Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34074753

RESUMEN

Forcing due to solar and volcanic variability, on the natural side, and greenhouse gas and aerosol emissions, on the anthropogenic side, are the main inputs to climate models. Reliable climate model simulations of past and future climate change depend crucially upon them. Here we analyze large ensembles of simulations using a comprehensive Earth System Model to quantify uncertainties in global climate change attributable to differences in prescribed forcings. The different forcings considered here are those used in the two most recent phases of the Coupled Model Intercomparison Project (CMIP), namely CMIP5 and CMIP6. We show significant differences in simulated global surface air temperature due to volcanic aerosol forcing in the second half of the 19th century and in the early 21st century. The latter arise from small-to-moderate eruptions incorporated in CMIP6 simulations but not in CMIP5 simulations. We also find significant differences in global surface air temperature and Arctic sea ice area due to anthropogenic aerosol forcing in the second half of the 20th century and early 21st century. These differences are as large as those obtained in different versions of an Earth System Model employing identical forcings. In simulations from 2015 to 2100, we find significant differences in the rates of projected global warming arising from CMIP5 and CMIP6 concentration pathways that differ slightly but are equivalent in terms of their nominal radiative forcing levels in 2100. Our results highlight the influence of assumptions about natural and anthropogenic aerosol loadings on carbon budgets, the likelihood of meeting Paris targets, and the equivalence of future forcing scenarios.

2.
Antimicrob Agents Chemother ; 65(11): e0098521, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34370589

RESUMEN

The widespread emergence of antibiotic resistance, including multidrug resistance in Gram-negative (G-) bacterial pathogens, poses a critical challenge to the current antimicrobial armamentarium. Antibody-drug conjugates (ADCs), primarily used in anticancer therapy, offer a promising treatment alternative due to their ability to deliver a therapeutic molecule while simultaneously activating the host immune response. The Cloudbreak platform is being used to develop ADCs to treat infectious diseases, composed of a therapeutic targeting moiety (TM) attached via a noncleavable linker to an effector moiety (EM) to treat infectious diseases. In this proof-of-concept study, 21 novel dimeric peptidic molecules (TMs) were evaluated for activity against a screening panel of G- pathogens. The activities of the TMs were not impacted by existing drug resistance. Potent TMs were conjugated to the Fc fragment of human IgG1 (EM), resulting in 4 novel ADCs. These ADCs were evaluated for immunoprophylactic efficacy in a neutropenic mouse model of deep thigh infection. In colistin-sensitive infections, 3 of the 4 ADCs offered protection similar to that of therapeutically dosed colistin, while CTC-171 offered enhanced protection. The efficacy of these ADCs was unchanged in colistin-resistant infections. Together, these results indicate that the ADCs used here are capable of potent binding to G- pathogens regardless of lipopolysaccharide (LPS) modifications that otherwise lead to antibiotic resistance and support further exploration of ADCs in the treatment of infections caused by drug-resistant G- bacteria.


Asunto(s)
Colistina , Infecciones por Bacterias Gramnegativas , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple , Bacterias Gramnegativas , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Lipopolisacáridos , Ratones
3.
Geophys Res Lett ; 48(20): e2021GL095882, 2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34924638

RESUMEN

Global aviation dropped precipitously during the covid-19 pandemic, providing an unprecedented opportunity to study aviation-induced cirrus (AIC). AIC is believed to be responsible for over half of aviation-related radiative forcing, but until now, its radiative impact has only been estimated from simulations. Here, we show that satellite observations of cirrus cloud do not exhibit a detectable global response to the dramatic aviation reductions of spring 2020. These results indicate that previous model-based estimates may overestimate AIC. In addition, we find no significant response of diurnal surface air temperature range to the 2020 aviation changes, reinforcing the findings of previous studies. Though aviation influences the climate through multiple pathways, our analysis suggests that its warming effect from cirrus changes may be smaller than previously estimated.

4.
Geophys Res Lett ; 48(8): e2020GL091883, 2021 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-34149115

RESUMEN

Many nations responded to the corona virus disease-2019 (COVID-19) pandemic by restricting travel and other activities during 2020, resulting in temporarily reduced emissions of CO2, other greenhouse gases and ozone and aerosol precursors. We present the initial results from a coordinated Intercomparison, CovidMIP, of Earth system model simulations which assess the impact on climate of these emissions reductions. 12 models performed multiple initial-condition ensembles to produce over 300 simulations spanning both initial condition and model structural uncertainty. We find model consensus on reduced aerosol amounts (particularly over southern and eastern Asia) and associated increases in surface shortwave radiation levels. However, any impact on near-surface temperature or rainfall during 2020-2024 is extremely small and is not detectable in this initial analysis. Regional analyses on a finer scale, and closer attention to extremes (especially linked to changes in atmospheric composition and air quality) are required to test the impact of COVID-19-related emission reductions on near-term climate.

5.
Proc Natl Acad Sci U S A ; 113(34): 9515-20, 2016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27512043

RESUMEN

The sequences of M proteins, the major surface-associated virulence factors of the widespread bacterial pathogen group A Streptococcus, are antigenically variable but have in common a strong propensity to form coiled coils. Paradoxically, these sequences are also replete with coiled-coil destabilizing residues. These features are evident in the irregular coiled-coil structure and thermal instability of M proteins. We present an explanation for this paradox through studies of the B repeats of the medically important M1 protein. The B repeats are required for interaction of M1 with fibrinogen (Fg) and consequent proinflammatory activation. The B repeats sample multiple conformations, including intrinsically disordered, dissociated, as well as two alternate coiled-coil conformations: a Fg-nonbinding register 1 and a Fg-binding register 2. Stabilization of M1 in the Fg-nonbinding register 1 resulted in attenuation of Fg binding as expected, but counterintuitively, so did stabilization in the Fg-binding register 2. Strikingly, these register-stabilized M1 proteins gained the ability to bind Fg when they were destabilized by a chaotrope. These results indicate that M1 stability is antithetical to Fg interaction and that M1 conformational dynamics, as specified by destabilizing residues, are essential for interaction. A "capture-and-collapse" model of association accounts for these observations, in which M1 captures Fg through a dynamic conformation and then collapses into a register 2-coiled coil as a result of stabilization provided by binding energy. Our results support the general conclusion that destabilizing residues are evolutionarily conserved in M proteins to enable functional interactions necessary for pathogenesis.


Asunto(s)
Aminoácidos/química , Antígenos Bacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas Portadoras/química , Fibrinógeno/química , Streptococcus pyogenes/química , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Sitios de Unión , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Fibrinógeno/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Termodinámica
6.
Nature ; 472(7341): 64-8, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21475196

RESUMEN

M1 protein, a major virulence factor of the leading invasive strain of group A Streptococcus, is sufficient to induce toxic-shock-like vascular leakage and tissue injury. These events are triggered by the formation of a complex between M1 and fibrinogen that, unlike M1 or fibrinogen alone, leads to neutrophil activation. Here we provide a structural explanation for the pathological properties of the complex formed between streptococcal M1 and human fibrinogen. A conformationally dynamic coiled-coil dimer of M1 was found to organize four fibrinogen molecules into a specific cross-like pattern. This pattern supported the construction of a supramolecular network that was required for neutrophil activation but was distinct from a fibrin clot. Disruption of this network into other supramolecular assemblies was not tolerated. These results have bearing on the pathophysiology of streptococcal toxic shock.


Asunto(s)
Proteínas Bacterianas/metabolismo , Fibrinógeno/química , Streptococcus pyogenes/patogenicidad , Factores de Virulencia/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/ultraestructura , Sitios de Unión , Cristalografía por Rayos X , Fibrinógeno/metabolismo , Fibrinógeno/ultraestructura , Humanos , Modelos Moleculares , Activación Neutrófila , Unión Proteica , Conformación Proteica , Choque Séptico/microbiología , Choque Séptico/fisiopatología , Streptococcus pyogenes/química , Virulencia , Factores de Virulencia/química
7.
Cell Microbiol ; 17(12): 1721-41, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26433203

RESUMEN

Group A Streptococcus (Streptococcus pyogenes), group B Streptococcus (Streptococcus agalactiae) and Streptococcus pneumoniae (pneumococcus) are host-adapted bacterial pathogens among the leading infectious causes of human morbidity and mortality. These microbes and related members of the genus Streptococcus produce an array of toxins that act against human cells or tissues, resulting in impaired immune responses and subversion of host physiological processes to benefit the invading microorganism. This toxin repertoire includes haemolysins, proteases, superantigens and other agents that ultimately enhance colonization and survival within the host and promote dissemination of the pathogen.


Asunto(s)
Toxinas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Infecciones Estreptocócicas/patología , Streptococcus agalactiae/patogenicidad , Streptococcus pneumoniae/patogenicidad , Streptococcus pyogenes/patogenicidad , Factores de Virulencia/metabolismo , Humanos , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/metabolismo , Streptococcus pneumoniae/metabolismo , Streptococcus pyogenes/metabolismo
8.
Clin Microbiol Rev ; 27(2): 264-301, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24696436

RESUMEN

Streptococcus pyogenes, also known as group A Streptococcus (GAS), causes mild human infections such as pharyngitis and impetigo and serious infections such as necrotizing fasciitis and streptococcal toxic shock syndrome. Furthermore, repeated GAS infections may trigger autoimmune diseases, including acute poststreptococcal glomerulonephritis, acute rheumatic fever, and rheumatic heart disease. Combined, these diseases account for over half a million deaths per year globally. Genomic and molecular analyses have now characterized a large number of GAS virulence determinants, many of which exhibit overlap and redundancy in the processes of adhesion and colonization, innate immune resistance, and the capacity to facilitate tissue barrier degradation and spread within the human host. This improved understanding of the contribution of individual virulence determinants to the disease process has led to the formulation of models of GAS disease progression, which may lead to better treatment and intervention strategies. While GAS remains sensitive to all penicillins and cephalosporins, rising resistance to other antibiotics used in disease treatment is an increasing worldwide concern. Several GAS vaccine formulations that elicit protective immunity in animal models have shown promise in nonhuman primate and early-stage human trials. The development of a safe and efficacious commercial human vaccine for the prophylaxis of GAS disease remains a high priority.


Asunto(s)
Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/patogenicidad , Factores de Virulencia/metabolismo , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Interacciones Huésped-Patógeno , Humanos , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/mortalidad , Vacunas Estreptocócicas/administración & dosificación , Vacunas Estreptocócicas/inmunología , Streptococcus pyogenes/genética , Virulencia , Factores de Virulencia/genética
9.
J Biol Chem ; 289(6): 3539-46, 2014 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-24356958

RESUMEN

Group A Streptococcus (GAS) is a leading human pathogen producing a diverse array of infections from simple pharyngitis ("strep throat") to invasive conditions, including necrotizing fasciitis and toxic shock syndrome. The surface-anchored GAS M1 protein is a classical virulence factor that promotes phagocyte resistance and exaggerated inflammation by binding host fibrinogen (Fg) to form supramolecular networks. In this study, we used a virulent WT M1T1 GAS strain and its isogenic M1-deficient mutant to examine the role of M1-Fg binding in a proximal step in GAS infection-interaction with the pharyngeal epithelium. Expression of the M1 protein reduced GAS adherence to human pharyngeal keratinocytes by 2-fold, and this difference was increased to 4-fold in the presence of Fg. In stationary phase, surface M1 protein cleavage by the GAS cysteine protease SpeB eliminated Fg binding and relieved its inhibitory effect on GAS pharyngeal cell adherence. In a mouse model of GAS colonization of nasal-associated lymphoid tissue, M1 protein expression was associated with an average 6-fold decreased GAS recovery in isogenic strain competition assays. Thus, GAS M1 protein-Fg binding reduces GAS pharyngeal cell adherence and colonization in a fashion that is counterbalanced by SpeB. Inactivation of SpeB during the shift to invasive GAS disease allows M1-Fg binding, increasing pathogen phagocyte resistance and proinflammatory activities.


Asunto(s)
Antígenos Bacterianos/metabolismo , Adhesión Bacteriana , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/metabolismo , Queratinocitos/metabolismo , Faringe/metabolismo , Infecciones Estreptocócicas/metabolismo , Streptococcus pyogenes/metabolismo , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Línea Celular , Modelos Animales de Enfermedad , Exotoxinas/genética , Exotoxinas/inmunología , Exotoxinas/metabolismo , Fibrinógeno/genética , Fibrinógeno/inmunología , Fibrinógeno/metabolismo , Humanos , Queratinocitos/microbiología , Queratinocitos/patología , Ratones , Faringe/microbiología , Faringe/patología , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/genética , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/patogenicidad
10.
J Biol Chem ; 289(46): 32303-32315, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25266727

RESUMEN

A recent analysis of group A Streptococcus (GAS) invasive infections in Australia has shown a predominance of M4 GAS, a serotype recently reported to lack the antiphagocytic hyaluronic acid (HA) capsule. Here, we use molecular genetics and bioinformatics techniques to characterize 17 clinical M4 isolates associated with invasive disease in children during this recent epidemiology. All M4 isolates lacked HA capsule, and whole genome sequence analysis of two isolates revealed the complete absence of the hasABC capsule biosynthesis operon. Conversely, M4 isolates possess a functional HA-degrading hyaluronate lyase (HylA) enzyme that is rendered nonfunctional in other GAS through a point mutation. Transformation with a plasmid expressing hasABC restored partial encapsulation in wild-type (WT) M4 GAS, and full encapsulation in an isogenic M4 mutant lacking HylA. However, partial encapsulation reduced binding to human complement regulatory protein C4BP, did not enhance survival in whole human blood, and did not increase virulence of WT M4 GAS in a mouse model of systemic infection. Bioinformatics analysis found no hasABC homologs in closely related species, suggesting that this operon was a recent acquisition. These data showcase a mutually exclusive interaction of HA capsule and active HylA among strains of this leading human pathogen.


Asunto(s)
Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/enzimología , Animales , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Membrana Celular/microbiología , Biología Computacional , Exotoxinas/metabolismo , Femenino , Prueba de Complementación Genética , Histidina Quinasa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Datos de Secuencia Molecular , Neutrófilos/microbiología , Mutación Puntual , Polisacárido Liasas/metabolismo , Polisacáridos/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Represoras/metabolismo , Virulencia
11.
Infect Immun ; 83(9): 3648-56, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26150541

RESUMEN

Recently, we showed that endothelial heparan sulfate facilitates entry of a bacterial pathogen into the central nervous system. Here, we show that normal bactericidal activity of neutrophils is influenced by the sulfation pattern of heparan sulfate. Inactivation of heparan sulfate uronyl 2-O-sulfotransferase (Hs2st) in neutrophils substantially reduced their bactericidal activity, and Hs2st deficiency rendered mice more susceptible to systemic infection with the pathogenic bacterium group B Streptococcus. Specifically, altered sulfation of heparan sulfate in mutant neutrophils affected formation of neutrophil extracellular traps while not influencing phagocytosis, production of reactive oxygen species, or secretion of granular proteases. Heparan sulfate proteoglycan(s) is present in neutrophil extracellular traps, modulates histone affinity, and modulates their microbial activity. Hs2st-deficient brain endothelial cells show enhanced binding to group B Streptococcus and are more susceptible to apoptosis, likely contributing to the observed increase in dissemination of group B Streptococcus into the brain of Hs2st-deficient mice following intravenous challenge. Taken together, our data provide strong evidence that heparan sulfate from both neutrophils and the endothelium plays important roles in modulating innate immunity.


Asunto(s)
Células Endoteliales/inmunología , Proteoglicanos de Heparán Sulfato/inmunología , Inmunidad Innata/inmunología , Neutrófilos/inmunología , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Trampas Extracelulares/inmunología , Proteoglicanos de Heparán Sulfato/metabolismo , Ratones , Microscopía Electrónica de Rastreo , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Sulfotransferasas/metabolismo
12.
Geophys Res Lett ; 42(13): 5485-5492, 2015 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-26937058

RESUMEN

Radiation parameterizations in GCMs are more accurate than their predecessorsErrors in estimates of 4 ×CO2 forcing are large, especially for solar radiationErrors depend on atmospheric state, so global mean error is unknown.

13.
Philos Trans A Math Phys Eng Sci ; 373(2054)2015 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-26438278

RESUMEN

We investigate the sensitivity of cloud feedbacks to the use of convective parametrizations by repeating the CMIP5/CFMIP-2 AMIP/AMIP + 4K uniform sea surface temperature perturbation experiments with 10 climate models which have had their convective parametrizations turned off. Previous studies have suggested that differences between parametrized convection schemes are a leading source of inter-model spread in cloud feedbacks. We find however that 'ConvOff' models with convection switched off have a similar overall range of cloud feedbacks compared with the standard configurations. Furthermore, applying a simple bias correction method to allow for differences in present-day global cloud radiative effects substantially reduces the differences between the cloud feedbacks with and without parametrized convection in the individual models. We conclude that, while parametrized convection influences the strength of the cloud feedbacks substantially in some models, other processes must also contribute substantially to the overall inter-model spread. The positive shortwave cloud feedbacks seen in the models in subtropical regimes associated with shallow clouds are still present in the ConvOff experiments. Inter-model spread in shortwave cloud feedback increases slightly in regimes associated with trade cumulus in the ConvOff experiments but is quite similar in the most stable subtropical regimes associated with stratocumulus clouds. Inter-model spread in longwave cloud feedbacks in strongly precipitating regions of the tropics is substantially reduced in the ConvOff experiments however, indicating a considerable local contribution from differences in the details of convective parametrizations. In both standard and ConvOff experiments, models with less mid-level cloud and less moist static energy near the top of the boundary layer tend to have more positive tropical cloud feedbacks. The role of non-convective processes in contributing to inter-model spread in cloud feedback is discussed.

14.
FASEB J ; 27(7): 2633-43, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23531597

RESUMEN

In Western countries, invasive infections caused by M1T1 serotype group A Streptococcus (GAS) are epidemiologically linked to mutations in the control of virulence regulatory 2-component operon (covRS). In indigenous communities and developing countries, severe GAS disease is associated with genetically diverse non-M1T1 GAS serotypes. Hypervirulent M1T1 covRS mutant strains arise through selection by human polymorphonuclear cells for increased expression of GAS virulence factors such as the DNase Sda1, which promotes neutrophil resistance. The GAS bacteremia isolate NS88.2 (emm 98.1) is a covS mutant that exhibits a hypervirulent phenotype and neutrophil resistance yet lacks the phage-encoded Sda1. Here, we have employed a comprehensive systems biology (genomic, transcriptomic, and proteomic) approach to identify NS88.2 virulence determinants that enhance neutrophil resistance in the non-M1T1 GAS genetic background. Using this approach, we have identified streptococcal collagen-like protein A and general stress protein 24 proteins as NS88.2 determinants that contribute to survival in whole blood and neutrophil resistance in non-M1T1 GAS. This study has revealed new factors that contribute to GAS pathogenicity that may play important roles in resisting innate immune defenses and the development of human invasive infections.


Asunto(s)
Proteínas Bacterianas/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Adhesión Bacteriana/genética , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Electroforesis en Gel Bidimensional , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano/genética , Genómica/métodos , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones , Viabilidad Microbiana/genética , Viabilidad Microbiana/inmunología , Mutación , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteoma/genética , Proteoma/metabolismo , Proteómica/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Virulencia/genética , Virulencia/inmunología
15.
Nat Med ; 13(8): 981-5, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17632528

RESUMEN

Most invasive bacterial infections are caused by species that more commonly colonize the human host with minimal symptoms. Although phenotypic or genetic correlates underlying a bacterium's shift to enhanced virulence have been studied, the in vivo selection pressures governing such shifts are poorly understood. The globally disseminated M1T1 clone of group A Streptococcus (GAS) is linked with the rare but life-threatening syndromes of necrotizing fasciitis and toxic shock syndrome. Mutations in the GAS control of virulence regulatory sensor kinase (covRS) operon are associated with severe invasive disease, abolishing expression of a broad-spectrum cysteine protease (SpeB) and allowing the recruitment and activation of host plasminogen on the bacterial surface. Here we describe how bacteriophage-encoded GAS DNase (Sda1), which facilitates the pathogen's escape from neutrophil extracellular traps, serves as a selective force for covRS mutation. The results provide a paradigm whereby natural selection exerted by the innate immune system generates hypervirulent bacterial variants with increased risk of systemic dissemination.


Asunto(s)
Desoxirribonucleasa I/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/patogenicidad , Animales , Supervivencia Celular , Desoxirribonucleasa I/genética , Humanos , Inmunidad Innata , Ratones , Neutrófilos/citología , Neutrófilos/microbiología , Fenotipo , Selección Genética , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/genética , Virulencia
16.
Vaccines (Basel) ; 12(4)2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38675764

RESUMEN

Vaccine development against group A Streptococcus (GAS) has gained traction in the last decade, fuelled by recognition of the significant worldwide burden of the disease. Several vaccine candidates are currently being evaluated in preclinical and early clinical studies. Here, we investigate two conjugate vaccine candidates that have shown promise in mouse models of infection. Two antigens, the J8 peptide from the conserved C-terminal end of the M protein, and the group A carbohydrate lacking N-acetylglucosamine side chain (ΔGAC) were each conjugated to arginine deiminase (ADI), an anchorless surface protein from GAS. Both conjugate vaccine candidates combined with alum adjuvant were tested in a non-human primate (NHP) model of pharyngeal infection. High antibody titres were detected against J8 and ADI antigens, while high background antibody titres in NHP sera hindered accurate quantification of ΔGAC-specific antibodies. The severity of pharyngitis and tonsillitis signs, as well as the level of GAS colonisation, showed no significant differences in NHPs immunised with either conjugate vaccine candidate compared to NHPs in the negative control group.

18.
FASEB J ; 26(11): 4675-84, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22878963

RESUMEN

The past 50 years has witnessed the emergence of new viral and bacterial pathogens with global effect on human health. The hyperinvasive group A Streptococcus (GAS) M1T1 clone, first detected in the mid-1980s in the United States, has since disseminated worldwide and remains a major cause of severe invasive human infections. Although much is understood regarding the capacity of this pathogen to cause disease, much less is known of the precise evolutionary events selecting for its emergence. We used high-throughput technologies to sequence a World Health Organization strain collection of serotype M1 GAS and reconstructed its phylogeny based on the analysis of core genome single-nucleotide polymorphisms. We demonstrate that acquisition of a 36-kb genome segment from serotype M12 GAS and the bacteriophage-encoded DNase Sda1 led to increased virulence of the M1T1 precursor and occurred relatively early in the molecular evolutionary history of this strain. The more recent acquisition of the phage-encoded superantigen SpeA is likely to have provided selection advantage for the global dissemination of the M1T1 clone. This study provides an exemplar for the evolution and emergence of virulent clones from microbial populations existing commensally or causing only superficial infection.


Asunto(s)
Evolución Biológica , Pandemias , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Exotoxinas/genética , Exotoxinas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Genoma Bacteriano , Salud Global , Interacciones Huésped-Patógeno , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Neutrófilos/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fagocitosis , Filogenia , Streptococcus pyogenes/clasificación , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Transcriptoma , Virulencia
19.
J Bacteriol ; 194(22): 6154-61, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22961854

RESUMEN

Group A Streptococcus (GAS) is a human-specific bacterial pathogen responsible for serious morbidity and mortality worldwide. The hyaluronic acid (HA) capsule of GAS is a major virulence factor, contributing to bloodstream survival through resistance to neutrophil and antimicrobial peptide killing and to in vivo pathogenicity. Capsule biosynthesis has been exclusively attributed to the ubiquitous hasABC hyaluronan synthase operon, which is highly conserved across GAS serotypes. Previous reports indicate that hasA, encoding hyaluronan synthase, and hasB, encoding UDP-glucose 6-dehydrogenase, are essential for capsule production in GAS. Here, we report that precise allelic exchange mutagenesis of hasB in GAS strain 5448, a representative of the globally disseminated M1T1 serotype, did not abolish HA capsule synthesis. In silico whole-genome screening identified a putative HasB paralog, designated HasB2, with 45% amino acid identity to HasB at a distant location in the GAS chromosome. In vitro enzymatic assays demonstrated that recombinant HasB2 is a functional UDP-glucose 6-dehydrogenase enzyme. Mutagenesis of hasB2 alone slightly decreased capsule abundance; however, a ΔhasB ΔhasB2 double mutant became completely acapsular. We conclude that HasB is not essential for M1T1 GAS capsule biogenesis due to the presence of a newly identified HasB paralog, HasB2, which most likely resulted from gene duplication. The identification of redundant UDP-glucose 6-dehydrogenases underscores the importance of HA capsule expression for M1T1 GAS pathogenicity and survival in the human host.


Asunto(s)
Cápsulas Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo , Operón/genética , Streptococcus pyogenes/metabolismo , Uridina Difosfato Glucosa Deshidrogenasa/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Clonación Molecular , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica , Genoma Bacteriano , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Insercional , Filogenia , Conformación Proteica , Streptococcus pyogenes/genética , Uridina Difosfato Glucosa Deshidrogenasa/genética
20.
Cell Microbiol ; 13(8): 1200-11, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21615663

RESUMEN

Invasive serotype M3 Streptococcus pyogenes are among the most frequently isolated organisms from patients suffering from invasive streptococcal disease and have the potential to invade primary human endothelial cells (EC) via a rapid and efficient mechanism. FbaB protein, the fibronectin-binding protein expressed by M3 S. pyogenes, was herein identified as a potent invasin for EC. By combining heterologous gene expression with allelic replacement, we demonstrate that FbaB is essential and sufficient to trigger EC invasion via a Rac1-dependent phagocytosis-like uptake. FbaB-mediated uptake follows the classical endocytic pathway with lysosomal destination. FbaB is demonstrated to be a streptococcal invasin exhibiting EC tropism. FbaB thus initiates a process that may contribute to the deep tissue tropism and spread of invasive S. pyogenes isolates into the vascular EC lining.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Células Endoteliales/microbiología , Interacciones Huésped-Patógeno , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/patogenicidad , Factores de Virulencia/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Adhesinas Bacterianas/genética , Proteínas Portadoras/genética , Células Cultivadas , Endocitosis , Fibronectinas/metabolismo , Humanos , Streptococcus pyogenes/genética , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA