Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.725
Filtrar
1.
Nat Commun ; 15(1): 3916, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38729927

RESUMEN

The UK observed a marked increase in scarlet fever and invasive group A streptococcal infection in 2022 with severe outcomes in children and similar trends worldwide. Here we report lineage M1UK to be the dominant source of invasive infections in this upsurge. Compared with ancestral M1global strains, invasive M1UK strains exhibit reduced genomic diversity and fewer mutations in two-component regulator genes covRS. The emergence of M1UK is dated to 2008. Following a bottleneck coinciding with the COVID-19 pandemic, three emergent M1UK clades underwent rapid nationwide expansion, despite lack of detection in previous years. All M1UK isolates thus-far sequenced globally have a phylogenetic origin in the UK, with dispersal of the new clades in Europe. While waning immunity may promote streptococcal epidemics, the genetic features of M1UK point to a fitness advantage in pathogenicity, and a striking ability to persist through population bottlenecks.


Asunto(s)
COVID-19 , Filogenia , Infecciones Estreptocócicas , Streptococcus pyogenes , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Streptococcus pyogenes/aislamiento & purificación , Reino Unido/epidemiología , Humanos , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/microbiología , COVID-19/epidemiología , Pandemias , Escarlatina/epidemiología , Escarlatina/microbiología , Mutación , Proteínas Represoras/genética , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Genoma Bacteriano , Europa (Continente)/epidemiología , Proteínas Bacterianas
2.
Nature ; 605(7910): 527-531, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35545676

RESUMEN

Gasdermins (GSDMs) are a family of pore-forming effectors that permeabilize the cell membrane during the cell death program pyroptosis1. GSDMs are activated by proteolytic removal of autoinhibitory carboxy-terminal domains, typically by caspase regulators1-9. However, no activator is known for one member of this family, GSDMA. Here we show that the major human pathogen group A Streptococcus (GAS) secretes a protease virulence factor, SpeB, that induces GSDMA-dependent pyroptosis. SpeB cleavage of GSDMA releases an active amino-terminal fragment that can insert into membranes to form lytic pores. GSDMA is primarily expressed in the skin10, and keratinocytes infected with SpeB-expressing GAS die of GSDMA-dependent pyroptosis. Mice have three homologues of human GSDMA, and triple-knockout mice are more susceptible to invasive infection by a pandemic hypervirulent M1T1 clone of GAS. These results indicate that GSDMA is critical in the immune defence against invasive skin infections by GAS. Furthermore, they show that GSDMs can act independently of host regulators as direct sensors of exogenous proteases. As SpeB is essential for tissue invasion and survival within skin cells, these results suggest that GSDMA can act akin to a guard protein that directly detects concerning virulence activities of microorganisms that present a severe infectious threat.


Asunto(s)
Piroptosis , Streptococcus pyogenes , Animales , Caspasas , Queratinocitos , Ratones , Proteínas Citotóxicas Formadoras de Poros , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/patogenicidad , Virulencia , Factores de Virulencia
3.
Sci Rep ; 11(1): 19011, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34561464

RESUMEN

Group A Streptoccocus (GAS) is among the most diverse of all human pathogens, responsible for a range of clinical manifestations, from mild superficial infections such as pharyngitis to serious invasive infections such as necrotising fasciitis and sepsis. The drivers of these different disease phenotypes are not known. The GAS cholesterol-dependent cytolysin, Streptolysin O (SLO), has well established cell and tissue destructive activity. We investigated the role of SLO in determining disease outcome in vivo, by using two different clinical lineages; the recently emerged hypervirulent outbreak emm type 32.2 strains, which result in sepsis, and the emm type 1.0 strains which cause septic arthritis. Using clinically relevant in vivo mouse models of sepsis and a novel septic arthritis model, we found that the amount and activity of SLO was vital in determining the course of infection. The emm type 32.2 strain produced large quantities of highly haemolytic SLO that resulted in rapid development of sepsis. By contrast, the reduced concentration and lower haemolytic activity of emm type 1.0 SLO led to translocation of bacteria from blood to joints. Importantly, sepsis associated strains that were attenuated by deletion or inhibition of SLO, then also translocated to the joint, confirming the key role of SLO in determining infection niche. Our findings demonstrate that SLO is key to in vivo phenotype and disease outcome. Careful consideration should be given to novel therapy or vaccination strategies that target SLO. Whilst neutralising SLO activity may reduce severe invasive disease, it has the potential to promote chronic inflammatory conditions such as septic arthritis.


Asunto(s)
Fenotipo , Infecciones Estreptocócicas/genética , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Estreptolisinas/metabolismo , Animales , Artritis Infecciosa/microbiología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/fisiología , Traslocación Bacteriana , Modelos Animales de Enfermedad , Fascitis Necrotizante/microbiología , Humanos , Ratones , Terapia Molecular Dirigida , Faringitis/microbiología , Pronóstico , Sepsis/microbiología , Infecciones Estreptocócicas/terapia , Estreptolisinas/fisiología
4.
mBio ; 12(4): e0164221, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34253064

RESUMEN

Adaptation of group A Streptococcus (GAS) to its human host is mediated by two-component systems that transduce external stimuli to regulate bacterial physiology. Among such systems, CsrRS (also known as CovRS) is the most extensively characterized for its role in regulating ∼10% of the GAS genome, including several virulence genes. Here, we show that extracellular magnesium and the human antimicrobial peptide LL-37 have opposing effects on the phosphorylation of the response regulator CsrR by the receptor kinase CsrS. Genetic inactivation of CsrS phosphatase or kinase activity, respectively, had similar but more pronounced effects on CsrR phosphorylation compared to growth in magnesium or LL-37. These changes in CsrR phosphorylation were correlated with the repression or activation of CsrR-regulated genes as assessed by NanoString analysis. Chromatin immunoprecipitation and DNA sequencing (ChIP-seq) revealed CsrR occupancy at CsrRS-regulated promoters and lower-affinity associations at many other locations on the GAS chromosome. Because ChIP-seq did not detect CsrR occupancy at promoters associated with some CsrR-regulated genes, we investigated whether these genes might be controlled indirectly by intermediate regulators whose expression is modulated by CsrR. Transcriptional profiling of mutant strains deficient in the expression of either of two previously uncharacterized transcription regulators in the CsrR regulon indicated that one or both proteins participated in the regulation of 22 of the 42 CsrR-regulated promoters for which no CsrR association was detected by ChIP-seq. Taken together, these results illuminate CsrRS-mediated regulation of GAS gene expression through modulation of CsrR phosphorylation, CsrR association with regulated promoters, and the control of intermediate transcription regulators. IMPORTANCE Group A Streptococcus (GAS) is an important public health threat as a cause of sore throat, skin infections, life-threatening invasive infections, and the postinfectious complications of acute rheumatic fever, a leading cause of acquired heart disease. This work characterizes CsrRS, a GAS system for the detection of environmental signals that enables adaptation of the bacteria for survival in the human throat by regulating the production of products that allow the bacteria to resist clearance by the human immune system. CsrRS consists of two proteins: CsrS, which is on the bacterial surface to detect specific stimuli, and CsrR, which receives signals from CsrS and, in response, represses or activates the expression of genes coding for proteins that enhance bacterial survival. Some of the genes regulated by CsrR encode proteins that are themselves regulators of gene expression, thereby creating a regulatory cascade.


Asunto(s)
Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Proteínas Quinasas/genética , Regulón/genética , Streptococcus pyogenes/genética , Adaptación Fisiológica/genética , Péptidos Catiónicos Antimicrobianos/farmacología , Humanos , Magnesio/farmacología , Fosforilación , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/efectos de los fármacos , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/patogenicidad , Catelicidinas
5.
Sci Rep ; 11(1): 12687, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34135390

RESUMEN

Group A Streptococcus (GAS) is a globally significant bacterial pathogen. The GAS genotyping gold standard characterises the nucleotide variation of emm, which encodes a surface-exposed protein that is recombinogenic and under immune-based selection pressure. Within a supervised learning methodology, we tested three random forest (RF) algorithms (Guided, Ordinary, and Regularized) and 53 GAS response regulator (RR) allele types to infer six genomic traits (emm-type, emm-subtype, tissue and country of sample, clinical outcomes, and isolate invasiveness). The Guided, Ordinary, and Regularized RF classifiers inferred the emm-type with accuracies of 96.7%, 95.7%, and 95.2%, using ten, three, and four RR alleles in the feature set, respectively. Notably, we inferred the emm-type with 93.7% accuracy using only mga2 and lrp. We demonstrated a utility for inferring emm-subtype (89.9%), country (88.6%), invasiveness (84.7%), but not clinical (56.9%), or tissue (56.4%), which is consistent with the complexity of GAS pathophysiology. We identified a novel cell wall-spanning domain (SF5), and proposed evolutionary pathways depicting the 'contrariwise' and 'likewise' chimeric deletion-fusion of emm and enn. We identified an intermediate strain, which provides evidence of the time-dependent excision of mga regulon genes. Overall, our workflow advances the understanding of the GAS mga regulon and its plasticity.


Asunto(s)
Algoritmos , Evolución Molecular , Variación Genética , Aprendizaje Automático , Regulón , Streptococcus pyogenes/genética , Alelos , Antígenos Bacterianos/genética , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Genes Bacterianos , Genoma Bacteriano , Humanos , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/clasificación , Streptococcus pyogenes/patogenicidad
6.
FEMS Microbiol Lett ; 368(13)2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34156082

RESUMEN

Streptococcus pyogenes is a Gram-positive human-specific pathogen that asymptomatically colonizes the human respiratory tract. The factors affecting the colonization to the host is not clearly understood. Adherence of the pathogen to host epithelial cell is the initial step for a successful colonization process. In the host, bacteria live in a polymicrobial community; thus, the signaling mediated between the bacteria plays a significant role in the colonization of the pathogen to the host. Thus, the effect of acyl-homoserine lactone, secreted by Gram-negative bacteria on the adhesion properties of S. pyogenes M3 strain was examined. N-(3-Oxododecanoyl)-L-homoserine lactone (Oxo-C12) increased the cell size as well as hydrophobicity of S. pyogenes. qPCR data revealed that the expression of sagA and hasA was negatively affected by Oxo-C12. Moreover, Oxo-C12 leads to changes in the morphological characteristic of S. pyogenes, further promoting adherence to host epithelia and biofilm formation on abiotic surface. The study demonstrates the role of Oxo-C12 as a factor that can promote virulence in S. pyogenes M3.


Asunto(s)
Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/crecimiento & desarrollo , Streptococcus pyogenes/patogenicidad , Acil-Butirolactonas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Transducción de Señal , Especificidad de la Especie , Streptococcus pyogenes/clasificación , Streptococcus pyogenes/metabolismo , Virulencia
8.
Pediatr Infect Dis J ; 40(7): 612-616, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34097654

RESUMEN

BACKGROUND: Group A Streptococcus can cause serious and sometimes life-threatening disease in children. The past few years have witnessed a rise in invasive group A Streptococcus infection (iGASi) for unclear reasons. This study attempted to describe the epidemiology, the clinical and demographic characteristics and the outcomes associated with iGASi in hospitalized children in central Israel. METHODS: We retrospectively analyzed the medical records of children <18 years old discharged with a diagnosis of iGASi between January 2012 and December 2019. Clinical, laboratory and microbiologic data, and immunization status were retrieved. The patients were divided into severe and nonsevere groups based on their clinical presentation. The emm type was determined at the national reference center. RESULTS: A total of 167 patients with 206 positive cultures for group A Streptococcus were identified. Hospitalizations for iGASi increased from 701 to 958 per 100,000 admissions between 2012-2015 and 2016-2019, respectively, representing an increase of 37%. The majority of the isolates were from the otolaryngologic system followed by blood, deep soft tissue and respiratory sites. Uncomplicated mastoiditis was the most common diagnosis, followed by bacteremia. Pneumonia was the main diagnosis in the severe group (39.4%). CONCLUSIONS: The admissions because of iGASi in children <18 years old increased during the last 8 years. Surveillance systems and prospective studies should be conducted to expend our understanding of the epidemiology of iGASi in children, better assess the pathogenesis and specific risk factors and monitor changes in emm-type distribution.


Asunto(s)
Hospitalización/estadística & datos numéricos , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/epidemiología , Adolescente , Distribución por Edad , Antibacterianos/uso terapéutico , Bacteriemia/epidemiología , Niño , Preescolar , Femenino , Humanos , Incidencia , Lactante , Israel/epidemiología , Masculino , Mastoiditis/epidemiología , Mastoiditis/microbiología , Neumonía Bacteriana/epidemiología , Estudios Prospectivos , Estudios Retrospectivos , Factores de Riesgo , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/tratamiento farmacológico , Streptococcus pyogenes/patogenicidad , Centros de Atención Terciaria/estadística & datos numéricos
9.
Infect Immun ; 89(8): e0018521, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-33972369

RESUMEN

M-type 28 (M28) Streptococcus pyogenes (group A Streptococcus [GAS]) strains are highly associated with life-threatening puerperal infections. Genome sequencing has revealed a large mobile genetic element, RD2, present in most M28 GAS isolates but not found widely in other serotypes. Previous studies have linked RD2 to the ability of M28 GAS to colonize the vaginal tract. A new study by Roshika and colleagues (R. Roshika, I. Jain, J. Medicielo, J. Wächter, J. L. Danger, P. Sumby, Infect Immun 89:e00722-20, 2021, https://doi.org/10.1128/IAI.00722-20) used gain-of-function mutants in three different GAS serotypes to help determine why RD2 appears to have a serotype preference and what that could mean for GAS mucosal colonization and pathogenesis.


Asunto(s)
Elementos Transponibles de ADN , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/clasificación , Streptococcus pyogenes/genética , Susceptibilidad a Enfermedades/inmunología , Genoma Bacteriano , Genómica/métodos , Interacciones Huésped-Patógeno/inmunología , Humanos , Serogrupo , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/patogenicidad , Factores de Virulencia/genética
10.
Int J Mol Sci ; 22(9)2021 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-33925779

RESUMEN

Paediatric inflammatory multisystem syndrome temporally associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (PIMS-TS) is a new systemic inflammatory disease that mainly affects children. Its course in many features resembles that of acute rheumatic fever (ARF). Therefore, it is interesting that the experiences with ARF can be used in the management of patients with PIMS-TS. The aim of the article is to analyse the current data on PIMS-TS in relation to ARF. PIMS-TS and ARF are associated with an abnormal immune response to specific pathogens (SARS-CoV-2 and group A streptococcus, respectively). The main symptoms of both diseases are fever and cardiac involvement. Current therapy for PIMS-TS is based on anti-inflammatory treatment: intravenous immunoglobulin (first-line), intravenous glucocorticoids (second-line), or biological therapy (third-line; including interleukin [IL]-1 antagonists, IL-6 receptor blockers, and anti-tumour necrosis factor agents). Vaccination might be good prophylaxis, but the efficacy and safety of the vaccines against SARS-CoV-2 have not yet been established in children. Interesting insights may be gained by considering PIMS-TS in light of what is known of ARF due to their similar courses, but there are still many unanswered questions surrounding this disease and its pathogenesis.


Asunto(s)
COVID-19/patología , Fiebre Reumática/patología , SARS-CoV-2/patogenicidad , Síndrome de Respuesta Inflamatoria Sistémica/patología , COVID-19/complicaciones , COVID-19/etiología , COVID-19/virología , Vacunas contra la COVID-19/inmunología , Citocinas/metabolismo , Glucocorticoides/uso terapéutico , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Fiebre Reumática/microbiología , SARS-CoV-2/aislamiento & purificación , Streptococcus pyogenes/patogenicidad , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Tratamiento Farmacológico de COVID-19
11.
Sci Rep ; 11(1): 8200, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859234

RESUMEN

Group A Streptococcus (GAS) causes 700 million infections and accounts for half a million deaths per year. Biofilm formation has been implicated in both pharyngeal and dermal GAS infections. In vitro, plate-based assays have shown that several GAS M-types form biofilms, and multiple GAS virulence factors have been linked to biofilm formation. Although the contributions of these plate-based studies have been valuable, most have failed to mimic the host environment, with many studies utilising abiotic surfaces. GAS is a human specific pathogen, and colonisation and subsequent biofilm formation is likely facilitated by distinct interactions with host tissue surfaces. As such, a host cell-GAS model has been optimised to support and grow GAS biofilms of a variety of GAS M-types. Improvements and adjustments to the crystal violet biofilm biomass assay have also been tailored to reproducibly detect delicate GAS biofilms. We propose 72 h as an optimal growth period for yielding detectable biofilm biomass. GAS biofilms formed are robust and durable, and can be reproducibly assessed via staining/washing intensive assays such as crystal violet with the aid of methanol fixation prior to staining. Lastly, SEM imaging of GAS biofilms formed by this model revealed GAS cocci chains arranged into three-dimensional aggregated structures with EPS matrix material. Taken together, we outline an efficacious GAS biofilm pharyngeal cell model that can support long-term GAS biofilm formation, with biofilms formed closely resembling those seen in vivo.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Faringe/microbiología , Streptococcus pyogenes/fisiología , Calibración , Técnicas de Cultivo de Célula/normas , Células Cultivadas , Humanos , Técnicas Microbiológicas/normas , Modelos Biológicos , Faringe/citología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/crecimiento & desarrollo , Streptococcus pyogenes/patogenicidad , Factores de Virulencia/metabolismo
12.
PLoS One ; 16(4): e0248201, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33914767

RESUMEN

Streptococcus pyogenes or group A Streptococcus (GAS) is a leading cause of bacterial pharyngitis, skin and soft tissue infections, life-threatening invasive infections, and the post-infectious autoimmune syndromes of acute rheumatic fever and post-streptococcal glomerulonephritis. Genetic manipulation of this important pathogen is complicated by resistance of the organism to genetic transformation. Very low transformation efficiency is attributed to recognition and degradation of introduced foreign DNA by a type I restriction-modification system encoded by the hsdRSM locus. DNA sequence analysis of this locus in ten GAS strains that had been previously transformed with an unrelated plasmid revealed that six of the ten harbored a spontaneous mutation in hsdR, S, or M. The mutations were all different, and at least five of the six were predicted to result in loss of function of the respective hsd gene product. The unexpected occurrence of such mutations in previously transformed isolates suggested that the process of transformation selects for spontaneous inactivating mutations in the Hsd system. We investigated the possibility of exploiting the increased transformability of hsd mutants by constructing a deletion mutation in hsdM in GAS strain 854, a clinical isolate representative of the globally dominant M1T1 clonal group. Mutant strain 854ΔhsdM exhibited a 5-fold increase in electrotransformation efficiency compared to the wild type parent strain and no obvious change in growth or off-target gene expression. We conclude that genetic transformation of GAS selects for spontaneous mutants in the hsdRSM restriction modification system. We propose that use of a defined hsdM mutant as a parent strain for genetic manipulation of GAS will enhance transformation efficiency and reduce the likelihood of selecting spontaneous hsd mutants with uncharacterized genotypes.


Asunto(s)
Proteínas Bacterianas/genética , Enzimas de Restricción-Modificación del ADN/genética , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Transformación Genética , Regulación Bacteriana de la Expresión Génica , Sitios Genéticos , Humanos , Modelos Moleculares , Mutación , Streptococcus pyogenes/patogenicidad , Virulencia
13.
Lancet Child Adolesc Health ; 5(6): 437-446, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33705693

RESUMEN

Indigenous children and young peoples live with an inequitable burden of acute rheumatic fever and rheumatic heart disease. In this Review, we focus on the epidemiological burden and lived experience of these conditions for Indigenous young peoples in Australia, New Zealand, and Canada. We outline the direct and indirect drivers of rheumatic heart disease risk and their mitigation. Specifically, we identify the opportunities and limitations of predominantly biomedical approaches to the primary, secondary, and tertiary prevention of disease among Indigenous peoples. We explain why these biomedical approaches must be coupled with decolonising approaches to address the underlying cause of disease. Initiatives underway to reduce acute rheumatic fever and rheumatic heart disease in Australia, New Zealand, and Canada are reviewed to identify how an Indigenous rights-based approach could contribute to elimination of rheumatic heart disease and global disease control goals.


Asunto(s)
Pueblos Indígenas/estadística & datos numéricos , Fiebre Reumática/epidemiología , Fiebre Reumática/prevención & control , Cardiopatía Reumática/epidemiología , Cardiopatía Reumática/prevención & control , Adolescente , Adulto , Australia/etnología , Investigación Biomédica/métodos , Canadá/etnología , Exposición a Riesgos Ambientales/efectos adversos , Carga Global de Enfermedades/estadística & datos numéricos , Accesibilidad a los Servicios de Salud/normas , Disparidades en Atención de Salud/etnología , Humanos , Incidencia , Nueva Zelanda/etnología , Fiebre Reumática/diagnóstico , Cardiopatía Reumática/diagnóstico , Factores de Riesgo , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/prevención & control , Streptococcus pyogenes/patogenicidad , Adulto Joven
14.
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
15.
Cell Rep ; 34(13): 108924, 2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33789094

RESUMEN

The arginine deiminase (ADI) pathway has been found in many kinds of bacteria and functions to supplement energy production and provide protection against acid stress. The Streptococcus pyogenes ADI pathway is upregulated upon exposure to various environmental stresses, including glucose starvation. However, there are several unclear points about the advantages to the organism for upregulating arginine catabolism. We show that the ADI pathway contributes to bacterial viability and pathogenesis under low-glucose conditions. S. pyogenes changes global gene expression, including upregulation of virulence genes, by catabolizing arginine. In a murine model of epicutaneous infection, S. pyogenes uses the ADI pathway to augment its pathogenicity by increasing the expression of virulence genes, including those encoding the exotoxins. We also find that arginine from stratum-corneum-derived filaggrin is a key substrate for the ADI pathway. In summary, arginine is a nutrient source that promotes the pathogenicity of S. pyogenes on the skin.


Asunto(s)
Arginina/metabolismo , Piel/microbiología , Streptococcus pyogenes/patogenicidad , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Filagrina , Regulación Bacteriana de la Expresión Génica , Células HaCaT , Humanos , Hidrolasas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Viabilidad Microbiana , Fosforilación , Piel/patología , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/genética , Transcriptoma/genética , Regulación hacia Arriba , Virulencia
16.
mBio ; 12(1)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33563838

RESUMEN

Group A Streptococcus (GAS; Streptococcus pyogenes) is a major human pathogen that causes streptococcal pharyngitis, skin and soft tissue infections, and life-threatening conditions such as streptococcal toxic-shock syndrome. During infection, GAS not only invades diverse host cells but also injects effector proteins such as NAD-glycohydrolase (Nga) into the host cells through a streptolysin O (SLO)-dependent mechanism without invading the cells; Nga and SLO are two major virulence factors that are associated with increased bacterial virulence. Here, we have shown that the invading GAS induces fragmentation of the Golgi complex and inhibits anterograde transport in the infected host cells through the secreted toxins SLO and Nga. GAS infection-induced Golgi fragmentation required both bacterial invasion and SLO-mediated Nga translocation into the host cytosol. The cellular Golgi network is critical for the sorting of surface molecules and is thus essential for the integrity of the epithelial barrier and for the immune response of macrophages to pathogens. In epithelial cells, inhibition of anterograde trafficking by invading GAS and Nga resulted in the redistribution of E-cadherin to the cytosol and an increase in bacterial translocation across the epithelial barrier. Moreover, in macrophages, interleukin-8 secretion in response to GAS infection was found to be suppressed by intracellular GAS and Nga. Our findings reveal a previously undescribed bacterial invasion-dependent function of Nga as well as a previously unrecognized GAS-host interaction that is associated with GAS pathogenesis.IMPORTANCE Two prominent virulence factors of group A Streptococcus (GAS), streptolysin O (SLO) and NAD-glycohydrolase (Nga), are linked to enhanced pathogenicity of the prevalent GAS strains. Recent advances show that SLO and Nga are important for intracellular survival of GAS in epithelial cells and macrophages. Here, we found that invading GAS disrupts the Golgi complex in host cells through SLO and Nga. We show that GAS-induced Golgi fragmentation requires bacterial invasion into host cells, SLO pore formation activity, and Nga NADase activity. GAS-induced Golgi fragmentation results in the impairment of the epithelial barrier and chemokine secretion in macrophages. This immune inhibition property of SLO and Nga by intracellular GAS indicates that the invasion of GAS is associated with virulence exerted by SLO and Nga.


Asunto(s)
Células Epiteliales/microbiología , Aparato de Golgi/patología , Interacciones Huésped-Patógeno/genética , NAD+ Nucleosidasa/genética , Streptococcus pyogenes/patogenicidad , Estreptolisinas/genética , Células A549 , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Citoplasma/microbiología , Aparato de Golgi/genética , Aparato de Golgi/microbiología , Células HeLa , Interacciones Huésped-Patógeno/inmunología , Humanos , Interleucina-8/inmunología , NAD+ Nucleosidasa/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/inmunología , Estreptolisinas/metabolismo , Células THP-1 , Factores de Virulencia
17.
Nature ; 590(7844): 29-31, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33469204

Asunto(s)
Autoanticuerpos/efectos adversos , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , COVID-19/complicaciones , COVID-19/fisiopatología , Modelos Inmunológicos , SARS-CoV-2/patogenicidad , Envejecimiento/inmunología , Anexina A2/inmunología , Autoanticuerpos/aislamiento & purificación , Enfermedades Autoinmunes/microbiología , Enfermedades Autoinmunes/virología , Linfocitos B/inmunología , Linfocitos B/patología , Coagulación Sanguínea/inmunología , Vasos Sanguíneos/inmunología , Vasos Sanguíneos/patología , Encéfalo/inmunología , Encéfalo/patología , COVID-19/etiología , COVID-19/inmunología , COVID-19/patología , COVID-19/virología , Enfermedad Crítica , Síndrome de Liberación de Citoquinas/complicaciones , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/patología , Síndrome de Liberación de Citoquinas/fisiopatología , Femenino , Cadenas HLA-DRB1/inmunología , Helicobacter pylori/inmunología , Helicobacter pylori/patogenicidad , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 4/patogenicidad , Humanos , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/inmunología , Masculino , Miocardio/inmunología , Miocardio/patología , Fosfolípidos/inmunología , Grupos Raciales , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación , Caracteres Sexuales , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/patogenicidad , Factores de Tiempo , Síndrome Post Agudo de COVID-19
18.
Int J Mol Sci ; 22(2)2021 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-33477598

RESUMEN

Acute glomerulonephritis (AGN) triggered by infection is still one of the major causes of acute kidney injury. During the previous two decades, there has been a major paradigm shift in the epidemiology of AGN. The incidence of poststreptococcal acute glomerulonephritis (PSAGN), which develops after the cure of group A Streptococcus infection in children has decreased, whereas adult AGN cases have been increasing, and those associated with nonstreptococcal infections, particularly infections by Staphylococcus, are now as common as PSAGN. In adult AGN patients, particularly older patients with comorbidities, infections are usually ongoing at the time when glomerulonephritis is diagnosed; thus, the term "infection-related glomerulonephritis (IRGN)" has recently been popularly used instead of "post-infectious AGN". The prognosis of children with PSAGN is generally considered excellent compared with that of adult IRGN cases. However, long-term epidemiological analysis demonstrated that an episode of PSAGN in childhood is a strong risk factor for chronic kidney disease (CKD), even after the complete remission of PSAGN. Although the precise mechanism of the transition from IRGN to CKD remains unknown, its clarification is important as it will lead to the prevention of CKD. In this review, we therefore focus on the possible factors that may contribute to the progression of IRGN into CKD. Four factors, namely, persistent infection, genetic background of the host's complement system, tubulointerstitial changes, and pre-existing histological damage, are discussed.


Asunto(s)
Lesión Renal Aguda/epidemiología , Glomerulonefritis/epidemiología , Insuficiencia Renal Crónica/epidemiología , Infecciones Estreptocócicas/epidemiología , Lesión Renal Aguda/microbiología , Niño , Glomerulonefritis/complicaciones , Glomerulonefritis/microbiología , Glomerulonefritis/patología , Humanos , Pronóstico , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/microbiología , Insuficiencia Renal Crónica/patología , Factores de Riesgo , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/patogenicidad
19.
Mol Microbiol ; 115(6): 1207-1228, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33325565

RESUMEN

Catabolite control protein A (CcpA) is a master regulator of carbon source utilization and contributes to the virulence of numerous medically important Gram-positive bacteria. Most functional assessments of CcpA, including interaction with its key co-factor HPr, have been performed in nonpathogenic bacteria. In this study we aimed to identify the in vivo DNA binding profile of CcpA and assess the extent to which HPr is required for CcpA-mediated regulation and DNA binding in the major human pathogen group A Streptococcus (GAS). Using a combination RNAseq/ChIP-seq approach, we found that CcpA affects transcript levels of 514 of 1667 GAS genes (31%) whereas direct DNA binding was identified for 105 GAS genes. Three of the directly regulated genes encode the key GAS virulence factors Streptolysin S, PrtS (IL-8 degrading proteinase), and SpeB (cysteine protease). Mutating CcpA Val301 to Ala (strain 2221-CcpA-V301A) abolished interaction between CcpA and HPr and impacted the transcript levels of 205 genes (40%) in the total CcpA regulon. By ChIP-seq analysis, CcpAV301A bound to DNA from 74% of genes bound by wild-type CcpA, but generally with lower affinity. These data delineate the direct CcpA regulon and clarify the HPr-dependent and independent activities of CcpA in a key pathogenic bacterium.


Asunto(s)
Proteínas Bacterianas/metabolismo , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Streptococcus pyogenes/metabolismo , Proteínas Bacterianas/genética , Proteínas Portadoras/metabolismo , Cromatina/genética , ADN Bacteriano/genética , Proteínas de Unión al ADN/genética , Exotoxinas/genética , Genoma Bacteriano/genética , Unión Proteica/fisiología , RNA-Seq , Proteínas Represoras/metabolismo , Serina Endopeptidasas/genética , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Estreptolisinas/genética , Virulencia/genética , Factores de Virulencia/genética
20.
Mol Med Rep ; 23(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33179113

RESUMEN

Rheumatic heart disease (RHD) is an autoimmune disease caused by rheumatic fever following group A hemolytic streptococcal infection and primarily affects the mitral valve. RHD is currently a major global health problem. However, the exact pathological mechanisms associated with RHD­induced cardiac valve damage remain to be elucidated. The endothelial­mesenchymal transition (EndMT) serves a key role in a number of diseases with an important role in cardiac fibrosis and the activin/Smad2 and 3 signaling pathway is involved in regulating the EndMT. Nevertheless, there are no studies to date, to the best of the authors' knowledge, investigating the association between RHD and EndMT. Thus, the aim of the current study was to investigate the potential role of EndMT in cardiac valve damage and assess whether activin/Smad2 and 3 signaling was activated during RHD­induced valvular injury in a rat model of RHD induced by inactivated Group A streptococci and complete Freund's adjuvant. Inflammation and fibrosis were assessed by hematoxylin and eosin and Sirius red staining. Serum cytokine and rheumatoid factor levels were measured using ELISA kits. Expression levels of activin/Smad2 and 3 signaling pathway­related factors [activin A, Smad2, Smad3, phosphorylated (p­)Smad2 and p­Smad3], EndMT­related factors [lymphoid enhancer factor­1 (LEF­1), Snail1, TWIST, zinc finger E­box­binding homeobox (ZEB)1, ZEB2, α smooth muscle actin (α­SMA) and type I collagen α 1 (COL1A1)], apoptosis­related markers (BAX and cleaved caspase­3) and valvular inflammation markers (NF­κB and p­NF­κB) were detected using reverse transcription­quantitative PCR and western blot analyses. Compared with the control group, the degree of valvular inflammation and fibrosis, serum levels of IL­6, IL­17, TNF­α and expression of apoptosis­related markers (BAX and cleaved caspase­3) and valvular inflammation marker (p­NF­κB), activin/Smad2 and 3 signaling pathway­related factors (activin A, p­Smad2 and p­Smad3), EndMT­related factors (LEF­1, Snail1, TWIST, ZEB 1, ZEB2, α­SMA and COL1A1) were significantly increased in the RHD group. These results suggested that the activin/Smad2 and 3 signaling pathway was activated during the development of valvular damage caused by RHD and that the EndMT is involved in RHD­induced cardiac valve damage.


Asunto(s)
Activinas/metabolismo , Válvula Mitral/patología , Cardiopatía Reumática/patología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Fibrosis , Adyuvante de Freund/efectos adversos , Válvula Mitral/metabolismo , Ratas , Cardiopatía Reumática/etiología , Cardiopatía Reumática/metabolismo , Transducción de Señal , Streptococcus pyogenes/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...