Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 18(7): e1010671, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35793394

RESUMEN

Blocking Plasmodium, the causative agent of malaria, at the asymptomatic pre-erythrocytic stage would abrogate disease pathology and prevent transmission. However, the lack of well-defined features within vaccine-elicited antibody responses that correlate with protection represents a major roadblock to improving on current generation vaccines. We vaccinated mice (BALB/cJ and C57BL/6J) with Py circumsporozoite protein (CSP), the major surface antigen on the sporozoite, and evaluated vaccine-elicited humoral immunity and identified immunological factors associated with protection after mosquito bite challenge. Vaccination achieved 60% sterile protection and otherwise delayed blood stage patency in BALB/cJ mice. In contrast, all C57BL/6J mice were infected similar to controls. Protection was mediated by antibodies and could be passively transferred from immunized BALB/cJ mice into naïve C57BL/6J. Dissection of the underlying immunological features of protection revealed early deficits in antibody titers and polyclonal avidity in C57BL/6J mice. Additionally, PyCSP-vaccination in BALB/cJ induced a significantly higher proportion of antigen-specific B-cells and class-switched memory B-cell (MBCs) populations than in C57BL/6J mice. Strikingly, C57BL/6J mice also had markedly fewer CSP-specific germinal center experienced B cells and class-switched MBCs compared to BALB/cJ mice. Analysis of the IgG γ chain repertoires by next generation sequencing in PyCSP-specific memory B-cell repertoires also revealed higher somatic hypermutation rates in BALB/cJ mice than in C57BL/6J mice. These findings indicate that the development of protective antibody responses in BALB/cJ mice in response to vaccination with PyCSP was associated with increased germinal center activity and somatic mutation compared to C57BL/6J mice, highlighting the key role B cell maturation may have in the development of vaccine-elicited protective antibodies against CSP.


Asunto(s)
Vacunas contra la Malaria , Malaria , Animales , Anticuerpos Antiprotozoarios , Formación de Anticuerpos , Centro Germinal , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Protozoarias/genética
2.
Cell Rep ; 36(5): 109489, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34348141

RESUMEN

Both subunit and attenuated whole-sporozoite vaccination strategies against Plasmodium infection have shown promising initial results in malaria-naive westerners but less efficacy in malaria-exposed individuals in endemic areas. Here, we demonstrate proof of concept by using a rodent malaria model in which non-neutralizing antibodies (nNAbs) can directly interfere with protective anti-circumsporozoite protein (CSP) humoral responses. We characterize a monoclonal antibody, RAM1, against Plasmodium yoelii sporozoite major surface antigen CSP. Unlike the canonical PyCSP repeat domain binding and neutralizing antibody (NAb) 2F6, RAM1 does not inhibit sporozoite traversal or entry of hepatocytes in vitro or infection in vivo. Although 2F6 and RAM1 bind non-overlapping regions of the CSP-repeat domain, pre-treatment with RAM1 abrogates the capacity of NAb to block sporozoite traversal and invasion in vitro. Importantly, RAM1 reduces the efficacy of the polyclonal humoral response against PyCSP in vivo. Collectively, our data provide a proof of concept that nNAbs can alter the efficacy of malaria vaccination.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antiprotozoarios/inmunología , Inmunidad Humoral , Estadios del Ciclo de Vida , Hígado/parasitología , Plasmodium yoelii/crecimiento & desarrollo , Plasmodium yoelii/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/aislamiento & purificación , Línea Celular , Epítopos/inmunología , Femenino , Cinética , Vacunas contra la Malaria/inmunología , Ratones Endogámicos BALB C , Modelos Biológicos , Unión Proteica , Dominios Proteicos , Proteínas Protozoarias/química , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología , Vacunas Sintéticas/inmunología
3.
J Biol Chem ; 293(3): 941-952, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29180448

RESUMEN

Clostridium difficile infection is the leading cause of hospital-acquired diarrhea and is mediated by the actions of two toxins, TcdA and TcdB. The toxins perturb host cell function through a multistep process of receptor binding, endocytosis, low pH-induced pore formation, and the translocation and delivery of an N-terminal glucosyltransferase domain that inactivates host GTPases. Infection studies with isogenic strains having defined toxin deletions have established TcdB as an important target for therapeutic development. Monoclonal antibodies that neutralize TcdB function have been shown to protect against C. difficile infection in animal models and reduce recurrence in humans. Here, we report the mechanism of TcdB neutralization by PA41, a humanized monoclonal antibody capable of neutralizing TcdB from a diverse array of C. difficile strains. Through a combination of structural, biochemical, and cell functional studies, involving X-ray crystallography and EM, we show that PA41 recognizes a single, highly conserved epitope on the TcdB glucosyltransferase domain and blocks productive translocation and delivery of the enzymatic cargo into the host cell. Our study reveals a unique mechanism of C. difficile toxin neutralization by a monoclonal antibody, which involves targeting a process that is conserved across the large clostridial glucosylating toxins. The PA41 antibody described here provides a valuable tool for dissecting the mechanism of toxin pore formation and translocation across the endosomal membrane.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Enterotoxinas/metabolismo , Anticuerpos Monoclonales/metabolismo , Toxinas Bacterianas/química , Células CACO-2 , Clostridioides difficile/enzimología , Cristalografía por Rayos X , Citosol/metabolismo , Enterotoxinas/química , Humanos , Concentración de Iones de Hidrógeno , Microscopía Electrónica , Rubidio/química , Proteína de Unión al GTP rac1/química , Proteína de Unión al GTP rac1/metabolismo
4.
FEMS Microbiol Rev ; 41(6): 723-750, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29048477

RESUMEN

Clostridium difficile is a bacterial pathogen that is the leading cause of nosocomial antibiotic-associated diarrhea and pseudomembranous colitis worldwide. The incidence, severity, mortality and healthcare costs associated with C. difficile infection (CDI) are rising, making C. difficile a major threat to public health. Traditional treatments for CDI involve use of antibiotics such as metronidazole and vancomycin, but disease recurrence occurs in about 30% of patients, highlighting the need for new therapies. The pathogenesis of C. difficile is primarily mediated by the actions of two large clostridial glucosylating toxins, toxin A (TcdA) and toxin B (TcdB). Some strains produce a third toxin, the binary toxin C. difficile transferase, which can also contribute to C. difficile virulence and disease. These toxins act on the colonic epithelium and immune cells and induce a complex cascade of cellular events that result in fluid secretion, inflammation and tissue damage, which are the hallmark features of the disease. In this review, we summarize our current understanding of the structure and mechanism of action of the C. difficile toxins and their role in disease.


Asunto(s)
Toxinas Bacterianas/toxicidad , Clostridioides difficile/química , Infecciones por Clostridium/patología , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/terapia , Humanos , Inmunidad/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología
5.
J Biol Chem ; 292(35): 14401-14412, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28705932

RESUMEN

Clostridium difficile is a clinically significant pathogen that causes mild-to-severe (and often recurrent) colon infections. Disease symptoms stem from the activities of two large, multidomain toxins known as TcdA and TcdB. The toxins can bind, enter, and perturb host cell function through a multistep mechanism of receptor binding, endocytosis, pore formation, autoproteolysis, and glucosyltransferase-mediated modification of host substrates. Monoclonal antibodies that neutralize toxin activity provide a survival benefit in preclinical animal models and prevent recurrent infections in human clinical trials. However, the molecular mechanisms involved in these neutralizing activities are unclear. To this end, we performed structural studies on a neutralizing monoclonal antibody, PA50, a humanized mAb with both potent and broad-spectrum neutralizing activity, in complex with TcdA. Electron microscopy imaging and multiangle light-scattering analysis revealed that PA50 binds multiple sites on the TcdA C-terminal combined repetitive oligopeptides (CROPs) domain. A crystal structure of two PA50 Fabs bound to a segment of the TcdA CROPs helped define a conserved epitope that is distinct from previously identified carbohydrate-binding sites. Binding of TcdA to the host cell surface was directly blocked by either PA50 mAb or Fab and suggested that receptor blockade is the mechanism by which PA50 neutralizes TcdA. These findings highlight the importance of the CROPs C terminus in cell-surface binding and a role for neutralizing antibodies in defining structural features critical to a pathogen's mechanism of action. We conclude that PA50 protects host cells by blocking the binding of TcdA to cell surfaces.


Asunto(s)
Antibacterianos/metabolismo , Anticuerpos Neutralizantes/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/enzimología , Enterocitos/metabolismo , Enterotoxinas/metabolismo , Glucosiltransferasas/metabolismo , Modelos Moleculares , Secuencia de Aminoácidos , Antibacterianos/química , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/metabolismo , Anticuerpos Neutralizantes/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/toxicidad , Sitios de Unión de Anticuerpos , Células CACO-2 , Secuencia Conservada , Cristalografía por Rayos X , Enterocitos/efectos de los fármacos , Enterotoxinas/química , Enterotoxinas/genética , Enterotoxinas/toxicidad , Mapeo Epitopo , Glucosiltransferasas/química , Glucosiltransferasas/genética , Glucosiltransferasas/toxicidad , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/toxicidad , Secuencias Repetitivas de Aminoácido
6.
PLoS Pathog ; 12(12): e1006070, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27942025

RESUMEN

Clostridium difficile infection affects a significant number of hospitalized patients in the United States. Two homologous exotoxins, TcdA and TcdB, are the major virulence factors in C. difficile pathogenesis. The toxins are glucosyltransferases that inactivate Rho family-GTPases to disrupt host cellular function and cause fluid secretion, inflammation, and cell death. Toxicity depends on receptor binding and subsequent endocytosis. TcdB has been shown to enter cells by clathrin-dependent endocytosis, but the mechanism of TcdA uptake is still unclear. Here, we utilize a combination of RNAi-based knockdown, pharmacological inhibition, and cell imaging approaches to investigate the endocytic mechanism(s) that contribute to TcdA uptake and subsequent cytopathic and cytotoxic effects. We show that TcdA uptake and cellular intoxication is dynamin-dependent but does not involve clathrin- or caveolae-mediated endocytosis. Confocal microscopy using fluorescently labeled TcdA shows significant colocalization of the toxin with PACSIN2-positive structures in cells during entry. Disruption of PACSIN2 function by RNAi-based knockdown approaches inhibits TcdA uptake and toxin-induced downstream effects in cells indicating that TcdA entry is PACSIN2-dependent. We conclude that TcdA and TcdB utilize distinct endocytic mechanisms to intoxicate host cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Toxinas Bacterianas/metabolismo , Infecciones por Clostridium/metabolismo , Endocitosis/fisiología , Enterotoxinas/metabolismo , Animales , Western Blotting , Células CACO-2 , Clatrina , Clostridioides difficile , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Procesamiento de Imagen Asistido por Computador , Ratones , Microscopía Confocal , Transporte de Proteínas/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Factores de Virulencia/metabolismo
7.
Sci Total Environ ; 538: 78-85, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26298250

RESUMEN

Escherichia coli is used to indicate fecal contamination in freshwater systems and is an indicator of the potential presence of human pathogens. However, naturalized E. coli strains that persist and grow in the environment confound the use of this bacterium as a fecal indicator. Here we examined the spatial and temporal distribution of E. coli in water and sediments of the Seven Mile Creek (SMC), a constructed, ephemeral watershed. E. coli concentrations showed variation by site and date, likely due to changes in temperature and rainfall. Horizontal fluorophore enhanced rep-PCR (HFERP) DNA fingerprint analyses indicated that E. coli populations were very diverse and consisted of transient and naturalized strains, which were especially prevalent in sediment. E. coli fingerprints from water and sediment collected in the same year clustered together with significant overlap, indicating exchange of strains between matrices. Isolates obtained during periods of flow, but not during non-flow conditions, clustered together regardless of sample site, indicating that transport between sites occurred. Naturalized E. coli strains were found in the SMC and strains become geographically isolated and distinct during non-flow conditions. Isolates collected during late spring to fall clustered together at each site, suggesting that temperature and growth of naturalized strains are likely factors affecting population dynamics. Results of this study show that newly introduced and naturalized E. coli strains are present in the SMC. Results of this study highlight an important concern for resource managers using this species for water quality monitoring.


Asunto(s)
Monitoreo del Ambiente , Escherichia coli/genética , Sedimentos Geológicos/microbiología , Microbiología del Agua , Contaminación del Agua/análisis , Dermatoglifia del ADN , ADN Bacteriano , Escherichia coli/clasificación , Escherichia coli/aislamiento & purificación , Variación Genética , Genotipo , Ohio , Reacción en Cadena de la Polimerasa
8.
Proc Natl Acad Sci U S A ; 112(22): 7073-8, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26038560

RESUMEN

Clostridium difficile is the leading cause of hospital-acquired diarrhea in the United States. The two main virulence factors of C. difficile are the large toxins, TcdA and TcdB, which enter colonic epithelial cells and cause fluid secretion, inflammation, and cell death. Using a gene-trap insertional mutagenesis screen, we identified poliovirus receptor-like 3 (PVRL3) as a cellular factor necessary for TcdB-mediated cytotoxicity. Disruption of PVRL3 expression by gene-trap mutagenesis, shRNA, or CRISPR/Cas9 mutagenesis resulted in resistance of cells to TcdB. Complementation of the gene-trap or CRISPR mutants with PVRL3 resulted in restoration of TcdB-mediated cell death. Purified PVRL3 ectodomain bound to TcdB by pull-down. Pretreatment of cells with a monoclonal antibody against PVRL3 or prebinding TcdB to PVRL3 ectodomain also inhibited cytotoxicity in cell culture. The receptor is highly expressed on the surface epithelium of the human colon and was observed to colocalize with TcdB in both an explant model and in tissue from a patient with pseudomembranous colitis. These data suggest PVRL3 is a physiologically relevant binding partner that can serve as a target for the prevention of TcdB-induced cytotoxicity in C. difficile infection.


Asunto(s)
Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Moléculas de Adhesión Celular/metabolismo , Clostridioides difficile/química , Enterotoxinas/toxicidad , Células Epiteliales/metabolismo , Análisis de Varianza , Anticuerpos Monoclonales/metabolismo , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Células CACO-2 , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Colon/metabolismo , Enterotoxinas/metabolismo , Prueba de Complementación Genética , Células HeLa , Humanos , Mutagénesis Insercional , Nectinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA