green, | |
Stefan J and Pirani, Ali and Aroutcheva, Alla and Weinstein, | |
Robert A", | |
abstract = "Background.: We examined whether disparities existed in | |
hospital-onset (HO) Staphylococcus aureus bloodstream infections | |
(BSIs) and used whole-genome sequencing (WGS) to identify factors | |
associated with USA300 transmission networks. Methods.: We | |
evaluated HO methicillin-susceptible S. aureus (MSSA) and HO | |
methicillin-resistant S. aureus (MRSA) BSIs for 2009-2013 at 2 | |
hospitals and used an adjusted incidence for modeling. WGS and | |
phylogenetic analyses were performed on a sample of USA300 BSI | |
isolates. Epidemiologic data were analyzed in the context of | |
phylogenetic reconstructions. Results.: On multivariate analysis, | |
male sex, African-American race, and non-Hispanic white | |
race/ethnicity were significantly associated with HO-MRSA BSIs | |
whereas Hispanic ethnicity was negatively associated (rate ratio, | |
0.41; P = .002). Intermixing of community-onset and HO-USA300 | |
strains on the phylogenetic tree indicates that these strains | |
derive from a common pool. African-American race was the only | |
factor associated with genomic clustering of isolates. | |
Conclusions.: In a multicenter assessment of HO-S. aureus BSIs, | |
African-American race was significantly associated with HO-MRSA | |
but not MSSA BSIs. There appears to be a nexus of USA300 | |
community and hospital transmission networks, with a community | |
factor being the primary driver. Our data suggest that HO-USA300 | |
BSIs likely are due to colonizing strains acquired in the | |
community before hospitalization. Therefore, prevention efforts | |
may need to extend to the community for maximal benefit.", | |
journal = "J. Infect. Dis.", | |
month = may, | |
year = 2017, | |
keywords = "MRSA; Whole genome sequencing; bacteremia.; | |
disparities;Staphylococcus;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Planet2016-jd, | |
title = "Global Spread of the {Community-Associated} | |
{Methicillin-Resistant} Staphylococcus aureus {USA300} Latin | |
American Variant", | |
author = "Planet, Paul J and Diaz, Lorena and Rios, Rafael and Arias, | |
Cesar A", | |
journal = "J. Infect. Dis.", | |
volume = 214, | |
number = 10, | |
pages = "1609--1610", | |
month = nov, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN", | |
language = "en", | |
original_id = "379c0306-8718-0c37-9696-1b028a15d914" | |
} | |
@ARTICLE{Popovich2016-nk, | |
title = "Genomic Epidemiology of {USA300} {Methicillin-Resistant} | |
Staphylococcus aureus in an Urban Community", | |
author = "Popovich, Kyle J and Snitkin, Evan and Green, Stefan J and | |
Aroutcheva, Alla and Hayden, Mary K and Hota, Bala and Weinstein, | |
Robert A", | |
abstract = "BACKGROUND: In a community, it is unknown what factors account | |
for transmission of methicillin-resistant Staphylococcus aureus | |
(MRSA). We integrated whole genome sequencing (WGS) and | |
epidemiologic data to identify factors associated with MRSA | |
transmission networks in an urban community. METHODS: WGS was | |
performed on colonizing USA300 MRSA isolates from 74 individuals | |
within 72 hours of admission to a public hospital in Chicago, IL. | |
Single nucleotide variants (SNVs) were used to reconstruct the | |
phylogeny of sequenced isolates, and epidemiologic data was | |
overlaid to identify factors associated with transmission | |
networks. RESULTS: The maximum within-patient SNV difference for | |
an individual with multisite colonization was 41 SNVs, with no | |
systematic divergence among body sites. We observed a minimum of | |
7 SNVs and maximum of 153 SNVs between isolates from different | |
individuals. We identified 4 pairs of individuals whose isolates | |
were within 40 SNVs of each other. Putting our isolates in the | |
context of previously sequenced USA300 isolates from other | |
communities, we identified a 13-member group and two 4-member | |
groups that represent samples from putative local transmission | |
networks. Individuals in these groups were more likely to be | |
African American, to be human immunodeficiency virus-infected, to | |
reside in high detainee release areas, and to be current users of | |
illicit drugs. CONCLUSIONS: Using WGS, we observed potential | |
transmission networks in an urban community and that certain | |
epidemiologic factors were associated with inclusion in these | |
networks. Future work with contact tracing and advanced molecular | |
diagnostics may allow for identification of MRSA ``epicenters'' | |
in the community where interventions can be targeted.", | |
journal = "Clin. Infect. Dis.", | |
volume = 62, | |
number = 1, | |
pages = "37--44", | |
month = jan, | |
year = 2016, | |
keywords = "MRSA; whole genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Connolly2017-su, | |
title = "The identification of Staphylococcus aureus factors required for | |
pathogenicity and growth in human blood", | |
author = "Connolly, John and Boldock, Emma and Prince, Lynne R and Renshaw, | |
Stephen A and Whyte, Moira K and Foster, Simon J", | |
abstract = "Staphylococcus aureus is a human commensal but also has | |
devastating potential as an opportunist pathogen. S. aureus | |
bacteraemia is often associated with an adverse outcome. To | |
identify potential targets for novel control approaches we have | |
identified S. aureus components that are required for growth on | |
human blood. An ordered transposon mutant library was screened, | |
identifying 9 genes involved specifically in haemolysis or growth | |
on human blood agar compared to the parental strain. Three genes | |
(purA, purB and pabA) were subsequently found to be required for | |
pathogenesis in the zebrafish embryo infection model. The pabA | |
growth defect was specific to the red blood cell component of | |
human blood, showing no growth difference compared to the | |
parental strain on human serum, human plasma, sheep or horse | |
blood. PabA is required in the tetrahydrofolate (THF) | |
biosynthesis pathway. The pabA growth defect was found to be due | |
to a combination of loss of THF-dependent dTMP production by the | |
enzyme ThyA and an increased demand for pyrimidines in human | |
blood. Our work highlights pabA and the pyrimidine salvage | |
pathway as potential targets for novel therapeutics and suggests | |
a previously undefined role for a human blood factor in the | |
activity of sulphonamide antibiotics.", | |
journal = "Infect. Immun.", | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;bacerial\_genetics;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Read-lab-shared", | |
language = "en" | |
} | |
@ARTICLE{Von_Dach2016-wg, | |
title = "Comparative Genomics of {Community-Associated} | |
{Methicillin-Resistant} Staphylococcus aureus Shows the Emergence | |
of Clone {ST8-USA300} in Geneva, Switzerland", | |
author = "Von Dach, Elodie and Diene, Seydina M and Fankhauser, Carolina | |
and Schrenzel, Jacques and Harbarth, Stephan and Fran{\c c}ois, | |
Patrice", | |
abstract = "BACKGROUND: Previous investigations of community-associated | |
methicillin-resistant Staphylococcus aureus(CA-MRSA) isolates | |
have revealed a wide diversity of genetic backgrounds, with only | |
sporadic occurrence of ST8-USA300, in Geneva, Switzerland. We | |
conducted a molecular epidemiologic analysis to identify the | |
origin of a sudden increase of ST8 PVL-positive isolates in | |
Geneva during 2013. METHODS: On the basis of prospective CA-MRSA | |
surveillance, we collected colonizing and infecting ST8-USA300 | |
isolates and compared them to non-ST8 CA-MRSA isolates. | |
Whole-genome sequencing (WGS) was performed for each isolate of | |
this collection, and discriminating molecular features were | |
linked to patient data. RESULTS: In 2013, 22 isolates with the | |
ST8-USA300 profile were identified among 46 cases of CA-MRSA. WGS | |
revealed 2 groups of strains that differed by the type of the | |
SCCmec IV element encoded and whether they harbored an arginine | |
catabolism mobile element (ACME) locus. ACME-negative strains | |
were mainly isolated from patients traveling in or originating | |
from South America. Single-nucleotide polymorphism positions in | |
isolate groups were used to infer their common ancestor, | |
determine their geographical origin, and trace their relatedness. | |
CONCLUSIONS: WGS allowed the identification of transmission | |
events and revealed that the increased prevalence of USA300 | |
CA-MRSA isolates resulted from multiple importation events from | |
the Americas but not from local clonal expansion of a successful | |
clone.", | |
journal = "J. Infect. Dis.", | |
volume = 213, | |
number = 9, | |
pages = "1370--1379", | |
month = may, | |
year = 2016, | |
keywords = "USA300; comparative genomics; epidemiological survey; genome | |
evolution; high throughput sequencing; importation event; | |
methicillin-resistant Staphylococcus | |
aureus;Staphylococcus;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN", | |
language = "en" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{Jamrozy2016-vu, | |
title = "Pan-genomic perspective on the evolution of the Staphylococcus | |
aureus {USA300} epidemic", | |
author = "Jamrozy, Dorota M and Harris, Simon R and Mohamed, Naglaa and | |
Peacock, Sharon J and Tan, Charles Y and Parkhill, Julian and | |
Anderson, Annaliesa S and Holden, Matthew T G", | |
abstract = "Staphylococcus aureus USA300 represents the dominant | |
community-associated methicillin-resistant S. aureus lineage in | |
the USA, where it is a major cause of skin and soft tissue | |
infections. Previous comparative genomic studies have described | |
the population structure and evolution of USA300 based on | |
geographically restricted isolate collections. Here, we | |
investigated the USA300 population by sequencing genomes of a | |
geographically distributed panel of 191 clinical S. aureus | |
isolates belonging to clonal complex 8 (CC8), derived from the | |
Tigecycline Evaluation and Surveillance Trial program. Isolates | |
were collected at 12 healthcare centres across nine USA states | |
in 2004, 2009 or 2010. Reconstruction of evolutionary | |
relationships revealed that CC8 was dominated by USA300 isolates | |
(154/191, 81 \%), which were heterogeneous and demonstrated | |
limited phylogeographic clustering. Analysis of the USA300 core | |
genomes revealed an increase in median pairwise SNP distance | |
from 62 to 98 between 2004 and 2010, with a stable pattern of | |
above average dN/dS ratios. The phylogeny of the USA300 | |
population indicated that early diversification events led to | |
the formation of nested clades, which arose through cumulative | |
acquisition of predominantly non-synonymous SNPs in various | |
coding sequences. The accessory genome of USA300 was largely | |
homogenous and consisted of elements previously associated with | |
this lineage. We observed an emergence of SCCmec negative and | |
ACME negative USA300 isolates amongst more recent samples, and | |
an increase in the prevalence of ϕSa5 prophage. Together, the | |
analysed S. aureus USA300 collection revealed an evolving | |
pan-genome through increased core genome heterogeneity and | |
temporal variation in the frequency of certain accessory | |
elements.", | |
journal = "Microb Genom", | |
publisher = "Microbiology Society", | |
volume = 2, | |
number = 5, | |
pages = "e000058", | |
month = may, | |
year = 2016, | |
keywords = "Evolution; MRSA; | |
USA300;Staphylococcus;pangenome;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers", | |
language = "en" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{Straus2017-ku, | |
title = "Origin, evolution, and global transmission of community-acquired | |
Staphylococcus aureus {ST8}", | |
author = "Strau{\ss}, Lena and Stegger, Marc and Akpaka, Patrick Eberechi | |
and Alabi, Abraham and Breurec, Sebastien and Coombs, Geoffrey | |
and Egyir, Beverly and Larsen, Anders Rhod and Laurent, Frederic | |
and Monecke, Stefan and Peters, Georg and Skov, Robert and | |
Strommenger, Birgit and Vandenesch, Fran{\c c}ois and Schaumburg, | |
Frieder and Mellmann, Alexander", | |
abstract = "USA300 is a pandemic clonal lineage of hypervirulent, | |
community-acquired, methicillin-resistant Staphylococcus aureus | |
(CA-MRSA) with specific molecular characteristics. Despite its | |
high clinical relevance, the evolutionary origin of USA300 | |
remained unclear. We used comparative genomics of 224 temporal | |
and spatial diverse S. aureus isolates of multilocus sequence | |
type (ST) 8 to reconstruct the molecular evolution and global | |
dissemination of ST8, including USA300. Analyses of core SNP | |
diversity and accessory genome variations showed that the | |
ancestor of all ST8 S. aureus most likely emerged in Central | |
Europe in the mid-19th century. From here, ST8 was exported to | |
North America in the early 20th century and progressively | |
acquired the USA300 characteristics Panton-Valentine leukocidin | |
(PVL), SCCmec IVa, the arginine catabolic mobile element (ACME), | |
and a specific mutation in capsular polysaccharide gene cap5E | |
Although the PVL-encoding phage ϕSa2USA was introduced into the | |
ST8 background only once, various SCCmec types were introduced to | |
ST8 at different times and places. Starting from North America, | |
USA300 spread globally, including Africa. African USA300 isolates | |
have aberrant spa-types (t112, t121) and form a monophyletic | |
group within the clade of North American USA300. Large parts of | |
ST8 methicillin-susceptible S. aureus (MSSA) isolated in Africa | |
represent a symplesiomorphic group of ST8 (i.e., a group | |
representing the characteristics of the ancestor), which are | |
rarely found in other world regions. Isolates previously | |
discussed as USA300 ancestors, including USA500 and a | |
``historic'' CA-MRSA from Western Australia, were shown to be | |
only distantly related to recent USA300 clones.", | |
journal = "Proc. Natl. Acad. Sci. U. S. A.", | |
month = nov, | |
year = 2017, | |
keywords = "Africa; CA-MRSA; USA300; comparative genomics; molecular | |
evolution;Staphylococcus;Staphylococcus\_aureus\_genome\_papers/USA300\_genome\_PRJN;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Spoor2013-nc, | |
title = "Livestock Origin for a Human Pandemic Clone of | |
{Community-Associated} {Methicillin-Resistant} Staphylococcus | |
aureus", | |
author = "Spoor, L E and Mcadam, P R and Weinert, L A and Rambaut, A and | |
Hasman, H and Aarestrup, F M and Kearns, A M and Larsen, A R and | |
Skov, R L and Fitzgerald, J R", | |
journal = "MBio", | |
volume = 4, | |
number = 4, | |
pages = "e00356--13--e00356--13", | |
year = 2013, | |
keywords = "Staphylococcus;imported\_from\_papers\_14Nov2013;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Kos2012-xa, | |
title = "Comparative Genomics of {Vancomycin-Resistant} Staphylococcus | |
aureus Strains and Their Positions within the Clade Most Commonly | |
Associated with {Methicillin-Resistant} S. aureus | |
{Hospital-Acquired} Infection in the United States", | |
author = "Kos, V N and Desjardins, C A and Griggs, A and Cerqueira, G and | |
Van Tonder, A and Holden, M T G and Godfrey, P and Palmer, K L | |
and Bodi, K and Mongodin, E F and Wortman, J and Feldgarden, M | |
and Lawley, T and Gill, S R and Haas, B J and Birren, B and | |
Gilmore, M S", | |
abstract = "ABSTRACT Staphylococcus aureus is a human commensal bacterium and | |
a prominent cause of infections globally. The high incidence of | |
S. aureus infections is compounded by the ability of the microbe | |
to readily acquire resistance to antibiotics. In the United | |
States, methicillin-resistant S. aureus (MRSA) is a leading cause | |
of morbidity and mortality by a single infectious agent. | |
Therapeutic options for severe MRSA infections are limited to a | |
few antibiotics to which the organism is typically susceptible, | |
including vancomycin. Acquisition of high-level vancomycin | |
resistance by MRSA is a major concern, but to date, there have | |
been only 12 vancomycin-resistant S. aureus (VRSA) isolates | |
reported in the United States and all belong to a phylogenetic | |
lineage known as clonal complex 5. To gain enhanced understanding | |
of the genetic characteristics conducive to the acquisition of | |
vancomycin resistance by S. aureus, V. N. Kos et al. performed | |
whole-genome sequencing of all 12 VRSA isolates and compared the | |
DNA sequences to the genomes of other S. aureus strains. The | |
findings provide new information about the evolutionary history | |
of VRSA and identify genetic features that may bear on the | |
relationship between S. aureus clonal complex 5 strains and the | |
acquisition of vancomycin resistance genes from enterococci.", | |
journal = "MBio", | |
volume = 3, | |
number = 3, | |
pages = "e00112--12--e00112--12", | |
year = 2012, | |
keywords = " | |
antibiotics;Staphylococcus;imported\_from\_papers\_14Nov2013;bacterial | |
comparative genomics;Teaching antibiotic resistance | |
genomics;R-M;Staph\_evolutionary\_history;antibiotcs-evolution-fitness;MRSA-VRSA-gene\_transfer;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Lowder2009-jk, | |
title = "Recent human-to-poultry host jump, adaptation, and pandemic | |
spread of Staphylococcus aureus", | |
author = "Lowder, V, Bethan and Guinane, Caitriona M and Ben Zakour, Nouri | |
L and Weinert, Lucy A and Conway-Morris, Andrew and Cartwright, | |
Robyn A and Simpson, A John and Rambaut, Andrew and N{\"u}bel, | |
Ulrich and Fitzgerald, J Ross", | |
abstract = "The impact of globalization on the emergence and spread of | |
pathogens is an important veterinary and public health issue. | |
Staphylococcus aureus is a notorious human pathogen associated | |
with serious nosocomial and community-acquired infections. In | |
addition, S. aureus is a major cause of animal diseases including | |
skeletal infections of poultry, which are a large economic burden | |
on the global broiler chicken industry. Here, we provide evidence | |
that the majority of S. aureus isolates from broiler chickens are | |
the descendants of a single human-to-poultry host jump that | |
occurred approximately 38 years ago (range, 30 to 63 years ago) | |
by a subtype of the worldwide human ST5 clonal lineage unique to | |
Poland. In contrast to human subtypes of the ST5 radiation, which | |
demonstrate strong geographic clustering, the poultry ST5 clade | |
was distributed in different continents, consistent with wide | |
dissemination via the global poultry industry distribution | |
network. The poultry ST5 clade has undergone genetic | |
diversification from its human progenitor strain by acquisition | |
of novel mobile genetic elements from an avian-specific accessory | |
gene pool, and by the inactivation of several proteins important | |
for human disease pathogenesis. These genetic events have | |
resulted in enhanced resistance to killing by chicken | |
heterophils, reflecting avian host-adaptive evolution. Taken | |
together, we have determined the evolutionary history of a major | |
new animal pathogen that has undergone rapid avian host | |
adaptation and intercontinental dissemination. These data provide | |
a new paradigm for the impact of human activities on the | |
emergence of animal pathogens.", | |
journal = "Proc. Natl. Acad. Sci. U. S. A.", | |
volume = 106, | |
number = 46, | |
pages = "19545--19550", | |
year = 2009, | |
keywords = "Staphylococcus;imported\_from\_papers\_14Nov2013;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Harris2010-uv, | |
title = "Evolution of {MRSA} During Hospital Transmission and | |
Intercontinental Spread", | |
author = "Harris, S R and Feil, E J and Holden, M T G and Quail, M A and | |
Nickerson, E K and Chantratita, N and Gardete, S and Tavares, A | |
and Day, N and Lindsay, J A and Edgeworth, J D and de Lencastre, | |
H and Parkhill, J and Peacock, S J and Bentley, S D", | |
abstract = "Current methods for differentiating isolates of predominant | |
lineages of pathogenic bacteria often do not provide sufficient | |
resolution to define precise relationships. Here, we describe a | |
high-throughput genomics approach that provides a high-resolution | |
view of the epidemiology and microevolution of a dominant strain | |
of methicillin-resistant Staphylococcus aureus (MRSA). This | |
approach reveals the global geographic structure within the | |
lineage, its intercontinental transmission through four decades, | |
and the potential to trace person-to-person transmission within a | |
hospital environment. The ability to interrogate and resolve | |
bacterial populations is applicable to a range of infectious | |
diseases, as well as microbial ecology.", | |
journal = "Science", | |
volume = 327, | |
number = 5964, | |
pages = "469--474", | |
year = 2010, | |
keywords = "Staphylococcus;imported\_from\_papers\_14Nov2013;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Howden2010-ek, | |
title = "Complete genome sequence of Staphylococcus aureus strain | |
{JKD6008}, an {ST239} clone of methicillin-resistant | |
Staphylococcus aureus with intermediate-level vancomycin | |
resistance", | |
author = "Howden, Benjamin P and Seemann, Torsten and Harrison, Paul F and | |
McEvoy, Chris R and Stanton, Jo-Ann L and Rand, Christy J and | |
Mason, Chris W and Jensen, Slade O and Firth, Neville and Davies, | |
John K and Johnson, Paul D R and Stinear, Timothy P", | |
abstract = "We report here the complete 2.92-Mb genome sequence of a clinical | |
isolate of methicillin-resistant Staphylococcus aureus subsp. | |
aureus that demonstrates intermediate-level vancomycin | |
resistance. The strain, named JKD6008, belongs to multilocus | |
sequence type 239 and was isolated from the bloodstream of a | |
patient in New Zealand in 2003.", | |
journal = "J. Bacteriol.", | |
volume = 192, | |
number = 21, | |
pages = "5848--5849", | |
year = 2010, | |
keywords = "Staphylococcus;imported\_from\_papers\_14Nov2013;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Gao2010-fv, | |
title = "Two novel point mutations in clinical Staphylococcus aureus | |
reduce linezolid susceptibility and switch on the stringent | |
response to promote persistent infection", | |
author = "Gao, Wei and Chua, Kyra and Davies, John K and Newton, Hayley J | |
and Seemann, Torsten and Harrison, Paul F and Holmes, Natasha E | |
and Rhee, Hyun-Woo and Hong, Jong-In and Hartland, Elizabeth L | |
and Stinear, Timothy P and Howden, Benjamin P", | |
abstract = "Staphylococcus aureus frequently invades the human bloodstream, | |
leading to life threatening bacteremia and often secondary foci | |
of infection. Failure of antibiotic therapy to eradicate | |
infection is frequently described; in some cases associated with | |
altered S. aureus antimicrobial resistance or the small colony | |
variant (SCV) phenotype. Newer antimicrobials, such as | |
linezolid, remain the last available therapy for some patients | |
with multi-resistant S. aureus infections. Using comparative and | |
functional genomics we investigated the molecular determinants | |
of resistance and SCV formation in sequential S. aureus isolates | |
from a patient who had a persistent and recurrent S. aureus | |
infection, after failed therapy with multiple antimicrobials, | |
including linezolid. Two point mutations in key staphylococcal | |
genes dramatically affected clinical behaviour of the bacterium, | |
altering virulence and antimicrobial resistance. Most | |
strikingly, a single nucleotide substitution in relA (SACOL1689) | |
reduced RelA hydrolase activity and caused accumulation of the | |
intracellular signalling molecule guanosine 3', | |
5'-bis(diphosphate) (ppGpp) and permanent activation of the | |
stringent response, which has not previously been reported in S. | |
aureus. Using the clinical isolate and a defined mutant with an | |
identical relA mutation, we demonstrate for the first time the | |
impact of an active stringent response in S. aureus, which was | |
associated with reduced growth, and attenuated virulence in the | |
Galleria mellonella model. In addition, a mutation in rlmN | |
(SACOL1230), encoding a ribosomal methyltransferase that | |
methylates 23S rRNA at position A2503, caused a reduction in | |
linezolid susceptibility. These results reinforce the exquisite | |
adaptability of S. aureus and show how subtle molecular changes | |
cause major alterations in bacterial behaviour, as well as | |
highlighting potential weaknesses of current antibiotic | |
treatment regimens.", | |
journal = "PLoS Pathog.", | |
publisher = "Public Library of Science", | |
volume = 6, | |
number = 6, | |
pages = "e1000944", | |
year = 2010, | |
keywords = "Staphylococcus;imported\_from\_papers\_14Nov2013;bacterial | |
comparative genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Young2012-vu, | |
title = "Evolutionary dynamics of Staphylococcus aureus during progression | |
from carriage to disease", | |
author = "Young, Bernadette C and Golubchik, Tanya and Batty, Elizabeth M | |
and Fung, Rowena and Larner-Svensson, Hanna and Votintseva, | |
Antonina A and Miller, Ruth R and Godwin, Heather and Knox, Kyle | |
and Everitt, Richard G and Iqbal, Zamin and Rimmer, Andrew J and | |
Cule, Madeleine and Ip, Camilla L C and Didelot, Xavier and | |
Harding, Rosalind M and Donnelly, Peter and Peto, Tim E and | |
Crook, Derrick W and Bowden, Rory and Wilson, Daniel J", | |
abstract = "Whole-genome sequencing offers new insights into the evolution of | |
bacterial pathogens and the etiology of bacterial disease. | |
Staphylococcus aureus is a major cause of bacteria-associated | |
mortality and invasive disease and is carried asymptomatically by | |
27\% of adults. Eighty percent of bacteremias match the carried | |
strain. However, the role of evolutionary change in the pathogen | |
during the progression from carriage to disease is incompletely | |
understood. Here we use high-throughput genome sequencing to | |
discover the genetic changes that accompany the transition from | |
nasal carriage to fatal bloodstream infection in an individual | |
colonized with methicillin-sensitive S. aureus. We found a | |
single, cohesive population exhibiting a repertoire of 30 | |
single-nucleotide polymorphisms and four insertion/deletion | |
variants. Mutations accumulated at a steady rate over a 13-mo | |
period, except for a cluster of mutations preceding the | |
transition to disease. Although bloodstream bacteria differed by | |
just eight mutations from the original nasally carried bacteria, | |
half of those mutations caused truncation of proteins, including | |
a premature stop codon in an AraC-family transcriptional | |
regulator that has been implicated in pathogenicity. Comparison | |
with evolution in two asymptomatic carriers supported the | |
conclusion that clusters of protein-truncating mutations are | |
highly unusual. Our results demonstrate that bacterial diversity | |
in vivo is limited but nonetheless detectable by whole-genome | |
sequencing, enabling the study of evolutionary dynamics within | |
the host. Regulatory or structural changes that occur during | |
carriage may be functionally important for pathogenesis; | |
therefore identifying those changes is a crucial step in | |
understanding the biological causes of invasive bacterial | |
disease.", | |
journal = "Proceedings of the National Academy of Sciences", | |
volume = 109, | |
number = 12, | |
pages = "4550--4555", | |
year = 2012, | |
keywords = "Staphylococcus;colonization;imported\_from\_papers\_14Nov2013;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Kser2012-yi, | |
title = "Rapid whole-genome sequencing for investigation of a neonatal | |
{MRSA} outbreak", | |
author = "K{\"o}ser, Claudio U and Holden, Matthew T G and Ellington, | |
Matthew J and Cartwright, Edward J P and Brown, Nicholas M and | |
Ogilvy-Stuart, Amanda L and Hsu, Li Yang and Chewapreecha, Claire | |
and Croucher, Nicholas J and Harris, Simon R and Sanders, Mandy | |
and Enright, Mark C and Dougan, Gordon and Bentley, Stephen D and | |
Parkhill, Julian and Fraser, Louise J and Betley, Jason R and | |
Schulz-Trieglaff, Ole B and Smith, Geoffrey P and Peacock, Sharon | |
J", | |
abstract = "BACKGROUND:Isolates of methicillin-resistant Staphylococcus | |
aureus (MRSA) belonging to a single lineage are often | |
indistinguishable by means of current typing techniques. | |
Whole-genome sequencing may provide improved resolution to define | |
transmission pathways and characterize outbreaks.METHODS:We | |
investigated a putative MRSA outbreak in a neonatal intensive | |
care unit. By using rapid high-throughput sequencing technology | |
with a clinically relevant turnaround time, we retrospectively | |
sequenced the DNA from seven isolates associated with the | |
outbreak and another seven MRSA isolates associated with carriage | |
of MRSA or bacteremia in the same hospital.RESULTS:We constructed | |
a phylogenetic tree by comparing single-nucleotide polymorphisms | |
(SNPs) in the core genome to a reference genome (an epidemic MRSA | |
clone, EMRSA-15 [sequence type 22]). This revealed a distinct | |
cluster of outbreak isolates and clear separation between these | |
and the nonoutbreak isolates. A previously missed transmission | |
event was detected between two patients with bacteremia who were | |
not part of the outbreak. We created an artificial ``resistome'' | |
of antibiotic-resistance genes and demonstrated concordance | |
between it and the results of phenotypic susceptibility testing; | |
we also created a ``toxome'' consisting of toxin genes. One | |
outbreak isolate had a hypermutator phenotype with a higher | |
number of SNPs than the other outbreak isolates, highlighting the | |
difficulty of imposing a simple threshold for the number of SNPs | |
between isolates to decide whether they are part of a recent | |
transmission chain.CONCLUSIONS:Whole-genome sequencing can | |
provide clinically relevant data within a time frame that can | |
influence patient care. The need for automated data | |
interpretation and the provision of clinically meaningful reports | |
represent hurdles to clinical implementation. (Funded by the U.K. | |
Clinical Research Collaboration Translational Infection Research | |
Initiative and others.).", | |
journal = "N. Engl. J. Med.", | |
volume = 366, | |
number = 24, | |
pages = "2267--2275", | |
year = 2012, | |
keywords = "Staphylococcus;imported\_from\_papers\_14Nov2013;Teaching | |
antibiotic resistance | |
genomics;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Kant2015-wt, | |
title = "Genome Sequences of Four Staphylococcus aureus Strains Isolated | |
from Bovine Mastitis", | |
author = "Kant, Ravi and Taponen, Suvi and Koort, Joanna and Paulin, Lars | |
and {\AA}vall-J{\"a}{\"a}skel{\"a}inen, Silja and Palva, Airi", | |
abstract = "Staphylococcus aureus is a major causative agent of mastitis in | |
dairy cows. The pathogenicity of S. aureus may vary; it is able | |
to cause severe clinical mastitis, but most often it is | |
associated with chronic subclinical mastitis. Here, we present | |
the genome assemblies of four S. aureus strains from bovine | |
mastitis.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 2, | |
month = apr, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Findley2014-rn, | |
title = "The skin microbiome: a focus on pathogens and their association | |
with skin disease", | |
author = "Findley, Keisha and Grice, Elizabeth A", | |
journal = "PLoS Pathog.", | |
volume = 10, | |
number = 10, | |
pages = "e1004436", | |
month = oct, | |
year = 2014, | |
keywords = "microbiome;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Jia2015-ri, | |
title = "Characterization and complete genome sequence analysis of | |
Staphylococcus aureus bacteriophage {JS01}", | |
author = "Jia, Hongying and Dong, Wenyang and Yuan, Lvfeng and Ma, Jiale | |
and Bai, Qinqin and Pan, Zihao and Lu, Chengping and Yao, Huochun", | |
abstract = "Staphylococcus aureus is a primary pathogen that causes bovine | |
mastitis resulting in serious economic losses and herd management | |
problems in dairy cows. A novel bacteriophage, JS01, specifically | |
infecting bovine S. aureus, was isolated from milk of | |
mastitis-affected cattle. TEM observation showed that it belonged | |
to the family Siphovirus. The JS01 strain demonstrated a broad | |
host range. The prediction result of PHACTS suggested that the | |
JS01 strain was temperate phage. The JS01 genome is 43,458 bp | |
long, with a GC content of 33.32\% and no tRNAs. Annotation and | |
functional analysis of the predicted ORFs revealed six functional | |
groups: structure and morphology, DNA replication and regulation, | |
packaging, lysogeny, lysis, and pathogenicity. Comparative | |
analysis between JS01, S. aureus MSSA476, and S. aureus prophage | |
PVL was also performed. The characterization and genomic analysis | |
of JS01 provide a better understanding of S. aureus-targeting | |
bacteriophages and useful information for the development of | |
phage-based biocontrol agents against S. aureus.", | |
journal = "Virus Genes", | |
volume = 50, | |
number = 2, | |
pages = "345--348", | |
month = apr, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Zhang2014-eo, | |
title = "Complete Genome Sequence of Staphylococcus aureus {XN108}, an | |
{ST239-MRSA-SCCmec} {III} Strain with Intermediate Vancomycin | |
Resistance Isolated in Mainland China", | |
author = "Zhang, Xia and Xu, Xiaomeng and Yuan, Wenchang and Hu, Qiwen and | |
Shang, Weilong and Hu, Xiaomei and Tong, Yigang and Rao, Xiancai", | |
abstract = "ST239-MRSA-SCCmec III (ST239, sequence type 239; MRSA, | |
methicillin-resistant Staphylococcus aureus; SCCmec III, | |
staphylococcal cassette chromosome mec type III) is the most | |
predominant clone of hospital-acquired methicillin-resistant S. | |
aureus in mainland China. We report here the complete genome | |
sequence of XN108, the first vancomycin-intermediate S. aureus | |
strain isolated from a steam-burned patient with a wound | |
infection.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 4, | |
month = jul, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Suhaili2014-nb, | |
title = "Draft Genome Sequence of {Methicillin-Resistant} Staphylococcus | |
aureus {KT/Y21}, a Sequence Type 772 ({ST772}) Strain Isolated | |
from a Pediatric Blood Sample in Terengganu, Malaysia", | |
author = "Suhaili, Zarizal and Lean, Soo-Sum and Yahya, Azifah and Mohd | |
Desa, Mohd Nasir and Ali, Abdul Manaf and Yeo, Chew Chieng", | |
abstract = "Here, we report the draft genome sequence of a | |
methicillin-resistant Staphylococcus aureus (MRSA) strain, | |
KT/Y21, isolated from a blood sample of a pediatric patient. This | |
strain belongs to sequence type 772 (ST772), harbors the | |
staphylococcal cassette chromosome mec element (SCCmec) type V, | |
and is positive for the Panton-Valentine leukocidin (PVL) | |
pathogenic determinant.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 2, | |
month = apr, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Swift2014-si, | |
title = "Complete Genome Sequence of Staphylococcus aureus Phage {GRCS}", | |
author = "Swift, Steven M and Nelson, Daniel C", | |
abstract = "The Staphylococcus aureus phage GRCS was isolated from a sewage | |
treatment facility in India and has shown potential for phage | |
therapy in a mouse model of bacteremia. Here, we report the | |
complete genome sequence of this bacteriophage.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 2, | |
month = apr, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Hernandez2014-ft, | |
title = "Genome Sequences of Sequence Type 45 ({ST45}) Persistent | |
{Methicillin-Resistant} Staphylococcus aureus ({MRSA}) Bacteremia | |
Strain 300-169 and {ST45} Resolving {MRSA} Bacteremia Strain | |
301-188", | |
author = "Hernandez, David and Seidl, Kati and Corvaglia, Anna-Rita and | |
Bayer, Arnold S and Xiong, Yan Q and Fran{\c c}ois, Patrice", | |
abstract = "Persistent methicillin-resistant Staphylococcus aureus (MRSA) | |
bacteremia (positive blood cultures after $\geq$7 days) | |
represents a challenging subset of invasive MRSA infections. The | |
comparison of genome sequences of persistent (300-169) and | |
resolving (301-188) MRSA bacteremia isolates with similar genetic | |
background (sequence type 45 [ST45]) will help us to better | |
understand underlying mechanisms of persistent MRSA bacteremia.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 2, | |
month = mar, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Velusamy2014-ja, | |
title = "Draft Genome Sequences of Staphylococcus aureus {AMRF1} ({ST22}) | |
and {AMRF2} ({ST672)}, Ocular {Methicillin-Resistant} Isolates", | |
author = "Velusamy, Nithya and Prakash, Logambiga and Sivakumar, Neelamegam | |
and Antony, Aju and Prajna, Lalitha and Mohankumar, Vidyarani and | |
Devarajan, Bharanidharan", | |
abstract = "Sequence type 22 (ST22) and ST672 are the two major emerging | |
clones of community-acquired methicillin-resistant Staphylococcus | |
aureus in India. ST672 strains were found to cause severe ocular | |
infections. We report the draft genome sequences of two emerging | |
strains of methicillin-resistant S. aureus, AMRF1 (ST22) and | |
AMRF2 (ST672), isolated from patients with ocular infections.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 2, | |
month = mar, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Gill2014-ui, | |
title = "Revised Genome Sequence of Staphylococcus aureus Bacteriophage | |
{K}", | |
author = "Gill, Jason J", | |
abstract = "Bacteriophage K is a member of the virulent Twort-like group of | |
myophages infecting Staphylococcus aureus. The revised sequence | |
presented here includes 12,436 bp of additional sequence not | |
present in the previously available phage K genome (GenBank | |
accession no. NC\_005880) and updated annotations, and has been | |
reopened at the predicted terminal repeat boundary.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 1, | |
month = jan, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Wang2014-mb, | |
title = "Genome Sequence of Staphylococcus aureus Strain {LCT-SA67}, a | |
Space Flight Strain with Altered Carbon Source Utilization | |
Properties", | |
author = "Wang, Junfeng and Li, Yinhu and Guo, Jun and Zhang, Xuelin and | |
Guo, Yinghua and Chang, De and Li, Tianzhi and Xu, Guogang and | |
Dai, Wenkui and Liu, Changting", | |
abstract = "An increasing number of studies have confirmed that space flight | |
environments can have a significant effect on a variety of | |
microbial properties. To explore the effect of these environments | |
on Staphylococcus aureus, we present the draft genome sequence of | |
an S. aureus strain, named LCT-SA67, which was isolated after | |
space flight.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 1, | |
month = feb, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Jia2013-jj, | |
title = "Complete Genome Sequence of Staphylococcus aureus Siphovirus | |
Phage {JS01}", | |
author = "Jia, Hongying and Bai, Qinqin and Yang, Yongchun and Yao, Huochun", | |
abstract = "Staphylococcus aureus is the most prevalent and economically | |
significant pathogen causing bovine mastitis. We isolated and | |
characterized one staphylophage from the milk of | |
mastitis-affected cattle and sequenced its genome. Transmission | |
electron microscopy (TEM) observation shows that it belongs to | |
the family Siphovirus. We announce here its complete genome | |
sequence and report major findings from the genomic analysis.", | |
journal = "Genome Announc.", | |
volume = 1, | |
number = 6, | |
month = nov, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Sabirova2014-ac, | |
title = "Complete Genome Sequences of Two Prolific {Biofilm-Forming} | |
Staphylococcus aureus Isolates Belonging to {USA300} and | |
{EMRSA-15} Clonal Lineages", | |
author = "Sabirova, J S and Xavier, B B and Hernalsteens, J-P and De Greve, | |
H and Ieven, M and Goossens, H and Malhotra-Kumar, S", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) causes serious | |
infections that are even more difficult to treat when associated | |
with a biofilm phenotype that facilitates evasion of the host | |
immune system and antibiotics. As a first step toward | |
understanding the mechanisms underlying biofilm formation, we | |
sequenced the genomes of two prolific biofilm-forming strains | |
belonging to the two most important globally disseminated clonal | |
lineages, USA300 and EMRSA-15.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 3, | |
month = jun, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Murali2014-ap, | |
title = "Genome Sequences of Four Clinical Staphylococcus aureus Strains | |
with Diverse Drug Resistance Profiles Isolated from Diabetic Foot | |
Ulcers", | |
author = "Murali, Thokur Sreepathy and Paul, Bobby and Parikh, Hersh and | |
Singh, Rana Pratap and Kavitha, Shettigar and Bhat, Manoj K and | |
Satyamoorthy, Kapaettu", | |
abstract = "Staphylococcus aureus is a major pathogen associated with | |
diabetic foot ulcer infections. To gain insight into their | |
pathogenicity and virulence potential, we report draft genome | |
sequences of four strains of Staphylococcus aureus, isolated from | |
diabetic foot ulcers, showing profiles with various degrees of | |
resistance to common antibiotics.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 2, | |
month = mar, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Kim2014-bx, | |
title = "Genome sequence of type strain of Staphylococcus aureus subsp. | |
aureus", | |
author = "Kim, Bong-Soo and Yi, Hana and Chun, Jongsik and Cha, Chang-Jun", | |
abstract = "BACKGROUND: Staphylococcus aureus is a pathogen that causes food | |
poisoning and community-associated infection with antibiotic | |
resistance. This species is an indigenous intestinal microbe | |
found in infants and not found in adult intestine. The relatively | |
small genome size and rapid evolution of antibiotic resistance | |
genes in the species have been drawing an increasing attention in | |
public health. To extend our understanding of the species and use | |
the genome data for comparative genomic studies, we sequenced the | |
type strain of S. aureus subsp. aureus DSM 20231T. RESULTS: | |
Seventeen contigs were generated using hybrid assembly of | |
sequences derived from the Roche 454 and Illumina systems. The | |
length of the genome sequence was 2,902,619 bases with a G + C | |
content of 32.8\%. Among the 2,550 annotated CDSs, 44 CDSs were | |
annotated to antibiotic resistance genes and 13 CDSs were related | |
to methicillin resistance. It is interesting to note that this | |
strain was first isolated in 1884 before methicillin was | |
generally used on patients. CONCLUSIONS: The present study | |
analyzed the genome sequence of S. aureus subsp. aureus type | |
strain as the reference sequence for comparative genomic analyses | |
of clinical isolates. Methicillin resistance genes found in the | |
genome indicate the presence of antibiotic resistance mechanism | |
prior to the usage of antibiotics. Further comparative genomic | |
studies of methicillin-resistant strains based on this reference | |
genome would help to understand the evolution of resistance | |
mechanism and dissemination of resistance genes.", | |
journal = "Gut Pathog.", | |
volume = 6, | |
number = 1, | |
pages = "6", | |
month = mar, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Tobes2013-xp, | |
title = "Noncontiguous Finished Genome Sequence of Staphylococcus aureus | |
{KLT6}, a Staphylococcal Enterotoxin {B-Positive} Strain Involved | |
in a Food Poisoning Outbreak in Switzerland", | |
author = "Tobes, Raquel and Manrique, Marina and Brozynska, Marta and | |
Stephan, Roger and Pareja, Eduardo and Johler, Sophia", | |
abstract = "We present the first complete genome sequence of a Staphylococcus | |
aureus strain assigned to clonal complex 12. The strain was | |
isolated in a food poisoning outbreak due to contaminated potato | |
salad in Switzerland in 2009, and it produces staphylococcal | |
enterotoxin B.", | |
journal = "Genome Announc.", | |
volume = 1, | |
number = 3, | |
month = may, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Stegger2013-hj, | |
title = "Genome Sequence of Staphylococcus aureus Strain {CA-347}, a | |
{USA600} {Methicillin-Resistant} Isolate", | |
author = "Stegger, Marc and Driebe, Elizabeth M and Roe, Chandler and | |
Lemmer, Darrin and Bowers, Jolene R and Engelthaler, David M and | |
Keim, Paul and Andersen, Paal S", | |
abstract = "The Staphylococcus aureus clonal lineage CC45 is a predominant | |
colonizer of healthy individuals in northern Europe and | |
constitutes a highly basal cluster of the S. aureus population. | |
Here, we report the complete genome sequence of S. aureus strain | |
CA-347 (NRS648), a representative of the methicillin-resistant | |
USA600 clone predominantly found in the United States.", | |
journal = "Genome Announc.", | |
volume = 1, | |
number = 4, | |
month = jul, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Chen2013-df, | |
title = "Complete genome sequence of Staphylococcus aureus Z172, a | |
vancomycin-intermediate and daptomycin-nonsusceptible | |
methicillin-resistant strain isolated in Taiwan", | |
author = "Chen, Feng-Jui and Lauderdale, Tsai-Ling and Wang, Lih-Shinn and | |
Huang, I-Wen", | |
abstract = "ABSTRACT We report the complete genome sequence of Z172, a | |
representative strain of sequence type 239-staphylococcal | |
cassette chromosome mec type III (ST239-SCCmec type III) | |
hospital-associated methicillin-resistant Staphylococcus aureus | |
in Taiwan. Strain Z172 ...", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 1, | |
number = 6, | |
pages = "e01011--13", | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{McDonald2013-xz, | |
title = "Draft Genome Sequence of {Methicillin-Susceptible} Staphylococcus | |
aureus Strain {06BA18369}, a Pathogen Associated with Skin and | |
Soft Tissue Infections in Northern Saskatchewan, Canada", | |
author = "McDonald, Ryan R and Golding, George R and Irvine, James and | |
Graham, Morag R and Tyler, Shaun and Mulvey, Michael R and | |
Levett, Paul N", | |
abstract = "Here, we announce the draft sequence of a representative | |
methicillin-susceptible Staphylococcus aureus (MSSA) isolate | |
(06BA18369) whose strain type (spa type t311) was commonly | |
isolated from skin and soft tissue coinfections with | |
Streptococcus pyogenes. This strain sequence provides insight | |
into a highly successful community-associated MSSA strain type.", | |
journal = "Genome Announc.", | |
volume = 1, | |
number = 3, | |
month = jun, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Corvaglia2013-gt, | |
title = "{Whole-Genome} Sequences of Two Staphylococcus aureus {ST398} | |
Strains of Human Origin, {S94} and {S100}", | |
author = "Corvaglia, Anna-Rita and Fran{\c c}ois, Patrice and Bertrand, | |
Xavier and Quentin, Roland and Hernandez, David and van der | |
Mee-Marquet, Nathalie", | |
abstract = "Sequence type 398 (ST398) Staphylococcus aureus was originally | |
associated with animal infection. We announce the complete genome | |
sequences of two ST398 methicillin-susceptible S. aureus strains | |
of human origin, S94 and S100. The genome sequences assist in the | |
characterization of interesting ST398 features related to host | |
specificities.", | |
journal = "Genome Announc.", | |
volume = 1, | |
number = 5, | |
month = aug, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Fraunholz2013-le, | |
title = "Complete Genome Sequence of Staphylococcus aureus 6850, a Highly | |
Cytotoxic and Clinically Virulent {Methicillin-Sensitive} Strain | |
with Distant Relatedness to Prototype Strains", | |
author = "Fraunholz, Martin and Bernhardt, J{\"o}rg and Schuldes, J{\"o}rg | |
and Daniel, Rolf and Hecker, Michael and Sinha, Bhanu", | |
abstract = "Staphylococcus aureus is a frequent human commensal bacterium and | |
pathogen. Here we report the complete genome sequence of strain | |
6850 (spa type t185; sequence type 50 [ST50]), a highly cytotoxic | |
and clinically virulent methicillin-sensitive strain from a | |
patient with complicated S. aureus bacteremia associated with | |
osteomyelitis and septic arthritis.", | |
journal = "Genome Announc.", | |
volume = 1, | |
number = 5, | |
month = sep, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Harro2013-ua, | |
title = "Draft Genome Sequence of the {Methicillin-Resistant} | |
Staphylococcus aureus Isolate {MRSA-M2}", | |
author = "Harro, Janette M and Daugherty, Sean and Bruno, Vincent M and | |
Jabra-Rizk, Mary Ann and Rasko, David A and Shirtliff, Mark E", | |
abstract = "We report the draft genome sequence of a methicillin-resistant | |
strain of Staphylococcus aureus, designated MRSA-M2. This | |
clinical isolate was obtained from an osteomyelitis patient | |
undergoing treatment at the University of Texas Medical Branch | |
(Galveston, TX). This strain is an ST30, spa type T019, agr III | |
strain and has been utilized as a model S. aureus strain in a | |
number of proteomic, transcriptomic, and animal model studies.", | |
journal = "Genome Announc.", | |
volume = 1, | |
number = 1, | |
month = jan, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Hernandez2014-ul, | |
title = "De novo finished 2.8 Mbp Staphylococcus aureus genome assembly | |
from 100 bp short and long range paired-end reads", | |
author = "Hernandez, David and Tewhey, Ryan and Veyrieras, Jean-Baptiste | |
and Farinelli, Laurent and {\O}ster{\aa}s, Magne and Fran{\c | |
c}ois, Patrice and Schrenzel, Jacques", | |
abstract = "MOTIVATION: Paired-end sequencing allows circumventing the | |
shortness of the reads produced by second generation sequencers | |
and is essential for de novo assembly of genomes. However, | |
obtaining a finished genome from short reads is still an open | |
challenge. We present an algorithm that exploits the pairing | |
information issued from inserts of potentially any length. The | |
method determines paths through an overlaps graph by using a | |
constrained search tree. We also present a method that | |
automatically determines suited overlaps cutoffs according to the | |
contextual coverage, reducing thus the need for manual | |
parameterization. Finally, we introduce an interactive mode that | |
allows querying an assembly at targeted regions. RESULTS: We | |
assess our methods by assembling two Staphylococcus aureus | |
strains that were sequenced on the Illumina platform. Using 100 | |
bp paired-end reads and minimal manual curation, we produce a | |
finished genome sequence for the previously undescribed isolate | |
SGH-10-168. AVAILABILITY AND IMPLEMENTATION: The presented | |
algorithms are implemented in the standalone Edena software, | |
freely available under the General Public License (GPLv3) at | |
www.genomic.ch/edena.php.", | |
journal = "Bioinformatics", | |
volume = 30, | |
number = 1, | |
pages = "40--49", | |
month = jan, | |
year = 2014, | |
keywords = "Staphylococcus;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Golubchik2013-cy, | |
title = "Within-host evolution of Staphylococcus aureus during | |
asymptomatic carriage", | |
author = "Golubchik, Tanya and Batty, Elizabeth M and Miller, Ruth R and | |
Farr, Helen and Young, Bernadette C and Larner-Svensson, Hanna | |
and Fung, Rowena and Godwin, Heather and Knox, Kyle and | |
Votintseva, Antonina and Everitt, Richard G and Street, Teresa | |
and Cule, Madeleine and Ip, Camilla L C and Didelot, Xavier and | |
Peto, Timothy E A and Harding, Rosalind M and Wilson, Daniel J | |
and Crook, Derrick W and Bowden, Rory", | |
abstract = "BACKGROUND: Staphylococcus aureus is a major cause of healthcare | |
associated mortality, but like many important bacterial | |
pathogens, it is a common constituent of the normal human body | |
flora. Around a third of healthy adults are carriers. Recent | |
evidence suggests that evolution of S. aureus during nasal | |
carriage may be associated with progression to invasive disease. | |
However, a more detailed understanding of within-host evolution | |
under natural conditions is required to appreciate the | |
evolutionary and mechanistic reasons why commensal bacteria such | |
as S. aureus cause disease. Therefore we examined in detail the | |
evolutionary dynamics of normal, asymptomatic carriage. | |
Sequencing a total of 131 genomes across 13 singly colonized | |
hosts using the Illumina platform, we investigated diversity, | |
selection, population dynamics and transmission during the | |
short-term evolution of S. aureus. PRINCIPAL FINDINGS: We | |
characterized the processes by which the raw material for | |
evolution is generated: micro-mutation (point mutation and small | |
insertions/deletions), macro-mutation (large | |
insertions/deletions) and the loss or acquisition of mobile | |
elements (plasmids and bacteriophages). Through an analysis of | |
synonymous, non-synonymous and intergenic mutations we | |
discovered a fitness landscape dominated by purifying selection, | |
with rare examples of adaptive change in genes encoding | |
surface-anchored proteins and an enterotoxin. We found evidence | |
for dramatic, hundred-fold fluctuations in the size of the | |
within-host population over time, which we related to the cycle | |
of colonization and clearance. Using a newly-developed | |
population genetics approach to detect recent transmission among | |
hosts, we revealed evidence for recent transmission between some | |
of our subjects, including a husband and wife both carrying | |
populations of methicillin-resistant S. aureus (MRSA). | |
SIGNIFICANCE: This investigation begins to paint a picture of | |
the within-host evolution of an important bacterial pathogen | |
during its prevailing natural state, asymptomatic carriage. | |
These results also have wider significance as a benchmark for | |
future systematic studies of evolution during invasive S. aureus | |
disease.", | |
journal = "PLoS One", | |
publisher = "Public Library of Science", | |
volume = 8, | |
number = 5, | |
pages = "e61319", | |
month = may, | |
year = 2013, | |
keywords = "evolution;colonization;Staphylococcus;within-host-evolution;Staph\_evolutionary\_history/USA300\_evolution;Staph\_evolutionary\_history;imported\_from\_papers\_14Nov2013;Staph\_evolutionary\_history/Staph\_population\_dynamics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Suzuki2012-cj, | |
title = "Comparative genomic analysis of the genus Staphylococcus | |
including Staphylococcus aureus and its newly described sister | |
species Staphylococcus simiae", | |
author = "Suzuki, Haruo and Lef{\'e}bure, Tristan and Bitar, Paulina | |
Pavinski and Stanhope, Michael J", | |
abstract = "BACKGROUND:Staphylococcus belongs to the Gram-positive low G + C | |
content group of the Firmicutes division of bacteria. | |
Staphylococcus aureus is an important human and veterinary | |
pathogen that causes a broad spectrum of diseases, and has | |
developed important multidrug resistant forms such as | |
methicillin-resistant S. aureus (MRSA). Staphylococcus simiae | |
was isolated from South American squirrel monkeys in 2000, and | |
is a coagulase-negative bacterium, closely related, and possibly | |
the sister group, to S. aureus. Comparative genomic analyses of | |
closely related bacteria with different phenotypes can provide | |
information relevant to understanding adaptation to host | |
environment and mechanisms of pathogenicity.RESULTS:We | |
determined a Roche/454 draft genome sequence for S. simiae and | |
included it in comparative genomic analyses with 11 other | |
Staphylococcus species including S. aureus. A genome based | |
phylogeny of the genus confirms that S. simiae is the sister | |
group to S. aureus and indicates that the most basal | |
Staphylococcus lineage is Staphylococcus pseudintermedius, | |
followed by Staphylococcus carnosus. Given the primary niche of | |
these two latter taxa, compared to the other species in the | |
genus, this phylogeny suggests that human adaptation evolved | |
after the split of S. carnosus. The two coagulase-positive | |
species (S. aureus and S. pseudintermedius) are not | |
phylogenetically closest but share many virulence factors | |
exclusively, suggesting that these genes were acquired by | |
horizontal transfer. Enrichment in genes related to mobile | |
elements such as prophage in S. aureus relative to S. simiae | |
suggests that pathogenesis in the S. aureus group has developed | |
by gene gain through horizontal transfer, after the split of S. | |
aureus and S. simiae from their common | |
ancestor.CONCLUSIONS:Comparative genomic analyses across 12 | |
Staphylococcus species provide hypotheses about lineages in | |
which human adaptation has taken place and contributions of | |
horizontal transfer in pathogenesis.", | |
journal = "BMC Genomics", | |
publisher = "BioMed Central Ltd", | |
volume = 13, | |
pages = "38", | |
year = 2012, | |
keywords = "Staphylococcus;imported\_from\_papers\_14Nov2013;bacterial | |
comparative genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Everitt2014-tp, | |
title = "Mobile elements drive recombination hotspots in the core genome | |
of Staphylococcus aureus", | |
author = "Everitt, Richard G and Didelot, Xavier and Batty, Elizabeth M and | |
Miller, Ruth R and Knox, Kyle and Young, Bernadette C and Bowden, | |
Rory and Auton, Adam and Votintseva, Antonina and | |
Larner-Svensson, Hanna and Charlesworth, Jane and Golubchik, | |
Tanya and Ip, Camilla L C and Godwin, Heather and Fung, Rowena | |
and Peto, Tim E A and Walker, A Sarah and Crook, Derrick W and | |
Wilson, Daniel J", | |
abstract = "Horizontal gene transfer is an important driver of bacterial | |
evolution, but genetic exchange in the core genome of clonal | |
species, including the major pathogen Staphylococcus aureus, is | |
incompletely understood. Here we reveal widespread homologous | |
recombination in S. aureus at the species level, in contrast to | |
its near-complete absence between closely related strains. We | |
discover a patchwork of hotspots and coldspots at fine scales | |
falling against a backdrop of broad-scale trends in rate | |
variation. Over megabases, homoplasy rates fluctuate 1.9-fold, | |
peaking towards the origin-of-replication. Over kilobases, we | |
find core recombination hotspots of up to 2.5-fold enrichment | |
situated near fault lines in the genome associated with mobile | |
elements. The strongest hotspots include regions flanking | |
conjugative transposon ICE6013, the staphylococcal cassette | |
chromosome (SCC) and genomic island $\nu$Sa$\alpha$. Mobile | |
element-driven core genome transfer represents an opportunity for | |
adaptation and challenges our understanding of the recombination | |
landscape in predominantly clonal pathogens, with important | |
implications for genotype-phenotype mapping.", | |
journal = "Nat. Commun.", | |
volume = 5, | |
pages = "3956", | |
month = may, | |
year = 2014, | |
keywords = " | |
Staphylococcus;evolution;teaching-tutorial;recombination;bacterial | |
comparative | |
genomics;Staph\_evolutionary\_history;Staph\_evolutionary\_history/Teaching\_Prokaryotic\_evolution;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Holt2011-bo, | |
title = "A Very {Early-Branching} Staphylococcus aureus Lineage Lacking | |
the Carotenoid Pigment Staphyloxanthin", | |
author = "Holt, D C and Holden, M T G and Tong, S Y C and Castillo-Ramirez, | |
S and Clarke, L and Quail, M A and Currie, B J and Parkhill, J | |
and Bentley, S D and Feil, E J and Giffard, P M", | |
journal = "Genome Biol. Evol.", | |
volume = 3, | |
number = 0, | |
pages = "881--895", | |
year = 2011, | |
keywords = "Staphylococcus;imported\_from\_papers\_14Nov2013;Staph\_evolutionary\_history;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Koch2014-bi, | |
title = "Evolution of Resistance to a {Last-Resort} Antibiotic in | |
Staphylococcus aureus via Bacterial Competition", | |
author = "Koch, Gudrun and Yepes, Ana and F{\"o}rstner, Konrad U and | |
Wermser, Charlotte and Stengel, Stephanie T and Modamio, Jennifer | |
and Ohlsen, Knut and Foster, Kevin R and Lopez, Daniel", | |
abstract = "Antibiotic resistance is a key medical concern, with antibiotic | |
use likely being an important cause. However, here we describe an | |
alternative route to clinically relevant antibiotic resistance | |
that occurs solely due to competitive interactions among | |
bacterial cells. We consistently observe that isolates of | |
Methicillin-resistant Staphylococcus aureus diversify | |
spontaneously into two distinct, sequentially arising strains. | |
The first evolved strain outgrows the parent strain via secretion | |
of surfactants and a toxic bacteriocin. The second is resistant | |
to the bacteriocin. Importantly, this second strain is also | |
resistant to intermediate levels of vancomycin. This so-called | |
VISA (vancomycin-intermediate S. aureus) phenotype is seen in | |
many hard-to-treat clinical isolates. This strain diversification | |
also occurs during in vivo infection in a mouse model, which is | |
consistent with the fact that both coevolved phenotypes resemble | |
strains commonly found in clinic. Our study shows how competition | |
between coevolving bacterial strains can generate antibiotic | |
resistance and recapitulate key clinical phenotypes.", | |
journal = "Cell", | |
volume = 158, | |
number = 5, | |
pages = "1060--1071", | |
month = aug, | |
year = 2014, | |
keywords = " | |
antibiotics;Staphylococcus;Staph\_evolutionary\_history;bacterial | |
comparative | |
genomics;antibiotcs-evolution-fitness;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Benson2014-jp, | |
title = "Evolution of hypervirulence by a {MRSA} clone through acquisition | |
of a transposable element", | |
author = "Benson, Meredith A and Ohneck, Elizabeth A and Ryan, Chanelle and | |
Alonzo, 3rd, Francis and Smith, Hannah and Narechania, Apurva and | |
Kolokotronis, Sergios-Orestis and Satola, Sarah W and Uhlemann, | |
Anne-Catrin and Sebra, Robert and Deikus, Gintaras and Shopsin, | |
Bo and Planet, Paul J and Torres, Victor J", | |
abstract = "Staphylococcus aureus has evolved as a pathogen that causes a | |
range of diseases in humans. There are two dominant modes of | |
evolution thought to explain most of the virulence differences | |
between strains. First, virulence genes may be acquired from | |
other organisms. Second, mutations may cause changes in the | |
regulation and expression of genes. Here we describe an | |
evolutionary event in which transposition of an IS element has a | |
direct impact on virulence gene regulation resulting in | |
hypervirulence. Whole-genome analysis of a methicillin-resistant | |
S. aureus (MRSA) strain USA500 revealed acquisition of a | |
transposable element (IS256) that is absent from close relatives | |
of this strain. Of the multiple copies of IS256 found in the | |
USA500 genome, one was inserted in the promoter sequence of | |
repressor of toxins (Rot), a master transcriptional regulator | |
responsible for the expression of virulence factors in S. aureus. | |
We show that insertion into the rot promoter by IS256 results in | |
the derepression of cytotoxin expression and increased virulence. | |
Taken together, this work provides new insight into evolutionary | |
strategies by which S. aureus is able to modify its virulence | |
properties and demonstrates a novel mechanism by which horizontal | |
gene transfer directly impacts virulence through altering toxin | |
regulation.", | |
journal = "Mol. Microbiol.", | |
volume = 93, | |
number = 4, | |
pages = "664--681", | |
month = aug, | |
year = 2014, | |
keywords = "Staphylococcus;toxicity;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers" | |
} | |
@ARTICLE{Chua2014-sh, | |
title = "Hyperexpression of alpha-hemolysin explains enhanced virulence of | |
sequence type 93 community-associated methicillin-resistant | |
Staphylococcus aureus", | |
author = "Chua, Kyra and Monk, Ian and Lin, Ya-Hsun and Seemann, Torsten | |
and Tuck, Kellie and Porter, Jessica and Stepnell, Justin and | |
Coombs, Geoffrey and Davies, John and Stinear, Timothy and | |
Howden, Benjamin", | |
abstract = "BACKGROUND:The community-associated methicillin-resistant S. | |
aureus (CA-MRSA) ST93 clone is becoming dominant in Australia and | |
is clinically highly virulent. In addition, sepsis and skin | |
infection models demonstrate that ST93 CA-MRSA is the most | |
virulent global clone of S. aureus tested to date. While the | |
determinants of virulence have been studied in other clones of | |
CA-MRSA, the basis for hypervirulence in ST93 CA-MRSA has not | |
been defined.RESULTS:Here, using a geographically and temporally | |
dispersed collection of ST93 isolates we demonstrate that the | |
ST93 population hyperexpresses key CA-MRSA exotoxins, in | |
particular alpha-hemolysin, in comparison to other global clones. | |
Gene deletion and complementation studies, and virulence | |
comparisons in a murine skin infection model, showed | |
unequivocally that increased expression of alpha-hemolysin is the | |
key staphylococcal virulence determinant for this clone. Genome | |
sequencing and comparative genomics of strains with divergent | |
exotoxin profiles demonstrated that, like other S. aureus clones, | |
the quorum sensing agr system is the master regulator of toxin | |
expression and virulence in ST93 CA-MRSA. However, we also | |
identified a previously uncharacterized AraC/XylS family | |
regulator (AryK) that potentiates toxin expression and virulence | |
in S. aureus.CONCLUSIONS:These data demonstrate that | |
hyperexpression of alpha-hemolysin mediates enhanced virulence in | |
ST93 CA-MRSA, and additional control of exotoxin production, in | |
particular alpha-hemolysin, mediated by regulatory systems other | |
than agr have the potential to fine-tune virulence in CA-MRSA.", | |
journal = "BMC Microbiol.", | |
volume = 14, | |
number = 1, | |
pages = "31", | |
year = 2014, | |
keywords = "Staphylococcus;toxicity;antibiotics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Long2014-hn, | |
title = "Absence of {Patient-to-Patient} Intrahospital Transmission of | |
Staphylococcus aureus as Determined by {Whole-Genome} Sequencing", | |
author = "Long, S Wesley and Beres, Stephen B and Olsen, Randall J and | |
Musser, James M", | |
abstract = "Nosocomial transmission of pathogens is a major health care | |
challenge. The increasing spread of antibiotic-resistant strains | |
represents an ongoing threat to public health. Previous | |
Staphylococcus aureus transmission studies have focused on | |
transmission of S. aureus between asymptomatic carriers or used | |
low-resolution typing methods such as multilocus sequence typing | |
(MLST) or spa typing. To identify patient-to-patient | |
intrahospital transmission using high-resolution genetic | |
analysis, we sequenced the genomes of a consecutive set of 398 S. | |
aureus isolates from sterile-site infections. The S. aureus | |
strains were collected from four hospitals in the Houston | |
Methodist Hospital System over a 6-month period. Importantly, we | |
discovered no evidence of transmission of S. aureus between | |
patients with sterile-site infections. The lack of intrahospital | |
transmission may reflect a fundamental difference between | |
day-to-day transmission events in the hospital setting and the | |
more frequently studied outbreak scenarios.IMPORTANCE Previous | |
studies have suggested that nosocomial transmission of S. aureus | |
is common. Our data revealed an unexpected lack of evidence for | |
intrahospital transmission of S. aureus between patients with | |
invasive infections. This finding has important implications for | |
hospital infection control and public health efforts. In | |
addition, our data demonstrate that highly related pools of S. | |
aureus strains exist in the community which may complicate | |
outbreak investigations.", | |
journal = "MBio", | |
volume = 5, | |
number = 5, | |
month = oct, | |
year = 2014, | |
keywords = "Staphylococcus;bacterial comparative | |
genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Parker2014-bc, | |
title = "Genome Sequence of Bacterial Interference Strain Staphylococcus | |
aureus {502A}", | |
author = "Parker, Dane and Narechania, Apurva and Sebra, Robert and Deikus, | |
Gintaras and Larussa, Samuel and Ryan, Chanelle and Smith, Hannah | |
and Prince, Alice and Mathema, Barun and Ratner, Adam J and | |
Kreiswirth, Barry and Planet, Paul J", | |
abstract = "Staphylococcus aureus 502A was a strain used in bacterial | |
interference programs during the 1960s and early 1970s. Infants | |
were deliberately colonized with 502A with the goal of preventing | |
colonization with more invasive strains. We present the completed | |
genome sequence of this organism.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 2, | |
month = apr, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Gordon2014-jf, | |
title = "Prediction of Staphylococcus aureus antimicrobial resistance by | |
whole-genome sequencing", | |
author = "Gordon, N C and Price, J R and Cole, K and Everitt, R and Morgan, | |
M and Finney, J and Kearns, A M and Pichon, B and Young, B and | |
Wilson, D J and Llewelyn, M J and Paul, J and Peto, T E A and | |
Crook, D W and Walker, A S and Golubchik, T", | |
abstract = "Whole-genome sequencing (WGS) could potentially provide a single | |
platform for extracting all the information required to predict | |
an organism's phenotype. However, its ability to provide accurate | |
predictions has not yet been demonstrated in large independent | |
studies of specific organisms. In this study, we aimed to develop | |
a genotypic prediction method for antimicrobial susceptibilities. | |
The whole genomes of 501 unrelated Staphylococcus aureus isolates | |
were sequenced, and the assembled genomes were interrogated using | |
BLASTn for a panel of known resistance determinants (chromosomal | |
mutations and genes carried on plasmids). Results were compared | |
with phenotypic susceptibility testing for 12 commonly used | |
antimicrobial agents (penicillin, methicillin, erythromycin, | |
clindamycin, tetracycline, ciprofloxacin, vancomycin, | |
trimethoprim, gentamicin, fusidic acid, rifampin, and mupirocin) | |
performed by the routine clinical laboratory. We investigated | |
discrepancies by repeat susceptibility testing and manual | |
inspection of the sequences and used this information to optimize | |
the resistance determinant panel and BLASTn algorithm. We then | |
tested performance of the optimized tool in an independent | |
validation set of 491 unrelated isolates, with phenotypic results | |
obtained in duplicate by automated broth dilution (BD Phoenix) | |
and disc diffusion. In the validation set, the overall | |
sensitivity and specificity of the genomic prediction method were | |
0.97 (95\% confidence interval [95\% CI], 0.95 to 0.98) and 0.99 | |
(95\% CI, 0.99 to 1), respectively, compared to standard | |
susceptibility testing methods. The very major error rate was | |
0.5\%, and the major error rate was 0.7\%. WGS was as sensitive | |
and specific as routine antimicrobial susceptibility testing | |
methods. WGS is a promising alternative to culture methods for | |
resistance prediction in S. aureus and ultimately other major | |
bacterial pathogens.", | |
journal = "J. Clin. Microbiol.", | |
volume = 52, | |
number = 4, | |
pages = "1182--1191", | |
month = apr, | |
year = 2014, | |
keywords = "antibiotics;Staphylococcus;Teaching antibiotic resistance | |
genomics;Read-lab-shared;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers;GWAS-bacteria" | |
} | |
@ARTICLE{Dean2014-fn, | |
title = "Identification of point mutations in clinical Staphylococcus | |
aureus strains that produce small-colony variants auxotrophic for | |
menadione", | |
author = "Dean, Melissa A and Olsen, Randall J and Long, S Wesley and | |
Rosato, Adriana E and Musser, James M", | |
abstract = "Staphylococcus aureus small-colony variants (SCVs) are implicated | |
in chronic and relapsing infections that are difficult to | |
diagnose and treat. Despite many years of study, the underlying | |
molecular mechanisms and virulence effect of the small-colony | |
phenotype remain incompletely understood. We sequenced the | |
genomes of five S. aureus SCV strains recovered from human | |
patients and discovered previously unidentified nonsynonymous | |
point mutations in three genes encoding proteins in the menadione | |
biosynthesis pathway. Analysis of genetic revertants and | |
complementation with wild-type alleles confirmed that these | |
mutations caused the SCV phenotype and decreased virulence for | |
mice.", | |
journal = "Infect. Immun.", | |
volume = 82, | |
number = 4, | |
pages = "1600--1605", | |
month = apr, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{McCarthy2014-np, | |
title = "Extensive horizontal gene transfer during Staphylococcus aureus | |
co-colonization in vivo", | |
author = "McCarthy, Alex J and Loeffler, Anette and Witney, Adam A and | |
Gould, Katherine A and Lloyd, David H and Lindsay, Jodi A", | |
abstract = "Staphylococcus aureus is a commensal and major pathogen of humans | |
and animals. Comparative genomics of S. aureus populations | |
suggests that colonization of different host species is | |
associated with carriage of mobile genetic elements (MGE), | |
particularly bacteriophages and plasmids capable of encoding | |
virulence, resistance, and immune evasion pathways. | |
Antimicrobial-resistant S. aureus of livestock are a potential | |
zoonotic threat to human health if they adapt to colonize humans | |
efficiently. We utilized the technique of experimental evolution | |
and co-colonized gnotobiotic piglets with both human- and | |
pig-associated variants of the lineage clonal complex 398, and | |
investigated growth and genetic changes over 16 days using whole | |
genome sequencing. The human isolate survived co-colonization on | |
piglets more efficiently than in vitro. During co-colonization, | |
transfer of MGE from the pig to the human isolate was detected | |
within 4 h. Extensive and repeated transfer of two bacteriophages | |
and three plasmids resulted in colonization with isolates | |
carrying a wide variety of mobilomes. Whole genome sequencing of | |
progeny bacteria revealed no acquisition of core genome | |
polymorphisms, highlighting the importance of MGE. Staphylococcus | |
aureus bacteriophage recombination and integration into novel | |
sites was detected experimentally for the first time. During | |
colonization, clones coexisted and diversified rather than a | |
single variant dominating. Unexpectedly, each piglet carried | |
unique populations of bacterial variants, suggesting limited | |
transmission of bacteria between piglets once colonized. Our data | |
show that horizontal gene transfer occurs at very high frequency | |
in vivo and significantly higher than that detectable in vitro.", | |
journal = "Genome Biol. Evol.", | |
volume = 6, | |
number = 10, | |
pages = "2697--2708", | |
month = oct, | |
year = 2014, | |
keywords = "bacteriophage; experimental evolution; host adaptation; plasmid; | |
population | |
dynamics;Staphylococcus;colonization;recombination;Staph\_evolutionary\_history/Staph\_population\_dynamics;Staph\_evolutionary\_history/staph | |
microbial interactions;bacterial comparative | |
genomics;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Kobayashi2012-he, | |
title = "Genomic analysis of the emergence of vancomycin-resistant | |
Staphylococcus aureus", | |
author = "Kobayashi, Scott D and Musser, James M and DeLeo, Frank R", | |
abstract = "Staphylococcus aureus is a human commensal bacterium and a | |
prominent cause of infections globally. The high incidence of S. | |
aureus infections is compounded by the ability of the microbe to | |
readily acquire resistance to antibiotics. In the United States, | |
methicillin-resistant S. aureus (MRSA) is a leading cause of | |
morbidity and mortality by a single infectious agent. Therapeutic | |
options for severe MRSA infections are limited to a few | |
antibiotics to which the organism is typically susceptible, | |
including vancomycin. Acquisition of high-level vancomycin | |
resistance by MRSA is a major concern, but to date, there have | |
been only 12 vancomycin-resistant S. aureus (VRSA) isolates | |
reported in the United States and all belong to a phylogenetic | |
lineage known as clonal complex 5. To gain enhanced understanding | |
of the genetic characteristics conducive to the acquisition of | |
vancomycin resistance by S. aureus, V. N. Kos et al. performed | |
whole-genome sequencing of all 12 VRSA isolates and compared the | |
DNA sequences to the genomes of other S. aureus strains. The | |
findings provide new information about the evolutionary history | |
of VRSA and identify genetic features that may bear on the | |
relationship between S. aureus clonal complex 5 strains and the | |
acquisition of vancomycin resistance genes from enterococci.", | |
journal = "MBio", | |
volume = 3, | |
number = 4, | |
month = jun, | |
year = 2012, | |
keywords = "Staphylococcus;antibiotcs-evolution-fitness;MRSA-VRSA-gene\_transfer;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Holden2013-bu, | |
title = "A genomic portrait of the emergence, evolution, and global spread | |
of a methicillin-resistant Staphylococcus aureus pandemic", | |
author = "Holden, Matthew T G and Hsu, Li Yang and Kurt, Kevin and Weinert, | |
Lucy A and Mather, Alison E and Harris, Simon R and Strommenger, | |
Birgit and Layer, Franziska and Witte, Wolfgang and de Lencastre, | |
Herminia and Skov, Robert and Westh, Henrik and Zemlickov{\'a}, | |
Helena and Coombs, Geoffrey and Kearns, Angela M and Hill, Robert | |
L R and Edgeworth, Jonathan and Gould, Ian and Gant, Vanya and | |
Cooke, Jonathan and Edwards, Giles F and McAdam, Paul R and | |
Templeton, Kate E and McCann, Angela and Zhou, Zhemin and | |
Castillo-Ram{\'\i}rez, Santiago and Feil, Edward J and Hudson, | |
Lyndsey O and Enright, Mark C and Balloux, Francois and Aanensen, | |
David M and Spratt, Brian G and Fitzgerald, J Ross and Parkhill, | |
Julian and Achtman, Mark and Bentley, Stephen D and N{\"u}bel, | |
Ulrich", | |
abstract = "The widespread use of antibiotics in association with | |
high-density clinical care has driven the emergence of | |
drug-resistant bacteria that are adapted to thrive in | |
hospitalized patients. Of particular concern are globally | |
disseminated methicillin-resistant Staphylococcus aureus (MRSA) | |
clones that cause outbreaks and epidemics associated with health | |
care. The most rapidly spreading and tenacious | |
health-care-associated clone in Europe currently is EMRSA-15, | |
which was first detected in the UK in the early 1990s and | |
subsequently spread throughout Europe and beyond. Using | |
phylogenomic methods to analyze the genome sequences for 193 S. | |
aureus isolates, we were able to show that the current pandemic | |
population of EMRSA-15 descends from a health-care-associated | |
MRSA epidemic that spread throughout England in the 1980s, which | |
had itself previously emerged from a primarily | |
community-associated methicillin-sensitive population. The | |
emergence of fluoroquinolone resistance in this EMRSA-15 subclone | |
in the English Midlands during the mid-1980s appears to have | |
played a key role in triggering pandemic spread, and occurred | |
shortly after the first clinical trials of this drug. | |
Genome-based coalescence analysis estimated that the population | |
of this subclone over the last 20 yr has grown four times faster | |
than its progenitor. Using comparative genomic analysis we | |
identified the molecular genetic basis of 99.8\% of the | |
antimicrobial resistance phenotypes of the isolates, highlighting | |
the potential of pathogen genome sequencing as a diagnostic tool. | |
We document the genetic changes associated with adaptation to the | |
hospital environment and with increasing drug resistance over | |
time, and how MRSA evolution likely has been influenced by | |
country-specific drug use regimens.", | |
journal = "Genome Res.", | |
volume = 23, | |
number = 4, | |
pages = "653--664", | |
year = 2013, | |
keywords = " | |
Staphylococcus;antibiotics;imported\_from\_papers\_14Nov2013;Staph\_evolutionary\_history;bacterial | |
comparative | |
genomics;antibiotcs-evolution-fitness;MRSA-VRSA-gene\_transfer;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Baba2002-mu, | |
title = "Genome and virulence determinants of high virulence | |
community-acquired {MRSA}", | |
author = "Baba, Tadashi and Takeuchi, Fumihiko and Kuroda, Makoto and | |
Yuzawa, Harumi and Aoki, Ken-Ichi and Oguchi, Akio and Nagai, | |
Yoshimi and Iwama, Natsuko and Asano, Kazuyuki and Naimi, Timothy | |
and Kuroda, Hiroko and Cui, Longzhu and Yamamoto, Kenji and | |
Hiramatsu, Keiichi", | |
abstract = "BACKGROUND: A new type of meticillin-resistant Staphylococcus | |
aureus (MRSA), designated community-acquired MRSA, is becoming | |
increasingly noticeable in the community, some strains of which | |
cause fatal infections in otherwise healthy individuals. By | |
contrast with hospital-acquired MRSA, community-acquired MRSA is | |
more susceptible to non b-lactam antibiotics. We investigated the | |
high virulence potential of certain strains of this bacterium. | |
METHODS: We ascertained the whole genome sequence of MW2, a | |
strain of community-acquired MRSA, by shotgun cloning and | |
sequencing. MW2 caused fatal septicaemia and septic arthritis in | |
a 16-month-old girl in North Dakota, USA, in 1998. The genome of | |
this strain was compared with those of hospital-acquired MRSA | |
strains, including N315 and Mu50. FINDINGS: Meticillin resistance | |
gene (mecA) in MW2 was carried by a novel allelic form (type IVa) | |
of staphylococcal cassette chromosome mec (SCCmec), by contrast | |
with type II in N315 and Mu50. Type IVa SCCmec did not carry any | |
of the multiple antibiotic resistance genes reported in type II | |
SCCmec. By contrast, 19 additional virulence genes were recorded | |
in the MW2 genome. All but two of these virulence genes were | |
noted in four of the seven genomic islands of MW2. | |
INTERPRETATION: MW2 carried a range of virulence and resistance | |
genes that was distinct from those displayed on the chromosomes | |
of extant S aureus strains. Most genes were carried by specific | |
allelic forms of genomic islands in the MW2 chromosome. The | |
combination of allelic forms of genomic islands is the genetic | |
basis that determines the pathogenicity of medically important | |
phenotypes of S aureus, including those of community-acquired | |
MRSA strains.", | |
journal = "Lancet", | |
volume = 359, | |
number = 9320, | |
pages = "1819--1827", | |
month = may, | |
year = 2002, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Rossi2014-hc, | |
title = "Transferable vancomycin resistance in a community-associated | |
{MRSA} lineage", | |
author = "Rossi, Fl{\'a}via and Diaz, Lorena and Wollam, Aye and Panesso, | |
Diana and Zhou, Yanjiao and Rincon, Sandra and Narechania, Apurva | |
and Xing, Galen and Di Gioia, Thais S R and Doi, Andr{\'e} and | |
Tran, Truc T and Reyes, Jinnethe and Munita, Jose M and Carvajal, | |
Lina P and Hernandez-Roldan, Alejandra and Brand{\~a}o, Denise | |
and van der Heijden, Inneke Marie and Murray, Barbara E and | |
Planet, Paul J and Weinstock, George M and Arias, Cesar A", | |
abstract = "We report the case of a patient from Brazil with a bloodstream | |
infection caused by a strain of methicillin-resistant | |
Staphylococcus aureus (MRSA) that was susceptible to vancomycin | |
(designated BR-VSSA) but that acquired the vanA gene cluster | |
during antibiotic therapy and became resistant to vancomycin | |
(designated BR-VRSA). Both strains belong to the sequence type | |
(ST) 8 community-associated genetic lineage that carries the | |
staphylococcal chromosomal cassette mec (SCCmec) type IVa and the | |
S. aureus protein A gene (spa) type t292 and are phylogenetically | |
related to MRSA lineage USA300. A conjugative plasmid of 55,706 | |
bp (pBRZ01) carrying the vanA cluster was identified and readily | |
transferred to other staphylococci. The pBRZ01 plasmid harbors | |
DNA sequences that are typical of the plasmid-associated | |
replication genes rep24 or rep21 described in | |
community-associated MRSA strains from Australia (pWBG745). The | |
presence and dissemination of community-associated MRSA | |
containing vanA could become a serious public health concern.", | |
journal = "N. Engl. J. Med.", | |
volume = 370, | |
number = 16, | |
pages = "1524--1531", | |
month = apr, | |
year = 2014, | |
keywords = "Staphylococcus;antibiotics;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Gill2005-zg, | |
title = "Insights on evolution of virulence and resistance from the | |
complete genome analysis of an early methicillin-resistant | |
Staphylococcus aureus strain and a biofilm-producing | |
methicillin-resistant Staphylococcus epidermidis strain", | |
author = "Gill, Steven R and Fouts, Derrick E and Archer, Gordon L and | |
Mongodin, Emmanuel F and Deboy, Robert T and Ravel, Jacques and | |
Paulsen, Ian T and Kolonay, James F and Brinkac, Lauren and | |
Beanan, Mauren and Dodson, Robert J and Daugherty, Sean C and | |
Madupu, Ramana and Angiuoli, Samuel V and Durkin, A Scott and | |
Haft, Daniel H and Vamathevan, Jessica and Khouri, Hoda and | |
Utterback, Terry and Lee, Chris and Dimitrov, George and Jiang, | |
Lingxia and Qin, Haiying and Weidman, Jan and Tran, Kevin and | |
Kang, Kathy and Hance, Ioana R and Nelson, Karen E and Fraser, | |
Claire M", | |
abstract = "Staphylococcus aureus is an opportunistic pathogen and the major | |
causative agent of numerous hospital- and community-acquired | |
infections. Staphylococcus epidermidis has emerged as a causative | |
agent of infections often associated with implanted medical | |
devices. We have sequenced the approximately 2.8-Mb genome of S. | |
aureus COL, an early methicillin-resistant isolate, and the | |
approximately 2.6-Mb genome of S. epidermidis RP62a, a | |
methicillin-resistant biofilm isolate. Comparative analysis of | |
these and other staphylococcal genomes was used to explore the | |
evolution of virulence and resistance between these two species. | |
The S. aureus and S. epidermidis genomes are syntenic throughout | |
their lengths and share a core set of 1,681 open reading frames. | |
Genome islands in nonsyntenic regions are the primary source of | |
variations in pathogenicity and resistance. Gene transfer between | |
staphylococci and low-GC-content gram-positive bacteria appears | |
to have shaped their virulence and resistance profiles. | |
Integrated plasmids in S. epidermidis carry genes encoding | |
resistance to cadmium and species-specific LPXTG surface | |
proteins. A novel genome island encodes multiple phenol-soluble | |
modulins, a potential S. epidermidis virulence factor. S. | |
epidermidis contains the cap operon, encoding the polyglutamate | |
capsule, a major virulence factor in Bacillus anthracis. | |
Additional phenotypic differences are likely the result of single | |
nucleotide polymorphisms, which are most numerous in cell | |
envelope proteins. Overall differences in pathogenicity can be | |
attributed to genome islands in S. aureus which encode | |
enterotoxins, exotoxins, leukocidins, and leukotoxins not found | |
in S. epidermidis.", | |
journal = "J. Bacteriol.", | |
volume = 187, | |
number = 7, | |
pages = "2426--2438", | |
month = apr, | |
year = 2005, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Diep2006-mj, | |
title = "Complete genome sequence of {USA300}, an epidemic clone of | |
community-acquired meticillin-resistant Staphylococcus aureus", | |
author = "Diep, Binh An and Gill, Steven R and Chang, Richard F and Phan, | |
Tiffany Haivan and Chen, Jason H and Davidson, Matthew G and Lin, | |
Felice and Lin, Jessica and Carleton, Heather A and Mongodin, | |
Emmanuel F and Sensabaugh, George F and Perdreau-Remington, | |
Fran{\c c}oise", | |
abstract = "BACKGROUND: USA300, a clone of meticillin-resistant | |
Staphylococcus aureus, is a major source of community-acquired | |
infections in the USA, Canada, and Europe. Our aim was to | |
sequence its genome and compare it with those of other strains of | |
S aureus to try to identify genes responsible for its distinctive | |
epidemiological and virulence properties. METHODS: We ascertained | |
the genome sequence of FPR3757, a multidrug resistant USA300 | |
strain, by random shotgun sequencing, then compared it with the | |
sequences of ten other staphylococcal strains. FINDINGS: Compared | |
with closely related S aureus, we noted that almost all of the | |
unique genes in USA300 clustered in novel allotypes of mobile | |
genetic elements. Some of the unique genes are involved in | |
pathogenesis, including Panton-Valentine leucocidin and molecular | |
variants of enterotoxin Q and K. The most striking feature of the | |
USA300 genome is the horizontal acquisition of a novel mobile | |
genetic element that encodes an arginine deiminase pathway and an | |
oligopeptide permease system that could contribute to growth and | |
survival of USA300. We did not detect this element, termed | |
arginine catabolic mobile element (ACME), in other S aureus | |
strains. We noted a high prevalence of ACME in S epidermidis, | |
suggesting not only that ACME transfers into USA300 from S | |
epidermidis, but also that this element confers a selective | |
advantage to this ubiquitous commensal of the human skin. | |
INTERPRETATION: USA300 has acquired mobile genetic elements that | |
encode resistance and virulence determinants that could enhance | |
fitness and pathogenicity.", | |
journal = "Lancet", | |
volume = 367, | |
number = 9512, | |
pages = "731--739", | |
month = mar, | |
year = 2006, | |
keywords = "Staphylococcus;bacterial comparative | |
genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Viana2015-wo, | |
title = "A single natural nucleotide mutation alters bacterial pathogen | |
host tropism", | |
author = "Viana, David and Comos, Mar{\'\i}a and McAdam, Paul R and Ward, | |
Melissa J and Selva, Laura and Guinane, Caitriona M and | |
Gonz{\'a}lez-Mu{\~n}oz, Beatriz M and Tristan, Anne and Foster, | |
Simon J and Ross Fitzgerald, J and Penad{\'e}s, Jos{\'e} R", | |
abstract = "Jose Penades and colleagues examine host adaptation and | |
specificity for Staphylococcus aureus with analysis of the | |
whole-genome sequences of strains isolated from humans or | |
rabbits. They find that a rabbit-specific clone evolved through | |
a human-to-rabbit host jump, enabled by a single mutation in | |
dltB.", | |
journal = "Nat. Genet.", | |
publisher = "Nature Publishing Group", | |
month = feb, | |
year = 2015, | |
keywords = "Staphylococcus;colonization;bacerial\_genetics;bacterial | |
comparative | |
genomics;Staph\_evolutionary\_history;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;GWAS-bacteria;Staph\_evolution\_toxins;Staph-animal-human" | |
} | |
@ARTICLE{Ward2014-bz, | |
title = "{Time-Scaled} Evolutionary Analysis of the Transmission and | |
Antibiotic Resistance Dynamics of Staphylococcus aureus Clonal | |
Complex 398", | |
author = "Ward, M J and Gibbons, C L and McAdam, P R and van Bunnik, B A D | |
and Girvan, E K and Edwards, G F and Fitzgerald, J R and | |
Woolhouse, M E J", | |
abstract = "Staphylococcus aureus clonal complex 398 (CC398) is associated | |
with disease in humans and livestock, and its origins and | |
transmission have generated considerable interest. We performed a | |
time-scaled phylogenetic analysis of CC398, including sequenced | |
isolates from the United Kingdom (Scotland), along with publicly | |
available genomes. Using state-of-the-art methods for mapping | |
traits onto phylogenies, we quantified transitions between host | |
species to identify sink and source populations for CC398 and | |
employed a novel approach to investigate the gain and loss of | |
antibiotic resistance in CC398 over time. We identified distinct | |
human- and livestock-associated CC398 clades and observed | |
multiple transmissions of CC398 from livestock to humans and | |
between countries, lending quantitative support to previous | |
reports. Of note, we identified a subclade within the | |
livestock-associated clade comprised of isolates from hospital | |
environments and newborn babies, suggesting that | |
livestock-associated CC398 is capable of onward transmission in | |
hospitals. In addition, our analysis revealed significant | |
differences in the dynamics of resistance to methicillin and | |
tetracycline related to contrasting historical patterns of | |
antibiotic usage between the livestock industry and human | |
medicine. We also identified significant differences in patterns | |
of gain and loss of different tetracycline resistance | |
determinants, which we ascribe to epistatic interactions between | |
the resistance genes and/or differences in the modes of | |
inheritance of the resistance determinants.", | |
journal = "Appl. Environ. Microbiol.", | |
volume = 80, | |
number = 23, | |
pages = "7275--7282", | |
month = dec, | |
year = 2014, | |
keywords = " | |
antibiotics;Staphylococcus;Staph\_evolutionary\_history;bacterial | |
comparative | |
genomics;antibiotcs-evolution-fitness;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Stinear2014-ay, | |
title = "Adaptive Change Inferred from Genomic Population Analysis of the | |
{ST93} Epidemic Clone of {Community-Associated} Methicillin | |
Resistant Staphylococcus aureus", | |
author = "Stinear, Timothy P. and Holt, Kathryn E. and Chua, Kyra and | |
Stepnell, Justin and Tuck, Kellie L. and Coombs, Geoffrey and | |
Harrison, Paul Francis and Seemann, Torsten and Howden, Benjamin | |
P.", | |
abstract = "Community associated methicillin resistant Staphylococcus aureus | |
(CA-MRSA) has emerged as a major public health problem around the | |
world. In Australia, ST93-IV[2B] is the dominant CA-MRSA clone | |
and displays significantly greater virulence than other S. | |
aureus. Here we have examined the evolution of ST93 via genomic | |
analysis of 12 MSSA and 44 MRSA ST93 isolates, collected from | |
around Australia over a 17-year period. Comparative analysis | |
revealed a core genome of 2.6 Mb, sharing greater than 99.7\% | |
nucleotide identity. The accessory genome was 0.45 Mb and | |
comprised additional mobile DNA elements, harboring resistance to | |
erythromycin, trimethoprim and tetracycline. Phylogenetic | |
inference revealed a molecular clock and suggested that a single | |
clone of methicillin susceptible, Panton-Valentine leukocidin | |
(PVL) positive, ST93 S. aureus likely spread from North Western | |
Australia in the early 1970s, acquiring methicillin resistance at | |
least twice in the mid 1990s. We also explored associations | |
between genotype and important MRSA phenotypes including | |
oxacillin MIC and production of exotoxins ($\alpha$-hemolysin, | |
$\delta$-hemolysin, PSM$\alpha$3, and PVL). High-level expression | |
of $\alpha$-hemolysin is a signature feature of ST93 and reduced | |
expression in eight isolates was readily explained by mutations | |
in the agr locus. However, subtle but significant decreases in | |
$\delta$-hemolysin were also noted over time that coincided with | |
decreasing oxacillin resistance and were independent of agr | |
mutations. The evolution of ST93 S. aureus is thus associated | |
with a reduction in both exotoxin expression and oxacillin MIC, | |
suggesting MRSA ST93 isolates are under pressure for adaptive | |
change.", | |
journal = "Genome Biol. Evol.", | |
month = jan, | |
year = 2014, | |
keywords = " | |
Staphylococcus;antibiotics;oxacillin;Staph\_evolutionary\_history;bacterial | |
comparative | |
genomics;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Paterson2015-nr, | |
title = "Capturing the cloud of diversity reveals complexity and | |
heterogeneity of {MRSA} carriage, infection and transmission", | |
author = "Paterson, Gavin K and Harrison, Ewan M and Murray, Gemma G R and | |
Welch, John J and Warland, James H and Holden, Matthew T G and | |
Morgan, Fiona J E and Ba, Xiaoliang and Koop, Gerrit and Harris, | |
Simon R and Maskell, Duncan J and Peacock, Sharon J and Herrtage, | |
Michael E and Parkhill, Julian and Holmes, Mark A", | |
abstract = "Genome sequencing is revolutionizing clinical microbiology and | |
our understanding of infectious diseases. Previous studies have | |
largely relied on the sequencing of a single isolate from each | |
individual. However, it is not clear what degree of bacterial | |
diversity exists within, and is transmitted between individuals. | |
Understanding this 'cloud of diversity' is key to accurate | |
identification of transmission pathways. Here, we report the deep | |
sequencing of methicillin-resistant Staphylococcus aureus among | |
staff and animal patients involved in a transmission network at a | |
veterinary hospital. We demonstrate considerable within-host | |
diversity and that within-host diversity may rise and fall over | |
time. Isolates from invasive disease contained multiple mutations | |
in the same genes, including inactivation of a global regulator | |
of virulence and changes in phage copy number. This study | |
highlights the need for sequencing of multiple isolates from | |
individuals to gain an accurate picture of transmission networks | |
and to further understand the basis of pathogenesis.", | |
journal = "Nat. Commun.", | |
volume = 6, | |
pages = "6560", | |
year = 2015, | |
keywords = "Staphylococcus;colonization;evolution;phage;bacterial comparative | |
genomics;Staph\_evolutionary\_history/staph microbial | |
interactions;Staph\_evolutionary\_history;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Moon2015-ue, | |
title = "Phage-mediated horizontal transfer of a Staphylococcus aureus | |
virulence-associated genomic island", | |
author = "Moon, Bo Youn and Park, Joo Youn and Hwang, Sun Yung and | |
Robinson, D Ashley and Thomas, Jonathan C and Fitzgerald, J Ross | |
and Park, Yong Ho and Seo, Keun Seok", | |
abstract = "Staphylococcus aureus is a major pathogen of humans and animals. | |
The capacity of S. aureus to adapt to different host species and | |
tissue types is strongly influenced by the acquisition of mobile | |
genetic elements encoding determinants involved in niche | |
adaptation. The genomic islands $\nu$Sa$\alpha$ and | |
$\nu$Sa$\beta$ are found in almost all S. aureus strains and are | |
characterized by extensive variation in virulence gene content. | |
However the basis for the diversity and the mechanism underlying | |
mobilization of the genomic islands between strains are | |
unexplained. Here, we demonstrated that the genomic island, | |
$\nu$Sa$\beta$, encoding an array of virulence factors including | |
staphylococcal superantigens, proteases, and leukotoxins, in | |
addition to bacteriocins, was transferrable in vitro to human and | |
animal strains of multiple S. aureus clones via a resident | |
prophage. The transfer of the $\nu$Sa$\beta$ appears to have been | |
accomplished by multiple conversions of transducing phage | |
particles carrying overlapping segments of the $\nu$Sa$\beta$. | |
Our findings solve a long-standing mystery regarding the | |
diversification and spread of the genomic island $\nu$Sa$\beta$, | |
highlighting the central role of bacteriophages in the pathogenic | |
evolution of S. aureus.", | |
journal = "Sci. Rep.", | |
volume = 5, | |
pages = "9784", | |
month = apr, | |
year = 2015, | |
keywords = "Staphylococcus;plasmid;phage;Staph\_evolutionary\_history;Read-lab-shared;MRSA-VRSA-gene\_transfer;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Hsu2015-dp, | |
title = "Evolutionary dynamics of methicillin-resistant Staphylococcus | |
aureus within a healthcare system", | |
author = "Hsu, Li-Yang and Harris, Simon R and Chlebowicz, Monika A and | |
Lindsay, Jodi A and Koh, Tse-Hsien and Krishnan, Prabha and Tan, | |
Thean-Yen and Hon, Pei-Yun and Grubb, Warren B and Bentley, | |
Stephen D and Parkhill, Julian and Peacock, Sharon J and Holden, | |
Matthew Tg", | |
abstract = "BACKGROUND: In the past decade, several countries have seen | |
gradual replacement of endemic multi-resistant | |
healthcare-associated methicillin-resistant Staphylococcus aureus | |
(MRSA) with clones that are more susceptible to antibiotic | |
treatment. One example is Singapore, where MRSA ST239, the | |
dominant clone since molecular profiling of MRSA began in the | |
mid-1980s, has been replaced by ST22 isolates belonging to | |
EMRSA-15, a recently emerged pandemic lineage originating from | |
Europe. RESULTS: We investigated the population structure of MRSA | |
in Singaporean hospitals spanning three decades, using whole | |
genome sequencing. Applying Bayesian phylogenetic methods we | |
report that prior to the introduction of ST22, the ST239 MRSA | |
population in Singapore originated from multiple introductions | |
from the surrounding region; it was frequently transferred within | |
the healthcare system resulting in a heterogeneous hospital | |
population. Following the introduction of ST22 around the | |
beginning of the millennium, this clone spread rapidly through | |
Singaporean hospitals, supplanting the endemic ST239 population. | |
Coalescent analysis revealed that although the genetic diversity | |
of ST239 initially decreased as ST22 became more dominant, from | |
2007 onwards the genetic diversity of ST239 began to increase | |
once more, which was not associated with the emergence of a | |
sub-clone of ST239. Comparative genomic analysis of the accessory | |
genome of the extant ST239 population identified that the | |
Arginine Catabolic Mobile Element arose multiple times, thereby | |
introducing genes associated with enhanced skin colonization into | |
this population. CONCLUSIONS: Our results clearly demonstrate | |
that, alongside clinical practice and antibiotic usage, | |
competition between clones also has an important role in driving | |
the evolution of nosocomial pathogen populations.", | |
journal = "Genome Biol.", | |
volume = 16, | |
number = 1, | |
pages = "81", | |
month = apr, | |
year = 2015, | |
keywords = "Staphylococcus;evolution;bacterial comparative | |
genomics;antibiotcs-evolution-fitness;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Hau2017-mi, | |
title = "Draft Genome Sequences of 50 {Methicillin-Resistant} | |
Staphylococcus aureus Sequence Type 5 Isolates Obtained from a | |
{U.S}. Hospital", | |
author = "Hau, Samantha J and Bayles, Darrell O and Alt, David P and | |
Nicholson, Tracy L", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) can be a | |
commensal or pathogen in humans. Pathogenicity and disease are | |
related to the acquisition of mobile genetic elements encoding | |
virulence and antimicrobial resistance genes. Here, we report | |
draft genome sequences for 50 clinical MRSA isolates from humans | |
with MRSA-related disease.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 44, | |
month = nov, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Guinane2010-pb, | |
title = "Evolutionary genomics of Staphylococcus aureus reveals insights | |
into the origin and molecular basis of ruminant host adaptation", | |
author = "Guinane, Caitriona M and Ben Zakour, Nouri L and Tormo-Mas, Maria | |
A and Weinert, Lucy A and Lowder, Bethan V and Cartwright, Robyn | |
A and Smyth, Davida S and Smyth, Cyril J and Lindsay, Jodi A and | |
Gould, Katherine A and Witney, Adam and Hinds, Jason and | |
Bollback, Jonathan P and Rambaut, Andrew and Penad{\'e}s, | |
Jos{\'e} R and Fitzgerald, J Ross", | |
abstract = "Phenotypic biotyping has traditionally been used to differentiate | |
bacteria occupying distinct ecological niches such as host | |
species. For example, the capacity of Staphylococcus aureus from | |
sheep to coagulate ruminant plasma, reported over 60 years ago, | |
led to the description of small ruminant and bovine S. aureus | |
ecovars. The great majority of small ruminant isolates are | |
represented by a single, widespread clonal complex (CC133) of S. | |
aureus, but its evolutionary origin and the molecular basis for | |
its host tropism remain unknown. Here, we provide evidence that | |
the CC133 clone evolved as the result of a human to ruminant host | |
jump followed by adaptive genome diversification. Comparative | |
whole-genome sequencing revealed molecular evidence for host | |
adaptation including gene decay and diversification of proteins | |
involved in host-pathogen interactions. Importantly, several | |
novel mobile genetic elements encoding virulence proteins with | |
attenuated or enhanced activity in ruminants were widely | |
distributed in CC133 isolates, suggesting a key role in its | |
host-specific interactions. To investigate this further, we | |
examined the activity of a novel staphylococcal pathogenicity | |
island (SaPIov2) found in the great majority of CC133 isolates | |
which encodes a variant of the chromosomally encoded von | |
Willebrand-binding protein (vWbp(Sov2)), previously demonstrated | |
to have coagulase activity for human plasma. Remarkably, we | |
discovered that SaPIov2 confers the ability to coagulate ruminant | |
plasma suggesting an important role in ruminant disease | |
pathogenesis and revealing the origin of a defining phenotype of | |
the classical S. aureus biotyping scheme. Taken together, these | |
data provide broad new insights into the origin and molecular | |
basis of S. aureus ruminant host specificity.", | |
journal = "Genome Biol. Evol.", | |
volume = 2, | |
pages = "454--466", | |
month = jul, | |
year = 2010, | |
keywords = "Staphylococcus;colonization;host-pathogen;bacterial comparative | |
genomics;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Kuroda2001-wn, | |
title = "Whole genome sequencing of meticillin-resistant Staphylococcus | |
aureus", | |
author = "Kuroda, M and Ohta, T and Uchiyama, I and Baba, T and Yuzawa, H | |
and Kobayashi, I and Cui, L and Oguchi, A and Aoki, K and Nagai, | |
Y and Lian, J and Ito, T and Kanamori, M and Matsumaru, H and | |
Maruyama, A and Murakami, H and Hosoyama, A and Mizutani-Ui, Y | |
and Takahashi, N K and Sawano, T and Inoue, R and Kaito, C and | |
Sekimizu, K and Hirakawa, H and Kuhara, S and Goto, S and | |
Yabuzaki, J and Kanehisa, M and Yamashita, A and Oshima, K and | |
Furuya, K and Yoshino, C and Shiba, T and Hattori, M and | |
Ogasawara, N and Hayashi, H and Hiramatsu, K", | |
abstract = "BACKGROUND: Staphylococcus aureus is one of the major causes of | |
community-acquired and hospital-acquired infections. It produces | |
numerous toxins including superantigens that cause unique disease | |
entities such as toxic-shock syndrome and staphylococcal scarlet | |
fever, and has acquired resistance to practically all | |
antibiotics. Whole genome analysis is a necessary step towards | |
future development of countermeasures against this organism. | |
METHODS: Whole genome sequences of two related S aureus strains | |
(N315 and Mu50) were determined by shot-gun random sequencing. | |
N315 is a meticillin-resistant S aureus (MRSA) strain isolated in | |
1982, and Mu50 is an MRSA strain with vancomycin resistance | |
isolated in 1997. The open reading frames were identified by use | |
of GAMBLER and GLIMMER programs, and annotation of each was done | |
with a BLAST homology search, motif analysis, and protein | |
localisation prediction. FINDINGS: The Staphylococcus genome was | |
composed of a complex mixture of genes, many of which seem to | |
have been acquired by lateral gene transfer. Most of the | |
antibiotic resistance genes were carried either by plasmids or by | |
mobile genetic elements including a unique resistance island. | |
Three classes of new pathogenicity islands were identified in the | |
genome: a toxic-shock-syndrome toxin island family, exotoxin | |
islands, and enterotoxin islands. In the latter two pathogenicity | |
islands, clusters of exotoxin and enterotoxin genes were found | |
closely linked with other gene clusters encoding putative | |
pathogenic factors. The analysis also identified 70 candidates | |
for new virulence factors. INTERPRETATION: The remarkable ability | |
of S aureus to acquire useful genes from various organisms was | |
revealed through the observation of genome complexity and | |
evidence of lateral gene transfer. Repeated duplication of genes | |
encoding superantigens explains why S aureus is capable of | |
infecting humans of diverse genetic backgrounds, eliciting severe | |
immune reactions. Investigation of many newly identified gene | |
products, including the 70 putative virulence factors, will | |
greatly improve our understanding of the biology of staphylococci | |
and the processes of infectious diseases caused by S aureus.", | |
journal = "Lancet", | |
volume = 357, | |
number = 9264, | |
pages = "1225--1240", | |
month = apr, | |
year = 2001, | |
keywords = "antibiotics;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Roach2015-ns, | |
title = "A Year of Infection in the Intensive Care Unit: Prospective Whole | |
Genome Sequencing of Bacterial Clinical Isolates Reveals Cryptic | |
Transmissions and Novel Microbiota", | |
author = "Roach, David J and Burton, Joshua N and Lee, Choli and | |
Stackhouse, Bethany and Butler-Wu, Susan M and Cookson, Brad T | |
and Shendure, Jay and Salipante, Stephen J", | |
abstract = "Bacterial whole genome sequencing holds promise as a disruptive | |
technology in clinical microbiology, but it has not yet been | |
applied systematically or comprehensively within a clinical | |
context. Here, over the course of one year, we performed | |
prospective collection and whole genome sequencing of nearly all | |
bacterial isolates obtained from a tertiary care hospital's | |
intensive care units (ICUs). This unbiased collection of 1,229 | |
bacterial genomes from 391 patients enables detailed exploration | |
of several features of clinical pathogens. A sizable fraction of | |
isolates identified as clinically relevant corresponded to | |
previously undescribed species: 12\% of isolates assigned a | |
species-level classification by conventional methods actually | |
qualified as distinct, novel genomospecies on the basis of | |
genomic similarity. Pan-genome analysis of the most frequently | |
encountered pathogens in the collection revealed substantial | |
variation in pan-genome size (1,420 to 20,432 genes) and the rate | |
of gene discovery (1 to 152 genes per isolate sequenced). | |
Surprisingly, although potential nosocomial transmission of | |
actively surveilled pathogens was rare, 8.7\% of isolates | |
belonged to genomically related clonal lineages that were present | |
among multiple patients, usually with overlapping hospital | |
admissions, and were associated with clinically significant | |
infection in 62\% of patients from which they were recovered. | |
Multi-patient clonal lineages were particularly evident in the | |
neonatal care unit, where seven separate Staphylococcus | |
epidermidis clonal lineages were identified, including one | |
lineage associated with bacteremia in 5/9 neonates. Our study | |
highlights key differences in the information made available by | |
conventional microbiological practices versus whole genome | |
sequencing, and motivates the further integration of microbial | |
genome sequencing into routine clinical care.", | |
journal = "PLoS Genet.", | |
volume = 11, | |
number = 7, | |
pages = "e1005413", | |
month = jul, | |
year = 2015, | |
keywords = "Staphylococcus;pangenome;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Burd2014-nc, | |
title = "Development of oxacillin resistance in a patient with recurrent | |
Staphylococcus aureus bacteremia", | |
author = "Burd, Eileen M and Alam, Mohammad Tauqeer and Passalacqua, Karla | |
D and Kalokhe, Ameeta S and Eaton, Molly E and Satola, Sarah W | |
and Kraft, Colleen S and Read, Timothy D", | |
abstract = "Whole-genome sequencing was used to compare longitudinal | |
isolates of Staphylococcus aureus that developed resistance to | |
oxacillin (MIC up to 16 $\mu$g/ml). The mecA gene was absent. A | |
novel 5-bp TATCC frameshift insertion in a gene encoding an ABC | |
transporter similar to that of the teichoic acid translocation | |
ATP-binding protein TagH and a 3-bp GCT nonframeshift insertion | |
in the pdhA pyruvate dehydrogenase gene were detected in the | |
oxacillin-resistant isolates.", | |
journal = "J. Clin. Microbiol.", | |
publisher = "American Society for Microbiology", | |
volume = 52, | |
number = 8, | |
pages = "3114--3117", | |
month = aug, | |
year = 2014, | |
keywords = "oxacillin;antibiotics;Staphylococcus;My | |
Publications;My\_Peer\_reviewed;Staphylococcus\_aureus\_genome\_papers" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{Alam2014-fv, | |
title = "Dissecting vancomycin-intermediate resistance in Staphylococcus | |
aureus using genome-wide association", | |
author = "Alam, Md Tauqeer and Petit, 3rd, Robert A and Crispelll, 3rd, | |
Emily K and Thornton, Timothy A and Conneely, Karen N and Jiang, | |
Yunxuan and Satola, Sarah W and Read, Timothy D", | |
abstract = "Vancomycin-intermediate Staphylococcus aureus (VISA) is | |
currently defined as having minimal inhibitory concentration | |
(MIC) of 4-8 µg/ml. VISA evolves through changes in multiple | |
genetic loci with at least 16 candidate genes identified in | |
clinical and in vitro-selected VISA strains. We report a | |
whole-genome comparative analysis of 49 vancomycin-sensitive S. | |
aureus and 26 VISA strains. Resistance to vancomycin was | |
determined by broth microdilution, Etest, and population | |
analysis profile-area under the curve (PAP-AUC). Genome-wide | |
association studies (GWAS) of 55,977 single-nucleotide | |
polymorphisms identified in one or more strains found one highly | |
significant association (P = 8.78 E-08) between a nonsynonymous | |
mutation at codon 481 (H481) of the rpoB gene and increased | |
vancomycin MIC. Additionally, we used a database of public S. | |
aureus genome sequences to identify rare mutations in candidate | |
genes associated with VISA. On the basis of these data, we | |
proposed a preliminary model called ECM+RMCG for the VISA | |
phenotype as a benchmark for future efforts. The model predicted | |
VISA based on the presence of a rare mutation in a set of | |
candidate genes (walKR, vraSR, graSR, and agrA) and/or three | |
previously experimentally verified mutations (including the rpoB | |
H481 locus) with an accuracy of 81\% and a sensitivity of 73\%. | |
Further, the level of resistance measured by both Etest and | |
PAP-AUC regressed positively with the number of mutations | |
present in a strain. This study demonstrated 1) the power of | |
GWAS for identifying common genetic variants associated with | |
antibiotic resistance in bacteria and 2) that rare mutations in | |
candidate gene, identified using large genomic data sets, can | |
also be associated with resistance phenotypes.", | |
journal = "Genome Biol. Evol.", | |
publisher = "Oxford University Press", | |
volume = 6, | |
number = 5, | |
pages = "1174--1185", | |
month = apr, | |
year = 2014, | |
keywords = "bacteria; genomics; phylogeny; whole-genome | |
sequencing;Staphylococcus;confirmed\_SA-resistance-mutation;My | |
Publications;antibiotcs-evolution-fitness;My\_Peer\_reviewed;Staphylococcus\_aureus\_genome\_papers;GWAS-bacteria" | |
} | |
@ARTICLE{Laabei2014-tg, | |
title = "Predicting the virulence of {MRSA} from its genome sequence", | |
author = "Laabei, Maisem and Recker, Mario and Rudkin, Justine K and | |
Aldeljawi, Mona and Gulay, Zeynep and Sloan, Tim J and Williams, | |
Paul and Endres, Jennifer L and Bayles, Kenneth W and Fey, Paul | |
D and Yajjala, Vijaya Kumar and Widhelm, Todd and Hawkins, Erica | |
and Lewis, Katie and Parfett, Sara and Scowen, Lucy and Peacock, | |
Sharon J and Holden, Matthew and Wilson, Daniel and Read, | |
Timothy D and van den Elsen, Jean and Priest, Nicholas K and | |
Feil, Edward J and Hurst, Laurence D and Josefsson, Elisabet and | |
Massey, Ruth C", | |
abstract = "Microbial virulence is a complex and often multifactorial | |
phenotype, intricately linked to a pathogen's evolutionary | |
trajectory. Toxicity, the ability to destroy host cell | |
membranes, and adhesion, the ability to adhere to human tissues, | |
are the major virulence factors of many bacterial pathogens, | |
including Staphylococcus aureus. Here, we assayed the toxicity | |
and adhesiveness of 90 MRSA (methicillin resistant S. aureus) | |
isolates and found that while there was remarkably little | |
variation in adhesion, toxicity varied by over an order of | |
magnitude between isolates, suggesting different evolutionary | |
selection pressures acting on these two traits. We performed a | |
genome-wide association study (GWAS) and identified a large | |
number of loci, as well as a putative network of epistatically | |
interacting loci, that significantly associated with toxicity. | |
Despite this apparent complexity in toxicity regulation, a | |
predictive model based on a set of significant single nucleotide | |
polymorphisms (SNPs) and insertion and deletions events (indels) | |
showed a high degree of accuracy in predicting an isolate's | |
toxicity solely from the genetic signature at these sites. Our | |
results thus highlight the potential of using sequence data to | |
determine clinically relevant parameters and have further | |
implications for understanding the microbial virulence of this | |
opportunistic pathogen.", | |
journal = "Genome Res.", | |
publisher = "Cold Spring Harbor Lab", | |
volume = 24, | |
number = 5, | |
pages = "839--849", | |
month = may, | |
year = 2014, | |
keywords = "bacerial\_genetics;Staphylococcus;toxicity;My | |
Publications;My\_Peer\_reviewed;Read-lab-shared;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers;GWAS-bacteria;Staph\_evolution\_toxins" | |
} | |
@ARTICLE{Passalacqua2012-yq, | |
title = "A mutation in the {PP2C} phosphatase gene in a Staphylococcus | |
aureus {USA300} clinical isolate with reduced susceptibility to | |
vancomycin and daptomycin", | |
author = "Passalacqua, Karla D and Satola, Sarah W and Crispell, Emily K | |
and Read, Timothy D", | |
journal = "Antimicrob. Agents Chemother.", | |
publisher = "American Society for Microbiology", | |
volume = 56, | |
number = 10, | |
pages = "5212--5223", | |
year = 2012, | |
keywords = "Staphylococcus;My | |
Publications;imported\_from\_papers\_14Nov2013;My\_Peer\_reviewed;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Nubel2010-gs, | |
title = "A timescale for evolution, population expansion, and spatial | |
spread of an emerging clone of methicillin-resistant | |
Staphylococcus aureus", | |
author = "N{\"u}bel, Ulrich and Dordel, Janina and Kurt, Kevin and | |
Strommenger, Birgit and Westh, Henrik and Shukla, Sanjay K and | |
Zemlickov{\'a}, Helena and Leblois, Rapha{\"e}l and Wirth, | |
Thierry and Jombart, Thibaut and Balloux, Fran{\c c}ois and | |
Witte, Wolfgang", | |
abstract = "Due to the lack of fossil evidence, the timescales of bacterial | |
evolution are largely unknown. The speed with which genetic | |
change accumulates in populations of pathogenic bacteria, | |
however, is a key parameter that is crucial for understanding the | |
emergence of traits such as increased virulence or antibiotic | |
resistance, together with the forces driving pathogen spread. | |
Methicillin-resistant Staphylococcus aureus (MRSA) is a common | |
cause of hospital-acquired infections. We have investigated an | |
MRSA strain (ST225) that is highly prevalent in hospitals in | |
Central Europe. By using mutation discovery at 269 genetic loci | |
(118,804 basepairs) within an international isolate collection, | |
we ascertained extremely low diversity among European ST225 | |
isolates, indicating that a recent population bottleneck had | |
preceded the expansion of this clone. In contrast, US isolates | |
were more divergent, suggesting they represent the ancestral | |
population. While diversity was low, however, our results | |
demonstrate that the short-term evolutionary rate in this natural | |
population of MRSA resulted in the accumulation of measurable DNA | |
sequence variation within two decades, which we could exploit to | |
reconstruct its recent demographic history and the spatiotemporal | |
dynamics of spread. By applying Bayesian coalescent methods on | |
DNA sequences serially sampled through time, we estimated that | |
ST225 had diverged since approximately 1990 (1987 to 1994), and | |
that expansion of the European clade began in 1995 (1991 to | |
1999), several years before the new clone was recognized. | |
Demographic analysis based on DNA sequence variation indicated a | |
sharp increase of bacterial population size from 2001 to 2004, | |
which is concordant with the reported prevalence of this strain | |
in several European countries. A detailed ancestry-based | |
reconstruction of the spatiotemporal dispersal dynamics suggested | |
a pattern of frequent transmission of the ST225 clone among | |
hospitals within Central Europe. In addition, comparative | |
genomics indicated complex bacteriophage dynamics.", | |
journal = "PLoS Pathog.", | |
volume = 6, | |
number = 4, | |
pages = "e1000855", | |
month = apr, | |
year = 2010, | |
keywords = "Staphylococcus;evolution;bacterial comparative | |
genomics;antibiotcs-evolution-fitness;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Harris2013-mf, | |
title = "Whole-genome sequencing for analysis of an outbreak of | |
meticillin-resistant Staphylococcus aureus: a descriptive study", | |
author = "Harris, Simon R and Cartwright, Edward J P and T{\"o}r{\"o}k, M | |
Est{\'e}e and Holden, Matthew T G and Brown, Nicholas M and | |
Ogilvy-Stuart, Amanda L and Ellington, Matthew J and Quail, | |
Michael A and Bentley, Stephen D and Parkhill, Julian and | |
Peacock, Sharon J", | |
abstract = "BACKGROUND: The emergence of meticillin-resistant Staphylococcus | |
aureus (MRSA) that can persist in the community and replace | |
existing hospital-adapted lineages of MRSA means that it is | |
necessary to understand transmission dynamics in terms of | |
hospitals and the community as one entity. We assessed the use | |
of whole-genome sequencing to enhance detection of MRSA | |
transmission between these settings. METHODS: We studied a | |
putative MRSA outbreak on a special care baby unit (SCBU) at a | |
National Health Service Foundation Trust in Cambridge, UK. We | |
used whole-genome sequencing to validate and expand findings | |
from an infection-control team who assessed the outbreak through | |
conventional analysis of epidemiological data and antibiogram | |
profiles. We sequenced isolates from all colonised patients in | |
the SCBU, and sequenced MRSA isolates from patients in the | |
hospital or community with the same antibiotic susceptibility | |
profile as the outbreak strain. FINDINGS: The hospital | |
infection-control team identified 12 infants colonised with MRSA | |
in a 6 month period in 2011, who were suspected of being linked, | |
but a persistent outbreak could not be confirmed with | |
conventional methods. With whole-genome sequencing, we | |
identified 26 related cases of MRSA carriage, and showed | |
transmission occurred within the SCBU, between mothers on a | |
postnatal ward, and in the community. The outbreak MRSA type was | |
a new sequence type (ST) 2371, which is closely related to ST22, | |
but contains genes encoding Panton-Valentine leucocidin. | |
Whole-genome sequencing data were used to propose and confirm | |
that MRSA carriage by a staff member had allowed the outbreak to | |
persist during periods without known infection on the SCBU and | |
after a deep clean. INTERPRETATION: Whole-genome sequencing | |
holds great promise for rapid, accurate, and comprehensive | |
identification of bacterial transmission pathways in hospital | |
and community settings, with concomitant reductions in | |
infections, morbidity, and costs. FUNDING: UK Clinical Research | |
Collaboration Translational Infection Research Initiative, | |
Wellcome Trust, Health Protection Agency, and the National | |
Institute for Health Research Cambridge Biomedical Research | |
Centre.", | |
journal = "Lancet Infect. Dis.", | |
publisher = "Elsevier", | |
volume = 13, | |
number = 2, | |
pages = "130--136", | |
month = feb, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Stegger2012-hy, | |
title = "Genome sequence of Staphylococcus aureus strain 11819-97, an | |
{ST80-IV} European community-acquired methicillin-resistant | |
isolate", | |
author = "Stegger, Marc and Price, Lance B and Larsen, Anders R and | |
Gillece, John D and Waters, Andrew E and Skov, Robert and | |
Andersen, Paal S", | |
abstract = "The European methicillin-resistant Staphylococcus aureus (MRSA) | |
clone ST80-IV has historically dominated community-associated | |
infections in major parts of Europe and is a lineage strongly | |
linked to skin and soft tissue infections. Here, we report the | |
genome sequence of an ST80-IV representative, 11819-97, isolated | |
from a skin infection in Denmark in 1997.", | |
journal = "J. Bacteriol.", | |
publisher = "Am Soc Microbiol", | |
volume = 194, | |
number = 6, | |
pages = "1625--1626", | |
month = mar, | |
year = 2012, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Treangen2014-oh, | |
title = "Complete Genome Sequence of the Quality Control Strain | |
Staphylococcus aureus subsp. aureus {ATCC} 25923", | |
author = "Treangen, Todd J and Maybank, Rosslyn A and Enke, Sana and | |
Friss, Mary Beth and Diviak, Lynn F and Karaolis, David K R and | |
Koren, Sergey and Ondov, Brian and Phillippy, Adam M and | |
Bergman, Nicholas H and Rosovitz, M J", | |
abstract = "Staphylococcus aureus subsp. aureus ATCC 25923 is commonly used | |
as a control strain for susceptibility testing to antibiotics | |
and as a quality control strain for commercial products. We | |
present the completed genome sequence for the strain, consisting | |
of the chromosome and a 27.5-kb plasmid.", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 2, | |
number = 6, | |
month = nov, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Price2014-jt, | |
title = "Whole-genome sequencing shows that patient-to-patient | |
transmission rarely accounts for acquisition of Staphylococcus | |
aureus in an intensive care unit", | |
author = "Price, James R and Golubchik, Tanya and Cole, Kevin and Wilson, | |
Daniel J and Crook, Derrick W and Thwaites, Guy E and Bowden, | |
Rory and Walker, A Sarah and Peto, Timothy E A and Paul, John | |
and Llewelyn, Martin J", | |
abstract = "BACKGROUND: Strategies to prevent Staphylococcus aureus | |
infection in hospitals focus on patient-to-patient transmission. | |
We used whole-genome sequencing to investigate the role of | |
colonized patients as the source of new S. aureus acquisitions, | |
and the reliability of identifying patient-to-patient | |
transmission using the conventional approach of spa typing and | |
overlapping patient stay. METHODS: Over 14 months, all | |
unselected patients admitted to an adult intensive care unit | |
(ICU) were serially screened for S. aureus. All available | |
isolates (n = 275) were spa typed and underwent whole-genome | |
sequencing to investigate their relatedness at high resolution. | |
RESULTS: Staphylococcus aureus was carried by 185 of 1109 | |
patients sampled within 24 hours of ICU admission (16.7\%); 59 | |
(5.3\%) patients carried methicillin-resistant S. aureus (MRSA). | |
Forty-four S. aureus (22 MRSA) acquisitions while on ICU were | |
detected. Isolates were available for genetic analysis from 37 | |
acquisitions. Whole-genome sequencing indicated that 7 of these | |
37 (18.9\%) were transmissions from other colonized patients. | |
Conventional methods (spa typing combined with overlapping | |
patient stay) falsely identified 3 patient-to-patient | |
transmissions (all MRSA) and failed to detect 2 acquisitions and | |
4 transmissions (2 MRSA). CONCLUSIONS: Only a minority of S. | |
aureus acquisitions can be explained by patient-to-patient | |
transmission. Whole-genome sequencing provides the resolution to | |
disprove transmission events indicated by conventional methods | |
and also to reveal otherwise unsuspected transmission events. | |
Whole-genome sequencing should replace conventional methods for | |
detection of nosocomial S. aureus transmission.", | |
journal = "Clin. Infect. Dis.", | |
publisher = "cid.oxfordjournals.org", | |
volume = 58, | |
number = 5, | |
pages = "609--618", | |
month = mar, | |
year = 2014, | |
keywords = "Staphylococcus aureus transmission; adult; intensive care unit; | |
spa typing; whole-genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Sass2012-jk, | |
title = "Genome sequence of Staphylococcus aureus {VC40}, a | |
vancomycin-and daptomycin-resistant strain, to study the | |
genetics of development of resistance to currently applied | |
last-resort antibiotics", | |
author = "Sass, Peter and Berscheid, Anne and Jansen, Andrea and | |
Oedenkoven, Marion and Szekat, Christiane and Strittmatter, Axel | |
and Gottschalk, Gerhard and Bierbaum, Gabriele", | |
abstract = "ABSTRACT The increasing emergence of multidrug-resistant | |
Staphylococcus aureus is a problem of global importance. Here, | |
we report the genome of S. aureus VC40, which is resistant to | |
the last-resort antibiotics vancomycin and daptomycin. Its | |
genome sequence ...", | |
journal = "J. Bacteriol.", | |
publisher = "Am Soc Microbiol", | |
volume = 194, | |
number = 8, | |
pages = "2107--2108", | |
year = 2012, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Ip2014-mq, | |
title = "Draft Genome Sequence of {Methicillin-Resistant} Staphylococcus | |
aureus {CUHK\_188} ({ST188)}, a Health {Care-Associated} | |
Bacteremic Isolate from Hong Kong", | |
author = "Ip, Margaret and Wang, Zheng and Lam, Wai Yip and Zhou, Haokui | |
and Tsui, Stephen", | |
abstract = "We report the draft genome sequence of a methicillin-resistant | |
Staphylococcus aureus strain designated CUHK\_188, isolated from | |
a bacteremic patient undergoing treatment at a university | |
teaching hospital in Hong Kong. This strain belongs to sequence | |
type 188 (ST188), with spa type t189 and staphylococcal cassette | |
chromosome mec type V.", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 2, | |
number = 2, | |
month = apr, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Huang2012-zq, | |
title = "Complete genome sequence of Staphylococcus aureus M013, a | |
pvl-positive, {ST59-SCCmec} type {V} strain isolated in Taiwan", | |
author = "Huang, Tzu-Wen and Chen, Feng-Jui and Miu, Wei-Chieh and Liao, | |
Tsai-Lien and Lin, Ann-Chi and Huang, I-Wen and Wu, Keh-Ming and | |
Tsai, Shih-Feng and Chen, Ying-Tsong and Lauderdale, Tsai-Ling | |
Yang", | |
abstract = "We report the complete genome sequence of M013, a representative | |
strain of a pvl-positive, sequence type 59-staphylococcal | |
cassette chromosome mec type V (ST59-SCCmec type V) | |
community-associated methicillin-resistant Staphylococcus aureus | |
(CA-MRSA) clone in Taiwan. Comparison of M013 with the genomes | |
of two CA-MRSA strains in the United States revealed major | |
differences in the regions covering several genomic islands and | |
prophages.", | |
journal = "J. Bacteriol.", | |
publisher = "Am Soc Microbiol", | |
volume = 194, | |
number = 5, | |
pages = "1256--1257", | |
month = mar, | |
year = 2012, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Bouchard2012-ud, | |
title = "Genome sequence of Staphylococcus aureus Newbould 305, a strain | |
associated with mild bovine mastitis", | |
author = "Bouchard, Damien and Peton, Vincent and Almeida, Sintia and Le | |
Mar{\'e}chal, Caroline and Miyoshi, Anderson and Azevedo, Vasco | |
and Berkova, Nadia and Rault, Lucie and Fran{\c c}ois, Patrice | |
and Schrenzel, Jacques and Even, Sergine and Hernandez, David | |
and Le Loir, Yves", | |
abstract = "Staphylococcus aureus is a major etiological agent of mastitis | |
in ruminants. We report here the genome sequence of bovine | |
strain Newbould 305, isolated in the 1950s in a case of bovine | |
mastitis and now used as a model strain able to reproducibly | |
induce chronic mastitis in cows.", | |
journal = "J. Bacteriol.", | |
publisher = "Am Soc Microbiol", | |
volume = 194, | |
number = 22, | |
pages = "6292--6293", | |
month = nov, | |
year = 2012, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Schijffelen2013-dh, | |
title = "Whole genome analysis of epidemiologically closely related | |
Staphylococcus aureus isolates", | |
author = "Schijffelen, Maarten and Konstantinov, Sergey R and Lina, | |
G{\'e}rard and Spiliopoulou, Iris and van Duijkeren, Engeline | |
and Brouwer, Ellen C and Fluit, Ad C", | |
abstract = "The change of the bacteria from colonizers to pathogens is | |
accompanied by a drastic change in expression profiles. These | |
changes may be due to environmental signals or to mutational | |
changes. We therefore compared the whole genome sequences of | |
four sets of S. aureus isolates. Three sets were from the same | |
patients. The isolates of each pair (S1800/S1805, S2396/S2395, | |
S2398/S2397, an isolate from colonization and an isolate from | |
infection, respectively) were obtained within <30 days of each | |
other and the isolate from infection caused skin infections. The | |
isolates were then compared for differences in gene content and | |
SNPs. In addition, a set of isolates from a colonized pig and a | |
farmer from the same farm at the same time (S0462 and S0460) | |
were analyzed. The isolates pair S1800/S1805 showed a difference | |
in a prophage, but these are easily lost or acquired. However, | |
S1805 contained an integrative conjugative element not present | |
in S1800. In addition, 92 SNPs were present in a variety of | |
genes and the isolates S1800 and S1805 were not considered a | |
pair. Between S2395/S2396 two SNPs were present: one was in an | |
intergenic region and one was a synonymous mutation in a | |
putative membrane protein. Between S2397/S2398 only one | |
synonymous mutation in a putative lipoprotein was found. The two | |
farm isolates were very similar and showed 12 SNPs in genes that | |
belong to a number of different functional categories. However, | |
we cannot pinpoint any gene that explains the change from | |
carrier status to infection. The data indicate that differences | |
between the isolate from infection and the colonizing isolate | |
for S2395/S2396 and S2397/S2398 exist as well as between | |
isolates from different hosts, but S1800/S1805 are not clonal.", | |
journal = "PLoS One", | |
publisher = "dx.plos.org", | |
volume = 8, | |
number = 10, | |
pages = "e78340", | |
month = oct, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Thomas2012-gq, | |
title = "Draft genome sequences of Staphylococcus aureus sequence type 34 | |
({ST34}) and {ST42} hybrids", | |
author = "Thomas, Jonathan C and Godfrey, Paul A and Feldgarden, Michael | |
and Robinson, D Ashley", | |
abstract = "Staphylococcus aureus is a major cause of | |
antimicrobial-resistant infections of humans. Hybrids of S. | |
aureus, which originate from large-scale chromosomal | |
recombinations between parents of distinct genetic backgrounds, | |
are of interest from clinical and evolutionary perspectives. We | |
present draft genome sequences of two S. aureus hybrids of | |
sequence type 34 (ST34) and ST42.", | |
journal = "J. Bacteriol.", | |
publisher = "Am Soc Microbiol", | |
volume = 194, | |
number = 10, | |
pages = "2740--2741", | |
month = may, | |
year = 2012, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Larner-Svensson2013-jf, | |
title = "Complete Genome Sequence of Staphylococcus aureus Strain M1, a | |
Unique {t024-ST8-IVa} Danish {Methicillin-Resistant} S. aureus | |
Clone", | |
author = "Larner-Svensson, Hanna and Worning, Peder and Bartels, Mette D | |
and Hestbjerg Hansen, Lars and Boye, Kit and Westh, Henrik", | |
abstract = "We report the genome sequence, in five contigs, of a | |
methicillin-resistant Staphylococcus aureus isolate designated | |
M1. This clinical isolate was from the index patient of a | |
methicillin-resistant Staphylococcus aureus (MRSA) outbreak in | |
Copenhagen, Denmark, that started in 2003. This strain is | |
sequence type 8 (ST8), spa type t024, and staphylococcal | |
cassette chromosome mec element (SCCmec) type IVa.", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 1, | |
number = 3, | |
month = jun, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Prabhakara2013-hi, | |
title = "Genome sequencing unveils a novel sea enterotoxin-carrying {PVL} | |
phage in Staphylococcus aureus {ST772} from India", | |
author = "Prabhakara, Sushma and Khedkar, Supriya and Shambat, Srikanth | |
Mairpady and Srinivasan, Rajalakshmi and Basu, Atanu and | |
Norrby-Teglund, Anna and Seshasayee, Aswin Sai Narain and | |
Arakere, Gayathri", | |
abstract = "Staphylococcus aureus is a major human pathogen, first | |
recognized as a leading cause of hospital-acquired infections. | |
Community-associated S. aureus (CA-SA) pose a greater threat due | |
to increase in severity of infection and disease among children | |
and healthy adults. CA-SA strains in India are genetically | |
diverse, among which is the sequence type (ST) 772, which has | |
now spread to Australia, Europe and Japan. Towards understanding | |
the genetic characteristics of ST772, we obtained draft genome | |
sequences of five relevant clinical isolates and studied the | |
properties of their PVL-carrying prophages, whose presence is a | |
defining hallmark of CA-SA. We show that this is a novel | |
prophage, which carries the structural genes of the hlb-carrying | |
prophage and includes the sea enterotoxin. This architecture | |
probably emerged early within the ST772 lineage, at least in | |
India. The sea gene, unique to ST772 PVL, despite having | |
promoter sequence characteristics typical of low expression, | |
appears to be highly expressed during early phase of growth in | |
laboratory conditions. We speculate that this might be a | |
consequence of its novel sequence context. The crippled nature | |
of the hlb-converting prophage in ST772 suggests that widespread | |
mobility of the sea enterotoxin might be a selective force | |
behind its 'transfer' to the PVL prophage. Wild type ST772 | |
strains induced strong proliferative responses as well as high | |
cytotoxic activity against neutrophils, likely mediated by | |
superantigen SEA and the PVL toxin respectively. Both | |
proliferation and cytotoxicity were markedly reduced in a cured | |
ST772 strain indicating the impact of the phage on virulence. | |
The presence of SEA alongside the genes for the immune | |
system-modulating PVL toxin may contribute to the success and | |
virulence of ST772.", | |
journal = "PLoS One", | |
publisher = "dx.plos.org", | |
volume = 8, | |
number = 3, | |
pages = "e60013", | |
month = mar, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Biswas2015-xv, | |
title = "Draft Genome Sequences of Five Clinical {Methicillin-Resistant} | |
Staphylococcus aureus Isolates and a {Methicillin-Resistant} | |
Staphylococcus epidermidis Isolate", | |
author = "Biswas, Rajesh Kumar and Kock, Marleen M and Adelowotan, Toyin | |
and Strasheim, Wilhelmina and Mahomed, Tanweer Goolam and | |
Salawu, Adeola and Ehlers, Marthie M", | |
abstract = "We report the complete draft genome sequences of five | |
individually isolated strains of methicillin-resistant | |
Staphylococcus aureus (MRSA) and a Staphylococcus epidermidis | |
strain. These clinically important isolates have staphylococcal | |
cassette chromosome mec type A, while Panton-Valentine | |
leukocidin (PVL) toxin coding genes were present in MRSA | |
isolates only.", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 3, | |
number = 4, | |
month = jul, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Sassi2014-nj, | |
title = "Draft Genome Sequence of Staphylococcus aureus subsp. aureus | |
Strain {HG003}, an {NCTC8325} Derivative", | |
author = "Sassi, Mohamed and Felden, Brice and Augagneur, Yoann", | |
abstract = "We report the draft genome sequence of a Staphylococcus aureus | |
NCTC8325 derivative, strain HG003. HG003 contains functional | |
global regulators rsbU and tcaR and is therefore considered as a | |
reference for studies of regulation and virulence. The genome is | |
composed of 2,797,898 bp and will be essential for subsequent | |
RNAseq analysis.", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 2, | |
number = 4, | |
month = aug, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Daligault2014-kl, | |
title = "Genome Assembly of {Methicillin-Resistant} Quality Control | |
Strain Staphylococcus aureus {CDC73-57501} ({ATCC} 29247)", | |
author = "Daligault, H E and Davenport, K W and Minogue, T D and | |
Bishop-Lilly, K A and Broomall, S M and Bruce, D C and Chain, P | |
S and Coyne, S R and Freitas, T and Frey, K G and Gibbons, H S | |
and Jaissle, J and Lo, C-C and Meincke, L and Munk, A C and | |
Redden, C L and Rosenzweig, C N and Johnson, S L", | |
abstract = "Staphylococcus aureus is a major cause of bacterial infections | |
in the United States, with high percentages of serious | |
infections resistant to a variety of $\beta$-lactam antibiotics. | |
Here, we present the scaffolded genome assembly into 16 contigs | |
of S. aureus CDC73-57501 (ATCC 29247), a methicillin-resistant | |
quality control strain.", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 2, | |
number = 5, | |
month = oct, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Stevens2015-lt, | |
title = "Complete and Assembled Genome Sequence of Staphylococcus aureus | |
{RKI4}, a {Food-Poisoning} Strain Exhibiting a Novel S. aureus | |
Pathogenicity Island Carrying seb", | |
author = "Stevens, Marc J A and Stephan, Roger and Johler, Sophia", | |
abstract = "The genome of Staphylococcus aureus RKI4, a strain isolated from | |
feces of a patient in a case of staphylococcal food poisoning, | |
was sequenced using combined Illumina and single-molecule | |
real-time sequencing. Hierarchical assembly of the genome | |
resulted in a 2,725,654-bp chromosome and a 17,905-bp mobile | |
genetic element.", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 3, | |
number = 4, | |
month = jul, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Harrison2013-vq, | |
title = "Whole genome sequencing identifies zoonotic transmission of | |
{MRSA} isolates with the novel mecA homologue mecC", | |
author = "Harrison, Ewan M and Paterson, Gavin K and Holden, Matthew T G | |
and Larsen, Jesper and Stegger, Marc and Larsen, Anders Rhod and | |
Petersen, Andreas and Skov, Robert L and Christensen, Judit | |
Marta and Bak Zeuthen, Anne and Heltberg, Ole and Harris, Simon | |
R and Zadoks, Ruth N and Parkhill, Julian and Peacock, Sharon J | |
and Holmes, Mark A", | |
abstract = "Several methicillin-resistant Staphylococcus aureus (MRSA) | |
lineages that carry a novel mecA homologue (mecC) have recently | |
been described in livestock and humans. In Denmark, two | |
independent human cases of mecC-MRSA infection have been linked | |
to a livestock reservoir. We investigated the molecular | |
epidemiology of the associated MRSA isolates using whole genome | |
sequencing (WGS). Single nucleotide polymorphisms (SNP) were | |
defined and compared to a reference genome to place the isolates | |
into a phylogenetic context. Phylogenetic analysis revealed two | |
distinct farm-specific clusters comprising isolates from the | |
human case and their own livestock, whereas human and animal | |
isolates from the same farm only differed by a small number of | |
SNPs, which supports the likelihood of zoonotic transmission. | |
Further analyses identified a number of genes and mutations that | |
may be associated with host interaction and virulence. This | |
study demonstrates that mecC-MRSA ST130 isolates are capable of | |
transmission between animals and humans, and underscores the | |
potential of WGS in epidemiological investigations and source | |
tracking of bacterial infections.", | |
journal = "EMBO Mol. Med.", | |
publisher = "embomolmed.embopress.org", | |
volume = 5, | |
number = 4, | |
pages = "509--515", | |
month = apr, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Nair2011-sm, | |
title = "Whole-genome sequencing of Staphylococcus aureus strain | |
{RN4220}, a key laboratory strain used in virulence research, | |
identifies mutations that affect not only virulence factors but | |
also the fitness of the strain", | |
author = "Nair, Dhanalakshmi and Memmi, Guido and Hernandez, David and | |
Bard, Jonathan and Beaume, Marie and Gill, Steven and Francois, | |
Patrice and Cheung, Ambrose L", | |
abstract = "ABSTRACT Staphylococcus aureus RN4220, a cloning intermediate, | |
is sometimes used in virulence, resistance, and metabolic | |
studies. Using whole- genome sequencing, we showed that RN4220 | |
differs from NCTC8325 and contains a number of genetic | |
polymorphisms ...", | |
journal = "J. Bacteriol.", | |
publisher = "Am Soc Microbiol", | |
volume = 193, | |
number = 9, | |
pages = "2332--2335", | |
year = 2011, | |
keywords = "Staphylococcus;imported\_from\_papers\_14Nov2013;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Li2011-rd, | |
title = "Complete genome sequence of Staphylococcus aureus T0131, an | |
{ST239-MRSA-SCCmec} type {III} clone isolated in China", | |
author = "Li, Yang and Cao, Boyang and Zhang, Yongfu and Zhou, Jiali and | |
Yang, Bin and Wang, Lei", | |
abstract = "We report here the complete genome sequence of Staphylococcus | |
aureus T0131, which is a multiresistant clinical isolate | |
recovered in China and the first sequenced epidemic | |
ST239-MRSA-SCCmec type III strain obtained in Asia. Comparison | |
with two published genomes of ST239 reveals the polymorphism | |
among strains of this type from different continents.", | |
journal = "J. Bacteriol.", | |
publisher = "Am Soc Microbiol", | |
volume = 193, | |
number = 13, | |
pages = "3411--3412", | |
month = jul, | |
year = 2011, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Chen2014-tq, | |
title = "Reduced susceptibility to vancomycin in isogenic Staphylococcus | |
aureus strains of sequence type 59: tracking evolution and | |
identifying mutations by whole-genome sequencing", | |
author = "Chen, Chih-Jung and Lin, Mei-Hui and Shu, Jwu-Ching and Lu, | |
Jang-Jih", | |
abstract = "Objectives Vancomycin-intermediate Staphylococcus aureus (VISA) | |
and heterogeneous VISA (hVISA) phenotypes are increasingly | |
reported in methicillin-resistant S. aureus (MRSA) strains of | |
distinct genetic backgrounds. This study tracked genetic | |
evolution during the ...", | |
journal = "J. Antimicrob. Chemother.", | |
publisher = "Br Soc Antimicrob Chemo", | |
volume = 69, | |
number = 2, | |
pages = "349--354", | |
year = 2014, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Van_Der_Mee-Marquet2013-sr, | |
title = "Whole-genome sequence of the ancestral animal-borne {ST398} | |
Staphylococcus aureus strain {S123}", | |
author = "Van Der Mee-Marquet, Nathalie and Hernandez, David and Bertrand, | |
Xavier and Quentin, Roland and Corvaglia, Anna-Rita and Fran{\c | |
c}ois, Patrice", | |
abstract = "ABSTRACT Sequence type 398 (ST398) Staphylococcus aureus was | |
originally reported in livestock. We announce the complete | |
genome sequence of an ST398 methicillin-susceptible S. aureus | |
strain of animal origin, S123. The genome sequence reveals a | |
wild-type ...", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 1, | |
number = 5, | |
pages = "e00692--13", | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Diene2015-nq, | |
title = "Comparative genomics to investigate the emergence of | |
community-associated methicillin-resistant Staphylococcus aureus | |
({CA-MRSA}) {USA300} clone in Geneva, Switzerland", | |
author = "Diene, S M and Von Dach, E and Fankhauser, C and Bonetti, E-J | |
and Schrenzel, J and Harbarth, S and Fran{\c c}ois, P", | |
abstract = "... Comparative genomics to investigate the emergence of | |
community-associated methicillin-resistant Staphylococcus aureus | |
(CA-MRSA) USA300 clone in Geneva, Switzerland. ... | |
Solexa-Illumina was used for whole genome sequencing (WGS) of | |
all USA300 isolated in 2013. ...", | |
journal = "Antimicrob. Resist. Infect. Control", | |
publisher = "BioMed Central", | |
volume = 4, | |
number = "Suppl 1", | |
pages = "P197", | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Castillo-Ramirez2012-to, | |
title = "Phylogeographic variation in recombination rates within a global | |
clone of {Methicillin-Resistant} Staphylococcus aureus ({MRSA})", | |
author = "Castillo-Ram{\'\i}rez, Santiago and Corander, Jukka and | |
Marttinen, Pekka and Aldeljawi, Mona and Hanage, William P and | |
Westh, Henrik and Boye, Kit and Gulay, Zeynep and Bentley, | |
Stephen D and Parkhill, Julian and Holden, Matthew T and Feil, | |
Edward J", | |
abstract = "ABSTRACT: BACKGROUND: Next-generation sequencing (NGS) is a | |
powerful tool for understanding both patterns of descent over | |
time and space (phylogeography) and the molecular processes | |
underpinning genome divergence in pathogenic bacteria. Here, we | |
describe a synthesis between these perspectives by employing a | |
recently developed Bayesian approach, BRATNextGen, for detecting | |
recombination on an expanded NGS dataset of the globally | |
disseminated methicillin-resistant Staphylococcus aureus (MRSA) | |
clone ST239. RESULTS: The data confirm strong geographical | |
clustering at continental, national and city scales and | |
demonstrate that the rate of recombination varies significantly | |
between phylogeographic sub-groups representing independent | |
introductions from Europe. These differences are most striking | |
when mobile non-core genes are included, but remain apparent | |
even when only considering the stable core genome. The | |
monophyletic ST239 sub-group corresponding to isolates from | |
South America shows heightened recombination, the sub-group | |
predominantly from Asia shows an intermediate level, and a very | |
low level of recombination is noted in a third sub-group | |
representing a large collection from Turkey. CONCLUSIONS: We | |
show that the rapid global dissemination of a single pathogenic | |
bacterial clone results in local variation in measured | |
recombination rates. Possible explanatory variables include the | |
size and time since emergence of each defined sub-population (as | |
determined by the sampling frame), variation in transmission | |
dynamics due to host movement, and changes in the bacterial | |
genome affecting the propensity for recombination.", | |
journal = "Genome Biol.", | |
publisher = "biomedcentral.com", | |
volume = 13, | |
number = 12, | |
pages = "R126", | |
month = dec, | |
year = 2012, | |
keywords = " | |
Staphylococcus;recombination;imported\_from\_papers\_14Nov2013;Staph\_evolutionary\_history;bacterial | |
comparative genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Boyle-Vavra2011-vl, | |
title = "Comparative genome sequencing of an isogenic pair of {USA800} | |
clinical methicillin-resistant Staphylococcus aureus isolates | |
obtained before and after daptomycin treatment failure", | |
author = "Boyle-Vavra, Susan and Jones, Marcus and Gourley, Brett L and | |
Holmes, Michael and Ruf, Rebecca and Balsam, Ashley R and | |
Boulware, David R and Kline, Susan and Jawahir, Selina and | |
Devries, Aaron and Peterson, Scott N and Daum, Robert S", | |
abstract = "We describe here a clinical daptomycin treatment failure in a | |
patient with recurrent methicillin-resistant Staphylococcus | |
aureus (MRSA) bacteremia in whom daptomycin was administered | |
after a failed empirical treatment course with vancomycin and | |
piperacillin-tazobactam. We had the opportunity to compare the | |
genome sequences of an isogenic pair of daptomycin-susceptible | |
and -resistant MRSA isolates obtained before and after | |
initiation of daptomycin therapy, respectively. The genotype of | |
both isolates was USA800, ST5, SCCmec type IV, agr type II. | |
There was no increase in cell wall thickness in the | |
daptomycin-resistant strain despite having decreased | |
susceptibility to both vancomycin and daptomycin. By comparing | |
the genome sequences by pyrosequencing, we identified a | |
polymorphism (S337L) in the tenth transmembrane segment of the | |
multiple peptide resistance factor, MprF, encoding lysyl | |
phosphatidylglycerol transferase. This enzyme has been shown | |
previously to promote repulsion of daptomycin at the cell | |
surface by addition of positively charged lysine to | |
phosphatidylglycerol. Also, the hlb open reading frame (ORF) | |
encoding the $\beta$-toxin was interrupted by a prophage in the | |
daptomycin-susceptible strain; this phage was missing in the | |
daptomycin-resistant isolate and the hlb ORF was restored. Loss | |
of the phage in the resistant isolate also resulted in loss of | |
the virulence factor genes clpP, scn, and sak. This is the first | |
study to use pyrosequencing to compare the genomes of a | |
daptomycin-susceptible/resistant MRSA isolate pair obtained | |
during failed daptomycin therapy in humans.", | |
journal = "Antimicrob. Agents Chemother.", | |
publisher = "Am Soc Microbiol", | |
volume = 55, | |
number = 5, | |
pages = "2018--2025", | |
month = may, | |
year = 2011, | |
keywords = " | |
Staphylococcus;phage;confirmed\_SA-resistance-mutation;imported\_from\_papers\_14Nov2013;bacterial | |
comparative genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Judge2015-kn, | |
title = "Early insights into the potential of the Oxford Nanopore {MinION} | |
for the detection of antimicrobial resistance genes", | |
author = "Judge, Kim and Harris, Simon R and Reuter, Sandra and Parkhill, | |
Julian and Peacock, Sharon J", | |
abstract = "OBJECTIVES: Genome sequencing will be increasingly used in the | |
clinical setting to tailor antimicrobial prescribing and inform | |
infection control outbreaks. A recent technological innovation | |
that could reduce the delay between pathogen sampling and data | |
generation is single molecule sequencing. An example of this | |
technology, which is undergoing evaluation through an early | |
access programme, is the Oxford Nanopore MinION. METHODS: We | |
undertook a feasibility study on six clinically significant | |
pathogens, comparing the MinION to the Illumina MiSeq and PacBio | |
RSII platforms. Genomic DNA was prepared and sequenced using the | |
MinION as instructed by the manufacturer, and Illumina MiSeq and | |
PacBio sequencing was performed using established methods. | |
RESULTS: An evaluation of the accuracy of the MinION based on | |
sequencing of an MRSA isolate showed that error rates were higher | |
in the MinION reads, but provided an even coverage across the | |
entire genome length. The MinION detected all of the expected | |
carbapenemases and ESBL genes in five Gram-negative isolates and | |
the mecA gene in an MRSA isolate. CONCLUSIONS: The MinION can | |
detect the presence of acquired resistance genes, but | |
improvements in accuracy are needed so that antimicrobial | |
resistance associated with mutations in chromosomal genes can be | |
identified.", | |
journal = "J. Antimicrob. Chemother.", | |
volume = 70, | |
number = 10, | |
pages = "2775--2778", | |
month = oct, | |
year = 2015, | |
keywords = "antibiotics;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Meric2015-oq, | |
title = "Ecological Overlap and Horizontal Gene Transfer in Staphylococcus | |
aureus and Staphylococcus epidermidis", | |
author = "M{\'e}ric, Guillaume and Miragaia, Maria and de Been, Mark and | |
Yahara, Koji and Pascoe, Ben and Mageiros, Leonardos and Mikhail, | |
Jane and Harris, Llinos G and Wilkinson, Thomas S and Rolo, Joana | |
and Lamble, Sarah and Bray, James E and Jolley, Keith A and | |
Hanage, William P and Bowden, Rory and Maiden, Martin C J and | |
Mack, Dietrich and de Lencastre, Herm{\'\i}nia and Feil, Edward J | |
and Corander, Jukka and Sheppard, Samuel K", | |
abstract = "The opportunistic pathogens Staphylococcus aureus and | |
Staphylococcus epidermidis represent major causes of severe | |
nosocomial infection, and are associated with high levels of | |
mortality and morbidity worldwide. These species are both common | |
commensals on the human skin and in the nasal pharynx, but are | |
genetically distinct, differing at 24\% average nucleotide | |
divergence in 1,478 core genes. To better understand the genome | |
dynamics of these ecologically similar staphylococcal species, we | |
carried out a comparative analysis of 324 S. aureus and S. | |
epidermidis genomes, including 83 novel S. epidermidis sequences. | |
A reference pan-genome approach and whole genome | |
multilocus-sequence typing revealed that around half of the | |
genome (1,478 genes) was shared between the species. Based on a | |
BratNextGen analysis, homologous recombination was found to have | |
impacted on 40\% of the core genes in S. epidermidis, but on only | |
24\% of the core genes in S. aureus. Homologous recombination | |
between the species is rare, with a maximum of 9 gene alleles | |
shared between any two S. epidermidis and S. aureus isolates. In | |
contrast, considerable interspecies admixture is noted in mobile | |
elements, in particular genes associated with the SaPIn1 | |
pathogenicity island, metal detoxification, and the | |
methicillin-resistance island SCCmec. Our data and analysis | |
provide a context for considering the nature of recombinational | |
boundaries between S. aureus and S. epidermidis and, the | |
selective forces that influence realized recombination between | |
these species.", | |
journal = "Genome Biol. Evol.", | |
volume = 7, | |
number = 5, | |
pages = "1313--1328", | |
month = apr, | |
year = 2015, | |
keywords = "Staphylococcus; ecology; evolution; nosocomial infections; | |
recombination;Staphylococcus;plasmid;recombination;pangenome;Staph\_evolutionary\_history;Staph\_evolutionary\_history/staph | |
microbial interactions;bacterial comparative | |
genomics;Read-lab-shared;Papers\_for\_Michelle;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers" | |
} | |
@ARTICLE{Bradley2015-zy, | |
title = "Rapid antibiotic-resistance predictions from genome sequence data | |
for Staphylococcus aureus and Mycobacterium tuberculosis", | |
author = "Bradley, Phelim and Gordon, N Claire and Walker, Timothy M and | |
Dunn, Laura and Heys, Simon and Huang, Bill and Earle, Sarah and | |
Pankhurst, Louise J and Anson, Luke and de Cesare, Mariateresa | |
and Piazza, Paolo and Votintseva, Antonina A and Golubchik, Tanya | |
and Wilson, Daniel J and Wyllie, David H and Diel, Roland and | |
Niemann, Stefan and Feuerriegel, Silke and Kohl, Thomas A and | |
Ismail, Nazir and Omar, Shaheed V and Smith, E Grace and Buck, | |
David and McVean, Gil and Walker, A Sarah and Peto, Tim E A and | |
Crook, Derrick W and Iqbal, Zamin", | |
abstract = "The rise of antibiotic-resistant bacteria has led to an urgent | |
need for rapid detection of drug resistance in clinical samples, | |
and improvements in global surveillance. Here we show how de | |
Bruijn graph representation of bacterial diversity can be used to | |
identify species and resistance profiles of clinical isolates. We | |
implement this method for Staphylococcus aureus and Mycobacterium | |
tuberculosis in a software package ('Mykrobe predictor') that | |
takes raw sequence data as input, and generates a | |
clinician-friendly report within 3 minutes on a laptop. For S. | |
aureus, the error rates of our method are comparable to | |
gold-standard phenotypic methods, with sensitivity/specificity of | |
99.1\%/99.6\% across 12 antibiotics (using an independent | |
validation set, n=470). For M. tuberculosis, our method predicts | |
resistance with sensitivity/specificity of 82.6\%/98.5\% | |
(independent validation set, n=1,609); sensitivity is lower here, | |
probably because of limited understanding of the underlying | |
genetic mechanisms. We give evidence that minor alleles improve | |
detection of extremely drug-resistant strains, and demonstrate | |
feasibility of the use of emerging single-molecule nanopore | |
sequencing techniques for these purposes.", | |
journal = "Nat. Commun.", | |
volume = 6, | |
pages = "10063", | |
month = dec, | |
year = 2015, | |
keywords = "Staphylococcus;antibiotics;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Garvey2016-cd, | |
title = "Improved understanding of an outbreak of meticillin-resistant | |
Staphylococcus aureus in a regional burns centre via whole-genome | |
sequencing", | |
author = "Garvey, M I and Pichon, B and Bradley, C W and Moiemen, N S and | |
Oppenheim, B and Kearns, A M", | |
journal = "J. Hosp. Infect.", | |
month = sep, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Bunter2016-ot, | |
title = "Wild type agr-negative livestock-associated {MRSA} exhibits high | |
adhesive capacity to human and porcine cells", | |
author = "B{\"u}nter, Julia P and Seth-Smith, Helena M B and R{\"u}egg, | |
Simon and Heikinheimo, Annamari and Borel, Nicole and Johler, | |
Sophia", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) is one of the | |
leading causes of nosocomial infections and a major public health | |
concern worldwide. During the last decade, MRSA of CC398 have | |
emerged as important colonizers of livestock. These strains also | |
represent an increasing cause of human infections. A recent study | |
reporting a new dominant spa type among MRSA from Finish | |
fattening pigs (CC398/t2741) identified a strain lacking both the | |
global virulence regulator gene locus agr and the adhesion gene | |
fnbB. The aim of this study was to characterize this | |
agr/fnbB-negative livestock-associated MRSA strain in terms of | |
growth, hemolysis and adhesive capacity, and to provide data on | |
its genomic background. To this end, growth curves and hemolysis | |
patterns were generated and adhesion assays on human keratinocyte | |
and porcine nasal mucosa cell lines were performed. Whole genome | |
sequencing was used to determine the nature and extent of the | |
relevant deletions in the livestock strains. For comparison, an | |
agr-positive, fnbB-negative CC398/t2741 strain from the same pig | |
herd, an agr/fnbB- positive CC398/t034 strain from another pig | |
herd and one human MRSA strain and its isogenic $\Delta$agr | |
knockout mutant were used. The agr-negative strains adhered | |
significantly better to human and porcine host cells than the | |
agr-positive control strains. For the agr-positive porcine MRSA | |
strains, cytotoxic effects on porcine mucosal cells were | |
observed. The strong adhesive capacity of the naturally | |
agr-negative livestock-associated MRSA, in combination with | |
diminished cytotoxic effects, is likely favorable for inducing | |
persistent colonization in pigs. Independently of the host cell | |
type, similar adhesive capacities of the naturally agr-negative | |
livestock-associated MRSA and the human MRSA strain were shown. | |
Our results indicate that loss of agr in the livestock-associated | |
MRSA strain investigated in this study may have increased its | |
potential to be transmitted to and amongst humans.", | |
journal = "Res. Microbiol.", | |
month = oct, | |
year = 2016, | |
keywords = "Adhesion; Livestock; MRSA; Staphylococcus aureus; | |
agr;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Senn2016-of, | |
title = "The Stealthy Superbug: the Role of Asymptomatic Enteric Carriage | |
in Maintaining a {Long-Term} Hospital Outbreak of {ST228} | |
{Methicillin-Resistant} Staphylococcus aureus", | |
author = "Senn, Laurence and Clerc, Olivier and Zanetti, Giorgio and | |
Basset, Patrick and Prod'hom, Guy and Gordon, Nicola C and | |
Sheppard, Anna E and Crook, Derrick W and James, Richard and | |
Thorpe, Harry A and Feil, Edward J and Blanc, Dominique S", | |
abstract = "UNLABELLED: Whole-genome sequencing (WGS) of 228 isolates was | |
used to elucidate the origin and dynamics of a long-term outbreak | |
of methicillin-resistant Staphylococcus aureus (MRSA) sequence | |
type 228 (ST228) SCCmec I that involved 1,600 patients in a | |
tertiary care hospital between 2008 and 2012. Combining of the | |
sequence data with detailed metadata on patient admission and | |
movement confirmed that the outbreak was due to the transmission | |
of a single clonal variant of ST228, rather than repeated | |
introductions of this clone into the hospital. We note that this | |
clone is significantly more frequently recovered from groin and | |
rectal swabs than other clones (P < 0.0001) and is also | |
significantly more transmissible between roommates (P < 0.01). | |
Unrecognized MRSA carriers, together with movements of patients | |
within the hospital, also seem to have played a major role. These | |
atypical colonization and transmission dynamics can help explain | |
how the outbreak was maintained over the long term. This | |
``stealthy'' asymptomatic colonization of the gut, combined with | |
heightened transmissibility (potentially reflecting a role for | |
environmental reservoirs), means the dynamics of this outbreak | |
share some properties with enteric pathogens such as | |
vancomycin-resistant enterococci or Clostridium difficile. | |
IMPORTANCE: Using whole-genome sequencing, we showed that a large | |
and prolonged outbreak of methicillin-resistant Staphylococcus | |
aureus was due to the clonal spread of a specific strain with | |
genetic elements adapted to the hospital environment. | |
Unrecognized MRSA carriers, the movement of patients within the | |
hospital, and the low detection with clinical specimens were also | |
factors that played a role in this occurrence. The atypical | |
colonization of the gut means the dynamics of this outbreak may | |
share some properties with enteric pathogens.", | |
journal = "MBio", | |
volume = 7, | |
number = 1, | |
pages = "e02039--15", | |
month = jan, | |
year = 2016, | |
keywords = " | |
Staphylococcus;Staph\_evolutionary\_history;Staph\_evolutionary\_history/staph | |
microbial interactions;bacterial comparative | |
genomics;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;Staph\_evolution\_toxins", | |
language = "en" | |
} | |
@ARTICLE{Ishii2015-ur, | |
title = "Phenotypic and genomic comparisons of highly | |
vancomycin-resistant Staphylococcus aureus strains developed | |
from multiple clinical {MRSA} strains by in vitro mutagenesis", | |
author = "Ishii, Kenichi and Tabuchi, Fumiaki and Matsuo, Miki and | |
Tatsuno, Keita and Sato, Tomoaki and Okazaki, Mitsuhiro and | |
Hamamoto, Hiroshi and Matsumoto, Yasuhiko and Kaito, Chikara and | |
Aoyagi, Tetsuji and Hiramatsu, Keiichi and Kaku, Mitsuo and | |
Moriya, Kyoji and Sekimizu, Kazuhisa", | |
abstract = "The development of vancomycin (VCM) resistance in Staphylococcus | |
aureus threatens global health. Studies of the VCM-resistance | |
mechanism and alternative therapeutic strategies are urgently | |
needed. We mutagenized S. aureus laboratory strains and | |
methicillin-resistant S. aureus (MRSA) with ethyl | |
methanesulfonate, and isolated mutants that exhibited high | |
resistance to VCM (minimum inhibitory concentration = 32 | |
$\mu$g/ml). These VCM-resistant strains were sensitive to | |
linezolid and rifampicin, and partly to arbekacin and | |
daptomycin. Beta-lactams had synergistic effects with VCM | |
against these mutants. VCM-resistant strains exhibited a 2-fold | |
increase in the cell wall thickness. Several genes were commonly | |
mutated among the highly VCM-resistant mutants. These findings | |
suggest that MRSA has a potential to develop high VCM resistance | |
with cell wall thickening by the accumulation of mutations.", | |
journal = "Sci. Rep.", | |
publisher = "ncbi.nlm.nih.gov", | |
volume = 5, | |
pages = "17092", | |
month = nov, | |
year = 2015, | |
keywords = "confirmed\_SA-resistance-mutation;Staphylococcus;antibiotics;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Chang2016-oa, | |
title = "Identifying the effect of patient sharing on between-hospital | |
genetic differentiation of methicillin-resistant Staphylococcus | |
aureus", | |
author = "Chang, Hsiao-Han and Dordel, Janina and Donker, Tjibbe and Worby, | |
Colin J and Feil, Edward J and Hanage, William P and Bentley, | |
Stephen D and Huang, Susan S and Lipsitch, Marc", | |
abstract = "BACKGROUND: Methicillin-resistant Staphylococcus aureus (MRSA) is | |
one of the most common healthcare-associated pathogens. To | |
examine the role of inter-hospital patient sharing on MRSA | |
transmission, a previous study collected 2,214 samples from 30 | |
hospitals in Orange County, California and showed by spa typing | |
that genetic differentiation decreased significantly with | |
increased patient sharing. In the current study, we focused on | |
the 986 samples with spa type t008 from the same population. | |
METHODS: We used genome sequencing to determine the effect of | |
patient sharing on genetic differentiation between hospitals. | |
Genetic differentiation was measured by between-hospital genetic | |
diversity, F ST , and the proportion of nearly identical isolates | |
between hospitals. RESULTS: Surprisingly, we found very similar | |
genetic diversity within and between hospitals, and no | |
significant association between patient sharing and genetic | |
differentiation measured by F ST . However, in contrast to F ST , | |
there was a significant association between patient sharing and | |
the proportion of nearly identical isolates between hospitals. We | |
propose that the proportion of nearly identical isolates is more | |
powerful at determining transmission dynamics than traditional | |
estimators of genetic differentiation (F ST ) when gene flow | |
between populations is high, since it is more responsive to | |
recent transmission events. Our hypothesis was supported by the | |
results from coalescent simulations. CONCLUSIONS: Our results | |
suggested that there was a high level of gene flow between | |
hospitals facilitated by patient sharing, and that the proportion | |
of nearly identical isolates is more sensitive to population | |
structure than F ST when gene flow is high.", | |
journal = "Genome Med.", | |
volume = 8, | |
number = 1, | |
pages = "18", | |
month = feb, | |
year = 2016, | |
keywords = "Staphylococcus;Staph\_evolutionary\_history;bacterial comparative | |
genomics;Teaching antibiotic resistance | |
genomics;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers" | |
} | |
@ARTICLE{Salipante2015-qz, | |
title = "Application of whole-genome sequencing for bacterial strain | |
typing in molecular epidemiology", | |
author = "Salipante, Stephen J and SenGupta, Dhruba J and Cummings, Lisa A | |
and Land, Tyler A and Hoogestraat, Daniel R and Cookson, Brad T", | |
abstract = "Nosocomial infections pose a significant threat to patient | |
health; however, the gold standard laboratory method for | |
determining bacterial relatedness (pulsed-field gel | |
electrophoresis [PFGE]) remains essentially unchanged 20 years | |
after its introduction. Here, we explored bacterial whole-genome | |
sequencing (WGS) as an alternative approach for molecular strain | |
typing. We compared WGS to PFGE for investigating presumptive | |
outbreaks involving three important pathogens: | |
vancomycin-resistant Enterococcus faecium (n=19), | |
methicillin-resistant Staphylococcus aureus (n=17), and | |
Acinetobacter baumannii (n=15). WGS was highly reproducible | |
(average$\leq$0.39 differences between technical replicates), | |
which enabled a functional, quantitative definition for | |
determining clonality. Strain relatedness data determined by PFGE | |
and WGS roughly correlated, but the resolution of WGS was | |
superior (P=5.6$\times$10(-8) to 0.016). Several discordant | |
results were noted between the methods. A total of 28.9\% of | |
isolates which were indistinguishable by PFGE were nonclonal by | |
WGS. For A. baumannii, a species known to undergo rapid | |
horizontal gene transfer, 16.2\% of isolate pairs considered | |
nonidentical by PFGE were clonal by WGS. Sequencing whole | |
bacterial genomes with single-nucleotide resolution demonstrates | |
that PFGE is prone to false-positive and false-negative results | |
and suggests the need for a new gold standard approach for | |
molecular epidemiological strain typing.", | |
journal = "J. Clin. Microbiol.", | |
volume = 53, | |
number = 4, | |
pages = "1072--1079", | |
month = apr, | |
year = 2015, | |
keywords = "experimental\_design;Staphylococcus;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Croucher2016-vz, | |
title = "Horizontal {DNA} Transfer Mechanisms of Bacteria as Weapons of | |
Intragenomic Conflict", | |
author = "Croucher, Nicholas J and Mostowy, Rafal and Wymant, Christopher | |
and Turner, Paul and Bentley, Stephen D and Fraser, Christophe", | |
abstract = "Horizontal DNA transfer (HDT) is a pervasive mechanism of | |
diversification in many microbial species, but its primary | |
evolutionary role remains controversial. Much recent research has | |
emphasised the adaptive benefit of acquiring novel DNA, but here | |
we argue instead that intragenomic conflict provides a coherent | |
framework for understanding the evolutionary origins of HDT. To | |
test this hypothesis, we developed a mathematical model of a | |
clonally descended bacterial population undergoing HDT through | |
transmission of mobile genetic elements (MGEs) and genetic | |
transformation. Including the known bias of transformation toward | |
the acquisition of shorter alleles into the model suggested it | |
could be an effective means of counteracting the spread of MGEs. | |
Both constitutive and transient competence for transformation | |
were found to provide an effective defence against parasitic | |
MGEs; transient competence could also be effective at permitting | |
the selective spread of MGEs conferring a benefit on their host | |
bacterium. The coordination of transient competence with | |
cell-cell killing, observed in multiple species, was found to | |
result in synergistic blocking of MGE transmission through | |
releasing genomic DNA for homologous recombination while | |
simultaneously reducing horizontal MGE spread by lowering the | |
local cell density. To evaluate the feasibility of the functions | |
suggested by the modelling analysis, we analysed genomic data | |
from longitudinal sampling of individuals carrying Streptococcus | |
pneumoniae. This revealed the frequent within-host coexistence of | |
clonally descended cells that differed in their MGE infection | |
status, a necessary condition for the proposed mechanism to | |
operate. Additionally, we found multiple examples of MGEs | |
inhibiting transformation through integrative disruption of genes | |
encoding the competence machinery across many species, providing | |
evidence of an ongoing ``arms race.'' Reduced rates of | |
transformation have also been observed in cells infected by MGEs | |
that reduce the concentration of extracellular DNA through | |
secretion of DNases. Simulations predicted that either mechanism | |
of limiting transformation would benefit individual MGEs, but | |
also that this tactic's effectiveness was limited by competition | |
with other MGEs coinfecting the same cell. A further observed | |
behaviour we hypothesised to reduce elimination by transformation | |
was MGE activation when cells become competent. Our model | |
predicted that this response was effective at counteracting | |
transformation independently of competing MGEs. Therefore, this | |
framework is able to explain both common properties of MGEs, and | |
the seemingly paradoxical bacterial behaviours of transformation | |
and cell-cell killing within clonally related populations, as the | |
consequences of intragenomic conflict between self-replicating | |
chromosomes and parasitic MGEs. The antagonistic nature of the | |
different mechanisms of HDT over short timescales means their | |
contribution to bacterial evolution is likely to be substantially | |
greater than previously appreciated.", | |
journal = "PLoS Biol.", | |
volume = 14, | |
number = 3, | |
pages = "e1002394", | |
month = mar, | |
year = 2016, | |
keywords = "systems\_biology;evolution;recombination;simulated-evolution;theory;Staph\_evolutionary\_history/Teaching\_Prokaryotic\_evolution;Staph\_evolutionary\_history;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Wang2014-uw, | |
title = "Comparative genomics of methicillin-resistant Staphylococcus | |
aureus {ST239}: distinct geographical variants in Beijing and | |
Hong Kong", | |
author = "Wang, Zheng and Zhou, Haokui and Wang, Hui and Chen, Hongbin and | |
Leung, K K and Tsui, Stephen and Ip, Margaret", | |
abstract = "BACKGROUND: The ST239 lineage is a globally disseminated, | |
multiply drug-resistant hospital-associated methicillin-resistant | |
Staphylococcus aureus (HA-MRSA). We performed whole-genome | |
sequencing of representative HA-MRSA isolates of the ST239 | |
lineage from bacteremic patients in hospitals in Hong Kong (HK) | |
and Beijing (BJ) and compared them with three published complete | |
genomes of ST239, namely T0131, TW20 and JKD6008. Orthologous | |
gene group (OGG) analyses of the Hong Kong and Beijing cluster | |
strains were also undertaken. RESULTS: Homology analysis, based | |
on highest-percentage nucleotide identity, indicated that HK | |
isolates were closely related to TW20, whereas BJ isolates were | |
more closely related to T0131 from Tianjin. Phylogenetic | |
analysis, incorporating a total of 30 isolates from different | |
continents, revealed that strains from HK clustered with TW20 | |
into the 'Asian clade', whereas BJ isolates and T0131 clustered | |
closely with strains of the 'Turkish clade' from Eastern Europe. | |
HK isolates contained the typical $\varphi$SP$\beta$-like | |
prophage with the SasX gene similar to TW20. In contrast, BJ | |
isolates contained a unique 15 kb PT1028-like prophage but lacked | |
$\varphi$SP$\beta$-like and $\varphi$SA1 prophages. Besides | |
distinct mobile genetic elements (MGE) in the two clusters, OGG | |
analyses and whole-genome alignment of these clusters highlighted | |
differences in genes located in the core genome, including the | |
identification of single nucleotide deletions in several genes, | |
resulting in frameshift mutations and the subsequent predicted | |
truncation of encoded proteins involved in metabolism and | |
antimicrobial resistance. CONCLUSIONS: Comparative genomics, | |
based on de novo assembly and deep sequencing of HK and BJ | |
strains, revealed different origins of the ST239 lineage in | |
northern and southern China and identified differences between | |
the two clades at single nucleotide polymorphism (SNP), core gene | |
and MGE levels. The results suggest that ST239 strains isolated | |
in Hong Kong since the 1990s belong to the Asian clade, present | |
mainly in southern Asia, whereas those that emerged in northern | |
China were of a distinct origin, reflecting the complexity of | |
dissemination and the dynamic evolution of this ST239 lineage.", | |
journal = "BMC Genomics", | |
volume = 15, | |
pages = "529", | |
month = jun, | |
year = 2014, | |
keywords = "Staphylococcus;bacterial comparative | |
genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Mader2016-da, | |
title = "Staphylococcus aureus Transcriptome Architecture: From Laboratory | |
to {Infection-Mimicking} Conditions", | |
author = "M{\"a}der, Ulrike and Nicolas, Pierre and Depke, Maren and | |
Pan{\'e}-Farr{\'e}, Jan and Debarbouille, Michel and van der | |
Kooi-Pol, Magdalena M and Gu{\'e}rin, Cyprien and D{\'e}rozier, | |
Sandra and Hiron, Aurelia and Jarmer, Hanne and Leduc, | |
Aur{\'e}lie and Michalik, Stephan and Reilman, Ewoud and | |
Schaffer, Marc and Schmidt, Frank and Bessi{\`e}res, Philippe and | |
Noirot, Philippe and Hecker, Michael and Msadek, Tarek and | |
V{\"o}lker, Uwe and van Dijl, Jan Maarten", | |
abstract = "Staphylococcus aureus is a major pathogen that colonizes about | |
20\% of the human population. Intriguingly, this Gram-positive | |
bacterium can survive and thrive under a wide range of different | |
conditions, both inside and outside the human body. Here, we | |
investigated the transcriptional adaptation of S. aureus HG001, a | |
derivative of strain NCTC 8325, across experimental conditions | |
ranging from optimal growth in vitro to intracellular growth in | |
host cells. These data establish an extensive repertoire of | |
transcription units and non-coding RNAs, a classification of 1412 | |
promoters according to their dependence on the RNA polymerase | |
sigma factors SigA or SigB, and allow identification of new | |
potential targets for several known transcription factors. In | |
particular, this study revealed a relatively low abundance of | |
antisense RNAs in S. aureus, where they overlap only 6\% of the | |
coding genes, and only 19 antisense RNAs not co-transcribed with | |
other genes were found. Promoter analysis and comparison with | |
Bacillus subtilis links the small number of antisense RNAs to a | |
less profound impact of alternative sigma factors in S. aureus. | |
Furthermore, we revealed that Rho-dependent transcription | |
termination suppresses pervasive antisense transcription, | |
presumably originating from abundant spurious transcription | |
initiation in this A+T-rich genome, which would otherwise affect | |
expression of the overlapped genes. In summary, our study | |
provides genome-wide information on transcriptional regulation | |
and non-coding RNAs in S. aureus as well as new insights into the | |
biological function of Rho and the implications of spurious | |
transcription in bacteria.", | |
journal = "PLoS Genet.", | |
volume = 12, | |
number = 4, | |
pages = "e1005962", | |
month = apr, | |
year = 2016, | |
keywords = "Staphylococcus;rna-seq;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers;microRNA\_in\_bacteria" | |
} | |
@ARTICLE{Carroll2013-gh, | |
title = "Draft Genome Sequence of Strain {CBD-635}, a | |
{Methicillin-Resistant} Staphylococcus aureus {USA100} Isolate", | |
author = "Carroll, Ronan K and Burda, Whittney N and Roberts, Jill C and | |
Peak, Kealy K and Cannons, Andrew C and Shaw, Lindsey N", | |
abstract = "We present the draft genome sequence of methicillin-resistant | |
Staphylococcus aureus strain CBD-635, from the USA100 lineage. | |
This is a sepsis isolate obtained from Tampa General Hospital. | |
This strain is spa type t003 and multilocus sequence typing | |
(MLST) type ST5, and it has been used by our group in the study | |
of novel antimicrobial chemotherapeutics.", | |
journal = "Genome Announc.", | |
volume = 1, | |
number = 4, | |
month = jul, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Wendt2014-oh, | |
title = "Transmission of methicillin-resistant Staphylococcus aureus | |
infection through solid organ transplantation: confirmation via | |
whole genome sequencing", | |
author = "Wendt, J M and Kaul, D and Limbago, B M and Ramesh, M and Cohle, | |
S and Denison, A M and Driebe, E M and Rasheed, J K and Zaki, S R | |
and Blau, D M and Paddock, C D and McDougal, L K and Engelthaler, | |
D M and Keim, P S and Roe, C C and Akselrod, H and Kuehnert, M J | |
and Basavaraju, S V", | |
abstract = "We describe two cases of donor-derived methicillin-resistant | |
Staphylococcus aureus (MRSA) bacteremia that developed after | |
transplantation of organs from a common donor who died from acute | |
MRSA endocarditis. Both recipients developed recurrent MRSA | |
infection despite appropriate antibiotic therapy, and required | |
prolonged hospitalization and hospital readmission. Comparison of | |
S. aureus whole genome sequence of DNA extracted from fixed donor | |
tissue and recipients' isolates confirmed donor-derived | |
transmission. Current guidelines emphasize the risk posed by | |
donors with bacteremia from multidrug-resistant organisms. This | |
investigation suggests that, particularly in the setting of donor | |
endocarditis, even a standard course of prophylactic antibiotics | |
may not be sufficient to prevent donor-derived infection.", | |
journal = "Am. J. Transplant", | |
volume = 14, | |
number = 11, | |
pages = "2633--2639", | |
month = nov, | |
year = 2014, | |
keywords = "Staphylococcus;Staph\_evolutionary\_history;bacterial comparative | |
genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Tong2015-xf, | |
title = "Genome sequencing defines phylogeny and spread of | |
methicillin-resistant Staphylococcus aureus in a high | |
transmission setting", | |
author = "Tong, Steven Y C and Holden, Matthew T G and Nickerson, Emma K | |
and Cooper, Ben S and K{\"o}ser, Claudio U and Cori, Anne and | |
Jombart, Thibaut and Cauchemez, Simon and Fraser, Christophe and | |
Wuthiekanun, Vanaporn and Thaipadungpanit, Janjira and Hongsuwan, | |
Maliwan and Day, Nicholas P and Limmathurotsakul, Direk and | |
Parkhill, Julian and Peacock, Sharon J", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) is a major | |
cause of nosocomial infection. Whole-genome sequencing of MRSA | |
has been used to define phylogeny and transmission in | |
well-resourced healthcare settings, yet the greatest burden of | |
nosocomial infection occurs in resource-restricted settings where | |
barriers to transmission are lower. Here, we study the flux and | |
genetic diversity of MRSA on ward and individual patient levels | |
in a hospital where transmission was common. We repeatedly | |
screened all patients on two intensive care units for MRSA | |
carriage over a 3-mo period. All MRSA belonged to multilocus | |
sequence type 239 (ST 239). We defined the population structure | |
and charted the spread of MRSA by sequencing 79 isolates from 46 | |
patients and five members of staff, including the first | |
MRSA-positive screen isolates and up to two repeat isolates where | |
available. Phylogenetic analysis identified a flux of distinct ST | |
239 clades over time in each intensive care unit. In total, five | |
main clades were identified, which varied in the carriage of | |
plasmids encoding antiseptic and antimicrobial resistance | |
determinants. Sequence data confirmed intra- and interwards | |
transmission events and identified individual patients who were | |
colonized by more than one clade. One patient on each unit was | |
the source of numerous transmission events, and deep sampling of | |
one of these cases demonstrated colonization with a ``cloud'' of | |
related MRSA variants. The application of whole-genome sequencing | |
and analysis provides novel insights into the transmission of | |
MRSA in under-resourced healthcare settings and has relevance to | |
wider global health.", | |
journal = "Genome Res.", | |
volume = 25, | |
number = 1, | |
pages = "111--118", | |
month = jan, | |
year = 2015, | |
keywords = "Staphylococcus;bacterial comparative | |
genomics;Staph\_evolutionary\_history;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Hernandez2013-xb, | |
title = "{Whole-Genome} Sequences of Staphylococcus aureus {ST398} | |
Strains of Animal Origin", | |
author = "Hernandez, David and van der Mee-Marquet, Nathalie and | |
Kluytmans, Jan and Donnio, Pierre-Yves and Quentin, Roland and | |
Corvaglia, Anna-Rita and Fran{\c c}ois, Patrice", | |
abstract = "Staphylococcus aureus sequence type 398 (ST398) was originally | |
associated with animal infections. We announce the complete | |
genome sequences of two ST398 methicillin-susceptible S. aureus | |
strains from the livestock environment. These genome sequences | |
assist in the characterization of interesting ST398 features | |
relying on host tropism and epidemiological settings.", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 1, | |
number = 5, | |
month = sep, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Barrado2013-dt, | |
title = "Draft Genome Sequence of Strain {SA\_ST125\_MupR} of | |
{Methicillin-Resistant} Staphylococcus aureus {ST125}, a Major | |
Clone in Spain", | |
author = "Barrado, Laura and Viedma, Esther and Vindel, Ana and Otero, | |
Joaqu{\'\i}n R and Chaves, Fernando", | |
abstract = "Here, we report the draft genome sequence of a | |
methicillin-resistant Staphylococcus aureus (MRSA) strain with | |
high-level mupirocin resistance (SA\_ST125\_MupR), isolated from | |
a patient with recurrent bacteremia. This strain belonged to | |
sequence type ST125, which was responsible for more than 50\% of | |
the health care-associated infections caused by MRSA in Spain.", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 1, | |
number = 4, | |
month = aug, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Stegger2013-lf, | |
title = "Genome analysis of Staphylococcus aureus {ST291}, a double locus | |
variant of {ST398}, reveals a distinct genetic lineage", | |
author = "Stegger, Marc and Aziz, Maliha and Chroboczek, Tomasz and Price, | |
Lance B and Ronco, Troels and Kiil, Kristoffer and Skov, Robert | |
L and Laurent, Frederic and Andersen, Paal S", | |
abstract = "Staphylococcus aureus ST291 has been reported as a homologue | |
recombinant double locus variant of the livestock associated S. | |
aureus ST398. However, whole genome sequencing show that ST291 | |
is a unique genetic lineage with highly variable content within | |
its accessory genome compared to both human and livestock | |
associated genome sequenced CC398s.", | |
journal = "PLoS One", | |
publisher = "dx.plos.org", | |
volume = 8, | |
number = 5, | |
pages = "e63008", | |
month = may, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Wang2012-hj, | |
title = "Whole-genome sequence of Staphylococcus aureus strain | |
{LCT-SA112}", | |
author = "Wang, Junfeng and Liu, Yanhong and Wan, Daiwei and Fang, | |
Xiangqun and Li, Tianzhi and Guo, Yinghua and Chang, De and Su, | |
Longxiang and Wang, Yajuan and Zhao, Jiao and Liu, Changting", | |
abstract = "Staphylococcus aureus is a facultative anaerobic Gram-positive | |
coccal bacterium. S. aureus is the most common species of | |
Staphylococcus to cause staphylococcal infections, which are | |
very common in clinical medicine. Here we report the genome | |
sequence of S. aureus strain LCT-SA112, which was isolated from | |
S. aureus subsp. aureus CGMCC 1.230.", | |
journal = "J. Bacteriol.", | |
publisher = "Am Soc Microbiol", | |
volume = 194, | |
number = 15, | |
pages = "4124", | |
month = aug, | |
year = 2012, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Golding2012-pq, | |
title = "whole-genome sequence of livestock-associated st398 | |
methicillin-resistant staphylococcus aureus Isolated from Humans | |
in Canada", | |
author = "Golding, George R and Bryden, Louis and Levett, Paul N and | |
McDonald, Ryan R and Wong, Alice and Graham, Morag R and Tyler, | |
Shaun and Van Domselaar, Gary and Mabon, Philip and Kent, | |
Heather and Butaye, Patrick and Smith, Tara C and Kadlec, | |
Kristina and Schwarz, Stefan and Weese, Scott J and Mulvey, | |
Michael R", | |
abstract = "Despite reports of high colonization rates of ST398 | |
livestock-associated methicillin-resistant Staphylococcus aureus | |
(LA-MRSA) among pigs and pig farmers, the incidence of LA-MRSA | |
infection in the general population in Canada appears to be rare | |
in comparison to that in some European countries. In this study, | |
the complete genome sequence of a Canadian representative | |
LA-MRSA isolate (08BA02176) from a human postoperative surgical | |
site infection was acquired and compared to the sequenced genome | |
of an LA-MRSA isolate (S0385) from Europe to identify genetic | |
traits that may explain differences in the success of these | |
particular strains in some locales.", | |
journal = "J. Bacteriol.", | |
publisher = "Am Soc Microbiol", | |
volume = 194, | |
number = 23, | |
pages = "6627--6628", | |
month = dec, | |
year = 2012, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{Prabhakara2012-aq, | |
title = "Draft genome sequence of Staphylococcus aureus 118 ({ST772)}, a | |
major disease clone from India", | |
author = "Prabhakara, Sushma and Khedkar, Supriya and Loganathan, Ramya | |
Malarini and Chandana, S and Gowda, Malali and Arakere, Gayathri | |
and Seshasayee, Aswin Sai Narain", | |
abstract = "We report the draft genome sequence of an ST772 Staphylococcus | |
aureus disease isolate carrying staphylococcal cassette | |
chromosome mec (SCCmec) type V from a pyomyositis patient. Our | |
de novo short read assembly is ∼2.8 Mb and encodes a unique | |
Panton-Valentine leukocidin (PVL) phage with structural genes | |
similar to those of ϕ7247PVL and novel lysogenic genes at the N | |
termini.", | |
journal = "J. Bacteriol.", | |
publisher = "Am Soc Microbiol", | |
volume = 194, | |
number = 14, | |
pages = "3727--3728", | |
month = jul, | |
year = 2012, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Peleg2012-gx, | |
title = "Whole genome characterization of the mechanisms of daptomycin | |
resistance in clinical and laboratory derived isolates of | |
Staphylococcus aureus", | |
author = "Peleg, Anton Y and Miyakis, Spiros and Ward, Doyle V and Earl, | |
Ashlee M and Rubio, Aileen and Cameron, David R and Pillai, | |
Satish and Moellering, Jr, Robert C and Eliopoulos, George M", | |
abstract = "BACKGROUND: Daptomycin remains one of our last-line | |
anti-staphylococcal agents. This study aims to characterize the | |
genetic evolution to daptomycin resistance in S. aureus. | |
METHODS: Whole genome sequencing was performed on a unique | |
collection of isogenic, clinical (21 strains) and laboratory (12 | |
strains) derived strains that had been exposed to daptomycin and | |
developed daptomycin-nonsusceptibility. Electron microscopy (EM) | |
and lipid membrane studies were performed on selected isolates. | |
RESULTS: On average, six coding region mutations were observed | |
across the genome in the clinical daptomycin exposed strains, | |
whereas only two mutations on average were seen in the | |
laboratory exposed pairs. All daptomycin-nonsusceptible strains | |
had a mutation in a phospholipid biosynthesis gene. This | |
included mutations in the previously described mprF gene, but | |
also in other phospholipid biosynthesis genes, including | |
cardiolipin synthase (cls2) and | |
CDP-diacylglycerol-glycerol-3-phosphate | |
3-phosphatidyltransferase (pgsA). EM and lipid membrane | |
composition analyses on two clinical pairs showed that the | |
daptomycin-nonsusceptible strains had a thicker cell wall and an | |
increase in membrane lysyl-phosphatidylglycerol. CONCLUSION: | |
Point mutations in genes coding for membrane phospholipids are | |
associated with the development of reduced susceptibility to | |
daptomycin in S. aureus. Mutations in cls2 and pgsA appear to be | |
new genetic mechanisms affecting daptomycin susceptibility in S. | |
aureus.", | |
journal = "PLoS One", | |
publisher = "dx.plos.org", | |
volume = 7, | |
number = 1, | |
pages = "e28316", | |
month = jan, | |
year = 2012, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Gasc2016-sq, | |
title = "Genome Sequence of Staphylococcus aureus Strain {HUK16}, Isolated | |
from {Hexachlorocyclohexane-Contaminated} Soil", | |
author = "Gasc, Cyrielle and Richard, Jean-Yves and Peyret, Pierre", | |
abstract = "Staphylococcus aureusstrain HUK16 has been isolated from | |
hexachlorocyclohexane (HCH)-long-term contaminated soil. The | |
genome of strain HUK16 was sequenced to understand the genetic | |
basis of its adaptation to HCH and to find the potential | |
metabolic pathways allowing it to degrade the pesticide. Here, we | |
report the annotated draft genome sequence (~2.7 Mbp) of this | |
strain.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 2, | |
month = apr, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Hishinuma2016-wu, | |
title = "Complete Genome Sequence of {Vancomycin-Intermediate} | |
Staphylococcus aureus Strain {MI} ({HIP5827})", | |
author = "Hishinuma, Tomomi and Katayama, Yuki and Matsuo, Miki and Sasaki, | |
Takashi and Hiramatsu, Keiichi", | |
abstract = "We report the complete genome sequence of vancomycin-intermediate | |
Staphylococcus aureus (VISA) strain MI (HIP5827).", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 2, | |
month = mar, | |
year = 2016, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Marques2016-wq, | |
title = "Genome Sequence of a Clinical Staphylococcus aureus Strain from a | |
Prosthetic Joint Infection", | |
author = "Marqu{\`e}s, Claire and Franceschi, Christine and Collin, | |
Val{\'e}rie and Laurent, Fr{\'e}d{\'e}ric and Chatellier, Sonia | |
and Forestier, Christiane", | |
abstract = "Here, we report the genome sequence ofStaphylococcus | |
aureusLYO-S2, an isolate with sequence type (ST) 45 that was | |
isolated in 2001 from a prosthetic joint infection.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 2, | |
month = apr, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Silva2016-il, | |
title = "Erratum for Silva et al., Draft Genome Sequences of | |
Staphylococcus aureus Strains Isolated from Subclinical Bovine | |
Mastitis in Brazil", | |
author = "Silva, Danielle Mendes and da Silva, M{\^o}nica Pacheco and | |
Vidigal, Pedro M Pereira and Barcelos, Rafael Mazioli and Klein, | |
Raphael Contelli and Aguilar, Ananda Pereira and Fabres-Klein, | |
Mary Hellen and Oliveira, Guilherme and Ribon, Andr{\'e}a | |
Oliveira Barros", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 2, | |
month = apr, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Abaev2016-sf, | |
title = "Draft Genome Sequences of Exfoliative Toxin {A-Producing} | |
Staphylococcus aureus Strains {B-7772} and {B-7777} | |
({CC8/ST2993}) and {B-7774} ({CC15/ST2126)}, Isolated in a | |
Maternity Hospital in the Central Federal District of Russia", | |
author = "Abaev, Igor and Skryabin, Yury and Kislichkina, Angelina and | |
Bogun, Alexandr and Korobova, Olga and Mayskaya, Nadezhda and | |
Shemyakin, Igor and Dyatlov, Ivan", | |
abstract = "Staphylococcus aureus clonal complex 8 (CC8) has not been | |
associated with staphylococcal scalded-skin syndrome (SSSS) in | |
newborns and exfoliative toxin genes. Here, we report the draft | |
genome sequences of exfoliative toxin A-producing B-7772, B-7777 | |
(both CC8), and B-7774 (CC15) strains associated with SSSS in | |
newborns.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 2, | |
month = mar, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Nielsen2016-bg, | |
title = "Fatal Septicemia Linked to Transmission of {MRSA} Clonal Complex | |
398 in Hospital and Nursing Home, Denmark", | |
author = "Nielsen, Rikke Thoft and Kemp, Michael and Holm, Anette and Skov, | |
Marianne Nielsine and Detlefsen, Mette and Hasman, Henrik and | |
Aarestrup, Frank M{\o}ller and Kaas, Rolf Sommer and Nielsen, | |
Jesper Boye and Westh, Henrik and Kolmos, Hans J{\o}rn", | |
abstract = "We describe 2 fatal cases of methicillin-resistant Staphylococcus | |
aureus (MRSA) clonal complex 398 septicemia in persons who had no | |
contact with livestock. Whole-genome sequencing of the isolated | |
MRSA strains strongly suggest that both were of animal origin and | |
that the patients had been infected through 2 independent | |
person-to-person transmission chains.", | |
journal = "Emerg. Infect. Dis.", | |
volume = 22, | |
number = 5, | |
pages = "900--902", | |
month = may, | |
year = 2016, | |
keywords = "antimicrobial resistance; bacteremia; bacteria; bacterial | |
infections; disease transmission; infectious; livestock; | |
methicillin-resistant Staphylococcus aureus; molecular | |
epidemiology; spa typing; | |
staphylococci;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Stine2016-na, | |
title = "Transmission Clusters of {Methicillin-Resistant} Staphylococcus | |
Aureus in {Long-Term} Care Facilities Based on {Whole-Genome} | |
Sequencing", | |
author = "Stine, O Colin and Burrowes, Shana and David, Sophia and Johnson, | |
J Kristie and Roghmann, Mary-Claire", | |
abstract = "OBJECTIVE To define how often methicillin-resistant | |
Staphylococcus aureus (MRSA) is spread from resident to resident | |
in long-term care facilities using whole-genome sequencing DESIGN | |
Prospective cohort study SETTING A long-term care facility | |
PARTICIPANTS Elderly residents in a long-term care facility | |
METHODS Cultures for MRSA were obtained weekly from multiple body | |
sites from residents with known MRSA colonization over 12-week | |
study periods. Simultaneously, cultures to detect MRSA | |
acquisition were obtained weekly from 2 body sites in residents | |
without known MRSA colonization. During the first 12-week cycle | |
on a single unit, we sequenced 8 MRSA isolates per swab for 2 | |
body sites from each of 6 residents. During the second 12-week | |
cycle, we sequenced 30 MRSA isolates from 13 residents with known | |
MRSA colonization and 3 residents who had acquired MRSA | |
colonization. RESULTS MRSA isolates from the same swab showed | |
little genetic variation between isolates with the exception of | |
isolates from wounds. The genetic variation of isolates between | |
body sites on an individual was greater than that within a single | |
body site with the exception of 1 sample, which had 2 unrelated | |
strains among the 8 isolates. In the second cycle, 10 of 16 | |
residents colonized with MRSA (63\%) shared 1 of 3 closely | |
related strains. Of the 3 residents with newly acquired MRSA, 2 | |
residents harbored isolates that were members of these clusters. | |
CONCLUSIONS Point prevalence surveys with whole-genome sequencing | |
of MRSA isolates may detect resident-to-resident transmission | |
more accurately than routine surveillance cultures for MRSA in | |
long-term care facilities. Infect. Control Hosp. Epidemiol. | |
2016;1-7.", | |
journal = "Infect. Control Hosp. Epidemiol.", | |
pages = "1--7", | |
month = mar, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Davis2016-zc, | |
title = "Complete genome of Staphylococcus aureus Tager 104 provides | |
evidence of its relation to modern systemic hospital-acquired | |
strains", | |
author = "Davis, 4th, Richard W and Brannen, Andrew D and Hossain, Mohammad | |
J and Monsma, Scott and Bock, Paul E and Nahrendorf, Matthias and | |
Mead, David and Lodes, Michael and Liles, Mark R and Panizzi, | |
Peter", | |
abstract = "BACKGROUND: Staphylococcus aureus (S. aureus) infections range in | |
severity due to expression of certain virulence factors encoded | |
on mobile genetic elements (MGE). As such, characterization of | |
these MGE, as well as single nucleotide polymorphisms, is of high | |
clinical and microbiological importance. To understand the | |
evolution of these dangerous pathogens, it is paramount to define | |
reference strains that may predate MGE acquisition. One such | |
candidate is S. aureus Tager 104, a previously uncharacterized | |
strain isolated from a patient with impetigo in 1947. RESULTS: We | |
show here that S. aureus Tager 104 can survive in the bloodstream | |
and infect na{\"\i}ve organs. We also demonstrate a procedure to | |
construct and validate the assembly of S. aureus genomes, using | |
Tager 104 as a proof-of-concept. In so doing, we bridged | |
confounding gap regions that limited our initial attempts to | |
close this 2.82 Mb genome, through integration of data from | |
Illumina Nextera paired-end, PacBio RS, and Lucigen NxSeq | |
mate-pair libraries. Furthermore, we provide independent | |
confirmation of our segmental arrangement of the Tager 104 genome | |
by the sole use of Lucigen NxSeq libraries filled by paired-end | |
MiSeq reads and alignment with SPAdes software. Genomic analysis | |
of Tager 104 revealed limited MGE, and a $\nu$Sa$\beta$ island | |
configuration that is reminiscent of other hospital acquired S. | |
aureus genomes. CONCLUSIONS: Tager 104 represents an | |
early-branching ancestor of certain hospital-acquired strains. | |
Combined with its earlier isolation date and limited content of | |
MGE, Tager 104 can serve as a viable reference for future | |
comparative genome studies.", | |
journal = "BMC Genomics", | |
volume = 17, | |
number = 1, | |
pages = "179", | |
month = mar, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Silva2016-rh, | |
title = "Draft Genome Sequences of Staphylococcus aureus Strains Isolated | |
from Subclinical Bovine Mastitis in Brazil", | |
author = "Silva, Danielle Mendes and da Silva, M{\^o}nica Pacheco and | |
Vidigal, Pedro M Pereira and Barcelos, Rafael Mazioli and Klein, | |
Raphael Contelli and Aguilar, Ananda Pereira and Fabres-Klein, | |
Mary Hellen and Oliveira, Guilherme and Ribon, Andr{\'e}a | |
Oliveira Barros", | |
abstract = "Here, we present the draft genome sequences of four | |
Staphylococcus aureus strains isolated from mastitic milk | |
collected from animals with subclinical manifestations. Three of | |
them were typed as sequence type 126 (ST126), a genotype with no | |
genome sequence available. ST126 is found in several herds of | |
southern Brazil and is described as a bovine pathogen strongly | |
associated with milk around the world.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 1, | |
month = feb, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Greninger2016-yp, | |
title = "{Whole-Genome} Sequencing of {Methicillin-Resistant} | |
Staphylococcus aureus Resistant to {Fifth-Generation} | |
Cephalosporins Reveals Potential {Non-mecA} Mechanisms of | |
Resistance", | |
author = "Greninger, Alexander L and Chatterjee, Som S and Chan, Liana C | |
and Hamilton, Stephanie M and Chambers, Henry F and Chiu, Charles | |
Y", | |
abstract = "Fifth-generation cephalosporins, ceftobiprole and ceftaroline, | |
are promising drugs for treatment of bacterial infections from | |
methicillin-resistant Staphylococcus aureus (MRSA). These | |
antibiotics are able to bind native PBP2a, the penicillin-binding | |
protein encoded by the mecA resistance determinant that mediates | |
broad class resistance to nearly all other beta-lactam | |
antibiotics, at clinically achievable concentrations. Mechanisms | |
of resistance to ceftaroline based on mecA mutations have been | |
previously described. Here we compare the genomes of 11 total | |
parent-daughter strains of Staphylococcus aureus for which | |
specific selection by serial passaging with ceftaroline or | |
ceftobiprole was used to identify novel non-mecA mechanisms of | |
resistance. All 5 ceftaroline-resistant strains, derived from 5 | |
different parental strains, contained mutations directly upstream | |
of the pbp4 gene (coding for the PBP4 protein), including four | |
with the same thymidine insertion located 377 nucleotides | |
upstream of the promoter site. In 4 of 5 independent | |
ceftaroline-driven selections, we also isolated mutations to the | |
same residue (Asn138) in PBP4. In addition, mutations in | |
additional candidate genes such as ClpX endopeptidase, PP2C | |
protein phosphatase and transcription terminator Rho, previously | |
undescribed in the context of resistance to ceftaroline or | |
ceftobiprole, were detected in multiple selections. These genomic | |
findings suggest that non-mecA mechanisms, while yet to be | |
encountered in the clinical setting, may also be important in | |
mediating resistance to 5th-generation cephalosporins.", | |
journal = "PLoS One", | |
volume = 11, | |
number = 2, | |
pages = "e0149541", | |
month = feb, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Khan2016-ez, | |
title = "Draft Genome Sequences of Two {Methicillin-Resistant} Clinical | |
Staphylococcus aureus Isolates", | |
author = "Khan, Saeed and Sung, Kidon and Iram, Saira and Nawaz, Mohamed | |
and Xu, Joshua and Marasa, Bernard", | |
abstract = "Here, we report the draft genome sequences of two | |
methicillin-resistant Staphylococcus aureus (MRSA) clinical | |
isolates, hospital-associated perirectal isolate 32S (ST 239) | |
from a colitis tracheostomy patient and community-associated MRSA | |
isolate 42S (ST 772) from a hepatic-splenomegaly patient in | |
Rawalpindi, Pakistan.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 1, | |
month = feb, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Holmes2016-yn, | |
title = "Genomic Analysis of Companion Rabbit Staphylococcus aureus", | |
author = "Holmes, Mark A and Harrison, Ewan M and Fisher, Elizabeth A and | |
Graham, Elizabeth M and Parkhill, Julian and Foster, Geoffrey and | |
Paterson, Gavin K", | |
abstract = "In addition to being an important human pathogen, Staphylococcus | |
aureus is able to cause a variety of infections in numerous other | |
host species. While the S. aureus strains causing infection in | |
several of these hosts have been well characterised, this is not | |
the case for companion rabbits (Oryctolagus cuniculus), where | |
little data are available on S. aureus strains from this host. To | |
address this deficiency we have performed antimicrobial | |
susceptibility testing and genome sequencing on a collection of | |
S. aureus isolates from companion rabbits. The findings show a | |
diverse S. aureus population is able to cause infection in this | |
host, and while antimicrobial resistance was uncommon, the | |
isolates possess a range of known and putative virulence factors | |
consistent with a diverse clinical presentation in companion | |
rabbits including severe abscesses. We additionally show that | |
companion rabbit isolates carry polymorphisms within dltB as | |
described as underlying host-adaption of S. aureus to farmed | |
rabbits. The availability of S. aureus genome sequences from | |
companion rabbits provides an important aid to understanding the | |
pathogenesis of disease in this host and in the clinical | |
management and surveillance of these infections.", | |
journal = "PLoS One", | |
volume = 11, | |
number = 3, | |
pages = "e0151458", | |
month = mar, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Kong2016-eb, | |
title = "{Whole-Genome} Sequencing for the Investigation of a Hospital | |
Outbreak of {MRSA} in China", | |
author = "Kong, Zhenzhen and Zhao, Peipei and Liu, Haibing and Yu, Xiang | |
and Qin, Yanyan and Su, Zhaoliang and Wang, Shengjun and Xu, | |
Huaxi and Chen, Jianguo", | |
abstract = "Staphylococcus aureus is a globally disseminated drug-resistant | |
bacterial species. It remains a leading cause of | |
hospital-acquired infection, primarily among immunocompromised | |
patients. In 2012, the Affiliated People's Hospital of Jiangsu | |
University experienced a putative outbreak of | |
methicillin-resistant S. aureus (MRSA) that affected 12 patients | |
in the Neurosurgery Department. In this study, whole-genome | |
sequencing (WGS) was used to gain insight into the epidemiology | |
of the outbreak caused by MRSA, and traditional bacterial | |
genotyping approaches were also applied to provide supportive | |
evidence for WGS. We sequenced the DNA from 6 isolates associated | |
with the outbreak. Phylogenetic analysis was constructed by | |
comparing single-nucleotide polymorphisms (SNPs) in the core | |
genome of 6 isolates in the present study and another 3 | |
referenced isolates from GenBank. Of the 6 MRSA sequences in the | |
current study, 5 belonged to the same group, clustering with | |
T0131, while the other one clustered closely with TW20. All of | |
the isolates were identified as ST239-SCCmecIII clones. | |
Whole-genome analysis revealed that four of the outbreak isolates | |
were more tightly clustered into a group and SA13002 together | |
with SA13009 were distinct from the outbreak strains, which were | |
considered non-outbreak strains. Based on the sequencing results, | |
the antibiotic-resistance gene status (present or absent) was | |
almost perfectly concordant with the results of phenotypic | |
susceptibility testing. Various toxin genes were also analyzed | |
successfully. Our analysis demonstrates that using traditional | |
molecular methods and WGS can facilitate the identification of | |
outbreaks and help to control nosocomial transmission.", | |
journal = "PLoS One", | |
volume = 11, | |
number = 3, | |
pages = "e0149844", | |
month = mar, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Sidjabat2015-nx, | |
title = "Draft Genome Sequences of Burkholderia pseudomallei and | |
Staphylococcus aureus, Isolated from a Patient with Chronic | |
Rhinosinusitis", | |
author = "Sidjabat, Hanna E and Cottrell, Kyra and Cervin, Anders", | |
abstract = "Here, we report the draft genome sequences of Burkholderia | |
pseudomallei and Staphylococcus aureus causing chronic | |
rhinosinusitis. Whole-genome sequencing determined the B. | |
pseudomallei as sequence type (ST) 1381 and the S. aureus as ST8. | |
B. pseudomallei possessed the blaOXA-59 gene. This study | |
illustrates the potential emergence of B. pseudomallei in cases | |
of chronic rhinosinusitis.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 5, | |
month = oct, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Sharma2015-mr, | |
title = "Draft Genome Sequence of a Staphylococcus aureus Strain Isolated | |
from a Cow with Clinical Mastitis", | |
author = "Sharma, Paresh and Reddy, D Peddi and Kumar, P Anand and | |
Gadicherla, Ramya and George, Neena and Bhandari, Vasundhra", | |
abstract = "We report here the draft genome of Staphylococcus aureus causing | |
clinical mastitis in a cow from India. It is a major causative | |
agent of mastitis and, further, livestock-associated strains are | |
emerging as a potential threat to public health, thereby | |
warranting studies to understand the genome of this deadly | |
pathogen.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 4, | |
month = aug, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Soni2015-tm, | |
title = "Draft Genome Sequence of {Methicillin-Sensitive} Staphylococcus | |
aureus {ATCC} 29213", | |
author = "Soni, Isha and Chakrapani, Harinath and Chopra, Sidharth", | |
abstract = "Staphylococcus aureus subsp. aureus ATCC 29213 is one of the most | |
commonly used strains in drug discovery research and for quality | |
control. We report the completed draft genome sequence for the | |
strain.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 5, | |
month = sep, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Marasa2015-ve, | |
title = "Draft Genome Sequence of {Methicillin-Resistant} Clinical | |
Staphylococcus aureus Isolate {51S} (Sequence Type 291)", | |
author = "Marasa, Bernard S and Khan, Saeed and Iram, Saira and Sung, Kidon | |
and Xu, Joshua", | |
abstract = "We report the draft genome sequence of a methicillin-resistant | |
clinical Staphylococcus aureus isolate with a novel spa type and | |
sequence type (ST291), isolated from a renal failure patient in | |
Rawalpindi, Pakistan.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 5, | |
month = sep, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Budd2015-ry, | |
title = "Extensive Genomic Diversity among {Bovine-Adapted} Staphylococcus | |
aureus: Evidence for a Genomic Rearrangement within {CC97}", | |
author = "Budd, Kathleen E and McCoy, Finola and Monecke, Stefan and | |
Cormican, Paul and Mitchell, Jennifer and Keane, Orla M", | |
abstract = "Staphylococcus aureus is an important pathogen associated with | |
both human and veterinary disease and is a common cause of bovine | |
mastitis. Genomic heterogeneity exists between S. aureus strains | |
and has been implicated in the adaptation of specific strains to | |
colonise particular mammalian hosts. Knowledge of the factors | |
required for host specificity and virulence is important for | |
understanding the pathogenesis and management of S. aureus | |
mastitis. In this study, a panel of mastitis-associated S. aureus | |
isolates (n = 126) was tested for resistance to antibiotics | |
commonly used to treat mastitis. Over half of the isolates (52\%) | |
demonstrated resistance to penicillin and ampicillin but all were | |
susceptible to the other antibiotics tested. S. aureus isolates | |
were further examined for their clonal diversity by Multi-Locus | |
Sequence Typing (MLST). In total, 18 different sequence types | |
(STs) were identified and eBURST analysis demonstrated that the | |
majority of isolates grouped into clonal complexes CC97, CC151 or | |
sequence type (ST) 136. Analysis of the role of recombination | |
events in determining S. aureus population structure determined | |
that ST diversification through nucleotide substitutions were | |
more likely to be due to recombination compared to point | |
mutation, with regions of the genome possibly acting as | |
recombination hotspots. DNA microarray analysis revealed a large | |
number of differences amongst S. aureus STs in their variable | |
genome content, including genes associated with capsule and | |
biofilm formation and adhesion factors. Finally, evidence for a | |
genomic arrangement was observed within isolates from CC97 with | |
the ST71-like subgroup showing evidence of an IS431 insertion | |
element having replaced approximately 30 kb of DNA including the | |
ica operon and histidine biosynthesis genes, resulting in | |
histidine auxotrophy. This genomic rearrangement may be | |
responsible for the diversification of ST71 into an emerging | |
bovine adapted subgroup.", | |
journal = "PLoS One", | |
volume = 10, | |
number = 8, | |
pages = "e0134592", | |
month = aug, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Shiroma2015-ho, | |
title = "First Complete Genome Sequences of Staphylococcus aureus subsp. | |
aureus Rosenbach 1884 ({DSM} 20231T), Determined by {PacBio} | |
{Single-Molecule} {Real-Time} Technology", | |
author = "Shiroma, Akino and Terabayashi, Yasunobu and Nakano, Kazuma and | |
Shimoji, Makiko and Tamotsu, Hinako and Ashimine, Noriko and | |
Ohki, Shun and Shinzato, Misuzu and Teruya, Kuniko and Satou, | |
Kazuhito and Hirano, Takashi", | |
abstract = "The first complete genome sequences of Staphylococcus aureus | |
subsp. aureus Rosenbach 1884 strain DSM 20231(T), the type strain | |
of the bacterium causing staphylococcal disease, were determined | |
using PacBio RS II. The sequences represent the chromosome | |
(2,755,072 bp long; G+C content, 32.86\%) and a plasmid (27,490 | |
bp long; G+C content, 30.69\%).", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 4, | |
month = jul, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Stephens2015-kc, | |
title = "Draft Genome Sequence of a {Community-Associated} | |
{Methicillin-Resistant} {Panton-Valentine} {Leukocidin-Positive} | |
Staphylococcus aureus Sequence Type 30 Isolate from a Pediatric | |
Patient with a Lung Infection in Brazil", | |
author = "Stephens, Craig and Cho, Paul Jang-Yeon and Afonso de Araujo, | |
Veronica and Gomes, Ivete Martins and de Azevedo Sias, Selma | |
Maria and Ara{\'u}jo Cardoso, Claudete A and Riley, Lee W and | |
Aguiar-Alves, F{\'a}bio", | |
abstract = "The sequence of methicillin-resistant Staphylococcus aureus | |
strain B6 (sequence type 30 [ST30], spa type t433, staphylococcal | |
chromosomal cassette mec element [SCCmec] type IVc, | |
Panton-Valentine leukocidin [PVL] positive), isolated from a | |
pediatric patient with a lung infection in Niter{\'o}i, Rio de | |
Janeiro, Brazil, is described here. The draft genome sequence | |
includes a 2.8-Mb chromosome, accompanied by a 20-kb plasmid | |
containing blaZ and two small cryptic plasmids.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 4, | |
month = aug, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Nemet2015-mo, | |
title = "Draft Genome Sequence of a Highly Virulent Rabbit Staphylococcus | |
aureus Strain", | |
author = "N{\'e}met, Zolt{\'a}n and Albert, Ervin and Nagy, Tibor and | |
Olasz, Ferenc and Barta, Endre and Kiss, J{\'a}nos and D{\'a}n, | |
{\'A}d{\'a}m and B{\'a}nyai, Kriszti{\'a}n and Hermans, Katleen | |
and Biksi, Imre", | |
abstract = "We report the draft genome sequence of Staphylococcus aureus | |
Sp17, a typical highly virulent (HV) rabbit strain. As current | |
medicine apparently fails to effectively reduce disease and | |
economical losses caused by this organism, it is essential to | |
gain better insight on its genomic arrangement.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 4, | |
month = jul, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Driebe2015-ia, | |
title = "Using Whole Genome Analysis to Examine Recombination across | |
Diverse Sequence Types of Staphylococcus aureus", | |
author = "Driebe, Elizabeth M and Sahl, Jason W and Roe, Chandler and | |
Bowers, Jolene R and Schupp, James M and Gillece, John D and | |
Kelley, Erin and Price, Lance B and Pearson, Talima R and Hepp, | |
Crystal M and Brzoska, Pius M and Cummings, Craig A and Furtado, | |
Manohar R and Andersen, Paal S and Stegger, Marc and Engelthaler, | |
David M and Keim, Paul S", | |
abstract = "Staphylococcus aureus is an important clinical pathogen worldwide | |
and understanding this organism's phylogeny and, in particular, | |
the role of recombination, is important both to understand the | |
overall spread of virulent lineages and to characterize | |
outbreaks. To further elucidate the phylogeny of S. aureus, 35 | |
diverse strains were sequenced using whole genome sequencing. In | |
addition, 29 publicly available whole genome sequences were | |
included to create a single nucleotide polymorphism (SNP)-based | |
phylogenetic tree encompassing 11 distinct lineages. All strains | |
of a particular sequence type fell into the same clade with clear | |
groupings of the major clonal complexes of CC8, CC5, CC30, CC45 | |
and CC1. Using a novel analysis method, we plotted the homoplasy | |
density and SNP density across the whole genome and found | |
evidence of recombination throughout the entire chromosome, but | |
when we examined individual clonal lineages we found very little | |
recombination. However, when we analyzed three branches of | |
multiple lineages, we saw intermediate and differing levels of | |
recombination between them. These data demonstrate that in S. | |
aureus, recombination occurs across major lineages that | |
subsequently expand in a clonal manner. Estimated mutation rates | |
for the CC8 and CC5 lineages were different from each other. | |
While the CC8 lineage rate was similar to previous studies, the | |
CC5 lineage was 100-fold greater. Fifty known virulence genes | |
were screened in all genomes in silico to determine their | |
distribution across major clades. Thirty-three genes were present | |
variably across clades, most of which were not constrained by | |
ancestry, indicating horizontal gene transfer or gene loss.", | |
journal = "PLoS One", | |
volume = 10, | |
number = 7, | |
pages = "e0130955", | |
month = jul, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Cameron2015-vs, | |
title = "Draft Genome Sequences of Clinical {Daptomycin-Nonsusceptible} | |
{Methicillin-Resistant} Staphylococcus aureus Strain {APS211} and | |
Its {Daptomycin-Susceptible} Progenitor {APS210}", | |
author = "Cameron, David R and Jiang, Jhih-Hang and Abbott, Iain J and | |
Spelman, Denis W and Peleg, Anton Y", | |
abstract = "To assess the genetic factors contributing to daptomycin | |
resistance in Staphylococcus aureus, the draft genome of a | |
clinically derived daptomycin-nonsusceptible isolate APS211 was | |
generated and compared to the draft sequence of its susceptible | |
progenitor strain APS210. Four genetic differences were | |
identified including a previously described mutation within the | |
mprF gene.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 3, | |
month = jun, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Crovadore2015-vb, | |
title = "{Whole-Genome} Sequences of 15 Strains of Staphylococcus aureus | |
subsp. aureus Isolated from Foodstuff and Human Clinical Samples", | |
author = "Crovadore, Julien and Calmin, Gautier and Tonacini, Jenna and | |
Chablais, Romain and Baumgartner, Andreas and Schnyder, Bruno and | |
Hodille, Elisabeth and Lefort, Fran{\c c}ois", | |
abstract = "The whole-genome sequences of 15 strains of Staphylococcus aureus | |
(10 strains isolated from foodstuff samples in Switzerland and | |
five from human clinical samples) were obtained by Illumina | |
sequencing. Most strains fit within the known diversity for the | |
species, but one (SA-120) possessed a higher G+C content and a | |
higher number of genes than usual.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 3, | |
month = jun, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{Botka2015-wl, | |
title = "Complete genome analysis of two new bacteriophages isolated from | |
impetigo strains of Staphylococcus aureus", | |
author = "Botka, Tibor and R{\r u}{\v z}i{\v c}kov{\'a}, Vladislava and | |
Kone{\v c}n{\'a}, Hana and Pant{\r u}{\v c}ek, Roman and | |
Rychl{\'\i}k, Ivan and Zdr{\'a}hal, Zbyn{\v e}k and Petr{\'a}{\v | |
s}, Petr and Do{\v s}ka{\v r}, Ji{\v r}{\'\i}", | |
abstract = "Exfoliative toxin A (ETA)-coding temperate bacteriophages are | |
leading contributors to the toxic phenotype of impetigo strains | |
of Staphylococcus aureus. Two distinct eta gene-positive | |
bacteriophages isolated from S. aureus strains which recently | |
caused massive outbreaks of pemphigus neonatorum in Czech | |
maternity hospitals were characterized. The phages, designated | |
ϕB166 and ϕB236, were able to transfer the eta gene into a | |
prophageless S. aureus strain which afterwards converted into an | |
ETA producer. Complete phage genome sequences were determined, | |
and a comparative analysis of five designed genomic regions | |
revealed major variances between them. They differed in the | |
genome size, number of open reading frames, genome architecture, | |
and virion protein patterns. Their high mutual sequence | |
similarity was detected only in the terminal regions of the | |
genome. When compared with the so far described eta phage | |
genomes, noticeable differences were found. Thus, both phages | |
represent two new lineages of as yet not characterized | |
bacteriophages of the Siphoviridae family having impact on | |
pathogenicity of impetigo strains of S. aureus.", | |
journal = "Virus Genes", | |
volume = 51, | |
number = 1, | |
pages = "122--131", | |
month = aug, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Planet2015-jk, | |
title = "Parallel Epidemics of {Community-Associated} | |
{Methicillin-Resistant} Staphylococcus aureus {USA300} Infection | |
in North and South America", | |
author = "Planet, Paul J and Diaz, Lorena and Kolokotronis, Sergios-Orestis | |
and Narechania, Apurva and Reyes, Jinnethe and Xing, Galen and | |
Rincon, Sandra and Smith, Hannah and Panesso, Diana and Ryan, | |
Chanelle and Smith, Dylan P and Guzman, Manuel and Zurita, | |
Jeannete and Sebra, Robert and Deikus, Gintaras and Nolan, Rathel | |
L and Tenover, Fred C and Weinstock, George M and Robinson, D | |
Ashley and Arias, Cesar A", | |
abstract = "BACKGROUND: The community-associated methicillin-resistant | |
Staphylococcus aureus (CA-MRSA) epidemic in the United States is | |
attributed to the spread of the USA300 clone. An epidemic of | |
CA-MRSA closely related to USA300 has occurred in northern South | |
America (USA300 Latin-American variant, USA300-LV). Using | |
phylogenomic analysis, we aimed to understand the relationships | |
between these 2 epidemics. METHODS: We sequenced the genomes of | |
51 MRSA clinical isolates collected between 1999 and 2012 from | |
the United States, Colombia, Venezuela, and Ecuador. Phylogenetic | |
analysis was used to infer the relationships and times since the | |
divergence of the major clades. RESULTS: Phylogenetic analyses | |
revealed 2 dominant clades that segregated by geographical | |
region, had a putative common ancestor in 1975, and originated in | |
1989, in North America, and in 1985, in South America. Emergence | |
of these parallel epidemics coincides with the independent | |
acquisition of the arginine catabolic mobile element (ACME) in | |
North American isolates and a novel copper and mercury resistance | |
(COMER) mobile element in South American isolates. CONCLUSIONS: | |
Our results reveal the existence of 2 parallel USA300 epidemics | |
that shared a recent common ancestor. The simultaneous rapid | |
dissemination of these 2 epidemic clades suggests the presence of | |
shared, potentially convergent adaptations that enhance fitness | |
and ability to spread.", | |
journal = "J. Infect. Dis.", | |
volume = 212, | |
number = 12, | |
pages = "1874--1882", | |
month = dec, | |
year = 2015, | |
keywords = "MRSA; USA300; USA300-LV; | |
epidemics;Staphylococcus;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers" | |
} | |
@ARTICLE{Khokhlova2015-lr, | |
title = "Healthcare- and {Community-Associated} {Methicillin-Resistant} | |
Staphylococcus aureus ({MRSA}) and Fatal Pneumonia with Pediatric | |
Deaths in Krasnoyarsk, Siberian Russia: Unique {MRSA's} Multiple | |
Virulence Factors, Genome, and Stepwise Evolution", | |
author = "Khokhlova, Olga E and Hung, Wei-Chun and Wan, Tsai-Wen and Iwao, | |
Yasuhisa and Takano, Tomomi and Higuchi, Wataru and Yachenko, | |
Svetlana V and Teplyakova, Olga V and Kamshilova, Vera V and | |
Kotlovsky, Yuri V and Nishiyama, Akihito and Reva, Ivan V and | |
Sidorenko, Sergey V and Peryanova, Olga V and Reva, Galina V and | |
Teng, Lee-Jene and Salmina, Alla B and Yamamoto, Tatsuo", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) is a common | |
multidrug-resistant (MDR) pathogen. We herein discussed MRSA and | |
its infections in Krasnoyarsk, Siberian Russia between 2007 and | |
2011. The incidence of MRSA in 3,662 subjects was 22.0\% and | |
2.9\% for healthcare- and community-associated MRSA (HA- and | |
CA-MRSA), respectively. The 15-day mortality rates for MRSA | |
hospital- and community-acquired pneumonia (HAP and CAP) were | |
6.5\% and 50\%, respectively. MRSA CAP cases included pediatric | |
deaths; of the MRSA pneumonia episodes available, $\geq$27.3\% | |
were associated with bacteremia. Most cases of HA-MRSA examined | |
exhibited ST239/spa3(t037)/SCCmecIII.1.1.2 (designated as | |
ST239Kras), while all CA-MRSA cases examined were | |
ST8/spa1(t008)/SCCmecIV.3.1.1(IVc) (designated as ST8Kras). | |
ST239Kras and ST8Kras strongly expressed cytolytic peptide | |
(phenol-soluble modulin $\alpha$, PSM$\alpha$; and | |
$\delta$-hemolysin, Hld) genes, similar to CA-MRSA. ST239Kras | |
pneumonia may have been attributed to a unique set of multiple | |
virulence factors (MVFs): toxic shock syndrome toxin-1 (TSST-1), | |
elevated PSM$\alpha$/Hld expression, $\alpha$-hemolysin, the | |
staphylococcal enterotoxin SEK/SEQ, the immune evasion factor | |
SCIN/SAK, and collagen adhesin. Regarding ST8Kras, SEA was | |
included in MVFs, some of which were common to ST239Kras. The | |
ST239Kras (strain OC3) genome contained: a completely unique | |
phage, $\varphi$Sa7-like (W), with no att repetition; S. aureus | |
pathogenicity island SaPI2R, the first TSST-1 gene-positive | |
(tst+) SaPI in the ST239 lineage; and a super copy of IS256 | |
($\geq$22 copies/genome). ST239Kras carried the Brazilian | |
SCCmecIII.1.1.2 and United Kingdom-type tst. ST239Kras and | |
ST8Kras were MDR, with the same levofloxacin resistance | |
mutations; small, but transmissible chloramphenicol resistance | |
plasmids spread widely enough to not be ignored. These results | |
suggest that novel MDR and MVF+ HA- and CA-MRSA (ST239Kras and | |
ST8Kras) emerged in Siberian Russia (Krasnoyarsk) associated with | |
fatal pneumonia, and also with ST239Kras, a new (Siberian | |
Russian) clade of the ST239 lineage, which was created through | |
stepwise evolution during its potential transmission route of | |
Brazil-Europe-Russia/Krasnoyarsk, thereby selective advantages | |
from unique MVFs and the MDR.", | |
journal = "PLoS One", | |
volume = 10, | |
number = 6, | |
pages = "e0128017", | |
month = jun, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Aalbaek2015-be, | |
title = "{Whole-Genome} Sequence of Staphylococcus aureus {S54F9} Isolated | |
from a Chronic Disseminated Porcine Lung Abscess and Used in | |
Human Infection Models", | |
author = "Aalb{\ae}k, Bent and Jensen, Louise Kruse and Jensen, Henrik | |
Elvang and Olsen, John Elmerdahl and Christensen, Henrik", | |
abstract = "We obtained a draft genome sequence of Staphylococcus aureus | |
strain S54F9, which was isolated from a chronic disseminated | |
porcine lung abscess and used in porcine infection models. Genes | |
coding for a number of toxins, including enterotoxins and | |
superantigen, were demonstrated in this strain.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 5, | |
month = oct, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Chang2015-gk, | |
title = "Isolation and Genome Characterization of the Virulent | |
Staphylococcus aureus Bacteriophage {SA97}", | |
author = "Chang, Yoonjee and Shin, Hakdong and Lee, Ju-Hoon and Park, Chul | |
Jong and Paik, Soon-Young and Ryu, Sangryeol", | |
abstract = "A novel bacteriophage that infects S. aureus, SA97, was isolated | |
and characterized. The phage SA97 belongs to the Siphoviridae | |
family, and the cell wall teichoic acid (WTA) was found to be a | |
host receptor of the phage SA97. Genome analysis revealed that | |
SA97 contains 40,592 bp of DNA encoding 54 predicted open reading | |
frames (ORFs), and none of these genes were related to virulence | |
or drug resistance. Although a few genes associated with lysogen | |
formation were detected in the phage SA97 genome, the phage SA97 | |
produced neither lysogen nor transductant in S. aureus. These | |
results suggest that the phage SA97 may be a promising candidate | |
for controlling S. aureus.", | |
journal = "Viruses", | |
volume = 7, | |
number = 10, | |
pages = "5225--5242", | |
month = oct, | |
year = 2015, | |
keywords = "Staphylococcus aureus; bacteriophage; genome | |
analysis;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Richards2015-mm, | |
title = "Persistent Staphylococcus aureus isolates from two independent | |
cases of bacteremia display increased bacterial fitness and novel | |
immune evasion phenotypes", | |
author = "Richards, R L and Haigh, R D and Pascoe, B and Sheppard, S K and | |
Price, F and Jenkins, D and Rajakumar, K and Morrissey, J A", | |
abstract = "Staphylococcus aureus bacteremia cases are complicated by | |
bacterial persistence and treatment failure despite the confirmed | |
in vitro susceptibility of the infecting strain to administered | |
antibiotics. A high incidence of methicillin-resistant S. aureus | |
(MRSA) bacteremia cases are classified as persistent and are | |
associated with poorer patient outcomes. It is still unclear how | |
S. aureus evades the host immune system and resists antibiotic | |
treatment for the prolonged duration of a persistent infection. | |
In this study, the genetic changes and associated phenotypic | |
traits specific to S. aureus persistent bacteremia were | |
identified by comparing temporally dispersed isolates from | |
persistent infections (persistent isolates) originating from two | |
independent persistent S. aureus bacteremia cases with the | |
initial infection isolates and with three resolved S. aureus | |
bacteremia isolates from the same genetic background. Several | |
novel traits were associated specifically with both independent | |
sets of persistent S. aureus isolates compared to both the | |
initial isolates and the isolates from resolved infections | |
(resolved isolates). These traits included (i) increased growth | |
under nutrient-poor conditions; (ii) increased tolerance of iron | |
toxicity; (iii) higher expression of cell surface proteins | |
involved in immune evasion and stress responses; and (iv) | |
attenuated virulence in a Galleria mellonella larva infection | |
model that was not associated with small-colony variation or | |
metabolic dormancy such as had been seen previously. Whole-genome | |
sequence analysis identified different single nucleotide | |
mutations within the mprF genes of all the isolates with the | |
adaptive persistence traits from both independent cases. Overall, | |
our data indicate a novel role for MprF function during | |
development of S. aureus persistence by increasing bacterial | |
fitness and immune evasion.", | |
journal = "Infect. Immun.", | |
volume = 83, | |
number = 8, | |
pages = "3311--3324", | |
month = aug, | |
year = 2015, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;biometals;antibiotcs-evolution-fitness;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers" | |
} | |
@ARTICLE{Sabat2015-jb, | |
title = "Whole-genome analysis of an oxacillin-susceptible {CC80} | |
mecA-positive Staphylococcus aureus clinical isolate: insights | |
into the mechanisms of cryptic methicillin resistance", | |
author = "Sabat, Artur J and Pournaras, Spyros and Akkerboom, Viktoria and | |
Tsakris, Athanassios and Grundmann, Hajo and Friedrich, Alexander | |
W", | |
abstract = "OBJECTIVES: The mec and bla systems, among other genetic factors, | |
are critical in regulating the expression of methicillin | |
resistance in Staphylococcus aureus. We examined by WGS a | |
naturally occurring oxacillin-susceptible mecA-positive S. aureus | |
isolate to identify the mechanism conferring oxacillin | |
susceptibility. METHODS: The mecA-positive oxacillin-susceptible | |
S. aureus isolate GR2 (penicillin and oxacillin MICs 0.094 and 1 | |
mg/L, respectively), belonging to clonal complex 80, was | |
characterized. DNA fragment libraries were sequenced on Roche 454 | |
and Illumina MiSeq sequencers and de novo assembly of the genome | |
was generated using SeqMan NGen software. Plasmid curing was | |
conducted by SDS treatment. Expression of mecA was quantified | |
without/with $\beta$-lactam pressure. RESULTS: The genome of GR2 | |
consisted of a 2 792 802 bp chromosome and plasmids pGR2A (28 895 | |
bp) and pGR2B (2473 bp). GR2 carried SCCmec type IV, with a | |
truncated/non-functional mecR1 gene and no mecI. A single copy of | |
the bla system, with an organization unique for S. aureus, was | |
found, harboured by plasmid pGR2A. Particularly, the blaZ gene | |
was orientated like its regulatory genes, blaI and blaR1, and a | |
gene encoding transposase IS66 was integrated between blaZ and | |
the regulatory genes deleting the 5'-end of blaR1; blaI, encoding | |
blaZ/mecA repressor, was intact. After plasmid loss, GR2 became | |
penicillin and oxacillin resistant (MICs 0.5 and 6 mg/L, | |
respectively). CONCLUSIONS: We can conclude that after exposure | |
to $\beta$-lactams, the non-functional BlaR1 does not cleave the | |
mecA repressor BlaI, derepression does not occur and mecA is not | |
efficiently expressed. Removal of the bla system after curing of | |
pGR2A allows constitutive expression of mecA, resulting in | |
oxacillin and penicillin resistance.", | |
journal = "J. Antimicrob. Chemother.", | |
volume = 70, | |
number = 11, | |
pages = "2956--2964", | |
month = nov, | |
year = 2015, | |
keywords = "Staphylococcus;oxacillin;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Hamed2015-om, | |
title = "Whole genome sequence typing and microarray profiling of nasal | |
and blood stream methicillin-resistant Staphylococcus aureus | |
isolates: Clues to phylogeny and invasiveness", | |
author = "Hamed, Mohamed and Nitsche-Schmitz, Daniel Patric and Ruffing, | |
Ulla and Steglich, Matthias and Dordel, Janina and Nguyen, Duy | |
and Brink, Jan-Hendrik and Chhatwal, Gursharan Singh and | |
Herrmann, Mathias and N{\"u}bel, Ulrich and Helms, Volkhard and | |
von M{\"u}ller, Lutz", | |
abstract = "Hospital-associated methicillin-resistant Staphylococcus aureus | |
(MRSA) infections are frequently caused by predominant clusters | |
of closely related isolates that cannot be discriminated by | |
conventional diagnostic typing methods. Whole genome sequencing | |
(WGS) and DNA microarray (MA) now allow for better discrimination | |
within a prevalent clonal complex (CC). This single center | |
exploratory study aims to distinguish invasive (blood stream | |
infection) and non-invasive (nasal colonization) MRSA isolates of | |
the same CC5 into phylogenetic- and virulence-associated | |
genotypic subgroups by WGS and MA. A cohort of twelve blood | |
stream and fifteen nasal MRSA isolates of CC5 (spa-types t003 and | |
t504) was selected. Isolates were propagated at the same period | |
of time from unrelated patients treated at the University of | |
Saarland Medical Center, Germany. Rooted phylotyping based on WGS | |
with core-genome single nucleotide polymorphism (SNP) analysis | |
revealed two local clusters of closely related CC5 subgroups | |
(t504 and Clade1 t003) which were separated from other local t003 | |
isolates and from unrelated CC5 MRSA reference isolates of German | |
origin. Phylogenetic subtyping was not associated with | |
invasiveness when comparing blood stream and nasal isolates. | |
Clustering based on MA profiles was not concordant with WGS | |
phylotyping, but MA profiles may identify subgroups of isolates | |
with nasal and blood stream origin. Among the new putative | |
virulence associated genes identified by WGS, the strongest | |
association with blood stream infections was shown for ebhB | |
mutants. Analysis of the core-genome together with the accessory | |
genome enables subtyping of closely related MRSA isolates | |
according to phylogeny and presumably also to the potential | |
virulence capacity of isolates.", | |
journal = "Infect. Genet. Evol.", | |
volume = 36, | |
pages = "475--482", | |
month = dec, | |
year = 2015, | |
keywords = "DNA microarray; Invasiveness; MRSA; Phylogeny; Staphylococcus | |
aureus; Whole genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Steinig2015-si, | |
title = "Single-molecule sequencing reveals the molecular basis of | |
multidrug-resistance in {ST772} methicillin-resistant | |
Staphylococcus aureus", | |
author = "Steinig, Eike J and Andersson, Patiyan and Harris, Simon R and | |
Sarovich, Derek S and Manoharan, Anand and Coupland, Paul and | |
Holden, Matthew T G and Parkhill, Julian and Bentley, Stephen D | |
and Robinson, D Ashley and Tong, Steven Y C", | |
abstract = "BACKGROUND: Methicillin-resistant Staphylococcus aureus (MRSA) is | |
a major cause of hospital-associated infection, but there is | |
growing awareness of the emergence of multidrug-resistant | |
lineages in community settings around the world. One such lineage | |
is ST772-MRSA-V, which has disseminated globally and is | |
increasingly prevalent in India. Here, we present the complete | |
genome sequence of DAR4145, a strain of the ST772-MRSA-V lineage | |
from India, and investigate its genomic characteristics in | |
regards to antibiotic resistance and virulence factors. RESULTS: | |
Sequencing using single-molecule real-time technology resulted in | |
the assembly of a single continuous chromosomal sequence, which | |
was error-corrected, annotated and compared to nine draft genome | |
assemblies of ST772-MRSA-V from Australia, Malaysia and India. We | |
discovered numerous and redundant resistance genes associated | |
with mobile genetic elements (MGEs) and known core genome | |
mutations that explain the highly antibiotic resistant phenotype | |
of DAR4145. Staphylococcal toxins and superantigens, including | |
the leukotoxin Panton-Valentinin Leukocidin, were predominantly | |
associated with genomic islands and the phage | |
$\varphi$-IND772PVL. Some of these mobile resistance and | |
virulence factors were variably present in other strains of the | |
ST772-MRSA-V lineage. CONCLUSIONS: The genomic characteristics | |
presented here emphasize the contribution of MGEs to the | |
emergence of multidrug-resistant and highly virulent strains of | |
community-associated MRSA. Antibiotic resistance was further | |
augmented by chromosomal mutations and redundancy of resistance | |
genes. The complete genome of DAR4145 provides a valuable | |
resource for future investigations into the global dissemination | |
and phylogeography of ST772-MRSA-V.", | |
journal = "BMC Genomics", | |
volume = 16, | |
pages = "388", | |
month = may, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Stipetic2015-tc, | |
title = "Draft Genome Sequence of Isolate Staphylococcus aureus | |
{LHSKBClinical}, Isolated from an Infected Hip", | |
author = "Stipetic, Laurence H and Hamilton, Graham and Dalby, Matthew J | |
and Davies, Robert L and Meek, R M Dominic and Ramage, Gordon and | |
Smith, David G E and Burgess, Karl E V", | |
abstract = "We report here the genome sequence of a clinical isolate of | |
Staphylococcus aureus from an orthopedic infection. | |
Phenotypically diverse Staphylococcus aureus strains are | |
associated with orthopedic infections and subsequent implant | |
failure, and some are highly resistant to antibiotics. This | |
genome sequence will support further analyses of strains causing | |
orthopedic infections.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 2, | |
month = apr, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Cazares-Dominguez2015-re, | |
title = "Vancomycin tolerant, methicillin-resistant Staphylococcus aureus | |
reveals the effects of vancomycin on cell wall thickening", | |
author = "C{\'a}zares-Dom{\'\i}nguez, Vicenta and Cruz-C{\'o}rdova, | |
Ariadnna and Ochoa, Sara A and Escalona, Gerardo and | |
Arellano-Galindo, Jos{\'e} and Rodr{\'\i}guez-Leviz, Alejandra | |
and Hern{\'a}ndez-Castro, Rigoberto and L{\'o}pez-Villegas, Edgar | |
O and Xicohtencatl-Cortes, Juan", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) is an | |
important opportunistic pathogen that causes both healthcare- and | |
community-acquired infections. An increase in the incidence of | |
these infections may lead to a substantial change in the rate of | |
vancomycin usage. Incidence of reduced susceptibility to | |
vancomycin has been increasing worldwide for the last few years, | |
conferring different levels of resistance to vancomycin as well | |
as producing changes in the cell wall structure. The aim of the | |
present study was to determine the effect of vancomycin on cell | |
wall thickening in clinical isolates of vancomycin-tolerant (VT) | |
MRSA obtained from pediatric patients. From a collection of 100 | |
MRSA clinical isolates from pediatric patients, 12\% (12/100) | |
were characterized as VT-MRSA, and from them, 41.66\% (5/12) | |
exhibited the heterogeneous vancomycin-intermediate S. aureus | |
(hVISA) phenotype. Multiplex-PCR assays revealed 66.66\% (8/12), | |
25\% (3/12), and 8.33\% (1/12) of the VT-MRSA isolates were | |
associated with agr group II, I, and III polymorphisms, | |
respectively; the II-mec gene was amplified from 83.3\% (10/12) | |
of the isolates, and the mecIVa gene was amplified from 16.66\% | |
(2/12) of the isolates. Pulsed field electrophoresis (PFGE) | |
fingerprint analysis showed 62\% similarity among the VT-MRSA | |
isolates. Thin transverse sections analyzed by transmission | |
electron microscopy (TEM) revealed an average increase of 24 nm | |
(105.55\%) in the cell wall thickness of VT-MRSA compared with | |
untreated VT-MRSA isolates. In summary, these data revealed that | |
the thickened cell walls of VT-MRSA clinical isolates with agr | |
type II and SCCmec group II polymorphisms are associated with an | |
adaptive resistance to vancomycin.", | |
journal = "PLoS One", | |
volume = 10, | |
number = 3, | |
pages = "e0118791", | |
month = mar, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Yamaguchi2015-vn, | |
title = "Evolution and single-nucleotide polymorphisms in | |
methicillin-resistant Staphylococcus aureus strains with reduced | |
susceptibility to vancomycin and daptomycin, based on | |
determination of the complete genome", | |
author = "Yamaguchi, Tetsuo and Suzuki, Shingo and Okamura, Sakiko and | |
Miura, Yuri and Tsukimori, Ayaka and Nakamura, Itaru and Ito, | |
Norihiko and Masuya, Anri and Shiina, Takashi and Matsumoto, | |
Tetsuya", | |
abstract = "We obtained a series of methicillin-resistant Staphylococcus | |
aureus isolates, including both daptomycin-susceptible strain TD1 | |
and daptomycin-resistant strain TD4, from a patient. We | |
determined the complete genome sequences of TD1 and TD4 using | |
next-generation sequencing, and only four single-nucleotide | |
polymorphisms (SNPs) were identified, one each in capB (E58K), | |
rpoB (H481Y), lytN (I16V), and mprF (V351E). We determined that | |
these four SNPs were sufficient to cause the strains to develop | |
daptomycin, vancomycin, and rifampin resistance.", | |
journal = "Antimicrob. Agents Chemother.", | |
volume = 59, | |
number = 6, | |
pages = "3585--3587", | |
month = mar, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Daum2015-li, | |
title = "{Whole-Genome} Sequence for {Methicillin-Resistant} | |
Staphylococcus aureus Strain {ATCC} {BAA-1680}", | |
author = "Daum, Luke T and Bumah, Violet V and Masson-Meyers, Daniela S and | |
Khubbar, Manjeet and Rodriguez, John D and Fischer, Gerald W and | |
Enwemeka, Chukuka S and Gradus, Steve and Bhattacharyya, Sanjib", | |
abstract = "We report here the whole-genome sequence of the USA300 strain of | |
methicillin-resistant Staphylococcus aureus (MRSA), designated | |
ATCC BAA-1680, and commonly referred to as community-associated | |
MRSA (CA-MRSA). This clinical MRSA isolate is commercially | |
available from the American Type Culture Collection (ATCC) and is | |
widely utilized as a control strain for research applications and | |
clinical diagnosis. The isolate was propagated in ATCC medium 18, | |
tryptic soy agar, and has been utilized as a model S. aureus | |
strain in several studies, including MRSA genetic analysis after | |
irradiation with 470-nm blue light.", | |
journal = "Genome Announc.", | |
volume = 3, | |
number = 2, | |
month = mar, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Anderson2014-qt, | |
title = "Whole Genome Sequencing of a {Methicillin-Resistant} | |
Staphylococcus aureus {Pseudo-Outbreak} in a Professional | |
Football Team", | |
author = "Anderson, Deverick J and Harris, Simon R and Godofsky, Eliot and | |
Toriscelli, Todd and Rude, Thomas H and Elder, Kevin and Sexton, | |
Daniel J and Pellman, Elliot J and Mayer, Thom and Fowler, Jr, | |
Vance G and Peacock, Sharon J", | |
abstract = "Two American football players on the same team were diagnosed | |
with methicillin-resistant Staphylococcus aureus (MRSA) skin and | |
soft tissue infections on the same day. Our investigation, | |
including whole genome sequencing, confirmed that players did not | |
transmit MRSA to one another nor did they acquire the MRSA from a | |
single source within the training facility.", | |
journal = "Open Forum Infect Dis", | |
volume = 1, | |
number = 3, | |
pages = "ofu096", | |
month = dec, | |
year = 2014, | |
keywords = "American football; MRSA; outbreak investigation; whole genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{Hattangady2015-we, | |
title = "Genomic, Transcriptomic and Metabolomic Studies of Two | |
{Well-Characterized}, {Laboratory-Derived} | |
{Vancomycin-Intermediate} Staphylococcus aureus Strains Derived | |
from the Same Parent Strain", | |
author = "Hattangady, Dipti S and Singh, Atul K and Muthaiyan, Arun and | |
Jayaswal, Radheshyam K and Gustafson, John E and Ulanov, | |
Alexander V and Li, Zhong and Wilkinson, Brian J and Pfeltz, | |
Richard F", | |
abstract = "Complete genome comparisons, transcriptomic and metabolomic | |
studies were performed on two laboratory-selected, | |
well-characterized vancomycin-intermediate Staphylococcus aureus | |
(VISA) derived from the same parent MRSA that have changes in | |
cell wall composition and decreased autolysis. A variety of | |
mutations were found in the VISA, with more in strain | |
13136p(-)m⁺V20 (vancomycin MIC = 16 µg/mL) than strain | |
13136p(-)m⁺V5 (MIC = 8 µg/mL). Most of the mutations have not | |
previously been associated with the VISA phenotype; some were | |
associated with cell wall metabolism and many with stress | |
responses, notably relating to DNA damage. The genomes and | |
transcriptomes of the two VISA support the importance of gene | |
expression regulation to the VISA phenotype. Similarities in | |
overall transcriptomic and metabolomic data indicated that the | |
VISA physiologic state includes elements of the stringent | |
response, such as downregulation of protein and nucleotide | |
synthesis, the pentose phosphate pathway and nutrient transport | |
systems. Gene expression for secreted virulence determinants was | |
generally downregulated, but was more variable for | |
surface-associated virulence determinants, although capsule | |
formation was clearly inhibited. The importance of activated | |
stress response elements could be seen across all three analyses, | |
as in the accumulation of osmoprotectant metabolites such as | |
proline and glutamate. Concentrations of potential cell wall | |
precursor amino acids and glucosamine were increased in the VISA | |
strains. Polyamines were decreased in the VISA, which may | |
facilitate the accrual of mutations. Overall, the studies confirm | |
the wide variability in mutations and gene expression patterns | |
that can lead to the VISA phenotype.", | |
journal = "Antibiotics (Basel)", | |
volume = 4, | |
number = 1, | |
pages = "76--112", | |
month = feb, | |
year = 2015, | |
keywords = "Staphylococcus aureus; VISA; genomics; metabolomics; | |
transcriptomics;Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Fluit2015-cw, | |
title = "Comparison of an {ST80} {MRSA} strain from the {USA} with | |
European {ST80} strains", | |
author = "Fluit, Ad C and Carpaij, Neeltje and Majoor, Eline A M and | |
Weinstein, Robert A and Aroutcheva, Alla and Rice, Thomas W and | |
Bonten, Marc J M and Willems, Rob J L", | |
abstract = "OBJECTIVES: A number of community-acquired MRSA (CA-MRSA) clonal | |
lineages dominate worldwide. ST80 was dominant in Europe and has | |
increasingly been described from the Middle East. Here we report | |
the whole genome sequence of the first ST80 CA-MRSA from the USA. | |
METHODS: CA-MRSA isolate S0924 was obtained from a patient | |
admitted to Cook County Hospital (Chicago, IL, USA) who came from | |
Syria; the isolate belonged to spa type t044 and ST80. The whole | |
genome sequence of S0924 was determined and compared with three | |
previously published whole genome sequences of ST80 CA-MRSA from | |
Europe and a newly sequenced ST80 CA-MRSA from the Netherlands | |
(S1475). RESULTS: Based on spa typing, SCCmec type and virulence | |
gene profile, this US ST80 isolate is indistinguishable from the | |
European CA-MRSA ST80 clone. SNP analysis within the conserved | |
core genome showed clear differences between the strains with up | |
to 144 SNPs differing between S0924 and strain S1800, an ST80 | |
MRSA from Greece. The gene content showed 21 regions of | |
difference between the US and European isolates, although these | |
were largely restricted to mobile genetic elements. Phylogenetic | |
reconstruction indicated that the European strains were more | |
closely related to each other than to the US strain. The SNP data | |
suggest that a common ancestor existed around two decades ago, | |
indicating that the US and European ST80 strains are clonally | |
linked. CONCLUSIONS: These data combined with the country of | |
origin of the patient suggest that ST80 S0924 was probably | |
relatively recently introduced into the USA from Syria.", | |
journal = "J. Antimicrob. Chemother.", | |
volume = 70, | |
number = 3, | |
pages = "664--669", | |
month = mar, | |
year = 2015, | |
keywords = "CA-MRSA; US; whole genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Guo2015-yz, | |
title = "Use of genome sequencing to assess nucleotide structure variation | |
of Staphylococcus aureus strains cultured in spaceflight on | |
{Shenzhou-X}, under simulated microgravity and on the ground", | |
author = "Guo, Jun and Han, Na and Zhang, Yuanyuan and Wang, Haiyin and | |
Zhang, Xuelin and Su, Longxiang and Liu, Chao and Li, Jia and | |
Chen, Chen and Liu, Changting", | |
abstract = "The extreme environment of space could affect microbial behavior | |
and may increase the risk of infectious disease during | |
spaceflight. However, the molecular genetic changes of | |
methicillin-resistant Staphylococcus aureus (MRSA) in response to | |
the spaceflight environment have not been fully clarified. In the | |
present study, we determined the draft genome sequences for an | |
ancestral S. aureus strain (LCT-SAO) isolated from a clinical | |
sample and three derivative strains, LCT-SAS, LCT-SAM and | |
LCT-SAG, cultured in parallel during the spaceflight Shenzhou-X, | |
under simulated microgravity and on the ground, respectively. To | |
evaluate the impact of short-term spaceflight on the MRSA | |
strains, comparative genomic analysis was implemented. | |
Genome-based mapping of toxin genes and antibiotic resistance | |
genes confirmed that these strains have the conventional | |
pathogenicity and resistance to drugs, as none of the strains | |
showed significant changes in these regions after culturing in | |
the three different environments; this result suggests that | |
spaceflight may not change bacterial virulence or drug | |
resistance. Thirty-nine strain-specific sequence variants (SVs) | |
were identified throughout the genomes, and the three derivatives | |
exhibited almost the same mutation rates. Fifty-nine percent of | |
SVs were located in the intergenic regions of the genomes, | |
indicating that S. aureus may have an extremely robust repair | |
mechanism responsible for recognizing and repairing DNA | |
replication mismatches. It is noteworthy that strain LCT-SAS, | |
cultured in space, presented the most unique SVs (n=9) and shared | |
the fewest SVs with LCT-SAM (n=5) and LCT-SAG (n=4). Furthermore, | |
we identified 10 potential deletion regions and 2 potential | |
insertion regions, with LCT-SAS appearing more fragile than other | |
strains by this measure. These results suggest that the | |
environment of space is inherently complicated, with multiple | |
variables, and cannot be simulated in a simple manner. Our | |
results represent the first analysis of nucleotide structure | |
variation of S. aureus strains in a spaceflight environment and | |
also provide a valuable insight for understanding the mutation | |
strategies of MRSA on earth.", | |
journal = "Microbiol. Res.", | |
volume = 170, | |
pages = "61--68", | |
month = jan, | |
year = 2015, | |
keywords = "Genome; High-throughput; SNP; Spaceflight; Staphylococcus | |
aureus;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Ramaraj2014-yk, | |
title = "Draft Genome Sequences of {Vancomycin-Susceptible} Staphylococcus | |
aureus Related to Heterogeneous {Vancomycin-Intermediate} S. | |
aureus", | |
author = "Ramaraj, Thiruvarangan and Matyi, Stephanie A and Sundararajan, | |
Anitha and Lindquist, Ingrid E and Devitt, Nicolas P and | |
Schilkey, Faye D and Lamichhane-Khadka, Reena and Hoyt, Peter R | |
and Mudge, Joann and Gustafson, John E", | |
abstract = "We report the draft genome sequences of three | |
vancomycin-susceptible methicillin-resistant Staphylococcus | |
aureus strains. S. aureus strain MV8 is a sequence type 8 (ST-8) | |
staphylococcal cassette chromosome mec element type IV (SCCmec | |
IV) derivative, while the other two strains (S. aureus MM25 and | |
MM61) are ST-5 SCCmec II strains. MM61 is also closely related to | |
the heterogeneous vancomycin-intermediate S. aureus strain MM66.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 5, | |
month = oct, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Kropp2014-ci, | |
title = "Draft Genome Sequence of a Staphylococcus aureus Isolate Taken | |
from the Blood of a Preterm Neonatal Blood Sepsis Patient", | |
author = "Kropp, K A and Lucid, A and Carroll, J and Belgrudov, V and | |
Walsh, P and Kelly, B and Templeton, K and Smith, C and | |
Dickinson, P and O'Driscoll, A and Ghazal, P and Sleator, R D", | |
abstract = "Herein, we report the draft genome sequence of Staphylococcus | |
aureus ED-NGS-1006, cultivated from a blood sample taken from a | |
neonatal sepsis patient at the Royal Infirmary in Edinburgh, | |
Scotland, United Kingdom.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 5, | |
month = sep, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Stegger2014-wr, | |
title = "Origin and evolution of European community-acquired | |
methicillin-resistant Staphylococcus aureus", | |
author = "Stegger, Marc and Wirth, Thierry and Andersen, Paal S and Skov, | |
Robert L and De Grassi, Anna and Sim{\~o}es, Patricia Martins and | |
Tristan, Anne and Petersen, Andreas and Aziz, Maliha and Kiil, | |
Kristoffer and Cirkovi{\'c}, Ivana and Udo, Edet E and del Campo, | |
Rosa and Vuopio-Varkila, Jaana and Ahmad, Norazah and Tokajian, | |
Sima and Peters, Georg and Schaumburg, Frieder and | |
Olsson-Liljequist, Barbro and Givskov, Michael and Driebe, | |
Elizabeth E and Vigh, Henrik E and Shittu, Adebayo and | |
Ramdani-Bougessa, Nadjia and Rasigade, Jean-Philippe and Price, | |
Lance B and Vandenesch, Francois and Larsen, Anders R and | |
Laurent, Frederic", | |
abstract = "UNLABELLED: Community-acquired methicillin-resistant | |
Staphylococcus aureus (CA-MRSA) was recognized in Europe and | |
worldwide in the late 1990s. Within a decade, several genetically | |
and geographically distinct CA-MRSA lineages carrying the small | |
SCCmec type IV and V genetic elements and the Panton-Valentine | |
leukocidin (PVL) emerged around the world. In Europe, the | |
predominant CA-MRSA strain belongs to clonal complex 80 (CC80) | |
and is resistant to kanamycin/amikacin and fusidic acid. CC80 was | |
first reported in 1993 but was relatively rare until the late | |
1990s. It has since been identified throughout North Africa, the | |
Middle East, and Europe, with recent sporadic reports in | |
sub-Saharan Africa. While strongly associated with skin and soft | |
tissue infections, it is rarely found among asymptomatic | |
carriers. Methicillin-sensitive S. aureus (MSSA) CC80 strains are | |
extremely rare except in sub-Saharan Africa. In the current | |
study, we applied whole-genome sequencing to a global collection | |
of both MSSA and MRSA CC80 isolates. Phylogenetic analyses | |
strongly suggest that the European epidemic CA-MRSA lineage is | |
derived from a PVL-positive MSSA ancestor from sub-Saharan | |
Africa. Moreover, the tree topology suggests a single acquisition | |
of both the SCCmec element and a plasmid encoding the fusidic | |
acid resistance determinant. Four canonical SNPs distinguish the | |
derived CA-MRSA lineage and include a nonsynonymous mutation in | |
accessory gene regulator C (agrC). These changes were associated | |
with a star-like expansion into Europe, the Middle East, and | |
North Africa in the early 1990s, including multiple cases of | |
cross-continent imports likely driven by human migrations. | |
IMPORTANCE: With increasing levels of CA-MRSA reported from most | |
parts of the Western world, there is a great interest in | |
understanding the origin and factors associated with the | |
emergence of these epidemic lineages. To trace the origin, | |
evolution, and dissemination pattern of the European CA-MRSA | |
clone (CC80), we sequenced a global collection of strains of the | |
S. aureus CC80 lineage. Our study determined that a single | |
descendant of a PVL-positive methicillin-sensitive ancestor | |
circulating in sub-Saharan Africa rose to become the dominant | |
CA-MRSA clone in Europe, the Middle East, and North Africa. In | |
the transition from a methicillin-susceptible lineage to a | |
successful CA-MRSA clone, it simultaneously became resistant to | |
fusidic acid, a widely used antibiotic for skin and soft tissue | |
infections, thus demonstrating the importance of antibiotic | |
selection in the success of this clone. This finding furthermore | |
highlights the significance of horizontal gene acquisitions and | |
underscores the combined importance of these factors for the | |
success of CA-MRSA.", | |
journal = "MBio", | |
volume = 5, | |
number = 5, | |
pages = "e01044--14", | |
month = aug, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Matyi2014-ug, | |
title = "Draft Genomes of Heterogeneous {Vancomycin-Intermediate} | |
Staphylococcus aureus Strain {MM66} and {MM66} Derivatives with | |
Altered Vancomycin Resistance Levels", | |
author = "Matyi, Stephanie A and Ramaraj, Thiruvarangan and Sundararajan, | |
Anitha and Lindquist, Ingrid E and Devitt, Nicolas P and | |
Schilkey, Faye D and Lamichhane-Khadka, Reena and Hoyt, Peter R | |
and Mudge, Joann and Gustafson, John E", | |
abstract = "The draft genomes of heterogeneous vancomycin-intermediate | |
Staphylococcus aureus (hVISA) strain MM66 and MM66 isolates | |
demonstrating altered vancomycin resistance levels were produced | |
in an effort to provide information on mutations contributing to | |
the vancomycin resistance levels observed in these strains.", | |
journal = "Genome Announc.", | |
volume = 2, | |
number = 4, | |
month = jul, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Jeon2014-nu, | |
title = "Complete Genome Sequence of the Siphoviral Bacteriophage | |
{YMC/09/04/R1988} {MRSA} {BP}: A lytic phage from a | |
methicillin-resistant Staphylococcus aureus isolate", | |
author = "Jeon, Jongsoo and D'Souza, Roshan and Hong, Sung Kuk and Lee, | |
Yangsoon and Yong, Dongeun and Choi, Jongrak and Lee, Kyungwon | |
and Chong, Yunsop", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) is an | |
increasing cause of serious infection, both in the community and | |
hospital settings. Despite sophisticated strategies and efforts, | |
the antibiotic options for treating MRSA infection have been | |
narrowed due to the limited number of newly developed | |
antimicrobials. Herein, we analyze the completely sequenced | |
genome of a novel virulent phage YMC/09/04/R1988 MRSA BP as a | |
potential alternative anti-MRSA agent, which lysed clinical | |
isolates from a patient admitted to the hospital due to hip | |
disarticulation. The phage contains a linear double-stranded DNA | |
genome of 44,459 bp in length, with 33.37\% GC content, 62 | |
predicted open reading frames (ORFs), and annotated functions of | |
only 23 ORFs that are associated with structural assembly, host | |
lysis, DNA replication, and modification. It showed a broad host | |
range (17 of 30 strains) against MRSA strains in clinical | |
isolates. This article is protected by copyright. All rights | |
reserved.", | |
journal = "FEMS Microbiol. Lett.", | |
month = aug, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{Keary2014-ke, | |
title = "Genome analysis of the staphylococcal temperate phage {DW2} and | |
functional studies on the endolysin and tail hydrolase", | |
author = "Keary, Ruth and McAuliffe, Olivia and Ross, R Paul and Hill, | |
Colin and O'Mahony, Jim and Coffey, Aidan", | |
abstract = "This study describes the genome of temperate Siphoviridae phage | |
DW2, which is routinely propagated on Staphylococcus aureus | |
DPC5246. The 41941 bp genome revealed an open reading frame | |
(ORF1) which has a high level of homology with members of the | |
resolvase subfamily of site-specific serine recombinase, involved | |
in chromosomal integration and excision. In contrast, the | |
majority of staphylococcal phages reported to date encode | |
tyrosine recombinases. Two putative genes encoded by phage DW2 | |
(ORF15 and ORF24) were highly homologous to the NWMN0273 and | |
NWMN0280 genes encoding virulence factors carried on the genome | |
of ϕNM4, a prophage in the genome of S. aureus Newman. Phage DW2 | |
also encodes proteins highly homologous to two well-characterized | |
Staphylococcus aureus pathogenicity island derepressors encoded | |
by the staphylococcal helper phage 80$\alpha$ indicating that it | |
may similarly act as a helper phage for mobility of pathogenicity | |
islands in S. aureus. This study also focused on the enzybiotic | |
potential of phage DW2. The structure of the putative endolysin | |
and tail hydrolase were investigated and used as the basis for a | |
cloning strategy to create recombinant peptidoglycan hydrolyzing | |
proteins. After overexpression in E. coli, four of these proteins | |
(LysDW2, THDW2, CHAPE1-153, and CHAPE1-163) were demonstrated to | |
have hydrolytic activity against peptidoglycan of S. aureus and | |
thus represent novel candidates for exploitation as enzybiotics.", | |
journal = "Bacteriophage", | |
volume = 4, | |
pages = "e28451", | |
month = mar, | |
year = 2014, | |
keywords = "Staphylococcus; bacteriophage; endolysin; serine recombinase; | |
virion-associated peptidoglycan hydrolase; | |
virulence;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{SenGupta2014-is, | |
title = "Whole-genome sequencing for high-resolution investigation of | |
methicillin-resistant Staphylococcus aureus epidemiology and | |
genome plasticity", | |
author = "SenGupta, Dhruba J and Cummings, Lisa A and Hoogestraat, Daniel R | |
and Butler-Wu, Susan M and Shendure, Jay and Cookson, Brad T and | |
Salipante, Stephen J", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) infections | |
pose a major challenge in health care, yet the limited | |
heterogeneity within this group hinders molecular investigations | |
of related outbreaks. Pulsed-field gel electrophoresis (PFGE) has | |
been the gold standard approach but is impractical for many | |
clinical laboratories and is often replaced with PCR-based | |
methods. Regardless, both approaches can prove problematic for | |
identifying subclonal outbreaks. Here, we explore the use of | |
whole-genome sequencing for clinical laboratory investigations of | |
MRSA molecular epidemiology. We examine the relationships of 44 | |
MRSA isolates collected over a period of 3 years by using | |
whole-genome sequencing and two PCR-based methods, multilocus | |
variable-number tandem-repeat analysis (MLVA) and spa typing. We | |
find that MLVA offers higher resolution than spa typing, as it | |
resolved 17 versus 12 discrete isolate groups, respectively. In | |
contrast, whole-genome sequencing reproducibly cataloged genomic | |
variants (131,424 different single nucleotide polymorphisms and | |
indels across the strain collection) that uniquely identified | |
each MRSA clone, recapitulating those groups but enabling | |
higher-resolution phylogenetic inferences of the epidemiological | |
relationships. Importantly, whole-genome sequencing detected a | |
significant number of variants, thereby distinguishing between | |
groups that were considered identical by both spa typing | |
(minimum, 1,124 polymorphisms) and MLVA (minimum, 193 | |
polymorphisms); this suggests that these more conventional | |
approaches can lead to false-positive identification of outbreaks | |
due to inappropriate grouping of genetically distinct strains. An | |
analysis of the distribution of variants across the MRSA genome | |
reveals 47 mutational hot spots (comprising ∼ 2.5\% of the | |
genome) that account for 23.5\% of the observed polymorphisms, | |
and the use of this selected data set successfully recapitulates | |
most epidemiological relationships in this pathogen group.", | |
journal = "J. Clin. Microbiol.", | |
volume = 52, | |
number = 8, | |
pages = "2787--2796", | |
month = aug, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Dordel2014-pp, | |
title = "Novel determinants of antibiotic resistance: identification of | |
mutated loci in highly methicillin-resistant subpopulations of | |
methicillin-resistant Staphylococcus aureus", | |
author = "Dordel, Janina and Kim, Choonkeun and Chung, Marilyn and Pardos | |
de la G{\'a}ndara, Mar{\'\i}a and Holden, Matthew T J and | |
Parkhill, Julian and de Lencastre, Herm{\'\i}nia and Bentley, | |
Stephen D and Tomasz, Alexander", | |
abstract = "We identified mutated genes in highly resistant subpopulations of | |
methicillin-resistant Staphylococcus aureus (MRSA) that are most | |
likely responsible for the historic failure of the $\beta$-lactam | |
family of antibiotics as therapeutic agents against these | |
important pathogens. Such subpopulations are produced during | |
growth of most clinical MRSA strains, including the four | |
historically early MRSA isolates studied here. Chromosomal DNA | |
was prepared from the highly resistant cells along with DNA from | |
the majority of cells (poorly resistant cells) followed by full | |
genome sequencing. In the highly resistant cells, mutations were | |
identified in 3 intergenic sequences and 27 genes representing a | |
wide range of functional categories. A common feature of these | |
mutations appears to be their capacity to induce high-level | |
$\beta$-lactam resistance and increased amounts of the resistance | |
protein PBP2A in the bacteria. The observations fit a recently | |
described model in which the ultimate controlling factor of the | |
phenotypic expression of $\beta$-lactam resistance in MRSA is a | |
RelA-mediated stringent response. IMPORTANCE It has been well | |
established that the level of antibiotic resistance (i.e., | |
minimum concentration of a $\beta$-lactam antibiotic needed to | |
inhibit growth) of a methicillin-resistant Staphylococcus aureus | |
(MRSA) strain depends on the transcription and translation of the | |
resistance protein PBP2A. Here we describe mutated loci in an | |
additional novel set of genetic determinants that appear to be | |
essential for the unusually high resistance levels typical of | |
subpopulations of staphylococci that are produced with unique low | |
frequency in most MRSA clinical isolates. We propose that | |
mutations in these determinants can trigger induction of the | |
stringent stress response which was recently shown to cause | |
increased transcription/translation of the resistance protein | |
PBP2A in parallel with the increased level of resistance.", | |
journal = "MBio", | |
volume = 5, | |
number = 2, | |
pages = "e01000", | |
month = apr, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Baines2015-ms, | |
title = "Convergent Adaptation in the Dominant Global Hospital Clone | |
{ST239} of {Methicillin-Resistant} Staphylococcus aureus", | |
author = "Baines, Sarah L and Holt, Kathryn E and Schultz, Mark B and | |
Seemann, Torsten and Howden, Brian O and Jensen, Slade O and van | |
Hal, Sebastiaan J and Coombs, Geoffrey W and Firth, Neville and | |
Powell, David R and Stinear, Timothy P and Howden, Benjamin P", | |
abstract = "UNLABELLED: Infections caused by highly successful clones of | |
hospital-associated methicillin-resistant Staphylococcus aureus | |
(HA-MRSA) are a major public health burden. The globally dominant | |
sequence type 239 (ST239) HA-MRSA clone has persisted in the | |
health care setting for decades, but the basis of its success has | |
not been identified. Taking a collection of 123 ST239 isolates | |
spanning 32 years, we have used population-based functional | |
genomics to investigate the evolution of this highly persistent | |
and successful clone. Phylogenetic reconstruction and population | |
modeling uncovered a previously unrecognized distinct clade of | |
ST239 that was introduced into Australia from Asia and has | |
perpetuated the epidemic in this region. Functional analysis | |
demonstrated attenuated virulence and enhanced resistance to | |
last-line antimicrobials, the result of two different phenomena, | |
adaptive evolution within the original Australian ST239 clade and | |
the introduction of a new clade displaying shifts in both | |
phenotypes. The genetic diversity between the clades allowed us | |
to employ genome-wide association testing and identify mutations | |
in other essential regulatory systems, including walKR, that | |
significantly associate with and may explain these key | |
phenotypes. The phenotypic convergence of two independently | |
evolving ST239 clades highlights the very strong selective | |
pressures acting on HA-MRSA, showing that hospital environments | |
have favored the accumulation of mutations in essential MRSA | |
genes that increase resistance to antimicrobials, attenuate | |
virulence, and promote persistence in the health care | |
environment. Combinations of comparative genomics and careful | |
phenotypic measurements of longitudinal collections of clinical | |
isolates are giving us the knowledge to intelligently address the | |
impact of current and future antibiotic usage policies and | |
practices on hospital pathogens globally. IMPORTANCE: | |
Methicillin-resistant Staphylococcus aureus (MRSA) is responsible | |
for innumerable drug-resistant health care-associated infections | |
globally. This study, the first to investigate the evolutionary | |
response of hospital-associated MRSA (HA-MRSA) over many decades, | |
demonstrates how MRSA can persist in a region through the | |
reintroduction of a previously unrecognized distinct clade. This | |
study also demonstrates the crucial adaptive responses of HA-MRSA | |
to the highly selective environment of the health care system, | |
the evolution of MRSA isolates to even higher levels of | |
antibiotic resistance at the cost of attenuated virulence. | |
However, in vivo persistence is maintained, resulting in a clone | |
of HA-MRSA able to resist almost all antimicrobial agents and | |
still cause invasive disease in the heavily compromised hosts | |
found in modern health care settings.", | |
journal = "MBio", | |
volume = 6, | |
number = 2, | |
pages = "e00080", | |
month = may, | |
year = 2015, | |
keywords = "Staphylococcus;Staph\_evolutionary\_history;bacterial comparative | |
genomics;antibiotcs-evolution-fitness;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;GWAS-bacteria" | |
} | |
@ARTICLE{Qu2014-cf, | |
title = "Whole genome analysis of a community-associated | |
methicillin-resistant Staphylococcus aureus {ST59} isolate from a | |
case of human sepsis and severe pneumonia in China", | |
author = "Qu, Tingting and Feng, Ye and Jiang, Yan and Zhu, Peiqiong and | |
Wei, Zeqing and Chen, Yan and Otto, Michael and Yu, Yunsong", | |
abstract = "We report a case of necrotizing pneumonia in a young patient | |
caused by community acquired-methicillin resistant Staphylococcus | |
aureus (CA-MRSA) in a teaching hospital in the People's Republic | |
of China. The patient had a typical clinical presentation and was | |
successfully treated with antibiotics and intravenous | |
immunoglobulin. A CA-MRSA strain, named SA268, was isolated from | |
the blood of the patient. The isolate was susceptible to most | |
antimicrobial agents, except cephalosporins, penicillins, and | |
$\beta$-lactamase inhibitor combinations. Multi-locus sequence | |
typing (MLST) assigned SA268 to ST59, a clone widely spread in | |
eastern Asia. The strain was positive for Panton Valentine | |
Leukocidin (PVL)-encoding genes and SCCmec type V. We sequenced | |
the complete genome of the SA268 isolate. The genome of SA268 was | |
almost identical to that of the Taiwanese ST59 CA-MRSA strains | |
M013 and SA957. However, we observed several differences in gene | |
composition, which included differences in the SCCmec element and | |
several lipoprotein genes that were present in the Taiwanese | |
strains but absent from SA268.", | |
journal = "PLoS One", | |
volume = 9, | |
number = 2, | |
pages = "e89235", | |
month = feb, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Elbir2013-tl, | |
title = "Staphylococcus aureus subsp. anaerobius strain {ST1464} genome | |
sequence", | |
author = "Elbir, Haitham and Robert, Catherine and Nguyen, Ti Thien and | |
Gimenez, Gr{\'e}gory and El Sanousi, Sulieman M and Flock, | |
Jan-Ingmar and Raoult, Didier and Drancourt, Michel", | |
abstract = "Staphylococcus aureus subsp. anaerobius is responsible for | |
Morel's disease in animals and a cause of abscess in humans. It | |
is characterized by a microaerophilic growth, contrary to the | |
other strains of S. aureus. The 2,604,446-bp genome (32.7\% GC | |
content) of S. anaerobius ST1464 comprises one chromosome and no | |
plasmids. The chromosome contains 2,660 open reading frames | |
(ORFs), 49 tRNAs and three complete rRNAs, forming one complete | |
operon. The size of ORFs ranges between 100 to 4,600 bp except | |
for two ORFs of 6,417 and 7,173 bp encoding segregation ATPase | |
and non-ribosomal peptide synthase, respectively. The chromosome | |
harbors Staphylococcus phage 2638A genome and incomplete | |
Staphylococcus phage genome PT1028, but no detectable CRISPRS. | |
The antibiotic resistance gene for tetracycline was found | |
although Staphylococcus aureus subsp. anaerobius is susceptible | |
to tetracycline in-vitro. Intact oxygen detoxification genes | |
encode superoxide dismutase and cytochrome quinol oxidase whereas | |
the catalase gene is impaired by a stop codon. Based on the | |
genome, in-silico multilocus sequence typing indicates that S. | |
aureus subsp. anaerobius emerged as a clone separated from all | |
other S. aureus strains, illustrating host-adaptation linked to | |
missing functions. Availability of S. aureus subsp. anaerobius | |
genome could prompt the development of post-genomic tools for its | |
rapid discrimination from S. aureus.", | |
journal = "Stand. Genomic Sci.", | |
volume = 9, | |
number = 1, | |
pages = "1--13", | |
month = oct, | |
year = 2013, | |
keywords = "Morel's disease; SOLiD; Staphylococcus aureus subsp. anaerobius; | |
genome;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Monecke2013-fe, | |
title = "Genome sequencing and molecular characterisation of | |
Staphylococcus aureus {ST772-MRSA-V}, ``Bengal Bay Clone''", | |
author = "Monecke, Stefan and Baier, Vico and Coombs, Geoffrey W and | |
Slickers, Peter and Ziegler, Albrecht and Ehricht, Ralf", | |
abstract = "BACKGROUND: The PVL-positive ST772-MRSA-V is an emerging | |
community-associated (CA-) MRSA clone that has been named Bengal | |
Bay Clone since most patients have epidemiological connections to | |
the Indian subcontinent. It is found increasingly common in other | |
areas of the world. METHODS: One isolate of ST772-MRSA-V was | |
sequenced using the Illumina Genome Analyzer System. After | |
initial assembling the multiple sequence contigs were analysed | |
using different in-house annotation scripts. Results were | |
compared to microarray hybridisation results of clinical isolates | |
of ST772-MRSA-V, of related strains and to another ST772-MRSA-V | |
genome sequence. RESULTS: According to MLST e-burst analysis, | |
ST772-MRSA-V belongs to Clonal Complex (CC)1, differing from ST1 | |
only in one MLST allele (pta-22). However, there are several | |
additional differences including agr alleles (group II rather | |
than III), capsule type (5 rather than 8), the presence of the | |
egc enterotoxin gene cluster and of the enterotoxin homologue ORF | |
CM14 as well as the absence of the enterotoxin H gene seh. | |
Enterotoxin genes sec and sel are present. ST772-MRSA-V harbours | |
the genes encoding enterotoxin A (sea) and PVL (lukS/F-PV). Both | |
are located on the same prophage. CONCLUSIONS: ST772-MRSA-V may | |
have emerged from the same lineage as globally spread CC1 and CC5 | |
strains. It has acquired a variety of virulence factors, and for | |
a CA-MRSA strain it has an unusually high number of genes | |
associated with antibiotic resistance.", | |
journal = "BMC Res. Notes", | |
volume = 6, | |
pages = "548", | |
month = dec, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Baek2013-yj, | |
title = "Genetic variation in the Staphylococcus aureus 8325 strain | |
lineage revealed by whole-genome sequencing", | |
author = "B{\ae}k, Kristoffer T and Frees, Dorte and Renzoni, Adriana and | |
Barras, Christine and Rodriguez, Natalia and Manzano, Caroline | |
and Kelley, William L", | |
abstract = "Staphylococcus aureus strains of the 8325 lineage, especially | |
8325-4 and derivatives lacking prophage, have been used | |
extensively for decades of research. We report herein the results | |
of our deep sequence analysis of strain 8325-4. Assignment of | |
sequence variants compared with the reference strain 8325 | |
(NRS77/PS47) required correction of errors in the 8325 reference | |
genome, and reassessment of variation previously attributed to | |
chemical mutagenesis of the restriction-defective RN4220. Using | |
an extensive strain pedigree analysis, we discovered that 8325-4 | |
contains 16 single nucleotide polymorphisms (SNP) arising prior | |
to the construction of RN4220. We identified 5 indels in 8325-4 | |
compared with 8325. Three indels correspond to expected $\Phi$11, | |
12, 13 excisions, one indel is explained by a sequence assembly | |
artifact, and the final indel ($\Delta$63bp) in the spa-sarS | |
intergenic region is common to only a sub-lineage of 8325-4 | |
strains including SH1000. This deletion was found to | |
significantly decrease (75\%) steady state sarS but not spa | |
transcript levels in post-exponential phase. The sub-lineage | |
8325-4 was also found to harbor 4 additional SNPs. We also found | |
large sequence variation between 8325, 8325-4 and RN4220 in a | |
cluster of repetitive hypothetical proteins (SA0282 homologs) | |
near the Ess secretion cluster. The overall 8325-4 SNP set | |
results in 17 alterations within coding sequences. Remarkably, we | |
discovered that all tested strains of the 8325-4 lineage lack | |
phenol soluble modulin $\alpha$3 (PSM$\alpha$3), a virulence | |
determinant implicated in neutrophil chemotaxis, biofilm | |
architecture and surface spreading. Collectively, our results | |
clarify and define the 8325-4 pedigree and reveal clear evidence | |
that mutations existing throughout all branches of this lineage, | |
including the widely used RN6390 and SH1000 strains, could | |
conceivably impact virulence regulation.", | |
journal = "PLoS One", | |
volume = 8, | |
number = 9, | |
pages = "e77122", | |
month = sep, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Matsuo2013-qu, | |
title = "Comprehensive identification of mutations responsible for | |
heterogeneous vancomycin-intermediate Staphylococcus aureus | |
({hVISA)-to-VISA} conversion in laboratory-generated {VISA} | |
strains derived from {hVISA} clinical strain Mu3", | |
author = "Matsuo, Miki and Cui, Longzhu and Kim, Jeeyoung and Hiramatsu, | |
Keiichi", | |
abstract = "Heterogeneous vancomycin-intermediate Staphylococcus aureus | |
(hVISA) spontaneously produces VISA cells within its cell | |
population at a frequency of 10(-6) or greater. We established a | |
total of 45 VISA mutant strains independently obtained from hVISA | |
Mu3 and its related strains by one-step vancomycin selection. We | |
then performed high-throughput whole-genome sequencing of the 45 | |
strains and their parent strains to identify the genes involved | |
in the hVISA-to-VISA phenotypic conversion. A comparative genome | |
study showed that all the VISA strains tested carried a unique | |
set of mutations. All of the 45 VISA strains carried 1 to 4 | |
mutations possibly affecting the expression of a total of 48 | |
genes. Among them, 32 VISA strains carried only one gene affected | |
by a single mutation. As many as 20 genes in more than eight | |
functional categories were affected in the 32 VISA strains, which | |
explained the extremely high rates of the hVISA-to-VISA | |
phenotypic conversion. Five genes, rpoB, rpoC, walK, pbp4, and | |
pp2c, were previously reported as being involved in vancomycin | |
resistance. Fifteen remaining genes were newly identified as | |
associated with vancomycin resistance in this study. The gene | |
most frequently affected (6 out of 32 strains) was cmk, which | |
encodes cytidylate kinase, followed closely by rpoB (5 out of | |
32), encoding the $\beta$ subunit of RNA polymerase. A mutation | |
prevalence study also revealed a sizable number of cmk mutants | |
among clinical VISA strains (7 out of 38 [18\%]). Reduced | |
cytidylate kinase activity in cmk mutant strains is proposed to | |
contribute to the hVISA-to-VISA phenotype conversion by | |
thickening the cell wall and reducing the cell growth rate.", | |
journal = "Antimicrob. Agents Chemother.", | |
volume = 57, | |
number = 12, | |
pages = "5843--5853", | |
month = dec, | |
year = 2013, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Chen2013-cu, | |
title = "Complete genome sequence of a {Pant{\'o}n-Valentine} | |
leukocidin-negative community-associated methicillin-resistant | |
Staphylococcus aureus strain of sequence type 72 from Korea", | |
author = "Chen, Yan and Chatterjee, Som S and Porcella, Stephen F and Yu, | |
Yun-Song and Otto, Michael", | |
abstract = "In the past decade, community-associated (CA-) infections with | |
methicillin-resistant Staphylococcus aureus (MRSA) have emerged | |
throughout the world. Different CA-MRSA strains dominate in | |
different geographical locations. Many CA-MRSA lineages contain | |
genes coding for the Pant{\'o}n-Valentine leukocidin. However, | |
the role of this leukotoxin in CA-MRSA pathogenesis is still | |
controversial. The genome sequences of two key PVL-positive | |
CA-MRSA strains (USA300, USA400) have been reported, but we lack | |
information on the more recently found PVL-negative CA-MRSA | |
strains. One such strain is the PVL-negative ST72, the main cause | |
of CA-MRSA infections in Korea. Here, we report the entire genome | |
sequence of CA-MRSA ST72 and analyze its gene content with a | |
focus on virulence factors. Our results show that this strain | |
does not have considerable differences in virulence factor | |
content compared to other CA-MRSA strains (USA300, USA400), | |
indicating that other toxins do not substitute for the lack of | |
PVL in ST72. This finding is in accordance with the notion that | |
differential expression of widespread virulence determinants, | |
rather than the acquisition of additional virulence factors on | |
mobile genetic elements, such as PVL, is responsible for the | |
increased virulence of CA- compared to hospital-associated MRSA.", | |
journal = "PLoS One", | |
volume = 8, | |
number = 8, | |
pages = "e72803", | |
month = aug, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Yoon2013-gy, | |
title = "Characterization and genomic analysis of two Staphylococcus | |
aureus bacteriophages isolated from poultry/livestock farms", | |
author = "Yoon, Hyunjin and Yun, Jiae and Lim, Jeong-A and Roh, Eunjung and | |
Jung, Kyu-Seok and Chang, Yoonjee and Ryu, Sangryeol and Heu, | |
Sunggi", | |
abstract = "Staphylococcus aureus is one of the most important pathogens, | |
causing various diseases in humans and animals. As | |
methicillin-resistant S. aureus (MRSA) has become increasingly | |
prevalent, controlling this pathogen with standard antibiotic | |
treatment has become challenging. Bacteriophages (phages) have | |
attracted interest as alternative antibacterial agents to control | |
MRSA. In this study, we isolated six S. aureus phages from soils | |
of poultry/livestock farms. Based on the results of host range | |
determination with 150 S. aureus strains and restriction enzyme | |
treatment of phage DNA, two phages, designated SP5 and SP6, were | |
selected for further characterization and genome sequencing. Both | |
SP5 and SP6 were classified as members of the family | |
Siphoviridae. The genome of SP5 comprises 43 305 bp and contains | |
63 ORFs, while the SP6 genome comprises 42 902 bp and contains 61 | |
ORFs. Although they have different host spectra, the phage | |
genomes exhibit high nucleotide similarity to each other. | |
Adsorption assay results suggested that the host range | |
determinants of the two phages are involved in both adsorption | |
and infection. Comparative genomic analyses of the two phages | |
provided evidence that the lysogenic/lytic control module and | |
tail proteins may be important for host specificity.", | |
journal = "J. Gen. Virol.", | |
volume = 94, | |
number = "Pt 11", | |
pages = "2569--2576", | |
month = nov, | |
year = 2013, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Botelho2016-yb, | |
title = "Complete genome sequence of an agr-dysfunctional variant of the | |
{ST239} lineage of the methicillin-resistant Staphylococcus | |
aureus strain {GV69} from Brazil", | |
author = "Botelho, Ana M N and Costa, Maiana O C and Beltrame, Cristiana O | |
and Ferreira, Fabienne A and C{\^o}rtes, Marina F and Bandeira, | |
Paula T and Lima, Nicholas C B and Souza, Rangel C and Almeida, | |
Luiz G P and Vasconcelos, Ana T R and Nicol{\'a}s, Marisa F and | |
Figueiredo, Agnes M S", | |
abstract = "Staphylococcus aureus is a versatile Gram-positive coccus | |
frequently found colonizing the skin and nasal membranes of | |
humans. The acquisition of the staphylococcal cassette chromosome | |
mec was a major milestone in the evolutionary path of | |
methicillin-resistant S. aureus. This genetic element carries the | |
mecA gene, the main determinant of methicillin resistance. MRSA | |
is involved in a plethora of opportunistic infectious diseases. | |
The accessory gene regulator is the major S. aureus quorum | |
sensing system, playing an important role in staphylococcal | |
virulence, including the development of biofilms. We report the | |
complete genome sequence (NCBI BioProject ID: PRJNA264181) of the | |
methicillin-resistant S. aureus strain GV69 (= CMVRS P4521), a | |
variant of the ST239 lineage that presents with a natural | |
attenuation of agr-RNAIII transcription and a moderate | |
accumulation of biofilm.", | |
journal = "Stand. Genomic Sci.", | |
volume = 11, | |
pages = "34", | |
month = may, | |
year = 2016, | |
keywords = "Complete genome sequence; Methicillin-resistant Staphylococcus | |
aureus; ST239; Skin hospital infection; agr | |
dysfunction;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Gaviria-Agudelo2015-oi, | |
title = "Genomic Heterogeneity of Methicillin Resistant Staphylococcus | |
aureus Associated with Variation in Severity of Illness among | |
Children with Acute Hematogenous Osteomyelitis", | |
author = "Gaviria-Agudelo, Claudia and Aroh, Chukwuemika and Tareen, | |
Naureen and Wakeland, Edward K and Kim, Minsoo and Copley, Lawson | |
A", | |
abstract = "INTRODUCTION: The association between severity of illness of | |
children with osteomyelitis caused by Methicillin-resistant | |
Staphylococcus aureus (MRSA) and genomic variation of the | |
causative organism has not been previously investigated. The | |
purpose of this study is to assess genomic heterogeneity among | |
MRSA isolates from children with osteomyelitis who have diverse | |
severity of illness. MATERIALS AND METHODS: Children with | |
osteomyelitis were prospectively studied between 2010 and 2011. | |
Severity of illness of the affected children was determined from | |
clinical and laboratory parameters. MRSA isolates were analyzed | |
with next generation sequencing (NGS) and optical mapping. | |
Sequence data was used for multi-locus sequence typing (MLST), | |
phylogenetic analysis by maximum likelihood (PAML), and | |
identification of virulence genes and single nucleotide | |
polymorphisms (SNP) relative to reference strains. RESULTS: The | |
twelve children studied demonstrated severity of illness scores | |
ranging from 0 (mild) to 9 (severe). All isolates were USA300, ST | |
8, SCC mec IVa MRSA by MLST. The isolates differed from reference | |
strains by 2 insertions (40 Kb each) and 2 deletions (10 and 25 | |
Kb) but had no rearrangements or copy number variations. There | |
was a higher occurrence of virulence genes among study isolates | |
when compared to the reference strains (p = 0.0124). There were | |
an average of 11 nonsynonymous SNPs per strain. PAML demonstrated | |
heterogeneity of study isolates from each other and from the | |
reference strains. DISCUSSION: Genomic heterogeneity exists among | |
MRSA isolates causing osteomyelitis among children in a single | |
community. These variations may play a role in the pathogenesis | |
of variation in clinical severity among these children.", | |
journal = "PLoS One", | |
volume = 10, | |
number = 6, | |
pages = "e0130415", | |
month = jun, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Aswani2016-td, | |
title = "Complete Genome Sequence of Staphylococcus aureus {MCRF184}, a | |
Necrotizing {Fasciitis-Causing} {Methicillin-Sensitive} Sequence | |
Type 45 Staphylococcus Strain", | |
author = "Aswani, Vijay and Mau, Bob and Shukla, Sanjay K", | |
abstract = "We report here the complete genome sequence of a highly virulent | |
methicillin-sensitive Staphylococcus aureus strain, MCRF184, | |
belonging to sequence type 45. This staphylococcal strain was | |
isolated from a surgical biopsy specimen from a patient with | |
necrotizing fasciitis.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 3, | |
month = may, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Johnston2016-tz, | |
title = "Genomic Signatures of Experimental Adaptation to Antimicrobial | |
Peptides in Staphylococcus aureus", | |
author = "Johnston, Paul R and Dobson, Adam J and Rolff, Jens", | |
abstract = "The evolution of resistance against antimicrobial peptides has | |
long been considered unlikely due to their mechanism of action, | |
yet experimental selection with AMPs results in rapid evolution | |
of resistance in several species of bacteria. Although numerous | |
studies have utilized mutant screens to identify loci that | |
determine AMP susceptibility, there is a dearth of data | |
concerning the genomic changes which accompany experimental | |
evolution of AMP resistance. Using genome re-sequencing we | |
analysed the mutations which arise during experimental evolution | |
of resistance to the cationic AMPs iseganan, melittin and | |
pexiganan, as well as to a combination of melittin and pexiganan, | |
or to the aminoglycoside antibiotic streptomycin. Analysis of 17 | |
independently replicated Staphylococcus aureus selection lines, | |
including unselected controls, showed that each AMP selected for | |
mutations at distinct loci. We identify mutations in genes | |
involved in the synthesis and maintenance of the cell envelope. | |
This includes genes previously identified from mutant screens for | |
AMP resistance, and genes involved in the response to AMPs and | |
cell-wall-active antibiotics. Furthermore, transposon insertion | |
mutants were used to verify that a number of the identified genes | |
are directly involved in determining AMP susceptibility. Strains | |
selected for AMP resistance under controlled experimental | |
evolution displayed consistent AMP-specific mutations in genes | |
which determine AMP susceptibility. This suggests that different | |
routes to evolve resistance are favored within a controlled | |
genetic background.", | |
journal = "G3", | |
month = apr, | |
year = 2016, | |
keywords = "Staphylococcus aureus; antimicrobial peptide resistance; | |
experimental | |
evolution;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Ugolotti2016-br, | |
title = "Whole-genome sequencing as standard practice for the analysis of | |
clonality in outbreaks of meticillin-resistant Staphylococcus | |
aureus in a paediatric setting", | |
author = "Ugolotti, E and Larghero, P and Vanni, I and Bandettini, R and | |
Tripodi, G and Melioli, G and Di Marco, E and Raso, A and | |
Biassoni, R", | |
abstract = "Meticillin-resistant Staphylococcus aureus (MRSA) is one of the | |
leading causes of hospital-associated infections. This study | |
investigated the potential use of whole-genome sequencing (WGS) | |
for surveillance purposes by re-examining MRSA strains related to | |
past outbreaks among hospitalized paediatric patients. WGS data | |
ameliorated the genotypic profile previously obtained with Sanger | |
sequencing and pulsed-field gel electrophoresis typing, and | |
discriminated between strains that were related and unrelated to | |
the outbreaks. This allowed strain clonality to be defined with a | |
higher level of resolution than achieved previously. This study | |
demonstrates the potential of WGS to trace hospital outbreaks, | |
which may lead to WGS becoming standard practice in outbreak | |
investigations.", | |
journal = "J. Hosp. Infect.", | |
month = apr, | |
year = 2016, | |
keywords = "Hospital-associated infections; MRSA; Paediatric hospital | |
outbreak; Whole-genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Iguchi2016-kk, | |
title = "Rapid Acquisition of Linezolid Resistance in | |
{Methicillin-Resistant} Staphylococcus aureus: Role of | |
Hypermutation and Homologous Recombination", | |
author = "Iguchi, Shigekazu and Mizutani, Tomonori and Hiramatsu, Keiichi | |
and Kikuchi, Ken", | |
abstract = "BACKGROUND: We previously reported the case of a 64-year-old man | |
with mediastinitis caused by Staphylococcus aureus in which the | |
infecting bacterium acquired linezolid resistance after only 14 | |
days treatment with linezolid. We therefore investigated relevant | |
clinical isolates for possible mechanisms of this rapid | |
acquisition of linezolid resistance. METHODS: Using clinical S. | |
aureus isolates, we assessed the in vitro mutation rate and | |
performed stepwise selection for linezolid resistance. To | |
investigate homologous recombination, sequences were determined | |
for each of the 23S ribosomal RNA (23S rRNA) loci; analyzed | |
sequences spanned the entirety of each 23S rRNA gene, including | |
domain V, as well as the 16S-23S intergenic spacer regions. We | |
additionally performed next-generation sequencing on clinical | |
strains to identify single-nucleotide polymorphisms compared to | |
the N315 genome. RESULTS: Strains isolated from the patient prior | |
to linezolid exposure (M5-M7) showed higher-level linezolid | |
resistance than N315, and the pre-exposure strain (M2) exhibited | |
more rapid acquisition of linezolid resistance than did N315. | |
However, the mutation rates of these and contemporaneous clinical | |
isolates were similar to those of N315, and the isolates did not | |
exhibit any mutations in hypermutation-related genes. Sequences | |
of the 23S rRNA genes and 16S-23S intergenic spacer regions were | |
identical among the pre- and post-exposure clinical strains. | |
Notably, all of the pre-exposure isolates harbored a recQ | |
missense mutation (Glu69Asp) with respect to N315; such a lesion | |
may have affected short sequence recombination (facilitating, for | |
example, recombination among rrn loci). We hypothesize that this | |
mechanism contributed to rapid acquisition of linezolid | |
resistance. CONCLUSIONS: Hypermutation and homologous | |
recombination of the ribosomal RNA genes, including 23S rRNA | |
genes, appear not to have been sources of the accelerated | |
acquisition of linezolid resistance observed in our clinical | |
case. Increased frequency of short sequence recombination may | |
have resulted from a recQ variant in the infecting organism.", | |
journal = "PLoS One", | |
volume = 11, | |
number = 5, | |
pages = "e0155512", | |
month = may, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Ward2016-qy, | |
title = "Identification of source and sink populations for the emergence | |
and global spread of the {East-Asia} clone of | |
community-associated {MRSA}", | |
author = "Ward, Melissa J and Goncheva, Mariya and Richardson, Emily and | |
McAdam, Paul R and Raftis, Emma and Kearns, Angela and Daum, | |
Robert S and David, Michael Z and Lauderdale, Tsai Ling and | |
Edwards, Giles F and Nimmo, Graeme R and Coombs, Geoffrey W and | |
Huijsdens, Xander and Woolhouse, Mark E J and Fitzgerald, J Ross", | |
abstract = "BACKGROUND: Our understanding of the factors influencing the | |
emergence, dissemination and global distribution of epidemic | |
clones of bacteria is limited. ST59 is a major epidemic clone of | |
community-associated MRSA in East Asia, responsible for extensive | |
morbidity and mortality, but has a much lower prevalence in other | |
parts of the world. The geographic origin of ST59 and its | |
international routes of dissemination are unclear and disputed in | |
the literature. RESULTS: To investigate the origin and spread of | |
the ST59 clone, we obtained whole genome sequences of isolates | |
from four continents, sampled over more than a decade, and | |
carried out a time-scaled phylogeographic analysis. We discover | |
that two distinct ST59 clades emerged concurrently, in East Asia | |
and the USA, but underwent clonal expansion at different times. | |
The East Asia clade was strongly enriched for gene determinants | |
associated with antibiotic resistance, consistent with regional | |
differences in antibiotic usage. Both clones spread independently | |
to Australia and Europe, and we found evidence of the persistence | |
of multi-drug resistance following export from East Asia. Direct | |
transfer of strains between Taiwan and the USA was not observed | |
in either direction, consistent with geographic niche exclusion. | |
CONCLUSIONS: Our results resolve a longstanding controversy | |
regarding the origin of the ST59 clone, revealing the major | |
global source and sink populations and routes for the spread of | |
multi-drug resistant clones. Additionally, our findings indicate | |
that diversification of the accessory genome of epidemic clones | |
partly reflects region-specific patterns of antibiotic usage, | |
which may influence bacterial fitness after transmission to | |
different geographic locations.", | |
journal = "Genome Biol.", | |
volume = 17, | |
number = 1, | |
pages = "160", | |
month = jul, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Lozano2012-xl, | |
title = "Expansion of a Plasmid Classification System for {Gram-Positive} | |
Bacteria and Determination of the Diversity of Plasmids in | |
Staphylococcus aureus Strains of Human, Animal, and Food Origins", | |
author = "Lozano, C and Garcia-Migura, L and Aspiroz, C and Zarazaga, M and | |
Torres, C and Aarestrup, F M", | |
journal = "Appl. Environ. Microbiol.", | |
volume = 78, | |
number = 16, | |
pages = "5948--5955", | |
month = aug, | |
year = 2012, | |
keywords = "Staphylococcus;plasmid;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Bosi2016-fz, | |
title = "Comparative genome-scale modelling of Staphylococcus aureus | |
strains identifies strain-specific metabolic capabilities linked | |
to pathogenicity", | |
author = "Bosi, Emanuele and Monk, Jonathan M and Aziz, Ramy K and Fondi, | |
Marco and Nizet, Victor and Palsson, Bernhard {\O}", | |
abstract = "Staphylococcus aureus is a preeminent bacterial pathogen capable | |
of colonizing diverse ecological niches within its human host. We | |
describe here the pangenome of S. aureus based on analysis of | |
genome sequences from 64 strains of S. aureus spanning a range of | |
ecological niches, host types, and antibiotic resistance | |
profiles. Based on this set, S. aureus is expected to have an | |
open pangenome composed of 7,411 genes and a core genome composed | |
of 1,441 genes. Metabolism was highly conserved in this core | |
genome; however, differences were identified in amino acid and | |
nucleotide biosynthesis pathways between the strains. | |
Genome-scale models (GEMs) of metabolism were constructed for the | |
64 strains of S. aureus These GEMs enabled a systems approach to | |
characterizing the core metabolic and panmetabolic capabilities | |
of the S. aureus species. All models were predicted to be | |
auxotrophic for the vitamins niacin (vitamin B3) and thiamin | |
(vitamin B1), whereas strain-specific auxotrophies were predicted | |
for riboflavin (vitamin B2), guanosine, leucine, methionine, and | |
cysteine, among others. GEMs were used to systematically analyze | |
growth capabilities in more than 300 different growth-supporting | |
environments. The results identified metabolic capabilities | |
linked to pathogenic traits and virulence acquisitions. Such | |
traits can be used to differentiate strains responsible for mild | |
vs. severe infections and preference for hosts (e.g., animals vs. | |
humans). Genome-scale analysis of multiple strains of a species | |
can thus be used to identify metabolic determinants of virulence | |
and increase our understanding of why certain strains of this | |
deadly pathogen have spread rapidly throughout the world.", | |
journal = "Proc. Natl. Acad. Sci. U. S. A.", | |
month = jun, | |
year = 2016, | |
keywords = "core genome; mathematical modeling; pangenome; pathogenicity; | |
systems | |
biology;Staphylococcus;pangenome;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Jaton2016-ll, | |
title = "Common skin infection due to {Panton-Valentine} | |
leucocidin-producing Staphylococcus aureus strains in asylum | |
seekers from Eritrea: a genome-based investigation of a suspected | |
outbreak", | |
author = "Jaton, L and Pillonel, T and Jaton, K and Dory, E and Prod'hom, G | |
and Blanc, D S and Tissot, F and Bodenmann, P and Greub, G", | |
abstract = "OBJECTIVES: Since late 2014, multiple cases of abscesses and | |
boils due to methicillin susceptible Staphylococcus aureus (MSSA) | |
expressing the Panton-Valentine leukocidin (PVL) were observed in | |
Eritrean asylum seekers in Lausanne, Switzerland. Strains | |
isolated from infected Eritrean and non-Eritrean patients were | |
compared by whole genome sequencing (WGS) to determine whether | |
these numerous cases results from an outbreak. METHODS: The | |
genome of S. aureus PVL-producing strains were sequenced and | |
compared. Clinical and epidemiological characteristics of | |
patients infected by PVL-producing strains were investigated. | |
RESULTS: This work reports 15 cases of infections due to | |
PVL-producing strains affecting mostly asylum seekers (n=10), | |
people working with refugees and/or exposed to Africans (n=3). | |
Most infections were due to closely related strains of CC152 | |
(n=8) and CC15 (n=3), two distantly related (>34,000 core single | |
nucleotide polymorphisms [SNPs]) clonal complexes. | |
Epidemiological link between the 15 cases could be ruled out by | |
WGS (33 to 172 core SNPs between the different strains of a given | |
complex). Altogether, these results reflect the probable high | |
incidence of CC15 and CC152 PVL-producing strains in eastern | |
Africa. CONCLUSIONS: Clinicians facing unusual skin infections in | |
African refugees (or in any person returning from this high | |
endemic region) should consider S. aureus PVL-producer before | |
suspecting rare infections such as leishmaniosis or | |
rickettsiosis. Clinicians should also remember that PVL are | |
frequently expressed by MSSA in some regions of the world and | |
that antibiotics efficient on the toxin expression, such as | |
clindamycin, represent the best therapeutic option.", | |
journal = "Clin. Microbiol. Infect.", | |
month = jun, | |
year = 2016, | |
keywords = "Panton-Valentine leukocidin; ST132; Staphylococcus aureus; genome | |
sequence; migrants; skin | |
infection;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Winstel2013-hw, | |
title = "Wall teichoic acid structure governs horizontal gene transfer | |
between major bacterial pathogens", | |
author = "Winstel, Volker and Liang, Chunguang and Sanchez-Carballo, | |
Patricia and Steglich, Matthias and Munar, Marta and Br{\"o}ker, | |
Barbara M and Penad{\'e}s, Jose R and N{\"u}bel, Ulrich and | |
Holst, Otto and Dandekar, Thomas and Peschel, Andreas and Xia, | |
Guoqing", | |
abstract = "Mobile genetic elements (MGEs) encoding virulence and resistance | |
genes are widespread in bacterial pathogens, but it has remained | |
unclear how they occasionally jump to new host species. | |
Staphylococcus aureus clones exchange MGEs such as S. aureus | |
pathogenicity islands (SaPIs) with high frequency via helper | |
phages. Here we report that the S. aureus ST395 lineage is | |
refractory to horizontal gene transfer (HGT) with typical S. | |
aureus but exchanges SaPIs with other species and genera | |
including Staphylococcus epidermidis and Listeria monocytogenes. | |
ST395 produces an unusual wall teichoic acid (WTA) resembling | |
that of its HGT partner species. Notably, distantly related | |
bacterial species and genera undergo efficient HGT with typical | |
S. aureus upon ectopic expression of S. aureus WTA. Combined with | |
genomic analyses, these results indicate that a 'glycocode' of | |
WTA structures and WTA-binding helper phages permits HGT even | |
across long phylogenetic distances thereby shaping the evolution | |
of Gram-positive pathogens.", | |
journal = "Nat. Commun.", | |
volume = 4, | |
pages = "2345", | |
year = 2013, | |
keywords = "Staphylococcus;evolution;recombination;phage;Staph\_evolutionary\_history;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Azarian2016-ta, | |
title = "Intrahost Evolution of {Methicillin-Resistant} Staphylococcus | |
aureus {USA300} Among Individuals With Reoccurring Skin and | |
{Soft-Tissue} Infections", | |
author = "Azarian, Taj and Daum, Robert S and Petty, Lindsay A and | |
Steinbeck, Jenny L and Yin, Zachary and Nolan, David and | |
Boyle-Vavra, Susan and Hanage, W P and Salemi, Marco and David, | |
Michael Z", | |
abstract = "BACKGROUND: Methicillin-resistant Staphylococcus aureus (MRSA) | |
USA300 is the leading cause of MRSA infections in the United | |
States and has caused an epidemic of skin and soft-tissue | |
infections. Recurrent infections with USA300 MRSA are common, yet | |
intrahost evolution during persistence on an individual has not | |
been studied. This gap hinders the ability to clinically manage | |
recurrent infections and reconstruct transmission networks. | |
METHODS: To characterize bacterial intrahost evolution, we | |
examined the clinical courses of 4 subjects with 3-6 recurrent | |
USA300 MRSA infections, using patient clinical data, including | |
antibiotic exposure history, and whole-genome sequencing and | |
phylogenetic analysis of all available MRSA isolates (n = 29). | |
RESULTS: Among sequential isolates, we found variability in | |
diversity, accumulation of mutations, and mobile genetic | |
elements. Selection for antimicrobial-resistant populations was | |
observed through both an increase in the number of plasmids | |
conferring multidrug resistance and strain replacement by a | |
resistant population. Two of 4 subjects had strain replacement | |
with a genetically distinct USA300 MRSA population. DISCUSSIONS: | |
During a 5-year period in 4 subjects, we identified development | |
of antimicrobial resistance, intrahost evolution, and strain | |
replacement among isolates from patients with recurrent MRSA | |
infections. This calls into question the efficacy of | |
decolonization to prevent recurrent infections and highlights the | |
adaptive potential of USA300 and the need for effective sampling.", | |
journal = "J. Infect. Dis.", | |
volume = 214, | |
number = 6, | |
pages = "895--905", | |
month = sep, | |
year = 2016, | |
keywords = "MRSA; Staphylococcus aureus; USA300; antibiotic resistance; | |
evolution; intrahost; phylogenetic; recurrent infections; skin | |
and soft-tissue infections; whole-genome sequencing; within | |
host;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Spoor2015-ub, | |
title = "Recombination-mediated remodelling of host--pathogen | |
interactions during Staphylococcus aureus niche adaptation", | |
author = "Spoor, Laura E and Richardson, Emily and Richards, Amy C and | |
Wilson, Gillian J and Mendonca, Chriselle and Gupta, Ravi Kr and | |
McAdam, Paul R and Nutbeam-Tuffs, Stephen and Black, Nikki S and | |
O'Gara, James P and Lee, Chia Y and Corander, Jukka and Ross | |
Fitzgerald, J", | |
abstract = "Large-scale recombination events have led to the emergence of | |
epidemic clones of several major bacterial pathogens. However, | |
the functional impact of the recombination on clonal success is | |
not understood. Here, we identified a novel widespread hybrid | |
clone (ST71) of livestock-associated Staphylococcus aureus that | |
evolved from an ancestor belonging to the major bovine lineage | |
CC97, through multiple large-scale recombination events with | |
other S. aureus lineages occupying the same ruminant niche. The | |
recombination events, affecting a 329 kb region of the | |
chromosome spanning the origin of replication, resulted in | |
allele replacement and loss or gain of an array of genes | |
influencing host--pathogen interactions. Of note, molecular | |
functional analyses revealed that the ST71 hybrid clone has | |
acquired multiple novel pathogenic traits associated with | |
acquired and innate immune evasion and bovine extracellular | |
matrix adherence. These findings provide a paradigm for the | |
impact of large-scale recombination events on the rapid | |
evolution of bacterial pathogens within defined ecological | |
niches.", | |
journal = "Microbial Genomics", | |
publisher = "Microbiology Society", | |
volume = 1, | |
number = 4, | |
month = oct, | |
year = 2015, | |
keywords = "recombination; remodelling; Staphylococcus aureus; | |
host--pathogen interactions; niche | |
adaptation;recombination;Staphylococcus;host-pathogen;Staph\_evolutionary\_history;Staph\_evolutionary\_history/staph | |
microbial interactions;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@UNPUBLISHED{Thorpe2016-nj, | |
title = "The large majority of intergenic sites in bacteria are | |
selectively constrained, even when known regulatory elements are | |
excluded", | |
author = "Thorpe, Harry A and Bayliss, Sion C and Hurst, Laurence D and | |
Feil, Edward J", | |
abstract = "There are currently no broad estimates of the overall strength | |
and direction of selection operating on intergenic variation in | |
bacteria. Here we address this using large whole genome sequence | |
datasets representing six diverse bacterial species; Escherichia | |
coli, Staphylococcus aureus, Salmonella enterica, Streptococcus | |
pneumoniae, Klebsiella pneumoniae, and Mycobacterium | |
tuberculosis. Excluding M. tuberculosis, we find that a high | |
proportion (62\%-79\%; mean 70\%) of intergenic sites are | |
selectively constrained, relative to synonymous sites. Non-coding | |
RNAs tend to be under stronger selective constraint than | |
promoters, which in turn are typically more constrained than | |
rho-independent terminators. Even when these regulatory elements | |
are excluded, the mean proportion of constrained intergenic sites | |
only falls to 69\%; thus our current understanding of the | |
functionality of intergenic regions (IGRs) in bacteria is | |
severely limited. Consistent with a role for positive as well as | |
negative selection on intergenic sites, we present evidence for | |
strong positive selection in Mycobacterium tuberculosis | |
promoters, underlining the key role of regulatory changes as an | |
adaptive mechanism in this highly monomorphic pathogen.", | |
journal = "bioRxiv", | |
pages = "069708", | |
month = jan, | |
year = 2016, | |
keywords = "evolution;Staphylococcus;bacterial comparative | |
genomics;Staph\_evolutionary\_history/Teaching\_Prokaryotic\_evolution;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;microRNA\_in\_bacteria", | |
language = "en" | |
} | |
@ARTICLE{Santos2016-wg, | |
title = "Draft Genome Sequence of the Aureocin {A53-Producing} Strain | |
Staphylococcus aureus {A53}", | |
author = "Santos, Olinda Cabral Silva and Duarte, Andreza Freitas Souza and | |
Albano, Rodolpho Mattos and Bastos, Maria Carmo Freire", | |
abstract = "Here, we present the 2,658,363-bp draft genome sequence of the | |
aureocin A53-producing strain Staphylococcus aureus A53. This | |
genome information may contribute to the optimal and rational | |
exploitation of aureocin A53 as an antimicrobial agent and to its | |
production in large scale.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 4, | |
month = aug, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Ward2016-pz, | |
title = "Identification of source and sink populations for the emergence | |
and global spread of the {East-Asia} clone of | |
community-associated {MRSA}", | |
author = "Ward, Melissa J and Goncheva, Mariya and Richardson, Emily and | |
McAdam, Paul R and Raftis, Emma and Kearns, Angela and Daum, | |
Robert S and David, Michael Z and Lauderdale, Tsai Ling and | |
Edwards, Giles F and Nimmo, Graeme R and Coombs, Geoffrey W and | |
Huijsdens, Xander and Woolhouse, Mark E J and Fitzgerald, J Ross", | |
abstract = "Our understanding of the factors influencing the emergence, | |
dissemination and global distribution of epidemic clones of | |
bacteria is limited. ST59 is a major epidemic clone of | |
community-associated MRSA in East Asia, responsible for extensive | |
morbidity and mortality, but has a much lower prevalence in other | |
parts of the world. The geographic origin of ST59 and its | |
international routes of dissemination are unclear and disputed in | |
the literature.", | |
journal = "Genome Biol.", | |
volume = 17, | |
number = 1, | |
pages = "1--13", | |
year = 2016, | |
keywords = " | |
Staphylococcus;antibiotics;Staph\_evolutionary\_history;bacterial | |
comparative | |
genomics;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Weber2016-fq, | |
title = "Complete Genome Sequences of Two {Methicillin-Sensitive} | |
Staphylococcus aureus Isolates Representing a Population Subset | |
Highly Prevalent in Human Colonization", | |
author = "Weber, Robert E and Layer, Franziska and Fuchs, Stephan and | |
Bender, Jennifer K and Fiedler, Stefan and Werner, Guido and | |
Strommenger, Birgit", | |
abstract = "Here, we report the high-quality draft genome sequences of two | |
methicillin-susceptible Staphylococcus aureus isolates, 08-02119 | |
and 08-02300. Belonging to sequence type 582 (ST582) and ST7, | |
both isolates are representatives of clonal lineages often | |
associated with asymptomatic colonization of humans.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 4, | |
month = jul, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Senghore2016-mp, | |
title = "Whole-genome sequencing reveals transmission of Staphylococcus | |
aureus from humans to green monkeys in The Gambia", | |
author = "Senghore, Madikay and Bayliss, Sion C and Kwambana-Adams, Brenda | |
A and Foster-Nyarko, Ebenezer and Manneh, Jainaba and Dione, | |
Michel and Badji, Henry and Ebruke, Chinelo and Doughty, Emma L | |
and Thorpe, Harry A and Jasinska, Anna J and Schmitt, Christopher | |
A and Cramer, Jennifer D and Turner, Trudy R and Weinstock, | |
George and Freimer, Nelson B and Pallen, Mark J and Feil, Edward | |
J and Antonio, Martin", | |
abstract = "Staphylococcus aureus is an important pathogen of humans and | |
animals. We genome-sequenced ninety S. aureus isolates from the | |
Gambia: 46 isolates from invasive disease in humans, 13 human | |
carriage isolates and 31 monkey carriage isolates. We inferred | |
multiple anthroponotic transmissions of S. aureus from humans to | |
green monkeys, Chlorocebus sabaeus in The Gambia over different | |
timescales. We report a novel monkey-associated clade of S. | |
aureus that emerged from a human-to-monkey switch estimated to | |
have occurred 2,700 years ago. Adaptation of this lineage to the | |
monkey host is accompanied by loss of phage-encoded genes that | |
are known to play an important role in human colonisation. We | |
also report recent anthroponotic transmission of the | |
well-characterised human lineages, ST6 and ST15, to monkeys, | |
probably because of steadily increasing encroachment of humans | |
into the monkeys' habitat. Although we have found no evidence of | |
transmission of S. aureus from monkeys to humans, as the two | |
species come into ever-closer contact there might be an increased | |
risk of additional inter-species exchanges of potential | |
pathogens. IMPORTANCE: The population structures of | |
Staphylococcus aureus in humans and monkeys in sub-Saharan Africa | |
have been previously described using MLST. However, these data | |
lack the power to accurately infer details regarding the origin | |
and maintenance of new adaptive lineages. Here, we describe the | |
use of whole genome sequencing to detect transmission of S. | |
aureus between humans and non-human primates and to document the | |
genetic changes accompanying host adaptation. We note that human | |
to monkey switches tend to be more common than the reverse, and | |
that a novel monkey associated clade is likely to have emerged | |
from such a switch approximately 2700 years ago. Moreover, | |
analysis of the accessory genome provides important clues as to | |
the genetic changes underpinning host adaptation, and in | |
particular show that human to monkey switches tend to be | |
associated with the loss of genes known to confer adaptation to | |
the human host.", | |
journal = "Appl. Environ. Microbiol.", | |
month = jul, | |
year = 2016, | |
keywords = "Staphylococcus;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Monecke2015-ob, | |
title = "A clonal complex 12 methicillin-resistant Staphylococcus aureus | |
strain, West Australian {MRSA-59}, harbors a novel | |
{pseudo-SCCmec} element", | |
author = "Monecke, Stefan and Coombs, Geoffrey W and Pearson, Julie and | |
Hotzel, Helmut and Slickers, Peter and Ehricht, Ralf", | |
abstract = "A West Australian methicillin-resistant Staphylococcus aureus | |
strain (WA MRSA-59) was characterized by microarray and | |
sequencing. Its pseudo-staphylococcal cassette chromosome mec | |
(SCCmec) element comprised dcs, Q9XB68-dcs, mvaS-SCC, Q5HJW6, | |
dru, ugpQ, ydeM, mecA-mecR-mecI, txbi mecI, tnp IS431, copA2-mco | |
(copper resistance), ydhK, arsC-arsB-arsR (arsenic resistance), | |
open reading frame PT43, and per-2. Recombinase genes, xylR | |
(mecR2), and PSM-mec (phenol-soluble modulin) were absent. We | |
suggest that mec complex A should be split into two subtypes. One | |
harbors PSM-mec and xylR (mecR2). It is found in SCCmec types II, | |
III, and VIII. The second subtype, described herein, is present | |
in WA MRSA-59 and some coagulase-negative staphylococci.", | |
journal = "Antimicrob. Agents Chemother.", | |
volume = 59, | |
number = 11, | |
pages = "7142--7144", | |
month = nov, | |
year = 2015, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Davis2015-op, | |
title = "Genome sequencing reveals strain dynamics of | |
methicillin-resistant Staphylococcus aureus in the same household | |
in the context of clinical disease in a person and a dog", | |
author = "Davis, Meghan F and Misic, Ana M and Morris, Daniel O and Moss, | |
John T and Tolomeo, Pam and Beiting, Daniel P and Nachamkin, | |
Irving and Lautenbach, Ebbing and Rankin, Shelley C", | |
abstract = "The strain dynamics of methicillin-resistant Staphylococcus | |
aureus (MRSA) isolates from people and the household dog were | |
investigated. The isolates were identified in the context of a | |
randomized controlled trial that tested household-wide | |
decolonization of people. Genotypic comparison of MRSA isolates | |
obtained from two household members, the dog, and home surfaces | |
over a three-month period failed to implicate the pet or the home | |
environment in recurrent colonization of the household members. | |
However, it did implicate the pet's bed in exposure of the dog | |
prior to the dog's infection. Whole genome sequencing was | |
performed to differentiate the isolates. This report also | |
describes introduction of diverse strains of MRSA into the | |
household within six weeks of cessation of harmonized | |
decolonization treatment of people and treatment for infection in | |
the dog. These findings suggest that community sources outside | |
the home may be important for recurrent MRSA colonization or | |
infection.", | |
journal = "Vet. Microbiol.", | |
volume = 180, | |
number = "3-4", | |
pages = "304--307", | |
month = nov, | |
year = 2015, | |
keywords = "Antimicrobial resistance; Canine; MRSA; SSI; | |
SSTI;Staphylococcus;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Yan2016-hn, | |
title = "Genetic features of livestock-associated Staphylococcus aureus | |
{ST9} isolates from Chinese pigs that carry the lsa(E) gene for | |
quinupristin/dalfopristin resistance", | |
author = "Yan, Xiaomei and Li, Zongwei and Chlebowicz, Monika A and Tao, | |
Xiaoxia and Ni, Ming and Hu, Yuan and Li, Zhen and Grundmann, | |
Hajo and Murray, Susan and Pascoe, Ben and Sheppard, Samuel K and | |
Bo, Xiaochen and Dijl, Jan Maarten van and Du, Pengcheng and | |
Zhang, Minli and You, Yuanhai and Yu, Xiaojie and Meng, Fanliang | |
and Wang, Shengqi and Zhang, Jianzhong", | |
abstract = "Whole-genome sequencing (WGS) was used to investigate the genetic | |
features of the recently identified lsa(E) gene in porcine S. | |
aureus ST9 isolates. Three quinupristin/dalfopristin-resistant | |
isolates harboring the lsa(E) gene (two MRSA and one MSSA) were | |
sequenced. Phylogenetic analysis of 184S. aureus genomes showed | |
that ST9 porcine isolates belong to a distinct sequence cluster. | |
Further analysis showed that all isolates were deficient in the | |
recently described type IV restriction-modification system and | |
SCCmec type XII was identified in the two MRSA isolates, which | |
included a rare class C2 mec gene complex. A 24kb $\Psi$SCC | |
fragment was found in the MRSA and MSSA isolates sharing 99\% | |
nucleotide sequence homology with the $\Psi$SCCJCSC6690 (O-2) | |
element of a ST9 MRSA isolate from Thailand (accession number | |
AB705453). Comparison of these ST9 isolates with 181 publically | |
available S. aureus genomes identified 24 genes present in all | |
(100\%) ST9 isolates, that were absent from the most closely | |
related human isolate. Our analysis suggests that the sequenced | |
quinupristin/dalfopristin-resistant ST9 lineage represent a | |
reservoir of mobile genetic elements associated with resistance | |
and virulence features.", | |
journal = "Int. J. Med. Microbiol.", | |
month = aug, | |
year = 2016, | |
keywords = "ABC transporters; Livestock-associated; | |
Quinupristin/dalfopristin; Resistance; ST9; Staphylococcus | |
aureus;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Hughes2015-sf, | |
title = "Clonal variation in high- and low-level phenotypic and genotypic | |
mupirocin resistance of {MRSA} isolates in south-east London", | |
author = "Hughes, John and Stabler, Richard and Gaunt, Michael and Karadag, | |
Tacim and Desai, Nergish and Betley, Jason and Ioannou, Avgousta | |
and Aryee, Anna and Hearn, Pasco and Marbach, Helene and Patel, | |
Amita and Otter, Jonathan A and Edgeworth, Jonathan D and Tosas | |
Auguet, Olga", | |
abstract = "OBJECTIVES: Both low-level mupirocin resistance (LMR) and | |
high-level mupirocin resistance (HMR) have been identified. The | |
aim of this study was to determine the epidemiology of LMR and | |
HMR in MRSA isolates at five hospitals that have used mupirocin | |
for targeted decolonization as part of successful institutional | |
control programmes. METHODS: All MRSA identified in three | |
microbiology laboratories serving five central and south-east | |
London hospitals and surrounding communities between November | |
2011 and February 2012 were included. HMR and LMR were determined | |
by disc diffusion testing. WGS was used to derive multilocus | |
sequence types (MLSTs) and the presence of HMR and LMR resistance | |
determinants. RESULTS: Prevalence of either HMR or LMR amongst | |
first healthcare episode isolates from 795 identified patients | |
was 9.69\% (95\% CI 7.72-11.96); LMR was 6.29\% (95\% CI | |
4.70-8.21) and HMR was 3.40\% (95\% CI 2.25-4.90). Mupirocin | |
resistance was not significantly different in isolates identified | |
from inpatients at each microbiology laboratory, but was more | |
common in genotypically defined 'hospital' rather than | |
'community' isolates (OR 3.17, 95\% CI 1.36-9.30, P = 0.002). LMR | |
was associated with inpatient stay, previous history of MRSA and | |
age $\geq$65 years; HMR was associated with age $\geq$65 years | |
and residential postcode outside London. LMR and HMR varied by | |
clone, with both being low in the dominant UK MRSA clone ST22 | |
compared with ST8, ST36 and ST239/241 for LMR and with ST8 and | |
ST36 for HMR. V588F mutation and mupA carriage had high | |
specificity (>97\%) and area under the curve (>83\%) to | |
discriminate phenotypic mupirocin resistance, but uncertainty | |
around the sensitivity point estimate was large (95\% CI | |
52.50\%-94.44\%). Mutations in or near the mupA gene were found | |
in eight isolates that carried mupA but were not HMR. | |
CONCLUSIONS: Mupirocin resistance was identified in <10\% of | |
patients and varied significantly by clone, implying that changes | |
in clonal epidemiology may have an important role in determining | |
the prevalence of resistance in conjunction with selection due to | |
mupirocin use.", | |
journal = "J. Antimicrob. Chemother.", | |
volume = 70, | |
number = 12, | |
pages = "3191--3199", | |
month = dec, | |
year = 2015, | |
keywords = "Staphylococcus;antibiotics;confirmed\_SA-resistance-mutation;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{Mellmann2016-os, | |
title = "{Real-Time} Genome Sequencing of Resistant Bacteria Provides | |
Precision Infection Control in an Institutional Setting", | |
author = "Mellmann, Alexander and Bletz, Stefan and B{\"o}king, Thomas and | |
Kipp, Frank and Becker, Karsten and Schultes, Anja and Prior, | |
Karola and Harmsen, Dag", | |
abstract = "The increasing prevalence of multidrug-resistant (MDR) bacteria | |
is a serious global challenge. Here, we studied prospectively | |
whether bacterial whole-genome sequencing (WGS) for real-time MDR | |
surveillance is technical feasible, returns actionable results, | |
and is cost-beneficial. WGS was applied to all MDR isolates of | |
four species (methicillin-resistant Staphylococcus aureus [MRSA], | |
vancomycin-resistant Enterococcus faecium, MDR Escherichia coli, | |
and MDR Pseudomonas aeruginosa) at the University Hospital | |
Muenster, Muenster, Germany, a tertiary care hospital with 1,450 | |
beds, during two 6-month intervals. Turnaround times (TAT) were | |
measured, and total costs for sequencing per isolate were | |
calculated. After cancelling prior policies of preemptive | |
isolation of patients harboring certain Gram-negative MDR | |
bacteria in risk areas, the second interval was conducted. During | |
interval I, 645 bacterial isolates were sequenced. From culture, | |
TATs ranged from 4.4 to 5.3 days, and costs were €202.49 per | |
isolate. During interval II, 550 bacterial isolates were | |
sequenced. Hospital-wide transmission rates of the two most | |
common species (MRSA and MDR E. coli) were low during interval I | |
(5.8\% and 2.3\%, respectively) and interval II (4.3\% and 5.0\%, | |
respectively). Cancellation of isolation of patients infected | |
with non-pan-resistant MDR E. coli in risk wards did not increase | |
transmission. Comparing sequencing costs with avoided costs | |
mostly due to fewer blocked beds during interval II, we saved in | |
excess of €200,000. Real-time microbial WGS in our institution | |
was feasible, produced precise actionable results, helped us to | |
monitor transmission rates that remained low following a | |
modification in isolation procedures, and ultimately saved costs.", | |
journal = "J. Clin. Microbiol.", | |
volume = 54, | |
number = 12, | |
pages = "2874--2881", | |
month = dec, | |
year = 2016, | |
keywords = "Staphylococcus;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers;Anti-infectives\_course", | |
language = "en" | |
} | |
@ARTICLE{Kleinert2016-mz, | |
title = "Draft Genome Sequences of Three Northern German Epidemic | |
Staphylococcus aureus ({ST247}) Strains Containing Multiple | |
Copies of {IS256}", | |
author = "Kleinert, Franziska and Kallies, Ren{\'e} and Zweynert, Annegret | |
and Bierbaum, Gabriele", | |
abstract = "We report the draft genome sequences of three multiresistant | |
Staphylococcus aureus strains of sequence type 247 (ST247). The | |
methicillin-resistant S. aureus (MRSA) SA1450/94 is vancomycin | |
susceptible, while the clinical MRSA isolate S. aureus SA137/93A | |
and its spontaneous laboratory mutant SA137/93G are characterized | |
by intermediate vancomycin susceptibility.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 5, | |
month = sep, | |
year = 2016, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Papers\_for\_Michelle;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers", | |
language = "en" | |
} | |
@ARTICLE{Cao2016-tc, | |
title = "Identification and functional study of type {III-A} {CRISPR-Cas} | |
systems in clinical isolates of Staphylococcus aureus", | |
author = "Cao, Linyan and Gao, Chun-Hui and Zhu, Jiade and Zhao, Liping and | |
Wu, Qingfa and Li, Min and Sun, Baolin", | |
abstract = "The CRISPR-Cas (clustered regularly interspaced short palindromic | |
repeats [CRISPR]-CRISPR associated proteins [Cas]) system can | |
provide prokaryote with immunity against invading mobile genetic | |
elements (MGEs) such as phages and plasmids, which are the main | |
sources of staphylococcal accessory genes. To date, only a few | |
Staphylococcus aureus strains containing CRISPR-Cas systems have | |
been identified, but no functional study in these strains has | |
been reported. In this study, 6 clinical isolates of S. aureus | |
with type III-A CRISPR-Cas systems were identified, and | |
whole-genome sequencing and functional study were conducted | |
subsequently. Genome sequence analysis revealed a close linkage | |
between the CRISPR-Cas system and the staphylococcal cassette | |
chromosome mec (SCCmec) element in five strains. Comparative | |
sequence analysis showed that the type III-A repeats are | |
conserved within staphylococci, despite of the decreased | |
conservation in trailer-end repeats. Highly homologous sequences | |
of some spacers were identified in staphylococcal MGEs, and | |
partially complementary sequences of spacers were mostly found in | |
the coding strand of lytic regions in staphylococcal phages. | |
Transformation experiments showed that S. aureus type III-A | |
CRISPR-Cas system can specifically prevent plasmid transfer in a | |
transcription-dependent manner. Base paring between crRNA and | |
target sequence, the endoribonuclease, and the Csm complex were | |
proved to be necessary for type III-A CRISPR-Cas immunity.", | |
journal = "Int. J. Med. Microbiol.", | |
month = aug, | |
year = 2016, | |
keywords = "Prophage; SCCmec; Staphylococcus aureus; Type III-A CRISPR-Cas | |
system;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Botelho2016-ed, | |
title = "Complete genome sequence of the {MRSA} isolate {HC1335} from | |
{ST239} lineage displaying a truncated {AgrC} histidine kinase | |
receptor", | |
author = "Botelho, Ana M N and Costa, Maiana O C and Beltrame, Cristiana O | |
and Ferreira, Fabienne A and Lima, Nicholas C B and Costa, Bruno | |
S S and de Morais, Guilherme L and Souza, Rangel C and Almeida, | |
Luiz G P and Vasconcelos, Ana T R and Nicol{\'a}s, Marisa F and | |
Figueiredo, Agnes M S", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) is still one | |
of the most important hospital pathogen globally. The | |
multiresistant isolates of the ST239-SCCmecIII lineage are spread | |
over large geographic regions, colonizing and infecting hospital | |
patients in virtually all continents. The balance between fitness | |
(adaptability) and virulence potential is likely to represent an | |
important issue in the clonal shift dynamics leading the success | |
of some specific MRSA clones over another. The accessory gene | |
regulator (agr) is the master quorum sensing system of | |
staphylococci playing a role in the global regulation of key | |
virulence factors. Consequently, agr inactivation in S. aureus | |
may represent a significant mechanism of genetic variability in | |
the adaptation of this healthcare-associated pathogen. We report | |
here the complete genome sequence of the methicillin-resistant S. | |
aureus, isolate HC1335, a variant of the ST239 lineage, which | |
presents a natural insertion of an IS256 transposase element in | |
the agrC gene encoding AgrC histidine kinase receptor.", | |
journal = "Genome Biol. Evol.", | |
month = sep, | |
year = 2016, | |
keywords = "IS256 insertion; ST239 lineage; agrC dysfunction; complete genome | |
sequence; methicillin-resistant Staphylococcus | |
aureus;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Harrison2016-eo, | |
title = "Transmission of methicillin-resistant Staphylococcus aureus in | |
long-term care facilities and their related healthcare networks", | |
author = "Harrison, Ewan M and Ludden, Catherine and Brodrick, Hayley J and | |
Blane, Beth and Brennan, Gr{\'a}inne and Morris, Dearbh{\'a}ile | |
and Coll, Francesc and Reuter, Sandra and Brown, Nicholas M and | |
Holmes, Mark A and O'Connell, Brian and Parkhill, Julian and | |
T{\"o}r{\"o}k, M Estee and Cormican, Martin and Peacock, Sharon J", | |
abstract = "BACKGROUND: Long-term care facilities (LTCF) are potential | |
reservoirs for methicillin-resistant Staphylococcus aureus | |
(MRSA), control of which may reduce MRSA transmission and | |
infection elsewhere in the healthcare system. Whole-genome | |
sequencing (WGS) has been used successfully to understand MRSA | |
epidemiology and transmission in hospitals and has the potential | |
to identify transmission between these and LTCF. METHODS: Two | |
prospective observational studies of MRSA carriage were conducted | |
in LTCF in England and Ireland. MRSA isolates were whole-genome | |
sequenced and analyzed using established methods. Genomic data | |
were available for MRSA isolated in the local healthcare systems | |
(isolates submitted by hospitals and general practitioners). | |
RESULTS: We sequenced a total of 181 MRSA isolates from the two | |
study sites. The majority of MRSA were multilocus sequence type | |
(ST)22. WGS identified one likely transmission event between | |
residents in the English LTCF and three putative transmission | |
events in the Irish LTCF. WGS also identified closely related | |
isolates present in colonized Irish residents and their immediate | |
environment. Based on phylogenetic reconstruction, closely | |
related MRSA clades were identified between the LTCF and their | |
healthcare referral network, together with putative MRSA | |
acquisition by LTCF residents during hospital admission. | |
CONCLUSIONS: These data confirm that MRSA is transmitted between | |
residents of LTCF and is both acquired and transmitted to others | |
in referral hospitals and beyond. Our data present compelling | |
evidence for the importance of environmental contamination in | |
MRSA transmission, reinforcing the importance of environmental | |
cleaning. The use of WGS in this study highlights the need to | |
consider infection control in hospitals and community healthcare | |
facilities as a continuum.", | |
journal = "Genome Med.", | |
volume = 8, | |
number = 1, | |
pages = "102", | |
month = oct, | |
year = 2016, | |
keywords = "Long-term care facilities; MRSA; Molecular epidemiology; Nursing | |
home; Staphylococcus aureus; Transmission; Whole-genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Van_Duijkeren2016-pd, | |
title = "Transmission of {MRSA} between humans and animals on duck and | |
turkey farms", | |
author = "van Duijkeren, E and Hengeveld, P and Zomer, T P and Landman, F | |
and Bosch, T and Haenen, A and van de Giessen, A", | |
abstract = "OBJECTIVES: The objectives of this study were to estimate the | |
prevalence of MRSA on duck and turkey farms, identify risk | |
factors for human carriage and study transmission between animals | |
and humans. METHODS: On 10 duck and 10 turkey farms, samples were | |
taken from animals, poultry houses, home residences and humans | |
and cultured using pre-enrichment and selective enrichment. MRSA | |
isolates were typed by spa typing and multiple-locus variable | |
number tandem repeat analysis (MLVA) typing. A subset of isolates | |
from animals and humans was investigated using whole-genome | |
mapping. RESULTS: MRSA was found on one duck farm and three | |
turkey farms. On duck farms, all humans were MRSA negative. On | |
turkey farms, 5 of 11 farmers, 2 of 32 family members and 15 of | |
49 samples from the home residences were MRSA positive. | |
Individuals with daily contact with turkeys were significantly | |
more often MRSA positive than individuals without daily contact. | |
All MRSA isolates belonged to livestock-associated MLVA complex | |
398, belonged to spa type t011, were negative for | |
Panton-Valentine leucocidin, were mecC negative and were mecA | |
positive. Whole-genome mapping proved a valuable tool to study | |
the transmission of livestock-associated MRSA and showed that on | |
two turkey farms the isolates from the animals and humans were | |
indistinguishable or closely related, indicating transmission. | |
CONCLUSIONS: MRSA carriage in individuals in daily contact with | |
turkeys was significantly higher than that in individuals only | |
living on the farms or than in the general Dutch population. | |
Therefore, persons with a high degree of contact with turkeys | |
have an increased risk of MRSA carriage, and we propose that they | |
should be screened prior to hospitalization in order to decrease | |
the risk of nosocomial transmission.", | |
journal = "J. Antimicrob. Chemother.", | |
volume = 71, | |
number = 1, | |
pages = "58--62", | |
month = jan, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Suhaili2016-gq, | |
title = "Draft genome sequence of Staphylococcus aureus {KT/312045}, an | |
{ST1-MSSA} {PVL} positive isolated from pus sample in East Coast | |
Malaysia", | |
author = "Suhaili, Zarizal and Lean, Soo-Sum and Mohamad, Noor Muzamil and | |
Rachman, Abdul R Abdul and Desa, Mohd Nasir Mohd and Yeo, Chew | |
Chieng", | |
abstract = "Most of the efforts in elucidating the molecular relatedness and | |
epidemiology of Staphylococcus aureus in Malaysia have been | |
largely focused on methicillin-resistant S. aureus (MRSA). | |
Therefore, here we report the draft genome sequence of the | |
methicillin-susceptible Staphylococcus aureus (MSSA) with | |
sequence type 1 (ST1), spa type t127 with Panton-Valentine | |
Leukocidin (pvl) pathogenic determinant isolated from pus sample | |
designated as KT/314250 strain. The size of the draft genome is | |
2.86 Mbp with 32.7\% of G + C content consisting 2673 coding | |
sequences. The draft genome sequence has been deposited in | |
DDBJ/EMBL/GenBank under the accession number AOCP00000000.", | |
journal = "Genom Data", | |
volume = 9, | |
pages = "111--112", | |
month = sep, | |
year = 2016, | |
keywords = "Methicillin-susceptible Staphylococcus aureus; Panton-Valentibe | |
Leukocidin; ST1-MSSA PVL positive Malaysia | |
isolates;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Azarian2016-bd, | |
title = "Genomic Epidemiology of {Methicillin-Resistant} Staphylococcus | |
aureus in a Neonatal Intensive Care Unit", | |
author = "Azarian, Taj and Maraqa, Nizar F and Cook, Robert L and Johnson, | |
Judith A and Bailey, Christine and Wheeler, Sarah and Nolan, | |
David and Rathore, Mobeen H and Morris, Jr, J Glenn and Salemi, | |
Marco", | |
abstract = "Despite infection prevention efforts, neonatal intensive care | |
unit (NICU) patients remain at risk of Methicillin-resistant | |
Staphylococcus aureus (MRSA) infection. Modes of transmission for | |
healthcare-associated (HA) and community-associated (CA) MRSA | |
remain poorly understood and may vary by genotype, hindering the | |
development of effective prevention and control strategies. From | |
2008-2010, all patients admitted to a level III NICU were | |
screened for MRSA colonization, and all available isolates were | |
spa-typed. Spa-type t008, the most prevalent CA- genotype in the | |
United States, spa-type t045, a HA- related genotype, and a | |
convenience sample of strains isolated from 2003-2011, underwent | |
whole-genome sequencing and phylodynamic analysis. Patient risk | |
factors were compared between colonized and noncolonized infants, | |
and virulence and resistance genes compared between spa-type t008 | |
and non-t008 strains. Epidemiological and genomic data were used | |
to estimate MRSA importations and acquisitions through | |
transmission reconstruction. MRSA colonization was identified in | |
9.1\% (177/1940) of hospitalized infants and associated with low | |
gestational age and birth weight. Among colonized infants, low | |
gestational age was more common among those colonized with t008 | |
strains. Our data suggest that approximately 70\% of | |
colonizations were the result of transmission events within the | |
NICU, with the remainder likely to reflect importations of | |
``outside'' strains. While risk of transmission within the NICU | |
was not affected by spa-type, patterns of acquisition and | |
importation differed between t008 and t045 strains. Phylodynamic | |
analysis showed the effective population size of spa-type t008 | |
has been exponentially increasing in both community and hospital, | |
with spa-type t008 strains possessed virulence genes not found | |
among t045 strains; t045 strains, in contrast, appeared to be of | |
more recent origin, with a possible hospital source. Our data | |
highlight the importance of both intra-NICU transmission and | |
recurrent introductions in maintenance of MRSA colonization | |
within the NICU environment, as well as spa-type-specific | |
differences in epidemiology.", | |
journal = "PLoS One", | |
volume = 11, | |
number = 10, | |
pages = "e0164397", | |
month = oct, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Wan2016-jx, | |
title = "Complete Circular Genome Sequence of Successful {ST8/SCCmecIV} | |
{Community-Associated} {Methicillin-Resistant} Staphylococcus | |
aureus ({OC8}) in Russia: {One-Megabase} Genomic Inversion, | |
{IS256's} Spread, and Evolution of Russia {ST8-IV}", | |
author = "Wan, Tsai-Wen and Khokhlova, Olga E and Iwao, Yasuhisa and | |
Higuchi, Wataru and Hung, Wei-Chun and Reva, Ivan V and Singur, | |
Olga A and Gostev, Vladimir V and Sidorenko, Sergey V and | |
Peryanova, Olga V and Salmina, Alla B and Reva, Galina V and | |
Teng, Lee-Jene and Yamamoto, Tatsuo", | |
abstract = "ST8/SCCmecIV community-associated methicillin-resistant | |
Staphylococcus aureus (CA-MRSA) has been a common threat, with | |
large USA300 epidemics in the United States. The global | |
geographical structure of ST8/SCCmecIV has not yet been fully | |
elucidated. We herein determined the complete circular genome | |
sequence of ST8/SCCmecIVc strain OC8 from Siberian Russia. We | |
found that 36.0\% of the genome was inverted relative to USA300. | |
Two IS256, oppositely oriented, at IS256-enriched hot spots were | |
implicated with the one-megabase genomic inversion (MbIN) and | |
vSa$\beta$ split. The behavior of IS256 was flexible: its | |
insertion site (att) sequences on the genome and junction | |
sequences of extrachromosomal circular DNA were all divergent, | |
albeit with fixed sizes. A similar multi-IS256 system was | |
detected, even in prevalent ST239 healthcare-associated MRSA in | |
Russia, suggesting IS256's strong transmission potential and | |
advantage in evolution. Regarding epidemiology, all ST8/SCCmecIVc | |
strains from European, Siberian, and Far Eastern Russia, examined | |
had MbIN, and geographical expansion accompanied divergent spa | |
types and resistance to fluoroquinolones, chloramphenicol, and | |
often rifampicin. Russia ST8/SCCmecIVc has been associated with | |
life-threatening infections such as pneumonia and sepsis in both | |
community and hospital settings. Regarding virulence, the OC8 | |
genome carried a series of toxin and immune evasion genes, a | |
truncated giant surface protein gene, and IS256 insertion | |
adjacent to a pan-regulatory gene. These results suggest that | |
unique single ST8/spa1(t008)/SCCmecIVc CA-MRSA (clade, Russia | |
ST8-IVc) emerged in Russia, and this was followed by large | |
geographical expansion, with MbIN as an epidemiological marker, | |
and fluoroquinolone resistance, multiple virulence factors, and | |
possibly a multi-IS256 system as selective advantages.", | |
journal = "PLoS One", | |
volume = 11, | |
number = 10, | |
pages = "e0164168", | |
month = oct, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers", | |
language = "en" | |
} | |
@ARTICLE{Bletz2015-ks, | |
title = "Ensuring backwards compatibility: traditional genotyping efforts | |
in the era of whole genome sequencing", | |
author = "Bletz, S and Mellmann, A and Rothg{\"a}nger, J and Harmsen, D", | |
abstract = "When using next-generation whole genome sequencing (WGS), | |
extraction of spa types from WGS data is essential for backwards | |
compatibility with Sanger sequencing-based spa typing of | |
methicillin-resistant Staphylococcus aureus (MRSA). We evaluated | |
WGS-based spa typing with a 2$\times$250 bp protocol in a diverse | |
collection of 423 MRSA isolates using two pipelines that executed | |
sequence quality-trimming and de novo assembly before spa typing. | |
The SeqSphere(+) pipeline correctly typed 419 isolates (99.1\%) | |
whereas the CLCbio pipeline succeeded in 249 isolates (58.9\%). | |
In summary, WGS combined with an optimized de novo assembly | |
enables nearly full compatibility with Sanger sequencing-based | |
spa typing data.", | |
journal = "Clin. Microbiol. Infect.", | |
volume = 21, | |
number = 4, | |
pages = "347.e1--4", | |
month = apr, | |
year = 2015, | |
keywords = "Backwards compatibility; Staphylococcus aureus; | |
methicillin-resistant Staphylococcus aureus; spa typing; whole | |
genome | |
sequencing;Staphylococcus;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Sarrou2016-yj, | |
title = "Characterization of a Novel lsa(E)- and {lnu(B)-Carrying} | |
Structure Located in the Chromosome of a Staphylococcus aureus | |
Sequence Type 398 Strain", | |
author = "Sarrou, S and Liakopoulos, A and Tsoumani, K and Sagri, E and | |
Mathiopoulos, K D and Tzouvelekis, L S and Miriagou, V and | |
Petinaki, E", | |
journal = "Antimicrob. Agents Chemother.", | |
volume = 60, | |
number = 2, | |
pages = "1164--1166", | |
month = feb, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Kaur2016-ib, | |
title = "Genome sequencing, assembly, annotation and analysis of | |
\textit{Staphylococcus xylosus} strain {DMB3-Bh1} reveals genes | |
responsible for pathogenicity", | |
author = "Kaur, Gurwinder and Arora, Amit and Sathyabama, Sathyaseelan and | |
Mubin, Nida and Verma, Sheenam and Mayilraj, Shanmugam and | |
Agrewala, Javed N", | |
abstract = "BACKGROUND: Staphylococcus xylosus is coagulase-negative | |
staphylococci (CNS), found occasionally on the skin of humans but | |
recurrently on other mammals. Recent reports suggest that this | |
commensal bacterium may cause diseases in humans and other | |
animals. In this study, we present the first report of whole | |
genome sequencing of S. xylosus strain DMB3-Bh1, which was | |
isolated from the stool of a mouse. RESULTS: The draft genome of | |
S. xylosus strain DMB3-Bh1 consisted of 2,81,0255 bp with G+C | |
content of 32.7 mol\%, 2623 predicted coding sequences (CDSs) and | |
58 RNAs. The final assembly contained 12 contigs of total size | |
2,81,0255 bp with N50 contig length of 4,37,962 bp and the | |
largest contig assembled measured 7,61,338 bp. Further, an | |
interspecies comparative genomic analysis through rapid | |
annotation using subsystem technology server was achieved with | |
Staphylococcus aureus RF122 that revealed 36 genes having | |
similarity with S. xylosus DMB3-Bh1. 35 genes encoded for | |
virulence, disease and defense and 1 gene encoded for phages, | |
prophages and transposable elements. CONCLUSIONS: These results | |
suggest co linearity in genes between S. xylosus DMB3-Bh1 and S. | |
aureus RF122 that contribute to pathogenicity and might be the | |
result of horizontal gene transfer. The study indicates that S. | |
xylosus DMB3-Bh1 may be a potential emerging pathogen for | |
rodents.", | |
journal = "Gut Pathog.", | |
volume = 8, | |
pages = "55", | |
month = nov, | |
year = 2016, | |
keywords = "CDSs; Coagulase-negative staphylococci; Disease and defense; | |
EzTaxon; Rapid annotation using subsystem technology (RAST); | |
Staphylococcus aureus; | |
Virulence;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Price2016-vt, | |
title = "Transmission of Staphylococcus aureus between healthcare | |
workers, the environment and patients in an intensive care unit: | |
a whole-genome sequencing based longitudinal cohort study", | |
author = "Price, J R and Cole, K and Bexley, A and Kostiou, V and Eyre, D | |
W and Golubchik, T and Wilson, D and Crook, D W and Walker, A S | |
and Peto, T and Llewelyn, M and Paul, J", | |
abstract = "UCL Discovery is UCL's open access repository, showcasing and | |
providing access to UCL research outputs from all UCL | |
disciplines.", | |
journal = "Lancet Infect. Dis.", | |
publisher = "Elsevier", | |
month = dec, | |
year = 2016, | |
keywords = "Staphylococcus;bacterial comparative | |
genomics;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Singh2016-ge, | |
title = "In Vitro Tolerance of A {Drug-Na{\"\i}ve} Staphylococcus aureus | |
{FDA209P} towards Vancomycin", | |
author = "Singh, Madhuri and Matsuo, Miki and Sasaki, Takashi and Morimoto, | |
Yuh and Hishinuma, Tomomi and Hiramatsu, Keiichi", | |
abstract = "The mechanisms underlying bacterial tolerance to antibiotics are | |
unclear. A possible adaptation strategy has now been explored by | |
exposure of drug-na{\"\i}ve methicillin-susceptible | |
Staphylococcus aureus strain FDA209P to vancomycin in vitro | |
Vancomycin-survivor strains, which appeared after 96 h of | |
exposure, were slow-growing derivatives of the parent strain. | |
Although the vancomycin minimum-inhibitory concentrations (MICs) | |
with the survivor strains were within the susceptible range, the | |
cytokilling effects of vancomycin at 20-fold MIC were | |
significantly lower than those in the parent strain. Whole-genome | |
sequencing demonstrated that ileS, encoding isoleucyl-tRNA | |
synthetase (IleRS), was mutated in two of the three | |
vancomycin-survivor strains. The IleRS Y723H mutation is located | |
close to the isoleucyl-tRNA contact site, and potentially affects | |
the binding affinity of IleRS for isoleucyl-tRNA, thereby | |
inhibiting protein synthesis and leading to vancomycin tolerance. | |
Introduction of the mutation encoding IleRS Y723H into FDA209P by | |
allelic replacement successfully transferred the | |
vancomycin-tolerance phenotype. We have identified mutation of | |
ileS as one of the bona fide genetic events leading to the | |
acquisition of vancomycin tolerance in S. aureus, potentially | |
acting via inhibition of the function of IleRS.", | |
journal = "Antimicrob. Agents Chemother.", | |
month = nov, | |
year = 2016, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Sharma2016-bu, | |
title = "{Livestock-Associated} Methicillin Resistant | |
\textit{Staphylococcus aureus} ({LA-MRSA}) Clonal Complex ({CC}) | |
398 Isolated from {UK} Animals belong to European Lineages", | |
author = "Sharma, Meenaxi and Nunez-Garcia, Javier and Kearns, Angela M and | |
Doumith, Michel and Butaye, Patrick R and Argud{\'\i}n, M Angeles | |
and Lahuerta-Marin, Angela and Pichon, Bruno and AbuOun, Manal | |
and Rogers, Jon and Ellis, Richard J and Teale, Christopher and | |
Anjum, Muna F", | |
abstract = "In recent years, there has been an increase in the number of | |
livestock-associated methicillin resistant Staphylococcus aureus | |
(LA-MRSA) clonal complex (CC) 398 recovered from S. aureus | |
isolated animals in the UK. To determine possible origins of 12 | |
LA-MRSA CC398 isolates collected after screening more than a | |
thousand S. aureus animal isolates from the UK between 2013 and | |
2015, whole genome sequences (WGS) of CC398 European, including | |
UK, and non-European isolates from diverse animal hosts were | |
compared. Phylogenetic reconstruction applied to WGS data to | |
assess genetic relatedness of all 89 isolates, clustered the 12 | |
UK CC398 LA-MRSA within the European sub-lineages, although on | |
different nodes; implicating multiple independent incursions into | |
the UK, as opposed to a single introduction followed by clonal | |
expansion. Three UK isolates from healthy pigs and one from | |
turkey clustered within the cassette chromosome recombinases ccr | |
C S. aureus protein A (spa)-type t011 European sub-lineage and | |
three UK isolates from horses within the ccrA2B2 t011 European | |
sub-lineage. The remaining UK isolates, mostly from pigs, | |
clustered within the t034 European lineage. Presence of | |
virulence, antimicrobial (AMR), heavy metal (HMR), and | |
disinfectant (DR) resistance genes were determined using an | |
in-house pipeline. Most, including UK isolates, harbored | |
resistance genes to $\geq$3 antimicrobial classes in addition to | |
$\beta$-lactams. HMR genes were detected in most European ccrC | |
positive isolates, with >80\% harboring czrC, encoding zinc and | |
cadmium resistance; in contrast ~60\% ccrC isolates within | |
non-European lineages and 6\% ccrA2B2 isolates showed this | |
characteristic. The UK turkey MRSA isolate did not harbor | |
$\varphi$AV$\beta$ avian prophage genes (SAAV\_2008 and | |
SAAV\_2009) present in US MSSA isolates from turkey and pigs. | |
Absence of some of the major human-associated MRSA toxigenic and | |
virulence genes in the UK LA-MRSA animal isolates was not | |
unexpected. Therefore, we can conclude that the 12 UK LA-MRSA | |
isolates collected in the past 2 years most likely represent | |
separate incursions into the UK from other European countries. | |
The presence of zinc and cadmium resistance in all nine food | |
animal isolates (pig and poultry), which was absent from the 3 | |
horse isolates may suggest heavy metal use/exposure has a | |
possible role in selection of some MRSA.", | |
journal = "Front. Microbiol.", | |
volume = 7, | |
pages = "1741", | |
month = nov, | |
year = 2016, | |
keywords = "UK CC398s; avian prophage genes; methicillin resistant | |
Staphylococcus aureus in animals; whole genome sequencing; zinc | |
and cadmium | |
resistance;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Meumann2016-kp, | |
title = "Whole genome sequencing to investigate a putative outbreak of | |
the virulent community-associated methicillin-resistant | |
Staphylococcus aureus {ST93} clone in a remote Indigenous | |
community", | |
author = "Meumann, Ella M and Andersson, Patiyan and Yeaman, Fiona and | |
Oldfield, Sarah and Lilliebridge, Rachael and Bentley, Stephen D | |
and Krause, Vicki and Beaman, Miles and Currie, Bart J and Holt, | |
Deborah C and Giffard, Philip M and Tong, Steven Y C", | |
abstract = "We report two cases of severe pneumonia due to ST93 MRSA | |
presenting from a remote Australian Indigenous community within | |
a 2-week period, and utilised whole genome sequences to | |
determine whether these were part of an outbreak. S. aureus was | |
isolated from 12 of 92 nasal swabs collected from 25 community | |
households (including the two index households); 1 isolate was | |
ST93. Three of 5 skin lesion S. aureus isolates obtained at the | |
community were ST93. Whole genome sequencing of the ST93 | |
isolates from this study and a further 20 ST93 isolates from the | |
same region suggested that recent transmission and progression | |
to disease had not taken place. The proximity in time and space | |
of the 2 severe pneumonia cases is likely a reflection of the | |
high burden of disease due to ST93 MRSA in this population where | |
skin infections and household crowding are common.", | |
journal = "Microbial Genomics", | |
publisher = "Microbiology Society", | |
month = nov, | |
year = 2016, | |
keywords = "colonization;Staphylococcus;bacterial comparative | |
genomics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Sabat2017-po, | |
title = "Complete-genome sequencing elucidates outbreak dynamics of | |
{CA-MRSA} {USA300} ({ST8-spa} t008) in an academic hospital of | |
Paramaribo, Republic of Suriname", | |
author = "Sabat, Artur J and Hermelijn, Sandra M and Akkerboom, Viktoria | |
and Juliana, Amadu and Degener, John E and Grundmann, Hajo and | |
Friedrich, Alexander W", | |
abstract = "We report the investigation of an outbreak situation of | |
methicillin-resistant Staphylococcus aureus (MRSA) that occurred | |
at the Academic Hospital Paramaribo (AZP) in the Republic of | |
Suriname from April to May 2013. We performed whole genome | |
sequencing with complete gap closure for chromosomes and plasmids | |
on all isolates. The outbreak involved 12 patients and 1 | |
healthcare worker/nurse at the AZP. In total 24 isolates were | |
investigated. spa typing, genome-wide single nucleotide | |
polymorphism (SNP) analysis, ad hoc whole genome multilocus | |
sequence typing (wgMLST), stable core genome MLST (cgMLST) and in | |
silico PFGE were used to determine phylogenetic relatedness and | |
to identify transmission. Whole-genome sequencing (WGS) showed | |
that all isolates were members of genomic variants of the North | |
American USA300 clone. However, WGS revealed a heterogeneous | |
population structure of USA300 circulating at the AZP. We | |
observed up to 8 SNPs or up to 5 alleles of difference by wgMLST | |
when the isolates were recovered from different body sites of the | |
same patient or if direct transmission between patients was most | |
likely. This work describes the usefulness of complete genome | |
sequencing of bacterial chromosomes and plasmids providing an | |
unprecedented level of detail during outbreak investigations not | |
being visible by using conventional typing methods.", | |
journal = "Sci. Rep.", | |
volume = 7, | |
pages = "41050", | |
month = jan, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Highlander2007-ff, | |
title = "Subtle genetic changes enhance virulence of methicillin resistant | |
and sensitive Staphylococcus aureus", | |
author = "Highlander, Sarah K and Hult{\'e}n, Kristina G and Qin, Xiang and | |
Jiang, Huaiyang and Yerrapragada, Shailaja and Mason, Jr, Edward | |
O and Shang, Yue and Williams, Tiffany M and Fortunov, R{\'e}gine | |
M and Liu, Yamei and Igboeli, Okezie and Petrosino, Joseph and | |
Tirumalai, Madhan and Uzman, Akif and Fox, George E and Cardenas, | |
Ana Maria and Muzny, Donna M and Hemphill, Lisa and Ding, Yan and | |
Dugan, Shannon and Blyth, Peter R and Buhay, Christian J and | |
Dinh, Huyen H and Hawes, Alicia C and Holder, Michael and Kovar, | |
Christie L and Lee, Sandra L and Liu, Wen and Nazareth, Lynne V | |
and Wang, Qiaoyan and Zhou, Jianling and Kaplan, Sheldon L and | |
Weinstock, George M", | |
abstract = "BACKGROUND: Community acquired (CA) methicillin-resistant | |
Staphylococcus aureus (MRSA) increasingly causes disease | |
worldwide. USA300 has emerged as the predominant clone causing | |
superficial and invasive infections in children and adults in the | |
USA. Epidemiological studies suggest that USA300 is more virulent | |
than other CA-MRSA. The genetic determinants that render | |
virulence and dominance to USA300 remain unclear. RESULTS: We | |
sequenced the genomes of two pediatric USA300 isolates: one | |
CA-MRSA and one CA-methicillin susceptible (MSSA), isolated at | |
Texas Children's Hospital in Houston. DNA sequencing was | |
performed by Sanger dideoxy whole genome shotgun (WGS) and 454 | |
Life Sciences pyrosequencing strategies. The sequence of the | |
USA300 MRSA strain was rigorously annotated. In USA300-MRSA 2658 | |
chromosomal open reading frames were predicted and 3.1 and 27 | |
kilobase (kb) plasmids were identified. USA300-MSSA contained a | |
20 kb plasmid with some homology to the 27 kb plasmid found in | |
USA300-MRSA. Two regions found in US300-MRSA were absent in | |
USA300-MSSA. One of these carried the arginine deiminase operon | |
that appears to have been acquired from S. epidermidis. The | |
USA300 sequence was aligned with other sequenced S. aureus | |
genomes and regions unique to USA300 MRSA were identified. | |
CONCLUSION: USA300-MRSA is highly similar to other MRSA strains | |
based on whole genome alignments and gene content, indicating | |
that the differences in pathogenesis are due to subtle changes | |
rather than to large-scale acquisition of virulence factor genes. | |
The USA300 Houston isolate differs from another sequenced USA300 | |
strain isolate, derived from a patient in San Francisco, in | |
plasmid content and a number of sequence polymorphisms. Such | |
differences will provide new insights into the evolution of | |
pathogens.", | |
journal = "BMC Microbiol.", | |
volume = 7, | |
pages = "99", | |
month = nov, | |
year = 2007, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Galata2016-sq, | |
title = "Comparing genome versus proteome-based identification of clinical | |
bacterial isolates", | |
author = "Galata, Valentina and Backes, Christina and Laczny, C{\'e}dric | |
Christian and Hemmrich-Stanisak, Georg and Li, Howard and Smoot, | |
Laura and Posch, Andreas Emanuel and Schmolke, Susanne and | |
Bischoff, Markus and von M{\"u}ller, Lutz and Plum, Achim and | |
Franke, Andre and Keller, Andreas", | |
abstract = "Whole-genome sequencing (WGS) is gaining importance in the | |
analysis of bacterial cultures derived from patients with | |
infectious diseases. Existing computational tools for WGS-based | |
identification have, however, been evaluated on previously | |
defined data relying thereby unwarily on the available taxonomic | |
information.Here, we newly sequenced 846 clinical gram-negative | |
bacterial isolates representing multiple distinct genera and | |
compared the performance of five tools (CLARK, Kaiju, Kraken, | |
DIAMOND/MEGAN and TUIT). To establish a faithful 'gold standard', | |
the expert-driven taxonomy was compared with identifications | |
based on matrix-assisted laser desorption/ionization | |
time-of-flight (MALDI-TOF) mass spectrometry (MS) analysis. | |
Additionally, the tools were also evaluated using a data set of | |
200 Staphylococcus aureus isolates.CLARK and Kraken (with k =31) | |
performed best with 626 (100\%) and 193 (99.5\%) correct species | |
classifications for the gram-negative and S aureus isolates, | |
respectively. Moreover, CLARK and Kraken demonstrated highest | |
mean F-measure values (85.5/87.9\% and 94.4/94.7\% for the two | |
data sets, respectively) in comparison with DIAMOND/MEGAN (71 and | |
85.3\%), Kaiju (41.8 and 18.9\%) and TUIT (34.5 and 86.5\%). | |
Finally, CLARK, Kaiju and Kraken outperformed the other tools by | |
a factor of 30 to 170 fold in terms of runtime.We conclude that | |
the application of nucleotide-based tools using k-mers-e.g. CLARK | |
or Kraken-allows for accurate and fast taxonomic characterization | |
of bacterial isolates from WGS data. Hence, our results suggest | |
WGS-based genotyping to be a promising alternative to the | |
MS-based biotyping in clinical settings. Moreover, we suggest | |
that complementary information should be used for the evaluation | |
of taxonomic classification tools, as public databases may suffer | |
from suboptimal annotations.", | |
journal = "Brief. Bioinform.", | |
month = dec, | |
year = 2016, | |
keywords = "MALDI-TOF MS; bacteria; taxonomy; whole-genome next-generation | |
sequencing;Staphylococcus;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Bakthavatchalam2016-zq, | |
title = "Novel Observations in 11 Heteroresistant | |
{Vancomycin-Intermediate} {Methicillin-Resistant} Staphylococcus | |
aureus Strains from South India", | |
author = "Bakthavatchalam, Yamuna Devi and Veeraraghavan, Balaji and Peter, | |
John Victor and Rajinikanth, Janakiraman and Inbanathan, Francis | |
Yesurajan and Devanga Ragupathi, Naveen Kumar and Rajamani Sekar, | |
Suresh Kumar", | |
abstract = "We report here the draft genome sequences of 11 heteroresistant | |
vancomycin-intermediate Staphylococcus aureus (hVISA) strains | |
from bloodstream infection. All strains harbor mutations in | |
vraSR, graSR, walKR, and/or tcaRAB and are often implicated as | |
the frequently mutated candidate genes in hVISA phenotypes.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 6, | |
month = dec, | |
year = 2016, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Berenger2016-gq, | |
title = "Draft {Whole-Genome} Sequence of a {Catalase-Negative} | |
Staphylococcus aureus subsp. aureus (Sequence Type 25) Strain | |
Isolated from a Patient with Endocarditis and Septic Arthritis", | |
author = "Berenger, Byron and Chen, Justin and Bernier, Anne-Marie and | |
Bernard, Kathryn", | |
abstract = "Staphylococcus aureus strains without catalase activity are rare, | |
challenging to identify with conventional biochemical methods, | |
and, despite a supposed decreased pathogenicity, can still cause | |
disease. The first whole-genome sequence of a catalase-negative | |
S. aureus isolate causing severe recurrent invasive infection | |
with two novel missense mutations in the katA gene is reported | |
here.", | |
journal = "Genome Announc.", | |
volume = 4, | |
number = 6, | |
month = dec, | |
year = 2016, | |
keywords = "Staphylococcus;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Peng2016-ga, | |
title = "{WalK(S221P)}, a naturally occurring mutation, confers vancomycin | |
resistance in {VISA} strain {XN108}", | |
author = "Peng, Huagang and Hu, Qiwen and Shang, Weilong and Yuan, Jizhen | |
and Zhang, Xiaopeng and Liu, Hui and Zheng, Ying and Hu, Zhen and | |
Yang, Yi and Tan, Li and Li, Shu and Hu, Xiaomei and Li, Ming and | |
Rao, Xiancai", | |
abstract = "OBJECTIVES: Vancomycin-intermediate Staphylococcus aureus (VISA) | |
strains have spread globally. We previously isolated an ST239 | |
VISA (XN108) with a vancomycin MIC of 12 mg/L. The mechanism for | |
XN108 resistance to vancomycin was investigated in this study. | |
METHODS: Genome comparison was performed to characterize | |
mutations that might contribute to the XN108 resistance | |
phenotype. The novel mutation WalK(S221P) was identified and | |
investigated using allelic replacement experiments. Vancomycin | |
susceptibilities, autolytic activities and morphologies of the | |
strains were examined. Autophosphorylation activities of WalK and | |
the WalK(S221P) mutant were determined in vitro with | |
[$\lambda$-(32)P]ATP, and binding activity of | |
WalK(S221P)-activated WalR to the promoter region of its target | |
gene lytM was determined by electrophoretic mobility shift assay. | |
RESULTS: Genome comparison revealed three mutations, GraS(T136I), | |
RpoB(H481N) and WalK(S221P), which might be responsible for | |
vancomycin resistance in XN108. The introduction of WalK(S221P) | |
to the vancomycin-susceptible strain N315 increased its | |
vancomycin MIC from 1.5 to 8 mg/L, whereas the allelic | |
replacement of WalK(S221P) with the native N315 WalK allele in | |
XN108 decreased its vancomycin MIC from 12 to 4 mg/L. The VISA | |
strains have thickened cell walls and decreased autolysis, | |
consistent with observed changes in the expression of genes | |
involved in cell wall metabolism and virulence regulation. | |
WalK(S221P) exhibited reduced autophosphorylation, which may lead | |
to reduced phosphorylation of WalR. WalK(S221P)-phosphorylated | |
WalR also exhibited a reduced capacity to bind to the lytM | |
promoter. CONCLUSIONS: The naturally occurring WalK(S221P) | |
mutation plays a key role in vancomycin resistance in XN108.", | |
journal = "J. Antimicrob. Chemother.", | |
month = dec, | |
year = 2016, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Mellmann2017-go, | |
title = "High Interlaboratory Reproducibility and Accuracy of | |
{Next-Generation} {Sequencing-Based} Bacterial Genotyping in a | |
{Ring-Trial}", | |
author = "Mellmann, Alexander and Andersen, Paal Skytt and Bletz, Stefan | |
and Friedrich, Alexander W and Kohl, Thomas A and Lilje, Berit | |
and Niemann, Stefan and Prior, Karola and Rossen, John W and | |
Harmsen, Dag", | |
abstract = "Today, next-generation whole genome sequencing (WGS) technology | |
is increasingly used to determine the genetic relationship of | |
bacteria on a nearly whole genome level for infection control | |
purposes and molecular surveillance. Here, we conducted a | |
multicenter ring-trial comprising five laboratories to determine | |
the reproducibility and accuracy of WGS-based typing.The | |
participating laboratories sequenced 20 blind-coded | |
Staphylococcus aureus DNA samples using the 250 bp paired-end | |
chemistry for library preparation in a single sequencing run on | |
an Illumina Miseq sequencer. Run acceptance criteria were | |
sequencing outputs > 5.6 Gb and Q30 read quality scores > 75\%. | |
Subsequently, spa typing, multilocus sequence typing (MLST), | |
ribosomal MLST, and core genome (cg)MLST were performed by the | |
participants. Moreover, cgMLST target sequence discrepancies in | |
comparison to the also included and published sequence of quality | |
control strain ATCC25923 were resolved using Sanger | |
sequencing.All five laboratories fulfilled the run acceptance | |
criteria in a single sequencing run without any repetition. Of | |
the in total 400 possible typing results, 394 reported spa types, | |
sequence types (ST), rSTs, and cgMLST cluster types were correct | |
and identical among all laboratories; only six typing results | |
were missing. Analysis of cgMLST allelic profiles corroborated | |
this high reproducibility: only 3 (0.0016\%) of in total 183,927 | |
cgMLST allele calls were wrong. Sanger sequencing confirmed all | |
the 12 discrepancies of the ring-trial results in comparison to | |
the published sequence of ATCC25923.In summary, this ring-trial | |
demonstrated the high reproducibility and accuracy of current | |
NGS-based bacterial typings for molecular surveillance when done | |
with nearly completely locked down methods.", | |
journal = "J. Clin. Microbiol.", | |
month = jan, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Kawada-Matsuo2016-tq, | |
title = "{C55} bacteriocin produced by {ETB-plasmid} positive | |
Staphylococcus aureus strains is a key factor for competition | |
with S. aureus strains", | |
author = "Kawada-Matsuo, Miki and Shammi, Fariha and Oogai, Yuichi and | |
Nakamura, Norifumi and Sugai, Motoyuki and Komatsuzawa, Hitoshi", | |
abstract = "Exfoliative toxin (ET) produced by Staphylococcus aureus is | |
closely associated with the onset of bullous impetigo. To date, | |
three ETs (ETA, ETB and ETD) have been identified. The gene | |
encoding ETB is located in a plasmid designated pETB. Bacteriocin | |
synthesis genes are also located in this plasmid and | |
pETB-positive strains reportedly produce the C55 bacteriocin. In | |
this study, the antibacterial activity against S. aureus strains | |
of the bacteriocin produced by the pETB-positive strain TY4 was | |
investigated. This bacteriocin demonstrated antibacterial | |
activity against all pETB-negative but not pETB-positive strains, | |
including TY4. Additionally, a TY4- strain from which the pETB | |
plasmid had been deleted exhibited susceptibility to the | |
bacteriocin. Further experiments revealed that two immunity | |
factors (orf 46-47 and orf 48) downstream of the bacteriocin | |
synthesis genes in the pETB plasmid are associated with immunity | |
against the bacteriocin produced by TY4. The TY4- with orf46-47 | |
strain exhibited complete resistance to bacteriocin, whereas the | |
TY4- with orf48 strain exhibited partial resistance. Whether | |
bacteriocin affects the proportion of each strain when | |
co-cultured with S. aureus strains was also investigated. When | |
TY4 or TY4- was co-cultured with 209P strain, which is | |
susceptible to the bacteriocin, the proportion of 209P | |
co-cultured with TY4 was significantly less than when 209P was | |
co-cultured with TY4-, whereas the proportion of TY4- with | |
orf46-48 co-cultured with TY4 was greater than with TY4-. These | |
results suggest that the C55 bacteriocin produced by | |
pETB-positive strains affects the proportion of each strain when | |
pETB-positive and -negative strains co-exist.", | |
journal = "Microbiol. Immunol.", | |
volume = 60, | |
number = 3, | |
pages = "139--147", | |
month = mar, | |
year = 2016, | |
keywords = "Staphylococcus aureus; bacteriocin; lantibiotics; two-component | |
system;Staphylococcus;Staph\_evolutionary\_history;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Stevens2017-ei, | |
title = "Draft Genome Sequence of Staphylococcus aureus 1608, a Strain | |
That Caused Toxic Mastitis in Twin Cows", | |
author = "Stevens, Marc J A and Stephan, Roger and Johler, Sophia", | |
abstract = "Staphylococcus aureus 1608 is a strain that caused a lethal | |
mastitis in cows. Here, the draft genome sequence of the strain | |
is presented.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 1, | |
month = jan, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Milheirico2017-lu, | |
title = "{MRSA} strains carrying the novel mecC gene: full genome | |
sequencing identifies in the genetic background several | |
determinants that modulate the resistant phenotype", | |
author = "Milheiri{\c c}o, Catarina and de Lencastre, Herm{\'\i}nia and | |
Tomasz, Alexander", | |
abstract = "Most MRSA strains are resistant to beta-lactam antibiotics due to | |
the presence of the mecA gene encoding an extra | |
penicillin-binding protein (PBP2A) that has low affinity to | |
virtually all beta-lactam antibiotics. Recently, a new resistance | |
determinant - the mecC gene - was identified in S. aureus | |
isolates recovered from humans and dairy cattle. Although having | |
typically low MICs to beta-lactam antibiotics, MRSA strains with | |
the mecC determinant are also capable of expressing high levels | |
of oxacillin resistance when in an optimal genetic background. In | |
order to test the impact of extensive beta-lactam selection on | |
the emergence of mecC carrying strains with high levels of | |
antibiotic resistance, we exposed the prototype mecC MRSA strain | |
LGA251 to increasing concentrations of oxacillin. LGA251 was able | |
to rapidly adapt to high concentrations of oxacillin in the | |
growth medium. In such laboratory mutants with increased levels | |
of oxacillin resistance, we identified mutations in genes with no | |
relationship to the mecC regulatory system indicating that the | |
genetic background plays an important role in the establishment | |
of the levels of oxacillin resistance. Our data also indicate | |
that the stringent stress response plays a critical role in the | |
beta-lactam antibiotic resistant phenotype of MRSA strains | |
carrying the mecC determinant.", | |
journal = "Antimicrob. Agents Chemother.", | |
month = jan, | |
year = 2017, | |
keywords = "oxacillin;Staphylococcus;antibiotics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Kennedy2010-fe, | |
title = "Complete nucleotide sequence analysis of plasmids in strains of | |
Staphylococcus aureus clone {USA300} reveals a high level of | |
identity among isolates with closely related core genome | |
sequences", | |
author = "Kennedy, Adam D and Porcella, Stephen F and Martens, Craig and | |
Whitney, Adeline R and Braughton, Kevin R and Chen, Liang and | |
Craig, Carly T and Tenover, Fred C and Kreiswirth, Barry N and | |
Musser, James M and DeLeo, Frank R", | |
abstract = "A community-associated methicillin-resistant Staphylococcus | |
aureus (CA-MRSA) strain known as pulsed-field type USA300 | |
(USA300) is epidemic in the United States. Previous comparative | |
whole-genome sequencing studies demonstrated that there has been | |
recent clonal emergence of a subset of USA300 isolates, which | |
comprise the epidemic clone. Although the core genomes of these | |
isolates are closely related, the level of diversity among USA300 | |
plasmids was not resolved. Inasmuch as these plasmids might | |
contribute to significant gene diversity among otherwise closely | |
related USA300 isolates, we performed de novo sequencing of | |
endogenous plasmids from 10 previously characterized USA300 | |
clinical isolates obtained from different geographic locations in | |
the United States. All isolates tested contained small (2- to | |
3-kb) and/or large (27- to 30-kb) plasmids. The large plasmids | |
encoded heavy metal and/or antimicrobial resistance elements, | |
including those that confer resistance to cadmium, bacitracin, | |
macrolides, penicillin, kanamycin, and streptothricin, although | |
all isolates were sensitive to minocycline, doxycycline, | |
trimethoprim-sulfamethoxazole, vancomycin, teicoplanin, and | |
linezolid. One of the USA300 isolates contained an archaic | |
plasmid that encoded staphylococcal enterotoxins R, J, and P. | |
Notably, the large plasmids (27 to 28 kb) from 8 USA300 | |
isolates--those that comprise the epidemic USA300 clone--were | |
virtually identical (99\% identity) and similar to a large | |
plasmid from strain USA300\_TCH1516 (a previously sequenced | |
USA300 strain from Houston, TX). These plasmids are largely | |
divergent from the 37-kb plasmid of FPR3757, the first sequenced | |
USA300 strain. The high level of plasmid sequence identity among | |
the majority of closely related USA300 isolates is consistent | |
with the recent clonal emergence hypothesis for USA300.", | |
journal = "J. Clin. Microbiol.", | |
volume = 48, | |
number = 12, | |
pages = "4504--4511", | |
month = dec, | |
year = 2010, | |
keywords = "plasmid;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Lima2017-ib, | |
title = "Genomic information on multidrug-resistant livestock-associated | |
methicillin-resistant Staphylococcus aureus {ST398} isolated from | |
a Brazilian patient with cystic fibrosis", | |
author = "Lima, Danielle F and Cohen, Renata Wf and Rocha, G{\'e}ssica A | |
and Albano, Rodolpho M and Marques, Elizabeth A and Le{\~a}o, | |
Robson S", | |
abstract = "Alarmingly, the isolation of methicillin-resistant Staphylococcus | |
aureus (MRSA) has been increasing among patients with cystic | |
fibrosis (CF). During a previous molecular characterisation of | |
MRSA isolates obtained from patients with CF from Rio de Janeiro, | |
Brazil, one isolate was identified as the ST398 clone, a | |
livestock-associated (LA) MRSA. In this study, we report the | |
draft genome sequence of an LA-MRSA ST398 clone isolated from a | |
patient with CF.", | |
journal = "Mem. Inst. Oswaldo Cruz", | |
volume = 112, | |
number = 1, | |
pages = "79--80", | |
month = jan, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Vestergaard2016-vt, | |
title = "{Genome-Wide} Identification of Antimicrobial Intrinsic | |
Resistance Determinants in \textit{Staphylococcus aureus}", | |
author = "Vestergaard, Martin and Leng, Bingfeng and Haaber, Jakob and | |
Bojer, Martin S and Vegge, Christina S and Ingmer, Hanne", | |
abstract = "The emergence of antimicrobial resistance severely threatens our | |
ability to treat bacterial infections. While acquired resistance | |
has received considerable attention, relatively little is known | |
of intrinsic resistance that allows bacteria to naturally | |
withstand antimicrobials. Gene products that confer intrinsic | |
resistance to antimicrobial agents may be explored for | |
alternative antimicrobial therapies, by potentiating the efficacy | |
of existing antimicrobials. In this study, we identified the | |
intrinsic resistome to a broad spectrum of antimicrobials in the | |
human pathogen, Staphylococcus aureus. We screened the Nebraska | |
Transposon Mutant Library of 1920 single-gene inactivations in S. | |
aureus strain JE2, for increased susceptibility to the | |
anti-staphylococcal antimicrobials (ciprofloxacin, oxacillin, | |
linezolid, fosfomycin, daptomycin, mupirocin, vancomycin, and | |
gentamicin). Sixty-eight mutants were confirmed by E-test to | |
display at least twofold increased susceptibility to one or more | |
antimicrobial agents. The majority of the identified genes have | |
not previously been associated with antimicrobial susceptibility | |
in S. aureus. For example, inactivation of genes encoding for | |
subunits of the ATP synthase, atpA, atpB, atpG and atpH, reduced | |
the minimum inhibitory concentration (MIC) of gentamicin 16-fold. | |
To elucidate the potential of the screen, we examined treatment | |
efficacy in the Galleria mellonella infection model. Gentamicin | |
efficacy was significantly improved, when treating larvae | |
infected with the atpA mutant compared to wild type cells with | |
gentamicin at a clinically relevant concentration. Our results | |
demonstrate that many gene products contribute to the intrinsic | |
antimicrobial resistance of S. aureus. Knowledge of these | |
intrinsic resistance determinants provides alternative targets | |
for compounds that may potentiate the efficacy of existing | |
antimicrobial agents against this important pathogen.", | |
journal = "Front. Microbiol.", | |
volume = 7, | |
pages = "2018", | |
month = dec, | |
year = 2016, | |
keywords = "Galleria mellonella; Staphylococcus aureus; antimicrobial agents; | |
intrinsic resistance; potentiator | |
targets;Staphylococcus;antibiotics;confirmed\_SA-resistance-mutation;oxacillin;Staphylococcus\_aureus\_genome\_papers;Michael\_F\_papers", | |
language = "en" | |
} | |
@ARTICLE{Capra2017-co, | |
title = "Genomic and transcriptomic comparison between Staphylococcus | |
aureus strains associated with high and low within herd | |
prevalence of intra-mammary infection", | |
author = "Capra, E and Cremonesi, P and Pietrelli, A and Puccio, S and | |
Luini, M and Stella, A and Castiglioni, B", | |
abstract = "BACKGROUND: Staphylococcus aureus (Staph. aureus) is one of the | |
major pathogens causing mastitis in dairy ruminants worldwide. | |
The chronic nature of Staph. aureus infection enhances the | |
contagiousness risk and diffusion in herds. In order to identify | |
the factors involved in intra-mammary infection (IMI) and | |
diffusion in dairy cows, we investigated the molecular | |
characteristics of two groups of Staph. aureus strains belonging | |
to ST8 and ST398, differing in clinical properties, through | |
comparison of whole genome and whole transcriptome sequencing. | |
RESULTS: The two groups of strains, one originated from high IMI | |
prevalence herds and the other from low IMI prevalence herds, | |
present a peculiar set of genes and polymorphisms related to | |
phenotypic features, such as bacterial invasion of mammary | |
epithelial cells and host adaptation. Transcriptomic analysis | |
supports the high propensity of ST8 strain to chronicity of | |
infection and to a higher potential cytotoxicity. CONCLUSIONS: | |
Our data are consistent with the invasiveness and host adaptation | |
feature for the strains GTB/ST8 associated to high within-herd | |
prevalence of mastitis. Variation in genes coding for surface | |
exposed proteins and those associated to virulence and defence | |
could constitute good targets for further research.", | |
journal = "BMC Microbiol.", | |
volume = 17, | |
number = 1, | |
pages = "21", | |
month = jan, | |
year = 2017, | |
keywords = "Genome; Mastitis; Next generation sequencing; Staphylococcus | |
aureus; Transcriptome; | |
Virulence;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Ledda2017-eh, | |
title = "Re-emergence of methicillin susceptibility in a resistant lineage | |
of Staphylococcus aureus", | |
author = "Ledda, Alice and Price, James R and Cole, Kevin and Llewelyn, | |
Martin J and Kearns, Angela M and Crook, Derrick W and Paul, John | |
and Didelot, Xavier", | |
abstract = "OBJECTIVES: MRSA is a leading cause of hospital-associated | |
infection. Acquired resistance is encoded by the mecA gene or its | |
homologue mecC, but little is known about the evolutionary | |
dynamics involved in gain and loss of resistance. The objective | |
of this study was to obtain an expanded understanding of | |
Staphylococcus aureus methicillin resistance microevolution in | |
vivo, by focusing on a single lineage. METHODS: We compared the | |
whole-genome sequences of 231 isolates from a single epidemic | |
lineage [clonal complex 30 (CC30) and spa-type t018] of S. aureus | |
that caused an epidemic in the UK. RESULTS: We show that | |
resistance to methicillin in this single lineage was gained on at | |
least two separate occasions, one of which led to a clonal | |
expansion around 1995 presumably caused by a selective advantage. | |
Resistance was, however, subsequently lost in vivo by nine | |
strains isolated between 2008 and 2012. We describe the genetic | |
mechanisms involved in this loss of resistance and the imperfect | |
relationship between genotypic and phenotypic resistance. | |
CONCLUSIONS: The recent re-emergence of methicillin | |
susceptibility in this epidemic lineage suggests a significant | |
fitness cost of resistance and reduced selective advantage | |
following the introduction in the mid-2000s of MRSA hospital | |
control measures throughout the UK.", | |
journal = "J. Antimicrob. Chemother.", | |
month = jan, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Uhlemann2017-db, | |
title = "Evolutionary Dynamics of Pandemic {Methicillin-Sensitive} | |
Staphylococcus aureus {ST398} and Its International Spread via | |
Routes of Human Migration", | |
author = "Uhlemann, Anne-Catrin and McAdam, Paul R and Sullivan, Sean B and | |
Knox, Justin R and Khiabanian, Hossein and Rabadan, Raul and | |
Davies, Peter R and Fitzgerald, J Ross and Lowy, Franklin D", | |
abstract = "Methicillin-susceptible Staphylococcus aureus (MSSA) accounts for | |
the majority of S. aureus infections globally, and yet | |
surprisingly little is known about its clonal evolution. We | |
applied comparative whole-genome sequencing (WGS) analyses to | |
epidemiologically and geographically diverse ST398-MSSA, a | |
pandemic lineage affecting both humans and livestock. Bayesian | |
phylogenetic analysis predicted divergence of human-associated | |
ST398-MSSA ~40 years ago. Isolates from Midwestern pigs and | |
veterinarians differed substantially from those in New York City | |
(NYC). Pig ST398 strains contained a large region of | |
recombination representing imports from multiple sequence types | |
(STs). Phylogeographic analyses supported the spread of | |
ST398-MSSA along local cultural and migratory links between parts | |
of the Caribbean, North America, and France, respectively. | |
Applying pairwise single-nucleotide polymorphism (SNP) distances | |
as a measure of genetic relatedness between isolates, we observed | |
that ST398 not only clustered in households but also frequently | |
extended across local social networks. Isolates collected from | |
environmental surfaces reflected the full diversity of colonizing | |
individuals, highlighting their potentially critical role as | |
reservoirs for transmission and diversification. Strikingly, we | |
observed high within-host SNP variability compared to our | |
previous studies on the dominant methicillin-resistant | |
Staphylococcus aureus (MRSA) clone USA300. Our data indicate that | |
the dynamics of colonization, persistence, and transmission | |
differ substantially between USA300-MRSA and ST398-MSSA. Taken | |
together, our study reveals local and international routes of | |
transmission for a major MSSA clone, indicating key impacts of | |
recombination and mutation on genetic diversification and | |
highlighting important ecological differences from epidemic | |
USA300. Our study demonstrates extensive local and international | |
routes of transmission for a major MSSA clone despite the lack of | |
substantial antibiotic resistance. IMPORTANCE: Unlike | |
methicillin-resistant Staphylococcus aureus (MRSA), surprisingly | |
little is known about the clonal evolution of | |
methicillin-susceptible S. aureus (MSSA), although these strains | |
account for the majority of S. aureus infections. To better | |
understand how MSSA spreads and becomes established in | |
communities, we applied comparative bacterial whole-genome | |
sequencing to pandemic ST398-MSSA, a clone of clinical importance | |
affecting humans and livestock in different geographic regions. | |
Phylogeographic analyses identified that ST398-MSSA spread along | |
local cultural and migratory links between parts of the | |
Caribbean, North America, and France, respectively. We observed | |
high within-host SNP variability compared to our previous studies | |
on the dominant MRSA clone USA300. Our data indicate that the | |
dynamics of colonization, persistence, and transmission differ | |
substantially between USA300 MRSA and ST398 MSSA.", | |
journal = "MBio", | |
volume = 8, | |
number = 1, | |
month = jan, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Escobar-Perez2017-wu, | |
title = "Emergence and spread of a new community-genotype | |
methicillin-resistant Staphylococcus aureus clone in Colombia", | |
author = "Escobar-Perez, Javier and Reyes, Niradiz and Marquez-Ortiz, | |
Ricaurte Alejandro and Rebollo, Juan and Pinz{\'o}n, Hernando and | |
Tovar, Catalina and Moreno-Casta{\~n}eda, Jaime and Corredor, | |
Zayda Lorena and Castro, Betsy Esperanza and Moncada, Maria | |
Victoria and Vanegas, Natasha", | |
abstract = "BACKGROUND: Community-genotype methicillin-resistant | |
Staphylococcus aureus (CG-MRSA) clones are a global concern due | |
to their resistance and increased virulence and their ability to | |
cause infections both hospitalized patients and healthy people in | |
the community. Here, we characterize 32 isolates of a new CG-MRSA | |
clone. These isolates were identified in four cities in Colombia, | |
South America. METHODS: The isolates were recovered from four | |
different epidemiological and prospective studies that were | |
conducted in several regions of Colombia. Molecular | |
characterizations included multilocus sequence typing; | |
pulsed-field gel electrophoresis; SCCmec, agr and spa typing; and | |
whole-genome sequencing. RESULTS: All isolates belonged to ST923 | |
(clonal complex 8), harbouring SCCmec IVa and a spa type t1635 | |
and lacking an arginine catabolism mobile element. The isolates | |
were classified as COL923, were resistant to at least one | |
non-beta-lactam antibiotic, and exhibited high frequencies | |
(>60\%) of resistance to macrolides and tetracycline. Using | |
whole-genome sequencing, we found that this new clone harbours | |
novel prophage 3 and beta-island structures and a slightly | |
different pathogenicity island 5. Moreover, isolates belonging to | |
the COL923 clone are grouped in a different clade than USA300 and | |
USA300-LV. CONCLUSION: Our results show the emergence and spread | |
of the COL923 clone in different cities in Colombia. This clone | |
is resistant to several antibiotics and possesses new structures | |
in its mobile genetic elements.", | |
journal = "BMC Infect. Dis.", | |
volume = 17, | |
number = 1, | |
pages = "108", | |
month = jan, | |
year = 2017, | |
keywords = "Colombia; Community; Methicillin-resistant Staphylococcus aureus; | |
New clone;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Eyre2012-xf, | |
title = "A pilot study of rapid benchtop sequencing of Staphylococcus | |
aureus and Clostridium difficile for outbreak detection and | |
surveillance", | |
author = "Eyre, David W and Golubchik, Tanya and Gordon, N Claire and | |
Bowden, Rory and Piazza, Paolo and Batty, Elizabeth M and Ip, | |
Camilla L C and Wilson, Daniel J and Didelot, Xavier and | |
O'Connor, Lily and Lay, Rochelle and Buck, David and Kearns, | |
Angela M and Shaw, Angela and Paul, John and Wilcox, Mark H and | |
Donnelly, Peter J and Peto, Tim E A and Walker, A Sarah and | |
Crook, Derrick W", | |
abstract = "OBJECTIVES: To investigate the prospects of newly available | |
benchtop sequencers to provide rapid whole-genome data in routine | |
clinical practice. Next-generation sequencing has the potential | |
to resolve uncertainties surrounding the route and timing of | |
person-to-person transmission of healthcare-associated infection, | |
which has been a major impediment to optimal management. DESIGN: | |
The authors used Illumina MiSeq benchtop sequencing to undertake | |
case studies investigating potential outbreaks of | |
methicillin-resistant Staphylococcus aureus (MRSA) and | |
Clostridium difficile. SETTING: Isolates were obtained from | |
potential outbreaks associated with three UK hospitals. | |
PARTICIPANTS: Isolates were sequenced from a cluster of eight | |
MRSA carriers and an associated bacteraemia case in an intensive | |
care unit, another MRSA cluster of six cases and two clusters of | |
C difficile. Additionally, all C difficile isolates from cases | |
over 6 weeks in a single hospital were rapidly sequenced and | |
compared with local strain sequences obtained in the preceding 3 | |
years. MAIN OUTCOME MEASURE: Whole-genome genetic relatedness of | |
the isolates within each epidemiological cluster. RESULTS: | |
Twenty-six MRSA and 15 C difficile isolates were successfully | |
sequenced and analysed within 5 days of culture. Both MRSA | |
clusters were identified as outbreaks, with most sequences in | |
each cluster indistinguishable and all within three single | |
nucleotide variants (SNVs). Epidemiologically unrelated isolates | |
of the same spa-type were genetically distinct ($\geq$21 SNVs). | |
In both C difficile clusters, closely epidemiologically linked | |
cases (in one case sharing the same strain type) were shown to be | |
genetically distinct ($\geq$144 SNVs). A reconstruction applying | |
rapid sequencing in C difficile surveillance provided early | |
outbreak detection and identified previously undetected probable | |
community transmission. CONCLUSIONS: This benchtop sequencing | |
technology is widely generalisable to human bacterial pathogens. | |
The findings provide several good examples of how rapid and | |
precise sequencing could transform identification of transmission | |
of healthcare-associated infection and therefore improve hospital | |
infection control and patient outcomes in routine clinical | |
practice.", | |
journal = "BMJ Open", | |
volume = 2, | |
number = 3, | |
month = jun, | |
year = 2012, | |
keywords = "Staphylococcus;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Harrison2014-mh, | |
title = "A shared population of epidemic methicillin-resistant | |
Staphylococcus aureus 15 circulates in humans and companion | |
animals", | |
author = "Harrison, Ewan M and Weinert, Lucy A and Holden, Matthew T G and | |
Welch, John J and Wilson, Katherine and Morgan, Fiona J E and | |
Harris, Simon R and Loeffler, Anette and Boag, Amanda K and | |
Peacock, Sharon J and Paterson, Gavin K and Waller, Andrew S and | |
Parkhill, Julian and Holmes, Mark A", | |
abstract = "UNLABELLED: Methicillin-resistant Staphylococcus aureus (MRSA) is | |
a global human health problem causing infections in both | |
hospitals and the community. Companion animals, such as cats, | |
dogs, and horses, are also frequently colonized by MRSA and can | |
become infected. We sequenced the genomes of 46 multilocus | |
sequence type (ST) 22 MRSA isolates from cats and dogs in the | |
United Kingdom and compared these to an extensive population | |
framework of human isolates from the same lineage. Phylogenomic | |
analyses showed that all companion animal isolates were | |
interspersed throughout the epidemic MRSA-15 (EMRSA-15) pandemic | |
clade and clustered with human isolates from the United Kingdom, | |
with human isolates basal to those from companion animals, | |
suggesting a human source for isolates infecting companion | |
animals. A number of isolates from the same veterinary hospital | |
clustered together, suggesting that as in human hospitals, | |
EMRSA-15 isolates are readily transmitted in the veterinary | |
hospital setting. Genome-wide association analysis did not | |
identify any host-specific single nucleotide polymorphisms (SNPs) | |
or virulence factors. However, isolates from companion animals | |
were significantly less likely to harbor a plasmid encoding | |
erythromycin resistance. When this plasmid was present in | |
animal-associated isolates, it was more likely to contain | |
mutations mediating resistance to clindamycin. This finding is | |
consistent with the low levels of erythromycin and high levels of | |
clindamycin used in veterinary medicine in the United Kingdom. | |
This study furthers the ``one health'' view of infectious | |
diseases that the pathogen pool of human and animal populations | |
are intrinsically linked and provides evidence that antibiotic | |
usage in animal medicine is shaping the population of a major | |
human pathogen. IMPORTANCE: Methicillin-resistant Staphylococcus | |
aureus (MRSA) is major problem in human medicine. Companion | |
animals, such as cats, dogs, and horses, can also become | |
colonized and infected by MRSA. Here, we demonstrate that a | |
shared population of an important and globally disseminated | |
lineage of MRSA can infect both humans and companion animals | |
without undergoing host adaptation. This suggests that companion | |
animals might act as a reservoir for human infections. We also | |
show that the isolates from companion animals have differences in | |
the presence of certain antibiotic resistance genes. This study | |
furthers the ``one health'' view of infectious diseases by | |
demonstrating that the pool of MRSA isolates in the human and | |
animal populations are shared and highlights how different | |
antibiotic usage patterns between human and veterinary medicine | |
can shape the population of bacterial pathogens.", | |
journal = "MBio", | |
volume = 5, | |
number = 3, | |
pages = "e00985--13", | |
month = may, | |
year = 2014, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Warne2016-sf, | |
title = "The {Ess/Type} {VII} secretion system of Staphylococcus aureus | |
shows unexpected genetic diversity", | |
author = "Warne, Ben and Harkins, Catriona P and Harris, Simon R and | |
Vatsiou, Alexandra and Stanley-Wall, Nicola and Parkhill, Julian | |
and Peacock, Sharon J and Palmer, Tracy and Holden, Matthew T G", | |
abstract = "BACKGROUND: Type VII protein secretion (T7SS) is a specialised | |
system for excreting extracellular proteins across bacterial cell | |
membranes and has been associated with virulence in | |
Staphylococcus aureus. The genetic diversity of the ess locus, | |
which encodes the T7SS, and the functions of proteins encoded | |
within it are poorly understood. RESULTS: We used whole genome | |
sequence data from 153 isolates representative of the diversity | |
of the species to investigate the genetic variability of T7SS | |
across S. aureus. The ess loci were found to comprise of four | |
distinct modules based on gene content and relative conservation. | |
Modules 1 and 4, comprising of the 5' and 3' modules of the ess | |
locus, contained the most conserved clusters of genes across the | |
species. Module 1 contained genes encoding the secreted protein | |
EsxA, and the EsaAB and EssAB components of the T7SS machinery, | |
and Module 4 contained two functionally uncharacterized conserved | |
membrane proteins. Across the species four variants of Module 2 | |
were identified containing the essC gene, each of which was | |
associated with a specific group of downstream genes. The most | |
diverse module of the ess locus was Module 3 comprising a highly | |
variable arrangement of hypothetical proteins. RNA-Seq was | |
performed on representatives of the four Module 2 variants and | |
demonstrated strain-specific differences in the levels of | |
transcription in the conserved Module 1 components and | |
transcriptional linkage Module 2, and provided evidence of the | |
expression of genes the variable regions of the ess loci. | |
CONCLUSIONS: The ess locus of S. aureus exhibits modularity and | |
organisational variation across the species and transcriptional | |
variation. In silico analysis of ess loci encoded hypothetical | |
proteins identified potential novel secreted substrates for the | |
T7SS. The considerable variety in operon arrangement between | |
otherwise closely related isolates provides strong evidence for | |
recombination at this locus. Comparison of these recombination | |
regions with each other, and with the genomes of other | |
Staphylococcal species, failed to identify evidence of intra- and | |
inter-species recombination, however the analysis identified a | |
novel T7SS in another pathogenic staphylococci, Staphylococcus | |
lugdunensis.", | |
journal = "BMC Genomics", | |
volume = 17, | |
pages = "222", | |
month = mar, | |
year = 2016, | |
keywords = "Secretion; Staphylococcus aureus; Type | |
VII;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Koskella2014-it, | |
title = "Bacteria-phage coevolution as a driver of ecological and | |
evolutionary processes in microbial communities", | |
author = "Koskella, Britt and Brockhurst, Michael A", | |
abstract = "Bacteria-phage coevolution, the reciprocal evolution between | |
bacterial hosts and the phages that infect them, is an important | |
driver of ecological and evolutionary processes in microbial | |
communities. There is growing evidence from both laboratory and | |
natural populations that coevolution can maintain phenotypic and | |
genetic diversity, increase the rate of bacterial and phage | |
evolution and divergence, affect community structure, and shape | |
the evolution of ecologically relevant bacterial traits. Although | |
the study of bacteria-phage coevolution is still in its infancy, | |
with open questions regarding the specificity of the interaction, | |
the gene networks of coevolving partners, and the relative | |
importance of the coevolving interaction in complex communities | |
and environments, there have recently been major advancements in | |
the field. In this review, we sum up our current understanding of | |
bacteria-phage coevolution both in the laboratory and in nature, | |
discuss recent findings on both the coevolutionary process itself | |
and the impact of coevolution on bacterial phenotype, diversity | |
and interactions with other species (particularly their | |
eukaryotic hosts), and outline future directions for the field.", | |
journal = "FEMS Microbiol. Rev.", | |
volume = 38, | |
number = 5, | |
pages = "916--931", | |
month = sep, | |
year = 2014, | |
keywords = "antagonistic; bacteriophage; host-parasite; infection; | |
resistance; species | |
interaction;phage;evolution;Staphylococcus;Papers\_for\_Michelle;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{McAdam2011-cn, | |
title = "Adaptive evolution of Staphylococcus aureus during chronic | |
endobronchial infection of a cystic fibrosis patient", | |
author = "McAdam, Paul R and Holmes, Anne and Templeton, Kate E and | |
Fitzgerald, J Ross", | |
abstract = "The molecular adaptation of Staphylococcus aureus to its host | |
during chronic infection is not well understood. Comparative | |
genome sequencing of 3 S. aureus isolates obtained sequentially | |
over 26 months from the airways of a cystic fibrosis patient, | |
revealed variation in phage content, and genetic polymorphisms in | |
genes which influence antibiotic resistance, and global | |
regulation of virulence. The majority of polymorphisms were | |
isolate-specific suggesting the existence of an heterogeneous | |
infecting population that evolved from a single infecting strain | |
of S. aureus. The genetic variation identified correlated with | |
differences in growth rate, hemolytic activity, and antibiotic | |
sensitivity, implying a profound effect on the ecology of S. | |
aureus. In particular, a high frequency of mutations in loci | |
associated with the alternate transcription factor SigB, were | |
observed. The identification of genes under diversifying | |
selection during long-term infection may inform the design of | |
novel therapeutics for the control of refractory chronic | |
infections.", | |
journal = "PLoS One", | |
volume = 6, | |
number = 9, | |
pages = "e24301", | |
month = sep, | |
year = 2011, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Donker2017-te, | |
title = "Population genetic structuring of methicillin-resistant | |
Staphylococcus aureus clone {EMRSA-15} within {UK} reflects | |
patient referral patterns", | |
author = "Donker, Tjibbe and Reuter, Sandra and Scriberras, James and | |
Reynolds, Rosy and Brown, Nicholas M and Est{\'e}e | |
T{\"o}r{\"o}k, M and James, Richard and {East of England | |
Microbiology Research Network} and Aanensen, David M and | |
Bentley, Stephen D and Holden, Matthew T G and Parkhill, Julian | |
and Spratt, Brian G and Peacock, Sharon J and Feil, Edward J and | |
Grundmann, Hajo", | |
abstract = "Antibiotic resistance forms a serious threat to the health of | |
hospitalised patients, rendering otherwise treatable bacterial | |
infections potentially life-threatening. A thorough | |
understanding of the mechanisms by which resistance spreads | |
between patients in different hospitals is required in order to | |
design effective control strategies. We measured the differences | |
between bacterial populations of 52 hospitals in the United | |
Kingdom and Ireland, using whole-genome sequences from 1085 MRSA | |
clonal complex 22 isolates collected between 1998 and 2012. The | |
genetic differences between bacterial populations were compared | |
with the number of patients transferred between hospitals and | |
their regional structure. The MRSA populations within single | |
hospitals, regions and countries were genetically distinct from | |
the rest of the bacterial population at each of these levels. | |
Hospitals from the same patient referral regions showed more | |
similar MRSA populations, as did hospitals sharing many | |
patients. Furthermore, the bacterial populations from different | |
time-periods within the same hospital were generally more | |
similar to each other than contemporaneous bacterial populations | |
from different hospitals. We conclude that, while a large part | |
of the dispersal and expansion of MRSA takes place among | |
patients seeking care in single hospitals, inter-hospital spread | |
of resistant bacteria is by no means a rare occurrence. | |
Hospitals are exposed to constant introductions of MRSA on a | |
number of levels: (1) most MRSA is received from hospitals that | |
directly transfer large numbers of patients, while (2) fewer | |
introductions happen between regions or (3) across national | |
borders, reflecting lower numbers of transferred patients. A | |
joint coordinated control effort between hospitals, is therefore | |
paramount for the national control of MRSA, antibiotic-resistant | |
bacteria and other hospital-associated pathogens.", | |
journal = "Microbial Genomics", | |
publisher = "Microbiology Society", | |
month = jul, | |
year = 2017, | |
keywords = "hospital network; antimicrobial Resistance; | |
MRSA;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{McMillan2017-xh, | |
title = "Draft Genome Sequences of 15 Staphylococcus aureus Isolates | |
Recovered from Raw Milk and Associated Milk Filters from | |
Victoria, Australia", | |
author = "McMillan, Kate and Allnutt, Theodore R and Fox, Edward M", | |
abstract = "This study describes draft whole genomes of 15 Staphylococcus | |
aureus isolates from dairy farms located in Victoria, Australia. | |
Two novel sequence types (ST3183 and ST3184) were identified | |
among these isolates.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 2, | |
month = jan, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Franck2017-tn, | |
title = "Staphylococcal aureus Enterotoxin {C} and {Enterotoxin-Like} {L} | |
Associated with Post-partum Mastitis", | |
author = "Franck, Kristina T and Gumpert, Heidi and Olesen, Bente and | |
Larsen, Anders R and Petersen, Andreas and Bangsborg, Jette and | |
Albertsen, Per and Westh, Henrik and Bartels, Mette D", | |
abstract = "Denmark is a low prevalence country with regard to methicillin | |
resistant Staphylococcus aureus (MRSA). In 2008 and 2014, two | |
neonatal wards in the Copenhagen area experienced outbreaks with | |
a typical community acquired MRSA belonging to the same spa type | |
and sequence type (t015:ST45) and both were PVL and ACME | |
negative. In outbreak 1, the isolates harbored SCCmec IVa and in | |
outbreak 2 SCCmec V. The clinical presentation differed between | |
the two outbreaks, as none of five MRSA positive mothers in | |
outbreak 1 had mastitis vs. five of six MRSA positive mothers in | |
outbreak 2 (p < 0.02). To investigate if whole-genome sequencing | |
could identify virulence genes associated with mastitis, | |
t015:ST45 isolates from Denmark (N = 101) were whole-genome | |
sequenced. Sequence analysis confirmed two separate outbreaks | |
with no sign of sustained spread into the community. Analysis of | |
the accessory genome between isolates from the two outbreaks | |
revealed a S. aureus pathogenicity island containing enterotoxin | |
C and enterotoxin-like L only in isolates from outbreak 2. | |
Enterotoxin C and enterotoxin-like L carrying S. aureus are | |
associated with bovine mastitis and our findings indicate that | |
these may also be important virulence factors for human mastitis.", | |
journal = "Front. Microbiol.", | |
volume = 8, | |
pages = "173", | |
month = feb, | |
year = 2017, | |
keywords = "MRSA; SaPITokyo12571; clinical presentation; enterotoxins; | |
pathogenicity islands; post-partum mastitis; virulence; | |
whole-genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Murray2017-uw, | |
title = "Recombination-mediated host-adaptation by avian Staphylococcus | |
aureus", | |
author = "Murray, Susan and Pascoe, Ben and M{\'e}ric, Guillaume and | |
Mageiros, Leonardos and Yahara, Koji and Hitchings, Matthew D and | |
Friedmann, Yasmin and Wilkinson, Thomas S and Gormley, Fraser J | |
and Mack, Dietrich and Bray, James E and Lamble, Sarah and | |
Bowden, Rory and Jolley, Keith A and Maiden, Martin C J and | |
Wendlandt, Sarah and Schwarz, Stefan and Corander, Jukka and Ross | |
Fitzgerald, J and Sheppard, Samuel K", | |
journal = "Genome Biol. Evol.", | |
month = feb, | |
year = 2017, | |
keywords = "Staphylococcus;recombination;colonization;plasmid;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human" | |
} | |
@ARTICLE{Bakthavatchalam2017-bb, | |
title = "Draft genome sequence of reduced teicoplanin-susceptible and | |
vancomycin-heteroresistant methicillin-resistant Staphylococcus | |
aureus from sepsis cases", | |
author = "Bakthavatchalam, Yamuna Devi and Veeraraghavan, Balaji and | |
Devanga Ragupathi, Naveen Kumar and Babu, Priyanka and Munuswamy, | |
Elakkiya and David, Thambu", | |
abstract = "Here we report the whole-genome shotgun sequence of six | |
methicillin-resistant Staphylococcus aureus (MRSA) showing | |
reduced susceptibility to both vancomycin and teicoplanin. The | |
typical Indian community-acquired MRSA (CA-MRSA) clone | |
ST772-MRSA-V-t657 was the most common genotype (3/6; 50\%), | |
followed by ST672-MRSA-IV (2/6; 33\%) and ST22-MRSA-IV (1/6; | |
17\%). All strains harboured a mutation in the tcaRAB operon, | |
vraSR, graSR and/or rpoB genes, which are frequently mutated | |
determinants in a heteroresistant vancomycin-intermediate S. | |
aureus (hVISA) phenotype.", | |
journal = "J Glob Antimicrob Resist", | |
volume = 8, | |
pages = "169--171", | |
month = feb, | |
year = 2017, | |
keywords = "MRSA; hVISA; | |
tcaA;Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Diene2017-oq, | |
title = "Prophages and adaptation of Staphylococcus aureus {ST398} to the | |
human clinic", | |
author = "Diene, Seydina M and Corvaglia, Anna Rita and Fran{\c c}ois, | |
Patrice and van der Mee-Marquet, Nathalie and {Regional Infection | |
Control Group of the Centre Region}", | |
abstract = "BACKGROUND: It has been suggested that prophages in the ST398 S. | |
aureus clone are responsible for expanding ST398's spectrum of | |
action and increasing its ability to cause human infections. We | |
carried out the first characterization of the various prophages | |
carried by 76 ST398 bloodstream infection (BSI) isolates obtained | |
over 9 years of observation. RESULTS: Whole-genome sequencing of | |
22 representative isolates showed (1) the presence of the | |
$\varphi$3-prophage and diverse genetic features typical of | |
animal-associated isolates (i.e., SCCmec XI element, Tn916 | |
transposon and non $\varphi$3-prophages) in a majority of BSI | |
isolates, (2) one BSI isolate devoid of the $\varphi$3-prophage | |
but otherwise similar to an animal-infecting isolate, (3) 35 | |
prophages carrying numerous genes previously associated with | |
virulence or immune evasion in animal models of staphylococcal | |
infections. The analysis of prophage content in all 76 BSI | |
isolates showed an increasing prevalence of polylysogeny over | |
time. Overall, over the course of the last 10 years, the BSI | |
isolates appear to have acquired increasing numbers of genetic | |
features previously shown to contribute to bacterial adaptation | |
and virulence in animal models of staphylococcal infections. | |
CONCLUSIONS: We hypothesize that lysogeny has played a | |
significant role in increasing the ability of the ST398 clone to | |
cause infections in humans. Our findings highlight the risk that | |
the ST398 lineage will increase its threat to public health by | |
continuing to acquire virulence and/or multiple | |
antibiotic-resistance genes from hospital-associated clones of | |
Staphylococcus aureus.", | |
journal = "BMC Genomics", | |
volume = 18, | |
number = 1, | |
pages = "133", | |
month = feb, | |
year = 2017, | |
keywords = "Bloodstream infections; CC398 lineage; Evolution; | |
Livestock-associated; Phage content; Prophage; Staphylococcus | |
aureus; | |
$\varphi$3-prophage;Staphylococcus;phage;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human", | |
language = "en" | |
} | |
@ARTICLE{Mannala2017-tj, | |
title = "Complete Genome and Plasmid Sequences of \textit{Staphylococcus | |
aureus} {EDCC} 5055 ({DSM} 28763), Used To Study | |
{Implant-Associated} Infections", | |
author = "Mannala, Gopala Krishna and Hain, Torsten and Spr{\"o}er, Cathrin | |
and Bunk, Boyke and Overmann, J{\"o}rg and Alt, Volker and | |
Domann, Eugen", | |
abstract = "Staphylococcus aureus EDCC 5055 (DSM 28763) is a human clinical | |
wound isolate intensively used to study implant-associated | |
infections in rabbit and rat infection models. Here, we report | |
its complete genome sequence (2,794,437 bp) along with that of | |
one plasmid (27,437 bp). This strain belongs to sequence type 8 | |
and contains a mecA gene.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 8, | |
month = feb, | |
year = 2017, | |
keywords = "Staphylococcus;plasmid;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Kpeli2017-lk, | |
title = "Genomic analysis of {ST88} community-acquired methicillin | |
resistant \textit{Staphylococcus aureus} in Ghana", | |
author = "Kpeli, Grace and Buultjens, Andrew H and Giulieri, Stefano and | |
Owusu-Mireku, Evelyn and Aboagye, Samuel Y and Baines, Sarah L | |
and Seemann, Torsten and Bulach, Dieter and Gon{\c c}alves da | |
Silva, Anders and Monk, Ian R and Howden, Benjamin P and | |
Pluschke, Gerd and Yeboah-Manu, Dorothy and Stinear, Timothy", | |
abstract = "BACKGROUND: The emergence and evolution of community-acquired | |
methicillin resistant Staphylococcus aureus (CA-MRSA) strains in | |
Africa is poorly understood. However, one particular MRSA lineage | |
called ST88, appears to be rapidly establishing itself as an | |
``African'' CA-MRSA clone. In this study, we employed whole | |
genome sequencing to provide more information on the genetic | |
background of ST88 CA-MRSA isolates from Ghana and to describe in | |
detail ST88 CA-MRSA isolates in comparison with other MRSA | |
lineages worldwide. METHODS: We first established a complete ST88 | |
reference genome (AUS0325) using PacBio SMRT sequencing. We then | |
used comparative genomics to assess relatedness among 17 ST88 | |
CA-MRSA isolates recovered from patients attending Buruli ulcer | |
treatment centres in Ghana, three non-African ST88s and 15 other | |
MRSA lineages. RESULTS: We show that Ghanaian ST88 forms a | |
discrete MRSA lineage (harbouring SCCmec-IV [2B]). Gene content | |
analysis identified five distinct genomic regions enriched among | |
ST88 isolates compared with the other S. aureus lineages. The | |
Ghanaian ST88 isolates had only 658 core genome SNPs and there | |
was no correlation between phylogeny and geography, suggesting | |
the recent spread of this clone. The lineage was also resistant | |
to multiple classes of antibiotics including $\beta$-lactams, | |
tetracycline and chloramphenicol. DISCUSSION: This study reveals | |
that S. aureus ST88-IV is a recently emerging and rapidly | |
spreading CA-MRSA clone in Ghana. The study highlights the | |
capacity of small snapshot genomic studies to provide actionable | |
public health information in resource limited settings. To our | |
knowledge this is the first genomic assessment of the ST88 | |
CA-MRSA clone.", | |
journal = "PeerJ", | |
volume = 5, | |
pages = "e3047", | |
month = feb, | |
year = 2017, | |
keywords = "CA-MRSA; Comparative genomics; MRSA; Phylogeography; ST88; | |
Staphylococcus aureus; Whole genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Piso2017-yj, | |
title = "A {Cross-Sectional} Study of Colonization Rates with | |
{Methicillin-Resistant} Staphylococcus aureus ({MRSA}) and | |
{Extended-Spectrum} {Beta-Lactamase} ({ESBL}) and | |
{Carbapenemase-Producing} Enterobacteriaceae in Four Swiss | |
Refugee Centres", | |
author = "Piso, Rein Jan and K{\"a}ch, Roman and Pop, Roxana and Zillig, | |
Daniela and Schibli, Urs and Bassetti, Stefano and Meinel, | |
Dominik and Egli, Adrian", | |
abstract = "BACKGROUND: The recent crisis of refugees seeking asylum in | |
European countries challenges public health on many levels. Most | |
refugees currently arrive from Syria, Afghanistan, or Eritrea. | |
Data about multidrug resistant bacteria (MDR) prevalence are not | |
present for these countries. However, when entering the European | |
heath care systems, data about colonisation rates regarding | |
highly resistant bacterial pathogens are important. METHODS: We | |
performed a cross-sectional screening in four Swiss refugee | |
centres to determine the colonization rates for MRSA and ESBL- | |
and carbapenemase-producing Enterobacteriaceae. We used | |
pharyngeal, nasal, and inguinal swabs for MRSA and rectal swabs | |
and urine for ESBL and carbapenemase screening using standard | |
microbiological procedures. Whole genome sequencing (WGS) was | |
used to determine the relatedness of MRSA isolates with high | |
resolution due to a suspected outbreak. RESULTS: 41/261(15.7\%) | |
refugees were colonized with MRSA. No differences regarding the | |
country of origin were observed. However, in a single centre | |
significantly more were colonized, which was confirmed to be a | |
recent local outbreak. 57/241 (23.7\%) refugees were colonized | |
with ESBL with significantly higher colonisation in persons | |
originating from the Middle East (35.1\%, p<0.001). No | |
carbapenemase producers were detected. CONCLUSION: The | |
colonisation rate of the refugees was about 10 times higher for | |
MRSA and 2-5 times higher for ESBL compared to the Swiss | |
population. Contact precaution is warranted for these persons if | |
they enter medical care. In cases of infections, MRSA and | |
ESBL-producing Enterobacteriaceae should be considered regarding | |
antibiotic treatment choices.", | |
journal = "PLoS One", | |
volume = 12, | |
number = 1, | |
pages = "e0170251", | |
month = jan, | |
year = 2017, | |
keywords = "Staphylococcus;antibiotics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@UNPUBLISHED{Young2017-di, | |
title = "Severe infections emerge from the microbiome by adaptive | |
evolution", | |
author = "Young, Bernadette C and Wu, Chieh-Hsi and Claire Gordon, N and | |
Cole, Kevin and Price, James R and Liu, Elian and Sheppard, Anna | |
and Perera, Sanuki and Charlesworth, Jane and Golubchik, Tanya | |
and Iqbal, Zamin and Bowden, Rory and Massey, Ruth and Paul, John | |
and Crook, Derrick W and Peto, Timothy E A and Sarah Walker, A | |
and Llewelyn, Martin and Wyllie, David H and Wilson, Daniel", | |
abstract = "Bacteria responsible for the greatest global mortality colonize | |
the human microbiome far more frequently than they cause severe | |
infections. Whether mutation and selection within the microbiome | |
precipitate infection is unknown. To address this question, we | |
investigated de novo mutation in 1163 Staphylococcus aureus | |
genomes from 105 infected patients with nose-colonization. We | |
report that 72\% of the infections emerged from the microbiome, | |
with infecting and nose-colonizing bacteria showing systematic | |
adaptive differences. We found 3.6-fold, 2.9-fold and 2.8-fold | |
enrichments of protein-altering variants in genes responding to | |
rsp, which regulates surface antigens and toxicity; agr, which | |
regulates quorum-sensing, toxicity and abscess formation; and | |
host-derived antimicrobial peptides, respectively. These adaptive | |
signatures were not observed in healthy carriers and differed | |
from prevailing species-level signals of selection, suggesting | |
disease-associated, short-term, within-host selection pressures. | |
Our results show that infection, like a cancer of the microbiome, | |
emerges through spontaneous adaptive evolution, raising new | |
possibilities for diagnosis and treatment.", | |
journal = "bioRxiv", | |
pages = "116681", | |
month = mar, | |
year = 2017, | |
keywords = "microbiome;Staphylococcus;evolution;systems\_biology;confirmed\_SA-resistance-mutation;within-host-evolution;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Murray2017-if, | |
title = "Recombination-mediated host-adaptation by avian Staphylococcus | |
aureus", | |
author = "Murray, Susan and Pascoe, Ben and M{\'e}ric, Guillaume and | |
Mageiros, Leonardos and Yahara, Koji and Hitchings, Matthew D and | |
Friedmann, Yasmin and Wilkinson, Thomas S and Gormley, Fraser J | |
and Mack, Dietrich and Bray, James E and Lamble, Sarah and | |
Bowden, Rory and Jolley, Keith A and Maiden, Martin C J and | |
Wendlandt, Sarah and Schwarz, Stefan and Corander, Jukka and Ross | |
Fitzgerald, J and Sheppard, Samuel K", | |
abstract = "Staphylococcus aureus are globally disseminated among farmed | |
chickens causing skeletal muscle infections, dermatitis and | |
septicaemia. The emergence of poultry-associated lineages has | |
involved zoonotic transmission from humans to chickens but | |
questions remain about the specific adaptations that promote | |
proliferation of chicken pathogens. We characterised genetic | |
variation in a population of genome-sequenced S. aureus isolates | |
of poultry and human origin. Genealogical analysis identified a | |
dominant poultry-associated sequence cluster within the CC5 | |
clonal complex. Poultry and human CC5 isolates were significantly | |
distinct from each other and more recombination events were | |
detected in the poultry isolates. We identified forty-four | |
recombination events in 33 genes along the branch extending to | |
the poultry-specific CC5 cluster, and 47 genes were found more | |
often in CC5 poultry isolates compared to those from humans. Many | |
of these gene sequences were common in chicken isolates from | |
other clonal complexes suggesting horizontal gene transfer among | |
poultry associated lineages. Consistent with functional | |
predictions for putative poultry-associated genes, poultry | |
isolates showed enhanced growth at 42oC and greater erythrocyte | |
lysis on chicken blood agar in comparison with human isolates. By | |
combining phenotype information with evolutionary analysis of | |
staphylococcal genomes, we provide evidence of adaptation, | |
following a human-to-poultry host transition. This has important | |
implications for the emergence and dissemination of new | |
pathogenic clones associated with modern agriculture.", | |
journal = "Genome Biol. Evol.", | |
month = feb, | |
year = 2017, | |
keywords = "Evolution; Genomics; Poultry infection; Recombination; | |
Staphylococcus;Staphylococcus;recombination;colonization;plasmid;Staphylococcus\_aureus\_genome\_papers;Staph-animal-human", | |
language = "en" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{Bayliss2017-kb, | |
title = "The use of Oxford Nanopore native barcoding for complete genome | |
assembly", | |
author = "Bayliss, Sion C and Hunt, Vicky L and Yokoyama, Maho and Thorpe, | |
Harry A and Feil, Edward J", | |
abstract = "Background: The Oxford Nanopore Technologies MinION(TM) is a | |
mobile DNA sequencer that can produce long read sequences with a | |
short turn-around time. Here we report the first demonstration of | |
single contig genome assembly using Oxford Nanopore native | |
barcoding when applied to a multiplexed library of 12 samples and | |
combined with existing Illumina short-read data. This paves the | |
way for the closure of multiple bacterial genomes from a single | |
MinION(TM) sequencing run, given the availability of existing | |
short-read data. The strain we used, MHO\_001, represents the | |
important community-acquired methicillin resistant Staphylococcus | |
aureus lineage USA300. Findings: Using a hybrid assembly of | |
existing short read and barcoded long read sequences from | |
multiplexed data, we completed a genome of the S. aureus USA300 | |
strain MHO\_001. The long-read data represented only ∼5-10\% of | |
an average MinION(TM) run (∼7x genomic coverage), but, using | |
standard tools, this was sufficient to complete the circular | |
chromosome of S. aureus strain MHO\_001 (2.86 Mb) and two | |
complete plasmids (27 Kb and 3 Kb). Minor differences were noted | |
when compared to USA300 reference genome, USA300\_FPR3757, | |
including the translocation, loss and gain of mobile genetic | |
elements. Conclusion: Here we demonstrate that MinION(TM) reads, | |
multiplexed using native barcoding, can be used in combination | |
with short-read data, to fully complete a bacterial genome. The | |
ability to complete multiple genomes, for which short-read data | |
is already available, from a single MinION(TM) run is set to | |
impact on our understanding of accessory genome content, plasmid | |
diversity and genome rearrangements.", | |
journal = "Gigascience", | |
month = feb, | |
year = 2017, | |
keywords = "MinION; Staphylococcus aureus; Whole genome sequencing; bacterial | |
genomics; hybrid assembly; long read; multiplexing; native | |
barcoding;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Hau2017-cu, | |
title = "Draft Genome Sequences of 14 \textit{Staphylococcus aureus} | |
Sequence Type 5 Isolates from California, {USA}", | |
author = "Hau, Samantha J and Bayles, Darrell O and Alt, David P and | |
Nicholson, Tracy L", | |
abstract = "Staphylococcus aureus is part of the human epithelial microbiota; | |
however, it is also a pathogen. The acquisition of mobile genetic | |
elements plays a role in the virulence of S. aureus isolates and | |
contributes to treatment failures. This report details the draft | |
genome sequences of 14 clinical S. aureus isolates.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 13, | |
month = mar, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Hau2017-yd, | |
title = "Complete Genome Sequences of Two \textit{Staphylococcus aureus} | |
Sequence Type 5 Isolates from California, {USA}", | |
author = "Hau, Samantha J and Bayles, Darrell O and Alt, David P and | |
Nicholson, Tracy L", | |
abstract = "Staphylococcus aureus causes a variety of human diseases ranging | |
in severity. The pathogenicity of S. aureus can be partially | |
attributed to the acquisition of mobile genetic elements. In this | |
report, we provide two complete genome sequences from human | |
clinical S. aureus isolates.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 13, | |
month = mar, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Balachandran2017-ld, | |
title = "Complete Genome Sequence of \textit{Staphylococcus aureus} Strain | |
Wood 46", | |
author = "Balachandran, Manasi and Riley, Matthew C and Bemis, David A and | |
Kania, Stephen A", | |
abstract = "Here, we report the first complete genome sequence of the | |
Staphylococcus aureus strain Wood 46. Wood 46 has played an | |
important role in understanding the virulence and pathogenesis of | |
S. aureus infections. This report will assist efforts in vaccine | |
development against methicillin-resistant S. aureus (MRSA) | |
infections.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 13, | |
month = mar, | |
year = 2017, | |
keywords = "Staphylococcus;phage;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Lin2017-aq, | |
title = "Nucleotide Sequence Variations in Autolysis Genes of {ST} 59 | |
{Methicillin-Resistant} Staphylococcus aureus Isolates", | |
author = "Lin, Lee-Chung and Shu, Jwu-Ching and Chang, Shih-Cheng and Ge, | |
Mao-Cheng and Liu, Tsui-Ping and Chen, Chien-Wei and Lu, Jang-Jih", | |
abstract = "Biofilm formation is a virulence factor of bacteria. The goal of | |
this study was to understand the mechanisms of biofilm formation | |
by methicillin-resistant Staphylococcus aureus (MRSA). | |
Whole-genome sequencing of eight MRSA strains was performed to | |
identify sequence variations in genes related to biofilm | |
formation. Thirty-one genes involved in MRSA biofilm formation | |
were analyzed and 11 amino acid sequence variations in four genes | |
related to autolysis were found. These variations include E121D | |
and H387 N in ArlS; Q117K, T424S, K428T, A509S, V752E, A754V, and | |
T771A in Atl; T184K in CidC; and D251N in CidR. Among the 26 | |
clinical MRSA isolates studied, 13 isolates were nonbiofilm | |
producers and were found to harbor these mutations. Furthermore, | |
all of these 13 isolates belonged to ST59. Ten of these 13 ST59 | |
isolates became able to produce biofilms when they were incubated | |
with extracellular DNA from MRSA N315. Results of this study | |
suggest that sequence variations in arlS, atl, cidC, and cidR | |
genes may render MRSA unable to produce biofilms. Further | |
investigations are needed to correlate these sequence variations | |
with the biofilm-forming ability of MRSA isolates.", | |
journal = "Microb. Drug Resist.", | |
month = mar, | |
year = 2017, | |
keywords = "MRSA; ST59; biofilm; | |
eDNA;Staphylococcus;phage;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Tosas_Auguet2016-co, | |
title = "Evidence for Community Transmission of {Community-Associated} but | |
Not {Health-Care-Associated} {Methicillin-Resistant} | |
Staphylococcus Aureus Strains Linked to Social and Material | |
Deprivation: Spatial Analysis of Cross-sectional Data", | |
author = "Tosas Auguet, Olga and Betley, Jason R and Stabler, Richard A and | |
Patel, Amita and Ioannou, Avgousta and Marbach, Helene and Hearn, | |
Pasco and Aryee, Anna and Goldenberg, Simon D and Otter, Jonathan | |
A and Desai, Nergish and Karadag, Tacim and Grundy, Chris and | |
Gaunt, Michael W and Cooper, Ben S and Edgeworth, Jonathan D and | |
Kypraios, Theodore", | |
abstract = "BACKGROUND: Identifying and tackling the social determinants of | |
infectious diseases has become a public health priority following | |
the recognition that individuals with lower socioeconomic status | |
are disproportionately affected by infectious diseases. In many | |
parts of the world, epidemiologically and genotypically defined | |
community-associated (CA) methicillin-resistant Staphylococcus | |
aureus (MRSA) strains have emerged to become frequent causes of | |
hospital infection. The aim of this study was to use spatial | |
models with adjustment for area-level hospital attendance to | |
determine the transmission niche of genotypically defined CA- and | |
health-care-associated (HA)-MRSA strains across a diverse region | |
of South East London and to explore a potential link between MRSA | |
carriage and markers of social and material deprivation. METHODS | |
AND FINDINGS: This study involved spatial analysis of | |
cross-sectional data linked with all MRSA isolates identified by | |
three National Health Service (NHS) microbiology laboratories | |
between 1 November 2011 and 29 February 2012. The cohort of | |
hospital-based NHS microbiology diagnostic services serves | |
867,254 usual residents in the Lambeth, Southwark, and Lewisham | |
boroughs in South East London, United Kingdom (UK). Isolates were | |
classified as HA- or CA-MRSA based on whole genome sequencing. | |
All MRSA cases identified over 4 mo within the three-borough | |
catchment area (n = 471) were mapped to small geographies and | |
linked to area-level aggregated socioeconomic and demographic | |
data. Disease mapping and ecological regression models were used | |
to infer the most likely transmission niches for each MRSA | |
genetic classification and to describe the spatial epidemiology | |
of MRSA in relation to social determinants. Specifically, we | |
aimed to identify demographic and socioeconomic population traits | |
that explain cross-area extra variation in HA- and CA-MRSA | |
relative risks following adjustment for hospital attendance data. | |
We explored the potential for associations with the English | |
Indices of Deprivation 2010 (including the Index of Multiple | |
Deprivation and several deprivation domains and subdomains) and | |
the 2011 England and Wales census demographic and socioeconomic | |
indicators (including numbers of households by deprivation | |
dimension) and indicators of population health. Both CA-and | |
HA-MRSA were associated with household deprivation (CA-MRSA | |
relative risk [RR]: 1.72 [1.03-2.94]; HA-MRSA RR: 1.57 | |
[1.06-2.33]), which was correlated with hospital attendance | |
(Pearson correlation coefficient [PCC] = 0.76). HA-MRSA was also | |
associated with poor health (RR: 1.10 [1.01-1.19]) and residence | |
in communal care homes (RR: 1.24 [1.12-1.37]), whereas CA-MRSA | |
was linked with household overcrowding (RR: 1.58 [1.04-2.41]) and | |
wider barriers, which represent a combined score for household | |
overcrowding, low income, and homelessness (RR: 1.76 | |
[1.16-2.70]). CA-MRSA was also associated with recent immigration | |
to the UK (RR: 1.77 [1.19-2.66]). For the area-level variation in | |
RR for CA-MRSA, 28.67\% was attributable to the spatial | |
arrangement of target geographies, compared with only 0.09\% for | |
HA-MRSA. An advantage to our study is that it provided a | |
representative sample of usual residents receiving care in the | |
catchment areas. A limitation is that relationships apparent in | |
aggregated data analyses cannot be assumed to operate at the | |
individual level. CONCLUSIONS: There was no evidence of community | |
transmission of HA-MRSA strains, implying that HA-MRSA cases | |
identified in the community originate from the hospital reservoir | |
and are maintained by frequent attendance at health care | |
facilities. In contrast, there was a high risk of CA-MRSA in | |
deprived areas linked with overcrowding, homelessness, low | |
income, and recent immigration to the UK, which was not | |
explainable by health care exposure. Furthermore, areas adjacent | |
to these deprived areas were themselves at greater risk of | |
CA-MRSA, indicating community transmission of CA-MRSA. This | |
ongoing community transmission could lead to CA-MRSA becoming the | |
dominant strain types carried by patients admitted to hospital, | |
particularly if successful hospital-based MRSA infection control | |
programmes are maintained. These results suggest that community | |
infection control programmes targeting transmission of CA-MRSA | |
will be required to control MRSA in both the community and | |
hospital. These epidemiological changes will also have | |
implications for effectiveness of risk-factor-based hospital | |
admission MRSA screening programmes.", | |
journal = "PLoS Med.", | |
volume = 13, | |
number = 1, | |
pages = "e1001944", | |
month = jan, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@UNPUBLISHED{Kanjilal2017-av, | |
title = "Increasing Antibiotic Susceptibility In Staphylococcus aureus In | |
Boston, Massachusetts, 2000-2014: An Observational Study", | |
author = "Kanjilal, Sanjat and Sater, Mohamad and Thayer, Maile and | |
Lagoudas, Georgia and Kim, Soohong and Blainey, Paul and Grad, | |
Yonatan", | |
abstract = "Background: Methicillin resistant S. aureus (MRSA) has been | |
declining over the past decade, but changes in S. aureus overall | |
and the implications for trends in antibiotic resistance remain | |
unclear. Objective: To determine whether the decline in rates of | |
infection by MRSA has been accompanied by changes in rates of | |
infection by methicillin susceptible, penicillin resistant S. | |
aureus (MSSA) and penicillin susceptible S. aureus (PSSA). We | |
test if these dynamics are associated with specific genetic | |
lineages and evaluate gains and losses of resistance at the | |
strain level. Methods: We conducted a 15 year retrospective | |
observational study at two tertiary care institutions in Boston, | |
MA of 31,589 adult inpatients with S. aureus infections. | |
Surveillance swabs and duplicate specimens were excluded. We also | |
sequenced a sample of contemporary isolates (n = 180) obtained | |
between January 2016 and July 2016. We determined changes in the | |
annual rates of infection per 1,000 inpatient admissions by S. | |
aureus subtype and in the annual mean antibiotic resistance by | |
subtype. We performed phylogenetic analysis to generate a | |
population structure and infer gain and loss of the genetic | |
determinants of resistance. Results: Of the 43,954 S. aureus | |
infections over the study period, 21,779 were MRSA, 17,565 MSSA | |
and 4,610 PSSA. After multivariate adjustment, annual rates of | |
infection by S. aureus declined from 2003 to 2014 by 2.9\% (95\% | |
CI, 1.6\%-4.3\%), attributable to an annual decline in MRSA of | |
9.1\% (95\% CI, 6.3\%-11.9\%) and in MSSA by 2.2\% (95\% CI, | |
0.4\%-4.0\%). PSSA increased over this time period by 4.6\% (95\% | |
CI, 3.0\%-6.3\%) annually. Resistance in S. aureus decreased from | |
2000 to 2014 by 0.86 antibiotics (95\% CI, 0.81-0.91). By | |
phylogenetic inference, 5/35 MSSA and 2/20 PSSA isolates in the | |
common MRSA lineages ST5/USA100 and ST8/USA300 arose from the | |
loss of genes conferring resistance. Conclusions and relevance: | |
At two large tertiary care centers in Boston, MA, S. aureus | |
infections have decreased in rate and have become more | |
susceptible to antibiotics, with a rise in PSSA making penicillin | |
an increasingly viable and important treatment option.", | |
journal = "bioRxiv", | |
pages = "125369", | |
month = apr, | |
year = 2017, | |
keywords = "Staphylococcus;antibiotics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{De_Carvalho2017-rz, | |
title = "Draft Genome Sequences of Two Clinical {Methicillin-Resistant} | |
\textit{Staphylococcus aureus} Strains Isolated from Healthy | |
Children in Brazil", | |
author = "de Carvalho, Suzi P and de Almeida, J{\'e}ssica B and de Freitas, | |
Leandro M and Guimaraes, Ana Marcia S and do Nascimento, | |
Na{\'\i}la C and Dos Santos, Andrea P and Messick, Joanne B and | |
Timenetsky, Jorge and Marques, Lucas M", | |
abstract = "We report here the draft genome sequences of two | |
community-associated methicillin-resistant Staphylococcus aureus | |
(CA-MRSA) strains, C18 and C80, isolated from healthy children | |
from day care centers. To our knowledge, these are the first | |
draft genome sequences of CA-MRSA ST398/CC398/SccmecV and CA-MRSA | |
ST5/CC5/SccmecIVa isolated from healthy children in Brazil.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 15, | |
month = apr, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Earls2017-dk, | |
title = "The recent emergence in hospitals of multidrug-resistant | |
community-associated sequence type 1 and spa type t127 | |
methicillin-resistant Staphylococcus aureus investigated by | |
whole-genome sequencing: Implications for screening", | |
author = "Earls, Megan R and Kinnevey, Peter M and Brennan, Gr{\'a}inne I | |
and Lazaris, Alexandros and Skally, Mairead and O'Connell, Brian | |
and Humphreys, Hilary and Shore, Anna C and Coleman, David C", | |
abstract = "Community-associated spa type t127/t922 methicillin-resistant | |
Staphylococcus aureus (MRSA) prevalence increased from 1\%-7\% in | |
Ireland between 2010-2015. This study tracked the spread of 89 | |
such isolates from June 2013-June 2016. These included 78 | |
healthcare-associated and 11 community associated-MRSA isolates | |
from a prolonged hospital outbreak (H1) (n = 46), 16 other | |
hospitals (n = 28), four other healthcare facilities (n = 4) and | |
community-associated sources (n = 11). Isolates underwent | |
antimicrobial susceptibility testing, DNA microarray profiling | |
and whole-genome sequencing. Minimum spanning trees were | |
generated following core-genome multilocus sequence typing and | |
pairwise single nucleotide variation (SNV) analysis was | |
performed. All isolates were sequence type 1 MRSA staphylococcal | |
cassette chromosome mec type IV (ST1-MRSA-IV) and 76/89 were | |
multidrug-resistant. Fifty isolates, including 40/46 from H1, | |
were high-level mupirocin-resistant, carrying a conjugative 39 kb | |
iles2-encoding plasmid. Two closely related ST1-MRSA-IV strains | |
(I and II) and multiple sporadic strains were identified. Strain | |
I isolates (57/89), including 43/46 H1 and all high-level | |
mupirocin-resistant isolates, exhibited $\leq$80 SNVs. Two strain | |
I isolates from separate H1 healthcare workers differed from | |
other H1/strain I isolates by 7-47 and 12-53 SNVs, respectively, | |
indicating healthcare worker involvement in this outbreak. Strain | |
II isolates (19/89), including the remaining H1 isolates, | |
exhibited $\leq$127 SNVs. For each strain, the pairwise SNVs | |
exhibited by healthcare-associated and community-associated | |
isolates indicated recent transmission of ST1-MRSA-IV within and | |
between multiple hospitals, healthcare facilities and communities | |
in Ireland. Given the interchange between healthcare-associated | |
and community-associated isolates in hospitals, the risk factors | |
that inform screening for MRSA require revision.", | |
journal = "PLoS One", | |
volume = 12, | |
number = 4, | |
pages = "e0175542", | |
month = apr, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Rolo2017-tl, | |
title = "Evidence for the evolutionary steps leading to mecA-mediated | |
$\beta$-lactam resistance in staphylococci", | |
author = "Rolo, Joana and Worning, Peder and Boye Nielsen, Jesper and | |
Sobral, Rita and Bowden, Rory and Bouchami, Ons and Damborg, | |
Peter and Guardabassi, Luca and Perreten, Vincent and Westh, | |
Henrik and Tomasz, Alexander and de Lencastre, Herm{\'\i}nia and | |
Miragaia, Maria", | |
abstract = "The epidemiologically most important mechanism of antibiotic | |
resistance in Staphylococcus aureus is associated with mecA-an | |
acquired gene encoding an extra penicillin-binding protein | |
(PBP2a) with low affinity to virtually all $\beta$-lactams. The | |
introduction of mecA into the S. aureus chromosome has led to the | |
emergence of methicillin-resistant S. aureus (MRSA) pandemics, | |
responsible for high rates of mortality worldwide. Nonetheless, | |
little is known regarding the origin and evolution of mecA. | |
Different mecA homologues have been identified in species | |
belonging to the Staphylococcus sciuri group representing the | |
most primitive staphylococci. In this study we aimed to identify | |
evolutionary steps linking these mecA precursors to the | |
$\beta$-lactam resistance gene mecA and the resistance phenotype. | |
We sequenced genomes of 106 S. sciuri, S. vitulinus and S. | |
fleurettii strains and determined their oxacillin susceptibility | |
profiles. Single-nucleotide polymorphism (SNP) analysis of the | |
core genome was performed to assess the genetic relatedness of | |
the isolates. Phylogenetic analysis of the mecA gene homologues | |
and promoters was achieved through nucleotide/amino acid sequence | |
alignments and mutation rates were estimated using a Bayesian | |
analysis. Furthermore, the predicted structure of mecA | |
homologue-encoded PBPs of oxacillin-susceptible and -resistant | |
strains were compared. We showed for the first time that | |
oxacillin resistance in the S. sciuri group has emerged multiple | |
times and by a variety of different mechanisms. Development of | |
resistance occurred through several steps including structural | |
diversification of the non-binding domain of native PBPs; changes | |
in the promoters of mecA homologues; acquisition of SCCmec and | |
adaptation of the bacterial genetic background. Moreover, our | |
results suggest that it was exposure to $\beta$-lactams in | |
human-created environments that has driven evolution of native | |
PBPs towards a resistance determinant. The evolution of | |
$\beta$-lactam resistance in staphylococci highlights the | |
numerous resources available to bacteria to adapt to the | |
selective pressure of antibiotics.", | |
journal = "PLoS Genet.", | |
volume = 13, | |
number = 4, | |
pages = "e1006674", | |
month = apr, | |
year = 2017, | |
keywords = "Staphylococcus;evolution;antibiotics;oxacillin;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{De_Almeida2017-qf, | |
title = "Draft Genome Sequence of {Methicillin-Resistant} | |
\textit{Staphylococcus aureus} Strain {LC33} Isolated from Human | |
Breast Milk", | |
author = "de Almeida, J{\'e}ssica B and de Carvalho, Suzi P and de Freitas, | |
Leandro M and Guimar{\~a}es, Ana Marcia S and do Nascimento, | |
Na{\'\i}la C and Dos Santos, Andrea P and Messick, Joanne B and | |
Timenetsky, Jorge and Marques, Lucas M", | |
abstract = "Here, we report the draft genome sequence of Staphylococcus | |
aureus strain LC33, isolated from human breast milk in Brazil. | |
This microorganism has been typed as ST1/t127/sccmecV. To our | |
knowledge, this is the first draft genome sequence of a | |
methicillin-resistant S. aureus strain isolated from human breast | |
milk.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 15, | |
month = apr, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Cunningham2017-vi, | |
title = "Comparison of Two {Whole-Genome} Sequencing Methods for Analysis | |
of Three {Methicillin-Resistant} \textit{Staphylococcus aureus} | |
Outbreaks", | |
author = "Cunningham, Scott A and Chia, Nicholas and Jeraldo, Patricio R | |
and Quest, Daniel J and Johnson, Julie A and Boxrud, Dave J and | |
Taylor, Angela J and Chen, Jun and Jenkins, Gregory D and | |
Drucker, Travis M and Nelson, Heidi and Patel, Robin", | |
abstract = "Whole genome sequencing (WGS) can provide excellent resolution in | |
global and local epidemiological investigations of Staphylococcus | |
aureus outbreaks. A variety of sequencing approaches and | |
analytical tools have been used; it is not clear which is ideal. | |
We compared two WGS strategies and two analytical approaches to | |
the standard method of SmaI restriction digestion pulsed-field | |
gel electrophoresis (PFGE) for typing S. aureus Forty-two S. | |
aureus isolates from three outbreaks and 12 reference isolates | |
were studied. Near-complete genomes, assembled de novo with | |
paired-end and long mate pair (8 Kb) libraries were first | |
assembled and analyzed utilizing an in-house assembly and | |
analytical informatics pipeline. In addition, paired-end data was | |
assembled and analyzed using a commercial software package. | |
Single nucleotide variant (SNP) analysis was performed using the | |
in-house pipeline. Two assembly strategies were used to generate | |
core genome multi-locus sequence typing (cgMLST) data. First, the | |
near-complete genome data generated with the in-house pipeline | |
was imported into the commercial software and used to perform | |
cgMLST analysis. Second, the commercial software was used to | |
assemble paired-end data and resolved assemblies were used to | |
perform cgMLST. Similar isolate clustering was observed using SNP | |
calling and cgMLST, regardless of data assembly strategy. All | |
methods provided more discrimination between outbreaks than did | |
PFGE. Overall, all of the evaluated WGS strategies yielded | |
statistically similar results for S. aureus typing.", | |
journal = "J. Clin. Microbiol.", | |
month = apr, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Millar2017-sn, | |
title = "Genomic Characterization of {USA300} {MRSA} to Evaluate | |
Intraclass Transmission and Recurrence of {SSTI} among High Risk | |
Military Trainees", | |
author = "Millar, Eugene V and Rice, Gregory K and Elassal, Emad M and | |
Schlett, Carey D and Bennett, Jason W and Redden, Cassie L and | |
Mor, Deepika and Law, Natasha N and Tribble, David R and | |
Hamilton, Theron and Ellis, Michael W and Bishop-Lilly, Kimberly | |
A", | |
abstract = "Background.: Military trainees are at increased risk for | |
methicillin-resistant Staphylococcus aureus (MRSA) skin and soft | |
tissue infection (SSTI). Whole genome sequencing (WGS) can refine | |
our understanding of MRSA transmission and microevolution in | |
congregate settings. Methods.: We conducted a prospective | |
case-control study of SSTI among US Army Infantry trainees at | |
Fort Benning, GA from July 2012-December 2014. We identified | |
clusters of USA300 MRSA SSTI within select training classes and | |
performed WGS on clinical isolates. We then linked genomic, | |
phylogenetic data, epidemiologic, and clinical data in order to | |
evaluate intra- and interclass disease transmission. Furthermore, | |
among cases of recurrent MRSA SSTI, we evaluated the intrahost | |
relatedness of infecting strains. Results.: Nine training classes | |
with $\geq$5 cases of USA300 MRSA SSTI were selected. Eighty | |
USA300 MRSA clinical isolates from 74 trainees, six (8.1\%) of | |
whom had recurrent infection, were subjected to WGS. We | |
identified 2719 single nucleotide variants (SNV). The overall | |
median (range) SNV difference between isolates was 173 (1-339). | |
Intraclass median SNV differences ranged from 23 to 245. Two | |
phylogenetic clusters were suggestive of interclass MRSA | |
transmission. One of these clusters stemmed from two classes that | |
were separated by a 13-month period but housed in the same | |
barracks. Among trainees with recurrent MRSA SSTI, the intrahost | |
median SNV difference was 7.5 (1-48). Conclusions.: Application | |
of WGS revealed intra- and interclass transmission of MRSA among | |
military trainees. An interclass cluster between two | |
non-contemporaneous classes suggests a long-term reservoir for | |
MRSA in this setting.", | |
journal = "Clin. Infect. Dis.", | |
month = apr, | |
year = 2017, | |
keywords = "Methicillin-resistant Staphylococcus aureus (MRSA); genomic | |
epidemiology; military; skin and soft tissue infection; whole | |
genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Moon2016-be, | |
title = "Draft Genome Sequences of a Unique {t324-ST541-V} | |
{Methicillin-Resistant} Staphylococcus aureus Strain from a Pig", | |
author = "Moon, Dong Chan and Kim, Byung-Yong and Nam, Hyang-Mi and Jang, | |
Geum-Chan and Jung, Suk-Chan and Lee, Hee-Soo and Park, Yong-Ho | |
and Lim, Suk-Kyung", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA), the major | |
causative agent of nosocomial infection, has also been reported | |
from non-human sources. A sequence type (ST) 541 MRSA isolate | |
designated K12PJN53 was isolated from a healthy pig in 2012. The | |
genome of K12PJN53 consists of 44 contiguous sequences (contigs), | |
totalling 2,880,108 bases with 32.88\% GC content. Among the | |
annotated contigs, 14, 17, and 18 contained genes related to | |
antimicrobial resistance, adherence, and toxin genes, | |
respectively. The genomic distance of strain K12PJN53 was close | |
to the ST398 strains. This is the first report of the draft | |
genome sequence of a novel livestock-associated MRSA ST541 | |
strain.", | |
journal = "J. Microbiol. Biotechnol.", | |
volume = 26, | |
number = 4, | |
pages = "799--805", | |
month = apr, | |
year = 2016, | |
keywords = "Genomics; MRSA; | |
Pig;Staphylococcus;antibiotics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Gordon2017-um, | |
title = "Whole Genome Sequencing reveals the contribution of long-term | |
carriers in Staphylococcus aureus outbreak investigation", | |
author = "Gordon, N C and Pichon, B and Golubchik, T and Wilson, D J and | |
Paul, J and Blanc, D S and Cole, K and Collins, J and Cortes, N | |
and Cubbon, M and Gould, F K and Jenks, P J and Llewelyn, M and | |
Nash, J Q and Orendi, J M and Paranthaman, K and Price, J and | |
Senn, L and Thomas, H L and Wyllie, S and Crook, D W and Peto, T | |
E A and Walker, A S and Kearns, A M", | |
abstract = "Whole genome sequencing (WGS) makes it possible to determine the | |
relatedness of bacterial isolates at high resolution, helping to | |
characterise outbreaks. However, for Staphylococcus aureus, | |
accumulation of within-host diversity during carriage might limit | |
interpretation of sequencing data.In this study, we hypothesised | |
the converse: that within-host diversity can in fact be exploited | |
to reveal the involvement of long-term carriers (LTCs) in | |
outbreaks. We analysed WGS data from 20 historical outbreaks, and | |
applied phylogenetic methods to assess genetic relatedness and | |
estimate time to most recent common ancestor (TMRCA). Findings | |
were compared with the routine investigation results and | |
epidemiological evidence.Outbreaks with epidemiological evidence | |
for an LTC source had a mean estimated TMRCA (adjusted for | |
outbreak duration) of 243 days (95\% CI 143-343), compared with | |
55 days (28-81) for outbreaks lacking epidemiological evidence | |
for an LTC (p=0.004). A threshold of 156 days predicted LTC | |
involvement with a sensitivity of 0.875 and a specificity of 1.We | |
also found 6/20 outbreaks included isolates with differing | |
antimicrobial susceptibility profiles, however, these had only | |
modestly increased pairwise diversity (mean 17.5 single | |
nucleotide variants (SNVs) (95\% CI 17.3-17.8) vs 12.7 SNVs | |
(12.5-12.8)) compared with isolates with identical antibiograms | |
(p<0.0001). Additionally, for 2 outbreaks, WGS identified 1 or | |
more isolates which were genetically distinct despite having the | |
outbreak PFGE pulsotype.Duration-adjusted TMRCA allowed the | |
involvement of LTCs in outbreaks to be identified and could be | |
used to decide whether screening for long-term carriage (e.g. in | |
healthcare workers) is warranted. Requiring identical | |
antibiograms to trigger investigation could miss important | |
contributors to outbreaks.", | |
journal = "J. Clin. Microbiol.", | |
month = may, | |
year = 2017, | |
keywords = "Staphylococcus;transmission-epidemiology;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Ankrum2017-pp, | |
title = "Population dynamics of \textit{Staphylococcus aureus} in Cystic | |
Fibrosis patients to determine transmission events utilizing | |
{WGS}", | |
author = "Ankrum, Andrea and Hall, Barry G", | |
abstract = "Strict infection control practices have been implemented for | |
healthcare visits by Cystic Fibrosis patients in an attempt to | |
prevent transmission of important pathogens. This study used | |
whole genome sequencing (WGS) to determine strain relatedness and | |
assess population dynamics of Staphylococcus aureus isolates from | |
a cohort of CF patients as assessed by strain relatedness. 311 S. | |
aureus isolates were collected from respiratory cultures of 115 | |
CF patients during a 22 month study period. Whole genome | |
sequencing was performed and using SNP analysis, phylogenetic | |
trees were assembled to determine relatedness between isolates. | |
MRSA phenotypes were predicted using PPFS2 and compared to the | |
observed phenotype. The accumulation of SNPs in multiple isolates | |
obtained over time from the same patient was examined to | |
determine if a genomic molecular clock could be calculated.Pairs | |
of isolates with $\leq$ 71 SNP differences were considered to be | |
the ``same'' strain. All of the ``same'' strain isolates were | |
either from the same patient or siblings pairs. There were 47 | |
examples of patients being superinfected with an unrelated | |
strain. Predicted MRSA phenotype was accurate in all but three | |
isolates. Mutation rates were unable to be determined because the | |
branching order in the phylogenetic tree was inconsistent with | |
the order of isolation.The observation that transmissions were | |
identified between sibling patients shows that WGS is an | |
effective tool for determining transmission between patients. The | |
observation that transmission only occurred between siblings | |
suggests that Staphylococcus aureus acquisition in our CF | |
population occurred outside the hospital environment and | |
indicates that current infection prevention efforts appear | |
effective.", | |
journal = "J. Clin. Microbiol.", | |
month = apr, | |
year = 2017, | |
keywords = "bacerial\_genetics;Staphylococcus;bacterial comparative | |
genomics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Mekonnen2017-oe, | |
title = "Signatures of cytoplasmic proteins in the exoproteome distinguish | |
community- and hospital-associated methicillin-resistant | |
Staphylococcus aureus {USA300} lineages", | |
author = "Mekonnen, Solomon A and Palma Medina, Laura M and Glasner, | |
Corinna and Tsompanidou, Eleni and de Jong, Anne and Grasso, | |
Stefano and Schaffer, Marc and M{\"a}der, Ulrike and Larsen, | |
Anders R and Gumpert, Heidi and Westh, Henrik and V{\"o}lker, Uwe | |
and Otto, Andreas and Becher, D{\"o}rte and van Dijl, Jan Maarten", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) is the common | |
name for a heterogeneous group of highly drug-resistant | |
staphylococci. Two major MRSA classes are distinguished based on | |
epidemiology, namely community-associated (CA) and | |
hospital-associated (HA) MRSA. Notably, the distinction of CA- | |
and HA-MRSA based on molecular traits remains difficult due to | |
the high genomic plasticity of S. aureus. Here we sought to | |
pinpoint global distinguishing features of CA- and HA-MRSA | |
through a comparative genome and proteome analysis of the | |
notorious MRSA lineage USA300. We show for the first time that | |
CA- and HA-MRSA isolates can be distinguished by two distinct | |
extracellular protein abundance clusters that are predictive not | |
only for epidemiologic behavior, but also for their growth and | |
survival within epithelial cells. This 'exoproteome profiling' | |
also groups more distantly related HA-MRSA isolates into the HA | |
exoproteome cluster. Comparative genome analysis suggests that | |
these distinctive features of CA- and HA-MRSA isolates relate | |
predominantly to the accessory genome. Intriguingly, the | |
identified exoproteome clusters differ in the relative abundance | |
of typical cytoplasmic proteins, suggesting that signatures of | |
cytoplasmic proteins in the exoproteome represent a new | |
distinguishing feature of CA- and HA-MRSA. Our comparative genome | |
and proteome analysis focuses attention on potentially | |
distinctive roles of 'liberated' cytoplasmic proteins in the | |
epidemiology and intracellular survival of CA- and HA-MRSA | |
isolates. Such extracellular cytoplasmic proteins were recently | |
invoked in staphylococcal virulence, but their implication in the | |
epidemiology of MRSA is unprecedented.", | |
journal = "Virulence", | |
pages = "0", | |
month = may, | |
year = 2017, | |
keywords = "MRSA; Staphylococcus; USA300; community; epithelial cells; | |
exoproteome; hospital; moonlighting; protein secretion; virulence | |
factor;Staphylococcus;controversial;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Makarova2017-yd, | |
title = "Complete Genome Sequence of the {Livestock-Associated} | |
{Methicillin-Resistant} Strain \textit{Staphylococcus aureus} | |
subsp. \textit{aureus} {08S00974} (Sequence Type 398)", | |
author = "Makarova, Olga and Johnston, Paul and Walther, Birgit and Rolff, | |
Jens and Roesler, Uwe", | |
abstract = "We report here the complete genome sequence of the | |
livestock-associated methicillin-resistant Staphylococcus aureus | |
strain 08S00974 from sequence type 398 (ST398 LA-MRSA) isolated | |
from a fatting pig at a farm in Germany.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 19, | |
month = may, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Makarova2017-tq, | |
title = "Complete Genome Sequence of the Disinfectant Susceptibility | |
Testing Reference Strain \textit{Staphylococcus aureus} subsp. | |
\textit{aureus} {ATCC} 6538", | |
author = "Makarova, Olga and Johnston, Paul and Walther, Birgit and Rolff, | |
Jens and Roesler, Uwe", | |
abstract = "We report here the complete genome sequence of the | |
methicillin-sensitive Staphylococcus aureus subsp. aureus strain | |
ATCC 6538 (FDA 209, DSM 799, WDCM 00032, and NCTC 10788).", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 19, | |
month = may, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Tewhey2012-yx, | |
title = "Genetic structure of community acquired methicillin-resistant | |
Staphylococcus aureus {USA300}", | |
author = "Tewhey, Ryan and Cannavino, Christopher R and Leake, John A D and | |
Bansal, Vikas and Topol, Eric J and Torkamani, Ali and Bradley, | |
John S and Schork, Nicholas J", | |
abstract = "BACKGROUND: Community-associated methicillin-resistant | |
Staphylococcus aureus (CA-MRSA) is a significant bacterial | |
pathogen that poses considerable clinical and public health | |
challenges. The majority of the CA-MRSA disease burden consists | |
of skin and soft tissue infections (SSTI) not associated with | |
significant morbidity; however, CA-MRSA also causes severe, | |
invasive infections resulting in significant morbidity and | |
mortality. The broad range of disease severity may be influenced | |
by bacterial genetic variation. RESULTS: We sequenced the | |
complete genomes of 36 CA-MRSA clinical isolates from the | |
predominant North American community acquired clonal type USA300 | |
(18 SSTI and 18 severe infection-associated isolates). While all | |
36 isolates shared remarkable genetic similarity, we found | |
greater overall time-dependent sequence diversity among SSTI | |
isolates. In addition, pathway analysis of non-synonymous | |
variations revealed increased sequence diversity in the putative | |
virulence genes of SSTI isolates. CONCLUSIONS: Here we report the | |
first whole genome survey of diverse clinical isolates of the | |
USA300 lineage and describe the evolution of the pathogen over | |
time within a defined geographic area. The results demonstrate | |
the close relatedness of clinically independent CA-MRSA isolates, | |
which carry implications for understanding CA-MRSA epidemiology | |
and combating its spread.", | |
journal = "BMC Genomics", | |
volume = 13, | |
pages = "508", | |
month = sep, | |
year = 2012, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Larsen2017-ln, | |
title = "Emergence of livestock-associated methicillin-resistant | |
Staphylococcus aureus bloodstream infections in Denmark", | |
author = "Larsen, Jesper and Petersen, Andreas and Larsen, Anders R and | |
Sieber, Raphael N and Stegger, Marc and Koch, Anders and | |
Aarestrup, Frank M and Price, Lance B and Skov, Robert L", | |
abstract = "Livestock-associated methicillin-resistant Staphylococcus aureus | |
clonal complex 398 (LA-MRSA CC398) is causing an increasing | |
number of skin and soft tissue infections (SSTIs) in Denmark and | |
other European countries with industrial pig production. Yet, its | |
impact on MRSA bloodstream infections (BSIs) has not been well | |
studied.We investigated the clinical epidemiology of all human | |
cases of LA-MRSA CC398 BSI during 2010--2015. Cases of LA-MRSA | |
CC398 BSI were compared to cases of BSI caused by other types of | |
MRSA and cases of SSTI caused by LA-MRSA CC398. Whole-genome | |
sequence analysis was used to assess the phylogenetic | |
relationship among LA-MRSA CC398 isolates from Danish pigs and | |
cases of BSI and SSTI.The number of LA-MRSA CC398 BSIs and SSTIs | |
increased over the years, peaking in 2014, where LA-MRSA CC398 | |
accounted for 16\% (7/44) and 21\% (211/985) of all MRSA BSIs and | |
SSTIs, corresponding to 1.2 and 37.4 cases of BSI and SSTI per | |
1,000,000 person-years, respectively. Most patients with LA-MRSA | |
CC398 BSI had no contact to livestock, although they tended to | |
live in rural areas. LA-MRSA CC398 caused 24.3 BSIs per 1,000 | |
SSTIs among people with no livestock contact, which is similar to | |
the ratio observed for other types of MRSA. Whole-genome sequence | |
analysis showed that most of the BSI and SSTI isolates were | |
closely related to Danish pig isolates.This study demonstrates | |
that the increasing number of LA-MRSA CC398 BSIs occurred in | |
parallel with a much larger wave of LA-MRSA CC398 SSTIs and an | |
expanding pig reservoir.", | |
journal = "Clin. Infect. Dis.", | |
month = may, | |
year = 2017, | |
keywords = "denmark; methicillin-resistant staphylococcus aureus; skin and | |
soft tissue infections; animals, farm; bloodstream infection; | |
genome; sequence analysis; family suidae; | |
epidemiology;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Park2017-ym, | |
title = "Molecular epidemiology of Staphylococcus aureus bacteremia in a | |
single large Minnesota medical center in 2015 as assessed using | |
{MLST}, core genome {MLST} and spa typing", | |
author = "Park, Kyung-Hwa and Greenwood-Quaintance, Kerryl E and Uhl, James | |
R and Cunningham, Scott A and Chia, Nicholas and Jeraldo, | |
Patricio R and Sampathkumar, Priya and Nelson, Heidi and Patel, | |
Robin", | |
abstract = "Staphylococcus aureus is a leading cause of bacteremia in | |
hospitalized patients. Whether or not S. aureus bacteremia (SAB) | |
is associated with clonality, implicating potential nosocomial | |
transmission, has not, however, been investigated. Herein, we | |
examined the epidemiology of SAB using whole genome sequencing | |
(WGS). 152 SAB isolates collected over the course of 2015 at a | |
single large Minnesota medical center were studied. | |
Staphylococcus protein A (spa) typing was performed by PCR/Sanger | |
sequencing; multilocus sequence typing (MLST) and core genome | |
MLST (cgMLST) were determined by WGS. Forty-eight isolates (32\%) | |
were methicillin-resistant S. aureus (MRSA). The isolates | |
encompassed 66 spa types, clustered into 11 spa clonal complexes | |
(CCs) and 10 singleton types. 88\% of 48 MRSA isolates belonged | |
to spa CC-002 or -008. Methicillin-susceptible S. aureus (MSSA) | |
isolates were more genotypically diverse, with 61\% distributed | |
across four spa CCs (CC-002, CC-012, CC-008 and CC-084). By MLST, | |
there was 31 sequence types (STs), including 18 divided into 6 | |
CCs and 13 singleton STs. Amongst MSSA isolates, the common MLST | |
clones were CC5 (23\%), CC30 (19\%), CC8 (15\%) and CC15 (11\%). | |
Common MRSA clones were CC5 (67\%) and CC8 (25\%); there were no | |
MRSA isolates in CC45 or CC30. By cgMLST analysis, there were 9 | |
allelic differences between two isolates, with the remaining 150 | |
isolates differing from each other by over 40 alleles. The two | |
isolates were retroactively epidemiologically linked by medical | |
record review. Overall, cgMLST analysis resulted in higher | |
resolution epidemiological typing than did multilocus sequence or | |
spa typing.", | |
journal = "PLoS One", | |
volume = 12, | |
number = 6, | |
pages = "e0179003", | |
month = jun, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{McClure2017-cr, | |
title = "Complete Genome Sequence of a {Community-Associated} | |
{Methicillin-Resistant} Staphylococcus aureus Hypervirulent | |
Strain, {USA300-C2406}, Isolated from a Patient with a Lethal | |
Case of Necrotizing Pneumonia", | |
author = "McClure, Jo-Ann and Zhang, Kunyan", | |
abstract = "USA300 is a predominant community-associated | |
methicillin-resistant Staphylococcus aureus strain causing | |
significant morbidity and mortality. We present here the full | |
annotated genome of a USA300 hypervirulent clinical strain, | |
USA300-C2406, isolated from a patient with a lethal case of | |
necrotizing pneumonia, to gain a better understanding of USA300 | |
hypervirulence.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 22, | |
month = jun, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Sabrina_Pankey2017-cu, | |
title = "Host-selected mutations converging on a global regulator drive | |
an adaptive leap towards symbiosis in bacteria", | |
author = "Sabrina Pankey, M and Foxall, Randi L and Ster, Ian M and Perry, | |
Lauren A and Schuster, Brian M and Donner, Rachel A and Coyle, | |
Matthew and Cooper, Vaughn S and Whistler, Cheryl A", | |
abstract = "Most bacteria that associate with animals do not cause harm, and | |
many are essential to health or provide other benefits. An | |
animal's immune system must permit these beneficial associations | |
and at the same time block harmful microbes. This ultimately | |
means that even beneficial bacteria must adapt to the immune | |
barriers that they encounter. Different species that live in a | |
close relationship with each other are known as symbionts. A | |
species of bacteria called Vibrio fischeri can form a mutually | |
beneficial symbiotic relationship with squid. The squid provide | |
food for the bacteria, but only the bacteria that successfully | |
navigate immune barriers and reach the squid's ``light organ'' | |
are fed. In return, the bacteria produce bioluminescence, making | |
the nocturnal squid appear like moonlight in the water. As the | |
bacteria reproduce, some individuals randomly acquire genetic | |
mutations, some of which might improve the bacteria's chances of | |
survival. Which mutations and associated traits allow bacteria | |
to beat out the competition and evolve to become animal | |
symbionts? To investigate, Pankey, Foxall et al. grew V. | |
fischeri bacteria from several ancestors that were poor at | |
colonizing squid. Groups of newly hatched squid selected | |
potential symbionts from the resulting mix of bacteria. The | |
selected symbionts were allowed to reproduce within the squid to | |
form a new population of bacteria and were later vented out for | |
a new batch of squid to sort through. This was repeated to | |
ultimately form a final group of bacteria that had passed | |
through 15 squid in turn. Unexpectedly, the bacteria in the | |
final group all found the same solution to help them adapt to | |
symbiotic life with the squid: mutations to the gene that | |
encodes a signaling protein called BinK. Eight distinct | |
mutations arose that dramatically changed how the bacteria | |
interacted with squid. The evolved bacteria created a coating | |
that hid them from squid immune cells and protected them from | |
chemicals that squid use to kill invaders. The mutations also | |
altered how the bacteria communicated with each other. This | |
adjusted the intensity of light that they produced for their | |
host to a more natural level, and improved their ability to grow | |
on squid-provided food. Overall, the results presented by | |
Pankey, Foxall et al. demonstrate that small genetic mutations | |
can transform non-symbionts into symbionts, enabling them to | |
evolve rapidly to form a symbiosis with a new host. This | |
demonstrates that these bacteria already had the ability to | |
coordinate the complex behaviors necessary to overcome the | |
multiple barriers provided to them by the squid immune system. | |
Other beneficial animal--bacteria associations are likely to | |
work on similar principles; the study exemplifies the utility of | |
experimental evolution systems and lays a foundation for further | |
work to investigate these principles in more detail.", | |
journal = "eLife Sciences", | |
publisher = "eLife Sciences Publications Limited", | |
volume = 6, | |
pages = "e24414", | |
month = apr, | |
year = 2017, | |
keywords = "Vibrio fischeri; Euprymna scolopes; experimental | |
evolution;evolution;Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Tabuchi2017-hq, | |
title = "D-cycloserine increases the effectiveness of vancomycin against | |
vancomycin-highly resistant Staphylococcus aureus", | |
author = "Tabuchi, Fumiaki and Matsumoto, Yasuhiko and Ishii, Masaki and | |
Tatsuno, Keita and Okazaki, Mitsuhiro and Sato, Tomoaki and | |
Moriya, Kyoji and Sekimizu, Kazuhisa", | |
abstract = "Vancomycin is a widely used clinical drug to treat for infection | |
by methicillin-resistant Staphylococcus aureus. Some patients | |
show a weak response to vancomycin treatment. We previously | |
reported that $\beta$-lactams increase the susceptibility to | |
vancomycin by vancomycin-highly resistant S. aureus (VRSA) | |
strains obtained following repeated in vitro mutagenesis and | |
vancomycin selection. Here we found that the susceptibility of | |
the VRSA strains to vancomycin was remarkably increased by | |
combined treatment with D-cycloserine. On the other hand, VRSA | |
did not show increased susceptibility to vancomycin in | |
combination with bacitracin, fosfomycin, erythromycin, | |
lincomycin, gentamicin, levofloxacin or nisin. Furthermore, in an | |
in vivo infection model with silkworms, combined treatment with | |
vancomycin and D-cycloserine exhibited therapeutic effects, | |
whereas treatment with each compound alone did not. These | |
findings suggest that combined treatment with vancomycin and | |
D-cycloserine could be therapeutically effective against | |
infectious diseases caused by VRSA.The Journal of Antibiotics | |
advance online publication, 7 June 2017; doi:10.1038/ja.2017.56.", | |
journal = "J. Antibiot.", | |
month = jun, | |
year = 2017, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Ronco2017-yo, | |
title = "Draft Genome Sequence of a Sequence Type 398 | |
{Methicillin-Resistant} \textit{Staphylococcus aureus} Isolate | |
from a Danish Dairy Cow with Mastitis", | |
author = "Ronco, Troels and Stegger, Marc and Pedersen, Karl", | |
abstract = "Livestock-associated (LA) methicillin-resistant Staphylococcus | |
aureus (MRSA) strains of sequence type 398 (ST398) colonize both | |
humans and various livestock species. In 2016, an ST398 LA-MRSA | |
isolate (Sa52) was collected from a Danish dairy cow with | |
mastitis, and here, we report the draft genome sequence of strain | |
Sa52.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 23, | |
month = jun, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{McClure2017-aj, | |
title = "Complete Genome Sequences of Five Representative | |
\textit{Staphylococcus aureus} {ST398} Strains from Five Major | |
Sequence Heterogeneity Groups of a Diverse Isolate Collection", | |
author = "McClure, Jo-Ann and Zhang, Kunyan", | |
abstract = "Staphylococcus aureus sequence type 398 (ST398) is a rapidly | |
emerging livestock-associated strain causing zoonotic disease in | |
humans. The course of pathogen evolution remains unclear, | |
prompting whole-genome comparative studies in attempts to | |
elucidate this issue. We present the full, annotated genomes of | |
five newly isolated representative ST398 strains from five major | |
sequence heterogeneity groups of our diverse isolate collection.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 23, | |
month = jun, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Panthee2017-vo, | |
title = "Draft Genome Sequence of the {Vancomycin-Resistant} Clinical | |
Isolate \textit{Staphylococcus aureus} {VRS3b}", | |
author = "Panthee, Suresh and Paudel, Atmika and Hamamoto, Hiroshi and | |
Sekimizu, Kazuhisa", | |
abstract = "We report here the draft genome sequence of the | |
vancomycin-resistant strain Staphylococcus aureus VRS3b. The | |
2.8-Mb genome, assembled into 46 contigs, harbored 2,915 putative | |
coding sequences. The G+C content of the genome was 32.7\%.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 22, | |
month = jun, | |
year = 2017, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{McClure2017-aw, | |
title = "Complete Genome Sequence of the {Methicillin-Resistant} | |
\textit{Staphylococcus aureus} Colonizing Strain {M92}", | |
author = "McClure, Jo-Ann and Zhang, Kunyan", | |
abstract = "M92 is a methicillin-resistant Staphylococcus aureus (MRSA) | |
colonizing strain belonging to ST239-MRSA-III. It frequently | |
shows local nasal colonization in our hospital staff, but has | |
never been associated with infection. We sequenced the complete | |
genome of M92, in order to compare it to highly virulent MRSA | |
strains to gain insight into MRSA virulence factors.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 23, | |
month = jun, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Basco2017-pi, | |
title = "Draft Genome Sequences of Two \textit{Staphylococcus aureus} | |
Strains Isolated in Succession from a Case of Bacteremia", | |
author = "Basco, M D S and Revollo, J R and McKinzie, P B and Agnihothram, | |
S and Kothari, A and Saccente, M and Hart, M E", | |
abstract = "Staphylococcus aureus strains MEH1 and MEH7 were successively | |
isolated from the blood of a patient with recurrent bacteremia. | |
The submitted draft genomes of strains MEH1 and MEH7 are | |
2,914,972 and 2,911,704 bp, respectively.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 23, | |
month = jun, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Larsen2017-yf, | |
title = "Emergence of livestock-associated methicillin-resistant | |
Staphylococcus aureus bloodstream infections in Denmark", | |
author = "Larsen, Jesper and Petersen, Andreas and Larsen, Anders R and | |
Sieber, Raphael N and Stegger, Marc and Koch, Anders and | |
Aarestrup, Frank M and Price, Lance B and Skov, Robert L and | |
{Danish MRSA Study Group}", | |
abstract = "Background: Livestock-associated methicillin-resistant | |
Staphylococcus aureus clonal complex 398 (LA-MRSA CC398) is | |
causing an increasing number of skin and soft tissue infections | |
(SSTIs) in Denmark and other European countries with industrial | |
pig production. Yet, its impact on MRSA bloodstream infections | |
(BSIs) has not been well studied. Methods: We investigated the | |
clinical epidemiology of all human cases of LA-MRSA CC398 BSI | |
during 2010-2015. Cases of LA-MRSA CC398 BSI were compared to | |
cases of BSI caused by other types of MRSA and cases of SSTI | |
caused by LA-MRSA CC398. Whole-genome sequence analysis was used | |
to assess the phylogenetic relationship among LA-MRSA CC398 | |
isolates from Danish pigs and cases of BSI and SSTI. Results: The | |
number of LA-MRSA CC398 BSIs and SSTIs increased over the years, | |
peaking in 2014, where LA-MRSA CC398 accounted for 16\% (7/44) | |
and 21\% (211/985) of all MRSA BSIs and SSTIs, corresponding to | |
1.2 and 37.4 cases of BSI and SSTI per 1,000,000 person-years, | |
respectively. Most patients with LA-MRSA CC398 BSI had no contact | |
to livestock, although they tended to live in rural areas. | |
LA-MRSA CC398 caused 24.3 BSIs per 1,000 SSTIs among people with | |
no livestock contact, which is similar to the ratio observed for | |
other types of MRSA. Whole-genome sequence analysis showed that | |
most of the BSI and SSTI isolates were closely related to Danish | |
pig isolates. Conclusions: This study demonstrates that the | |
increasing number of LA-MRSA CC398 BSIs occurred in parallel with | |
a much larger wave of LA-MRSA CC398 SSTIs and an expanding pig | |
reservoir.", | |
journal = "Clin. Infect. Dis.", | |
month = may, | |
year = 2017, | |
keywords = "MRSA; bacteremia.; humans; livestock; | |
zoonosis;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Larsen2017-yf, | |
title = "Emergence of livestock-associated methicillin-resistant | |
Staphylococcus aureus bloodstream infections in Denmark", | |
author = "Larsen, Jesper and Petersen, Andreas and Larsen, Anders R and | |
Sieber, Raphael N and Stegger, Marc and Koch, Anders and | |
Aarestrup, Frank M and Price, Lance B and Skov, Robert L and | |
{Danish MRSA Study Group}", | |
abstract = "Background: Livestock-associated methicillin-resistant | |
Staphylococcus aureus clonal complex 398 (LA-MRSA CC398) is | |
causing an increasing number of skin and soft tissue infections | |
(SSTIs) in Denmark and other European countries with industrial | |
pig production. Yet, its impact on MRSA bloodstream infections | |
(BSIs) has not been well studied. Methods: We investigated the | |
clinical epidemiology of all human cases of LA-MRSA CC398 BSI | |
during 2010-2015. Cases of LA-MRSA CC398 BSI were compared to | |
cases of BSI caused by other types of MRSA and cases of SSTI | |
caused by LA-MRSA CC398. Whole-genome sequence analysis was used | |
to assess the phylogenetic relationship among LA-MRSA CC398 | |
isolates from Danish pigs and cases of BSI and SSTI. Results: The | |
number of LA-MRSA CC398 BSIs and SSTIs increased over the years, | |
peaking in 2014, where LA-MRSA CC398 accounted for 16\% (7/44) | |
and 21\% (211/985) of all MRSA BSIs and SSTIs, corresponding to | |
1.2 and 37.4 cases of BSI and SSTI per 1,000,000 person-years, | |
respectively. Most patients with LA-MRSA CC398 BSI had no contact | |
to livestock, although they tended to live in rural areas. | |
LA-MRSA CC398 caused 24.3 BSIs per 1,000 SSTIs among people with | |
no livestock contact, which is similar to the ratio observed for | |
other types of MRSA. Whole-genome sequence analysis showed that | |
most of the BSI and SSTI isolates were closely related to Danish | |
pig isolates. Conclusions: This study demonstrates that the | |
increasing number of LA-MRSA CC398 BSIs occurred in parallel with | |
a much larger wave of LA-MRSA CC398 SSTIs and an expanding pig | |
reservoir.", | |
journal = "Clin. Infect. Dis.", | |
month = may, | |
year = 2017, | |
keywords = "denmark; methicillin-resistant staphylococcus aureus; skin and | |
soft tissue infections; animals, farm; bloodstream infection; | |
genome; sequence analysis; family suidae; | |
epidemiology;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Feng2017-gr, | |
title = "Genome comparisons of two Taiwanese community-associated | |
methicillin-resistant Staphylococcus aureus {ST59} clones support | |
the multi-origin theory of {CA-MRSA}", | |
author = "Feng, Ye and Chen, Hsiu-Ling and Chen, Chih-Jung and Chen, | |
Chyi-Liang and Chiu, Cheng-Hsun", | |
abstract = "Sequence type (ST) 59 is an epidemic lineage of | |
community-associated methicillin-resistant Staphylococcus aureus | |
(CA-MRSA) in Asia. Two ST59 clones are prevalent in Taiwan: the | |
Taiwan clone (TW) causes severe infections, whereas the | |
Asian-Pacific clone (AP) is usually commensal. In this study, we | |
sequenced the genome and transcriptome of the representative | |
strains of these two clones and found their differences to focus | |
on three mobile genetic elements: TW carries SCCmec Type VT, | |
Panton-Valentine leucocidin (PVL)-encoding prophage $\Phi$Sa2, | |
whereas AP carries SCCmec Type IV and staphylokinase | |
(SAK)-encoding prophage $\Phi$Sa3. The anti-virulent role of SAK | |
was confirmed using murine skin and bloodstream infection models. | |
$\Phi$Sa3 usually integrates into the hlb gene, but in AP was | |
found to be integrated at the genomic island $\nu$Sa$\beta$. The | |
mutation of the attB site ``TGTATCCAAACTGG'' to | |
``TGTATCCGAATTGG'' led to a failure in the integration of | |
$\Phi$Sa3 in hlb, prompting atypical integration at other sites. | |
The sak gene possessed remarkably different patterns of | |
distribution among the different STs of S. aureus. We conclude | |
that the atypical integration of $\Phi$Sa3 may help S. aureus | |
adapt to the human host habitat and that the subsequent loss of | |
$\Phi$Sa3 contributes toward the development of a virulent | |
CA-MRSA lineage for wider horizontal transmission.", | |
journal = "Infect. Genet. Evol.", | |
volume = 54, | |
pages = "60--65", | |
month = jun, | |
year = 2017, | |
keywords = "Anti-virulence factor; Bacteriophage; CA-MRSA; Clone; | |
Staphylokinase;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Edslev2017-oi, | |
title = "Identification of a {PVL-negative} {SCCmec-IVa} sub-lineage of | |
the methicillin-resistant Staphylococcus aureus {CC80} lineage: | |
Understanding the clonal origin of {CA-MRSA}", | |
author = "Edslev, Sofie Marie and Westh, Henrik Torkil and Andersen, Paal | |
Skytt and Skov, Robert and Kobayashi, Nobumichi and Bartels, | |
Mette Damkj{\ae}r and Vandenesh, Francois and Petersen, Andreas | |
and Worning, Peder and Larsen, Anders Rhod and Stegger, Marc", | |
abstract = "OBJECTIVES: Community-acquired MRSA (CA-MRSA) isolates belonging | |
to clonal complex 80 (CC80) are recognized as the European | |
CA-MRSA. The prevailing European CA-MRSA clone carries a type IVc | |
staphylococcal cassette chromosome mec (SCCmec) and expresses | |
Panton-Valentine leukocidin (PVL). Recently, a significant | |
increase of PVL-negative CC80 MRSA has been observed in Denmark. | |
The aim of this study was to examine the genetics and the | |
epidemiology of these, and to compare them to the European | |
CA-MRSA clone in order to understand the emergence of | |
PVL-negative CC80 MRSA. METHODS: Phylogenetic analysis of the | |
CC80 S. aureus lineage was conducted from whole-genome sequences | |
of 217 isolates (23 MSSA and 194 MRSA) from 22 countries. All | |
isolates were further genetically characterized in regard to | |
resistance determinants and PVL carriage, and epidemiological | |
data was obtained for selected isolates. RESULTS: The | |
phylogenetic analysis revealed the existence of three distinct | |
clades of the CC80 lineage: i) an MSSA clade encompassing | |
Sub-Saharan African isolates (n=13); ii) a derived clade | |
encompassing the European CA-MRSA SCCmec-IVc clone (n=185); and | |
iii) a novel and genetically distinct clade encompassing MRSA | |
SCCmec-IVa isolates (n=19). All isolates in the novel clade were | |
PVL negative, but carried remnant parts (8 -12 kb) of the | |
PVL-encoding prophage $\Phi$Sa2, and were susceptible to fusidic | |
acid and kanamycin/amikacin. Geospatial mapping could link these | |
isolates to regions in the Middle East, Asia and South Pacific. | |
CONCLUSIONS: This study reports the emergence of a novel CC80 | |
CA-MRSA sub-lineage, showing that the CC80 lineage is more | |
diverse than previously assumed.", | |
journal = "Clin. Microbiol. Infect.", | |
month = jun, | |
year = 2017, | |
keywords = "Antibiotic resistance; CA-MRSA; Epidemiology; Evolution; | |
Genetics; Phylogeny; | |
SCCmec;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Byrd2017-yt, | |
title = "Staphylococcus aureus and Staphylococcus epidermidis strain | |
diversity underlying pediatric atopic dermatitis", | |
author = "Byrd, Allyson L and Deming, Clay and Cassidy, Sara K B and | |
Harrison, Oliver J and Ng, Weng-Ian and Conlan, Sean and | |
Program, Nisc Comparative Sequencing and Belkaid, Yasmine and | |
Segre, Julia A and Kong, Heidi H", | |
abstract = "The heterogeneous course, severity, and treatment responses | |
among patients with atopic dermatitis (AD; eczema) highlight the | |
complexity of this multifactorial disease. Prior studies have | |
used traditional typing methods on cultivated isolates or | |
sequenced a bacterial marker gene to study the skin microbial | |
communities of AD patients. Shotgun metagenomic sequence | |
analysis provides much greater resolution, elucidating multiple | |
levels of microbial community assembly ranging from kingdom to | |
species and strain-level diversification. We analyzed microbial | |
temporal dynamics from a cohort of pediatric AD patients sampled | |
throughout the disease course. Species-level investigation of AD | |
flares showed greater Staphylococcus aureus predominance in | |
patients with more severe disease and Staphylococcus epidermidis | |
predominance in patients with less severe disease. At the strain | |
level, metagenomic sequencing analyses demonstrated clonal S. | |
aureus strains in more severe patients and heterogeneous S. | |
epidermidis strain communities in all patients. To investigate | |
strain-level biological effects of S. aureus, we topically | |
colonized mice with human strains isolated from AD patients and | |
controls. This cutaneous colonization model demonstrated S. | |
aureus strain--specific differences in eliciting skin | |
inflammation and immune signatures characteristic of AD | |
patients. Specifically, S. aureus isolates from AD patients with | |
more severe flares induced epidermal thickening and expansion of | |
cutaneous T helper 2 (TH2) and TH17 cells. Integrating | |
high-resolution sequencing, culturing, and animal models | |
demonstrated how functional differences of staphylococcal | |
strains may contribute to the complexity of AD disease.", | |
journal = "Sci. Transl. Med.", | |
publisher = "American Association for the Advancement of Science", | |
volume = 9, | |
number = 397, | |
pages = "eaal4651", | |
month = jul, | |
year = 2017, | |
keywords = "Staphylococcus;immunology-immunogenomics;fungus;grants;Staphylococcus\_aureus\_genome\_papers;skin\_shotgun\_microbiome", | |
language = "en" | |
} | |
@ARTICLE{Moradigaravand2017-mi, | |
title = "Evolution of the \textit{Staphylococcus argenteus} {ST2250} Clone | |
in Northeastern Thailand Is Linked with the Acquisition of | |
{Livestock-Associated} Staphylococcal Genes", | |
author = "Moradigaravand, Danesh and Jamrozy, Dorota and Mostowy, Rafal and | |
Anderson, Annaliesa and Nickerson, Emma K and Thaipadungpanit, | |
Janjira and Wuthiekanun, Vanaporn and Limmathurotsakul, Direk and | |
Tandhavanant, Sarunporn and Wikraiphat, Chanthiwa and Wongsuvan, | |
Gumphol and Teerawattanasook, Nittaya and Jutrakul, Yaowaruk and | |
Srisurat, Nuttiya and Chaimanee, Prajuab and Eoin West, T and | |
Blane, Beth and Parkhill, Julian and Chantratita, Narisara and | |
Peacock, Sharon J", | |
abstract = "Staphylococcus argenteus is a newly named species previously | |
described as a divergent lineage of Staphylococcus aureus that | |
has recently been shown to have a global distribution. Despite | |
growing evidence of the clinical importance of this species, | |
knowledge about its population epidemiology and genomic | |
architecture is limited. We used whole-genome sequencing to | |
evaluate and compare S. aureus (n = 251) and S. argenteus (n = | |
68) isolates from adults with staphylococcal sepsis at several | |
hospitals in northeastern Thailand between 2006 and 2013. The | |
majority (82\%) of the S. argenteus isolates were of multilocus | |
sequence type 2250 (ST2250). S. aureus was more diverse, although | |
43\% of the isolates belonged to ST121. Bayesian analysis | |
suggested an S. argenteus ST2250 substitution rate of 4.66 (95\% | |
confidence interval [CI], 3.12 to 6.38) mutations per genome per | |
year, which was comparable to the S. aureus ST121 substitution | |
rate of 4.07 (95\% CI, 2.61 to 5.55). S. argenteus ST2250 emerged | |
in Thailand an estimated 15 years ago, which contrasts with the | |
S. aureus ST1, ST88, and ST121 clades that emerged around 100 to | |
150 years ago. Comparison of S. argenteus ST2250 genomes from | |
Thailand and a global collection indicated a single introduction | |
into Thailand, followed by transmission to local and more distant | |
countries in Southeast Asia and further afield. S. argenteus and | |
S. aureus shared around half of their core gene repertoire, | |
indicating a high level of divergence and providing strong | |
support for their classification as separate species. Several | |
gene clusters were present in ST2250 isolates but absent from the | |
other S. argenteus and S. aureus study isolates. These included | |
multiple exotoxins and antibiotic resistance genes that have been | |
linked previously with livestock-associated S. aureus, consistent | |
with a livestock reservoir for S. argenteus These genes appeared | |
to be associated with plasmids and mobile genetic elements and | |
may have contributed to the biological success of | |
ST2250.IMPORTANCE In this study, we used whole-genome sequencing | |
to understand the genome evolution and population structure of a | |
systematic collection of ST2250 S. argenteus isolates. A newly | |
identified ancestral species of S. aureus, S. argenteus has | |
become increasingly known as a clinically important species that | |
has been reported recently across various countries. Our results | |
indicate that S. argenteus has spread at a relatively rapid pace | |
over the past 2 decades across northeastern Thailand and acquired | |
multiple exotoxin and antibiotic resistance genes that have been | |
linked previously with livestock-associated S. aureus Our | |
findings highlight the clinical importance and potential | |
pathogenicity of S. argenteus as a recently emerging pathogen.", | |
journal = "MBio", | |
volume = 8, | |
number = 4, | |
month = jul, | |
year = 2017, | |
keywords = "Staphylococcus argenteus; Staphylococcus aureus; antibiotic | |
resistance; genomic | |
epidemiology;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Weterings2017-wc, | |
title = "Next-generation sequence analysis reveals methicillin-resistance | |
transfer to a methicillin-susceptible \textit{Staphylococcus | |
aureus} ({MSSA}) strain that subsequently caused a | |
methicillin-resistant \textit{Staphylococcus aureus} ({MRSA}) | |
outbreak: a descriptive study", | |
author = "Weterings, Veronica and Bosch, Thijs and Witteveen, Sandra and | |
Landman, Fabian and Schouls, Leo and Kluytmans, Jan", | |
abstract = "Resistance to methicillin in Staphylococcus aureus is primarily | |
caused by the mecA gene, which is carried on a mobile genetic | |
element: the staphylococcal cassette chromosome mec (SCCmec). | |
Horizontal transfer of this element is supposed to play an | |
important factor in the emergence of new clones of | |
methicillin-resistant Staphylococcus aureus (MRSA), but has been | |
rarely observed in real-time.In 2012, an outbreak occurred | |
involving a healthcare worker (HCW) and three patients all | |
carrying a fusidic-acid resistant MRSA strain. The husband of the | |
HCW was screened for MRSA carriage, but only a | |
methicillin-susceptible S. aureus (MSSA) was detected, which was | |
also resistant for fusidic acid. Multiple Locus Variable-number | |
Tandem Repeat Analysis (MLVA)-typing showed that both the MSSA | |
and the MRSA isolates were MT4053-MC0005. This finding led to the | |
hypothesis that the MSSA strain acquired the SCCmec and | |
subsequently caused an outbreak. To support this hypothesis, | |
next-generation sequencing of the MSSA and MRSA isolates was | |
performed.This study showed that the MSSA isolate clustered | |
closely with the outbreak isolates based on whole-genome | |
multilocus sequence typing (wgMLST) and the single nucleotide | |
polymorphism (SNP) analysis, with a distance of 17 genes and 44 | |
SNPs, respectively. Remarkably, there were relatively large | |
differences in mobile genetic elements in strains within and | |
between individuals.The limited genetic distance between the MSSA | |
and the MRSA isolates in combination with a clear epidemiologic | |
link, support the hypothesis that the MSSA isolate acquired a | |
SCCmec and that the resulting MRSA strain caused an | |
outbreak.IMPORTANCE This report shows that acquisition of the | |
staphylococcal cassette chromosome mec (SCCmec) by a | |
methicillin-susceptible S. aureus does occur. In this case it | |
concerned a healthcare worker (HCW) and this subsequently caused | |
an outbreak in a vulnerable group of patients. The whole genome | |
sequence based analysis further showed that considerable genetic | |
variability occurred in a relatively short time span. These | |
findings are important to reconsider the exclusion of HCW with | |
regard to control measures for methicillin-resistant S. aureus | |
Also the genetic variation in related microorganisms is likely to | |
be larger than currently assumed.", | |
journal = "J. Clin. Microbiol.", | |
month = jul, | |
year = 2017, | |
keywords = "Staphylococcus;antibiotics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{DeFrancesco2017-dv, | |
title = "Genome-wide screen for genes involved in {eDNA} release during | |
biofilm formation by \textit{Staphylococcus aureus}", | |
author = "DeFrancesco, Alicia S and Masloboeva, Nadezda and Syed, Adnan K | |
and DeLoughery, Aaron and Bradshaw, Niels and Li, Gene-Wei and | |
Gilmore, Michael S and Walker, Suzanne and Losick, Richard", | |
abstract = "Staphylococcus aureus is a leading cause of both nosocomial and | |
community-acquired infection. Biofilm formation at the site of | |
infection reduces antimicrobial susceptibility and can lead to | |
chronic infection. During biofilm formation, a subset of cells | |
liberate cytoplasmic proteins and DNA, which are repurposed to | |
form the extracellular matrix that binds the remaining cells | |
together in large clusters. Using a strain that forms robust | |
biofilms in vitro during growth under glucose supplementation, we | |
carried out a genome-wide screen for genes involved in the | |
release of extracellular DNA (eDNA). A high-density transposon | |
insertion library was grown under biofilm-inducing conditions, | |
and the relative frequency of insertions was compared between | |
genomic DNA (gDNA) collected from cells in the biofilm and eDNA | |
from the matrix. Transposon insertions into genes encoding | |
functions necessary for eDNA release were identified by reduced | |
representation in the eDNA. On direct testing, mutants of some of | |
these genes exhibited markedly reduced levels of eDNA and a | |
concomitant reduction in cell clustering. Among the genes with | |
robust mutant phenotypes were gdpP, which encodes a | |
phosphodiesterase that degrades the second messenger | |
cyclic-di-AMP, and xdrA, the gene for a transcription factor | |
that, as revealed by RNA-sequencing analysis, influences the | |
expression of multiple genes, including many involved in cell | |
wall homeostasis. Finally, we report that growth in | |
biofilm-inducing medium lowers cyclic-di-AMP levels and does so | |
in a manner that depends on the gdpP phosphodiesterase gene.", | |
journal = "Proc. Natl. Acad. Sci. U. S. A.", | |
month = jul, | |
year = 2017, | |
keywords = "Staphylococcus aureus; biofilm; cyclic-di-AMP; | |
eDNA;Staphylococcus;tn-seq;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Putonti2017-qz, | |
title = "Draft Genome Sequences of Two {ATCC} \textit{Staphylococcus | |
aureus} subsp. \textit{aureus} Strains", | |
author = "Putonti, Catherine and Kalesinskas, Laurynas and Cudone, Evan and | |
Engelbrecht, Kathleen C and Koenig, David W and Wolfe, Alan J", | |
abstract = "Draft genome sequences for Staphylococcus aureus subsp. aureus | |
Rosenbach ATCC 14458 and ATCC 27217 strains were investigated. | |
The genome sizes were 2,880,761 bp and 2,759,100 bp, | |
respectively. Strain ATCC 14458 was assembled into 39 contigs, | |
including 3 plasmids, and strain ATCC 27217 was assembled into 25 | |
contigs, including 2 plasmids.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 27, | |
month = jul, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Sahibzada2017-gl, | |
title = "Transmission of highly virulent community-associated {MRSA} | |
{ST93} and livestock-associated {MRSA} {ST398} between humans and | |
pigs in Australia", | |
author = "Sahibzada, S and Abraham, S and Coombs, G W and Pang, S and | |
Hern{\'a}ndez-Jover, M and Jordan, D and Heller, J", | |
abstract = "Pigs have been recognised as a reservoir of livestock associated | |
methicillin-resistant Staphylococcus aureus (LA-MRSA) in Europe, | |
Asia and North America. However, little is known about the | |
presence and distribution of MRSA in the Australian pig | |
population and pig industry. This study describes the presence, | |
distribution and molecular characteristics of the human adapted | |
Australian CA-MRSA ST93 isolated from pigs, people, and the | |
environment within a piggery. Isolates were subjected to | |
antibiotic susceptibility testing, DNA microarray, whole genome | |
sequencing, multi locus sequence typing, virulence and resistance | |
gene characterization and phylogenetic analysis. MRSA were | |
isolated from 60\% (n = 52) of farm workers where 84\% of | |
isolates returned ST93 and the rest ST398. Of the thirty-one pig | |
isolates tested further, an equal number of ST398 and ST93 (15 | |
each) and one as ST30-V were identified. Four of six | |
environmental isolates were identified as ST93 and two as ST398. | |
This study has identified for the first time in Australia the | |
occurrence of CA-MRSA ST93 and LA-MRSA ST398 amongst farm | |
workers, pigs, and the farm environment. Comparative genome | |
analysis indicates that ST398 is likely to have been introduced | |
into Australia from Europe or North America. This study also | |
reports the first linezolid resistant MRSA isolated in Australia.", | |
journal = "Sci. Rep.", | |
volume = 7, | |
number = 1, | |
pages = "5273", | |
month = jul, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Ward2016-el, | |
title = "Identification of source and sink populations for the emergence | |
and global spread of the {East-Asia} clone of | |
community-associated {MRSA}", | |
author = "Ward, Melissa J and Goncheva, Mariya and Richardson, Emily and | |
McAdam, Paul R and Raftis, Emma and Kearns, Angela and Daum, | |
Robert S and David, Michael Z and Lauderdale, Tsai Ling and | |
Edwards, Giles F and Nimmo, Graeme R and Coombs, Geoffrey W and | |
Huijsdens, Xander and Woolhouse, Mark E J and Fitzgerald, J Ross", | |
abstract = "BACKGROUND: Our understanding of the factors influencing the | |
emergence, dissemination and global distribution of epidemic | |
clones of bacteria is limited. ST59 is a major epidemic clone of | |
community-associated MRSA in East Asia, responsible for extensive | |
morbidity and mortality, but has a much lower prevalence in other | |
parts of the world. The geographic origin of ST59 and its | |
international routes of dissemination are unclear and disputed in | |
the literature. RESULTS: To investigate the origin and spread of | |
the ST59 clone, we obtained whole genome sequences of isolates | |
from four continents, sampled over more than a decade, and | |
carried out a time-scaled phylogeographic analysis. We discover | |
that two distinct ST59 clades emerged concurrently, in East Asia | |
and the USA, but underwent clonal expansion at different times. | |
The East Asia clade was strongly enriched for gene determinants | |
associated with antibiotic resistance, consistent with regional | |
differences in antibiotic usage. Both clones spread independently | |
to Australia and Europe, and we found evidence of the persistence | |
of multi-drug resistance following export from East Asia. Direct | |
transfer of strains between Taiwan and the USA was not observed | |
in either direction, consistent with geographic niche exclusion. | |
CONCLUSIONS: Our results resolve a longstanding controversy | |
regarding the origin of the ST59 clone, revealing the major | |
global source and sink populations and routes for the spread of | |
multi-drug resistant clones. Additionally, our findings indicate | |
that diversification of the accessory genome of epidemic clones | |
partly reflects region-specific patterns of antibiotic usage, | |
which may influence bacterial fitness after transmission to | |
different geographic locations.", | |
journal = "Genome Biol.", | |
volume = 17, | |
number = 1, | |
pages = "160", | |
month = jul, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Hayden2016-uc, | |
title = "Chlorhexidine and Mupirocin Susceptibility of | |
{Methicillin-Resistant} Staphylococcus aureus Isolates in the | |
{REDUCE-MRSA} Trial", | |
author = "Hayden, Mary K and Lolans, Karen and Haffenreffer, Katherine and | |
Avery, Taliser R and Kleinman, Ken and Li, Haiying and Kaganov, | |
Rebecca E and Lankiewicz, Julie and Moody, Julia and Septimus, | |
Edward and Weinstein, Robert A and Hickok, Jason and Jernigan, | |
John and Perlin, Jonathan B and Platt, Richard and Huang, Susan S", | |
abstract = "Whether targeted or universal decolonization strategies for the | |
control of methicillin-resistant Staphylococcus aureus (MRSA) | |
select for resistance to decolonizing agents is unresolved. The | |
REDUCE-MRSA trial (ClinicalTrials registration no. NCT00980980) | |
provided an opportunity to investigate this question. REDUCE-MRSA | |
was a 3-arm, cluster-randomized trial of either screening and | |
isolation without decolonization, targeted decolonization with | |
chlorhexidine and mupirocin, or universal decolonization without | |
screening to prevent MRSA infection in intensive-care unit (ICU) | |
patients. Isolates from the baseline and intervention periods | |
were collected and tested for susceptibility to chlorhexidine | |
gluconate (CHG) by microtiter dilution; mupirocin susceptibility | |
was tested by Etest. The presence of the qacA or qacB gene was | |
determined by PCR and DNA sequence analysis. A total of 3,173 | |
isolates were analyzed; 2 were nonsusceptible to CHG (MICs, 8 | |
$\mu$g/ml), and 5/814 (0.6\%) carried qacA or qacB At baseline, | |
7.1\% of MRSA isolates expressed low-level mupirocin resistance, | |
and 7.5\% expressed high-level mupirocin resistance. In a | |
mixed-effects generalized logistic regression model, the odds of | |
mupirocin resistance among clinical MRSA isolates or MRSA | |
isolates acquired in an ICU in intervention versus baseline | |
periods did not differ across arms, although estimates were | |
imprecise due to small numbers. Reduced susceptibility to | |
chlorhexidine and carriage of qacA or qacB were rare among MRSA | |
isolates in the REDUCE-MRSA trial. The odds of mupirocin | |
resistance were no different in the intervention versus baseline | |
periods across arms, but the confidence limits were broad, and | |
the results should be interpreted with caution.", | |
journal = "J. Clin. Microbiol.", | |
volume = 54, | |
number = 11, | |
pages = "2735--2742", | |
month = nov, | |
year = 2016, | |
keywords = "Staphylococcus;chlorhexidine;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Sun2017-ib, | |
title = "Characterization of a {PVL-negative} community-acquired | |
methicillin-resistant Staphylococcus aureus strain of sequence | |
type 88 in China", | |
author = "Sun, Lu and Wu, Dandan and Chen, Yan and Wang, Qian and Wang, | |
Haiping and Yu, Yunsong", | |
abstract = "Sequence type 88 community-acquired methicillin-resistant | |
Staphylococcus aureus (CA-MRSA) strain SR434, isolated from an | |
outpatient with skin and soft tissue infection, was subjected to | |
whole genome sequencing, antimicrobial susceptibility testing, | |
mouse skin infection model and hemolysis analysis to identify its | |
virulence and resistance determinants. MRSA strain SR434 is | |
resistant to clindamycin, erythromycin and fosfomycin. Four | |
plasmids with resistance genes were identified in this strain, | |
including a 20,658bp blaZ-carrying plasmid, a 2473bp | |
ermC-carrying plasmid, a 2622bp fosB7-carrying plasmid (86\% | |
identity with plasmid in a ST2590 MRSA strain) and a 4817bp | |
lnuA-carrying plasmid (99\% identity with pLNU4 from bovine | |
coagulase-nagetive Staphylococci). This strain contains | |
staphylococcal cassette chromosome mec type IV and does not | |
contain arginine catabolic mobile element or | |
Panton-Valentine-Leukocidin. SR434 harbors genomic islands | |
$\nu$Sa$\alpha$, $\nu$Sa$\beta$, $\nu$Sa$\gamma$ and $\Phi$Sa3 | |
and pathogenicity islands $\nu$Sa2 that carries genes encoding | |
toxic shock syndrome toxin 1, superantigen enterotoxin C and | |
superantigen enterotoxin L. Mouse skin infection model results | |
show that SR434 had similar virulence potential causing invasive | |
skin infection as a PVL-negative epidemic Korea clone HL1 (ST72). | |
CA-MRSA strain of ST88 lineage might be a great concern for its | |
high virulence. PVL has limited contribution to virulence | |
phenotype among this lineage.", | |
journal = "Int. J. Med. Microbiol.", | |
month = jul, | |
year = 2017, | |
keywords = "CA-MRSA; Genome; High virulence; | |
ST88;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Harkins2017-uw, | |
title = "Methicillin-resistant Staphylococcus aureus emerged long before | |
the introduction of methicillin into clinical practice", | |
author = "Harkins, Catriona P and Pichon, Bruno and Doumith, Michel and | |
Parkhill, Julian and Westh, Henrik and Tomasz, Alexander and de | |
Lencastre, Herminia and Bentley, Stephen D and Kearns, Angela M | |
and Holden, Matthew T G", | |
abstract = "BACKGROUND: The spread of drug-resistant bacterial pathogens | |
poses a major threat to global health. It is widely recognised | |
that the widespread use of antibiotics has generated selective | |
pressures that have driven the emergence of resistant strains. | |
Methicillin-resistant Staphylococcus aureus (MRSA) was first | |
observed in 1960, less than one year after the introduction of | |
this second generation beta-lactam antibiotic into clinical | |
practice. Epidemiological evidence has always suggested that | |
resistance arose around this period, when the mecA gene encoding | |
methicillin resistance carried on an SCCmec element, was | |
horizontally transferred to an intrinsically sensitive strain of | |
S. aureus. RESULTS: Whole genome sequencing a collection of the | |
first MRSA isolates allows us to reconstruct the evolutionary | |
history of the archetypal MRSA. We apply Bayesian phylogenetic | |
reconstruction to infer the time point at which this early MRSA | |
lineage arose and when SCCmec was acquired. MRSA emerged in the | |
mid-1940s, following the acquisition of an ancestral type I | |
SCCmec element, some 14 years before the first therapeutic use of | |
methicillin. CONCLUSIONS: Methicillin use was not the original | |
driving factor in the evolution of MRSA as previously thought. | |
Rather it was the widespread use of first generation beta-lactams | |
such as penicillin in the years prior to the introduction of | |
methicillin, which selected for S. aureus strains carrying the | |
mecA determinant. Crucially this highlights how new drugs, | |
introduced to circumvent known resistance mechanisms, can be | |
rendered ineffective by unrecognised adaptations in the bacterial | |
population due to the historic selective landscape created by the | |
widespread use of other antibiotics.", | |
journal = "Genome Biol.", | |
volume = 18, | |
number = 1, | |
pages = "130", | |
month = jul, | |
year = 2017, | |
keywords = "Antibiotic resistance; MRSA; Staphylococcus | |
aureus;Staphylococcus;antibiotics;transmission-epidemiology;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Arias2017-iu, | |
title = "Molecular Epidemiology and Phylogenomics of | |
\textit{Staphylococcus aureus} Bacteremia in {Latin-America}: A | |
Prospective Cohort Multicenter Study in Nine Countries", | |
author = "Arias, Cesar A and Reyes, Jinnethe and Carvajal, Lina Paola and | |
Rincon, Sandra and Diaz, Lorena and Panesso, Diana and Ibarra, | |
Gabriel and Rios, Rafael and Munita, Jose M and Salles, Mauro J | |
and Alvarez-Moreno, Carlos and Labarca, Jaime and Garcia, | |
Coralith and Luna, Carlos M and Mejia-Villatoro, Carlos and | |
Zurita, Jeannete and Guzman-Blanco, Manuel and Rodriguez-Noriega, | |
Eduardo and Narechania, Apurva and Rojas, Laura J and Planet, | |
Paul J and Weinstock, George M and Gotuzzo, Eduardo and Seas, | |
Carlos", | |
abstract = "Staphylococcus aureus are important pathogens causing a spectrum | |
of diseases ranging from mild skin and soft tissue infections to | |
life-threatening conditions. Bloodstream infections are | |
particularly important and the treatment approach is complicated | |
by the presence of methicillin resistance among S. aureus | |
isolates (MRSA). Emergence of new genetic lineages of MRSA has | |
occurred in Latin America (LA) with the rise and dissemination of | |
the community-associated USA300 Latin American Variant | |
(USA300-LV). Here, we prospectively characterized bloodstream | |
MRSA recovered from selected hospitals in 9 Latin American | |
countries. All isolates were typed by pulsed field gel | |
electrophoresis (PFGE) and subjected to antibiotic susceptibility | |
testing. Whole genome sequencing was performed in 96 MRSA | |
representatives. MRSA represented 45\% of all isolates (out of | |
1,185 S. aureus). The majority of MRSA isolates belonged to | |
clonal cluster (CC) 5. In Colombia and Ecuador, most isolates | |
($\geq$ 72\%) belonged to the USA300-LV lineage (CC8). | |
Phylogenetic reconstructions indicated that MRSA isolates from | |
participating hospitals belonged to three major clades. Clade A | |
grouped isolates with ST5, ST105, and ST1011 (mostly SCCmec I and | |
II). Clade B included ST8, ST88, ST97, and ST72 strains (SCCmec | |
IV, subtypes a, b, and c/E) and clade C grouped mostly | |
Argentinian MRSA belonging to ST30. In summary, CC5 MRSA was | |
prevalent in bloodstream infections in LA with the exception of | |
Colombia and Ecuador where USA300-LV is now the dominant lineage. | |
Clonal replacement appears to be a common phenomenon and | |
continuous surveillance is crucial to identify changes in the | |
molecular epidemiology of MRSA.", | |
journal = "Antimicrob. Agents Chemother.", | |
month = jul, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Hung2016-fz, | |
title = "Molecular Evolutionary Pathways toward Two Successful | |
{Community-Associated} but {Multidrug-Resistant} {ST59} | |
{Methicillin-Resistant} Staphylococcus aureus Lineages in Taiwan: | |
Dynamic Modes of Mobile Genetic Element Salvages", | |
author = "Hung, Wei-Chun and Wan, Tsai-Wen and Kuo, Yu-Chia and Yamamoto, | |
Tatsuo and Tsai, Jui-Chang and Lin, Yu-Tzu and Hsueh, Po-Ren and | |
Teng, Lee-Jene", | |
abstract = "Clonal complex 59 (CC59) Staphylococcus aureus in Taiwan includes | |
both methicillin-susceptible S. aureus (MSSA) and | |
methicillin-resistant S. aureus (MRSA). As the most prominent | |
community-associated MRSA (CA-MRSA) in Taiwan, CC59 has two major | |
clones characterized as PVL-negative SCCmec IV (carrying the | |
staphylococcal cassette chromosome mec IV but Panton-Valentine | |
leukocidin-negative) and PVL-positive SCCmec V (5C2\&5). We | |
investigated the drug resistance, phylogeny and the distribution | |
and sequence variation of SCCmec, staphylococcal bacteriophage | |
$\varphi$SA3, genomic island $\nu$Sa$\beta$ and MES (an | |
enterococcal mobile genetic element conferring multidrug | |
resistance) in 195 CC59 S. aureus. Sequencing and PCR mapping | |
revealed that all of the CC59/SCCmec V (5C2\&5) MRSA strains had | |
acquired MESPM1 or its segregants, and obtained a | |
$\varphi$SA3-related fragment in $\nu$Sa$\beta$. In contrast, | |
MES6272-2 and MES4578, which showed gentamicin resistance that | |
was not encoded by MESPM1, were dominant in SCCmec IVg MRSA. | |
Translocation of a whole $\varphi$SA3 into $\nu$Sa$\beta$ instead | |
of only a $\varphi$SA3-related fragment was common in SCCmec IVg | |
MRSA. However, the non-subtype-g SCCmec IV MRSA (SCCmec IVa is | |
the major) still carried MES and $\nu$Sa$\beta$ structures | |
similar to those in SCCmec V (5C2\&5) MRSA. A minimum spanning | |
tree constructed by multiple-locus variable-number tandem repeat | |
analysis revealed that SCCmec IVg MRSA and SCCmec V (5C2\&5) MRSA | |
grouped respectively in two major clades. The CC59 MSSA was | |
equally distributed among the two clades, while the non-subtype-g | |
SCCmec IV MRSA mostly clustered with SCCmec V (5C2\&5) MRSA. Our | |
findings strongly suggest that CC59 MSSA acquired divergent | |
mobile genetic elements and evolved to SCCmec IVg MRSA and SCCmec | |
V (5C2\&5) MRSA/non-subtype-g SCCmec IV MRSA independently. The | |
evolutionary history of CC59 S. aureus explains how mobile | |
genetic elements increase the antimicrobial resistance and | |
virulence and contribute to the success of CA-MRSA in Taiwan.", | |
journal = "PLoS One", | |
volume = 11, | |
number = 9, | |
pages = "e0162526", | |
month = sep, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Werth2017-wu, | |
title = "Emergence of dalbavancin non-susceptible, vancomycin-intermediate | |
Staphylococcus aureus ({VISA}) after treatment of {MRSA} central | |
line-associated bloodstream infection with a dalbavancin- and | |
vancomycin-containing regimen", | |
author = "Werth, Brian J and Jain, Rupali and Hahn, Andrew and Cummings, | |
Lisa and Weaver, Tatiana and Waalkes, Adam and Sengupta, Dhruba | |
and Salipante, Stephen J and Rakita, Robert M and Butler-Wu, | |
Susan M", | |
abstract = "OBJECTIVES: Dalbavancin is a long-acting lipoglycopeptide with | |
activity against Gram-positives, including methicillin-resistant | |
Staphylococcus aureus(MRSA). The potential for lipoglycopeptides, | |
with half-lives greater than one-week, to select for resistance | |
is unknown. Here we explore a case of MRSA central | |
line-associated blood stream infection in which dalbavancin and | |
vancomycin non-susceptibility emerged in a urine isolate | |
collected after the patient was treated with vancomycin and | |
dalbavancin sequentially. METHODS: Isolates from blood and urine | |
underwent susceptibility testing, and whole genome sequencing | |
(WGS). The blood isolate was subjected to successive passage in | |
vitro in the presence of escalating dalbavancin concentrations | |
and the emergent isolate was subjected to repeat susceptibility | |
testing and WGS. RESULTS: The blood isolate was fully susceptible | |
to vancomycin, however, MICs of the urine isolate to dalbavancin, | |
vancomycin, telavancin and daptomycin were $\geq$4-fold higher | |
than the blood-derived strain. Both strains were | |
indistinguishable by spa and variable number tandem repeat (VNTR) | |
typing, and WGS revealed only 7 variants, indicating clonality. | |
Four variants affected genes, including a non-synonymous change | |
in yvqF, which has been implicated in glycopeptide resistance. | |
Vancomycin and dalbavancin non-susceptibility emerged in the | |
blood isolate after successive passage in vitro in the presence | |
of dalbavancin, and WGS identified a single non-synonymous | |
variant in yvqF. CONCLUSIONS: This is the first case in which | |
VISA has emerged in the context of a dalbavancin-containing | |
regimen. The selection for cross-resistance to vancomycin in | |
vitro by dalbavancin exposure alone is troubling. Clinicians | |
should be aware of the possibility for emergence of dalbavancin | |
non-susceptibility and glycopeptide cross-resistance arising | |
following therapy.", | |
journal = "Clin. Microbiol. Infect.", | |
month = aug, | |
year = 2017, | |
keywords = "CLABSI; cross-resistance; glycopeptide resistance; | |
lipoglycopeptide;Staphylococcus;confirmed\_SA-resistance-mutation;Papers\_for\_Michelle;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Hau2017-ag, | |
title = "Complete Genome Sequence of {Livestock-Associated} | |
{Methicillin-Resistant} \textit{Staphylococcus aureus} Sequence | |
Type 398 Isolated from Swine in the United States", | |
author = "Hau, Samantha J and Bayles, Darrell O and Alt, David P and Frana, | |
Timothy S and Nicholson, Tracy L", | |
abstract = "Methicillin-resistant Staphylococcus aureus (MRSA) colonizes and | |
causes disease in many animal species. Livestock-associated MRSA | |
(LA-MRSA) isolates are represented by isolates of the sequence | |
type 398 (ST398). These isolates are considered to be livestock | |
adapted. This report provides the complete genome sequence of one | |
swine-associated LA-MRSA ST398 isolate from the United States.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 32, | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Hau2017-vj, | |
title = "Complete Genome Sequence of a {Livestock-Associated} | |
{Methicillin-Resistant} \textit{Staphylococcus aureus} Sequence | |
Type 5 Isolate from the United States", | |
author = "Hau, Samantha J and Bayles, Darrell O and Alt, David P and Frana, | |
Timothy S and Nicholson, Tracy L", | |
abstract = "Livestock-associated methicillin-resistant Staphylococcus aureus | |
(LA-MRSA) may be the largest MRSA reservoir outside the hospital | |
setting. One concern with LA-MRSA is the acquisition of novel | |
mobile genetic elements by these isolates. Here, we report the | |
complete genome sequence of a swine LA-MRSA sequence type 5 | |
isolate from the United States.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 32, | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Harrison2017-fy, | |
title = "Genomic surveillance reveals low prevalence of | |
livestock-associated methicillin-resistant Staphylococcus aureus | |
in the East of England", | |
author = "Harrison, Ewan M and Coll, Francesc and Toleman, Michelle S and | |
Blane, Beth and Brown, Nicholas M and T{\"o}r{\"o}k, M Estee and | |
Parkhill, Julian and Peacock, Sharon J", | |
abstract = "Livestock-associated methicillin-resistant Staphylococcus aureus | |
(LA-MRSA) is an emerging problem in many parts of the world. | |
LA-MRSA has been isolated previously from animals and humans in | |
the United Kingdom (UK), but the prevalence is unknown. The aim | |
of this study was to determine the prevalence and to describe the | |
molecular epidemiology of LA-MRSA isolated in the East of England | |
(broadly Cambridge and the surrounding area). We accessed whole | |
genome sequence data for 2,283 MRSA isolates from 1,465 people | |
identified during a 12-month prospective study between 2012 and | |
2013 conducted in the East of England, United Kingdom. This | |
laboratory serves four hospitals and 75 general practices. We | |
screened the collection for multilocus sequence types (STs) and | |
for host specific resistance and virulence factors previously | |
associated with LA-MRSA. We identified 13 putative LA-MRSA | |
isolates from 12 individuals, giving an estimated prevalence of | |
0.82\% (95\% CI 0.47\% to 1.43\%). Twelve isolates were mecC-MRSA | |
(ten CC130, one ST425 and one ST1943) and single isolate was | |
ST398. Our data demonstrate a low burden of LA-MRSA in the East | |
of England, but the detection of mecC-MRSA and ST398 indicates | |
the need for vigilance. Genomic surveillance provides a mechanism | |
to detect and track the emergence and spread of MRSA clones of | |
human importance.", | |
journal = "Sci. Rep.", | |
volume = 7, | |
number = 1, | |
pages = "7406", | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Caldelari2017-tn, | |
title = "Complete Genome Sequence and Annotation of the | |
\textit{Staphylococcus aureus} Strain {HG001}", | |
author = "Caldelari, Isabelle and Chane-Woon-Ming, B{\'e}atrice and Noirot, | |
C{\'e}line and Moreau, Karen and Romby, Pascale and Gaspin, | |
Christine and Marzi, Stefano", | |
abstract = "Staphylococcus aureus is an opportunistic Gram-positive pathogen | |
responsible for a wide range of infections from minor skin | |
abscesses to life-threatening diseases. Here, we report the draft | |
genome assembly and current annotation of the HG001 strain, a | |
derivative of the RN1 (NCT8325) strain with restored rbsU (a | |
positive activator of SigB).", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 32, | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Grinberg2017-ax, | |
title = "Genomic epidemiology of methicillin-susceptible Staphylococcus | |
aureus across colonisation and skin and soft tissue infection", | |
author = "Grinberg, Alex and Biggs, Patrick J and Zhang, Ji and Ritchie, | |
Stephen and Oneroa, Zachary and O'Neil, Charlotte and Karkaba, | |
Ali and Velathanthiri, Niluka S and Coombs, Geoffrey W", | |
abstract = "OBJECTIVES: Staphylococcus aureus skin and soft tissue infection | |
(Sa-SSTI) places a significant burden on healthcare systems. New | |
Zealand has a high incidence of Sa-SSTI, and here most morbidity | |
is caused by a polyclonal methicillin-susceptible (MSSA) | |
population. However, MSSA also colonise asymptomatically the | |
cornified epithelia of approximately 20\% of the population, and | |
their divide between commensalism and pathogenicity is poorly | |
understood. We aimed to see whether MSSA is genetically | |
differentiated across asymptomatic colonisation and SSTI; and | |
given the close interactions between people and pets, whether | |
strains isolated from pets differ from human strains. METHODS: We | |
compared the genomes of contemporaneous asymptomatic colonisation | |
and clinical MSSA isolates obtained in New Zealand from humans | |
and pets. RESULTS: Core and accessory genome comparisons revealed | |
a homogeneous bacterial population across colonisation, disease, | |
humans, and pets. The rate of MSSA colonisation in dogs was | |
comparatively low (5.4\%). CONCLUSIONS: In New Zealand, most | |
Sa-SSTI morbidity is caused by a random sample of the colonising | |
MSSA population, consistent with the opportunistic infection | |
paradigm rather than a model distinguishing pathogenic from | |
innocuous strains. Thus, studies of host factors determining | |
colonisation and immune-escape may be more beneficial than | |
comparative virulence studies. Contact with house-hold pets may | |
pose low zoonotic risk.", | |
journal = "J. Infect.", | |
month = aug, | |
year = 2017, | |
keywords = "SSTI; Skin and soft tissue infection; Staphylococcus aureus; | |
Whole genome sequencing; | |
Zoonoses;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Recker2017-lp, | |
title = "Clonal differences in Staphylococcus aureus | |
bacteraemia-associated mortality", | |
author = "Recker, Mario and Laabei, Maisem and Toleman, Michelle S and | |
Reuter, Sandra and Saunderson, Rebecca B and Blane, Beth and | |
T{\"o}r{\"o}k, M Estee and Ouadi, Khadija and Stevens, Emily and | |
Yokoyama, Maho and Steventon, Joseph and Thompson, Luke and | |
Milne, Gregory and Bayliss, Sion and Bacon, Leann and Peacock, | |
Sharon J and Massey, Ruth C", | |
abstract = "The bacterium Staphylococcus aureus is a major human pathogen for | |
which the emergence of antibiotic resistance is a global public | |
health concern. Infection severity, and in particular | |
bacteraemia-associated mortality, has been attributed to several | |
host-related factors, such as age and the presence of | |
comorbidities. The role of the bacterium in infection severity is | |
less well understood, as it is complicated by the multifaceted | |
nature of bacterial virulence, which has so far prevented a | |
robust mapping between genotype, phenotype and infection outcome. | |
To investigate the role of bacterial factors in contributing to | |
bacteraemia-associated mortality, we phenotyped a collection of | |
sequenced clinical S. aureus isolates from patients with | |
bloodstream infections, representing two globally important | |
clonal types, CC22 and CC30. By adopting a genome-wide | |
association study approach we identified and functionally | |
verified several genetic loci that affect the expression of | |
cytolytic toxicity and biofilm formation. By analysing the pooled | |
data comprising bacterial genotype and phenotype together with | |
clinical metadata within a machine-learning framework, we found | |
significant clonal differences in the determinants most | |
predictive of poor infection outcome. Whereas elevated cytolytic | |
toxicity in combination with low levels of biofilm formation was | |
predictive of an increased risk of mortality in infections by | |
strains of a CC22 background, these virulence-specific factors | |
had little influence on mortality rates associated with CC30 | |
infections. Our results therefore suggest that different clones | |
may have adopted different strategies to overcome host responses | |
and cause severe pathology. Our study further demonstrates the | |
use of a combined genomics and data analytic approach to enhance | |
our understanding of bacterial pathogenesis at the individual | |
level, which will be an important step towards personalized | |
medicine and infectious disease management.A genome-wide | |
association approach identifies differential biofilm and | |
virulence attributes associated with mortality in two | |
Staphylococcus aureus clonal complexes.", | |
journal = "Nat Microbiol", | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Kleinert2017-cf, | |
title = "Influence of {IS\textit{256}} on genome variability and formation | |
of {SCV} in \textit{Staphylococcus aureus}", | |
author = "Kleinert, Franziska and Kallies, Ren{\'e} and Hort, Michael and | |
Zweynert, Annegret and Szekat, Christiane and Nagel, Michael and | |
Bierbaum, Gabriele", | |
abstract = "Staphylococcus aureus has acquired resistance to nearly all | |
antibiotics used in clinical practice. Whereas some resistance | |
mechanisms are conferred by uptake of resistance genes, others | |
evolve by mutation. Here, the insertion sequence 256 (IS256) has | |
been shown to play a role, e. g. in S. aureus strains displaying | |
intermediate resistance to vancomycin (VISA).To characterize the | |
IS256 insertion sites in the genomes of two closely related | |
sequence type (ST) 247 VISA strains, all insertions were mapped | |
in both VISA and a susceptible control strain. The results showed | |
that the three ST247 strains contained the highest number so far | |
of IS256 insertions of all sequenced S. aureus strains. | |
Furthermore, in contrast to the other IS elements in these | |
genomes, the IS256 insertion sites were not identical in the | |
closely related strains, indicating a high transposition | |
frequency of IS256 When IS256 was introduced into a laboratory | |
strain which was then cultured in the presence of antibiotics, it | |
was possible to isolate small colony variants (SCV) that | |
possessed IS256 insertions in guaA and hemY that displayed | |
increased resistance to vancomycin and aminoglycosides, | |
respectively. For these clones, a very rapid reversion to the | |
wild type was observed, that resembled the fast reversion of | |
clinical SCV. The reversion was caused by excision of IS256 in a | |
small number of fast growing clones that quickly outcompeted the | |
SCVs in broth cultures. In conclusion, the presence of IS256 | |
confers a strong genomic plasticity that is useful for adaptation | |
to antibiotic stress.", | |
journal = "Antimicrob. Agents Chemother.", | |
month = jun, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Panthee2017-to, | |
title = "Genomic analysis of vancomycin-resistant Staphylococcus aureus | |
{VRS3b} and its comparison with other {VRSA} isolates", | |
author = "Panthee, Suresh and Hamamoto, Hiroshi and Paudel, Atmika and | |
Sekimizu, Kazuhisa", | |
abstract = "High-level vancomycin resistance among Staphylococcus aureus | |
poses a grave threat to global health as the treatment options | |
for this pathogen are very limited. A detailed evaluation of the | |
genetic background of vancomycin-resistant S. aureus (VRSA) is | |
expected to facilitate the understanding of its origin and | |
pathogenicity. In this study, we performed the genetic analysis | |
of the clinical VRSA isolates and identified the genetic basis of | |
resistance to multiple antibiotics among these strains, based on | |
the available draft genome sequences. In addition, we generated | |
the draft genome of the strain VRS3b, which was considered to be | |
same as VRS3a based on its isolation from the same patient. We | |
found that strain VRS3b did not harbor the genes responsible for | |
tetracycline and gentamicin, which was further confirmed by the | |
sensitivity towards these antibiotics. Our results suggest that | |
the strains VRS3a and VRS3b are different from the view of | |
antibiotic resistance and highlight the possibility of generation | |
of two distinct VRSA strains from the same patient.", | |
journal = "Drug Discov. Ther.", | |
month = apr, | |
year = 2017, | |
keywords = "Staphylococcus aureus; genomics; methicillin; resistance; | |
vancomycin;Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Chow2017-ed, | |
title = "{MRSA} Transmission Dynamics Among Interconnected Acute, | |
{Intermediate-Term}, and {Long-Term} Healthcare Facilities in | |
Singapore", | |
author = "Chow, Angela and Lim, Vanessa W and Khan, Ateeb and Pettigrew, | |
Kerry and Lye, David C B and Kanagasabai, Kala and Phua, Kelvin | |
and Krishnan, Prabha and Ang, Brenda and Marimuthu, Kalisvar and | |
Hon, Pei-Yun and Koh, Jocelyn and Leong, Ian and Parkhill, Julian | |
and Hsu, Li-Yang and Holden, Matthew T G", | |
abstract = "Background: Methicillin-resistant Staphylococcus aureus (MRSA) is | |
the most common healthcare-associated multidrug-resistant | |
organism. Despite the interconnectedness between acute care | |
hospitals (ACHs) and intermediate- and long-term care facilities | |
(ILTCFs), the transmission dynamics of MRSA between healthcare | |
settings is not well understood. Methods: We conducted a | |
cross-sectional study in a network comprising an ACH and 5 | |
closely affiliated ILTCFs in Singapore. A total of 1700 | |
inpatients were screened for MRSA over a 6-week period in 2014. | |
MRSA isolates underwent whole-genome sequencing, with a pairwise | |
single-nucleotide polymorphism (Hamming distance) cutoff of 60 | |
core genome single-nucleotide polymorphisms used to define recent | |
transmission clusters (clades) for the 3 major clones. Results: | |
MRSA prevalence was significantly higher in intermediate-term | |
(29.9\%) and long-term (20.4\%) care facilities than in the ACH | |
(11.8\%) (P < .001). The predominant clones were sequence type | |
[ST] 22 (n = 183; 47.8\%), ST45 (n = 129; 33.7\%), and ST239 (n = | |
26; 6.8\%), with greater diversity of STs in ILTCFs relative to | |
the ACH. A large proportion of the clades in ST22 (14 of 21 | |
clades; 67\%) and ST45 (7 of 13; 54\%) included inpatients from | |
the ACH and ILTCFs. The most frequent source of the interfacility | |
transmissions was the ACH (n = 28 transmission events; 36.4\%). | |
Conclusions: MRSA transmission dynamics between the ACH and | |
ILTCFs were complex. The greater diversity of STs in ILTCFs | |
suggests that the ecosystem in such settings might be more | |
conducive for intrafacility transmission events. ST22 and ST45 | |
have successfully established themselves in ILTCFs. The | |
importance of interconnected infection prevention and control | |
measures and strategies cannot be overemphasized.", | |
journal = "Clin. Infect. Dis.", | |
volume = 64, | |
number = "suppl\_2", | |
pages = "S76--S81", | |
month = may, | |
year = 2017, | |
keywords = "MRSA; intermediate-care facilities; long-term care facilities.; | |
transmission; whole-genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Canovas2016-ts, | |
title = "{Cross-Talk} between Staphylococcus aureus and Other | |
Staphylococcal Species via the agr Quorum Sensing System", | |
author = "Canovas, Jaime and Baldry, Mara and Bojer, Martin S and Andersen, | |
Paal S and Grzeskowiak, Piotr K and Stegger, Marc and Damborg, | |
Peter and Olsen, Christian A and Ingmer, Hanne", | |
abstract = "Staphylococci are associated with both humans and animals. While | |
most are non-pathogenic colonizers, Staphylococcus aureus is an | |
opportunistic pathogen capable of causing severe infections. S. | |
aureus virulence is controlled by the agr quorum sensing system | |
responding to secreted auto-inducing peptides (AIPs) sensed by | |
AgrC, a two component histidine kinase. agr loci are found also | |
in other staphylococcal species and for Staphylococcus | |
epidermidis, the encoded AIP represses expression of agr | |
regulated virulence genes in S. aureus. In this study we aimed to | |
better understand the interaction between staphylococci and S. | |
aureus, and show that this interaction may eventually lead to the | |
identification of new anti-virulence candidates to target S. | |
aureus infections. Here we show that culture supernatants of 37 | |
out of 52 staphylococcal isolates representing 17 different | |
species inhibit S. aureus agr. The dog pathogen, Staphylococcus | |
schleiferi, expressed the most potent inhibitory activity and was | |
active against all four agr classes found in S. aureus. By | |
employing a S. aureus strain encoding a constitutively active AIP | |
receptor we show that the activity is mediated via agr. | |
Subsequent cloning and heterologous expression of the S. | |
schleiferi AIP in S. aureus demonstrated that this molecule was | |
likely responsible for the inhibitory activity, and further proof | |
was provided when pure synthetic S. schleiferi AIP was able to | |
completely abolish agr induction of an S. aureus reporter strain. | |
To assess impact on S. aureus virulence, we co-inoculated S. | |
aureus and S. schleiferi in vivo in the Galleria mellonella wax | |
moth larva, and found that expression of key S. aureus virulence | |
factors was abrogated. Our data show that the S. aureus agr locus | |
is highly responsive to other staphylococcal species suggesting | |
that agr is an inter-species communication system. Based on these | |
results we speculate that interactions between S. aureus and | |
other colonizing staphylococci will significantly influence the | |
ability of S. aureus to cause infection, and we propose that | |
other staphylococci are potential sources of compounds that can | |
be applied as anti-virulence therapy for combating S. aureus | |
infections.", | |
journal = "Front. Microbiol.", | |
volume = 7, | |
pages = "1733", | |
month = nov, | |
year = 2016, | |
keywords = "Staphylococcus aureus; Staphylococcus schleiferi; agr; | |
anti-virulence therapy; auto-inducing peptide; cross-talk; quorum | |
sensing; quorum sensing | |
inhibition;Staphylococcus;Papers\_for\_Michelle;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Roe2016-uc, | |
title = "Whole genome {SNP} typing to investigate methicillin-resistant | |
\textit{Staphylococcus aureus} carriage in a health-care provider | |
as the source of multiple surgical site infections", | |
author = "Roe, Chandler C and Horn, Kimberly S and Driebe, Elizabeth M and | |
Bowers, Jolene and Terriquez, Joel A and Keim, Paul and | |
Engelthaler, David M", | |
abstract = "BACKGROUND: Prevention of nosocomial transmission of infections | |
is a central responsibility in the healthcare environment, and | |
accurate identification of transmission events presents the first | |
challenge. Phylogenetic analysis based on whole genome sequencing | |
provides a high-resolution approach for accurately relating | |
isolates to one another, allowing precise identification or | |
exclusion of transmission events and sources for nearly all | |
cases. We sequenced 24 methicillin-resistant Staphylococcus | |
aureus (MRSA) genomes to retrospectively investigate a suspected | |
point source of three surgical site infections (SSIs) that | |
occurred over a one-year period. The source of transmission was | |
believed to be a surgical team member colonized with MRSA, | |
involved in all surgeries preceding the SSI cases, who was | |
subsequently decolonized. Genetic relatedness among isolates was | |
determined using whole genome single nucleotide polymorphism | |
(SNP) data. RESULTS: Whole genome SNP typing (WGST) revealed 283 | |
informative SNPs between the surgical team member's isolate and | |
the closest SSI isolate. The second isolate was 286 and the third | |
was thousands of SNPs different, indicating the nasal carriage | |
strain from the surgical team member was not the source of the | |
SSIs. Given the mutation rates estimated for S. aureus, none of | |
the SSI isolates share a common ancestor within the past 16 | |
years, further discounting any common point source for these | |
infections. The decolonization procedures and resources spent on | |
the point source infection control could have been prevented if | |
WGST was performed at the time of the suspected transmission, | |
instead of retrospectively. CONCLUSIONS: Whole genome sequence | |
analysis is an ideal method to exclude isolates involved in | |
transmission events and nosocomial outbreaks, and coupling this | |
method with epidemiological data can determine if a transmission | |
event occurred. These methods promise to direct infection control | |
resources more appropriately.", | |
journal = "Hereditas", | |
volume = 153, | |
pages = "11", | |
month = nov, | |
year = 2016, | |
keywords = "MRSA; Nasal carriage; Surgical site infections; Transmission; | |
Whole genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Post2016-tm, | |
title = "Characterisation of nasal methicillin-resistant Staphylococcus | |
aureus isolated from international human and veterinary surgeons", | |
author = "Post, Virginia and Harris, Llinos and Morgenstern, Mario and | |
Richards, R Geoff and Sheppard, Samuel K and Moriarty, Fintan", | |
abstract = "Nasal colonization with methicillin-resistant Staphylococcus | |
aureus is poorly described for surgeons, despite the increased | |
exposure to nosocomial pathogens and at-risk patients. This study | |
investigated the molecular epidemiology and antimicrobial | |
resistance of 26 MRSA isolates cultured from the nares of an | |
international cross-sectional study of 1,166 human and 60 | |
veterinary surgeons. All isolates were subjected to agr-, spa- | |
and MLST typing and the presence of 22 virulence factors were | |
screened for by PCR. Additionally, biofilm-forming ability, | |
haemolytic activity, staphyloxanthin production and antibiotic | |
resistance were determined. The genome of a rifampicin resistant | |
MRSA was sequenced. Approximately half of the isolates belonged | |
to well-described clonal lineages, ST1, ST5, ST8, ST45 and ST59, | |
that have been previously associated with severe infections and | |
increased patient mortality. Two of the 3 veterinarian MRSA | |
belonged to epidemic livestock-associated MRSA clonal lineages | |
(ST398 and ST8) previously associated with high transmission | |
potential between animals and humans. The isolates did not | |
display any consistent virulence gene pattern, and 35\% of the | |
isolates carried at least one of: the Panton-Valentine leukocidin | |
lukFS-PV; the exfoliative toxin eta; or the toxic shock syndrome | |
tst genes. Resistance to rifampicin was detected in one | |
veterinarian isolate, and was found to be due to 3 mutations in | |
the rpoB gene. Surgeons occupy a critical position in the | |
healthcare profession due to their close contact with patients. | |
In this study, surgeons were found to be colonized with MRSA at | |
low rates that are similar to the general population, and the | |
colonising strains were often common clonal lineages.", | |
journal = "J. Med. Microbiol.", | |
month = dec, | |
year = 2016, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Costa2013-dt, | |
title = "Complete genome sequence of a variant of the | |
methicillin-resistant Staphylococcus aureus {ST239} lineage, | |
strain {BMB9393}, displaying superior ability to accumulate | |
ica-independent biofilm", | |
author = "Costa, Maiana Oliveira Cerqueira and Beltrame, Cristiana | |
Ossaille and Ferreira, Fabienne Antunes and Botelho, Ana Maria | |
Nunes and Lima, Nicholas Costa Barroso and Souza, Rangel Celso | |
and de Almeida, Luiz Gonzaga Paula and Vasconcelos, Ana Tereza | |
Ribeiro and Nicol{\'a}s, Marisa Fabiana and Figueiredo, Agnes | |
Marie S{\'a}", | |
abstract = "ABSTRACT Biofilm is considered an important virulence factor in | |
nosocomial infections. Herein, we report the complete genome | |
sequence of a variant of methicillin-resistant Staphylococcus | |
aureus, strain BMB9393, which is highly disseminated in Brazil. | |
This ...", | |
journal = "Genome Announc.", | |
publisher = "Am Soc Microbiol", | |
volume = 1, | |
number = 4, | |
pages = "e00576--13", | |
year = 2013, | |
keywords = "Staphylococcus;experimental\_design;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Lee2017-xz, | |
title = "Emerging multidrug resistance in community-associated | |
Staphylococcus aureus involved in skin and soft tissue infections | |
and nasal colonization", | |
author = "Lee, Grace C and Dallas, Steven D and Wang, Yufeng and Olsen, | |
Randall J and Lawson, Kenneth A and Wilson, James and Frei, | |
Christopher R", | |
abstract = "Background:Staphylococcus aureus is a major pathogen causing | |
significant morbidity and mortality worldwide. The emergence of | |
MDR S. aureus strains in the community setting has major | |
implications in disease management. However, data regarding the | |
occurrence and patterns of MDR community-associated S. aureus | |
sub-clones is limited.Objectives: To use whole-genome sequences | |
to describe the diversity and distribution of resistance | |
mechanisms among community-associated S. aureus | |
isolates.Methods:S. aureus isolates from skin and soft tissue | |
infections (SSTIs) and nasal colonization were collected from | |
patients within 10 primary care clinics from 2007 to 2015. The | |
Illumina Miseq platform was used to determine the genome | |
sequences for 144 S. aureus isolates. Phylogenetic and | |
bioinformatics analyses were performed using in silico tools. The | |
resistome was assembled and compared with the phenotypically | |
derived antibiogram.Results: Approximately one-third of S. aureus | |
isolates in the South Texas primary care setting were MDR. A | |
higher proportion of SSTI isolates were MDR in comparison with | |
nasal colonization isolates. Individuals with MDR S. aureus SSTIs | |
were more likely to be African American and obese. Furthermore, | |
S. aureus populations are able to acquire and lose antimicrobial | |
resistance genes. USA300 strains were differentiated by a stable | |
chromosomal mutation in gyrA conferring quinolone resistance. The | |
resistomes were highly predictive of antimicrobial resistance | |
phenotypes.Conclusions: These findings highlight the high | |
prevalence and epidemiological factors associated with MDR S. | |
aureus strains in the community setting and demonstrate the | |
utility of next-generation sequencing to potentially quicken | |
antimicrobial resistance detection and surveillance for targeted | |
interventions.", | |
journal = "J. Antimicrob. Chemother.", | |
month = jul, | |
year = 2017, | |
keywords = "staphylococcus aureus; drug resistance, microbial; nose; skin and | |
soft tissue infections; microbial colonization; community; drug | |
resistance, multiple; phenotype; genome; genes; mutation; clone | |
cells; african american; chromosomes; obesity; primary health | |
care; disease management; surveillance, medical; | |
antibiogram;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Bayer2016-hc, | |
title = "Dysregulation of mprF and {dltABCD} expression among | |
daptomycin-non-susceptible {MRSA} clinical isolates", | |
author = "Bayer, Arnold S and Mishra, Nagendra N and Cheung, Ambrose L and | |
Rubio, Aileen and Yang, Soo-Jin", | |
abstract = "BACKGROUND: In small series or individual reports, SNPs within | |
the mprF ORF and dysregulation of its expression in | |
Staphylococcus aureus have been linked to daptomycin resistance | |
(DAP-R) via a proposed gain-in-function mechanism. Similarly, | |
dysregulation of dltABCD has also been associated with DAP-R. | |
METHODS: Using 22 well-characterized, isogenic | |
daptomycin-susceptible (DAP-S)/DAP-R clinical MRSA strain pairs, | |
we assessed potential relationships of the DAP-R phenotype with: | |
(i) regulation of mprF transcription; (ii) regulation of dltABCD | |
transcription; (iii) expression of the two-component regulatory | |
system, graRS (upstream regulator for both mprF and dltABCD | |
transcription); (iv) SNPs within the graRS promoter or its ORF; | |
and (v) altered mprF transcription and lysyl-phosphatidylglycerol | |
(L-PG) synthesis. RESULTS: Enhanced expression of mprF occurred | |
with SNPs in highly distinct and well-chronicled MprF domain 'hot | |
spots' and rarely occurred without such mutations. Increased | |
expression and/or dysregulation of mprF and dltABCD were not | |
uncommon in DAP-R strains, occurring in 27\% of strains for each | |
gene. In these latter strains, neither graRS expression profiles | |
nor polymorphic sequences within the graRS promoter or ORF could | |
be significantly linked to altered transcription of mprF or dlt. | |
CONCLUSIONS: Although graRS can co-regulate mprF and dltABCD | |
expression, loci outside of this regulon appear to be involved in | |
dysregulation of these latter two genes and the DAP-R phenotype. | |
Finally, DAP-R strains exhibiting significantly altered mprF | |
transcription profiles produced significantly increased levels of | |
L-PG.", | |
journal = "J. Antimicrob. Chemother.", | |
volume = 71, | |
number = 8, | |
pages = "2100--2104", | |
month = aug, | |
year = 2016, | |
keywords = "confirmed\_SA-resistance-mutation;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Stevens2017-ff, | |
title = "Draft Genome Sequence of \textit{Staphylococcus aureus} S681, a | |
{Tetracycline-Sensitive} {Livestock-Associated} | |
{Methicillin-Resistant} Clonal Complex 398 Strain", | |
author = "Stevens, Marc J A and Stephan, Roger and Johler, Sophia", | |
abstract = "We present the draft genome sequence of an atypical | |
tetracycline-susceptible livestock-associated | |
methicillin-resistant Staphylococcus aureus (MRSA) strain. It | |
contains 2,817,340 bp and 2,858 coding sequences, including 6 | |
rRNA operons, 56 tRNAs, and 4 noncoding RNA (ncRNA) genes. The | |
strain harbors a tet(M) gene, but 15 point mutations in amino | |
acids are present that likely impair the functionality of TetM.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 33, | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Ugolotti2017-fa, | |
title = "Genomic characterization of a paediatric {MRSA} outbreak by Next | |
Generation Sequencing", | |
author = "Ugolotti, Elisabetta and Di Marco, Eddi and Bandettini, Roberto | |
and Biassoni, Roberto", | |
abstract = "INTRODUCTION: Twelve strains of meticillin-resistant | |
Staphylococcus aureus (MRSA) isolated during a suspected outbreak | |
in a Paediatric Intensive Care Unit were analyzed by Whole Genome | |
Sequence (WGS). AIM: To define the clonality of MRSA strains to a | |
high discriminative power and to evaluate the presence of genetic | |
determinants responsible for antibiotic resistance and for | |
virulence. RESULTS: Ten out of 12 strains belonged to the MLST | |
ST2625, while the other two were ST8. Among the ST2625 strains, | |
analysis based on 1126 genes showed that they were clonal sharing | |
more than 98.3\% of allelic identities and one of them was | |
isolated from a health care worker. All the ST2625 strains were | |
characterized by a SCC-Mec cassette IVa and the Resistoma | |
analysis indicated a correspondence between phenotypic and | |
genotypic characteristics. The study of 63 genes associated with | |
the virulence was correlated to the pattern of clonality shown. | |
CONCLUSION: This analysis confirmed the occurrence of an | |
outbreak, which allowed us to strictly enforce standard infection | |
control measures that promptly terminated the outbreak.", | |
journal = "J. Hosp. Infect.", | |
month = aug, | |
year = 2017, | |
keywords = "MRSA; Paediatric Nosocomial Infections; Resistoma; Staphylococcus | |
aureus; Toxoma; Whole-genome | |
sequencing;Staphylococcus;transmission-epidemiology;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Satoo2017-ue, | |
title = "Complete Sequence of a \textit{Staphylococcus aureus} Clonal | |
Complex 81 Strain, the Dominant Lineage in Food Poisoning | |
Outbreaks in Japan", | |
author = "Sato'o, Yusuke and Hisatsune, Junzo and Hirakawa, Hideki and Ono, | |
Hisaya K and Omoe, Katsuhiko and Sugai, Motoyuki", | |
abstract = "Staphylococcus aureus No. 10 is an isolate from a staphylococcal | |
food poisoning outbreak in Japan, classified as clonal complex 81 | |
subtype 1. It preferentially produces larger quantities of | |
staphylococcal enterotoxin A (SEA) and staphylococcal enterotoxin | |
H (SEH) in foods and media. Here, we report the complete | |
annotated genome sequence of the chromosome and a plasmid.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 34, | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Sivaraman2017-uf, | |
title = "Draft Genome Sequence of a {Methicillin-Resistant} | |
\textit{Staphylococcus aureus} Isolate (Sequence Type 1) from | |
Seafood", | |
author = "Sivaraman, G K and Vanik, Deesha and Visnuvinayagam, S and | |
Prasad, M M and Ravishankar, C N", | |
abstract = "The draft genome sequence of a methicillin-resistant | |
Staphylococcus aureus (MRSA) isolate (sequence type 1 [ST 1]) | |
from the salted dried ribbonfish from Gujarat, India, is reported | |
here. Staphylococcus genus-specific genes were present in this | |
MRSA isolate. The whole-genome sequence of this strain contains | |
2,797 protein-coding genes and 80 RNAs within the 2.85-Mb genome.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 34, | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Lee2017-lr, | |
title = "Emerging multidrug resistance in community-associated | |
Staphylococcus aureus involved in skin and soft tissue infections | |
and nasal colonization", | |
author = "Lee, Grace C and Dallas, Steven D and Wang, Yufeng and Olsen, | |
Randall J and Lawson, Kenneth A and Wilson, James and Frei, | |
Christopher R", | |
abstract = "Background: Staphylococcus aureus is a major pathogen causing | |
significant morbidity and mortality worldwide. The emergence of | |
MDR S. aureus strains in the community setting has major | |
implications in disease management. However, data regarding the | |
occurrence and patterns of MDR community-associated S. aureus | |
sub-clones is limited. Objectives: To use whole-genome sequences | |
to describe the diversity and distribution of resistance | |
mechanisms among community-associated S. aureus isolates. | |
Methods: S. aureus isolates from skin and soft tissue infections | |
(SSTIs) and nasal colonization were collected from patients | |
within 10 primary care clinics from 2007 to 2015. The Illumina | |
Miseq platform was used to determine the genome sequences for 144 | |
S. aureus isolates. Phylogenetic and bioinformatics analyses were | |
performed using in silico tools. The resistome was assembled and | |
compared with the phenotypically derived antibiogram. Results: | |
Approximately one-third of S. aureus isolates in the South Texas | |
primary care setting were MDR. A higher proportion of SSTI | |
isolates were MDR in comparison with nasal colonization isolates. | |
Individuals with MDR S. aureus SSTIs were more likely to be | |
African American and obese. Furthermore, S. aureus populations | |
are able to acquire and lose antimicrobial resistance genes. | |
USA300 strains were differentiated by a stable chromosomal | |
mutation in gyrA conferring quinolone resistance. The resistomes | |
were highly predictive of antimicrobial resistance phenotypes. | |
Conclusions: These findings highlight the high prevalence and | |
epidemiological factors associated with MDR S. aureus strains in | |
the community setting and demonstrate the utility of | |
next-generation sequencing to potentially quicken antimicrobial | |
resistance detection and surveillance for targeted interventions.", | |
journal = "J. Antimicrob. Chemother.", | |
volume = 72, | |
number = 9, | |
pages = "2461--2468", | |
month = sep, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Hansen2017-hf, | |
title = "Whole Genome Sequencing of Danish \textit{Staphylococcus | |
argenteus} Reveals a Genetically Diverse Collection with Clear | |
Separation from \textit{Staphylococcus aureus}", | |
author = "Hansen, Thomas A and Bartels, Mette D and H{\o}gh, Silje V and | |
Dons, Lone E and Pedersen, Michael and Jensen, Th{\o}ger G and | |
Kemp, Michael and Skov, Marianne N and Gumpert, Heidi and | |
Worning, Peder and Westh, Henrik", | |
abstract = "Staphylococcus argenteus (S. argenteus) is a newly identified | |
Staphylococcus species that has been misidentified as | |
Staphylococcus aureus (S. aureus) and is clinically relevant. We | |
identified 25 S. argenteus genomes in our collection of whole | |
genome sequenced S. aureus. These genomes were compared to | |
publicly available genomes and a phylogeny revealed seven | |
clusters corresponding to seven clonal complexes. The genome of | |
S. argenteus was found to be different from the genome of S. | |
aureus and a core genome analysis showed that ~33\% of the total | |
gene pool was shared between the two species, at 90\% homology | |
level. An assessment of mobile elements shows flow of SCCmec | |
cassettes, plasmids, phages, and pathogenicity islands, between | |
S. argenteus and S. aureus. This dataset emphasizes that S. | |
argenteus and S. aureus are two separate species that share | |
genetic material.", | |
journal = "Front. Microbiol.", | |
volume = 8, | |
pages = "1512", | |
month = aug, | |
year = 2017, | |
keywords = "S. argenteus; S. aureus; SargPI; SargPID7903; Staphylococcus; | |
phage; | |
plasmid;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Jo2016-qi, | |
title = "Phenotypic and genotypic characterisation of multiple | |
antibiotic-resistant Staphylococcus aureus exposed to | |
subinhibitory levels of oxacillin and levofloxacin", | |
author = "Jo, Ara and Ahn, Juhee", | |
abstract = "BACKGROUND: The emergence and spread of multidrug resistant | |
methicillin-resistant Staphylococcus aureus (MDR-MRSA) has | |
serious health consequences in the presence of sub-MIC | |
antibiotics. Therefore, this study was designed to evaluate | |
$\beta$-lactamase activity, efflux activity, biofilm formation, | |
and gene expression pattern in Staphylococcus aureus KACC 10778, | |
S. aureus ATCC 15564, and S. aureus CCARM 3080 exposed to | |
sublethal concentrations of levofloxacin and oxacillin. RESULTS: | |
The decreased MICs were observed in S. aureus KACC and S. aureus | |
ATCC when exposed to levofloxacin and oxacillin, while and S. | |
aureus CCARM remained resistance to streptomycin (512 $\mu$g/mL) | |
in the presence of levofloxacin and imipenem (>512 $\mu$g/mL) in | |
the presence of oxacillin. The considerable increase in | |
extracellular and membrane-bound $\beta$-lactamase activities was | |
observed in S. aureus ATCC exposed to oxacillin (>26 | |
$\mu$mol/min/mL). The antibiotic susceptibility of all strains | |
exposed to EPIs (CCCP and PA$\beta$N) varied depending on the | |
classes of antibiotics. The relative expression levels of | |
adhesion-related genes (clfA, clfB, fnbA, fnnB, and icaD), | |
efflux-related genes (norB, norC, and qacA/B), and enterotoxin | |
gene (sec) were increased more than 5-fold in S. aureus CCARM. | |
The eno and qacA/B genes were highly overexpressed by more than | |
12- and 9-folds, respectively, in S. aureus CCARM exposed to | |
levofloxacin. The antibiotic susceptibility, lactamase activity, | |
biofilm-forming ability, efflux activity, and gene expression | |
pattern varied with the intrinsic antibiotic resistance of S. | |
aureus KACC, S. aureus ATCC, and S. aureus CCARM exposed to | |
levofloxacin and oxacillin. CONCLUSIONS: This study would provide | |
useful information for better understating of combination therapy | |
related to antibiotic resistance mechanisms and open the door for | |
designing effective antibiotic treatment protocols to prevent | |
excessive use of antibiotics in clinical practice.", | |
journal = "BMC Microbiol.", | |
volume = 16, | |
number = 1, | |
pages = "170", | |
month = jul, | |
year = 2016, | |
keywords = "Antibiotic resistance; Biofilm; Efflux pump; Gene expression; | |
Lactamase; Levofloxacin; Oxacillin; Staphylococcus | |
aureus;Staphylococcus;antibiotics;oxacillin;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Hisatsune2017-ce, | |
title = "Complete Genome Sequence of Systemically Disseminated Sequence | |
Type 8 Staphylococcal Cassette Chromosome \textit{mec} Type {IVl} | |
{Community-Acquired} {Methicillin-Resistant} | |
\textit{Staphylococcus aureus}", | |
author = "Hisatsune, Junzo and Hagiya, Hideharu and Shiota, Sumiko and | |
Sugai, Motoyuki", | |
abstract = "Staphylococcus aureus JH4899, a community-acquired | |
methicillin-resistant Staphylococcus aureus (CA-MRSA) isolate | |
collected from a patient with systematically disseminated | |
infection, is classified as sequence type 8 and carries the | |
staphylococcal cassette chromosome mec type IVl (SCCmecIVl). It | |
produces TSST-1, SEC, a newly discovered enterotoxin (SE1), and | |
epidermal cell differentiation inhibitor A (EDIN-A). Here, we | |
present the complete genome sequence of the chromosome and a | |
plasmid harboring the se1 and ednA genes.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 35, | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Zhang2017-bb, | |
title = "Genome Sequence of \textit{Staphylococcus aureus} {PX03}, an | |
{Acetoin-Producing} Strain with a {Small-Sized} Genome", | |
author = "Zhang, Ge and Wang, Qian and Su, Yulong and Li, Shugui and Liu, | |
Haobao", | |
abstract = "Staphylococcus aureus PX03 can produce acetoin efficiently. Here, | |
we present a 2.38-Mb assembly of its genome sequence, which might | |
provide further insights into the molecular mechanism of its | |
acetoin biosynthesis to further improve its biotechnological | |
applications.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 37, | |
month = sep, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Lebughe2017-qk, | |
title = "The Impact of the \textit{Staphylococcus aureus} Virulome on | |
Infection in a Developing Country: A Cohort Study", | |
author = "Lebughe, Marthe and Phaku, Patrick and Niemann, Silke and Mumba, | |
Dieudonn{\'e} and Peters, Georg and Muyembe-Tamfum, Jean-Jacques | |
and Mellmann, Alexander and Strau{\ss}, Lena and Schaumburg, | |
Frieder", | |
abstract = "We performed a cohort study to analyze the virulome of | |
Staphylococcus aureus from the Democratic Republic of the Congo | |
using whole genome sequencing and to assess its impact on the | |
course of S. aureus infections. Community-associated S. aureus | |
from nasal colonization (n = 100) and infection (n = 86) were | |
prospectively collected. Phenotypic susceptibility testing and | |
WGS was done for each isolate. WGS data were used to screen for | |
79 different virulence factors and for genotyping purposes (spa | |
typing, multilocus sequence typing). The majority of the 79 | |
virulence factors were equally distributed among isolates from | |
colonization and infection. Panton-Valentine leukocidin (PVL) and | |
the non-truncated hemolysin $\beta$ were associated with skin and | |
soft tissue infection (SSTI) and recurrence of disease but did | |
not influence the course of infection (i.e., mortality, surgical | |
intervention). For the first time, we show that not only PVL but | |
also hemolysin $\beta$ could contribute to the development of | |
SSTI in PVL-endemic areas such as Africa.", | |
journal = "Front. Microbiol.", | |
volume = 8, | |
pages = "1662", | |
month = aug, | |
year = 2017, | |
keywords = "Africa; Staphylococcus aureus; infection; virulome; whole genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Sassi2017-mx, | |
title = "An outbreak in intravenous drug users due to {USA300} | |
{Latin-American} variant community-acquired methicillin-resistant | |
Staphylococcus aureus in France as early as 2007", | |
author = "Sassi, M and Felden, B and Revest, M and Tattevin, P and | |
Augagneur, Y and Donnio, P-Y", | |
abstract = "Intravenous drug users are at increased risk of Staphylococcus | |
aureus infections. Most cases are related to clones prevalent in | |
the community. We report an outbreak of community-acquired | |
methicillin-resistant Staphylococcus aureus infections that | |
occurred from 2007 to 2009 in intravenous drug users and their | |
close contacts in Northwestern France. Clinical and molecular | |
investigations suggested that the clones were more similar than | |
those usually isolated in the American continent although none of | |
the patients traveled abroad or had contact with individuals who | |
had traveled to the Americas. Then, a retrospective whole genome | |
sequencing and phylogenetic analyses demonstrated that the | |
strains isolated from the first case belong to the USA300 | |
Latin-American variant clone, based on the absence of arginine | |
catabolic mobile element (ACME), and the presence of copper and | |
mercury resistance mobile element (COMER), a distinctive feature | |
of the South American variant. Our study shows genetic evidence | |
for introduction of this clone as early as 2007 in France. This | |
report also illustrates the importance of genome sequencing to | |
finely characterize and monitor the emergence of unexpected S. | |
aureus clones among high-risk populations, especially when living | |
in promiscuity.", | |
journal = "Eur. J. Clin. Microbiol. Infect. Dis.", | |
month = sep, | |
year = 2017, | |
keywords = "Staphylococcus;transmission-epidemiology;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Katayama2017-uq, | |
title = "Prevalence of Slow-growth Vancomycin Non-susceptibility In | |
Methicillin-resistant Staphylococcus aureus", | |
author = "Katayama, Yuki and Azechi, Takuya and Miyazaki, Motoyasu and | |
Takata, Tohru and Sekine, Miwa and Matsui, Hidehito and Hanaki, | |
Hideaki and Yahara, Koji and Sasano, Hiroshi and Asakura, Kota | |
and Takaku, Tomoiku and Ochiai, Tomonori and Komatsu, Norio and | |
Chambers, Henry F", | |
abstract = "We have previsously reported a novel phenotype of | |
vancomycin-intermediate Staphylococcus aureus (VISA) strains, | |
``slow-VISA,'' in which colonies appear only after 72 h of | |
incubation. Slow-VISA strains can be difficult to detect because | |
prolonged incubation is required and the phenotype is unstable. | |
To develop a method for detection of slow-VISA isolates, we | |
studied 23 slow-VISA isolates derived from heteroVISA clinical | |
strain Mu3. We identified single nucleotide polymorphisms (SNP) | |
in genes involved in various pathways such as purine/pyrimidine | |
synthesis, cell metabolism, and cell-wall peptidoglycan | |
synthesis, which have been implicated in the stringent response. | |
We found that mupirocin, which also induces a stringent response, | |
caused stable expression of vancomycin resistance. On the basis | |
of these results, we developed a method for detection of | |
slow-VISA using mupirocin 0.032 $\mu$g/ml (Patent Application No. | |
PCT/JP2017/008975filed on March 7, 2017). Using this method, we | |
detected 53 (15.6\%) slow-VISA isolates among clinical | |
methicillin-resistant S.aureus (MRSA) isolates. In contrast, the | |
VISA phenotype was detected in fewer than 1\% of isolates. | |
Deep-sequencing analysis showed that slow-VISA clones are present | |
in small numbers among hVISA isolates, and proliferate in the | |
presence of vancomycin. This slow-VISA subpopulation could | |
account in part for the recurrence and persistence of MRSA | |
infection.", | |
journal = "Antimicrob. Agents Chemother.", | |
month = aug, | |
year = 2017, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Stanczak-Mrozek2015-ds, | |
title = "Within-host diversity of {MRSA} antimicrobial resistances", | |
author = "Stanczak-Mrozek, Kinga I and Manne, Anusha and Knight, Gwenan M | |
and Gould, Katherine and Witney, Adam A and Lindsay, Jodi A", | |
abstract = "OBJECTIVES: MRSA is a major antimicrobial resistance (AMR) | |
pathogen. The reservoir of infecting isolates is colonization, | |
which is the site of evolutionary selection. The aim was to | |
identify if AMRs in colonizing MRSA populations diversified and | |
potential mechanisms of resistance gene transfer in vivo. | |
METHODS: Nasal swabs from 38 MRSA carriers admitted to hospital | |
were plated and 20 individual colonies from each patient tested | |
for phenotypic antibiotic susceptibility and genetically for | |
lineage, carriage of four prophages and three plasmid families. | |
Free bacteriophages were detected in swabs as well as their | |
capacity for transducing resistance genes. RESULTS: Nine (24\%) | |
patients carried phenotypic AMR variants and 24 (63\%) carried | |
prophage and plasmid variants. If a single colony was selected | |
for testing, the probability of detecting all AMR in that patient | |
was 87\%. Sixty-four different AMR and mobile genetic element | |
(MGE) profiles were detected, mostly in the MRSA CC22 background | |
(where CC stands for clonal complex), with up to 8 profiles per | |
patient. Nearly half of the patients carried detectable free | |
bacteriophages and phages successfully transduced resistance | |
genes between laboratory and patient isolates in vitro. WGS | |
showed MRSA core genomes were stable, while AMR and MGEs varied. | |
CONCLUSIONS: 'Clouds' of MRSA variants that have acquired or lost | |
AMR and MGEs are common in nasal colonizing populations and | |
bacteriophages may play an important role in gene transfer. | |
Accurate estimation of AMR and genetic variability has | |
implications for diagnostics, epidemiology, antimicrobial | |
stewardship and understanding the evolutionary selection of AMR | |
in colonizing populations.", | |
journal = "J. Antimicrob. Chemother.", | |
volume = 70, | |
number = 8, | |
pages = "2191--2198", | |
month = aug, | |
year = 2015, | |
keywords = "horizontal gene transfer; mobile genetic elements; transduction; | |
whole-genome | |
sequencing;Staphylococcus;bacerial\_genetics;colonization;phage;Staph\_evolutionary\_history;Staph\_evolutionary\_history/staph | |
microbial | |
interactions;antibiotcs-evolution-fitness;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Aanensen2016-lz, | |
title = "{Whole-Genome} Sequencing for Routine Pathogen Surveillance in | |
Public Health: a Population Snapshot of Invasive Staphylococcus | |
aureus in Europe", | |
author = "Aanensen, David M and Feil, Edward J and Holden, Matthew T G and | |
Dordel, Janina and Yeats, Corin A and Fedosejev, Artemij and | |
Goater, Richard and Castillo-Ram{\'\i}rez, Santiago and Corander, | |
Jukka and Colijn, Caroline and Chlebowicz, Monika A and Schouls, | |
Leo and Heck, Max and Pluister, Gerlinde and Ruimy, Raymond and | |
Kahlmeter, Gunnar and {\AA}hman, Jenny and Matuschek, Erika and | |
Friedrich, Alexander W and Parkhill, Julian and Bentley, Stephen | |
D and Spratt, Brian G and Grundmann, Hajo and {European SRL | |
Working Group}", | |
abstract = "UNLABELLED: The implementation of routine whole-genome sequencing | |
(WGS) promises to transform our ability to monitor the emergence | |
and spread of bacterial pathogens. Here we combined WGS data from | |
308 invasive Staphylococcus aureus isolates corresponding to a | |
pan-European population snapshot, with epidemiological and | |
resistance data. Geospatial visualization of the data is made | |
possible by a generic software tool designed for public health | |
purposes that is available at the project URL | |
(http://www.microreact.org/project/EkUvg9uY?tt=rc). Our analysis | |
demonstrates that high-risk clones can be identified on the basis | |
of population level properties such as clonal relatedness, | |
abundance, and spatial structuring and by inferring virulence and | |
resistance properties on the basis of gene content. We also show | |
that in silico predictions of antibiotic resistance profiles are | |
at least as reliable as phenotypic testing. We argue that this | |
work provides a comprehensive road map illustrating the three | |
vital components for future molecular epidemiological | |
surveillance: (i) large-scale structured surveys, (ii) WGS, and | |
(iii) community-oriented database infrastructure and analysis | |
tools. IMPORTANCE: The spread of antibiotic-resistant bacteria is | |
a public health emergency of global concern, threatening medical | |
intervention at every level of health care delivery. Several | |
recent studies have demonstrated the promise of routine | |
whole-genome sequencing (WGS) of bacterial pathogens for | |
epidemiological surveillance, outbreak detection, and infection | |
control. However, as this technology becomes more widely adopted, | |
the key challenges of generating representative national and | |
international data sets and the development of bioinformatic | |
tools to manage and interpret the data become increasingly | |
pertinent. This study provides a road map for the integration of | |
WGS data into routine pathogen surveillance. We emphasize the | |
importance of large-scale routine surveys to provide the | |
population context for more targeted or localized investigation | |
and the development of open-access bioinformatic tools to provide | |
the means to combine and compare independently generated data | |
with publicly available data sets.", | |
journal = "MBio", | |
volume = 7, | |
number = 3, | |
month = may, | |
year = 2016, | |
keywords = "cloning;Staphylococcus;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Reuter2016-yp, | |
title = "Building a genomic framework for prospective {MRSA} surveillance | |
in the United Kingdom and the Republic of Ireland", | |
author = "Reuter, Sandra and T{\"o}r{\"o}k, M Est{\'e}e and Holden, Matthew | |
T G and Reynolds, Rosy and Raven, Kathy E and Blane, Beth and | |
Donker, Tjibbe and Bentley, Stephen D and Aanensen, David M and | |
Grundmann, Hajo and Feil, Edward J and Spratt, Brian G and | |
Parkhill, Julian and Peacock, Sharon J", | |
abstract = "The correct interpretation of microbial sequencing data applied | |
to surveillance and outbreak investigation depends on accessible | |
genomic databases to provide vital genetic context. Our aim was | |
to construct and describe a United Kingdom MRSA database | |
containing over 1000 methicillin-resistant Staphylococcus aureus | |
(MRSA) genomes drawn from England, Northern Ireland, Wales, | |
Scotland, and the Republic of Ireland over a decade. We sequenced | |
1013 MRSA submitted to the British Society for Antimicrobial | |
Chemotherapy by 46 laboratories between 2001 and 2010. Each | |
isolate was assigned to a regional healthcare referral network in | |
England and was otherwise grouped based on country of origin. | |
Phylogenetic reconstructions were used to contextualize MRSA | |
outbreak investigations and to detect the spread of resistance. | |
The majority of isolates (n = 783, 77\%) belonged to CC22, which | |
contains the dominant United Kingdom epidemic clone (EMRSA-15). | |
There was marked geographic structuring of EMRSA-15, consistent | |
with widespread dissemination prior to the sampling decade | |
followed by local diversification. The addition of MRSA genomes | |
from two outbreaks and one pseudo-outbreak demonstrated the | |
certainty with which outbreaks could be confirmed or refuted. We | |
identified local and regional differences in antibiotic | |
resistance profiles, with examples of local expansion, as well as | |
widespread circulation of mobile genetic elements across the | |
bacterial population. We have generated a resource for the future | |
surveillance and outbreak investigation of MRSA in the United | |
Kingdom and Ireland and have shown the value of this during | |
outbreak investigation and tracking of antimicrobial resistance.", | |
journal = "Genome Res.", | |
volume = 26, | |
number = 2, | |
pages = "263--270", | |
month = feb, | |
year = 2016, | |
keywords = "Staphylococcus;Staph\_evolutionary\_history/staph microbial | |
interactions;Read-lab-shared;Staphylococcus\_aureus\_genome\_papers;Staph\_evolution\_toxins", | |
language = "en" | |
} | |
@ARTICLE{Dekker2016-rk, | |
title = "{Next-Generation} Epidemiology: Using {Real-Time} Core Genome | |
Multilocus Sequence Typing To Support Infection Control Policy", | |
author = "Dekker, John P and Frank, Karen M", | |
abstract = "Multidrug-resistant bacteria are responsible for substantial | |
morbidity and mortality worldwide. Tracking the nosocomial spread | |
of resistant bacteria is critical to infection control. Mellmann | |
et al. (J. Clin. Microbiol. 54:2874-2881, 2016, | |
http://dx.doi.org/10.1128/JCM.00790-16) have described | |
prospective whole-genome sequencing with core genome multilocus | |
sequencing typing (cgMLST) analysis for real-time surveillance | |
and have addressed the practical aspects of implementing this | |
type of operation in the hospital setting.", | |
journal = "J. Clin. Microbiol.", | |
volume = 54, | |
number = 12, | |
pages = "2850--2853", | |
month = dec, | |
year = 2016, | |
keywords = "Staphylococcus;clinical-diagnosis-genomics;Staphylococcus\_aureus\_genome\_papers;Anti-infectives\_course", | |
language = "en" | |
} | |
@ARTICLE{Hau2017-rx, | |
title = "Draft Genome Sequences of Nine {Livestock-Associated} | |
{Methicillin-Resistant} \textit{Staphylococcus aureus} Sequence | |
Type 5 Isolates Obtained from Humans after {Short-Term} Swine | |
Contact", | |
author = "Hau, Samantha J and Bayles, Darrell O and Alt, David P and Frana, | |
Timothy S and Nicholson, Tracy L", | |
abstract = "Livestock-associated methicillin-resistant Staphylococcus aureus | |
(LA-MRSA) sequence type 5 (ST5) has raised concerns surrounding | |
the potential for these isolates to colonize or cause disease in | |
humans with swine contact. Here, we report draft genome sequences | |
for nine LA-MRSA ST5 isolates obtained from humans after short | |
term swine contact.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 41, | |
month = oct, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Donker2017-qu, | |
title = "Population genetic structuring of methicillin-resistant | |
\textit{Staphylococcus aureus} clone {EMRSA-15} within {UK} | |
reflects patient referral patterns", | |
author = "Donker, Tjibbe and Reuter, Sandra and Scriberras, James and | |
Reynolds, Rosy and Brown, Nicholas M and T{\"o}r{\"o}k, M | |
Est{\'e}e and James, Richard and Network, East Of England | |
Microbiology Research and Aanensen, David M and Bentley, Stephen | |
D and Holden, Matthew T G and Parkhill, Julian and Spratt, Brian | |
G and Peacock, Sharon J and Feil, Edward J and Grundmann, Hajo", | |
abstract = "Antibiotic resistance forms a serious threat to the health of | |
hospitalised patients, rendering otherwise treatable bacterial | |
infections potentially life-threatening. A thorough understanding | |
of the mechanisms by which resistance spreads between patients in | |
different hospitals is required in order to design effective | |
control strategies. We measured the differences between bacterial | |
populations of 52 hospitals in the United Kingdom and Ireland, | |
using whole-genome sequences from 1085 MRSA clonal complex 22 | |
isolates collected between 1998 and 2012. The genetic differences | |
between bacterial populations were compared with the number of | |
patients transferred between hospitals and their regional | |
structure. The MRSA populations within single hospitals, regions | |
and countries were genetically distinct from the rest of the | |
bacterial population at each of these levels. Hospitals from the | |
same patient referral regions showed more similar MRSA | |
populations, as did hospitals sharing many patients. Furthermore, | |
the bacterial populations from different time-periods within the | |
same hospital were generally more similar to each other than | |
contemporaneous bacterial populations from different hospitals. | |
We conclude that, while a large part of the dispersal and | |
expansion of MRSA takes place among patients seeking care in | |
single hospitals, inter-hospital spread of resistant bacteria is | |
by no means a rare occurrence. Hospitals are exposed to constant | |
introductions of MRSA on a number of levels: (1) most MRSA is | |
received from hospitals that directly transfer large numbers of | |
patients, while (2) fewer introductions happen between regions or | |
(3) across national borders, reflecting lower numbers of | |
transferred patients. A joint coordinated control effort between | |
hospitals, is therefore paramount for the national control of | |
MRSA, antibiotic-resistant bacteria and other hospital-associated | |
pathogens.", | |
journal = "Microb Genom", | |
volume = 3, | |
number = 7, | |
pages = "e000113", | |
month = jul, | |
year = 2017, | |
keywords = "MRSA; antimicrobial Resistance; hospital | |
network;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Yu2017-kx, | |
title = "Complete Genome Sequence of Super {Biofilm-Elaborating} | |
\textit{Staphylococcus aureus} Isolated in Japan", | |
author = "Yu, Liansheng and Hisatsune, Junzo and Hirakawa, Hideki and | |
Mizumachi, Emiri and Toyoda, Atsushi and Yahara, Koji and Sugai, | |
Motoyuki", | |
abstract = "Staphylococcus aureus JP080, previously named TF2758, is a | |
clinical isolate from an atheroma and a super biofilm-elaborating | |
strain whose biofilm elaboration is dependent solely on | |
polysaccharide poly-N-acetylglucosamine/polysaccharide | |
intercellular adhesin (PNAG/PIA). Here, we report the complete | |
genome sequence of strain JP080, which consists of one chromosome | |
and one circular plasmid.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 41, | |
month = oct, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Hau2017-mo, | |
title = "Draft Genome Sequences of Nine {Livestock-Associated} | |
{Methicillin-Resistant} \textit{Staphylococcus aureus} Sequence | |
Type 5 Isolates from Humans with {Long-Term} Swine Contact", | |
author = "Hau, Samantha J and Bayles, Darrell O and Alt, David P and | |
Davies, Peter R and Haan, Jisun S and Nicholson, Tracy L", | |
abstract = "Humans have been found to harbor livestock-associated | |
methicillin-resistant Staphylococcus aureus (LA-MRSA) isolates. | |
LA-MRSA isolates are considered adapted to colonizing livestock | |
and less pathogenic in humans than their hospital- and | |
community-acquired counterparts. Here, we present nine LA-MRSA | |
sequence type 5 isolates from veterinarians with long-term swine | |
contact.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 41, | |
month = oct, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Toleman2017-de, | |
title = "Investigation of a Cluster of Sequence Type 22 | |
{Methicillin-Resistant} Staphylococcus aureus Transmission in a | |
Community Setting", | |
author = "Toleman, Michelle S and Watkins, Emmeline R and Williams, Tom and | |
Blane, Beth and Sadler, Belinda and Harrison, Ewan M and Coll, | |
Francesc and Parkhill, Julian and Nazareth, Bernadette and Brown, | |
Nicholas M and Peacock, Sharon J", | |
abstract = "BackgroundWhole-genome sequencing (WGS) has typically been used | |
to confirm or refute hospital/ward outbreaks of | |
methicillin-resistant Staphylococcus aureus (MRSA) identified | |
through routine practice. However, appropriately targeted WGS | |
strategies that identify routinely ``undetectable'' transmission | |
remain the ultimate aim.MethodsWGS of MRSA isolates sent to a | |
regional microbiological laboratory was performed as part of a | |
12-month prospective observational study. Phylogenetic analyses | |
identified a genetically related cluster of E-MRSA15 isolated | |
from patients registered to the same general practice (GP) | |
surgery. This led to an investigation to identify epidemiological | |
links, find additional cases, and determine potential for ongoing | |
transmission.ResultsWe identified 15 MRSA-positive individuals | |
with 27 highly related MRSA isolates who were linked to the GP | |
surgery, 2 of whom died with MRSA bacteremia. Of the 13 cases | |
that were further investigated, 11 had attended a leg | |
ulcer/podiatry clinic. Cases lacked epidemiological links to | |
hospitals, suggesting that transmission occurred elsewhere. | |
Environmental and staff screening at the GP surgery did not | |
identify an ongoing source of infection.ConclusionsSurveillance | |
in the United Kingdom shows that the proportion of MRSA | |
bacteremias apportioned to hospitals is decreasing, suggesting | |
the need for greater focus on the detection of MRSA outbreaks and | |
transmission in the community. This case study confirms that the | |
typically nosocomial lineage (E-MRSA15) can transmit within | |
community settings. Our study exemplifies the continued | |
importance of WGS in detecting outbreaks, including those which | |
may be missed by routine practice, and suggests that universal | |
WGS of bacteremia isolates may help detect outbreaks in | |
low-surveillance settings.", | |
journal = "Clin. Infect. Dis.", | |
month = oct, | |
year = 2017, | |
keywords = "genome; methicillin-resistant staphylococcus aureus; community; | |
disease outbreaks; surgical procedures, operative; | |
bacteremia;Staphylococcus;grants;interesting;transmission-epidemiology;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Coll_undated-fs, | |
title = "and the community", | |
author = "Coll, Francesc and Harrison, Ewan M and Toleman, Michelle S and | |
Reuter, Sandra and Raven, Kathy E and Blane, Beth and Palmer, | |
Beverley and Kappeler, A Ruth M and Brown, Nicholas M and | |
Est{\'e}e T{\"o}r{\"o}k, M and Parkhill, Julian and Peacock, | |
Sharon J", | |
keywords = "transmission-epidemiology;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Donker2017-qu, | |
title = "Population genetic structuring of methicillin-resistant | |
\textit{Staphylococcus aureus} clone {EMRSA-15} within {UK} | |
reflects patient referral patterns", | |
author = "Donker, Tjibbe and Reuter, Sandra and Scriberras, James and | |
Reynolds, Rosy and Brown, Nicholas M and Est{\'e}e | |
T{\"o}r{\"o}k, M and James, Richard and {East of England | |
Microbiology Research Network} and Aanensen, David M and | |
Bentley, Stephen D and Holden, Matthew T G and Parkhill, Julian | |
and Spratt, Brian G and Peacock, Sharon J and Feil, Edward J and | |
Grundmann, Hajo", | |
abstract = "Antibiotic resistance forms a serious threat to the health of | |
hospitalised patients, rendering otherwise treatable bacterial | |
infections potentially life-threatening. A thorough | |
understanding of the mechanisms by which resistance spreads | |
between patients in different hospitals is required in order to | |
design effective control strategies. We measured the differences | |
between bacterial populations of 52 hospitals in the United | |
Kingdom and Ireland, using whole-genome sequences from 1085 MRSA | |
clonal complex 22 isolates collected between 1998 and 2012. The | |
genetic differences between bacterial populations were compared | |
with the number of patients transferred between hospitals and | |
their regional structure. The MRSA populations within single | |
hospitals, regions and countries were genetically distinct from | |
the rest of the bacterial population at each of these levels. | |
Hospitals from the same patient referral regions showed more | |
similar MRSA populations, as did hospitals sharing many | |
patients. Furthermore, the bacterial populations from different | |
time-periods within the same hospital were generally more | |
similar to each other than contemporaneous bacterial populations | |
from different hospitals. We conclude that, while a large part | |
of the dispersal and expansion of MRSA takes place among | |
patients seeking care in single hospitals, inter-hospital spread | |
of resistant bacteria is by no means a rare occurrence. | |
Hospitals are exposed to constant introductions of MRSA on a | |
number of levels: (1) most MRSA is received from hospitals that | |
directly transfer large numbers of patients, while (2) fewer | |
introductions happen between regions or (3) across national | |
borders, reflecting lower numbers of transferred patients. A | |
joint coordinated control effort between hospitals, is therefore | |
paramount for the national control of MRSA, antibiotic-resistant | |
bacteria and other hospital-associated pathogens.", | |
journal = "Microbial Genomics", | |
publisher = "Microbiology Society", | |
volume = 3, | |
number = 7, | |
pages = "e000113", | |
month = jul, | |
year = 2017, | |
keywords = "MRSA; antimicrobial Resistance; hospital | |
network;Staphylococcus;Teaching antibiotic resistance | |
genomics;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Kumar2017-ri, | |
title = "Tuning of the Lethal Response to Multiple Stressors with a | |
{Single-Site} Mutation during Clinical Infection by | |
Staphylococcus aureus", | |
author = "Kumar, Krishan and Chen, John and Drlica, Karl and Shopsin, Bo", | |
abstract = "The agr system of Staphylococcus aureus promotes invasion of host | |
tissues, and as expected, agents that block agr quorum sensing | |
have anti-infective properties. Paradoxically, agr-defective | |
mutants are frequently recovered from patients, especially those | |
persistently infected with S. aureus. We found that an agr | |
deficiency increased survival of cultured bacteria during severe | |
stress, such as treatment with gentamicin, ciprofloxacin, heat, | |
or low pH. With daptomycin, deletion of agr decreased survival. | |
Therefore, agr activity can be either detrimental or protective, | |
depending on the type of lethal stress. Deletion of agr had no | |
effect on the ability of the antimicrobials to block bacterial | |
growth, indicating that agr effects are limited to lethal action. | |
Thus, the effect of an agr deletion is on bacterial tolerance, | |
not resistance. For gentamicin and daptomycin, activity can be | |
altered by agr-regulated secreted factors. For ciprofloxacin, a | |
detrimental function was downregulation of glutathione peroxidase | |
(bsaA), an enzyme responsible for defense against oxidative | |
stress. Deficiencies in agr and bsaA were epistatic for survival, | |
consistent with agr having a destructive role mediated by | |
reactive oxygen species. Enhanced susceptibility to lethal stress | |
by wild-type agr, particularly antimicrobial stress, helps | |
explain why inactivating mutations in S. aureus agr commonly | |
occur in hospitalized patients during infection. Moreover, the | |
agr quorum-sensing system of S. aureus provides a clinically | |
relevant example in which a single-step change in the response to | |
severe stress alters the evolutionary path of a pathogen during | |
infection.IMPORTANCE When phenotypes produced in response to an | |
environmental stress are inadequate to buffer against that | |
stress, changes that do buffer may become genetically encoded by | |
natural selection. A clinically relevant example is seen with S. | |
aureus mutants that are deficient in the key virulence regulator | |
agr. Paradoxically, defects in agr are selected during serious | |
hospital infection and have been associated with worse outcome. | |
The current work helps resolve this paradox: agr mutants are | |
often less readily killed by lethal stressors without affecting | |
MIC, a phenomenon known as tolerance. Our results indicate that | |
tolerance, which would not be detected as resistance, can be | |
selected in clinical settings. The data also support the ideas | |
that (i) S. aureus broadly hedges against environmental change | |
and stress through genome plasticity, (ii) reactive oxygen can be | |
involved in the self-destructive response in bacteria, and (iii) | |
therapeutic targeting of agr and virulence can be | |
counterproductive.", | |
journal = "MBio", | |
volume = 8, | |
number = 5, | |
month = nov, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Hau2017-di, | |
title = "Draft Genome Sequences of One {Methicillin-Sensitive} and Seven | |
{Methicillin-Resistant} \textit{Staphylococcus aureus} Sequence | |
Type 5 Isolates Obtained in California", | |
author = "Hau, Samantha J and Bayles, Darrell O and Alt, David P and | |
Nicholson, Tracy L", | |
abstract = "Staphylococcus aureus is a commensal bacterium of humans that can | |
cause a spectrum of diseases. An isolate's capacity to cause | |
disease is partially attributed to the acquisition of novel | |
mobile genetic elements. This report provides the draft genome | |
sequence of one methicillin-susceptible and seven | |
methicillin-resistant clinical human S. aureus isolates.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 42, | |
month = oct, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Lepuschitz2017-oz, | |
title = "Draft Genome Sequence of a {Community-Acquired} | |
{Methicillin-Resistant} \textit{Staphylococcus aureus} {USA300} | |
Isolate from a River Sample", | |
author = "Lepuschitz, Sarah and Mach, Robert and Springer, Burkhard and | |
Allerberger, Franz and Ruppitsch, Werner", | |
abstract = "The increasing emergence of multiresistant bacteria in health | |
care settings in the community and in the environment represents | |
a major health threat worldwide. Here, we report the draft genome | |
sequence of a community-acquired methicillin-resistant | |
Staphylococcus aureus (CA-MRSA) USA300 isolate (W1) from a small | |
river in southern Austria.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 42, | |
month = oct, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Nemet2017-kj, | |
title = "Draft Genome Sequence of an Atypical Highly Virulent Rabbit | |
\textit{Staphylococcus aureus} Strain", | |
author = "N{\'e}met, Zolt{\'a}n and Albert, Ervin and Nagy, Tibor and | |
Olasz, Ferenc and Barta, Endre and Kiss, J{\'a}nos and D{\'a}n, | |
{\'A}d{\'a}m and B{\'a}nyai, Kriszti{\'a}n and Hermans, Katleen | |
and Biksi, Imre", | |
abstract = "Rabbit staphylococcosis is one of the most important diseases in | |
industrial rabbit production. We report here the draft genome | |
sequence of Staphylococcus aureus strain 380/11, an atypical | |
highly virulent (aHV) rabbit Staphylococcus aureus strain.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 42, | |
month = oct, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Mohamed2017-ly, | |
title = "First Complete Genome Sequence of {Methicillin-Resistant} | |
\textit{Staphylococcus aureus} Strain {SO-1977} Isolated from | |
Khartoum, Sudan", | |
author = "Mohamed, Sofia B and Ali, Mohamed S and Alamir, Faisal M and | |
Alyas, Tahani B and Ahmed, Abdallah E and Seed, Almeen O and | |
Omer, Rihab A", | |
abstract = "Methicillin-resistant Staphylococcus aureus is increasingly | |
becoming resistant to most antibiotics and consequently has | |
become a challenging public health problem in Sudan. The present | |
study documented the first complete genome sequence of strain | |
SO-1977, isolated from a contaminated wound in Sudan.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 42, | |
month = oct, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Senok2017-ku, | |
title = "Investigating a rare methicillin-resistant \textit{Staphylococcus | |
aureus} strain: first description of genome sequencing and | |
molecular characterization of {CC15-MRSA}", | |
author = "Senok, Abiola C and Somily, Ali M and Slickers, Peter and Raji, | |
Muhabat A and Garaween, Ghada and Shibl, Atef and Monecke, Stefan | |
and Ehricht, Ralf", | |
abstract = "PURPOSE: Methicillin resistant Staphylococcus aureus CC15 strains | |
(CC15-MRSA) have only been sporadically described in literature. | |
This study was carried out to describe the genetic make-up for | |
this rare MRSA strain. METHODS: Four CC15-MRSA isolates collected | |
in Riyadh, Saudi Arabia, between 2013 and 2014 were studied. Two | |
isolates were from clinical infection and 2 from retail meat | |
products. Whole genome sequencing was carried out using Illumina | |
HiSeq2500 genome analyzer. RESULTS: All the CC15-MRSA isolates | |
had the multilocus sequence typing profile ST1535, | |
13-13-1-1-81-11-13, which is a single locus variant of ST15. Of | |
the 6 contigs related to the SCC element, one comprised a | |
recombinase gene ccrAA, ccrC-PM1, fusC and a helicase, another | |
one included mvaS, dru, mecA and 1 had yobV and Q4LAG7. The SCC | |
element had 5 transposase genes, namely 3 identical paralogs of | |
tnpIS431 and 2 identical paralogs of tnpIS256. Two identical | |
copies of a tnpIS256-based insertion element flank the aacA-aphD | |
gene. Two copies of this insertion element were present with 1 | |
located in the SCC element and another inserted into the sasC | |
gene. A short 3 kb region, which lacks any bacteriophage | |
structural genes and site-specific DNA integrase, was inserted | |
into the hlb gene. The hsdM and the 5'-part of the hsdS gene are | |
replaced by a copy of the hsdM/hsdS paralogs from $\nu$Sa$\beta$ | |
giving rise to a new chimeric paralog of hsdS in $\nu$Sa$\alpha$. | |
CONCLUSION: CC15-MRSA shows a novel SCCmecV/SCCfus composite | |
element. Its variant of hsdM/hsdS probably facilitated uptake of | |
foreign mobile genetic elements that promoted emergence of | |
CC15-MRSA. Close surveillance is needed to monitor spread and | |
emergence of further CC15 MRSA strains.", | |
journal = "Infect. Drug Resist.", | |
volume = 10, | |
pages = "307--315", | |
month = oct, | |
year = 2017, | |
keywords = "MLST; MRSA; SCCmec; Saudi Arabia; clonal complex; whole genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Kanjilal2017-dv, | |
title = "Trends in antibiotic susceptibility in Staphylococcus aureus in | |
Boston, Massachusetts, 2000-2014", | |
author = "Kanjilal, Sanjat and Sater, Mohamad R Abdul and Thayer, Maile and | |
Lagoudas, Georgia K and Kim, Soohong and Blainey, Paul C and | |
Grad, Yonatan H", | |
abstract = "The rate of infection by methicillin-resistant S. aureus (MRSA) | |
has declined over the past decade, but it is unclear whether this | |
represents a decline in S. aureus infections overall. To evaluate | |
trends in the annual rates of infection by S. aureus subtype and | |
mean antibiotic resistance, we conducted a 15-year retrospective | |
observational study at two tertiary care institutions in Boston, | |
MA of 31 753 adult inpatients with S. aureus isolated from | |
clinical specimens. We inferred gain and loss of methicillin | |
resistance through genome sequencing of 180 isolates from 2016. | |
Annual rates of infection by S. aureus declined from 2003 to 2014 | |
by 4.2\% (2.7\%-5.6\%), attributable to an annual decline in MRSA | |
of 10.9\% (9.3\%-12.6\%). Penicillin-susceptible S. aureus (PSSA) | |
increased by 6.1\% (4.2\%-8.1\%) annually and rates of | |
methicillin-susceptible, penicillin-resistant S. aureus (MSSA) | |
did not change. Resistance in S. aureus decreased from 2000 to | |
2014 by 0.8 antibiotics (0.7-0.8). Within common MRSA clonal | |
complexes, 3/14 MSSA and 2/21 PSSA isolates arose from the loss | |
of resistance-conferring genes. Overall, in two tertiary care | |
institutions in Boston, MA, a decline in S. aureus infections has | |
been accompanied by a shift towards increased antibiotic | |
susceptibility. The rise in PSSA makes penicillin an increasingly | |
viable treatment option.", | |
journal = "J. Clin. Microbiol.", | |
month = nov, | |
year = 2017, | |
keywords = "Staphylococcus;antibiotics;transmission-epidemiology;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Zhang2017-ar, | |
title = "Preliminary comparative genomics revealed pathogenic potential | |
and international spread of Staphylococcus argenteus", | |
author = "Zhang, Dao-Feng and Zhi, Xiao-Yang and Zhang, Jing and Paoli, | |
George C and Cui, Yan and Shi, Chunlei and Shi, Xianming", | |
abstract = "BACKGROUND: Staphylococcus argenteus and S. schweitzeri, were | |
recently proposed as novel species within S. aureus complex | |
(SAC). S. argenteus has been reported in many countries and can | |
threaten human health. S. schweitzeri has not been associated | |
with human infections, but has been isolated from non-human | |
primates. Questions regarding the evolution of pathogenicity of | |
these two species will remain elusive until an exploratory | |
evolutionary framework is established. RESULTS: We present | |
genomic comparison analysis among members of SAC based on a | |
pan-genome definition, which included 15 S. argenteus genomes | |
(five newly sequenced), six S. schweitzeri genomes and 30 | |
divergent S. aureus genomes. The three species had divergent core | |
genomes and rare interspecific recombination was observed among | |
the core genes. However, some subtypes of staphylococcal cassette | |
chromosome mec (SCCmec) elements and prophages were present in | |
different species. Of 111 tested virulence genes of S. aureus, 85 | |
and 86 homologous genes were found in S. argenteus and S. | |
schweitzeri, respectively. There was no difference in virulence | |
gene content among the three species, but the sequence of most | |
core virulence genes was divergent. Analysis of the agr locus and | |
the genes in the capsular polysaccharides biosynthetic operon | |
revealed that they both diverged before the speciation of SAC | |
members. Furthermore, the widespread geographic distribution of | |
S. argenteus, sequence type 2250, showed ambiguous | |
biogeographical structure among geographically isolated | |
populations, demonstrating an international spread of this | |
pathogen. CONCLUSIONS: S. argenteus has spread among several | |
countries, and invasive infections and persistent carriage may be | |
not limited to currently reported regions. S. argenteus probably | |
had undergone a recent host adaption and can cause human | |
infections with a similar pathogenic potential.", | |
journal = "BMC Genomics", | |
volume = 18, | |
number = 1, | |
pages = "808", | |
month = oct, | |
year = 2017, | |
keywords = "Agr; Biogeographical structure; Capsular polysaccharides; | |
Comparative genomics; Staphylococcus argenteus; Staphylococcus | |
aureus; Staphylococcus schweitzeri; Virulence | |
gene;Staphylococcus;confirmed\_SA-resistance-mutation;pangenome;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Amissah2017-yh, | |
title = "Methicillin Resistant \textit{Staphylococcus aureus} Transmission | |
in a Ghanaian Burn Unit: The Importance of Active Surveillance in | |
{Resource-Limited} Settings", | |
author = "Amissah, Nana Ama and Buultjens, Andrew H and Ablordey, Anthony | |
and van Dam, Lieke and Opoku-Ware, Ampomah and Baines, Sarah L | |
and Bulach, Dieter and Tetteh, Caitlin S and Prah, Isaac and van | |
der Werf, Tjip S and Friedrich, Alexander W and Seemann, Torsten | |
and van Dijl, Jan Maarten and Stienstra, Ymkje and Stinear, | |
Timothy P and Rossen, John W", | |
abstract = "Objectives:Staphylococcus aureus infections in burn patients can | |
lead to serious complications and death. The frequency of S. | |
aureus infection is high in low- and middle-income countries | |
presumably due to limited resources, misuse of antibiotics and | |
poor infection control. The objective of the present study was to | |
apply population genomics to precisely define, for the first | |
time, the transmission of antibiotic resistant S. aureus in a | |
resource-limited setting in sub-Saharan Africa. | |
Methods:Staphylococcus aureus surveillance was performed amongst | |
burn patients and healthcare workers during a 7-months survey | |
within the burn unit of the Korle Bu Teaching Hospital in Ghana. | |
Results: Sixty-six S. aureus isolates (59 colonizing and 7 | |
clinical) were obtained from 31 patients and 10 healthcare | |
workers. Twenty-one of these isolates were ST250-IV | |
methicillin-resistant S. aureus (MRSA). Notably, 25 (81\%) of the | |
31 patients carried or were infected with S. aureus within 24 h | |
of admission. Genome comparisons revealed six distinct S. aureus | |
clones circulating in the burn unit, and demonstrated multiple | |
transmission events between patients and healthcare workers. | |
Further, the collected S. aureus isolates exhibited a wide range | |
of genotypic resistances to antibiotics, including trimethoprim | |
(21\%), aminoglycosides (33\%), oxacillin (33\%), chloramphenicol | |
(50\%), tetracycline (59\%) and fluoroquinolones (100\%). | |
Conclusion: Population genomics uncovered multiple transmission | |
events of S. aureus, especially MRSA, within the investigated | |
burn unit. Our findings highlight lapses in infection control and | |
prevention, and underscore the great importance of active | |
surveillance to protect burn victims against multi-drug resistant | |
pathogens in resource-limited settings.", | |
journal = "Front. Microbiol.", | |
volume = 8, | |
pages = "1906", | |
month = oct, | |
year = 2017, | |
keywords = "Staphylococcus aureus; burn patients; comparative genomics; | |
healthcare workers; transmission | |
events;Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Coll2017-jb, | |
title = "Longitudinal genomic surveillance of {MRSA} in the {UK} reveals | |
transmission patterns in hospitals and the community", | |
author = "Coll, Francesc and Harrison, Ewan M and Toleman, Michelle S and | |
Reuter, Sandra and Raven, Kathy E and Blane, Beth and Palmer, | |
Beverley and Kappeler, A Ruth M and Brown, Nicholas M and | |
T{\"o}r{\"o}k, M Est{\'e}e and Parkhill, Julian and Peacock, | |
Sharon J", | |
abstract = "Genome sequencing has provided snapshots of the transmission of | |
methicillin-resistant Staphylococcus aureus (MRSA) during | |
suspected outbreaks in isolated hospital wards. Scale-up to | |
populations is now required to establish the full potential of | |
this technology for surveillance. We prospectively identified all | |
individuals over a 12-month period who had at least one | |
MRSA-positive sample processed by a routine diagnostic | |
microbiology laboratory in the East of England, which received | |
samples from three hospitals and 75 general practitioner (GP) | |
practices. We sequenced at least 1 MRSA isolate from 1465 | |
individuals (2282 MRSA isolates) and recorded epidemiological | |
data. An integrated epidemiological and phylogenetic analysis | |
revealed 173 transmission clusters containing between 2 and 44 | |
cases and involving 598 people (40.8\%). Of these, 118 clusters | |
(371 people) involved hospital contacts alone, 27 clusters (72 | |
people) involved community contacts alone, and 28 clusters (157 | |
people) had both types of contact. Community- and | |
hospital-associated MRSA lineages were equally capable of | |
transmission in the community, with instances of spread in | |
households, long-term care facilities, and GP practices. Our | |
study provides a comprehensive picture of MRSA transmission in a | |
sampled population of 1465 people and suggests the need to review | |
existing infection control policy and practice.", | |
journal = "Sci. Transl. Med.", | |
volume = 9, | |
number = 413, | |
month = oct, | |
year = 2017, | |
keywords = "Staphylococcus;confirmed\_SA-resistance-mutation;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Wan2017-xr, | |
title = "Genomic comparison between Staphylococcus aureus {GN} strains | |
clinically isolated from a familial infection case: {IS1272} | |
transposition through a novel inverted repeat-replacing mechanism", | |
author = "Wan, Tsai-Wen and Higuchi, Wataru and Khokhlova, Olga E and Hung, | |
Wei-Chun and Iwao, Yasuhisa and Wakayama, Masataka and Inomata, | |
Noriyoshi and Takano, Tomomi and Lin, Yu-Tzu and Peryanova, Olga | |
V and Kojima, Kenji K and Salmina, Alla B and Teng, Lee-Jene and | |
Yamamoto, Tatsuo", | |
abstract = "A bacterial insertion sequence (IS) is a mobile DNA sequence | |
carrying only the transposase gene (tnp) that acts as a mutator | |
to disrupt genes, alter gene expressions, and cause genomic | |
rearrangements. ``Canonical'' ISs have historically been | |
characterized by their terminal inverted repeats (IRs), which may | |
form a stem-loop structure, and duplications of a short (non-IR) | |
target sequence at both ends, called target site duplications | |
(TSDs). The IS distributions and virulence potentials of | |
Staphylococcus aureus genomes in familial infection cases are | |
unclear. Here, we determined the complete circular genome | |
sequences of familial strains from a Panton-Valentine leukocidin | |
(PVL)-positive ST50/agr4 S. aureus (GN) infection of a 4-year old | |
boy with skin abscesses. The genomes of the patient strain (GN1) | |
and parent strain (GN3) were rich for ``canonical'' IS1272 with | |
terminal IRs, both having 13 commonly-existing copies | |
(ce-IS1272). Moreover, GN1 had a newly-inserted IS1272 | |
(ni-IS1272) on the PVL-converting prophage, while GN3 had two | |
copies of ni-IS1272 within the DNA helicase gene and near rot. | |
The GN3 genome also had a small deletion. The targets of | |
ni-IS1272 transposition were IR structures, in contrast with | |
previous ``canonical'' ISs. There were no TSDs. Based on a | |
database search, the targets for ce-IS1272 were IRs or | |
``non-IRs''. IS1272 included a larger structure with tandem | |
duplications of the left (IRL) side sequence; tnp included minor | |
cases of a long fusion form and truncated form. One ce-IS1272 was | |
associated with the segments responsible for immune evasion and | |
drug resistance. Regarding virulence, GN1 expressed cytolytic | |
peptides (phenol-soluble modulin $\alpha$ and $\delta$-hemolysin) | |
and PVL more strongly than some other familial strains. These | |
results suggest that IS1272 transposes through an IR-replacing | |
mechanism, with an irreversible process unlike that of | |
``canonical'' transpositions, resulting in genomic variations, | |
and that, among the familial strains, the patient strain has | |
strong virulence potential based on community-associated | |
virulence factors.", | |
journal = "PLoS One", | |
volume = 12, | |
number = 11, | |
pages = "e0187288", | |
month = nov, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Stieber2017-ig, | |
title = "{PVL} overexpression due to genomic rearrangements and mutations | |
in the S. aureus reference strain {ATCC25923}", | |
author = "Stieber, Bettina and Sabat, Artur and Monecke, Stefan and | |
Slickers, Peter and Akkerboom, Viktoria and M{\"u}ller, Elke and | |
Friedrich, Alexander W and Ehricht, Ralf", | |
abstract = "OBJECTIVE: ATCC25923 is a Staphylococcus aureus strain that is | |
positive for the Panton Valentin leukocidin. It has been used for | |
decades as reference strain. We observed that two separately | |
maintained clones of ATCC25923 (``G477 and G478'') differed | |
grossly in the expression of this toxin. For that reason, both | |
clones were sequenced using an Illumina MiSeq instrument. After | |
assembling, the final sequences were analyzed and mapped to a | |
previously published ATCC25923 sequence (GenBank CP009361) using | |
bl2seq from the NCBI Blast2 package. RESULTS: The genomes of G477 | |
and G478 size 2778,859 and 2792,213 nucleotides, respectively. | |
Both genomes include a circular plasmid of 27,490 nucleotides. | |
The sequence of the G477 chromosome maps nearly exactly to | |
CP009361. G478 has a slightly larger size because of the presence | |
of an additional transposable element tnp13k. The second copy of | |
that tnp13k element is located in an intergenic region between | |
the genes mazF and rsbU. The sequences of the ATCC25923 clones | |
G477 and G478 differ mainly in the insertion of a second tnp13k | |
element between the genes mazF and rsbU. That insertion may lead | |
to a different transcription of that genome region resulting in | |
upregulation of the expression of the Panton-Valentine leukocidin | |
in the ATCC25923 clone G478.", | |
journal = "BMC Res. Notes", | |
volume = 10, | |
number = 1, | |
pages = "576", | |
month = nov, | |
year = 2017, | |
keywords = "Panton-Valentine leukocidin (PVL); Protein expression; Sequence | |
mutation; Staphylococcus | |
aureus;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Takeuchi2016-jc, | |
title = "The Presence of Two {Receptor-Binding} Proteins Contributes to | |
the Wide Host Range of Staphylococcal {Twort-Like} Phages", | |
author = "Takeuchi, Ippei and Osada, Keita and Azam, Aa Haeruman and | |
Asakawa, Hiroaki and Miyanaga, Kazuhiko and Tanji, Yasunori", | |
abstract = "UNLABELLED: Thanks to their wide host range and virulence, | |
staphylococcal bacteriophages (phages) belonging to the genus | |
Twortlikevirus (staphylococcal Twort-like phages) are regarded as | |
ideal candidates for clinical application for Staphylococcus | |
aureus infections due to the emergence of antibiotic-resistant | |
bacteria of this species. To increase the usability of these | |
phages, it is necessary to understand the mechanism underlying | |
host recognition, especially the receptor-binding proteins (RBPs) | |
that determine host range. In this study, we found that the | |
staphylococcal Twort-like phage $\Phi$SA012 possesses at least | |
two RBPs. Genomic analysis of five mutant phages of $\Phi$SA012 | |
revealed point mutations in orf103, in a region unique to | |
staphylococcal Twort-like phages. Phages harboring mutated ORF103 | |
could not infect S. aureus strains in which wall teichoic acids | |
(WTAs) are glycosylated with $\alpha$-N-acetylglucosamine | |
($\alpha$-GlcNAc). A polyclonal antibody against ORF103 also | |
inhibited infection by $\Phi$SA012 in the presence of | |
$\alpha$-GlcNAc, suggesting that ORF103 binds to $\alpha$-GlcNAc. | |
In contrast, a polyclonal antibody against ORF105, a short tail | |
fiber component previously shown to be an RBP, inhibited phage | |
infection irrespective of the presence of $\alpha$-GlcNAc. | |
Immunoelectron microscopy indicated that ORF103 is a tail fiber | |
component localized at the bottom of the baseplate. From these | |
results, we conclude that ORF103 binds $\alpha$-GlcNAc in WTAs, | |
whereas ORF105, the primary RBP, is likely to bind the WTA | |
backbone. These findings provide insight into the infection | |
mechanism of staphylococcal Twort-like phages. IMPORTANCE: | |
Staphylococcus phages belonging to the genus Twortlikevirus | |
(called staphylococcal Twort-like phages) are considered | |
promising agents for control of Staphylococcus aureus due to | |
their wide host range and highly lytic capabilities. Although | |
staphylococcal Twort-like phages have been studied widely for | |
therapeutic purposes, the host recognition process of | |
staphylococcal Twort-like phages remains unclear. This work | |
provides new findings about the mechanisms of host recognition of | |
the staphylococcal Twort-like phage $\Phi$SA012. The details of | |
the host recognition mechanism of $\Phi$SA012 will allow us to | |
analyze the mechanisms of infection and expand the utility of | |
staphylococcal Twort-like phages for the control of S. aureus.", | |
journal = "Appl. Environ. Microbiol.", | |
volume = 82, | |
number = 19, | |
pages = "5763--5774", | |
month = oct, | |
year = 2016, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Sivaraman2017-ta, | |
title = "Draft Genome Sequence of a {Methicillin-Resistant} Sequence Type | |
39 Staphylococcal Isolate Obtained from Seafood", | |
author = "Sivaraman, G K and Vanik, Deesha and Visnuvinayagam, S and | |
Prasad, Mothadaka Mukteswar and Murugadas, V and Nadella, Ranjit | |
Kumar and Ravishankar, C N", | |
abstract = "The draft genome sequence of a methicillin-resistant | |
Staphylococcus aureus (MRSA) sequence type 39 (ST 39) isolate | |
obtained from the dried ribbonfish of Gujarat, India, is reported | |
here. Staphylococcus-specific genes were present in this MRSA | |
isolate. The whole-genome sequence of this strain contains 2,693 | |
protein-coding genes and 70 RNAs within the 2.82-Mb genome.", | |
journal = "Genome Announc.", | |
volume = 5, | |
number = 45, | |
month = nov, | |
year = 2017, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Shylo_E_Wardyn2018-vx, | |
title = "{Whole-Genome} Analysis of Recurrent Staphylococcus aureus | |
{t571/ST398} Infection in Farmer, Iowa, {USA}", | |
author = "{Shylo E. Wardyn} and {Marc Stegger} and {Lance B. Price} and | |
{Tara C. Smith}", | |
abstract = "Staphylococcus aureus strain sequence type (ST) 398 has emerged | |
during the last decade, largely among persons who have contact | |
with swine or other livestock. Although colonization with ST398 | |
is common in livestock workers, infections are not frequently | |
documented. We report recurrent ST398-IIa infection in an Iowa | |
farmer in contact with swine and cattle.", | |
journal = "Emerging Infectious Disease journal", | |
volume = 24, | |
number = 1, | |
year = 2018, | |
keywords = "antibiotic resistance; antimicrobial resistance; bacteria; | |
farmer; farming; Iowa; livestock; methicillin-resistant | |
Staphylococcus aureus; methicillin-sensitive S. aureus; MRSA; | |
MSSA; sequence types; swine; United States; whole-genome | |
analysis; | |
zoonosis;Staphylococcus;Staphylococcus\_aureus\_genome\_papers" | |
} | |
@ARTICLE{Lilje2017-hx, | |
title = "Whole-genome sequencing of bloodstream Staphylococcus aureus | |
isolates does not distinguish bacteraemia from endocarditis", | |
author = "Lilje, Berit and Rasmussen, Rasmus Vedby and Dahl, Anders and | |
Stegger, Marc and Skov, Robert Leo and Fowler, Vance G and Ng, | |
Kim Lee and Kiil, Kristoffer and Larsen, Anders Rhod and | |
Petersen, Andreas and Johansen, Helle Krogh and Sch{\o}nheyder, | |
Henrik Carl and Arpi, Magnus and Rosenvinge, Flemming S and | |
Korup, Eva and H{\o}st, Ulla and Hassager, Christian and Gill, | |
Sabine Ute Alice and Hansen, Thomas Fritz and Johannesen, Thor | |
Bech and Smit, Jesper and S{\o}gaard, Peter and Skytt Andersen, | |
Paal and Eske-Bruun, Niels", | |
abstract = "Most Staphylococcus aureus isolates can cause invasive disease | |
given the right circumstances, but it is unknown if some isolates | |
are more likely to cause severe infections than others. S. aureus | |
bloodstream isolates from 120 patients with definite infective | |
endocarditis and 121 with S. aureus bacteraemia without infective | |
endocarditis underwent whole-genome sequencing. Genome-wide | |
association analysis was performed using a variety of | |
bioinformatics approaches including SNP analysis, accessory | |
genome analysis and k-mer based analysis. Core and accessory | |
genome analyses found no association with either of the two | |
clinical groups. In this study, the genome sequences of S. aureus | |
bloodstream isolates did not discriminate between bacteraemia and | |
infective endocarditis. Based on our study and the current | |
literature, it is not convincing that a specific S. aureus | |
genotype is clearly associated to infective endocarditis in | |
patients with S. aureus bacteraemia.", | |
journal = "Microb Genom", | |
volume = 3, | |
number = 11, | |
month = nov, | |
year = 2017, | |
keywords = "Staphylococcus aureus; bacteraemia; genome-wide association; | |
infective | |
endocarditis;Staphylococcus;Staphylococcus\_aureus\_genome\_papers;GWAS-bacteria", | |
language = "en" | |
} | |
@ARTICLE{Carter2017-go, | |
title = "Topical antibiotic use co-selects for the carriage of mobile | |
genetic elements conferring resistance to unrelated | |
antimicrobials in \textit{Staphylococcus aureus}", | |
author = "Carter, Glen P and Schultz, Mark B and Baines, Sarah L and | |
Goncalves da Silva, Anders and Heffernan, Helen and Tiong, Audrey | |
and Pham, Peter H and Monk, Ian R and Stinear, Timothy P and | |
Howden, Benjamin P and Williamson, Deborah A", | |
abstract = "Topical antibiotics such as mupirocin and fusidic acid are | |
commonly used in the prevention and treatment of skin infections, | |
particularly those caused by staphylococci. However, widespread | |
use of these agents is associated with increased resistance to | |
these agents, potentially limiting their efficacy. Of particular | |
concern is the observation that resistance to topical antibiotics | |
is often associated with multidrug resistance, suggesting that | |
topical antibiotics may play a role in the emergence of | |
multidrug-resistant (MDR) strains.New Zealand (NZ) has some of | |
the highest globally recorded rates of topical antibiotic usage | |
and resistance. Using a combination of Pacific Biosciences Single | |
Molecule Real Time (SMRT) whole genome sequencing, Illumina short | |
read sequencing and Bayesian phylogenomic modelling on 118 new | |
multilocus sequence type 1 (ST1) community Staphylococcus aureus | |
isolates from New Zealand and 61 publically available | |
international ST1 genome sequences, we demonstrate a strong | |
correlation between the clinical introduction of topical | |
antibiotics and the emergence of MDR ST1 S. aureus We also | |
provide in vitro experimental evidence showing that exposure to | |
topical antibiotics can lead to the rapid selection of MDR S. | |
aureus isolates carrying plasmids that confer resistance to | |
multiple unrelated antibiotics, from within a mixed population of | |
competitor strains. These findings have important implications | |
regarding the impact of the indiscriminate use of topical | |
antibiotics.", | |
journal = "Antimicrob. Agents Chemother.", | |
month = dec, | |
year = 2017, | |
keywords = "Staphylococcus;antibiotics;chlorhexidine;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Chen2018-bt, | |
title = "Draft Genome Sequence of {USA100} {Methicillin-Resistant} | |
\textit{Staphylococcus aureus} Strain 209", | |
author = "Chen, Yingfeng and Crosby, Heidi A and Oosthuysen, 2nd, Wilhelm F | |
and Diekema, Daniel J and Kelley, Scott T and Horswill, Alexander | |
R", | |
abstract = "USA100 strains are significant contributors to the overall burden | |
of health care-associated methicillin-resistant Staphylococcus | |
aureus (MRSA) infections. Strain 209 is a representative MRSA | |
isolate that serves as a model organism for agr type II studies | |
and USA100 virulence assessments. We present a draft genome | |
sequence of this strain.", | |
journal = "Genome Announc.", | |
volume = 6, | |
number = 1, | |
month = jan, | |
year = 2018, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Challagundla2018-vw, | |
title = "Range Expansion and the Origin of {USA300} North American | |
Epidemic {Methicillin-Resistant} Staphylococcus aureus", | |
author = "Challagundla, Lavanya and Luo, Xiao and Tickler, Isabella A and | |
Didelot, Xavier and Coleman, David C and Shore, Anna C and | |
Coombs, Geoffrey W and Sordelli, Daniel O and Brown, Eric L and | |
Skov, Robert and Larsen, Anders Rhod and Reyes, Jinnethe and | |
Robledo, Iraida E and Vazquez, Guillermo J and Rivera, Raul and | |
Fey, Paul D and Stevenson, Kurt and Wang, Shu-Hua and Kreiswirth, | |
Barry N and Mediavilla, Jose R and Arias, Cesar A and Planet, | |
Paul J and Nolan, Rathel L and Tenover, Fred C and Goering, | |
Richard V and Robinson, D Ashley", | |
abstract = "The USA300 North American epidemic (USA300-NAE) clone of | |
methicillin-resistant Staphylococcus aureus has caused a wave of | |
severe skin and soft tissue infections in the United States since | |
it emerged in the early 2000s, but its geographic origin is | |
obscure. Here we use the population genomic signatures expected | |
from the serial founder effects of a geographic range expansion | |
to infer the origin of USA300-NAE and identify polymorphisms | |
associated with its spread. Genome sequences from 357 isolates | |
from 22 U.S. states and territories and seven other countries are | |
compared. We observe two significant signatures of range | |
expansion, including decreases in genetic diversity and increases | |
in derived allele frequency with geographic distance from the | |
Pennsylvania region. These signatures account for approximately | |
half of the core nucleotide variation of this clone, occur genome | |
wide, and are robust to heterogeneity in temporal sampling of | |
isolates, human population density, and recombination detection | |
methods. The potential for positive selection of a gyrA | |
fluoroquinolone resistance allele and several intergenic regions, | |
along with a 2.4 times higher recombination rate in a resistant | |
subclade, is noted. These results are the first to show a pattern | |
of genetic variation that is consistent with a range expansion of | |
an epidemic bacterial clone, and they highlight a rarely | |
considered but potentially common mechanism by which genetic | |
drift may profoundly influence bacterial genetic | |
variation.IMPORTANCE The process of geographic spread of an | |
origin population by a series of smaller populations can result | |
in distinctive patterns of genetic variation. We detect these | |
patterns for the first time with an epidemic bacterial clone and | |
use them to uncover the clone's geographic origin and variants | |
associated with its spread. We study the USA300 clone of | |
methicillin-resistant Staphylococcus aureus, which was first | |
noticed in the early 2000s and subsequently became the leading | |
cause of skin and soft tissue infections in the United States. | |
The eastern United States is the most likely origin of epidemic | |
USA300. Relatively few variants, which include an antibiotic | |
resistance mutation, have persisted during this clone's spread. | |
Our study suggests that an early chapter in the genetic history | |
of this epidemic bacterial clone was greatly influenced by random | |
subsampling of isolates during the clone's geographic spread.", | |
journal = "MBio", | |
volume = 9, | |
number = 1, | |
month = jan, | |
year = 2018, | |
keywords = "epidemics; fluoroquinolones; founder effects; genetic drift; | |
population genetics; range | |
expansion;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Delgado-Baquerizo2018-zw, | |
title = "A global atlas of the dominant bacteria found in soil", | |
author = "Delgado-Baquerizo, Manuel and Oliverio, Angela M and Brewer, | |
Tess E and Benavent-Gonz{\'a}lez, Alberto and Eldridge, David J | |
and Bardgett, Richard D and Maestre, Fernando T and Singh, | |
Brajesh K and Fierer, Noah", | |
abstract = "The immense diversity of soil bacterial communities has stymied | |
efforts to characterize individual taxa and document their | |
global distributions. We analyzed soils from 237 locations | |
across six continents and found that only 2\% of bacterial | |
phylotypes (~500 phylotypes) consistently accounted for almost | |
half of the soil bacterial communities worldwide. Despite the | |
overwhelming diversity of bacterial communities, relatively few | |
bacterial taxa are abundant in soils globally. We clustered | |
these dominant taxa into ecological groups to build the first | |
global atlas of soil bacterial taxa. Our study narrows down the | |
immense number of bacterial taxa to a ``most wanted'' list that | |
will be fruitful targets for genomic and cultivation-based | |
efforts aimed at improving our understanding of soil microbes | |
and their contributions to ecosystem functioning.", | |
journal = "Science", | |
publisher = "American Association for the Advancement of Science", | |
volume = 359, | |
number = 6373, | |
pages = "320--325", | |
month = jan, | |
year = 2018, | |
keywords = "microbiome;climate;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Becker2018-xs, | |
title = "{Plasmid-Encoded} Transferable {mecB-Mediated} Methicillin | |
Resistance in Staphylococcus aureus", | |
author = "Becker, Karsten and van Alen, Sarah and Idelevich, Evgeny A and | |
Schleimer, Nina and Seggewi{\ss}, Jochen and Mellmann, Alexander | |
and Kaspar, Ursula and Peters, Georg", | |
abstract = "During cefoxitin-based nasal screening, phenotypically | |
categorized methicillin-resistant Staphylococcus aureus (MRSA) | |
was isolated and tested negative for the presence of the mecA and | |
mecC genes as well as for the SCCmec-orfX junction region. The | |
isolate was found to carry a mecB gene previously described for | |
Macrococcus caseolyticus but not for staphylococcal species. The | |
gene is flanked by $\beta$-lactam regulatory genes similar to | |
mecR, mecI, and blaZ and is part of an 84.6-kb | |
multidrug-resistance plasmid that harbors genes encoding | |
additional resistances to aminoglycosides (aacA-aphD, aphA, and | |
aadK) as well as macrolides (ermB) and tetracyclines (tetS). This | |
further plasmidborne $\beta$-lactam resistance mechanism harbors | |
the putative risk of acceleration or reacceleration of MRSA | |
spread, resulting in broad ineffectiveness of $\beta$-lactams as | |
a main therapeutic application against staphylococcal infections.", | |
journal = "Emerg. Infect. Dis.", | |
volume = 24, | |
number = 2, | |
pages = "242--248", | |
month = feb, | |
year = 2018, | |
keywords = "MRSA; Macrococcus; Staphylococcaceae; Staphylococcus aureus; | |
antibacterial agents; antimicrobial resistance; bacteria; mecB; | |
methicillin-resistant Staphylococcus aureus; microbial genome; | |
plasmids; | |
staphylococci;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Ronco2018-qi, | |
title = "Genomic investigation of Staphylococcus aureus isolates from bulk | |
tank milk and dairy cows with clinical mastitis", | |
author = "Ronco, Troels and Klaas, Ilka C and Stegger, Marc and Svennesen, | |
Line and Astrup, L{\ae}rke B and Farre, Michael and Pedersen, | |
Karl", | |
abstract = "Staphylococcus aureus is one of the most common pathogens that | |
cause mastitis in dairy cows. Various subtypes, virulence genes | |
and mobile genetic elements have been associated with isolates | |
from bulk tank milk and clinical mastitis. So far, no Danish | |
cattle associated S. aureus isolates have been whole-genome | |
sequenced and further analyzed. Thus, the main objective was to | |
investigate the population structure and genomic content of | |
isolates from bulk tank milk and clinical mastitis, using | |
whole-genome sequencing. This may reveal the origin of strains | |
that cause clinical mastitis. S. aureus isolates from bulk tank | |
milk (n = 94) and clinical mastitis (n = 63) were collected from | |
91 and 24 different farms, respectively and whole-genome | |
sequenced. The genomic content was analyzed and a phylogenetic | |
tree based on single nucleotide polymorphisms was constructed. In | |
general, the isolates from both bulk tank milk and clinical | |
mastitis were of similar genetic background. This suggests that | |
dairy cows are natural carriers of the S. aureus subtypes that | |
cause clinical mastitis if the right conditions are present and | |
that a broad range of subtypes cause mastitis. A phylogenetic | |
cluster that mostly consisted of ST151 isolates carried three | |
mobile genetic elements that were primarily found in this group. | |
The prevalence of resistance genes was generally low. However, | |
the first ST398 methicillin resistant S. aureus isolate from a | |
Danish dairy cow with clinical mastitis was detected.", | |
journal = "Vet. Microbiol.", | |
volume = 215, | |
pages = "35--42", | |
month = feb, | |
year = 2018, | |
keywords = "Bulk tank milk; Clinical mastitis; Genomic analysis; S. aureus; | |
Virulence and resistance | |
genes;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Choe2018-me, | |
title = "Genome-scale analysis of Methicillin-resistant Staphylococcus | |
aureus {USA300} reveals a tradeoff between pathogenesis and drug | |
resistance", | |
author = "Choe, Donghui and Szubin, Richard and Dahesh, Samira and Cho, | |
Suhyung and Nizet, Victor and Palsson, Bernhard and Cho, | |
Byung-Kwan", | |
abstract = "Staphylococcus aureus infection is a rising public health care | |
threat. S. aureus is believed to have elaborate regulatory | |
networks that orchestrate its virulence. Despite its importance, | |
the systematic understanding of the transcriptional landscape of | |
S. aureus is limited. Here, we describe the primary transcriptome | |
landscape of an epidemic USA300 isolate of community-acquired | |
methicillin-resistant S. aureus. We experimentally determined | |
1,861 transcription start sites with their principal promoter | |
elements, including well-conserved -35 and -10 elements and | |
weakly conserved -16 element and 5' untranslated regions | |
containing AG-rich Shine-Dalgarno sequence. In addition, we | |
identified 225 genes whose transcription was initiated from | |
multiple transcription start sites, suggesting potential | |
regulatory functions at transcription level. Along with the | |
transcription unit architecture derived by integrating the | |
primary transcriptome analysis with operon prediction, the | |
measurement of differential gene expression revealed the | |
regulatory framework of the virulence regulator Agr, the | |
SarA-family transcriptional regulators, and $\beta$-lactam | |
resistance regulators. Interestingly, we observed a complex | |
interplay between virulence regulation, $\beta$-lactam | |
resistance, and metabolism, suggesting a possible tradeoff | |
between pathogenesis and drug resistance in the USA300 strain. | |
Our results provide platform resource for the location of | |
transcription initiation and an in-depth understanding of | |
transcriptional regulation of pathogenesis, virulence, and | |
antibiotic resistance in S. aureus.", | |
journal = "Sci. Rep.", | |
volume = 8, | |
number = 1, | |
pages = "2215", | |
month = feb, | |
year = 2018, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Suligoy2018-de, | |
title = "Mutation of Agr Is Associated with the Adaptation | |
ofStaphylococcus aureusto the Host during Chronic Osteomyelitis", | |
author = "Suligoy, Carlos M and Lattar, Santiago M and Noto Llana, | |
Mari{\'a}ngeles and Gonz{\'a}lez, Cintia D and Alvarez, | |
Luc{\'\i}a P and Robinson, D Ashley and G{\'o}mez, Marisa I and | |
Buzzola, Fernanda R and Sordelli, Daniel O", | |
abstract = "Selection pressures exerted onStaphylococcus aureusby host | |
factors may lead to the emergence of mutants better adapted to | |
the evolving conditions at the infection site. This study was | |
aimed at identifying the changes that occur inS. aureusexposed to | |
the host defense mechanisms during chronic osteomyelitis and | |
evaluating whether these changes affect the virulence of the | |
organism. Genome assessment of twoS. aureusisolates collected 13 | |
months apart (HU-85a and HU-85c) from a host with chronic | |
osteomyelitis was made by whole genome sequencing. Agr | |
functionality was assessed by qRT-PCR. Isolates were tested in a | |
rat model of osteomyelitis and the bacterial load (CFU/tibia) and | |
the morphometric osteomyelitic index (OI) were determined. The | |
ability of the isolates to trigger the release of proinflammatory | |
cytokines was determined on macrophages in culture. Persistence | |
ofS. aureuswithin the host resulted in anagrCframeshift mutation | |
that likely led to the observed phenotype. The capacity to cause | |
bone tissue damage and trigger proinflammatory cytokines by | |
macrophages of theagr-deficient, unencapsulated derivative | |
(HU-85c) was decreased when compared with those of the isogenic | |
CP8-capsulated parental strain (HU-85a). By comparison, no | |
significant differences were found in the bacterial load or the | |
OI from rats challenged with isogenic Reynolds strains [CP5, CP8, | |
and non-typeable (NT)], indicating that lack of CP expression | |
alone was not likely responsible for the reduced capacity to | |
cause tissue damage in HU-85c compared with HU-85a. The | |
production of biofilm was significantly increased in the isogenic | |
derivative HU-85c. Lack ofagr-dependent factors makesS. | |
aureusless virulent during chronic osteomyelitis and alteration | |
of theagrfunctionality seems to permit better adaptation ofS. | |
aureusto the chronically infected host.", | |
journal = "Front. Cell. Infect. Microbiol.", | |
volume = 8, | |
pages = "18", | |
month = feb, | |
year = 2018, | |
keywords = "Staphylococcus aureus; adaptation; agr; biofilm; capsular | |
polysaccharide; chronic; infection; | |
osteomyelitis;Staphylococcus;host-pathogen;within-host-evolution;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Argemi2018-ot, | |
title = "Comparative Genomics and Identification of an | |
{Enterotoxin-Bearing} Pathogenicity Island, {SEPI-1/SECI-1}, in | |
Staphylococcus epidermidis Pathogenic Strains", | |
author = "Argemi, Xavier and Nanoukon, Chim{\`e}ne and Affolabi, Dissou and | |
Keller, Daniel and Hansmann, Yves and Riegel, Philippe and | |
Baba-Moussa, Lamine and Pr{\'e}vost, Gilles", | |
abstract = "Staphylococcus epidermidis is a leading cause of nosocomial | |
infections, majorly resistant to beta-lactam antibiotics, and may | |
transfer several mobile genetic elements among the members of its | |
own species, as well as toStaphylococcus aureus; however, a | |
genetic exchange fromS. aureustoS. epidermidisremains | |
controversial. We recently identified two pathogenic clinical | |
strains ofS. epidermidisthat produce a staphylococcal enterotoxin | |
C3-like (SEC) similar to that byS. aureuspathogenicity islands. | |
This study aimed to determine the genetic environment of the | |
SEC-coding sequence and to identify the mobile genetic elements. | |
Whole-genome sequencing and annotation of theS. | |
epidermidisstrains were performed using Illumina technology and a | |
bioinformatics pipeline for assembly, which provided evidence | |
that the SEC-coding sequences were located in a composite | |
pathogenicity island that was previously described in theS. | |
epidermidisstrain FRI909, called SePI-1/SeCI-1, with 83.8-89.7\% | |
nucleotide similarity. Various other plasmids were identified, | |
particularly p\_3\_95 and p\_4\_95, which carry antibiotic | |
resistance genes (hsrAanddfrG, respectively), and share | |
homologies with SAP085A and pUSA04-2-SUR11, two plasmids | |
described inS. aureus. Eventually, one complete prophage was | |
identified, $\Phi$SE90, sharing 30 out of 52 coding sequences | |
with theAcinetobacterphage vB\_AbaM\_IME200. Thus, the | |
SePI-1/SeCI-1 pathogenicity island was identified in two | |
pathogenic strains ofS. epidermidisthat produced a SEC | |
enterotoxin causing septic shock. These findings suggest the | |
existence of in vivo genetic exchange fromS. aureustoS. | |
epidermidis.", | |
journal = "Toxins", | |
volume = 10, | |
number = 3, | |
month = feb, | |
year = 2018, | |
keywords = "Staphylococcus aureus; Staphylococcus epidermidis; core genome; | |
enterotoxin; pan-genome; pathogenicity | |
island;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Kumburu2018-uw, | |
title = "Hospital Epidemiology of {Methicillin-ResistantStaphylococcus} | |
aureusin a Tertiary Care Hospital in Moshi, Tanzania, as | |
Determined by Whole Genome Sequencing", | |
author = "Kumburu, Happiness H and Sonda, Tolbert and Leekitcharoenphon, | |
Pimlapas and van Zwetselaar, Marco and Lukjancenko, Oksana and | |
Alifrangis, Michael and Lund, Ole and Mmbaga, Blandina T and | |
Kibiki, Gibson and Aarestrup, Frank M", | |
abstract = "Objective: To determine molecular epidemiology of | |
methicillin-resistantS. aureusin Tanzania using whole genome | |
sequencing. Methods: DNA from 33Staphylococcusspecies was | |
recovered from subcultured archivedStaphylococcusisolates. Whole | |
genome sequencing was performed on Illumina Miseq using | |
paired-end 2 $\times$ 250 bp protocol. Raw sequence data were | |
analyzed using online tools. Results: Full susceptibility to | |
vancomycin and chloramphenicol was observed. Thirteen isolates | |
(43.3\%) resisted cefoxitin and other antimicrobials tested. | |
Multilocus sequence typing revealed 13 different sequence types | |
among the 30S. aureusisolates, with ST-8 (n= seven, 23\%) being | |
the most common. Gene detection inS.aureusstains were as | |
follows:mecA, 10 (33.3\%);pvl, 5 (16.7\%);tst, 2 (6.7\%). The SNP | |
difference among the six Tanzanian ST-8 MRSA isolates ranged from | |
24 to 196 SNPs and from 16 to 446 SNPs when using the | |
USA300\_FPR3757 or the USA500\_2395 as a reference, respectively. | |
The mutation rate was 1.38 $\times$ 10-11SNPs/site/year or 1.4 | |
$\times$ 10-6SNPs/site/year as estimated by USA300\_FPR3757 or | |
the USA500\_2395, respectively. Conclusion: S. aureus isolates | |
causing infections in hospitalized patients in Moshi are highly | |
diverse and epidemiologically unrelated. Temporal phylogenetic | |
analysis provided better resolution on transmission and | |
introduction of MRSA and it may be important to include this in | |
future routines.", | |
journal = "Biomed Res. Int.", | |
volume = 2018, | |
pages = "2087693", | |
month = jan, | |
year = 2018, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Bazaid2018-tp, | |
title = "Fatty Acid Supplementation Reverses the Small Colony Variant | |
Phenotype in {Triclosan-Adapted} Staphylococcus aureus: Genetic, | |
Proteomic and Phenotypic Analyses", | |
author = "Bazaid, Abdulrahman S and Forbes, Sarah and Humphreys, Gavin J | |
and Ledder, Ruth G and O'Cualain, Ronan and McBain, Andrew J", | |
abstract = "Staphylococcus aureus can develop a small colony variant (SCV) | |
phenotype in response to sub-lethal exposure to the biocide | |
triclosan. In the current study, whole genome sequencing was | |
performed and changes in virulence were investigated in five | |
Staphylococcus aureus strains following repeated exposure to | |
triclosan. Following exposure, 4/5 formed SCV and exhibited point | |
mutations in the triclosan target gene fabI with 2/4 SCVs showing | |
mutations in both fabI and fabD. The SCV phenotype was in all | |
cases immediately reversed by nutritional supplementation with | |
fatty acids or by repeated growth in the absence of triclosan, | |
although fabI mutations persisted in 3/4 reverted SCVs. | |
Virulence, determined using keratinocyte invasion and Galleria | |
mellonella pathogenicity assays was significantly (p < 0.05) | |
attenuated in 3/4 SCVs and in the non-SCV triclosan-adapted | |
bacterium. Proteomic analysis revealed elevated FabI in 2/3 SCV | |
and down-regulation in a protein associated with virulence in 1/3 | |
SCV. In summary, attenuated keratinocyte invasion and larval | |
virulence in triclosan-induced SCVs was associated with decreases | |
in growth rate and virulence factor expression. Mutation occurred | |
in fabI, which encodes the main triclosan target in all SCVs and | |
the phenotype was reversed by fatty acid supplementation, | |
demonstrating an association between fatty acid metabolism and | |
triclosan-induced SCV.", | |
journal = "Sci. Rep.", | |
volume = 8, | |
number = 1, | |
pages = "3876", | |
month = mar, | |
year = 2018, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{You2018-er, | |
title = "Genomic differences between nasal Staphylococcus aureus from hog | |
slaughterhouse workers and their communities", | |
author = "You, Yaqi and Song, Li and Nonyane, Bareng A S and Price, Lance B | |
and Silbergeld, Ellen K", | |
abstract = "New human pathogens can emerge from the livestock-human interface | |
and spread into human populations through many pathways including | |
livestock products. Occupational contact with livestock is a risk | |
factor for exposure to those pathogens and may cause further | |
spreading of those pathogens in the community. The current study | |
used whole genome sequencing to explore nasal Staphylococcus | |
aureus obtained from hog slaughterhouse workers and their | |
community members, all of whom resided in a livestock-dense | |
region in rural North Carolina. Sequence data were analyzed for | |
lineage distribution, pathogenicity-related genomic features, and | |
mobile genetic elements. We observed evidence of nasal S. aureus | |
differences between hog workers and non-workers. Nasal S. aureus | |
from hog workers showed a greater lineage diversity than nasal S. | |
aureus from community residents. Hog worker isolates were less | |
likely to carry the $\varphi$Sa3 prophage and human-specific | |
immune evasion cluster genes than community resident isolates | |
($\varphi$Sa3 prophage: 54.5\% vs. 91.7\%, Benjamini-Hochberg | |
(BH) corrected p = 0.035; immune evasion cluster genes: 66.7\% | |
vs. 100\%, BH p = 0.021). Hog worker isolates had a lower | |
prevalence and diversity of enterotoxins than community resident | |
isolates, particularly lacking the enterotoxin gene cluster | |
(39.4\% vs. 70.8\%, BH p = 0.125). Moreover, hog worker isolates | |
harbored more diverse antibiotic resistance genes, with a higher | |
prevalence of carriage of multiple resistance genes, than | |
community resident isolates (75.8\% vs. 29.2\%, BH p = 0.021). | |
Phylogenetic analysis of all ST5 isolates, the most abundant | |
lineage in the collection, further supported separation of | |
isolates from hog workers and non-workers. Together, our | |
observations suggest impact of occupational contact with | |
livestock on nasal S. aureus colonization and highlight the need | |
for further research on the complex epidemiology of S. aureus at | |
the livestock-human interface.", | |
journal = "PLoS One", | |
volume = 13, | |
number = 3, | |
pages = "e0193820", | |
month = mar, | |
year = 2018, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Worthing2018-ee, | |
title = "Qac genes and biocide tolerance in clinical veterinary | |
methicillin-resistant and methicillin-susceptible Staphylococcus | |
aureus and Staphylococcus pseudintermedius", | |
author = "Worthing, Kate A and Marcus, Alan and Abraham, Sam and Trott, | |
Darren J and Norris, Jacqueline M", | |
abstract = "Qac genes are associated with increased tolerance to quaternary | |
ammonium compounds and other cationic biocides such as | |
chlorhexidine. This study aimed to determine whether qac genes | |
and increased biocide tolerance were present in 125 clinical | |
methicillin-resistant and susceptible veterinary staphylococci. A | |
total of 125 methicillin-resistant Staphylococcus aureus (MRSA) | |
and methicillin-resistant and -susceptible Staphylococcus | |
pseudintermedius (MRSP and MSSP) from three archived Australian | |
veterinary staphylococci collections underwent whole genome | |
sequencing, multilocus sequence typing and qac gene screening. | |
Two MRSA isolates (12\%) harboured qacA/B genes; both isolates | |
were ST8 from horses. QacJ, qacG and smr genes were identified in | |
28/90 (31\%) MRSP and 1/18 (6\%) MSSP isolates. ST71 MRSP was | |
significantly more likely to harbour qac genes than other MRSP | |
clones (p < 0.05). A random subset of 31 isolates underwent | |
minimum bactericidal concentration (MBC) testing against | |
F10SCTM(benzalkonium chloride and biguanide), and | |
HexaconTM(chlorhexidine gluconate), with and without the addition | |
of bovine serum albumin (BSA) as an in vitro substitute for | |
organic matter contamination. Qac genes were not associated with | |
increased phenotypic biocide tolerance but biocide efficacy was | |
significantly affected by the presence of BSA. In the absence of | |
BSA, all MBC values were well below the recommended usage | |
concentration. When BSA was present, regardless of qac gene | |
presence, 50\% of MRSA and 43\% of MRSP had an F10SCTMMBC above | |
the recommended concentration for general disinfection. Qac genes | |
did not confer increased in vitro biocide tolerance to veterinary | |
staphylococci. Organic matter contamination must be minimized to | |
ensure the efficacy of biocides against MRSA and MRSP.", | |
journal = "Vet. Microbiol.", | |
volume = 216, | |
pages = "153--158", | |
month = mar, | |
year = 2018, | |
keywords = "Biocide tolerance; Methicillin-resistance; Qac genes; | |
Staphylococcus; Veterinary; | |
Zoonosis;Staphylococcus;chlorhexidine;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
% The entry below contains non-ASCII chars that could not be converted | |
% to a LaTeX equivalent. | |
@ARTICLE{Anson2018-he, | |
title = "{DNA} extraction from primary liquid blood cultures for | |
bloodstream infection diagnosis using whole genome sequencing", | |
author = "Anson, Luke W and Chau, Kevin and Sanderson, Nicholas and | |
Hoosdally, Sarah and Bradley, Phelim and Iqbal, Zamin and Phan, | |
Hang and Foster, Dona and Oakley, Sarah and Morgan, Marcus and | |
Peto, Tim E A and {Modernizing Medical Microbiology Informatics | |
Group Mmmig} and Crook, Derrick W and Pankhurst, Louise J", | |
abstract = "PURPOSE: Speed of bloodstream infection diagnosis is vital to | |
reduce morbidity and mortality. Whole genome sequencing (WGS) | |
performed directly from liquid blood culture could provide | |
single-assay species and antibiotic susceptibility prediction; | |
however, high inhibitor and human cell/DNA concentrations limit | |
pathogen recovery. We develop a method for the preparation of | |
bacterial DNA for WGS-based diagnostics direct from liquid blood | |
culture. METHODOLOGY: We evaluate three commercial DNA extraction | |
kits: BiOstic Bacteraemia, Amplex Hyplex and MolYsis Plus. | |
Differential centrifugation, filtration, selective lysis and | |
solid-phase reversible immobilization bead clean-up are tested to | |
improve human cells/DNA and inhibitor removal. Using WGS | |
(Illumina/MinION), we assess human DNA removal, pathogen | |
recovery, and predict species and antibiotic susceptibility | |
inpositive blood cultures of 44 Gram-negative and 54 | |
Staphylococcus species.Results/Key findings. BiOstic kit | |
extractions yield the greatest mean DNA concentration, 94-301 ng | |
µl-1, versus 0-2.5 ng µl-1using Amplex and MolYsis kits. However, | |
we note higher levels of inhibition (260/280 ratio 0.9-2.1) and | |
human DNA (0.0-4.4$\times$106 copies) in BiOstic extracts. | |
Differential centrifugation (2000 g, 1 min) prior to BiOstic | |
extraction reduces human DNA by 63-89 \% with selective lysis | |
minimizing by a further 62 \%. Post-extraction bead clean-up | |
lowers inhibition. Overall, 67 \% of sequenced samples (Illumina | |
MiSeq) contain 93 \% concordance between WGS-based species and | |
susceptibility predictions and clinical diagnosis. If >60 \% of | |
sequencing reads are human (7/98 samples) susceptibility | |
prediction becomes compromised. Novel MinION-based WGS (n=9) | |
currently gives rapid species identification but not | |
susceptibility prediction. CONCLUSION: Our method for DNA | |
preparation allows WGS-based diagnosis direct from blood culture | |
bottles, providing species and antibiotic susceptibility | |
prediction in a single assay.", | |
journal = "J. Med. Microbiol.", | |
volume = 67, | |
number = 3, | |
pages = "347--357", | |
month = mar, | |
year = 2018, | |
keywords = "bacteraemia; bloodstream infection; sepsis; whole genome | |
sequencing;Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} | |
@ARTICLE{Purves2018-im, | |
title = "A horizontally gene transferred copper resistance locus confers | |
hyper-resistance to antibacterial copper toxicity and enables | |
survival of community acquired methicillin resistant | |
Staphylococcus aureus {USA300} in macrophages", | |
author = "Purves, Joanne and Thomas, Jamie and Riboldi, Gustavo Pelicoli | |
and Zapotoczna, Marta and Tarrant, Emma and Andrew, Peter W and | |
Londo{\~n}o, Alejandra and Planet, Paul J and Geoghegan, Joan A | |
and Waldron, Kevin J and Morrissey, Julie A", | |
abstract = "Excess copper is highly toxic and forms part of the host innate | |
immune system's antibacterial arsenal, accumulating at sites of | |
infection and acting within macrophages to kill engulfed | |
pathogens. We show for the first time that a novel, horizontally | |
gene transferred copper resistance locus (copXL), uniquely | |
associated with the SCCmec elements of the highly virulent, | |
epidemic, community acquired methicillin resistant Staphylococcus | |
aureus (CA-MRSA) USA300, confers copper hyper-resistance. These | |
genes are additional to existing core genome copper resistance | |
mechanisms, and are not found in typical S. aureus lineages, but | |
are increasingly identified in emerging pathogenic isolates. Our | |
data show that CopX, a putative P1B-3-ATPase efflux transporter, | |
and CopL, a novel lipoprotein, confer copper hyper-resistance | |
compared to typical S. aureus strains. The copXL genes form an | |
operon that is tightly repressed in low copper environments by | |
the copper regulator CsoR. Significantly, CopX and CopL are | |
important for S. aureus USA300 intracellular survival within | |
macrophages. Therefore the emergence of new S. aureus clones with | |
the copXL locus has significant implications for public health | |
because these genes confer increased resistance to antibacterial | |
copper toxicity, enhancing bacterial fitness by altering S. | |
aureus interaction with innate immunity. This article is | |
protected by copyright. All rights reserved.", | |
journal = "Environ. Microbiol.", | |
month = mar, | |
year = 2018, | |
keywords = "Staphylococcus;Staphylococcus\_aureus\_genome\_papers", | |
language = "en" | |
} |
Sign up for free
to join this conversation on GitHub.
Already have an account?
Sign in to comment