Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 300(1): 105525, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38043800

RESUMEN

The innate antiviral response to RNA viruses is initiated by sensing of viral RNAs by RIG-I-like receptors and elicits type I interferon (IFN) production, which stimulates the expression of IFN-stimulated genes that orchestrate the antiviral response to prevent systemic infection. Negative regulation of type I IFN and its master regulator, transcription factor IRF7, is essential to maintain immune homeostasis. We previously demonstrated that AIP (aryl hydrocarbon receptor interacting protein) functions as a negative regulator of the innate antiviral immune response by binding to and sequestering IRF7 in the cytoplasm, thereby preventing IRF7 transcriptional activation and type I IFN production. However, it remains unknown how AIP inhibition of IRF7 is regulated. We show here that the kinase TBK1 phosphorylates AIP and Thr40 serves as the primary target for TBK1 phosphorylation. AIP Thr40 plays critical roles in regulating AIP stability and mediating its interaction with IRF7. The AIP phosphomimetic T40E exhibited increased proteasomal degradation and enhanced interaction with IRF7 compared with wildtype AIP. AIP T40E also blocked IRF7 nuclear translocation, which resulted in reduced type I IFN production and increased viral replication. In sharp contrast, AIP phosphonull mutant T40A had impaired IRF7 binding, and stable expression of AIP T40A in AIP-deficient mouse embryonic fibroblasts elicited a heightened type I IFN response and diminished RNA virus replication. Taken together, these results demonstrate that TBK1-mediated phosphorylation of AIP at Thr40 functions as a molecular switch that enables AIP to interact with and inhibit IRF7, thus preventing overactivation of type I IFN genes by IRF7.


Asunto(s)
Inmunidad Innata , Factor 7 Regulador del Interferón , Interferón Tipo I , Proteínas Serina-Treonina Quinasas , Infecciones por Virus ARN , Virus ARN , Receptores de Hidrocarburo de Aril , Animales , Ratones , Fibroblastos , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Virus ARN/inmunología , Infecciones por Virus ARN/inmunología , Humanos , Células HEK293
2.
Sci Transl Med ; 13(612): eabg3459, 2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34550727

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating disease of premature infants, whose pathogenesis remains incompletely understood, although activation of the Gram-negative bacterial receptor Toll-like receptor 4 (TLR4) on the intestinal epithelium plays a critical role. Patients with NEC typically display gastrointestinal dysmotility before systemic disease is manifest, suggesting that dysmotility could drive NEC development. Both intestinal motility and inflammation are governed by the enteric nervous system, a network of enteric neurons and glia. We hypothesized here that enteric glia loss in the premature intestine could lead to dysmotility, exaggerated TLR4 signaling, and NEC development. We found that intestinal motility is reduced early in NEC in mice, preceding the onset of intestinal inflammation, whereas pharmacologic restoration of intestinal motility reduced NEC severity. Ileal samples from mouse, piglet, and human NEC revealed enteric glia depletion, and glia-deficient mice (Plp1ΔDTR, Sox10ΔDTR, and BdnfΔDTR) showed increased NEC severity compared with wild-type mice. Mice lacking TLR4 on enteric glia (Sox10-Tlr4ko) did not show NEC-induced enteric glia depletion and were protected from NEC. Mechanistically, brain-derived neurotrophic factor (BDNF) from enteric glia restrained TLR4 signaling on the intestine to prevent NEC. BDNF was reduced in mouse and human NEC, and BDNF administration reduced both TLR4 signaling and NEC severity in enteric glia­deficient mice. Last, we identified an agent (J11) that enhanced enteric glial BDNF release, inhibited intestinal TLR4, restored motility, and prevented NEC in mice. Thus, enteric glia loss might contribute to NEC through intestinal dysmotility and increased TLR4 activation, suggesting enteric glia therapies for this disorder.


Asunto(s)
Enterocolitis Necrotizante , Receptor Toll-Like 4 , Humanos , Recién Nacido , Neuroglía , Receptor Toll-Like 4/genética , Animales , Ratones
3.
Nat Commun ; 12(1): 1042, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589625

RESUMEN

Necrotizing enterocolitis (NEC) is a disease of premature infants characterized by acute intestinal necrosis. Current dogma suggests that NEC develops in response to post-natal dietary and bacterial factors, and so a potential role for in utero factors in NEC remains unexplored. We now show that during pregnancy, administration of a diet rich in the aryl hydrocarbon receptor (AHR) ligand indole-3-carbinole (I3C), or of breast milk, activates AHR and prevents NEC in newborn mice by reducing Toll-like receptor 4 (TLR4) signaling in the newborn gut. Protection from NEC requires activation of AHR in the intestinal epithelium which is reduced in mouse and human NEC, and is independent of leukocyte activation. Finally, we identify an AHR ligand ("A18") that limits TLR4 signaling in mouse and human intestine, and prevents NEC in mice when administered during pregnancy. In summary, AHR signaling is critical in NEC development, and maternally-delivered, AHR-based therapies may alleviate NEC.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Enterocolitis Necrotizante/genética , Indoles/administración & dosificación , Leche Humana/fisiología , Receptores de Hidrocarburo de Aril/genética , Receptor Toll-Like 4/genética , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/inmunología , Dieta/métodos , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/prevención & control , Femenino , Regulación de la Expresión Génica , Humanos , Recién Nacido , Recien Nacido Prematuro , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ligandos , Exposición Materna , Ratones , Embarazo , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/inmunología , Transducción de Señal , Porcinos , Receptor Toll-Like 4/inmunología
4.
Sci Transl Med ; 13(575)2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33408187

RESUMEN

Necrotizing enterocolitis (NEC) causes acute intestinal necrosis in premature infants and is associated with severe neurological impairment. In NEC, Toll-like receptor 4 is activated in the intestinal epithelium, and NEC-associated brain injury is characterized by microglial activation and white matter loss through mechanisms that remain unclear. We now show that the brains of mice and humans with NEC contained CD4+ T lymphocytes that were required for the development of brain injury. Inhibition of T lymphocyte influx into the brains of neonatal mice with NEC reduced inflammation and prevented myelin loss. Adoptive intracerebroventricular delivery of gut T lymphocytes from mice with NEC into Rag1 -/- recipient mice lacking CD4+ T cells resulted in brain injury. Brain organoids derived from mice with or without NEC and from human neuronal progenitor cells revealed that IFN-γ release by CD4+ T lymphocytes induced microglial activation and myelin loss in the organoids. IFN-γ knockdown in CD4+ T cells derived from mice with NEC abrogated the induction of NEC-associated brain injury after adoptive transfer to naïve Rag1 -/- recipient mice. T cell receptor sequencing revealed that NEC mouse brain-derived T lymphocytes shared homology with gut T lymphocytes from NEC mice. Intraperitoneal injection of NEC gut-derived CD4+ T lymphocytes into naïve Rag1 -/- recipient mice induced brain injury, suggesting that gut-derived T lymphocytes could mediate neuroinflammation in NEC. These findings indicate that NEC-associated brain injury may be induced by gut-derived IFN-γ-releasing CD4+ T cells, suggesting that early management of intestinal inflammation in children with NEC could improve neurological outcomes.


Asunto(s)
Enterocolitis Necrotizante , Animales , Encéfalo , Inflamación , Mucosa Intestinal , Ratones , Linfocitos T Reguladores
6.
Pediatr Res ; 89(1): 91-101, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32221473

RESUMEN

BACKGROUND: Necrotizing enterocolitis (NEC) develops through exaggerated toll-like receptor 4 (TLR4) signaling in the intestinal epithelium. Breast milk is rich in non-digestible oligosaccharides and prevents NEC through unclear mechanisms. We now hypothesize that the human milk oligosaccharides 2'-fucosyllactose (2'-FL) and 6'-sialyllactose (6'-SL) can reduce NEC through inhibition of TLR4 signaling. METHODS: NEC was induced in newborn mice and premature piglets and infant formula was supplemented with 2'-FL, 6'-SL, or lactose. Intestinal tissue was obtained at surgical resection. HMO inhibition of TLR4 was assessed in IEC-6 enterocytes, mice, and human tissue explants and via in silico modeling. RESULTS: Supplementation of infant formula with either 2'-FL and/or 6'-SL, but not the parent sugar lactose, reduced NEC in mice and piglets via reduced apoptosis, inflammation, weight loss, and histological appearance. Mechanistically, both 2'-FL and 6'-SL, but not lactose, reduced TLR4-mediated nuclear factor kappa light-chain enhancer of activated B cells (NF-kB) inflammatory signaling in the mouse and human intestine. Strikingly, in silico modeling revealed 2'-FL and 6'-SL, but not lactose, to dock into the binding pocket of the TLR4-MD2 complex, explaining their ability to inhibit TLR4 signaling. CONCLUSIONS: 2'-FL and 6'-SL, but not lactose, prevent NEC in mice and piglet models and attenuate NEC inflammation in the human ileum, in part through TLR4 inhibition. IMPACT: Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants that occurs in the setting of bacterial colonization of the gut and administration of formula feeds and activation by the innate immune receptor toll-like receptor 4 (TLR4). Breast milk prevents NEC through unclear mechanisms. We now show that breast milk-enriched human milk oligosaccharides (HMOs) that are derived from lactose prevent NEC through inhibition of TLR4. The human milk oligosaccharides 2'-FL and 6'-SL, but not the backbone sugar lactose, prevent NEC in mice and piglets. 2'-FL and 6'-SL but not lactose inhibited TLR4 signaling in cultured enterocytes, in enteroids derived from mouse intestine, and in human intestinal explants obtained at the time of surgical resection for patients with NEC. In seeking the mechanisms involved, 2'-FL and 6'-SL but not lactose were found to directly bind to TLR4, explaining the inhibition and protection against NEC. These findings may impact clinical practice by suggesting that administration of HMOs could serve as a preventive strategy for premature infants at risk for NEC development.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Íleon/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Lactosa/análogos & derivados , Leche Humana/química , Receptor Toll-Like 4/antagonistas & inhibidores , Trisacáridos/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Humanos , Íleon/inmunología , Íleon/metabolismo , Íleon/patología , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Lactosa/aislamiento & purificación , Lactosa/farmacología , Ratones , Simulación del Acoplamiento Molecular , Transducción de Señal , Sus scrofa , Receptor Toll-Like 4/metabolismo , Trisacáridos/aislamiento & purificación , Pérdida de Peso/efectos de los fármacos
7.
Cell Mol Gastroenterol Hepatol ; 9(3): 403-423, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31756560

RESUMEN

BACKGROUND & AIMS: Necrotizing enterocolitis (NEC) is a devastating disease of premature infants characterized by Toll-like receptor 4 (TLR4)-dependent intestinal inflammation and enterocyte death. Given that necroptosis is a proinflammatory cell death process that is linked to bacterial signaling, we investigated its potential role in NEC, and the mechanisms involved. METHODS: Human and mouse NEC intestine were analyzed for necroptosis gene expression (ie, RIPK1, RIPK3, and MLKL), and protein activation (phosphorylated RIPK3). To evaluate a potential role for necroptosis in NEC, the effects of genetic (ie, Ripk3 knockout or Mlkl knockout) or pharmacologic (ie, Nec1s) inhibition of intestinal inflammation were assessed in a mouse NEC model, and a possible upstream role of TLR4 was assessed in Tlr4-deficient mice. The NEC-protective effects of human breast milk and its constituent milk oligosaccharides on necroptosis were assessed in a NEC-in-a-dish model, in which mouse intestinal organoids were cultured as either undifferentiated or differentiated epithelium in the presence of NEC bacteria and hypoxia. RESULTS: Necroptosis was activated in the intestines of human and mouse NEC in a TLR4-dependent manner, and was up-regulated specifically in differentiated epithelium of the immature ileum. Inhibition of necroptosis genetically and pharmacologically reduced intestinal-epithelial cell death and mucosal inflammation in experimental NEC, and ex vivo in the NEC-in-a-dish system. Strikingly, the addition of human breast milk, or the human milk oligosaccharide 2 fucosyllactose in the ex vivo system, reduced necroptosis and inflammation. CONCLUSIONS: Necroptosis is activated in the intestinal epithelium upon TLR4 signaling and is required for NEC development, and explains in part the protective effects of breast milk.


Asunto(s)
Enterocolitis Necrotizante/patología , Enterocitos/patología , Mucosa Intestinal/patología , Leche Humana/química , Necroptosis/inmunología , Animales , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/inmunología , Enterocitos/efectos de los fármacos , Enterocitos/inmunología , Femenino , Humanos , Recién Nacido , Mucosa Intestinal/efectos de los fármacos , Ratones , Ratones Noqueados , Necroptosis/efectos de los fármacos , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Trisacáridos/farmacología , Trisacáridos/uso terapéutico , Regulación hacia Arriba
8.
Sci Transl Med ; 10(471)2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30541786

RESUMEN

Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease of the premature infant. One of the most important long-term complications observed in children who survive NEC early in life is the development of profound neurological impairments. However, the pathways leading to NEC-associated neurological impairments remain unknown, thus limiting the development of prevention strategies. We have recently shown that NEC development is dependent on the expression of the lipopolysaccharide receptor Toll-like receptor 4 (TLR4) on the intestinal epithelium, whose activation by bacteria in the newborn gut leads to mucosal inflammation. Here, we hypothesized that damage-induced production of TLR4 endogenous ligands in the intestine might lead to activation of microglial cells in the brain and promote cognitive impairments. We identified a gut-brain signaling axis in an NEC mouse model in which activation of intestinal TLR4 signaling led to release of high-mobility group box 1 in the intestine that, in turn, promoted microglial activation in the brain and neurological dysfunction. We further demonstrated that an orally administered dendrimer-based nanotherapeutic approach to targeting activated microglia could prevent NEC-associated neurological dysfunction in neonatal mice. These findings shed light on the molecular pathways leading to the development of NEC-associated brain injury, provide a rationale for early removal of diseased intestine in NEC, and indicate the potential of targeted therapies that protect the developing brain in the treatment of NEC in early childhood.


Asunto(s)
Encéfalo/patología , Disfunción Cognitiva/etiología , Enterocolitis Necrotizante/complicaciones , Microglía/patología , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , Administración Oral , Animales , Animales Recién Nacidos , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Encéfalo/ultraestructura , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/patología , Dendrímeros/química , Proteína HMGB1/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Ratones Endogámicos C57BL , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Vaina de Mielina/ultraestructura , Receptor Toll-Like 4/metabolismo
9.
Shock ; 47(1): 22-32, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27488085

RESUMEN

Necrotizing enterocolitis (NEC) is the most devastating gastrointestinal disease of the premature infant. We have recently shown that NEC development occurs after an increase in proinflammatory CD4Th17 (Th17) cells and reduced anti-inflammatory forkhead box P3 regulatory T cells (Tregs) to the premature small intestine of mice and humans, which can be experimentally reversed in mice by administration of all-trans retinoic acid (ATRA). We have also shown that NEC is characterized by apoptosis of Lgr5-positive intestinal stem cells (ISCs-Lgr5 cells) within the crypts of Lieberkühn, which are subsequently essential for intestinal homeostasis. We now hypothesize that the normal lymphocyte balance within the lamina propria of the intestine can be achieved via administration of ATRA which restores mucosal integrity by preventing the loss of ISCs. Using both in vivo and in vitro strategies, we now demonstrate that Th17 recruitment and Treg depletion lead to increased apoptosis within ISC niches, significantly impairing proliferative capacity and mucosal healing. ATRA exerted its protective effects by preventing T cell imbalance, ultimately leading to the protection of the ISC pool preventing the development of NEC in mice. These findings raise the exciting possibility that dietary manipulations could prevent and treat NEC by modulating lymphocyte balance and the ISC pool within the newborn small intestine.


Asunto(s)
Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/prevención & control , Intestinos/citología , Linfocitos/citología , Linfocitos/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Tretinoina/uso terapéutico , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Citometría de Flujo , Inmunohistoquímica , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Madre/citología
10.
Br J Nutr ; 116(7): 1175-1187, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27609061

RESUMEN

Necrotising enterocolitis (NEC) is a common disease in premature infants characterised by intestinal ischaemia and necrosis. The only effective preventative strategy against NEC is the administration of breast milk, although the protective mechanisms remain unknown. We hypothesise that an abundant human milk oligosaccharide (HMO) in breast milk, 2'-fucosyllactose (2'FL), protects against NEC by enhancing intestinal mucosal blood flow, and we sought to determine the mechanisms underlying this protection. Administration of HMO-2'FL protected against NEC in neonatal wild-type mice, resulted in a decrease in pro-inflammatory markers and preserved the small intestinal mucosal architecture. These protective effects occurred via restoration of intestinal perfusion through up-regulation of the vasodilatory molecule endothelial nitric oxide synthase (eNOS), as administration of HMO-2'FL to eNOS-deficient mice or to mice that received eNOS inhibitors did not protect against NEC, and by 16S analysis HMO-2'FL affected the microbiota of the neonatal mouse gut, although these changes do not seem to be the primary mechanism of protection. Induction of eNOS by HMO-2'FL was also observed in cultured endothelial cells, providing a link between eNOS and HMO in the endothelium. These data demonstrate that HMO-2'FL protects against NEC in part through maintaining mesenteric perfusion via increased eNOS expression, and suggest that the 2'FL found in human milk may be mediating some of the protective benefits of breast milk in the clinical setting against NEC.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Enfermedades del Prematuro/fisiopatología , Leche Humana/química , Circulación Esplácnica/efectos de los fármacos , Trisacáridos/administración & dosificación , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/fisiopatología , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Recién Nacido , Mucosa Intestinal/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/efectos de los fármacos , Óxido Nítrico/análisis , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/fisiología
11.
J Immunol ; 197(3): 859-71, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27307558

RESUMEN

We seek to define the mechanisms leading to the development of lung disease in the setting of neonatal necrotizing enterocolitis (NEC), a life-threatening gastrointestinal disease of premature infants characterized by the sudden onset of intestinal necrosis. NEC development in mice requires activation of the LPS receptor TLR4 on the intestinal epithelium, through its effects on modulating epithelial injury and repair. Although NEC-associated lung injury is more severe than the lung injury that occurs in premature infants without NEC, the mechanisms leading to its development remain unknown. In this study, we now show that TLR4 expression in the lung gradually increases during postnatal development, and that mice and humans with NEC-associated lung inflammation express higher levels of pulmonary TLR4 than do age-matched controls. NEC in wild-type newborn mice resulted in significant pulmonary injury that was prevented by deletion of TLR4 from the pulmonary epithelium, indicating a role for pulmonary TLR4 in lung injury development. Mechanistically, intestinal epithelial TLR4 activation induced high-mobility group box 1 release from the intestine, which activated pulmonary epithelial TLR4, leading to the induction of the neutrophil recruiting CXCL5 and the influx of proinflammatory neutrophils to the lung. Strikingly, the aerosolized administration of a novel carbohydrate TLR4 inhibitor prevented CXCL5 upregulation and blocked NEC-induced lung injury in mice. These findings illustrate the critical role of pulmonary TLR4 in the development of NEC-associated lung injury, and they suggest that inhibition of this innate immune receptor in the neonatal lung may prevent this devastating complication of NEC.


Asunto(s)
Enterocolitis Necrotizante/complicaciones , Lesión Pulmonar/etiología , Mucosa Respiratoria/metabolismo , Receptor Toll-Like 4/biosíntesis , Animales , Animales Recién Nacidos , Quimiocina CXCL5/metabolismo , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Proteína HMGB1/metabolismo , Humanos , Inmunohistoquímica , Recién Nacido , Lesión Pulmonar/inmunología , Lesión Pulmonar/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Mucosa Respiratoria/inmunología
12.
J Biol Chem ; 290(23): 14729-39, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-25911105

RESUMEN

The transcription factor IRF7 (interferon regulatory factor 7) is a key regulator of type I interferon and plays essential roles in restricting virus infection and spread. IRF7 activation is tightly regulated to prevent excessive inflammation and autoimmunity; however, how IRF7 is suppressed by negative regulators remains poorly understood. Here, we have identified AIP (aryl hydrocarbon receptor interacting protein) as a new binding partner of IRF7. The interaction between AIP and IRF7 is enhanced upon virus infection, and AIP potently inhibits IRF7-induced type I IFN (IFNα/ß) production. Overexpression of AIP blocks virus-induced activation of IFN, whereas knockdown of AIP by siRNA potentiates virally activated IFN production. Consistently, AIP-deficient murine embryonic fibroblasts are highly resistant to virus infection because of increased production of IFNα/ß. AIP inhibits IRF7 function by antagonizing the nuclear localization of IRF7. Together, our study identifies AIP as a novel inhibitor of IRF7 and a negative regulator of innate antiviral signaling.


Asunto(s)
Factor 7 Regulador del Interferón/inmunología , Interferón-alfa/inmunología , Interferón beta/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Virosis/inmunología , Animales , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética , Regulación hacia Arriba , Virosis/genética
13.
J Virol ; 85(14): 6955-62, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21593151

RESUMEN

Human T cell leukemia virus type 1 (HTLV-1) inhibits host antiviral signaling pathways although the underlying mechanisms are unclear. Here we found that the HTLV-1 Tax oncoprotein induced the expression of SOCS1, an inhibitor of interferon signaling. Tax required NF-κB, but not CREB, to induce the expression of SOCS1 in T cells. Furthermore, Tax interacted with SOCS1 in both transfected cells and in HTLV-1-transformed cell lines. Although SOCS1 is normally a short-lived protein, in the presence of Tax, the stability of SOCS1 was greatly increased. Accordingly, Tax enhanced the replication of a heterologous virus, vesicular stomatitis virus (VSV), in a SOCS1-dependent manner. Surprisingly, Tax required SOCS1 to inhibit RIG-I-dependent antiviral signaling, but not the interferon-induced JAK/STAT pathway. Inhibition of SOCS1 by RNA-mediated interference in the HTLV-1-transformed cell line MT-2 resulted in increased IFN-ß expression accompanied by reduced HTLV-1 replication and p19(Gag) levels. Taken together, our results reveal that Tax inhibits antiviral signaling, in part, by hijacking an interferon regulatory protein.


Asunto(s)
Productos del Gen tax/fisiología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Inmunidad Innata/fisiología , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Proteínas Supresoras de la Señalización de Citocinas/biosíntesis , Secuencia de Bases , Línea Celular , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 Supresora de la Señalización de Citocinas , Técnicas del Sistema de Dos Híbridos , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA