Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Crohns Colitis ; 18(9): 1381-1393, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-38572716

RESUMEN

BACKGROUND: Faecal microbiota transplantation [FMT] shows some efficacy in treating patients with ulcerative colitis [UC], although variability has been observed among donors and treatment regimens. We investigated the effect of FMT using rationally selected donors after pretreatment with budesonide or placebo in active UC. METHODS: Patients ≥18 years old with mild to moderate active UC were randomly assigned to 3 weeks of budesonide [9 mg] or placebo followed by 4-weekly infusions of a donor faeces suspension. Two donors were selected based on microbiota composition, regulatory T cell induction and short-chain fatty acid production in mice. The primary endpoint was engraftment of donor microbiota after FMT. In addition, clinical efficacy was assessed. RESULTS: In total, 24 patients were enrolled. Pretreatment with budesonide did not increase donor microbiota engraftment [p = 0.56] nor clinical response, and engraftment was not associated with clinical response. At week 14, 10/24 [42%] patients achieved [partial] remission. Remarkably, patients treated with FMT suspensions from one donor were associated with clinical response [80% of responders, p < 0.05] but had lower overall engraftment of donor microbiota. Furthermore, differences in the taxonomic composition of the donors and the engraftment of certain taxa were associated with clinical response. CONCLUSION: In this small study, pretreatment with budesonide did not significantly influence engraftment or clinical response after FMT. However, clinical response appeared to be donor-dependent. Response to FMT may be related to transfer of specific strains instead of overall engraftment, demonstrating the need to characterize mechanisms of actions of strains that maximize therapeutic benefit in UC.


Asunto(s)
Budesonida , Colitis Ulcerosa , Trasplante de Microbiota Fecal , Humanos , Trasplante de Microbiota Fecal/métodos , Budesonida/uso terapéutico , Budesonida/administración & dosificación , Colitis Ulcerosa/terapia , Colitis Ulcerosa/microbiología , Femenino , Masculino , Adulto , Proyectos Piloto , Persona de Mediana Edad , Resultado del Tratamiento , Microbioma Gastrointestinal/efectos de los fármacos , Método Doble Ciego , Selección de Donante/métodos , Antiinflamatorios/uso terapéutico , Heces/microbiología
2.
JAMA ; 329(16): 1356-1366, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37060545

RESUMEN

Importance: The effect of rationally defined nonpathogenic, nontoxigenic, commensal strains of Clostridia on prevention of Clostridioides difficile infection (CDI) is unknown. Objective: To determine the efficacy of VE303, a defined bacterial consortium of 8 strains of commensal Clostridia, in adults at high risk for CDI recurrence. The primary objective was to determine the recommended VE303 dosing for a phase 3 trial. Design, Setting, and Participants: Phase 2, randomized, double-blind, placebo-controlled, dose-ranging study conducted from February 2019 to September 2021 at 27 sites in the US and Canada. The study included 79 participants aged 18 years or older who were diagnosed with laboratory-confirmed CDI with 1 or more prior CDI episodes in the last 6 months and those with primary CDI at high risk for recurrence (defined as aged ≥75 years or ≥65 years with ≥1 risk factors: creatinine clearance <60 mL/min/1.73 m2, proton pump inhibitor use, remote [>6 months earlier] CDI history). Interventions: Participants were randomly assigned to high-dose VE303 (8.0 × 109 colony-forming units [CFUs]) (n = 30), low-dose VE303 (1.6 × 109 CFUs) (n = 27), or placebo capsules (n = 22) orally once daily for 14 days. Main Outcomes and Measures: The primary efficacy end point was the proportion of participants with CDI recurrence at 8 weeks using a combined clinical and laboratory definition. The primary efficacy end point was analyzed in 3 prespecified analyses, using successively broader definitions for an on-study CDI recurrence: (1) diarrhea consistent with CDI plus a toxin-positive stool sample; (2) diarrhea consistent with CDI plus a toxin-positive, polymerase chain reaction-positive, or toxigenic culture-positive stool sample; and (3) diarrhea consistent with CDI plus laboratory confirmation or (in the absence of a stool sample) treatment with a CDI-targeted antibiotic. Results: Baseline characteristics were similar across the high-dose VE303 (n = 29; 1 additional participant excluded from efficacy analysis), low-dose VE303 (n = 27), and placebo (n = 22) groups. The participants' median age was 63.5 years (range, 24-96); 70.5% were female; and 1.3% were Asian, 1.3% Black, 2.6% Hispanic, and 96.2% White. CDI recurrence rates through week 8 (using the efficacy analysis 3 definition) were 13.8% (4/29) for high-dose VE303, 37.0% (10/27) for low-dose VE303, and 45.5% (10/22) for placebo (P = .006, high-dose VE303 vs placebo). Conclusions and Relevance: Among adults with laboratory-confirmed CDI with 1 or more prior CDI episodes in the last 6 months and those with primary CDI at high risk for recurrence, high-dose VE303 prevented recurrent CDI compared with placebo. A larger, phase 3 study is needed to confirm these findings. Trial Registration: ClinicalTrials.gov Identifier: NCT03788434.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium , Probióticos , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Antibacterianos/administración & dosificación , Antibacterianos/uso terapéutico , Infecciones por Clostridium/complicaciones , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/prevención & control , Infecciones por Clostridium/terapia , Diarrea/etiología , Diarrea/microbiología , Diarrea/prevención & control , Diarrea/terapia , Heces/química , Heces/microbiología , Microbioma Gastrointestinal , Probióticos/administración & dosificación , Probióticos/uso terapéutico , Recurrencia , Reinfección/prevención & control , Simbiosis , Resultado del Tratamiento , Método Doble Ciego , Toxinas Bacterianas/análisis , Adulto Joven , Anciano , Anciano de 80 o más Años
3.
Nature ; 609(7927): 582-589, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36071157

RESUMEN

Increased levels of proteases, such as trypsin, in the distal intestine have been implicated in intestinal pathological conditions1-3. However, the players and mechanisms that underlie protease regulation in the intestinal lumen have remained unclear. Here we show that Paraprevotella strains isolated from the faecal microbiome of healthy human donors are potent trypsin-degrading commensals. Mechanistically, Paraprevotella recruit trypsin to the bacterial surface through type IX secretion system-dependent polysaccharide-anchoring proteins to promote trypsin autolysis. Paraprevotella colonization protects IgA from trypsin degradation and enhances the effectiveness of oral vaccines against Citrobacter rodentium. Moreover, Paraprevotella colonization inhibits lethal infection with murine hepatitis virus-2, a mouse coronavirus that is dependent on trypsin and trypsin-like proteases for entry into host cells4,5. Consistently, carriage of putative genes involved in trypsin degradation in the gut microbiome was associated with reduced severity of diarrhoea in patients with SARS-CoV-2 infection. Thus, trypsin-degrading commensal colonization may contribute to the maintenance of intestinal homeostasis and protection from pathogen infection.


Asunto(s)
Microbioma Gastrointestinal , Intestino Grueso , Simbiosis , Tripsina , Administración Oral , Animales , Sistemas de Secreción Bacterianos , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Bacteroidetes/aislamiento & purificación , Bacteroidetes/metabolismo , COVID-19/complicaciones , Citrobacter rodentium/inmunología , Diarrea/complicaciones , Heces/microbiología , Microbioma Gastrointestinal/genética , Humanos , Inmunoglobulina A/metabolismo , Intestino Grueso/metabolismo , Intestino Grueso/microbiología , Ratones , Virus de la Hepatitis Murina/metabolismo , Virus de la Hepatitis Murina/patogenicidad , Proteolisis , SARS-CoV-2/patogenicidad , Tripsina/metabolismo , Internalización del Virus
4.
Cell Host Microbe ; 30(4): 583-598.e8, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35421353

RESUMEN

Manipulation of the gut microbiota via fecal microbiota transplantation (FMT) has shown clinical promise in diseases such as recurrent Clostridioides difficile infection (rCDI). However, the variable nature of this approach makes it challenging to describe the relationship between fecal strain colonization, corresponding microbiota changes, and clinical efficacy. Live biotherapeutic products (LBPs) consisting of defined consortia of clonal bacterial isolates have been proposed as an alternative therapeutic class because of their promising preclinical results and safety profile. We describe VE303, an LBP comprising 8 commensal Clostridia strains under development for rCDI, and its early clinical development in healthy volunteers (HVs). In a phase 1a/b study in HVs, VE303 is determined to be safe and well-tolerated at all doses tested. VE303 strains optimally colonize HVs if dosed over multiple days after vancomycin pretreatment. VE303 promotes the establishment of a microbiota community known to provide colonization resistance.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium , Microbiota , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/terapia , Trasplante de Microbiota Fecal/métodos , Voluntarios Sanos , Humanos
5.
Gastroenterology ; 161(4): 1218-1228.e5, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34126062

RESUMEN

BACKGROUND & AIMS: Patients with multiple recurrent Clostridioides difficile infection (rCDI) have a disturbed gut microbiota that can be restored by fecal microbiota transplantation (FMT). Despite extensive screening, healthy feces donors may carry bacteria in their intestinal tract that could have long-term health effects, such as potentially procarcinogenic polyketide synthase-positive (pks+) Escherichia coli. Here, we aim to determine whether the pks abundance and persistence of pks+E coli is influenced by pks status of the donor feces. METHODS: In a cohort of 49 patients with rCDI treated with FMT and matching donor samples-the largest cohort of its kind, to our knowledge-we retrospectively screened fecal metagenomes for pks+E coli and compared the presence of pks in patients before and after treatment and to their respective donors. RESULTS: The pks island was more prevalent (P = .026) and abundant (P < .001) in patients with rCDI (pre-FMT, 27 of 49 [55%]; median, 0.46 reads per kilobase per million [RPKM] pks) than in healthy donors (3 of 8 donors [37.5%], 11 of 38 samples [29%]; median, 0.01 RPKM pks). The pks status of patients post-FMT depended on the pks status of the donor suspension with which the patient was treated (P = .046). Particularly, persistence (8 of 9 cases) or clearance (13 of 18) of pks+E coli in pks+ patients was correlated to pks in the donor (P = .004). CONCLUSIONS: We conclude that FMT contributes to pks+E coli persistence or eradication in patients with rCDI but that donor-to-patient transmission of pks+E coli is unlikely.


Asunto(s)
Clostridioides difficile/patogenicidad , Infecciones por Clostridium/terapia , Escherichia coli/crecimiento & desarrollo , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Adulto , Anciano , Anciano de 80 o más Años , Infecciones por Clostridium/diagnóstico , Infecciones por Clostridium/microbiología , Disbiosis , Escherichia coli/enzimología , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Trasplante de Microbiota Fecal/efectos adversos , Femenino , Humanos , Masculino , Metagenoma , Metagenómica , Persona de Mediana Edad , Sintasas Poliquetidas/genética , Sintasas Poliquetidas/metabolismo , Reinfección , Estudios Retrospectivos , Factores de Tiempo , Resultado del Tratamiento
7.
Nature ; 565(7741): 600-605, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30675064

RESUMEN

There is a growing appreciation for the importance of the gut microbiota as a therapeutic target in various diseases. However, there are only a handful of known commensal strains that can potentially be used to manipulate host physiological functions. Here we isolate a consortium of 11 bacterial strains from healthy human donor faeces that is capable of robustly inducing interferon-γ-producing CD8 T cells in the intestine. These 11 strains act together to mediate the induction without causing inflammation in a manner that is dependent on CD103+ dendritic cells and major histocompatibility (MHC) class Ia molecules. Colonization of mice with the 11-strain mixture enhances both host resistance against Listeria monocytogenes infection and the therapeutic efficacy of immune checkpoint inhibitors in syngeneic tumour models. The 11 strains primarily represent rare, low-abundance components of the human microbiome, and thus have great potential as broadly effective biotherapeutics.


Asunto(s)
Adenocarcinoma/inmunología , Adenocarcinoma/terapia , Bacterias/clasificación , Linfocitos T CD8-positivos/inmunología , Microbioma Gastrointestinal/inmunología , Listeriosis/prevención & control , Simbiosis/inmunología , Adenocarcinoma/patología , Animales , Antígenos CD/metabolismo , Bacterias/inmunología , Bacterias/aislamiento & purificación , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Células Dendríticas/inmunología , Heces/microbiología , Femenino , Voluntarios Sanos , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Cadenas alfa de Integrinas/metabolismo , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/microbiología , Masculino , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Elife ; 72018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29664397

RESUMEN

Manipulation of the gut microbiota holds great promise for the treatment of diseases. However, a major challenge is the identification of therapeutically potent microbial consortia that colonize the host effectively while maximizing immunologic outcome. Here, we propose a novel workflow to select optimal immune-inducing consortia from microbiome compositicon and immune effectors measurements. Using published and newly generated microbial and regulatory T-cell (Treg) data from germ-free mice, we estimate the contributions of twelve Clostridia strains with known immune-modulating effect to Treg induction. Combining this with a longitudinal data-constrained ecological model, we predict the ability of every attainable and ecologically stable subconsortium in promoting Treg activation and rank them by the Treg Induction Score (TrIS). Experimental validation of selected consortia indicates a strong and statistically significant correlation between predicted TrIS and measured Treg. We argue that computational indexes, such as the TrIS, are valuable tools for the systematic selection of immune-modulating bacteriotherapeutics.


Asunto(s)
Firmicutes/inmunología , Interacciones Microbiota-Huesped , Inmunidad Celular , Consorcios Microbianos , Linfocitos T Reguladores/inmunología , Animales , Simulación por Computador , Activación de Linfocitos , Ratones
9.
Cell Host Microbe ; 19(3): 311-22, 2016 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-26962942

RESUMEN

Human immunodeficiency virus (HIV) infection is associated with increased intestinal translocation of microbial products and enteropathy as well as alterations in gut bacterial communities. However, whether the enteric virome contributes to this infection and resulting immunodeficiency remains unknown. We characterized the enteric virome and bacterial microbiome in a cohort of Ugandan patients, including HIV-uninfected or HIV-infected subjects and those either treated with anti-retroviral therapy (ART) or untreated. Low peripheral CD4 T cell counts were associated with an expansion of enteric adenovirus sequences and this increase was independent of ART treatment. Additionally, the enteric bacterial microbiome of patients with lower CD4 T counts exhibited reduced phylogenetic diversity and richness with specific bacteria showing differential abundance, including increases in Enterobacteriaceae, which have been associated with inflammation. Thus, immunodeficiency in progressive HIV infection is associated with alterations in the enteric virome and bacterial microbiome, which may contribute to AIDS-associated enteropathy and disease progression.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/microbiología , Síndrome de Inmunodeficiencia Adquirida/virología , Bacterias/aislamiento & purificación , Microbioma Gastrointestinal , Microbiota , Virus/aislamiento & purificación , Síndrome de Inmunodeficiencia Adquirida/complicaciones , Síndrome de Inmunodeficiencia Adquirida/inmunología , Antirretrovirales/uso terapéutico , Bacterias/clasificación , Bacterias/genética , Linfocitos T CD4-Positivos/inmunología , Variación Genética , Enteropatía por VIH/etiología , Voluntarios Sanos , Humanos , Filogenia , Uganda , Virus/clasificación , Virus/genética
10.
Cell ; 160(3): 447-60, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25619688

RESUMEN

Decreases in the diversity of enteric bacterial populations are observed in patients with Crohn's disease (CD) and ulcerative colitis (UC). Less is known about the virome in these diseases. We show that the enteric virome is abnormal in CD and UC patients. In-depth analysis of preparations enriched for free virions in the intestine revealed that CD and UC were associated with a significant expansion of Caudovirales bacteriophages. The viromes of CD and UC patients were disease and cohort specific. Importantly, it did not appear that expansion and diversification of the enteric virome was secondary to changes in bacterial populations. These data support a model in which changes in the virome may contribute to intestinal inflammation and bacterial dysbiosis. We conclude that the virome is a candidate for contributing to, or being a biomarker for, human inflammatory bowel disease and speculate that the enteric virome may play a role in other diseases.


Asunto(s)
Caudovirales/aislamiento & purificación , Colitis Ulcerosa/virología , Enfermedad de Crohn/virología , Disbiosis/virología , Microviridae/aislamiento & purificación , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Estudios de Casos y Controles , Caudovirales/genética , Estudios de Cohortes , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Colitis Ulcerosa/terapia , Enfermedad de Crohn/microbiología , Enfermedad de Crohn/patología , Enfermedad de Crohn/terapia , Disbiosis/microbiología , Disbiosis/patología , Disbiosis/terapia , Heces/microbiología , Heces/virología , Humanos , Metagenoma , Microviridae/genética
11.
Science ; 347(6219): 269-73, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25431489

RESUMEN

Norovirus gastroenteritis is a major public health burden worldwide. Although fecal shedding is important for transmission of enteric viruses, little is known about the immune factors that restrict persistent enteric infection. We report here that although the cytokines interferon-α (IFN-α) and IFN-ß prevented the systemic spread of murine norovirus (MNoV), only IFN-λ controlled persistent enteric infection. Infection-dependent induction of IFN-λ was governed by the MNoV capsid protein and correlated with diminished enteric persistence. Treatment of established infection with IFN-λ cured mice in a manner requiring nonhematopoietic cell expression of the IFN-λ receptor, Ifnlr1, and independent of adaptive immunity. These results suggest the therapeutic potential of IFN-λ for curing virus infections in the gastrointestinal tract.


Asunto(s)
Infecciones por Caliciviridae/tratamiento farmacológico , Infecciones por Caliciviridae/inmunología , Citocinas/inmunología , Citocinas/uso terapéutico , Gastroenteritis/inmunología , Norovirus/inmunología , Norovirus/fisiología , Inmunidad Adaptativa , Animales , Infecciones por Caliciviridae/virología , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo , Células Cultivadas , Citocinas/biosíntesis , Heces/virología , Gastroenteritis/tratamiento farmacológico , Gastroenteritis/virología , Inmunidad Innata , Interferón-alfa/biosíntesis , Interferón-alfa/inmunología , Interferón beta/biosíntesis , Interferón beta/inmunología , Ratones , Ratones Endogámicos C57BL , Replicación Viral , Esparcimiento de Virus
12.
Proc Natl Acad Sci U S A ; 111(21): 7741-6, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24821797

RESUMEN

A coding polymorphism (Thr300Ala) in the essential autophagy gene, autophagy related 16-like 1 (ATG16L1), confers increased risk for the development of Crohn disease, although the mechanisms by which single disease-associated polymorphisms contribute to pathogenesis have been difficult to dissect given that environmental factors likely influence disease initiation in these patients. Here we introduce a knock-in mouse model expressing the Atg16L1 T300A variant. Consistent with the human polymorphism, T300A knock-in mice do not develop spontaneous intestinal inflammation, but exhibit morphological defects in Paneth and goblet cells. Selective autophagy is reduced in multiple cell types from T300A knock-in mice compared with WT mice. The T300A polymorphism significantly increases caspase 3- and caspase 7-mediated cleavage of Atg16L1, resulting in lower levels of full-length Atg16Ll T300A protein. Moreover, Atg16L1 T300A is associated with decreased antibacterial autophagy and increased IL-1ß production in primary cells and in vivo. Quantitative proteomics for protein interactors of ATG16L1 identified previously unknown nonoverlapping sets of proteins involved in ATG16L1-dependent antibacterial autophagy or IL-1ß production. These findings demonstrate how the T300A polymorphism leads to cell type- and pathway-specific disruptions of selective autophagy and suggest a mechanism by which this polymorphism contributes to disease.


Asunto(s)
Proteínas Portadoras/genética , Enfermedad de Crohn/inmunología , Células de Paneth/patología , Polimorfismo de Nucleótido Simple/genética , Infecciones por Salmonella/inmunología , Animales , Autofagia/genética , Proteínas Relacionadas con la Autofagia , Western Blotting , Cromatografía Liquida , Enfermedad de Crohn/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnicas de Sustitución del Gen , Células Caliciformes/patología , Ratones , Proteómica , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Masas en Tándem
13.
Gastroenterology ; 146(6): 1459-69, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24508599

RESUMEN

Advanced sequencing techniques have shown that bacteria are not the only complex and important microbes in the human intestine. Nonbacterial organisms, particularly the virome and the mycobiome, are important regulators of intestinal immunity and inflammation. The virome is mucosal and systemic; it can alter the host response to bacteria and interact with host genes and bacteria to contribute to disease pathogenesis. The human mycobiome is also complex and can contribute to intestinal inflammation. We review what has recently been learned about the nonbacterial and nonarchaeal microbes in the gastrointestinal tract, discussing their potential effects on health and disease and analytical approaches for their study. Studies of associations between the microbiome and intestinal pathology should incorporate kingdom-agnostic approaches if we are to fully understand intestinal health and disease.


Asunto(s)
Hongos/genética , Intestinos/microbiología , Intestinos/virología , Metagenoma , Metagenómica , Microbiota , Virus/genética , Animales , Susceptibilidad a Enfermedades , Hongos/clasificación , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/virología , Regulación Fúngica de la Expresión Génica , Regulación Viral de la Expresión Génica , Humanos , Metagenómica/métodos , Virus/clasificación
14.
J Virol ; 85(23): 12216-26, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21917951

RESUMEN

HIV-1-infected cells are partially resistant to anti-HIV cytotoxic T lymphocytes (CTLs) due to the effects of the HIV Nef protein on antigen presentation by major histocompatibility complex class I (MHC-I), and evidence has been accumulating that this function of Nef is important in vivo. HIV Nef disrupts the normal expression of MHC-I by stabilizing a protein-protein interaction between the clathrin adaptor protein AP-1 and the MHC-I cytoplasmic tail. There is also evidence that Nef activates a phosphatidylinositol 3 kinase (PI3K)-dependent GTPase, ADP ribosylation factor 6 (ARF-6), to stimulate MHC-I internalization. However, the relative importance of these two pathways is unclear. Here we report that a GTPase required for AP-1 activity (ARF-1) was needed for Nef to disrupt MHC-I surface levels, whereas no significant requirement for ARF-6 was observed in Nef-expressing T cell lines and in HIV-infected primary T cells. An ARF-1 inhibitor blocked the ability of Nef to recruit AP-1 to the MHC-I cytoplasmic tail, and a dominant active ARF-1 mutant stabilized the Nef-MHC-I-AP-1 complex. These data support a model in which Nef and ARF-1 stabilize an interaction between MHC-I and AP-1 to disrupt the presentation of HIV-1 epitopes to CTLs.


Asunto(s)
Factor 1 de Ribosilacion-ADP/metabolismo , Factores de Ribosilacion-ADP/metabolismo , Infecciones por VIH/virología , Antígeno HLA-A2/metabolismo , Linfocitos T/metabolismo , Linfocitos T/virología , Factor de Transcripción AP-1/metabolismo , Factor 1 de Ribosilacion-ADP/antagonistas & inhibidores , Factor 1 de Ribosilacion-ADP/genética , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/antagonistas & inhibidores , Factores de Ribosilacion-ADP/genética , Presentación de Antígeno , Western Blotting , Células Cultivadas , Citoplasma/metabolismo , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Infecciones por VIH/genética , Infecciones por VIH/inmunología , VIH-1/genética , VIH-1/patogenicidad , Antígeno HLA-A2/genética , Humanos , Inmunoprecipitación , Unión Proteica , Transporte de Proteínas , Linfocitos T/inmunología , Factor de Transcripción AP-1/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
15.
Nat Immunol ; 12(10): 975-83, 2011 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-21874023

RESUMEN

APOBEC3G (A3G) is an intrinsic antiviral factor that inhibits the replication of human immunodeficiency virus (HIV) by deaminating cytidine residues to uridine. This causes guanosine-to-adenosine hypermutation in the opposite strand and results in inactivation of the virus. HIV counteracts A3G through the activity of viral infectivity factor (Vif), which promotes degradation of A3G. We report that viral protein R (Vpr), which interacts with a uracil glycosylase, also counteracted A3G by diminishing the incorporation of uridine. However, this process resulted in activation of the DNA-damage-response pathway and the expression of natural killer (NK) cell-activating ligands. Our results show that pathogen-induced deamination of cytidine and the DNA-damage response to virus-mediated repair of the incorporation of uridine enhance the recognition of HIV-infected cells by NK cells.


Asunto(s)
Citidina Desaminasa/fisiología , VIH/inmunología , Células Asesinas Naturales/inmunología , Linfocitos T/virología , Desaminasa APOBEC-3G , Células Cultivadas , Citotoxicidad Inmunológica , Daño del ADN , Productos del Gen vpr/fisiología , Humanos , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Uridina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...