Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Am J Pathol ; 190(4): 862-873, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32200972

RESUMO

Group A streptococcus (GAS) is a major pathogen that impacts health and economic affairs worldwide. Although the oropharynx is the primary site of infection, GAS can colonize the female genital tract and cause severe diseases, such as puerperal sepsis, neonatal infections, and necrotizing myometritis. Our understanding of how GAS genes contribute to interaction with the primate female genital tract is limited by the lack of relevant animal models. Using two genome-wide transposon mutagenesis screens, we identified 69 GAS genes required for colonization of the primate vaginal mucosa in vivo and 96 genes required for infection of the uterine wall ex vivo. We discovered a common set of 39 genes important for GAS fitness in both environments. They include genes encoding transporters, surface proteins, transcriptional regulators, and metabolic pathways. Notably, the genes that encode the surface-exclusion protein (SpyAD) and the immunogenic secreted protein 2 (Isp2) were found to be crucial for GAS fitness in the female primate genital tract. Targeted gene deletion confirmed that isogenic mutant strains ΔspyAD and Δisp2 are significantly impaired in ability to colonize the primate genital tract and cause uterine wall pathologic findings. Our studies identified novel GAS genes that contribute to female reproductive tract interaction that warrant translational research investigation.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Membrana/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidade , Doenças Vaginais/microbiologia , Animais , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Feminino , Regulação Bacteriana da Expressão Gênica , Macaca fascicularis , Proteínas de Membrana/genética , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes/metabolismo , Doenças Vaginais/patologia , Virulência
2.
Am J Pathol ; 190(8): 1680-1690, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32473109

RESUMO

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2, has spread globally, and no proven treatments are available. Convalescent plasma therapy has been used with varying degrees of success to treat severe microbial infections for >100 years. Patients (n = 25) with severe and/or life-threatening COVID-19 disease were enrolled at the Houston Methodist hospitals from March 28, 2020, to April 14, 2020. Patients were transfused with convalescent plasma, obtained from donors with confirmed severe acute respiratory syndrome coronavirus 2 infection who had recovered. The primary study outcome was safety, and the secondary outcome was clinical status at day 14 after transfusion. Clinical improvement was assessed on the basis of a modified World Health Organization six-point ordinal scale and laboratory parameters. Viral genome sequencing was performed on donor and recipient strains. At day 7 after transfusion with convalescent plasma, nine patients had at least a one-point improvement in clinical scale, and seven of those were discharged. By day 14 after transfusion, 19 (76%) patients had at least a one-point improvement in clinical status, and 11 were discharged. No adverse events as a result of plasma transfusion were observed. Whole genome sequencing data did not identify a strain genotype-disease severity correlation. The data indicate that administration of convalescent plasma is a safe treatment option for those with severe COVID-19 disease.


Assuntos
Infecções por Coronavirus/terapia , Pneumonia Viral/terapia , Adulto , Idoso , Betacoronavirus/genética , COVID-19 , Feminino , Humanos , Imunização Passiva , Aplicação de Novas Drogas em Teste , Masculino , Pessoa de Meia-Idade , Pandemias , SARS-CoV-2 , Texas , Sequenciamento Completo do Genoma , Adulto Jovem , Soroterapia para COVID-19
3.
J Bacteriol ; 202(23)2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-32958630

RESUMO

Streptococcus agalactiae (group B streptococcus [GBS]) is a major cause of infections in newborns, pregnant women, and immunocompromised patients. GBS strain CNCTC10/84 is a clinical isolate that has high virulence in animal models of infection and has been used extensively to study GBS pathogenesis. Two unusual features of this strain are hyperhemolytic activity and hypo-CAMP factor activity. These two phenotypes are typical of GBS strains that are functionally deficient in the CovR-CovS two-component regulatory system. A previous whole-genome sequencing study found that strain CNCTC10/84 has intact covR and covS regulatory genes. We investigated CovR-CovS regulation in CNCTC10/84 and discovered that a single-nucleotide insertion in a homopolymeric tract in the covR promoter region underlies the strong hemolytic activity and weak CAMP activity of this strain. Using isogenic mutant strains, we demonstrate that this single-nucleotide insertion confers significantly decreased expression of covR and covS and altered expression of CovR-CovS-regulated genes, including that of genes encoding ß-hemolysin and CAMP factor. This single-nucleotide insertion also confers significantly increased GBS survival in human whole blood ex vivoIMPORTANCE Group B streptococcus (GBS) is the leading cause of neonatal sepsis, pneumonia, and meningitis. GBS strain CNCTC10/84 is a highly virulent blood isolate that has been used extensively to study GBS pathogenesis for over 20 years. Strain CNCTC10/84 has an unusually strong hemolytic activity, but the genetic basis is unknown. In this study, we discovered that a single-nucleotide insertion in an intergenic homopolymeric tract is responsible for the elevated hemolytic activity of CNCTC10/84.


Assuntos
Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Regulação Bacteriana da Expressão Gênica , Hemólise , Histidina Quinase/genética , Histidina Quinase/metabolismo , Humanos , Fenótipo , Mutação Puntual , Regiões Promotoras Genéticas , Infecções Estreptocócicas/sangue , Streptococcus agalactiae/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
4.
Infect Immun ; 88(10)2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32747604

RESUMO

Streptococcus agalactiae (group B streptococcus, or GBS) is a common cause of bacteremia and sepsis in newborns, pregnant women, and immunocompromised patients. The molecular mechanisms used by GBS to survive and proliferate in blood are not well understood. Here, using a highly virulent GBS strain and transposon-directed insertion site sequencing (TraDIS), we performed genome-wide screens to discover novel GBS genes required for bacterial survival in human whole blood and plasma. The screen identified 85 and 41 genes that are required for GBS growth in whole blood and plasma, respectively. A common set of 29 genes was required in both whole blood and plasma. Targeted gene deletion confirmed that (i) genes encoding methionine transporter (metP) and manganese transporter (mtsA) are crucial for GBS survival in whole blood and plasma, (ii) gene W903_1820, encoding a small multidrug export family protein, contributes significantly to GBS survival in whole blood, (iii) the shikimate pathway gene aroA is essential for GBS growth in whole blood and plasma, and (iv) deletion of srr1, encoding a fibrinogen-binding adhesin, increases GBS survival in whole blood. Our findings provide new insight into the GBS-host interactions in human blood.


Assuntos
Bacteriemia/microbiologia , Genes Bacterianos , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/genética , Proteínas de Bactérias/genética , Aptidão Genética , Genoma Bacteriano/genética , Humanos , Viabilidade Microbiana/genética , Mutagênese Insercional , Mutação , Streptococcus agalactiae/crescimento & desenvolvimento , Streptococcus agalactiae/patogenicidade , Virulência/genética
5.
Am J Pathol ; 189(10): 2002-2018, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31369755

RESUMO

Two-component systems (TCSs) are signal transduction proteins that enable bacteria to respond to external stimuli by altering the global transcriptome. Accessory proteins interact with TCSs to fine-tune their activity. In group A Streptococcus (GAS), regulator of Cov (RocA) is an accessory protein that functions with the control of virulence regulator/sensor TCS, which regulates approximately 15% of the GAS transcriptome. Whole-genome sequencing analysis of serotype M28 GAS strains collected from invasive infections in humans identified a higher number of missense (amino acid-altering) and nonsense (protein-truncating) polymorphisms in rocA than expected. We hypothesized that polymorphisms in RocA alter the global transcriptome and virulence of serotype M28 GAS. We used naturally occurring clinical isolates with rocA polymorphisms (n = 48), an isogenic rocA deletion mutant strain, and five isogenic rocA polymorphism mutant strains to perform genome-wide transcript analysis (RNA sequencing), in vitro virulence factor assays, and mouse and nonhuman primate pathogenesis studies to test this hypothesis. Results demonstrated that polymorphisms in rocA result in either a subtle transcriptome change, causing a wild-type-like virulence phenotype, or a substantial transcriptome change, leading to a significantly increased virulence phenotype. Each polymorphism had a unique effect on the global GAS transcriptome. Taken together, our data show that naturally occurring polymorphisms in one gene encoding an accessory protein can significantly alter the global transcriptome and virulence phenotype of GAS, an important human pathogen.


Assuntos
Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Miosite/patologia , Polimorfismo de Nucleotídeo Único , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/patogenicidade , Transativadores/genética , Animais , Proteínas de Bactérias/metabolismo , Camundongos , Miosite/epidemiologia , Miosite/microbiologia , Infecções Estreptocócicas/complicações , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Transcriptoma , Virulência , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
6.
Infect Immun ; 85(5)2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28264907

RESUMO

Studies of the human pathogen group A Streptococcus (GAS) define the carrier phenotype to be an increased ability to adhere to and persist on epithelial surfaces and a decreased ability to cause disease. We tested the hypothesis that a single amino acid change (Arg135Gly) in a highly conserved sensor kinase (LiaS) of a poorly defined GAS regulatory system contributes to a carrier phenotype through increased pilus production. When introduced into an emm serotype-matched invasive strain, the carrier allele (the gene encoding the LiaS protein with an arginine-to-glycine change at position 135 [liaSR135G]) recapitulated a carrier phenotype defined by an increased ability to adhere to mucosal surfaces and a decreased ability to cause disease. Gene transcript analyses revealed that the liaS mutation significantly altered transcription of the genes encoding pilus in the presence of bacitracin. Elimination of pilus production in the isogenic carrier mutant decreased its ability to colonize the mouse nasopharynx and to adhere to and be internalized by cultured human epithelial cells and restored the virulence phenotype in a mouse model of necrotizing fasciitis. We also observed significantly reduced survival of the isogenic carrier mutant compared to that of the parental invasive strain after exposure to human neutrophils. Elimination of pilus in the isogenic carrier mutant increased the level of survival after exposure to human neutrophils to that for the parental invasive strain. Together, our data demonstrate that the carrier mutation (liaSR135G) affects pilus expression. Our data suggest new mechanisms of pilus gene regulation in GAS and that the invasiveness associated with pilus gene regulation in GAS differs from the enhanced invasiveness associated with increased pilus production in other bacterial pathogens.


Assuntos
Portador Sadio/microbiologia , Fímbrias Bacterianas/genética , Histidina Quinase/genética , Mutação de Sentido Incorreto , Biogênese de Organelas , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/patogenicidade , Animais , Aderência Bacteriana , Células Cultivadas , Células Epiteliais/microbiologia , Feminino , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Viabilidade Microbiana , Nasofaringe/microbiologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Streptococcus pyogenes/fisiologia , Transcrição Gênica
7.
Surg Endosc ; 31(2): 852-860, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27387174

RESUMO

BACKGROUND: Surgical energy has been widely implemented because of ease of use, effective hemostasis, and surgical dissection. Studies demonstrate its use to be an independent risk factor for postoperative wound infection. Methicillin-resistant Staphylococcus aureus (MRSA) is the most common bacteria found in postoperative mesh infection. No reports are available on the sequelae of surgical energy use for open ventral hernia repair (oVHR) with mesh. We hypothesized that increasing amounts of surgical energy will result in higher infectious burden after oVHR with composite multifilament polyester mesh (Parietex™ PCO). METHODS: New Zealand rabbits underwent bridging oVHR with Parietex™ PCO and were divided into three surgical treatment groups: (1) scalpel alone, (2) 120 J of energy, and (3) 600 J of energy. The bioprosthesis was then inoculated with 105 colony-forming units of MRSA. Rabbits were survived for 7 days with daily physical examination. Complete blood count, basci metabolic panel, and blood cultures were performed on postoperative days one, four, and seven. Surviving rabbits were killed, and meshes explanted for MRSA colony counts. RESULTS: Rabbits receiving the most surgical energy developed signs and symptoms of severe sepsis and wound necrosis within 24 h. In comparison, rabbits receiving no surgical energy had significantly less MRSA recovered from explanted mesh, significantly less bacteremia, and fewer adhesions. CONCLUSIONS: Increased use of surgical energy promoted greater colonization, exaggerated septic response to bacterial contamination, and more severe adhesions. In the absence of devitalized tissue, rabbits can effectively limit bacterial contamination. These findings support the surgical principles of proper tissue handling and highlight the detrimental effects of indiscriminant surgical energy usage, thus emphasizing the importance of programs such as Fundamental Use of Surgical Energy.


Assuntos
Colágeno , Eletrocoagulação/métodos , Hérnia Ventral/cirurgia , Herniorrafia/métodos , Staphylococcus aureus Resistente à Meticilina , Poliésteres , Infecções Relacionadas à Prótese/epidemiologia , Sepse/epidemiologia , Infecções Estafilocócicas/epidemiologia , Infecção da Ferida Cirúrgica/epidemiologia , Animais , Feminino , Infecções Relacionadas à Prótese/microbiologia , Coelhos , Fatores de Risco , Sepse/microbiologia , Infecções Estafilocócicas/microbiologia , Telas Cirúrgicas , Infecção da Ferida Cirúrgica/microbiologia
8.
Am J Pathol ; 185(2): 462-71, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25476528

RESUMO

Single-nucleotide polymorphisms (SNPs) are the most common source of genetic variation within a species; however, few investigations demonstrate how naturally occurring SNPs may increase strain virulence. We recently used group A Streptococcus as a model pathogen to study bacteria strain genotype-patient disease phenotype relationships. Whole-genome sequencing of approximately 800 serotype M59 group A Streptococcus strains, recovered during an outbreak of severe invasive infections across North America, identified a disproportionate number of SNPs in the gene encoding multiple gene regulator of group A Streptococcus (mga). Herein, we report results of studies designed to test the hypothesis that the most commonly occurring SNP, encoding a replacement of arginine for histidine at codon 201 of Mga (H201R), significantly increases virulence. Whole transcriptome analysis revealed that the H201R replacement significantly increased expression of mga and 54 other genes, including many proven virulence factors. Compared to the wild-type strain, a H201R isogenic mutant strain caused significantly larger skin lesions in mice. Serial quantitative bacterial culture and noninvasive magnetic resonance imaging also demonstrated that the isogenic H201R strain was significantly more virulent in a nonhuman primate model of joint infection. These findings show that the H201R replacement in Mga increases the virulence of M59 group A Streptococcus and provide new insight to how a naturally occurring SNP in bacteria contributes to human disease phenotypes.


Assuntos
Proteínas de Bactérias , Artropatias , Mutação de Sentido Incorreto , Polimorfismo de Nucleotídeo Único , Infecções Estreptocócicas , Streptococcus pyogenes , Substituição de Aminoácidos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Feminino , Genoma Bacteriano , Humanos , Artropatias/genética , Artropatias/metabolismo , Artropatias/microbiologia , Artropatias/patologia , Camundongos , Camundongos Pelados , Infecções Estreptocócicas/genética , Infecções Estreptocócicas/metabolismo , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/patogenicidade
9.
Infect Immun ; 83(6): 2382-95, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25824840

RESUMO

Whole-genome sequencing analysis of ∼800 strains of group A Streptococcus (GAS) found that the gene encoding the multiple virulence gene regulator of GAS (mga) is highly polymorphic in serotype M59 strains but not in strains of other serotypes. To help understand the molecular mechanism of gene regulation by Mga and its contribution to GAS pathogenesis in serotype M59 GAS, we constructed an isogenic mga mutant strain. Transcriptome studies indicated a significant regulatory influence of Mga and altered metabolic capabilities conferred by Mga-regulated genes. We assessed the phosphorylation status of Mga in GAS cell lysates with Phos-tag gels. The results revealed that Mga is phosphorylated at histidines in vivo. Using phosphomimetic and nonphosphomimetic substitutions at conserved phosphoenolpyruvate:carbohydrate phosphotransferase regulation domain (PRD) histidines of Mga, we demonstrated that phosphorylation-mimicking aspartate replacements at H207 and H273 of PRD-1 and at H327 of PRD-2 are inhibitory to Mga-dependent gene expression. Conversely, non-phosphorylation-mimicking alanine substitutions at H273 and H327 relieved inhibition, and the mutant strains exhibited a wild-type phenotype. The opposing regulatory profiles observed for phosphorylation- and non-phosphorylation-mimicking substitutions at H273 extended to global gene regulation by Mga. Consistent with these observations, the H273D mutant strain attenuated GAS virulence, whereas the H273A strain exhibited a wild-type virulence phenotype in a mouse model of necrotizing fasciitis. Together, our results demonstrate phosphoregulation of Mga and its direct link to virulence in M59 GAS strains. These data also lay a foundation toward understanding how naturally occurring gain-of-function variations in mga, such as H201R, may confer an advantage to the pathogen and contribute to M59 GAS pathogenesis.


Assuntos
Fasciite Necrosante/microbiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Streptococcus pyogenes/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Camundongos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Fosforilação , Conformação Proteica , Streptococcus pyogenes/genética , Virulência
10.
Infect Immun ; 83(12): 4750-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26416912

RESUMO

Group A streptococcus (GAS), the causative agent of pharyngitis and necrotizing fasciitis, secretes the potent cysteine protease SpeB. Several lines of evidence suggest that SpeB is an important virulence factor. SpeB is expressed in human infections, protects mice from lethal challenge when used as a vaccine, and contributes significantly to tissue destruction and dissemination in animal models. However, recent descriptions of mutations in genes implicated in SpeB production have led to the idea that GAS may be under selective pressure to decrease secreted SpeB protease activity during infection. Thus, two divergent hypotheses have been proposed. One postulates that SpeB is a key contributor to pathogenesis; the other, that GAS is under selection to decrease SpeB during infection. In order to distinguish between these alternative hypotheses, we performed casein hydrolysis assays to measure the SpeB protease activity secreted by 6,775 GAS strains recovered from infected humans. The results demonstrated that 84.3% of the strains have a wild-type SpeB protease phenotype. The availability of whole-genome sequence data allowed us to determine the relative frequencies of mutations in genes implicated in SpeB production. The most abundantly mutated genes were direct transcription regulators. We also sequenced the genomes of 2,954 GAS isolates recovered from nonhuman primates with experimental necrotizing fasciitis. No mutations that would result in a SpeB-deficient phenotype were identified. Taken together, these data unambiguously demonstrate that the great majority of GAS strains recovered from infected humans secrete wild-type levels of SpeB protease activity. Our data confirm the important role of SpeB in GAS pathogenesis and help end a long-standing controversy.


Assuntos
Proteínas de Bactérias/genética , Exotoxinas/genética , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Streptococcus pyogenes/enzimologia , Streptococcus pyogenes/genética , Animais , Proteínas de Bactérias/metabolismo , Caseínas/química , Monitoramento Epidemiológico , Europa (Continente)/epidemiologia , Exotoxinas/metabolismo , Fasciite Necrosante/epidemiologia , Fasciite Necrosante/microbiologia , Fasciite Necrosante/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mutação , Faringite/epidemiologia , Faringite/microbiologia , Faringite/patologia , Primatas , Proteólise , Sorotipagem , Infecções Estreptocócicas/epidemiologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/classificação , Streptococcus pyogenes/patogenicidade , Transcrição Gênica , Estados Unidos/epidemiologia , Virulência
11.
J Clin Microbiol ; 52(12): 4210-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25253790

RESUMO

Large hospital-based clinical laboratories must be prepared to rapidly investigate potential infectious disease outbreaks. To challenge the ability of our molecular diagnostics laboratory to use whole-genome sequencing in a potential outbreak scenario and identify impediments to these efforts, we studied 84 invasive serotype emm59 group A streptococcus (GAS) strains collected in the United States. We performed a rapid-response exercise to the mock outbreak scenario using whole-genome sequencing, genome-wide transcript analysis, and mouse virulence studies. The protocol changes installed in response to the lessons learned were tested in a second iteration. The initial investigation was completed in 9 days. Whole-genome sequencing showed that the invasive infections were caused by multiple subclones of epidemic emm59 GAS strains likely spread to the United States from Canada. The phylogenetic tree showed a strong temporal-spatial structure with diversity in mobile genetic element content, features that are useful for identifying closely related strains and possible transmission events. The genome data informed the epidemiology, identifying multiple patients who likely acquired the organisms through direct person-to-person transmission. Transcriptome analysis unexpectedly revealed significantly altered expression of genes encoding a two-component regulator and the hyaluronic acid capsule virulence factor. Mouse infection studies confirmed a high-virulence capacity of these emm59 organisms. Whole-genome sequencing, coupled with transcriptome analysis and animal virulence studies, can be rapidly performed in a clinical environment to effectively contribute to patient care decisions and public health maneuvers.


Assuntos
Defesa Civil , Surtos de Doenças , Ensaio de Proficiência Laboratorial , Infecções Estreptocócicas/diagnóstico , Infecções Estreptocócicas/epidemiologia , Streptococcus pyogenes/isolamento & purificação , Animais , Canadá , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Genoma Bacteriano , Pesquisa sobre Serviços de Saúde , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Camundongos , Técnicas de Diagnóstico Molecular/métodos , Streptococcus pyogenes/classificação , Streptococcus pyogenes/genética , Estados Unidos
12.
J Clin Microbiol ; 51(4): 1272-7, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23345298

RESUMO

Next-generation sequencing technology is available to many clinical laboratories; however, it is not yet widely used in routine microbiology practice. To demonstrate the feasibility of using whole-genome sequencing in a routine clinical microbiology workflow, we sequenced the genome of every organism isolated in our laboratory for 1 day.


Assuntos
Bactérias/classificação , Bactérias/genética , Infecções Bacterianas/diagnóstico , Técnicas Bacteriológicas/métodos , Genoma Bacteriano , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Técnicas de Diagnóstico Molecular/métodos , Bactérias/isolamento & purificação , Infecções Bacterianas/microbiologia , Humanos
13.
Am J Pathol ; 180(4): 1522-34, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22330677

RESUMO

Group A Streptococcus (GAS) causes an exceptionally broad range of infections in humans, from relatively mild pharyngitis and skin infections to life-threatening necrotizing fasciitis and toxic shock syndrome. An epidemic of severe invasive human infections caused by type emm59 GAS, heretofore an exceedingly rare cause of disease, spread west to east across Canada over a 3-year period (2006 to 2008). By sequencing the genomes of 601 epidemic, historic, and other emm59 organisms, we discovered that a recently emerged, genetically distinct emm59 clone is responsible for the Canadian epidemic. Using near-real-time genome sequencing, we were able to show spread of the Canadian epidemic clone into the United States. The extensive genome data permitted us to identify patterns of geographic dissemination as well as links between emm59 subclonal lineages that cause infections. Mouse and nonhuman primate models of infection demonstrated that the emerged clone is unusually virulent. Transmission of epidemic emm59 strains may have occurred primarily by skin contact, as suggested by an experimental model of skin transmission. In addition, the emm59 strains had a significantly impaired ability to persist in human saliva and to colonize the oropharynx of mice, and seldom caused human pharyngitis. Our study contributes new information to the rapidly emerging field of molecular pathogenomics of bacterial epidemics and illustrates how full-genome data can be used to precisely illuminate the landscape of strain dissemination during a bacterial epidemic.


Assuntos
Infecções Estreptocócicas/epidemiologia , Streptococcus pyogenes/genética , Animais , Canadá/epidemiologia , DNA Bacteriano/genética , Modelos Animais de Doenças , Epidemias , Fasciite Necrosante/microbiologia , Fasciite Necrosante/patologia , Feminino , Genoma Bacteriano , Humanos , Sequências Repetidas Invertidas/genética , Macaca fascicularis , Masculino , Camundongos , Camundongos Pelados , Faringite/epidemiologia , Faringite/microbiologia , Filogenia , Saliva/microbiologia , Análise de Sequência de DNA/métodos , Dermatopatias Bacterianas/microbiologia , Dermatopatias Bacterianas/patologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/transmissão , Streptococcus pyogenes/classificação , Streptococcus pyogenes/crescimento & desenvolvimento , Streptococcus pyogenes/patogenicidade , Estados Unidos/epidemiologia , Virulência/genética
14.
Proc Natl Acad Sci U S A ; 107(2): 888-93, 2010 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-20080771

RESUMO

Single-nucleotide changes are the most common cause of natural genetic variation among members of the same species, but there is remarkably little information bearing on how they alter bacterial virulence. We recently discovered a single-nucleotide mutation in the group A Streptococcus genome that is epidemiologically associated with decreased human necrotizing fasciitis ("flesh-eating disease"). Working from this clinical observation, we find that wild-type mtsR function is required for group A Streptococcus to cause necrotizing fasciitis in mice and nonhuman primates. Expression microarray analysis revealed that mtsR inactivation results in overexpression of PrsA, a chaperonin involved in posttranslational maturation of SpeB, an extracellular cysteine protease. Isogenic mutant strains that overexpress prsA or lack speB had decreased secreted protease activity in vivo and recapitulated the necrotizing fasciitis-negative phenotype of the DeltamtsR mutant strain in mice and monkeys. mtsR inactivation results in increased PrsA expression, which in turn causes decreased SpeB secreted protease activity and reduced necrotizing fasciitis capacity. Thus, a naturally occurring single-nucleotide mutation dramatically alters virulence by dysregulating a multiple gene virulence axis. Our discovery has broad implications for the confluence of population genomics and molecular pathogenesis research.


Assuntos
Fasciite Necrosante/genética , Polimorfismo de Nucleotídeo Único , Virulência/genética , Animais , Fasciite Necrosante/imunologia , Fasciite Necrosante/prevenção & controle , Variação Genética , Humanos , Macaca fascicularis/microbiologia , Masculino , Camundongos , Neutrófilos/fisiologia , Sorotipagem , Choque Séptico/microbiologia , Streptococcus pyogenes/genética , Regulação para Cima
15.
J Infect Dis ; 205(11): 1719-29, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22262791

RESUMO

Whole-genome sequencing of serotype M3 group A streptococci (GAS) from oropharyngeal and invasive infections in Ontario recently showed that the gene encoding regulator of protease B (RopB) is highly polymorphic in this population. To test the hypothesis that ropB is under diversifying selective pressure among all serotype M3 GAS strains, we sequenced this gene in 1178 strains collected from different infection types, geographic regions, and time periods. The results confirmed our hypothesis and discovered a significant association between mutant ropB alleles, decreased activity of its major regulatory target SpeB, and pharyngitis. Additionally, isoallelic strains with ropB polymorphisms were significantly less virulent in a mouse model of necrotizing fasciitis. These studies provide a model strategy for applying whole-genome sequencing followed by deep single-gene sequencing to generate new insight to the rapid evolution and virulence regulation of human pathogens.


Assuntos
Proteínas de Bactérias/genética , Faringite/microbiologia , Polimorfismo Genético , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/isolamento & purificação , Fatores de Virulência/genética , Animais , Proteínas de Bactérias/metabolismo , DNA Bacteriano/química , DNA Bacteriano/genética , Modelos Animais de Doenças , Fasciite Necrosante/microbiologia , Fasciite Necrosante/patologia , Humanos , Camundongos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Faringite/patologia , Análise de Sequência de DNA , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/classificação , Virulência , Fatores de Virulência/metabolismo
16.
Surg Infect (Larchmt) ; 22(9): 910-922, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33944615

RESUMO

Background: Infected hernia mesh is a cause of post-operative morbidity. Nitric oxide (NO) plays a key role in the endogenous immune response to infection. We sought to study the efficacy of a NO-releasing mesh against methicillin-resistant Staphylococcus aureus (MRSA). We hypothesized that a NO-releasing polyester mesh would decrease MRSA colonization and proliferation. Materials and Methods: A composite polyester mesh functionalized with N-diazeniumdiolate silica nanoparticles was synthesized and characterized. N-diazeniumdiolate silica parietex composite (NOSi) was inoculated with 104,106, or 108 colony forming units (CFUs) of MRSA and a dose response was quantified in a soy tryptic broth assay. Utilizing a rat model of contaminated hernia repair, implanted mesh was inoculated with MRSA, recovered, and CFUs were quantified. Clinical metrics of erythema, mesh contracture, and adhesion severity were then characterized. Results: Methicillin-resistant Staphylococcus aureus CFUs demonstrated a dose-dependent response to NOSi in vitro. In vivo, quantified CFUs showed a dose-dependent response to NOSi-PCO. Treated rats had fewer severe adhesions, less erythema, and reduced mesh contracture. Conclusions: We demonstrate the efficacy of a NO-releasing mesh to treat MRSA in vitro and in vivo. Creation of a novel class of antimicrobial prosthetics offers new strategies for reconstructing contaminated abdominal wall defects and other procedures that benefit from deploying synthetic prostheses in contaminated environments.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Nanopartículas , Infecções Estafilocócicas , Animais , Óxido Nítrico , Poliésteres , Ratos , Dióxido de Silício , Infecções Estafilocócicas/prevenção & controle , Telas Cirúrgicas
17.
Microb Pathog ; 49(6): 336-47, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20674736

RESUMO

Group A Streptococcus (GAS), a human-specific pathogen, is best known for causing pharyngitis ("strep-throat") and necrotizing fasciitis ("flesh-eating disease"). However, the organism is also an uncommon but important cause of community-acquired bronchopneumonia, an infection with an exceptionally high mortality rate. Inasmuch as little is known about the molecular pathogenesis of GAS lower respiratory tract infection, we sought to develop a relevant human infection model. Nine cynomolgus macaques were infected by intra-bronchial instillation of either sterile saline or GAS (10(5) or 10(7) CFU). Animals were continuously monitored and sacrificed at five days post-inoculation. Serial bronchial alveolar lavage specimens and tissues collected at necropsy were used for histologic and immunohistochemical examination, quantitative microbial culture, lung and blood biomarker analysis, and in vivo GAS gene expression studies. The lower respiratory tract disease observed in cynomolgus macaques mimicked the clinical and pathological features of severe GAS bronchopneumonia in humans. This new monkey model will be useful for testing hypotheses bearing on the molecular pathogenesis of GAS in the lower respiratory tract.


Assuntos
Broncopneumonia/veterinária , Doenças dos Macacos/microbiologia , Doenças dos Macacos/patologia , Infecções Estreptocócicas/veterinária , Streptococcus pyogenes/isolamento & purificação , Animais , Biomarcadores/sangue , Líquido da Lavagem Broncoalveolar/microbiologia , Broncopneumonia/microbiologia , Broncopneumonia/patologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Imuno-Histoquímica , Pulmão/patologia , Macaca fascicularis , Masculino , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/genética
18.
mBio ; 11(1)2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32071274

RESUMO

A fundamental goal of contemporary biomedical research is to understand the molecular basis of disease pathogenesis and exploit this information to develop targeted and more-effective therapies. Necrotizing myositis caused by the bacterial pathogen Streptococcus pyogenes is a devastating human infection with a high mortality rate and few successful therapeutic options. We used dual transcriptome sequencing (RNA-seq) to analyze the transcriptomes of S. pyogenes and host skeletal muscle recovered contemporaneously from infected nonhuman primates. The in vivo bacterial transcriptome was strikingly remodeled compared to organisms grown in vitro, with significant upregulation of genes contributing to virulence and altered regulation of metabolic genes. The transcriptome of muscle tissue from infected nonhuman primates (NHPs) differed significantly from that of mock-infected animals, due in part to substantial changes in genes contributing to inflammation and host defense processes. We discovered significant positive correlations between group A streptococcus (GAS) virulence factor transcripts and genes involved in the host immune response and inflammation. We also discovered significant correlations between the magnitude of bacterial virulence gene expression in vivo and pathogen fitness, as assessed by previously conducted genome-wide transposon-directed insertion site sequencing (TraDIS). By integrating the bacterial RNA-seq data with the fitness data generated by TraDIS, we discovered five new pathogen genes, namely, S. pyogenes 0281 (Spy0281 [dahA]), ihk-irr, slr, isp, and ciaH, that contribute to necrotizing myositis and confirmed these findings using isogenic deletion-mutant strains. Taken together, our study results provide rich new information about the molecular events occurring in severe invasive infection of primate skeletal muscle that has extensive translational research implications.IMPORTANCE Necrotizing myositis caused by Streptococcus pyogenes has high morbidity and mortality rates and relatively few successful therapeutic options. In addition, there is no licensed human S. pyogenes vaccine. To gain enhanced understanding of the molecular basis of this infection, we employed a multidimensional analysis strategy that included dual RNA-seq and other data derived from experimental infection of nonhuman primates. The data were used to target five streptococcal genes for pathogenesis research, resulting in the unambiguous demonstration that these genes contribute to pathogen-host molecular interactions in necrotizing infections. We exploited fitness data derived from a recently conducted genome-wide transposon mutagenesis study to discover significant correlation between the magnitude of bacterial virulence gene expression in vivo and pathogen fitness. Collectively, our findings have significant implications for translational research, potentially including vaccine efforts.


Assuntos
Fasciite Necrosante/microbiologia , Miosite/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Transcriptoma , Fatores de Virulência/genética , Animais , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Músculo Esquelético/microbiologia , Músculo Esquelético/patologia , Miosite/genética , Miosite/metabolismo , Primatas , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , Streptococcus pyogenes/patogenicidade , Virulência/genética , Fatores de Virulência/metabolismo
19.
PLoS One ; 15(3): e0229064, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32214338

RESUMO

Streptococcus pyogenes is a strict human pathogen responsible for more than 700 million infections annually worldwide. Strains of serotype M28 S. pyogenes are typically among the five more abundant types causing invasive infections and pharyngitis in adults and children. Type M28 strains also have an unusual propensity to cause puerperal sepsis and neonatal disease. We recently discovered that a one-nucleotide indel in an intergenic homopolymeric tract located between genes Spy1336/R28 and Spy1337 altered virulence in a mouse model of infection. In the present study, we analyzed size variation in this homopolymeric tract and determined the extent of heterogeneity in the number of tandemly-repeated 79-amino acid domains in the coding region of Spy1336/R28 in large samples of strains recovered from humans with invasive infections. Both repeat sequence elements are highly polymorphic in natural populations of M28 strains. Variation in the homopolymeric tract results in (i) changes in transcript levels of Spy1336/R28 and Spy1337 in vitro, (ii) differences in virulence in a mouse model of necrotizing myositis, and (iii) global transcriptome changes as shown by RNAseq analysis of isogenic mutant strains. Variation in the number of tandem repeats in the coding sequence of Spy1336/R28 is responsible for size variation of R28 protein in natural populations. Isogenic mutant strains in which genes encoding R28 or transcriptional regulator Spy1337 are inactivated are significantly less virulent in a nonhuman primate model of necrotizing myositis. Our findings provide impetus for additional studies addressing the role of R28 and Spy1337 variation in pathogen-host interactions.


Assuntos
Proteínas de Bactérias/genética , Fasciite Necrosante/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/isolamento & purificação , Virulência/genética , Animais , Modelos Animais de Doenças , Fasciite Necrosante/patologia , Regulação Bacteriana da Expressão Gênica , Heterogeneidade Genética , Humanos , Camundongos , Polimorfismo Genético , Infecções Estreptocócicas/patologia , Transcriptoma , Fatores de Virulência/genética
20.
JCI Insight ; 5(11)2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32493846

RESUMO

Streptococcus pyogenes (group A streptococcus; GAS) causes 600 million cases of pharyngitis annually worldwide. There is no licensed human GAS vaccine despite a century of research. Although the human oropharynx is the primary site of GAS infection, the pathogenic genes and molecular processes used to colonize, cause disease, and persist in the upper respiratory tract are poorly understood. Using dense transposon mutant libraries made with serotype M1 and M28 GAS strains and transposon-directed insertion sequencing, we performed genome-wide screens in the nonhuman primate (NHP) oropharynx. We identified many potentially novel GAS fitness genes, including a common set of 115 genes that contribute to fitness in both genetically distinct GAS strains during experimental NHP pharyngitis. Targeted deletion of 4 identified fitness genes/operons confirmed that our newly identified targets are critical for GAS virulence during experimental pharyngitis. Our screens discovered many surface-exposed or secreted proteins - substrates for vaccine research - that potentially contribute to GAS pharyngitis, including lipoprotein HitA. Pooled human immune globulin reacted with purified HitA, suggesting that humans produce antibodies against this lipoprotein. Our findings provide new information about GAS fitness in the upper respiratory tract that may assist in translational research, including developing novel vaccines.


Assuntos
Genes Bacterianos , Faringite , Infecções Estreptocócicas , Streptococcus pyogenes , Fatores de Virulência , Animais , Modelos Animais de Doenças , Estudo de Associação Genômica Ampla , Humanos , Macaca fascicularis , Faringite/genética , Faringite/metabolismo , Faringite/microbiologia , Faringite/patologia , Infecções Estreptocócicas/genética , Infecções Estreptocócicas/metabolismo , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/patogenicidade , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa