Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 156(1-2): 97-108, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24439371

RESUMEN

Successful infection depends on the ability of the pathogen to gain nutrients from the host. The extracellular pathogenic bacterium group A Streptococcus (GAS) causes a vast array of human diseases. By using the quorum-sensing sil system as a reporter, we found that, during adherence to host cells, GAS delivers streptolysin toxins, creating endoplasmic reticulum stress. This, in turn, increases asparagine (ASN) synthetase expression and the production of ASN. The released ASN is sensed by the bacteria, altering the expression of ∼17% of GAS genes of which about one-third are dependent on the two-component system TrxSR. The expression of the streptolysin toxins is strongly upregulated, whereas genes linked to proliferation are downregulated in ASN absence. Asparaginase, a widely used chemotherapeutic agent, arrests GAS growth in human blood and blocks GAS proliferation in a mouse model of human bacteremia. These results delineate a pathogenic pathway and propose a therapeutic strategy against GAS infections.


Asunto(s)
Percepción de Quorum , Infecciones Estreptocócicas/microbiología , Streptococcus/metabolismo , Animales , Asparagina/metabolismo , Aspartatoamoníaco Ligasa/genética , Aspartatoamoníaco Ligasa/metabolismo , Bacteriemia/microbiología , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Streptococcus/citología , Streptococcus/patogenicidad , Transcripción Genética , Factores de Virulencia/genética
2.
World J Microbiol Biotechnol ; 37(4): 67, 2021 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-33748926

RESUMEN

Infectious diseases are one of the main grounds of death and disabilities in human beings globally. Lack of effective treatment and immunization for many deadly infectious diseases and emerging drug resistance in pathogens underlines the need to either develop new vaccines or sufficiently improve the effectiveness of currently available drugs and vaccines. In this review, we discuss the application of advanced tools like bioinformatics, genomics, proteomics and associated techniques for a rational vaccine design.


Asunto(s)
Vacunas Bacterianas , Desarrollo de Medicamentos , Bacterias , Biología Computacional , Genómica , Humanos , Inmunización , Proteómica
3.
Cell Microbiol ; 21(1): e12956, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30239106

RESUMEN

Group A Streptococcus (GAS) is a human pathogen that causes infections ranging from mild to fulminant and life-threatening. Biofilms have been implicated in acute GAS soft-tissue infections such as necrotising fasciitis (NF). However, most in vitro models used to study GAS biofilms have been designed to mimic chronic infections and insufficiently recapitulate in vivo conditions along with the host-pathogen interactions that might influence biofilm formation. Here, we establish and characterise an in vitro model of GAS biofilm development on mammalian cells that simulates microcolony formation observed in a mouse model of human NF. We show that on mammalian cells, GAS forms dense aggregates that display hallmark biofilm characteristics including a 3D architecture and enhanced tolerance to antibiotics. In contrast to abiotic-grown biofilms, host-associated biofilms require the expression of secreted GAS streptolysins O and S (SLO, SLS) that induce endoplasmic reticulum (ER) stress in the host. In an in vivo mouse model, the streptolysin null mutant is attenuated in both microcolony formation and bacterial spread, but pretreatment of soft-tissue with an ER stressor restores the ability of the mutant to form wild-type-like microcolonies that disseminate throughout the soft tissue. Taken together, we have identified a new role of streptolysin-driven ER stress in GAS biofilm formation and NF disease progression.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Fascitis Necrotizante/microbiología , Streptococcus pyogenes/crecimiento & desarrollo , Streptococcus pyogenes/metabolismo , Estreptolisinas/metabolismo , Animales , Línea Celular , Humanos , Ratones , Modelos Teóricos
4.
Biochem J ; 475(17): 2847-2860, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30049896

RESUMEN

Group A Streptococcus (GAS; Streptococcus pyogenes) causes a wide range of infections, including pharyngitis, impetigo, and necrotizing fasciitis, and results in over half a million deaths annually. GAS ScpC (SpyCEP), a 180-kDa surface-exposed, subtilisin-like serine protease, acts as an essential virulence factor that helps S. pyogenes evade the innate immune response by cleaving and inactivating C-X-C chemokines. ScpC is thus a key candidate for the development of a vaccine against GAS and other pathogenic streptococcal species. Here, we report the crystal structures of full-length ScpC wild-type, the inactive mutant, and the ScpC-AEBSF inhibitor complex. We show ScpC to be a multi-domain, modular protein consisting of nine structural domains, of which the first five constitute the PR + A region required for catalytic activity. The four unique C-terminal domains of this protein are similar to collagen-binding and pilin proteins, suggesting an additional role for ScpC as an adhesin that might mediate the attachment of S. pyogenes to various host tissues. The Cat domain of ScpC is similar to subtilisin-like proteases with significant difference to dictate its specificity toward C-X-C chemokines. We further show that ScpC does not undergo structural rearrangement upon maturation. In the ScpC-inhibitor complex, the bound inhibitor breaks the hydrogen bond between active-site residues, which is essential for catalysis. Guided by our structure, we designed various epitopes and raised antibodies capable of neutralizing ScpC activity. Collectively, our results demonstrate the structure, maturation process, inhibition, and substrate recognition of GAS ScpC, and reveal the presence of functional domains at the C-terminal region.


Asunto(s)
Proteínas Bacterianas/química , Serina Endopeptidasas/química , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/patogenicidad , Factores de Virulencia/química , Proteínas Bacterianas/genética , Cristalografía por Rayos X , Dominios Proteicos , Serina Endopeptidasas/genética , Streptococcus pyogenes/genética , Factores de Virulencia/genética
5.
J Clin Microbiol ; 52(6): 2003-10, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24671796

RESUMEN

Streptococcus pyogenes (group A Streptococcus [GAS]) causes a wide variety of diseases, ranging from mild noninvasive to severe invasive infections. Mutations in regulatory components have been implicated in the switch from colonization to invasive phenotypes. The inactivation of the sil locus, composed of six genes encoding a quorum-sensing complex, gives rise to a highly invasive strain. However, studies conducted on limited collections of GAS strains suggested that sil prevalence is around 15%; furthermore, whereas a correlation between the presence of sil and the genetic background was suggested, no link between the presence of a functional sil locus and the invasive status was assessed. We established a collection of 637 nonredundant strains covering all emm genotypes present in France and of known clinical history; 68%, 22%, and 10% were from invasive infections, noninvasive infections, and asymptomatic carriage, respectively. Among the 637 strains, 206 were sil positive. The prevalence of the sil locus varied according to the emm genotype, being present in >85% of the emm4, emm18, emm32, emm60, emm87, and emm90 strains and absent from all emm1, emm28, and emm89 strains. A random selection based on 2009 French epidemiological data indicated that 16% of GAS strains are sil positive. Moreover, due to mutations leading to truncated proteins, only 9% of GAS strains harbor a predicted functional sil system. No correlation was observed between the presence or absence of a functional sil locus and the strain invasiveness status.


Asunto(s)
Técnicas de Tipificación Bacteriana , Sitios Genéticos , Técnicas de Genotipaje , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , ADN Bacteriano/genética , Femenino , Francia/epidemiología , Humanos , Lactante , Masculino , Persona de Mediana Edad , Epidemiología Molecular , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/patogenicidad , Virulencia , Adulto Joven
6.
J Immunol ; 187(11): 6022-31, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22025550

RESUMEN

Group A Streptococcus (GAS) causes diverse infections in humans, ranging from mild to life-threatening invasive diseases, such as necrotizing fasciitis (NF), a rapidly progressing deep tissue infection. Despite prompt treatments, NF remains a significant cause of morbidity and mortality, even in previously healthy individuals. The early recruitment of leukocytes is crucial to the outcome of NF; however, although the role of polymorphonuclear neutrophils (PMNs) in host defense against NF is well established, the role of recruited macrophages remains poorly defined. Using a cutaneous murine model mimicking human NF, we found that mice deficient in TNF-α were highly susceptible to s.c. infections with GAS, and a paucity of macrophages, but not PMNs, was demonstrated. To test whether the effects of TNF-α on the outcome of infection are mediated by macrophages/monocytes, we systemically depleted C57BL/6 mice of monocytes by pharmacological and genetic approaches. Systemic monocyte depletion substantially increased bacterial dissemination from soft tissues without affecting the number of recruited PMNs or altering the bacterial loads in soft tissues. Enhanced GAS dissemination could be reverted by either i.v. injection of monocytes or s.c. administration of peritoneal macrophages. These experiments demonstrated that recruited macrophages play a key role in defense against the extracellular pathogen GAS by limiting its spread from soft tissues.


Asunto(s)
Fascitis Necrotizante/inmunología , Macrófagos/inmunología , Infecciones Estreptocócicas/inmunología , Animales , Separación Celular , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Streptococcus pyogenes/inmunología
7.
Front Cell Infect Microbiol ; 13: 1168530, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37545859

RESUMEN

Introduction: GBS may cause a devastating disease in newborns. In early onset disease of the newborn the bacteria are acquired from the colonized mother during delivery. We characterized type VII secretion system (T7SS), exporting small proteins of the WXG100 superfamily, in group B Streptococci (GBS) isolates from pregnant colonized women and newborns with early onset disease (EOD) to better understand T7SS contribution to virulence in these different clinical scenarios. Methods: GBS genomes [N=33, 17 EOD isolates (serotype III/ST17) and 16 colonizing isolates (12 serotype VI/ST1, one serotype VI/ST19, one serotype VI/ST6, and two serotype 3/ST19)] were analyzed for presence of T7SS genes and genes encoding WXG100 proteins. We also perform bioinformatic analysis. Galleria mellonella larvae were used to compare virulence between colonizing, EOD, and mutant EOD isolates. The EOD isolate number 118659 (III/ST17) was used for knocking out the essC gene encoding a membrane-bound ATPase, considered the driver of T7SS. Results: Most GBS T7SS loci encoded core component genes: essC, membrane-embedded proteins (essA; essB), modulators of T7SS activity (esaA; esaB; esaC) and effectors: [esxA (SAG1039); esxB (SAG1030)].Bioinformatic analysis indicated that based on sequence type (ST) the clinicalGBS isolates encode at least three distinct subtypes of T7SS machinery. In all ST1isolates we identified two copies of esxA gene (encoding putative WXG100proteins), when only 23.5% of the ST17 isolates harbored the esxA gene. Five ST17isolates encoded two copies of the essC gene. Orphaned WXG100 molecule(SAG0230), distinct from T7SS locus, were found in all tested strains, except inST17 strains where the locus was found in only 23.5% of the isolates. In ST6 andST19 isolates most of the structure T7SS genes were missing. EOD isolates demonstrated enhanced virulence in G. mellonella modelcompared to colonizing isolates. The 118659DessC strain was attenuated in itskilling ability, and the larvae were more effective in eradicating 118659DessC. Conclusions: We demonstrated that T7SS plays a role during infection. Knocking out the essC gene, considered the driver of T7SS, decreased the virulence of ST17 responsible for EOD, causing them to be less virulent comparable to the virulence observed in colonizing isolates.


Asunto(s)
Infecciones Estreptocócicas , Sistemas de Secreción Tipo VII , Humanos , Recién Nacido , Femenino , Embarazo , Mujeres Embarazadas , Sistemas de Secreción Tipo VII/genética , Sistemas de Secreción Tipo VII/metabolismo , Virulencia/genética , Streptococcus agalactiae/genética , Serogrupo , Proteínas de la Membrana/genética , Infecciones Estreptocócicas/microbiología
8.
Bio Protoc ; 12(24)2022 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-36618091

RESUMEN

Group A streptococcus (GAS) is a Gram-positive human pathogen that causes invasive infections with mild to life-threatening severity, like toxic shock syndrome, rheumatic heart disease, and necrotizing fasciitis (NF). NF is characterized by a clinical presentation of widespread tissue destruction due to the rapid spread of GAS infection into fascial planes. Despite quick medical interventions, mortality from NF is high. The early onset of the disease is difficult to diagnose because of non-specific clinical symptoms. Moreover, the unavailability of an effective vaccine against GAS warrants a genuine need for alternative treatments against GAS NF. One endoplasmic reticulum stress signaling pathway (PERK pathway) gets triggered in the host upon GAS infection. Bacteria utilize asparagine release as an output of this pathway for its pathogenesis. We reported that the combination of sub-cutaneous (SC) and intraperitoneal (IP) administration of PERK pathway inhibitors (GSK2656157 and ISRIB) cures local as well as systemic GAS infection in a NF murine model, by reducing asparagine release at the infection site. This protocol's methodology is detailed below. This protocol was validated in: Sci Transl Med (2021), DOI: 10.1126/scitranslmed.abd7465.

9.
Methods Mol Biol ; 2427: 185-200, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35619035

RESUMEN

Group A streptococcus (GAS) necrotizing fasciitis (NF) causes high morbidity and mortality despite prompt intravenous administration of antibiotics, surgical soft-tissue debridement, and supportive treatment in the intensive care unit. Since there is no effective vaccine against GAS infections, a comprehensive understanding of NF pathogenesis is required to design more efficient treatments. To increase our understanding of NF pathogenesis, we need a reliable animal model that mirrors, at least in part, the infectious process in humans. This chapter describes a reliable murine model of human NF that mimics the histopathology observed in humans, namely the destruction of soft tissue, a paucity of infiltrating neutrophils, and the presence of many gram-positive cocci at the center of the infection.


Asunto(s)
Fascitis Necrotizante , Infecciones de los Tejidos Blandos , Infecciones Estreptocócicas , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Fascitis Necrotizante/tratamiento farmacológico , Fascitis Necrotizante/patología , Ratones , Infecciones de los Tejidos Blandos/tratamiento farmacológico , Streptococcus pyogenes
10.
J Biol Chem ; 285(36): 27798-805, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20562101

RESUMEN

Streptococcus pyogenes expresses the LPXTG motif-containing cell envelope serine protease SpyCep (also called ScpC, PrtS) that degrades and inactivates the major chemoattractant interleukin 8 (IL-8), thereby impairing host neutrophil recruitment. In this study, we identified a novel function of SpyCep: the ability to mediate uptake into primary human endothelial cells. SpyCep triggered its uptake into endothelial cells but not into human epithelial cells originating from pharynx or lung, indicating an endothelial cell-specific uptake mechanism. SpyCep mediated cellular invasion by an endosomal/lysosomal pathway distinct from the caveolae-mediated invasion pathway of S. pyogenes. Recombinant expression and purification of proteolytically active SpyCep and a series of subfragments allowed functional dissection of the domains responsible for endothelial cell invasion and IL-8 degradation. The N-terminal PR domain was sufficient to mediate endothelial cell invasion, whereas for IL-8-degrading activity, the protease domain and the flanking A domain were required. A polyclonal rabbit serum raised against the recombinant protease efficiently blocked the invasion-mediating activity of SpyCep but not its proteolytic function, further indicating that SpyCep-mediated internalization is independent from its enzymatic activity. SpyCep may thus specifically mediate its own uptake as secreted protein into human endothelial cells.


Asunto(s)
Células Endoteliales/metabolismo , Interleucina-8/metabolismo , Péptido Hidrolasas/metabolismo , Streptococcus pyogenes/enzimología , Animales , Anticuerpos/inmunología , Línea Celular , Clonación Molecular , Endocitosis , Endosomas/metabolismo , Células Endoteliales/citología , Humanos , Lisosomas/metabolismo , Péptido Hidrolasas/química , Péptido Hidrolasas/genética , Péptido Hidrolasas/inmunología , Estructura Terciaria de Proteína , Transporte de Proteínas , Streptococcus pyogenes/genética
11.
PLoS Pathog ; 5(11): e1000651, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19893632

RESUMEN

Group A streptococcus (GAS) causes a wide variety of human diseases, and at the same time, GAS can also circulate without producing symptoms, similar to its close commensal relative, group G streptococcus (GGS). We previously identified, by transposon-tagged mutagenesis, the streptococcal invasion locus (sil). sil is a quorum-sensing regulated locus which is activated by the autoinducer peptide SilCR through the two-component system SilA-SilB. Here we characterize the DNA promoter region necessary for SilA-mediated activation. This site is composed of two direct repeats of 10 bp, separated by a spacer of 11 bp. Fusion of this site to gfp allowed us to systematically introduce single-base substitutions in the repeats region and to assess the relative contribution of various positions to promoter strength. We then developed an algorithm giving different weights to these positions, and performed a chromosome-wide bioinformatics search which was validated by transcriptome analysis. We identified 13 genes, mostly bacteriocin related, that are directly under the control of SilA. Having developed the ability to quantify SilCR signaling via GFP accumulation prompted us to search for GAS and GGS strains that sense and produce SilCR. While the majority of GAS strains lost sil, all GGS strains examined still possess the locus and approximately 63% are able to respond to exogenously added SilCR. By triggering the autoinduction circle using a minute concentration of synthetic SilCR, we identified GAS and GGS strains that are capable of sensing and naturally producing SilCR, and showed that SilCR can be sensed across these streptococci species. These findings suggest that sil may be involved in colonization and establishment of commensal host-bacterial relationships.


Asunto(s)
Sitios Genéticos/genética , Regiones Promotoras Genéticas/genética , Percepción de Quorum/genética , Streptococcus pyogenes/genética , Secuencia de Bases , Biología Computacional/métodos , Perfilación de la Expresión Génica , Sitios Genéticos/fisiología , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Streptococcus pyogenes/fisiología
12.
Sci Transl Med ; 13(605)2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-34349034

RESUMEN

Group A streptococcus (GAS) is among the top 10 causes of mortality from an infectious disease, producing mild to invasive life-threatening manifestations. Necrotizing fasciitis (NF) is characterized by a rapid GAS spread into fascial planes followed by extensive tissue destruction. Despite prompt treatments of antibiotic administration and tissue debridement, mortality from NF is still high. Moreover, there is no effective vaccine against GAS, and early diagnosis of NF is problematic because its clinical presentations are not specific. Thus, there is a genuine need for effective treatments against GAS NF. Previously, we reported that GAS induces endoplasmic reticulum (ER) stress to gain asparagine from the host. Here, we demonstrate that GAS-mediated asparagine induction and release occur through the PERK-eIF2α-ATF4 branch of the unfolded protein response. Inhibitors of PERK or integrated stress response (ISR) blocked the formation and release of asparagine by infected mammalian cells, and exogenously added asparagine overcame this inhibition. Moreover, in a murine model of NF, we show that the inhibitors minimized mortality when mice were challenged with a lethal dose of GAS and reduced bacterial counts and lesion size when mice were challenged with a sublethal dose. Immunohistopathology studies demonstrated that PERK/ISR inhibitors protected mice by enabling neutrophil infiltration into GAS-infected fascia and reducing the pro-inflammatory response that causes tissue damage. Inhibitor treatment was also effective in mice when started at 12 hours after infection. We conclude that host metabolic alteration induced by PERK or ISR inhibitors is a promising therapeutic strategy to treat highly invasive GAS infections.


Asunto(s)
Fascitis Necrotizante , Infecciones Estreptocócicas , Animales , Asparagina , Fascitis Necrotizante/tratamiento farmacológico , Ratones , Infecciones Estreptocócicas/tratamiento farmacológico , Streptococcus pyogenes , Respuesta de Proteína Desplegada
13.
Cell Rep ; 34(9): 108766, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33657368

RESUMEN

Group A Streptococcus (GAS) causes diverse human diseases, including life-threatening soft-tissue infections. It is accepted that the human antimicrobial peptide LL-37 protects the host by killing GAS. Here, we show that GAS extracellular protease ScpC N-terminally cleaves LL-37 into two fragments of 8 and 29 amino acids, preserving its bactericidal activity. At sub-bactericidal concentrations, the cleavage inhibits LL-37-mediated neutrophil chemotaxis, shortens neutrophil lifespan, and eliminates P2X7 and EGF receptors' activation. Mutations at the LL-37 cleavage site protect the peptide from ScpC-mediated splitting, maintaining all its functions. The mouse LL-37 ortholog CRAMP is neither cleaved by ScpC nor does it activate P2X7 or EGF receptors. Treating wild-type or CRAMP-null mice with sub-bactericidal concentrations of the non-cleavable LL-37 analogs promotes GAS clearance that is abolished by the administration of either P2X7 or EGF receptor antagonists. We demonstrate that LL-37-mediated activation of host receptors is critical for defense against GAS soft-tissue infections.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Receptores ErbB/metabolismo , Neutrófilos/microbiología , Receptores Purinérgicos P2X7/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/patogenicidad , Animales , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas Bacterianas/metabolismo , Catelicidinas/genética , Catelicidinas/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Serina Endopeptidasas/metabolismo , Transducción de Señal , Infecciones Estreptocócicas/tratamiento farmacológico , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/metabolismo , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/genética , Especificidad por Sustrato
14.
Nat Commun ; 11(1): 3545, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32669564

RESUMEN

Group A Streptococcus (GAS) infection causes a range of diseases, but vaccine development is hampered by the high number of serotypes. Here, using reverse vaccinology the authors identify SPy_2191 as a cross-protective vaccine candidate. From 18 initially identified surface proteins, only SPy_2191 is conserved, surface-exposed and inhibits both GAS adhesion and invasion. SPy_2191 immunization in mice generates bactericidal antibodies resulting in opsonophagocytic killing of prevalent and invasive GAS serotypes of different geographical regions, including M1 and M49 (India), M3.1 (Israel), M1 (UK) and M1 (USA). Resident splenocytes show higher interferon-γ and tumor necrosis factor-α secretion upon antigen re-stimulation, suggesting activation of cell-mediated immunity. SPy_2191 immunization significantly reduces streptococcal load in the organs and confers ~76-92% protection upon challenge with invasive GAS serotypes. Further, it significantly suppresses GAS pharyngeal colonization in mice mucosal infection model. Our findings suggest that SPy_2191 can act as a universal vaccine candidate against GAS infections.


Asunto(s)
Proteínas Bacterianas/inmunología , Protección Cruzada/inmunología , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Adhesión Bacteriana/inmunología , Línea Celular , Clonación Molecular , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunogenicidad Vacunal , Ratones , Pruebas de Neutralización , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Serogrupo , Infecciones Estreptocócicas/microbiología , Vacunas Estreptocócicas/administración & dosificación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
15.
Cell Host Microbe ; 23(3): 312-323.e6, 2018 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-29544095

RESUMEN

Bacteria use quorum sensing (QS) to regulate gene expression. We identified a group A Streptococcus (GAS) strain possessing the QS system sil, which produces functional bacteriocins, through a sequential signaling pathway integrating host and bacterial signals. Host cells infected by GAS release asparagine (ASN), which is sensed by the bacteria to alter its gene expression and rate of proliferation. We show that upon ASN sensing, GAS upregulates expression of the QS autoinducer peptide SilCR. Initial SilCR expression activates the autoinduction cycle for further SilCR production. The autoinduction process propagates throughout the GAS population, resulting in bacteriocin production. Subcutaneous co-injection of mice with a bacteriocin-producing strain and the globally disseminated M1T1 GAS clone results in M1T1 killing within soft tissue. Thus, by sensing host signals, a fraction of a bacterial population can trigger an autoinduction mechanism mediated by QS, which acts on the entire bacterial community to outcompete other bacteria within the infection.


Asunto(s)
Bacteriocinas/metabolismo , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus/metabolismo , Streptococcus/patogenicidad , Animales , Asparagina/metabolismo , Proteínas Bacterianas , Bacteriocinas/genética , Línea Celular , ADN Bacteriano/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos/genética , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Percepción de Quorum , Transducción de Señal , Streptococcus/genética , Streptococcus/aislamiento & purificación , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
16.
Genome Announc ; 5(11)2017 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-28302774

RESUMEN

Here, we report the complete genome sequence of the Streptococcus pyogenes emm14 strain JS95, isolated from a patient with necrotizing fasciitis. The streptococcal invasion locus (sil), the first quorum-sensing system characterized in S. pyogenes, was identified in this strain.

17.
Lancet ; 363(9410): 696-703, 2004 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-15001327

RESUMEN

BACKGROUND: Necrotising soft-tissue infections due to group A streptococcus (GAS) are rare (about 0.2 cases per 100000 people). The disease progresses rapidly, causing severe necrosis and hydrolysis of soft tissues. Histopathological analysis of necrotic tissue debrided from two patients (one with necrotising fasciitis and one with myonecrosis) showed large quantities of bacteria but no infiltrating neutrophils. We aimed to investigate whether the poor neutrophil chemotaxis was linked with the ability of group A streptococcus (GAS) to degrade host chemokines. METHODS: We did RT-PCR, ELISA, and dot-blot assays to establish whether GAS induces synthesis of interleukin 8 mRNA, but subsequently degrades the released chemokine protein. Class-specific protease inhibitors were used to characterise the protease that degraded the chemokine. We used a mouse model of human soft-tissue infections to investigate the pathogenic relevance of GAS chemokine degradation, and to test the therapeutic effect of a GAS pheromone peptide (SilCR) that downregulates activity of chemokine protease. FINDINGS: The only isolates from the necrotic tissue were two beta-haemolytic GAS strains of an M14 serotype. A trypsin-like protease released by these strains degraded human interleukin 8 and its mouse homologue MIP2. When innoculated subcutaneously in mice, these strains produced a fatal necrotic soft-tissue infection that had reduced neutrophil recruitment to the site of injection. The M14 GAS strains have a missense mutation in the start codon of silCR, which encodes a predicted 17 aminoacid pheromone peptide, SilCR. Growth of the M14 strain in the presence of SilCR abrogated chemokine proteolysis. When SilCR was injected together with the bacteria, abundant neutrophils were recruited to the site of infection, bacteria were cleared without systemic spread, and the mice survived. The therapeutic effect of SilCR was also obtained in mice challenged with M1 and M3 GAS strains, a leading cause of invasive infections. INTERPRETATION: The unusual reduction in neutrophils in necrotic tissue of people with GAS soft-tissue infections is partly caused by a GAS protease that degrades interleukin 8. In mice, degradation can be controlled by administration of SilCR, which downregulates GAS chemokine protease activity. This downregulation increases neutrophil migration to the site of infection, preventing bacterial spread and development of a fulminant lethal systemic infection.


Asunto(s)
Quimiocinas/inmunología , Fascitis Necrotizante/microbiología , Feromonas/fisiología , Infecciones de los Tejidos Blandos/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/enzimología , Adulto , Anciano , Animales , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Endopeptidasas/genética , Endopeptidasas/inmunología , Fascitis Necrotizante/inmunología , Fascitis Necrotizante/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Infiltración Neutrófila/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones de los Tejidos Blandos/inmunología , Infecciones de los Tejidos Blandos/metabolismo , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/metabolismo , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/aislamiento & purificación
18.
Artículo en Inglés | MEDLINE | ID: mdl-25136516

RESUMEN

The connection between bacterial pathogens and unfolded protein response (UPR) is poorly explored. In this review we highlight the evidence showing that group A streptococcus (GAS) induces endoplasmic reticulum (ER) stress and UPR through which it captures the amino acid asparagine (ASN) from the host. GAS acts extracellularly and during adherence to host cells it delivers the hemolysin toxins; streptolysin O (SLO) and streptolysin S (SLS). By poorly understood pathways, these toxins trigger UPR leading to the induction of the transcriptional regulator ATF4 and consequently to the upregulation of asparagine synthetase (ASNS) transcription leading to production and release of ASN. GAS senses ASN and alters gene expression profile accordingly, and increases the rate of multiplication. We suggest that induction of UPR by GAS and by other bacterial pathogens represent means through which bacterial pathogens gain nutrients from the host, obviating the need to become internalized or inflict irreversible cell damage.


Asunto(s)
Estrés del Retículo Endoplásmico , Infecciones Estreptocócicas/metabolismo , Streptococcus pyogenes/fisiología , Respuesta de Proteína Desplegada , Animales , Asparagina/metabolismo , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/microbiología , Estreptolisinas/metabolismo
20.
Cell Host Microbe ; 4(2): 170-8, 2008 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-18692776

RESUMEN

Interleukin-8 (IL-8) promotes neutrophil-mediated host defense through its chemoattractant and immunostimulatory activities. The Group A Streptococcus (GAS) protease SpyCEP (also called ScpC) cleaves IL-8, and SpyCEP expression is strongly upregulated in vivo in the M1T1 GAS strains associated with life-threatening systemic disease including necrotizing fasciitis. Coupling allelic replacement with heterologous gene expression, we show that SpyCEP is necessary and sufficient for IL-8 degradation. SpyCEP decreased IL-8-dependent neutrophil endothelial transmigration and bacterial killing, the latter by reducing neutrophil extracellular trap formation. The knockout mutant lacking SpyCEP was attenuated for virulence in murine infection models, and SpyCEP expression conferred protection to coinfecting bacteria. We also show that the zoonotic pathogen Streptococcus iniae possesses a functional homolog of SpyCEP (CepI) that cleaves IL-8, promotes neutrophil resistance, and contributes to virulence. By inactivating the multifunctional host defense peptide IL-8, the SpyCEP protease impairs neutrophil clearance mechanisms, contributing to the pathogenesis of invasive streptococcal infection.


Asunto(s)
Interacciones Huésped-Patógeno , Interleucina-8/metabolismo , Neutrófilos/inmunología , Péptido Hidrolasas/metabolismo , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/enzimología , Animales , Células Cultivadas , Células Endoteliales/microbiología , Humanos , Ratones , Ratones Endogámicos C57BL , Neutrófilos/microbiología , Péptido Hidrolasas/genética , Piel/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus/enzimología , Streptococcus/genética , Streptococcus/fisiología , Streptococcus pyogenes/genética , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA