Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 20(7): e1012282, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38990812

RESUMEN

Haemophilus influenzae is a human respiratory pathogen and inhabits the human respiratory tract as its only niche. Despite this, the molecular mechanisms that allow H. influenzae to establish persistent infections of human epithelia are not well understood. Here, we have investigated how H. influenzae adapts to the host environment and triggers the host immune response using a human primary cell-based infection model that closely resembles human nasal epithelia (NHNE). Physiological assays combined with dualRNAseq revealed that NHNE from five healthy donors all responded to H. influenzae infection with an initial, 'unproductive' inflammatory response that included a strong hypoxia signature but did not produce pro-inflammatory cytokines. Subsequently, an apparent tolerance to large extracellular and intraepithelial burdens of H. influenzae developed, with NHNE transcriptional profiles resembling the pre-infection state. This occurred in parallel with the development of intraepithelial bacterial populations, and appears to involve interruption of NFκB signalling. This is the first time that large-scale, persistence-promoting immunomodulatory effects of H. influenzae during infection have been observed, and we were able to demonstrate that only infections with live, but not heat-killed H. influenzae led to immunomodulation and reduced expression of NFκB-controlled cytokines such as IL-1ß, IL-36γ and TNFα. Interestingly, NHNE were able to re-activate pro-inflammatory responses towards the end of the 14-day infection, resulting in release of IL-8 and TNFα. In addition to providing first molecular insights into mechanisms enabling persistence of H. influenzae in the host, our data further indicate the presence of infection stage-specific gene expression modules, highlighting fundamental similarities between immune responses in NHNE and canonical immune cells, which merit further investigation.


Asunto(s)
Células Epiteliales , Infecciones por Haemophilus , Haemophilus influenzae , Humanos , Haemophilus influenzae/inmunología , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/microbiología , Células Epiteliales/microbiología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Mucosa Nasal/microbiología , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Tolerancia Inmunológica , Células Cultivadas , Citocinas/metabolismo
2.
PLoS Pathog ; 18(1): e1010209, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35085362

RESUMEN

Haemophilus influenzae (Hi) infections are associated with recurring acute exacerbations of chronic respiratory diseases in children and adults including otitis media, pneumonia, chronic obstructive pulmonary disease and asthma. Here, we show that persistence and recurrence of Hi infections are closely linked to Hi metabolic properties, where preferred growth substrates are aligned to the metabolome of human airway epithelial surfaces and include lactate, pentoses, and nucleosides, but not glucose that is typically used for studies of Hi growth in vitro. Enzymatic and physiological investigations revealed that utilization of lactate, the preferred Hi carbon source, required the LldD L-lactate dehydrogenase (conservation: 98.8% of strains), but not the two redox-balancing D-lactate dehydrogenases Dld and LdhA. Utilization of preferred substrates was directly linked to Hi infection and persistence. When unable to utilize L-lactate or forced to rely on salvaged guanine, Hi showed reduced extra- and intra-cellular persistence in a murine model of lung infection and in primary normal human nasal epithelia, with up to 3000-fold attenuation observed in competitive infections. In contrast, D-lactate dehydrogenase mutants only showed a very slight reduction compared to the wild-type strain. Interestingly, acetate, the major Hi metabolic end-product, had anti-inflammatory effects on cultured human tissue cells in the presence of live but not heat-killed Hi, suggesting that metabolic endproducts also influence HI-host interactions. Our work provides significant new insights into the critical role of metabolism for Hi persistence in contact with host cells and reveals for the first time the immunomodulatory potential of Hi metabolites.


Asunto(s)
Infecciones por Haemophilus/metabolismo , Haemophilus influenzae/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Animales , Interacciones Huésped-Patógeno/fisiología , Humanos , Ratones
3.
PLoS Pathog ; 14(10): e1007348, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30321240

RESUMEN

Human Group IIA secreted phospholipase A2 (hGIIA) is an acute phase protein with bactericidal activity against Gram-positive bacteria. Infection models in hGIIA transgenic mice have suggested the importance of hGIIA as an innate defense mechanism against the human pathogens Group A Streptococcus (GAS) and Group B Streptococcus (GBS). Compared to other Gram-positive bacteria, GAS is remarkably resistant to hGIIA activity. To identify GAS resistance mechanisms, we exposed a highly saturated GAS M1 transposon library to recombinant hGIIA and compared relative mutant abundance with library input through transposon-sequencing (Tn-seq). Based on transposon prevalence in the output library, we identified nine genes, including dltA and lytR, conferring increased hGIIA susceptibility. In addition, seven genes conferred increased hGIIA resistance, which included two genes, gacH and gacI that are located within the Group A Carbohydrate (GAC) gene cluster. Using GAS 5448 wild-type and the isogenic gacI mutant and gacI-complemented strains, we demonstrate that loss of the GAC N-acetylglucosamine (GlcNAc) side chain in the ΔgacI mutant increases hGIIA resistance approximately 10-fold, a phenotype that is conserved across different GAS serotypes. Increased resistance is associated with delayed penetration of hGIIA through the cell wall. Correspondingly, loss of the Lancefield Group B Carbohydrate (GBC) rendered GBS significantly more resistant to hGIIA-mediated killing. This suggests that the streptococcal Lancefield antigens, which are critical determinants for streptococcal physiology and virulence, are required for the bactericidal enzyme hGIIA to exert its bactericidal function.


Asunto(s)
Antibacterianos/farmacología , Pared Celular/metabolismo , Fosfolipasas A2 Grupo II/inmunología , Inmunidad Innata/efectos de los fármacos , Polisacáridos Bacterianos/farmacología , Infecciones Estreptocócicas/microbiología , Streptococcus/inmunología , Actividad Bactericida de la Sangre , Fosfolipasas A2 Grupo II/sangre , Fosfolipasas A2 Grupo II/genética , Interacciones Huésped-Patógeno , Humanos , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/enzimología , Streptococcus/patogenicidad
4.
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
5.
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
6.
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
7.
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
8.
J Infect Dis ; 210(8): 1325-38, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24799598

RESUMEN

Streptococcus pyogenes ranks among the main causes of mortality from bacterial infections worldwide. Currently there is no vaccine to prevent diseases such as rheumatic heart disease and invasive streptococcal infection. The streptococcal M protein that is used as the substrate for epidemiological typing is both a virulence factor and a vaccine antigen. Over 220 variants of this protein have been described, making comparisons between proteins difficult, and hindering M protein-based vaccine development. A functional classification based on 48 emm-clusters containing closely related M proteins that share binding and structural properties is proposed. The need for a paradigm shift from type-specific immunity against S. pyogenes to emm-cluster based immunity for this bacterium should be further investigated. Implementation of this emm-cluster-based system as a standard typing scheme for S. pyogenes will facilitate the design of future studies of M protein function, streptococcal virulence, epidemiological surveillance, and vaccine development.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Vacunas Estreptocócicas/inmunología , Streptococcus pyogenes/clasificación , Streptococcus pyogenes/fisiología , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Clonación Molecular , Datos de Secuencia Molecular , Filogenia , Proteínas Recombinantes
9.
Curr Top Microbiol Immunol ; 368: 207-42, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23250780

RESUMEN

Currently there is no commercial Group A Streptococcus (GAS; S. pyogenes) vaccine available. The development of safe GAS vaccines is challenging, researchers are confronted with obstacles such as the occurrence of many unique serotypes (there are greater than 150 M types), antigenic variation within the same serotype, large variations in the geographical distribution of serotypes, and the production of antibodies cross-reactive with human tissue which can lead to host auto-immune disease. Cell wall anchored, cell membrane associated, secreted and anchorless proteins have all been targeted as GAS vaccine candidates. As GAS is an exclusively human pathogen, the quest for an efficacious vaccine is further complicated by the lack of an animal model which mimics human disease and can be consistently and reproducibly colonized by multiple GAS strains.


Asunto(s)
Vacunas Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/inmunología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Humanos
10.
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
11.
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.

12.
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
13.
Nanomedicine ; 9(7): 935-44, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23428988

RESUMEN

A novel vaccine development platform that enables the site-specific conjugation of synthetic lipid adjuvants to recombinant proteins was produced. This technology facilitates the simple and efficient production of homogeneous, chemically-defined, semisynthetic lipoprotein vaccines. Using a polytope 'string-of-beads' approach, a synthetic gene incorporating seven Streptococcus pyogenes M protein strain-specific antigens, and a conserved M protein antigen (J14) was produced, expressed, and attached to a lipoamino acid based adjuvant (lipid core peptide; LCP). Nanoparticles (40 nm diameter) of an optimal size for stimulating antibody-mediated immunity were formed upon the addition of these lipoproteins to aqueous buffer (PBS). Systemic antigen-specific IgG antibodies were raised against all eight antigens in C57BL/6J mice, without the need to formulate with additional adjuvant. These antibodies bound cell surface M proteins of S. pyogenes strains represented within the polytope sequence, with higher antibody levels observed where a dendritic cell targeting peptide (DCpep) was incorporated within the LCP adjuvant. FROM THE CLINICAL EDITOR: In this study, a novel vaccine development system is presented, combining adjuvants with recombinant protein antigens, and presenting the antigen in a nanoparticle system optimized for antibody production. They demonstrate efficient vaccination in a murine model system without the need for additional adjuvants.


Asunto(s)
Adyuvantes Inmunológicos/química , Lípidos/química , Nanopartículas/química , Vacunas Estreptocócicas/inmunología , Animales , Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Técnica del Anticuerpo Fluorescente , Inmunidad , Lipoproteínas/química , Maleimidas/química , Ratones , Ratones Endogámicos C57BL , Nanopartículas/ultraestructura , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/síntesis química , Vacunas Estreptocócicas/química , Streptococcus pyogenes/inmunología
14.
J Cell Signal ; 3(4): 193-206, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36777036

RESUMEN

Oxidative stress (OS) in the airway epithelium is associated with inflammation, cell damage, and mitochondrial dysfunction that may initiate or worsen respiratory disease. Redox regulation maintains the equilibrium of pro-oxidant/antioxidant reactions but can be disturbed by environmental exposures. The mechanism(s) underlying the induction and impact of OS on airway epithelium and how these influences on respiratory disease is poorly understood. The aim of this study was to develop a stress response model in primary human nasal epithelial cells (NECs) grown at the air-liquid interface (ALI) into a well-differentiated epithelium and to use this model to investigate the mechanisms underlying OS. Hydrogen peroxide (H2O2) was used to induce acute OS and the responses were measured with trans epithelial electrical resistance (TEER), membrane permeability, cell death (LDH release), mitochondrial reactive oxygen species (mtROS) generation, redox status (GSH/GSSG ratio), cellular ATP, and signaling pathways (SIRT1, FOXO3, p53, p21, PINK1, PARKIN, NRF2). Following 25 mM (sensitive) or 50mM (resistant) H2O2 exposure, cell integrity decreased (p<0.05), GSH/GSSG ratio reduced (p<0.05), and ATP production declined by 83% (p<0.05) in the sensitive and 55% (p<0.05) in the resistant group; mtROS production increased 3.4-fold (p<0.001). Significant inter-individual differences between healthy humans with regards to susceptibility to OS, and differential activation of various pathways (FOXO3, PARKIN) were observed. These intra-individual differences in susceptibility to OS may be attributed to resistant individuals having more mitochondria or greater mitochondrial function.

15.
16.
Sci Rep ; 9(1): 2627, 2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30796268

RESUMEN

Respiratory disease is a major cause of morbidity and mortality in patients with ataxia-telangiectasia (A-T) who are prone to recurrent sinopulmonary infections, bronchiectasis, pulmonary fibrosis, and pulmonary failure. Upper airway infections are common in patients and S. pneumoniae is associated with these infections. We demonstrate here that the upper airway microbiome in patients with A-T is different from that to healthy controls, with S. pneumoniae detected largely in patients only. Patient-specific airway epithelial cells and differentiated air-liquid interface cultures derived from these were hypersensitive to infection which was at least in part due to oxidative damage since it was partially reversed by catalase. We also observed increased levels of the pro-inflammatory cytokines IL-8 and TNF-α (inflammasome-independent) and a decreased level of the inflammasome-dependent cytokine IL-ß in patient cells. Further investigation revealed that the ASC-Caspase 1 signalling pathway was defective in A-T airway epithelial cells. These data suggest that the heightened susceptibility of these cells to S. pneumoniae infection is due to both increased oxidative damage and a defect in inflammasome activation, and has implications for lung disease in these patients.


Asunto(s)
Ataxia Telangiectasia/patología , Células Epiteliales/patología , Inmunidad Innata , Pulmón/patología , Estrés Oxidativo , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/patología , Streptococcus pneumoniae/fisiología , Adolescente , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Estudios de Casos y Controles , Diferenciación Celular , Células Cultivadas , Niño , Preescolar , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Femenino , Humanos , Inflamasomas/metabolismo , Inflamación/patología , Pulmón/microbiología , Masculino , Nariz/patología
17.
mBio ; 9(1)2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382733

RESUMEN

Classification of streptococci is based upon expression of unique cell wall carbohydrate antigens. All serotypes of group A Streptococcus (GAS; Streptococcus pyogenes), a leading cause of infection-related mortality worldwide, express the group A carbohydrate (GAC). GAC, the classical Lancefield antigen, is comprised of a polyrhamnose backbone with N-acetylglucosamine (GlcNAc) side chains. The immunodominant GlcNAc epitope of GAC is the basis of all rapid diagnostic testing for GAS infection. We previously identified the 12-gene GAC biosynthesis gene cluster and determined that the glycosyltransferase GacI was required for addition of the GlcNAc side chain to the polyrhamnose core. Loss of the GAC GlcNAc epitope in serotype M1 GAS resulted in attenuated virulence in two animal infection models and increased GAS sensitivity to killing by whole human blood, serum, neutrophils, and antimicrobial peptides. Here, we report that the GAC biosynthesis gene cluster is ubiquitous among 520 GAS isolates from global sources, representing 105 GAS emm serotypes. Isogenic ΔgacI mutants were constructed in M2, M3, M4, M28, and M89 backgrounds and displayed an array of phenotypes in susceptibility to killing by whole human blood, baby rabbit serum, human platelet releasate, human neutrophils, and antimicrobial peptide LL-37. The contribution of the GlcNAc side chain to GAS survival in vivo also varied by strain, demonstrating that it is not a prerequisite for virulence in the murine infection model. Thus, the relative contribution of GAC to virulence in non-M1 serotypes appears to depend on the quorum of other virulence factors that each strain possesses.IMPORTANCE The Lancefield group A carbohydrate (GAC) is the species-defining antigen for group A Streptococcus (GAS), comprising ~50% of the cell wall of this major human pathogen. We previously showed that the GlcNAc side chain of GAC contributes to the innate immune resistance and animal virulence phenotypes of the globally disseminated strain of serotype M1 GAS. Here, we use isogenic mutagenesis to examine the role of GAC GlcNAc in five additional medically relevant GAS serotypes. Overall, the GlcNAc side chain of GAC contributes to the innate immune resistance of GAS, but the relative contribution varies among individual strains. Moreover, the GAC GlcNAc side chain is not a universal prerequisite for GAS virulence in the animal model.


Asunto(s)
Acetilglucosamina/metabolismo , Antígenos Bacterianos/metabolismo , Pared Celular/metabolismo , Polisacáridos Bacterianos/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/patogenicidad , Factores de Virulencia/metabolismo , Animales , Antígenos Bacterianos/genética , Actividad Bactericida de la Sangre , Modelos Animales de Enfermedad , Eliminación de Gen , Humanos , Ratones , Polisacáridos Bacterianos/genética , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/genética , Análisis de Supervivencia , Virulencia , Factores de Virulencia/genética
18.
PLoS One ; 11(6): e0156639, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27310707

RESUMEN

The C-terminal region of the M-protein of Streptococcus pyogenes is a major target for vaccine development. The major feature is the C-repeat region, consisting of 35-42 amino acid repeat units that display high but not perfect identity. SV1 is a S. pyogenes vaccine candidate that incorporates five 14mer amino acid sequences (called J14i variants) from differing C-repeat units in a single recombinant construct. Here we show that the J14i variants chosen for inclusion in SV1 are the most common variants in a dataset of 176 unique M-proteins. Murine antibodies raised against SV1 were shown to bind to each of the J14i variants present in SV1, as well as variants not present in the vaccine. Antibodies raised to the individual J14i variants were also shown to bind to multiple but different combinations of J14i variants, supporting the underlying rationale for the design of SV1. A Lewis Rat Model of valvulitis was then used to assess the capacity of SV1 to induce deleterious immune response associated with rheumatic heart disease. In this model, both SV1 and the M5 positive control protein were immunogenic. Neither of these antibodies were cross-reactive with cardiac myosin or collagen. Splenic T cells from SV1/CFA and SV1/alum immunized rats did not proliferate in response to cardiac myosin or collagen. Subsequent histological examination of heart tissue showed that 4 of 5 mice from the M5/CFA group had valvulitis and inflammatory cell infiltration into valvular tissue, whereas mice immunised with SV1/CFA, SV1/alum showed no sign of valvulitis. These results suggest that SV1 is a safe vaccine candidate that will elicit antibodies that recognise the vast majority of circulating GAS M-types.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Antígenos Bacterianos/inmunología , Cardiopatía Reumática/prevención & control , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/administración & dosificación , Streptococcus pyogenes/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Alumbre/administración & dosificación , Animales , Antígenos Bacterianos/genética , Colágeno/genética , Colágeno/metabolismo , Femenino , Expresión Génica , Válvulas Cardíacas/efectos de los fármacos , Válvulas Cardíacas/inmunología , Válvulas Cardíacas/microbiología , Válvulas Cardíacas/patología , Ratones , Ratones Endogámicos BALB C , Miosinas/genética , Miosinas/metabolismo , Ratas , Ratas Endogámicas Lew , Secuencias Repetitivas de Aminoácido , Cardiopatía Reumática/inmunología , Cardiopatía Reumática/microbiología , Cardiopatía Reumática/patología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/microbiología , Bazo/patología , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Vacunas Estreptocócicas/biosíntesis , Vacunas Estreptocócicas/inmunología , Streptococcus pyogenes/efectos de los fármacos , Streptococcus pyogenes/genética , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/microbiología , Linfocitos T/patología , Vacunas Sintéticas
19.
J Mol Med (Berl) ; 94(2): 219-33, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26411873

RESUMEN

UNLABELLED: Inhibitory CD33-related Siglec receptors regulate immune cell activation upon engaging ubiquitous sialic acids (Sias) on host cell surface glycans. Through molecular mimicry, Sia-expressing pathogen group B Streptococcus binds inhibitory human Siglec-9 (hSiglec-9) to blunt neutrophil activation and promote bacterial survival. We unexpectedly discovered that hSiglec-9 also specifically binds high molecular weight hyaluronan (HMW-HA), another ubiquitous host glycan, through a region of its terminal Ig-like V-set domain distinct from the Sia-binding site. HMW-HA recognition by hSiglec-9 limited neutrophil extracellular trap (NET) formation, oxidative burst, and apoptosis, defining HMW-HA as a regulator of neutrophil activation. However, the pathogen group A Streptococcus (GAS) expresses a HMW-HA capsule that engages hSiglec-9, blocking NET formation and oxidative burst, thereby promoting bacterial survival. Thus, a single inhibitory lectin receptor detects two distinct glycan "self-associated molecular patterns" to maintain neutrophil homeostasis, and two leading human bacterial pathogens have independently evolved molecular mimicry to exploit this immunoregulatory mechanism. KEY MESSAGE: HMW-HA is the first example of a non-sialic acid containing glycan to be recognized by CD33-related Siglecs. HMW-HA engagement of hSiglec-9 attenuates neutrophil activation. Group A Streptococcus exploits hSiglec-9 recognition via its polysaccharide HMW-HA capsule to subvert neutrophil killing.


Asunto(s)
Antígenos CD/metabolismo , Interacciones Huésped-Patógeno , Ácido Hialurónico/metabolismo , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Animales , Antígenos CD/química , Antígenos CD/genética , Apoptosis/genética , Apoptosis/inmunología , Bacterias/inmunología , Bacterias/metabolismo , Quimiotaxis de Leucocito/inmunología , Trampas Extracelulares/genética , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Ácido Hialurónico/química , Inmunidad Innata , Fragmentos Fc de Inmunoglobulinas/metabolismo , Peso Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión , Estallido Respiratorio/inmunología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/química , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/genética , Streptococcus/fisiología
20.
mBio ; 7(3)2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27302756

RESUMEN

UNLABELLED: Group A Streptococcus (GAS) is an important human pathogen responsible for both superficial infections and invasive diseases. Autoimmune sequelae may occur upon repeated infection. For this reason, development of a vaccine against GAS represents a major challenge, since certain GAS components may trigger autoimmunity. We formulated three combination vaccines containing the following: (i) streptolysin O (SLO), interleukin 8 (IL-8) protease (Streptococcus pyogenes cell envelope proteinase [SpyCEP]), group A streptococcal C5a peptidase (SCPA), arginine deiminase (ADI), and trigger factor (TF); (ii) the conserved M-protein-derived J8 peptide conjugated to ADI; and (iii) group A carbohydrate lacking the N-acetylglucosamine side chain conjugated to ADI. We compared these combination vaccines to a "gold standard" for immunogenicity, full-length M1 protein. Vaccines were adjuvanted with alum, and mice were immunized on days 0, 21, and 28. On day 42, mice were challenged via cutaneous or subcutaneous routes. High-titer antigen-specific antibody responses with bactericidal activity were detected in mouse serum samples for all vaccine candidates. In comparison with sham-immunized mice, all vaccines afforded protection against cutaneous challenge. However, only full-length M1 protein provided protection in the subcutaneous invasive disease model. IMPORTANCE: This set of experiments demonstrates the inherent variability of mouse models for the characterization of GAS vaccine candidate protective efficacy. Such variability poses an important challenge for GAS vaccine development, as advancement of candidates to human clinical trials requires strong evidence of efficacy. This study highlights the need for an open discussion within the field regarding standardization of animal models for GAS vaccine development.


Asunto(s)
Bacteriemia/prevención & control , Enfermedades Cutáneas Bacterianas/prevención & control , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/administración & dosificación , Vacunas Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Alumbre/administración & dosificación , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Bacteriemia/inmunología , Actividad Bactericida de la Sangre , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , Enfermedades Cutáneas Bacterianas/inmunología , Infecciones Estreptocócicas/inmunología , Resultado del Tratamiento , Vacunas Combinadas/administración & dosificación , Vacunas Combinadas/inmunología , Vacunas Conjugadas/administración & dosificación , Vacunas Conjugadas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA