Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 9: 2366, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30429846

RESUMEN

Background: Inherited CARD9 deficiency constitutes a primary immunodeficiency predisposing uniquely to chronic and invasive fungal infections. Certain mutations are shown to negatively impact CARD9 protein expression and/or NF-κB activation, but the underlying biochemical mechanism remains to be fully understood. Objectives: To investigate a possible founder origin of a known CARD9 R70W mutation in five families of Turkish origin. To explore the biochemical mechanism of immunodeficiency by R70W CARD9. Methods: We performed haplotype analysis using microsatellite markers and SNPs. We designed a model system exploiting a gain-of-function (GOF) CARD9 L213LI mutant that triggers constitutive NF-κB activation, analogous to an oncogenic CARD11 mutant, to study NF-κB signaling and signalosome formation. We performed reporter assays, immunoprecipitation and confocal imaging on HEK cells overexpressing different CARD9 variants. Results: We identified a common haplotype, thus providing evidence for a common Turkish founder. CARD9 R70W failed to activate NF-κB and abrogated NF-κB activation by WT CARD9 and by GOF CARD9. Notably, R70W CARD9 also exerted negative effects on NF-κB activation by CARD10, CARD11, and CARD14. Consistent with the NF-κB results, the R70W mutation prevented GOF CARD9 to pull down the signalosome partner proteins BCL10 and MALT1. This reflected into drastic reduction of BCL10 filamentous assemblies in a cellular context. Indeed, structural analysis revealed that position R70 in CARD9 maps at the putative interface between successive CARD domains in CARD9 filaments. Conclusions: The R70W mutation in CARD9 prevents NF-κB activation by inhibiting productive interactions with downstream BCL10 and MALT1, necessary for assembly of the filamentous CARD9-BCL10-MALT1 signalosome.


Asunto(s)
Proteína 10 de la LLC-Linfoma de Células B/metabolismo , Proteínas Adaptadoras de Señalización CARD/genética , Efecto Fundador , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , Mutación , FN-kappa B/metabolismo , Transducción de Señal , Proteínas Adaptadoras de Señalización CARD/química , Línea Celular , Susceptibilidad a Enfermedades , Femenino , Mutación con Ganancia de Función , Humanos , Masculino , Modelos Moleculares , Linaje , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
3.
Immunity ; 48(1): 45-58.e6, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29287995

RESUMEN

Interleukin-23 (IL-23), an IL-12 family cytokine, plays pivotal roles in pro-inflammatory T helper 17 cell responses linked to autoimmune and inflammatory diseases. Despite intense therapeutic targeting, structural and mechanistic insights into receptor complexes mediated by IL-23, and by IL-12 family members in general, have remained elusive. We determined a crystal structure of human IL-23 in complex with its cognate receptor, IL-23R, and revealed that IL-23R bound to IL-23 exclusively via its N-terminal immunoglobulin domain. The structural and functional hotspot of this interaction partially restructured the helical IL-23p19 subunit of IL-23 and restrained its IL-12p40 subunit to cooperatively bind the shared receptor IL-12Rß1 with high affinity. Together with structural insights from the interaction of IL-23 with the inhibitory antibody briakinumab and by leveraging additional IL-23:antibody complexes, we propose a mechanistic paradigm for IL-23 and IL-12 whereby cognate receptor binding to the helical cytokine subunits primes recruitment of the shared receptors via the IL-12p40 subunit.


Asunto(s)
Sudunidad beta 1 del Receptor de Interleucina-12/metabolismo , Interleucina-23/metabolismo , Receptores de Interleucina/metabolismo , Animales , Calorimetría/métodos , Línea Celular , Humanos , Interferometría/métodos , Subunidad p40 de la Interleucina-12/metabolismo , Masculino , Ratones , Unión Proteica/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
4.
Gastroenterology ; 153(4): 1054-1067, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28642198

RESUMEN

BACKGROUND: Intestinal fibrosis resulting in (sub)obstruction is a common complication of Crohn's disease (CD). Rho kinases (ROCKs) play multiple roles in TGFß-induced myofibroblast activation that could be therapeutic targets. Because systemic ROCK inhibition causes cardiovascular side effects, we evaluated the effects of a locally acting ROCK inhibitor (AMA0825) on intestinal fibrosis. METHODS: Fibrosis was assessed in mouse models using dextran sulfate sodium (DSS) and adoptive T-cell transfer. The in vitro and ex vivo effects of AMA0825 were studied in different cell types and in CD biopsy cultures. RESULTS: ROCK is expressed in fibroblastic, epithelial, endothelial, and muscle cells of the human intestinal tract and is activated in inflamed and fibrotic tissue. Prophylactic treatment with AMA0825 inhibited myofibroblast accumulation, expression of pro-fibrotic factors, and accumulation of fibrotic tissue without affecting clinical disease activity and histologic inflammation in 2 models of fibrosis. ROCK inhibition reversed established fibrosis in a chronic DSS model and impeded ex vivo pro-fibrotic protein secretion from stenotic CD biopsies. AMA0825 reduced TGFß1-induced activation of myocardin-related transcription factor (MRTF) and p38 mitogen-activated protein kinase (MAPK), down-regulating matrix metalloproteinases, collagen, and IL6 secretion from fibroblasts. In these cells, ROCK inhibition potentiated autophagy, which was required for the observed reduction in collagen and IL6 production. AMA0825 did not affect pro-inflammatory cytokine secretion from other ROCK-positive cell types, corroborating the selective in vivo effect on fibrosis. CONCLUSIONS: Local ROCK inhibition prevents and reverses intestinal fibrosis by diminishing MRTF and p38 MAPK activation and increasing autophagy in fibroblasts. Overall, our results show that local ROCK inhibition is promising for counteracting fibrosis as an add-on therapy for CD.


Asunto(s)
Íleon/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/prevención & control , Obstrucción Intestinal/prevención & control , Miofibroblastos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Traslado Adoptivo , Animales , Autofagia/efectos de los fármacos , Estudios de Casos y Controles , Colágeno/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Activación Enzimática , Fibrosis , Humanos , Íleon/enzimología , Íleon/inmunología , Íleon/patología , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/patología , Interleucina-6/metabolismo , Obstrucción Intestinal/inducido químicamente , Obstrucción Intestinal/enzimología , Obstrucción Intestinal/patología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones Endogámicos C57BL , Miofibroblastos/enzimología , Miofibroblastos/inmunología , Miofibroblastos/patología , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/trasplante , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Quinasas Asociadas a rho/metabolismo
5.
Histochem Cell Biol ; 148(1): 85-93, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28265783

RESUMEN

Colonic adenocarcinoma-derived Caco-2 and T84 epithelial cell lines are frequently used as in vitro model systems of functional epithelial barriers. Both are utilised interchangeably despite evidence that differentiated Caco-2 cells are more reminiscent of small intestinal enterocytes than of colonocytes, whereas differentiated T84 cells are less well characterised. The aim of this study was, therefore, to further characterise and compare differentiated Caco-2 and T84 cells. The objectives were to (1) compare the brush border morphology, (2) measure the expression of enterocyte- and colonocyte-specific genes and (3) compare their response to butyrate, which is dependent on the monocarboxylate transporter 1 (MCT1), an apical protein expressed primarily in colonocytes. T84 microvilli were significantly shorter than those of Caco-2 cells, which is a characteristic difference between small intestinal enterocytes and colonocytes. Also, enterocyte-associated brush border enzymes expressed in differentiated Caco-2 cells were not increased during T84 maturation, whereas colonic markers such as MCT1 were more abundant in differentiated T84 cells compared to differentiated Caco-2 cells. Consequently, T84 cells displayed a dose-responsive improvement of barrier function towards butyrate, which was absent in Caco-2 cells. On the other hand, differences in epithelial toll-like receptor expression between Caco-2 and T84 monolayers did not result in a corresponding differential functional response. We conclude that differentiated Caco-2 and T84 cells have distinct morphological, biochemical and functional characteristics, suggesting that T84 cells do not acquire the biochemical signature of mature small intestinal enterocytes like Caco-2 cells, but retain much of their original colonic characteristics throughout differentiation. These findings can help investigators select the appropriate intestinal epithelial cell line for specific in vitro research purposes.


Asunto(s)
Colon/patología , Modelos Biológicos , Animales , Células CACO-2 , Humanos , Células Tumorales Cultivadas
6.
Lab Invest ; 97(5): 519-529, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28165466

RESUMEN

Bile acids regulate the expression of intestinal bile acid transporters and are natural ligands for nuclear receptors controlling inflammation. Accumulating evidence suggests that signaling through these receptors is impaired in inflammatory bowel disease. We investigated whether tauroursodeoxycholic acid (TUDCA), a secondary bile acid with cytoprotective properties, regulates ileal nuclear receptor and bile acid transporter expression and assessed its therapeutic potential in an experimental model of Crohn's disease (CD). Gene expression of the nuclear receptors farnesoid X receptor, pregnane X receptor and vitamin D receptor and the bile acid transporters apical sodium-dependent bile acid transporter and organic solute transporter α and ß was analyzed in Caco-2 cell monolayers exposed to tumor necrosis factor (TNF)α, in ileal tissue of TNFΔARE/WT mice and in inflamed ileal biopsies from CD patients by quantitative real-time polymerase chain reaction. TNFΔARE/WT mice and wild-type littermates were treated with TUDCA or placebo for 11 weeks and ileal histopathology and expression of the aforementioned genes were determined. Exposing Caco-2 cell monolayers to TNFα impaired the mRNA expression of nuclear receptors and bile acid transporters, whereas co-incubation with TUDCA antagonized their downregulation. TNFΔARE/WT mice displayed altered ileal bile acid homeostasis that mimicked the situation in human CD ileitis. Administration of TUDCA attenuated ileitis and alleviated the downregulation of nuclear receptors and bile acid transporters in these mice. These results show that TUDCA protects bile acid homeostasis under inflammatory conditions and suppresses CD-like ileitis. Together with previous observations showing similar efficacy in experimental colitis, we conclude that TUDCA could be a promising therapeutic agent for inflammatory bowel disease, warranting a clinical trial.


Asunto(s)
Enfermedad de Crohn/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Homeostasis/efectos de los fármacos , Ileítis/metabolismo , Ácido Tauroquenodesoxicólico/farmacología , Adulto , Animales , Ácidos y Sales Biliares/metabolismo , Células CACO-2 , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Íleon/efectos de los fármacos , Íleon/patología , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Receptores Citoplasmáticos y Nucleares/metabolismo , Adulto Joven
7.
Appl Environ Microbiol ; 83(7)2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28115375

RESUMEN

The promising results seen in studies of secondary bile acids in experimental colitis suggest that they may represent an attractive and safe class of drugs for the treatment of inflammatory bowel diseases (IBD). However, the exact mechanism by which bile acid therapy confers protection from colitogenesis is currently unknown. Since the gut microbiota plays a crucial role in the pathogenesis of IBD, and exogenous bile acid administration may affect the community structure of the microbiota, we examined the impact of the secondary bile acid ursodeoxycholic acid (UDCA) and its taurine or glycine conjugates on the fecal microbial community structure during experimental colitis. Daily oral administration of UDCA, tauroursodeoxycholic acid (TUDCA), or glycoursodeoxycholic acid (GUDCA) equally lowered the severity of dextran sodium sulfate-induced colitis in mice, as evidenced by reduced body weight loss, colonic shortening, and expression of inflammatory cytokines. Illumina sequencing demonstrated that bile acid therapy during colitis did not restore fecal bacterial richness and diversity. However, bile acid therapy normalized the colitis-associated increased ratio of Firmicutes to Bacteroidetes Interestingly, administration of bile acids prevented the loss of Clostridium cluster XIVa and increased the abundance of Akkermansia muciniphila, bacterial species known to be particularly decreased in IBD patients. We conclude that UDCA, which is an FDA-approved drug for cholestatic liver disorders, could be an attractive treatment option to reduce dysbiosis and ameliorate inflammation in human IBD.IMPORTANCE Secondary bile acids are emerging as attractive candidates for the treatment of inflammatory bowel disease. Although bile acids may affect the intestinal microbial community structure, which significantly contributes to the course of these inflammatory disorders, the impact of bile acid therapy on the fecal microbiota during colitis has not yet been considered. Here, we studied the alterations in the fecal microbial abundance in colitic mice following the administration of secondary bile acids. Our results show that secondary bile acids reduce the severity of colitis and ameliorate colitis-associated fecal dysbiosis at the phylum level. This study indicates that secondary bile acids might act as a safe and effective drug for inflammatory bowel disease.


Asunto(s)
Disbiosis/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Ácido Tauroquenodesoxicólico/uso terapéutico , Ácido Ursodesoxicólico/análogos & derivados , Ácido Ursodesoxicólico/uso terapéutico , Animales , Bacteroides/efectos de los fármacos , Colon/microbiología , Sulfato de Dextran/administración & dosificación , Modelos Animales de Enfermedad , Heces/microbiología , Firmicutes/efectos de los fármacos , Humanos , Ratones , Taurina/química , Ácido Tauroquenodesoxicólico/administración & dosificación , Ácido Ursodesoxicólico/administración & dosificación , Ácido Ursodesoxicólico/química
8.
J Pathol ; 241(4): 547-558, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27981571

RESUMEN

Prolyl hydroxylase domain-containing proteins (PHDs) regulate the adaptation of cells to hypoxia. Pan-hydroxylase inhibition is protective in experimental colitis, in which PHD1 plays a prominent role. However, it is currently unknown how PHD1 targeting regulates this protection and which cell type(s) are involved. Here, we demonstrated that Phd1 deletion in endothelial and haematopoietic cells (Phd1f/f Tie2:cre) protected mice from dextran sulphate sodium (DSS)-induced colitis, with reduced epithelial erosions, immune cell infiltration, and colonic microvascular dysfunction, whereas the response of Phd2f/+ Tie2:cre and Phd3f/f Tie2:cre mice to DSS was similar to that of their littermate controls. Using bone marrow chimeras and cell-specific cre mice, we demonstrated that ablation of Phd1 in haematopoietic cells but not in endothelial cells was both necessary and sufficient to inhibit experimental colitis. This effect relied, at least in part, on skewing of Phd1-deficient bone marrow-derived macrophages towards an anti-inflammatory M2 phenotype. These cells showed an attenuated nuclear factor-κB-dependent response to lipopolysaccharide (LPS), which in turn diminished endothelial chemokine expression. In addition, Phd1 deficiency in dendritic cells significantly reduced interleukin-1ß production in response to LPS. Taken together, our results further support the development of selective PHD1 inhibitors for ulcerative colitis, and identify haematopoietic cells as their primary target. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Macrófagos/metabolismo , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colon/efectos de los fármacos , Colon/patología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Eliminación de Gen , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , Procolágeno-Prolina Dioxigenasa/deficiencia , Procolágeno-Prolina Dioxigenasa/genética
9.
J Crohns Colitis ; 11(2): 229-236, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27484096

RESUMEN

BACKGROUND AND AIMS: Butyricicoccus is a butyrate-producing clostridial cluster IV genus whose numbers are reduced in the stool of ulcerative colitis [UC] patients. Conditioned medium of Butyricicoccus [B.] pullicaecorum prevents tumour necrosis factor alpha [TNFα]-induced increase in epithelial permeability in vitro. Since butyrate influences intestinal barrier integrity, we further investigated the relationship between the abundance of mucosa-associated Butyricicoccus and the expression of butyrate-regulated tight junction [TJ] genes. METHODS: Tight junction protein 1 [TJP1], occludin [OCLN], claudin-1 [CLDN1], and Butyricicoccus 16S rRNA expression was analysed in a collection of colonic biopsies of healthy controls and UC patients with active disease. The effect of butyrate and B. pullicaecorum conditioned medium on TJ gene expression was investigated in TNFα-stimulated Caco-2 monolayers and inflamed mucosal biopsies of UC patients. RESULTS: TJP1 expression was significantly decreased in inflamed UC mucosa, whereas CLDN1 mRNA levels were increased. OCLN did not differ significantly between the groups. Mucosa-associated Butyricicoccus 16S rRNA transcripts were reduced in active UC patients compared with healthy controls. Interestingly, Butyricicoccus activity negatively correlated with CLDN1 expression. Butyrate reversed the inflammation-induced increase of CLDN1 protein levels, and stimulation of inflamed UC biopsies with B. pullicaecorum conditioned medium normalized CLDN1 mRNA levels. CONCLUSIONS: Butyricicoccus is a mucosa-associated bacterial genus under-represented in colonic mucosa of patients with active UC, whose activity inversely correlates with CLDN1 expression. Butyrate and B. pullicaecorum conditioned medium reduce CLDN1 expression, supporting its use as a pharmabiotic preserving epithelial TJ integrity.


Asunto(s)
Claudina-1/metabolismo , Colitis Ulcerosa , Eubacterium , Mucosa Intestinal , Ocludina/metabolismo , Uniones Estrechas , Proteína de la Zonula Occludens-1/metabolismo , Adulto , Biopsia/métodos , Butiratos/metabolismo , Células CACO-2 , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Eubacterium/aislamiento & purificación , Eubacterium/fisiología , Heces/microbiología , Femenino , Interacciones Huésped-Patógeno/fisiología , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Gravedad del Paciente , ARN Ribosómico 16S/análisis , Estadística como Asunto , Uniones Estrechas/metabolismo , Uniones Estrechas/microbiología , Uniones Estrechas/patología , Factor de Necrosis Tumoral alfa/metabolismo
10.
Lab Invest ; 94(12): 1419-30, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25310532

RESUMEN

Ulcerative colitis (UC) is characterized by increased epithelial cell death and subsequent breakdown of the intestinal epithelial barrier, which perpetuates chronic intestinal inflammation. Since fecal bile acid dysmetabolism is associated with UC and tauroursodeoxycholic acid (TUDCA) has been shown to improve murine colitis, we evaluated the effect of TUDCA on intestinal epithelial cell death in a mouse model of UC-like barrier dysfunction elicited by dextran sulfate sodium (DSS). We identified the prevention of colonic caspase-3 induction, a key proapoptotic marker which was also over-activated in UC, as the earliest event resulting in a clear clinical benefit. Whereas vehicle-treated mice showed a cumulative mortality of 40%, all TUDCA-treated mice survived the DSS experiment during a 14-day follow-up period. In line with a barrier protective effect, TUDCA decreased bacterial translocation to the spleen and stimulated mucin production. Similarly, TUDCA inhibited lipopolysaccharide-induced intestinal permeability and associated enterocyte apoptosis. The anti-apoptotic effect was confirmed in vitro by a dose-dependent inhibition of both receptor-dependent (using tumor necrosis factor and Fas ligand) and receptor-independent (staurosporine) caspase-3 induction in HT29 colonic epithelial cells. These data imply that caspase-3 activation is an early marker of colitis that is prevented by TUDCA treatment. These data, together with the previously reported beneficial effect in colitis, suggest that TUDCA could be an add-on strategy to current immunosuppressive treatment of UC patients.


Asunto(s)
Apoptosis/efectos de los fármacos , Colitis/prevención & control , Células Epiteliales/patología , Mucosa Intestinal/efectos de los fármacos , Ácido Tauroquenodesoxicólico/uso terapéutico , Adolescente , Adulto , Anciano , Animales , Caspasa 3/metabolismo , Niño , Colitis/inducido químicamente , Sulfato de Dextran , Células Epiteliales/metabolismo , Femenino , Células HT29 , Humanos , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/fisiología , Receptores de Vitronectina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...