Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int Immunol ; 31(10): 669-683, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31132297

RESUMEN

Previous studies have shown that inhibition of receptor-interacting serine/threonine kinase (RICK) (also known as RIP2) results in amelioration of experimental colitis. This role has largely been attributed to nucleotide-binding oligomerization domain 2 (NOD2) signaling since the latter is considered a major inducer of RICK activation. In this study, we explored the molecular mechanisms accounting for RICK-mediated inhibition of inflammatory bowel disease (IBD). In an initial series of studies focused on trinitrobenzene sulfonic acid (TNBS)-colitis and dextran sodium sulfate (DSS)-colitis we showed that down-regulation of intestinal RICK expression in NOD2-intact mice by intra-rectal administration of a plasmid expressing RICK-specific siRNA was accompanied by down-regulation of pro-inflammatory cytokine responses in the colon and protection of the mice from experimental colitis. Somewhat surprisingly, intra-rectal administration of RICK-siRNA also inhibited TNBS-colitis and DSS-colitis in NOD2-deficient and in NOD1/NOD2-double deficient mice. In complementary studies of humans with IBD we found that expression of RICK, cellular inhibitor of apoptosis protein 2 (cIAP2) and downstream signaling partners were markedly increased in inflamed tissue of IBD compared to controls without marked elevations of NOD1 or NOD2 expression. In addition, the increase in RICK expression correlated with disease activity and pro-inflammatory cytokine responses. These studies thus suggest that NOD1- or NOD2-independenent activation of RICK plays a major role in both murine experimental colitis and human IBD.


Asunto(s)
Inflamación/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Animales , Humanos , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/inmunología , Transducción de Señal/inmunología
2.
Immunity ; 33(3): 313-25, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20870174

RESUMEN

The molecular mechanisms underlying retinoic acid (RA) augmentation of T cell receptor (TCR) and transforming growth factor-ß (TGF-ß)-induced Foxp3 transcription and inhibition of the latter by cytokines such as IL-27 were here shown to be related processes involving modifications of baseline (TGF-ß-induced) phosphorylated Smad3 (pSmad3) binding to a conserved enhancer region (enhancer I). RA augmentation involved the binding of retinoic acid receptor (RAR) and retinoid X receptor (RXR) to a dominant site in enhancer I and a subordinate site in the promoter. This led to increased histone acetylation in the region of the Smad3 binding site and increased binding of pSmad3. Cytokine (IL-27) inhibition involved binding of pStat3 to a gene silencer in a second conserved enhancer region (enhancer II) downstream from enhancer I; this led to loss of pSmad3 binding to enhancer I. Thus, control of accessibility and binding of pSmad3 provides a common framework for positive and negative regulation of TGF-ß-induced Foxp3 transcription.


Asunto(s)
Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Proteína smad3/metabolismo , Animales , Sitios de Unión , Femenino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Antígenos de Linfocitos T/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Receptores X Retinoide/fisiología , Factor de Transcripción AP-1/fisiología , Transcripción Genética , Factor de Crecimiento Transformador beta/fisiología , Tretinoina/farmacología
3.
Immunity ; 30(6): 860-74, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19501001

RESUMEN

Missense mutations of the gene encoding NLRP3 are associated with autoinflammatory disorders characterized with excessive production of interleukin-1beta (IL-1beta). Here we analyzed the immune responses of gene-targeted mice carrying a mutation in the Nlrp3 gene equivalent to the human mutation associated with Muckle-Wells Syndrome. We found that antigen-presenting cells (APCs) from such mice produced massive amounts of IL-1beta upon stimulation with microbial stimuli in the absence of ATP. This was likely due to a diminished inflammasome activation threshold that allowed a response to the small amount of agonist. Moreover, the Nlrp3 gene-targeted mice exhibited skin inflammation characterized by neutrophil infiltration and a Th17 cytokine-dominant response, which originated from hematopoietic cells. The inflammation of Nlrp3 gene-targeted mice resulted from excess IL-1beta production from APCs, which augmented Th17 cell differentiation. These results demonstrate that the NLRP3 mutation leads to inflammasome hyperactivation and consequently Th17 cell-dominant immunopathology in autoinflammation.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Proteínas Portadoras/metabolismo , Inflamación/genética , Interleucina-1beta/inmunología , Macrófagos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/metabolismo , Animales , Células Presentadoras de Antígenos/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Diferenciación Celular/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Inflamación/inmunología , Interleucina-18/biosíntesis , Interleucina-18/inmunología , Interleucina-1beta/biosíntesis , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Mutación/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Piel/inmunología , Piel/metabolismo , Piel/patología , Linfocitos T Colaboradores-Inductores/metabolismo
4.
Immunol Rev ; 260(1): 249-60, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24942694

RESUMEN

The discovery that polymorphisms in the NOD2 (nucleotide-binding oligomerization domain containing 2) gene are associated with a greatly increased risk for the development of Crohn's disease has provided a means to achieve a deeper understanding of the dysregulation of mucosal immune responses to the commensal intestinal organisms that is thought to underlie this disease. NOD2 is a NOD-like receptor (NLR) family member that senses and responds to bacterial wall peptides; thus, the most widely held view of the relation of the NOD2 polymorphisms with Crohn's disease is that these polymorphisms lead to deficient immune responses to gut bacteria, and these, in turn, lead to quantitative or qualitative changes in the bacterial population in the gut lumen or lamina propria that cause inflammation at this site. Initially, this view was based mainly on the observation that defective NOD2 function can result in reduced α-defensin production by intestinal Paneth cells and that such impairment leads to loss of host defense against gut bacteria. In this review, we reconsider this possibility and marshal evidence that it is not in fact likely to be a prime element of Crohn's disease causation. More recently, evidence has been accumulating that the NOD2 dysfunction leads to Crohn's inflammation by inducing changes in the gut microbiome that influence immune effector or regulatory function. We review the strengths and weaknesses of this emerging hypothesis. Finally, we consider the possibility that NOD2 dysfunction can lead to inflammation because of a second and somewhat overlooked aspect of its function, that as an immunoregulator of innate immune responses. In particular, we review the body of evidence that NOD2 stimulation activates a cross-tolerance response that downregulates and thus prevents excessive TLR responses that cause Crohn's inflammation.


Asunto(s)
Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Predisposición Genética a la Enfermedad , Proteína Adaptadora de Señalización NOD2/genética , Polimorfismo Genético , Animales , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/microbiología , Defensinas/biosíntesis , Estudios de Asociación Genética , Humanos , Inmunidad Innata , Inmunomodulación , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Intestinos/inmunología , Intestinos/microbiología , Microbiota , Proteína Adaptadora de Señalización NOD2/deficiencia , Proteína Adaptadora de Señalización NOD2/metabolismo , Células de Paneth/metabolismo
5.
Nat Rev Immunol ; 6(1): 9-20, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16493424

RESUMEN

The NOD (nucleotide-binding oligomerization domain) proteins NOD1 and NOD2 have important roles in innate immunity as sensors of microbial components derived from bacterial peptidoglycan. The importance of these molecules is underscored by the fact that mutations in the gene that encodes NOD2 occur in a subpopulation of patients with Crohn's disease, and NOD1 has also been shown to participate in host defence against infection with Helicobacter pylori. Here, we focus on the molecular interactions between these NOD proteins and other intracellular molecules to elucidate the mechanisms by which NOD1 and NOD2 contribute to the maintenance of mucosal homeostasis and the induction of mucosal inflammation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Humanos , Inmunidad Mucosa/genética , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Adaptadora de Señalización NOD1 , Proteína Adaptadora de Señalización NOD2 , Transducción de Señal/genética
6.
J Immunol ; 192(8): 3969-80, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24634488

RESUMEN

Spontaneous amelioration of inflammation (often accompanied by fibrosis) is a well-known, but poorly understood, outcome of many chronic inflammatory processes. We studied this phenomenon in a chronic trinitrobenzene sulfonic acid-induced colitis model, an experimental colitis in mice that we showed to ultimately undergo spontaneous resolution, despite continued trinitrobenzene sulfonic acid stimulation. Analysis of the mechanism of this resolution revealed that it was critically dependent on IL-13 activation of STAT6, followed by phosphorylation (inactivation) of glycogen synthase kinase-3ß, at least in part via STAT6 induction of p38 MAPK. Such glycogen synthase kinase-3ß inactivation causes changes in CREB and p65 DNA-binding activity that favors decreased proinflammatory IL-17 production and increased anti-inflammatory IL-10 production. Thus, in this case, IL-13 acts as a molecular switch that leads to resolution of inflammation.


Asunto(s)
Colitis/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Interleucina-13/metabolismo , Animales , Enfermedad Crónica , Colitis/inducido químicamente , Colitis/genética , Colitis/inmunología , Colitis/patología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Interleucina-13/antagonistas & inhibidores , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fosforilación , Transducción de Señal , Ácido Trinitrobencenosulfónico/efectos adversos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Nat Med ; 12(1): 99-106, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16327802

RESUMEN

Interleukin (IL)-13 is a major inducer of fibrosis in many chronic infectious and autoimmune diseases. In studies of the mechanisms underlying such induction, we found that IL-13 induces transforming growth factor (TGF)-beta(1) in macrophages through a two-stage process involving, first, the induction of a receptor formerly considered to function only as a decoy receptor, IL-13Ralpha(2). Such induction requires IL-13 (or IL-4) and tumor necrosis factor (TNF)-alpha. Second, it involves IL-13 signaling through IL-13Ralpha(2) to activate an AP-1 variant containing c-jun and Fra-2, which then activates the TGFB1 promoter. In vivo, we found that prevention of IL-13Ralpha(2) expression reduced production of TGF-beta(1) in oxazolone-induced colitis and that prevention of IL-13Ralpha(2) expression, Il13ra2 gene silencing or blockade of IL-13Ralpha(2) signaling led to marked downregulation of TGF-beta(1) production and collagen deposition in bleomycin-induced lung fibrosis. These data suggest that IL-13Ralpha(2) signaling during prolonged inflammation is an important therapeutic target for the prevention of TGF-beta(1)-mediated fibrosis.


Asunto(s)
Interleucina-13/fisiología , Macrófagos/metabolismo , Receptores de Interleucina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Bleomicina/farmacología , Western Blotting , Linaje de la Célula , Colitis/metabolismo , Colitis/patología , Colágeno/metabolismo , Citocinas/metabolismo , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Etanercept , Fibrosis/metabolismo , Citometría de Flujo , Silenciador del Gen , Vectores Genéticos , Humanos , Inmunoglobulina G/farmacología , Inflamación , Interleucina-13/metabolismo , Subunidad alfa1 del Receptor de Interleucina-13 , Luciferasas/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , FN-kappa B/metabolismo , Oxazolona/metabolismo , Oxazolona/farmacología , Regiones Promotoras Genéticas , ARN Interferente Pequeño/metabolismo , Receptores de Interleucina-13 , Receptores del Factor de Necrosis Tumoral , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo , Factor de Crecimiento Transformador beta1 , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
8.
Front Immunol ; 13: 988862, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36189261

RESUMEN

The studies described here provide an analysis of the pathogenesis of Blau syndrome and thereby the function of NOD2 as seen through the lens of its dysfunction resulting from Blau-associated NOD2 mutations in its nucleotide-binding domain (NBD). As such, this analysis also sheds light on the role of NOD2 risk polymorphisms in the LRR domain occurring in Crohn's disease. The main finding was that Blau NOD2 mutations precipitate a loss of canonical NOD2 signaling via RIPK2 and that this loss has two consequences: first, it results in defective NOD2 ligand (MDP)-mediated NF-κB activation and second, it disrupts NOD2-mediated cross-regulation whereby NOD2 downregulates concomitant innate (TLR) responses. Strong evidence is also presented favoring the view that NOD2-mediated cross-regulation is under mechanistic control by IRF4 and that failure to up-regulate this factor because of faulty NOD2 signaling is the proximal cause of defective cross-regulation and the latter's effect on Blau syndrome inflammation. Overall, these studies highlight the role of NOD2 as a regulatory factor and thus provide additional insight into its function in inflammatory disease. Mutations in the nucleotide binding domain of the CARD15 (NOD2) gene underlie the granulomatous inflammation characterizing Blau syndrome (BS). In studies probing the mechanism of this inflammation we show here that NOD2 plasmids expressing various Blau mutations in HEK293 cells result in reduced NOD2 activation of RIPK2 and correspondingly reduced NOD2 activation of NF-κB. These in vitro studies of NOD2 signaling were accompanied by in vivo studies showing that BS-NOD2 also exhibit defects in cross-regulation of innate responses underlying inflammation. Thus, whereas over-expressed intact NOD2 suppresses TNBS-colitis, over-expressed BS-NOD2 does not; in addition, whereas administration of NOD2 ligand (muramyl dipeptide, MDP) suppresses DSS-colitis in Wild Type (WT) mice it fails to do so in homozygous or heterozygous mice bearing a NOD2 Blau mutation. Similarly, mice bearing a Blau mutation exhibit enhanced anti-collagen antibody-induced arthritis. The basis of such cross-regulatory failure was revealed in studies showing that MDP-stimulated cells bearing BS-NOD2 exhibit a reduced capacity to signal via RIPK2 as well as a reduced capacity to up-regulate IRF4, a factor shown previously to mediate NOD2 suppression of NF-κB activation. Indeed, TLR-stimulated cells bearing a Blau mutation exhibited enhanced in vitro cytokine responses that are quieted by lentivirus transduction of IRF4. In addition, enhanced anti-collagen-induced joint inflammation in mice bearing a Blau mutation was accompanied by reduced IRF4 expression in inflamed joint tissue and IRF4 expression was reduced in MDP-stimulated cells from BS patients. Thus, inflammation characterizing Blau syndrome are caused, at least in part, by faulty canonical signaling and reduce IRF4-mediated cross-regulation.


Asunto(s)
Artritis , Colitis , Proteína Adaptadora de Señalización NOD2/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacología , Animales , Artritis/genética , Colitis/inducido químicamente , Citocinas/metabolismo , Células HEK293 , Humanos , Inflamación/genética , Ligandos , Ratones , Mutación , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD2/genética , Nucleótidos/metabolismo , Sarcoidosis , Sinovitis , Uveítis
9.
J Clin Invest ; 118(2): 545-59, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18188453

RESUMEN

The mechanisms underlying the susceptibility of individuals with caspase recruitment domain 15 (CARD15) mutations and corresponding abnormalities of nucleotide-binding oligomerization domain 2 (NOD2) protein to Crohn disease are still poorly understood. One possibility is based on previous studies showing that muramyl dipeptide (MDP) activation of NOD2 negatively regulates TLR2 responses and that absence of such regulation leads to heightened Th1 responses. We now report that administration of MDP protects mice from the development of experimental colitis by downregulating multiple TLR responses, not just TLR2. The basis of these in vivo findings was suggested by in vitro studies of DCs, in which we showed that prestimulation of cells with MDP reduces cytokine responses to multiple TLR ligands and this reduction is dependent on enhanced IFN regulatory factor 4 (IRF4) activity. Further studies of mouse models of colitis showed that this inhibitory role of IRF4 does in fact apply to MDP-mediated protection from colitis, since neither IRF4-deficient mice nor mice treated with siRNA specific for IRF4 were protected. These findings indicate that MDP activation of NOD2 regulates innate responses to intestinal microflora by downregulating multiple TLR responses and suggest that the absence of such regulation leads to increased susceptibility to Crohn disease.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Colitis/prevención & control , Enfermedad de Crohn/inmunología , Proteína Adaptadora de Señalización NOD2/metabolismo , Animales , Colitis/inducido químicamente , Colitis/inmunología , Enfermedad de Crohn/tratamiento farmacológico , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Inmunidad Innata/efectos de los fármacos , Factores Reguladores del Interferón/antagonistas & inhibidores , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Ligandos , Ratones , Ratones Mutantes , Proteína Adaptadora de Señalización NOD2/genética , ARN Interferente Pequeño/farmacología , Receptores Toll-Like/antagonistas & inhibidores , Ácido Trinitrobencenosulfónico/toxicidad
10.
J Exp Med ; 198(8): 1179-88, 2003 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-14557415

RESUMEN

Interleukin (IL)-10 and transforming growth factor (TGF)-beta1 are suppressor cytokines that frequently occur together during a regulatory T cell response. Here we used a one gene doxycycline (Dox)-inducible plasmid encoding TGF-beta1 to analyze this association and test its utility. In initial studies, we showed that intranasal administration of this plasmid (along with Dox) led to the appearance of TGF-beta1-producing cells (in spleen and lamina propria) and the almost concomitant appearance of IL-10-producing cells. Moreover, we showed that these cells exert Dox-regulated suppression of the T helper cell (Th)1-mediated inflammation in trinitrobenzene sulfonic acid colitis. In subsequent in vitro studies using retroviral TGF-beta1 expression, we established that IL-10 production by Th1 cells occurs after exposure to TGF-beta1 from either an endogenous or exogenous source. In addition, using a self-inactivating retrovirus luciferase reporter construct we showed that TGF-beta1 induces Smad4, which then binds to and activates the IL-10 promoter. Furthermore, intranasal TGF-beta1 plasmid administration ameliorates bleomycin-induced fibrosis in wild-type but not IL-10-deficient mice, strongly suggesting that the amelioration is IL-10 dependent and that IL-10 protects mice from TGF-beta1-mediated fibrosis. Taken together, these findings suggest that the induction of IL-10 by TGF-beta1 is not fortuitous, but instead fulfills important requirements of TGF-beta1 function after its secretion by regulatory T cells.


Asunto(s)
Proteínas de Unión al ADN/farmacología , Fibrosis/prevención & control , Interleucina-10/metabolismo , Linfocitos T/metabolismo , Transactivadores/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Bleomicina , Línea Celular , Proteínas de Unión al ADN/metabolismo , Electroporación , Fibrosis/inducido químicamente , Regulación de la Expresión Génica , Tolerancia Inmunológica , Macrófagos , Masculino , Ratones , Proteínas Smad , Células TH1/inmunología , Transactivadores/metabolismo , Transfección , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1
11.
Curr Opin Gastroenterol ; 26(4): 310-7, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20473158

RESUMEN

PURPOSE OF REVIEW: To encapsulate our current understanding of the proinflammatory cytokines responsible for the inflammation underlying Crohn's disease and the prospect of using this information to devise therapy for this condition based on inhibition of these cytokines. RECENT FINDINGS: Current research is shedding new light on the role of both T helper cell (Th)1 and Th17 responses in the pathogenesis of Crohn's disease. Initial studies conducted a decade ago highlighted the view that Crohn's disease inflammation is caused by an interleukin-12-driven Th1 response, which resulted in the generation of interferon-gamma, which then served as the main inflammatory mediator. In recent years, however, this view has been largely eclipsed by studies, conducted mainly in murine models, showing that a Th17 response is the main cause of Crohn's disease inflammation through the production of interleukin-17. Now, a somewhat more balanced view is emerging, which holds that interferon-gamma is still a major proinflammatory cytokine in Crohn's disease, although it may arise from both the Th1 and Th17-mediated responses at different phases of the inflammatory process. SUMMARY: The new findings continue to support the idea that anti-interleukin-12p40, an antibody that inhibits both the Th1 and Th17 response, is logically the most potent anticytokine for the treatment of Crohn's disease.


Asunto(s)
Enfermedad de Crohn/inmunología , Citocinas/inmunología , Animales , Citocinas/antagonistas & inhibidores , Citocinas/fisiología , Modelos Animales de Enfermedad , Humanos , Interleucina-12/antagonistas & inhibidores , Interleucina-12/inmunología , Interleucina-12/fisiología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/inmunología , Interleucina-17/fisiología , Interleucina-23/antagonistas & inhibidores , Interleucina-23/inmunología , Interleucina-23/fisiología , Interleucinas/antagonistas & inhibidores , Interleucinas/inmunología , Interleucinas/fisiología , Linfotoxina-alfa/inmunología , Células TH1/inmunología , Interleucina-22
12.
J Clin Invest ; 130(4): 1793-1807, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31895698

RESUMEN

Bruton tyrosine kinase (BTK) is present in a wide variety of cells and may thus have important non-B cell functions. Here, we explored the function of this kinase in macrophages with studies of its regulation of the NLR family, pyrin domain-containing 3 (NLRP3) inflammasome. We found that bone marrow-derived macrophages (BMDMs) from BTK-deficient mice or monocytes from patients with X-linked agammaglobulinemia (XLA) exhibited increased NLRP3 inflammasome activity; this was also the case for BMDMs exposed to low doses of BTK inhibitors such as ibrutinib and for monocytes from patients with chronic lymphocytic leukemia being treated with ibrutinib. In mechanistic studies, we found that BTK bound to NLRP3 during the priming phase of inflammasome activation and, in doing so, inhibited LPS- and nigericin-induced assembly of the NLRP3 inflammasome during the activation phase of inflammasome activation. This inhibitory effect was caused by BTK inhibition of protein phosphatase 2A-mediated (PP2A-mediated) dephosphorylation of Ser5 in the pyrin domain of NLRP3. Finally, we show that BTK-deficient mice were subject to severe experimental colitis and that such colitis was normalized by administration of anti-IL-ß or anakinra, an inhibitor of IL-1ß signaling. Together, these studies strongly suggest that BTK functions as a physiologic inhibitor of NLRP3 inflammasome activation and explain why patients with XLA are prone to develop Crohn's disease.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/deficiencia , Enfermedad de Crohn , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Adolescente , Adulto , Agammaglobulinemia Tirosina Quinasa/metabolismo , Agammaglobulinemia/enzimología , Agammaglobulinemia/genética , Agammaglobulinemia/patología , Anciano , Anciano de 80 o más Años , Animales , Niño , Enfermedad de Crohn/enzimología , Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/enzimología , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Inflamasomas/genética , Interleucina-1beta/genética , Masculino , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética
13.
Gastroenterology ; 135(6): 2003-13, 2013.e1-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18938165

RESUMEN

BACKGROUND & AIMS: Previous studies have shown that fibrosis developing in chronic experimental colitis is driven by interleukin (IL)-13 signaling via IL-13R alpha(2) and the production of transforming growth factor (TGF)-beta1. In the present study, we sought to determine the fibrogenic downstream events set in motion by such signaling. METHODS: Experimental colitis with late-onset intestinal fibrosis was induced by weekly intrarectal administration of trinitrobenzene sulfonic acid (TNBS) to BALB/c mice. Blockade of IL-13 signaling via IL-13R alpha(2) and TGF-beta1 signaling was achieved by the administration of small interfering RNA or decoy oligonucleotides that target promoter sequences of signaling components of these receptors. Effects of blockade were determined by enzyme-linked immunosorbent assay or Western blotting detecting specific key fibrogenic factors and by measurement of collagen production. RESULTS: Initially, we showed that abrogation of IL-13 activity via blockade of IL-13R alpha(2) and TGF-beta1 signaling results in severe inhibition of expression of colonic insulin-like growth factor (IGF)-I and early growth response gene (Egr)-1, factors known to initiate and sustain fibrosis. We then showed that Egr-1 was necessary early in the fibrotic process for caspase-mediated apoptosis of myofibroblasts and the production of urokinase plasminogen activator, a protein that enhances TGF-beta1 activation. Finally, we showed that IGF-I (together with TGF-beta1) acts later in the process to stimulate myofibroblasts to deposit collagen in the colon. CONCLUSIONS: These studies establish that IL-13 signaling via the IL-13R alpha(2) is a key initiation point for a complex fibrotic program in the colon consisting of TGF-beta1 activation, IGF-I and Egr-1 expression, myofibroblast apoptosis, and myofibroblast production of collagen.


Asunto(s)
Colitis/metabolismo , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Interleucina-13/metabolismo , Animales , Apoptosis , Western Blotting , Células Cultivadas , Colitis/inducido químicamente , Colitis/patología , Colon/metabolismo , Colon/patología , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/metabolismo , Fibrosis/patología , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Factor I del Crecimiento Similar a la Insulina/genética , Subunidad alfa2 del Receptor de Interleucina-13/antagonistas & inhibidores , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , ARN/genética , Transducción de Señal , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Ácido Trinitrobencenosulfónico/toxicidad , Dedos de Zinc
14.
Gastroenterology ; 135(5): 1612-1623.e5, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18765239

RESUMEN

BACKGROUND & AIMS: Previous studies have indicated that a defective epithelial barrier leads to inflammation of the underlying lamina propria. Nevertheless, it is likely that physiologic breaks in the barrier must occur for homeostatic regulatory T cells to develop. We determined the effect of agents that disrupt epithelial tight junctions (ethanol and AT1002, a Vibrio cholerae zonula occludens toxin hexapeptide) on regulatory T-cell induction and resistance to induction of colitis by trinitrobenzene sulfonic acid (TNBS). METHODS: The effects of ethanol and AT1002 on colon immune function were evaluated by their capacity to induce direct phenotypic or functional changes in effector and regulatory cell populations and their indirect effect on the development of TNBS-induced colitis. The basis of regulatory cell development was evaluated with in vitro studies of isolated dendritic cell populations. The role of innate immunity was evaluated by in vivo gene silencing studies utilizing Toll-like receptor (TLR)-2-specific small interfering RNA (siRNA). RESULTS: Both ethanol and AT1002 induced persistent latency-associated peptide-positive CD4(+) regulatory T cells that, as shown in adoptive transfer studies, render mice resistant to the induction of TNBS colitis. The development of these cells requires the presence of an intact microflora and the activity of CD11c(+) dendritic cells. Their induction is also influenced by innate immune factors operating through TLR-2, because attenuation of TLR-2 signaling by in vivo TLR-2 siRNA administration prevents their development. CONCLUSIONS: A mild and/or transient breach in epithelial barrier function leads to dominant regulatory T-cell responses that protect the mucosa from inflammation.


Asunto(s)
Colitis/inmunología , Inmunidad Celular/fisiología , Mucosa Intestinal/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Antiinfecciosos Locales/farmacología , Western Blotting , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Colitis/metabolismo , Colitis/patología , Modelos Animales de Enfermedad , Etanol/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Oligopéptidos/farmacología , ARN Interferente Pequeño/genética , Receptor Toll-Like 2/efectos de los fármacos , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo
15.
J Clin Invest ; 115(11): 3057-71, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16239967

RESUMEN

The Th1 and Th2 T cell responses that underlie inflammatory bowel diseases (IBDs) are likely to depend on NF-kappaB transcriptional activity. We explored this possibility in studies in which we determined the capacity of NF-kappaB decoy oligodeoxynucleotides (decoy ODNs) to treat various murine models of IBD. In initial studies, we showed that i.r. (intrarectal) or i.p. administration of decoy ODNs encapsulated in a viral envelope prevented and treated a model of acute trinitrobenzene sulfonic acid-induced (TNBS-induced) colitis, as assessed by clinical course and effect on Th1 cytokine production. In further studies, we showed that NF-kappaB decoy ODNs were also an effective treatment of a model of chronic TNBS-colitis, inhibiting both the production of IL-23/IL-17 and the development of fibrosis that characterizes this model. Treatment of TNBS-induced inflammation by i.r. administration of NF-kappaB decoy ODNs did not inhibit NF-kappaB in extraintestinal organs and resulted in CD4+ T cell apoptosis, suggesting that such treatment is highly focused and durable. Finally, we showed that NF-kappaB decoy ODNs also prevented and treated oxazolone-colitis and thus affect a Th2-mediated inflammatory process. In each case, decoy administration led to inflammation-clearing effects, suggesting a therapeutic potency applicable to human IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inmunología , FN-kappa B/genética , Oligodesoxirribonucleótidos/farmacología , Células TH1/inmunología , Células Th2/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , ADN/metabolismo , Femenino , Fibrosis , Vectores Genéticos , Enfermedades Inflamatorias del Intestino/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Membrana Mucosa/citología , Membrana Mucosa/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/genética , Virus Sendai
16.
Gastroenterology ; 133(5): 1510-21, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17915219

RESUMEN

BACKGROUND & AIMS: Mutations in the CARD15 gene encoding NOD2 are susceptibility factors in Crohn's disease. We explored the mechanism of this susceptibility using mice that over express NOD2. METHODS: Cellular and molecular responses of mice bearing an NOD2 transgene or administered plasmids that express wild-type and mutated NOD2 constructs were examined. RESULTS: In initial studies, we showed that splenocytes from NOD2 transgenic mice as compared with littermate controls exhibit decreased interleukin (IL)-12p70 responses to peptidoglycan (PGN), a TLR2 ligand that contains muramyl dipeptide, but not other TLR ligands; in contrast, IL-12 responses to PAM(3)CSK(4), a TLR2 ligand that does not contain muramyl dipeptide, were normal. Similarly, transgenic mice as compared with controls exhibited greatly decreased IL-12p40 responses to intraperitoneal administration of PGN but not to lipopolysaccharide. In further studies, we showed using electrophoretic mobility shift assay that PGN-stimulated cells from transgenic mice exhibited decreased activation of nuclear factor kappaB. Finally, in a series of studies on the effect of the NOD2 on susceptibility to induced colitis, we found that (1) transgenic mice were highly resistant to induction of PGN colitis and partially resistant to induction of trinitrobenzene sulfonic acid (TNBS) colitis and (2) mice administered a plasmid expressing a wild-type NOD2 gene were completely resistant to TNBS colitis whereas mice administered a plasmid expressing an NOD2 gene with the Crohn's disease frameshift mutation were only slightly resistant to TNBS colitis. CONCLUSIONS: These data offer new evidence that NOD2 mutations contribute to inflammatory bowel disease by causing excessive TLR2 cytokine responses.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/farmacología , Adyuvantes Inmunológicos/farmacología , Colitis/genética , Regulación hacia Abajo/efectos de los fármacos , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/fisiología , Receptor Toll-Like 2/genética , Animales , Células Cultivadas , Colitis/inducido químicamente , Colitis/inmunología , Mutación del Sistema de Lectura/genética , Inmunidad Innata/efectos de los fármacos , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/citología , Monocitos/metabolismo , FN-kappa B/metabolismo , Peptidoglicano , Plásmidos/genética , Bazo/citología , Bazo/metabolismo , Receptor Toll-Like 2/metabolismo , Ácido Trinitrobencenosulfónico
17.
Front Immunol ; 9: 2566, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30455704

RESUMEN

It is logical to assume that a major pro-inflammatory mechanism, i.e., the NLRP3 inflammasome would play a prominent role in the pathogenesis of the Inflammatory Bowel Disease (IBD) in humans. However, while both studies of murine models of gut disease and patients provide data that the main cytokine product generated by this inflammasome, IL-1ß, does in fact contribute to inflammation in IBD, there is no evidence that IL-1ß plays a decisive or prominent role in "ordinary" patients with IBD (Crohn's disease). On the other hand, there are several definable point mutations that result in over-active NLRP3 inflammasome activity and in these cases, the gut inflammation is driven by IL-1ß and is treatable by biologic agents that block the effects of this cytokine.


Asunto(s)
Enfermedad de Crohn/patología , Tracto Gastrointestinal/patología , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Humanos , Inflamación/patología , Interleucina-18/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteínas de Neoplasias/genética , Transducción de Señal/inmunología
18.
Sci Transl Med ; 10(444)2018 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-29875204

RESUMEN

The LRRK2/MUC19 gene region constitutes a high-risk genetic locus for the occurrence of both inflammatory bowel diseases (IBDs) and Parkinson's disease. We show that dendritic cells (DCs) from patients with Crohn's disease (CD) and lymphoblastoid cell lines derived from patients without CD but bearing a high-risk allele (rs11564258) at this locus as heterozygotes exhibited increased LRRK2 expression in vitro. To investigate the immunological consequences of this increased LRRK2 expression, we conducted studies in transgenic mice overexpressing Lrrk2 and showed that these mice exhibited more severe colitis induced by dextran sodium sulfate (DSS) than did littermate control animals. This increase in colitis severity was associated with lamina propria DCs that showed increased Dectin-1-induced NF-κB activation and proinflammatory cytokine secretion. Colitis severity was driven by LRRK2 activation of NF-κB pathway components including the TAK1 complex and TRAF6. Next, we found that membrane-associated LRRK2 (in association with TAB2) caused inactivation of Beclin-1 and inhibition of autophagy. HCT116 colon epithelial cells lacking Beclin-1 exhibited increased LRRK2 expression compared to wild-type cells, suggesting that inhibition of autophagy potentially could augment LRRK2 proinflammatory signaling. We then showed that LRRK2 inhibitors decreased Dectin-1-induced TNF-α production by mouse DCs and ameliorated DSS-induced colitis, both in control and Lrrk2 transgenic animals. Finally, we demonstrated that LRRK2 inhibitors blocked TNF-α production by cultured DCs from patients with CD. Our findings suggest that normalization of LRRK2 activation could be a therapeutic approach for treating IBD, regardless of whether a LRRK2 risk allele is involved.


Asunto(s)
Autofagia , Colitis/inmunología , Colitis/patología , Inmunidad , Lectinas Tipo C/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Animales , Beclina-1/metabolismo , Células de la Médula Ósea/metabolismo , Colon/patología , Enfermedad de Crohn/enzimología , Enfermedad de Crohn/patología , Citocinas/metabolismo , Células Dendríticas/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Humanos , Inflamación/patología , Ratones Transgénicos , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Proteolisis , Transducción de Señal , Transcripción Genética
19.
J Clin Invest ; 128(5): 1793-1806, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29408806

RESUMEN

In these studies, we evaluated the contribution of the NLRP3 inflammasome to Crohn's disease (CD) in a kindred containing individuals having a missense mutation in CARD8, a protein known to inhibit this inflammasome. Whole exome sequencing and PCR studies identified the affected individuals as having a V44I mutation in a single allele of the T60 isoform of CARD8. The serum levels of IL-1ß in the affected individuals were increased compared with those in healthy controls, and their peripheral monocytes produced increased amounts of IL-1ß when stimulated by NLRP3 activators. Immunoblot studies probing the basis of these findings showed that mutated T60 CARD8 failed to downregulate the NLRP3 inflammasome because it did not bind to NLRP3 and inhibit its oligomerization. In addition, these studies showed that mutated T60 CARD8 exerted a dominant-negative effect by its capacity to bind to and form oligomers with unmutated T60 or T48 CARD8 that impeded their binding to NLRP3. Finally, inflammasome activation studies revealed that intact but not mutated CARD8 prevented NLRP3 deubiquitination and serine dephosphorylation. CD due to a CARD8 mutation was not effectively treated by anti-TNF-α, but did respond to IL-1ß inhibitors. Thus, patients with anti-TNF-α-resistant CD may respond to this treatment option.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/inmunología , Enfermedad de Crohn/inmunología , Inflamasomas/inmunología , Mutación con Pérdida de Función , Monocitos/inmunología , Mutación Missense , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteínas de Neoplasias/inmunología , Sustitución de Aminoácidos , Proteínas Adaptadoras de Señalización CARD/genética , Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Femenino , Células HEK293 , Humanos , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Masculino , Monocitos/patología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteínas de Neoplasias/genética , Fosforilación/genética , Fosforilación/inmunología , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Ubiquitinación/genética , Ubiquitinación/inmunología , Secuenciación Completa del Genoma
20.
Exp Ther Med ; 16(5): 4277-4282, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30344701

RESUMEN

Administration of bone marrow-derived mesenchymal stem cells (MSCs) is a possible treatment for graft-versus-host disease (GVHD) following allogeneic hematopoietic stem cell transplantation and other inflammatory conditions. To address the mechanism of immunosuppression by MSCs, in particular those derived from adipose tissue (AMSCs), AMSCs were isolated from three different mouse strains, and the suppressive capacity of the AMSCs thus obtained to suppress interferon (IFN)-γ generation in mixed lymphocyte reaction cultures serving as an in vitro model of GVHD were assessed. It was revealed that the AMSCs had a potent capacity to suppress IFN-γ production regardless of their strain of origin and that such suppression was not associated with production of interleukin-10. In addition, the results demonstrated that ß2-microglobulin (ß2m)-deficient AMSCs from ß2m-/- mice were also potent suppressor cells, verifying the fact that the mechanism underlying the suppression by AMSCs is independent of major histocompatibility complex (MHC) class I expression or MHC compatibility. As AMSCs appear to have immunosuppressive properties, AMSCs may be a useful source of biological suppressor cells for the control of GVHD in humans.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA