Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Inflamm Res ; 68(3): 223-230, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30564895

RESUMEN

OBJECTIVE AND DESIGN: Protease activity of MALT lymphoma-translocation protein 1 (Malt1) plays an important role in the development of colitis, but the detailed mechanism has not been fully elucidated. METHOD: Effects of Malt1 protease on the activation of T cells and the development of experimental colitis was investigated using Malt1 protease-deficient (PD) mouse. RESULTS: IL-2 production from CD4+ T cells of Malt1 PD mice was decreased compared with that of wild-type (WT) mice. Intraperitoneal injection of anti-CD3 antibody into Malt1 PD mouse induced less productions of IL-17 in the plasma, as well as the colonic gene expression of IL-17A, compared with WT mice, whereas IFN-γ production was not impaired. In naïve T-cell transfer colitis model, Malt1 PD T cells induced less disease severity than WT T cells. Then, reduction in the populations of Th17 and Th1/17 cells was observed in the mesenteric lymph nodes of the recipient mice transferred with Malt1 PD T cells, whereas those of Th1 cells were not impaired. IL-17A expression in the colon was also decreased in the mouse receiving Malt1 PD T cells. CONCLUSIONS: Inactivation of Malt1 protease activity abrogates Th17 and Th1/17 cell activation, resulting in the amelioration of experimental colitis.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Citocinas/inmunología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/trasplante , Colitis/inmunología , Colon/inmunología , Femenino , Ganglios Linfáticos/inmunología , Ratones Endogámicos BALB C , Ratones SCID
2.
Inflamm Res ; 68(6): 493-509, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30972425

RESUMEN

OBJECTIVE AND DESIGN: To evaluate the potency of RORγt blockade for treatment of Inflammatory Bowel Disease (IBD), the efficacy of TAK-828F, a novel RORγt inverse agonist, in anti-TNF-α mAb non-responsive mouse colitis model and effect of TAK-828F on IL-17 production in peripheral mononuclear blood cells (PBMCs) of anti-TNF-α naive and treatment-failure patients of IBD was investigated. METHODS AND RESULTS: The colitis model showed Th17-dependent pathogenicity and response to anti-IL-12/23p40 monoclonal antibody (mAb), but no response to anti-TNF-α mAb. In the model, TAK-828F, at oral dosages of 1 and 3 mg/kg, inhibited progression of colitis and reduced the immune reaction that characterize Th17 cells. Anti-IL-17A mAb showed neither efficacy nor change in the T cell population and colonic gene expression in the model. In the normal mouse, a 4-week treatment of TAK-828F at 30 mg/kg did not severely reduce lymphocyte cell counts in peripheral and intestinal mucosa, which was observed in RORγ-/- mice. TAK-828F strongly inhibited IL-17 gene expression with IC50 values from 21.4 to 34.4 nmol/L in PBMCs from anti-TNF mAb naive and treatment-failure patients of IBD. CONCLUSIONS: These results indicate that RORγt blockade would provide an effective approach for treating refractory patients with IBD by blocking IL-23/Th17 pathway.


Asunto(s)
Acetatos/farmacología , Acetatos/uso terapéutico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Leucocitos Mononucleares/efectos de los fármacos , Naftiridinas/farmacología , Naftiridinas/uso terapéutico , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Adolescente , Adulto , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Células Cultivadas , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/genética , Interleucina-17/inmunología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Leucocitos Mononucleares/inmunología , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Persona de Mediana Edad , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adulto Joven
3.
Biochem Biophys Res Commun ; 483(1): 183-190, 2017 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-28039053

RESUMEN

Dedicator of cytokinesis 2 (DOCK2) is a key molecule for lymphocyte activation and migration. DOCK2 interacts with Ras-related C3 botulinus toxin substrate 1 (Rac1, GTPase) and mediates the GDP-GTP exchange reaction, indicating that inhibitors against protein-protein interaction (PPI) between DOCK2 and Rac1 would be good drug candidates for treating immune-related disorders. Here, we report DOCK2-selective PPI inhibitory peptides discovered using random peptide T7 phage display technology. These peptides inhibited DOCK2 activity at nanomolar concentrations and were delivered to intracellular compartments by combination with cell-penetrating peptide (CPP). Consequently, one peptide, R4-DCpep-2(V2W/K4R/ox)-NH2 (Ac-RRRRCWARYHGYPWCRRRR-NH2), inhibited migration in human B lymphocyte MINO cell line at IC50 = 120 nM. To our knowledge, this is the first report of a DOCK2-selective peptide inhibitor; this study will contribute to the development of novel DOCK2-targeting immunosuppressive drugs.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Linfoma de Células B/tratamiento farmacológico , Péptidos/química , Péptidos/farmacología , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Sistema Libre de Células , Evaluación Preclínica de Medicamentos/métodos , Proteínas Activadoras de GTPasa , Humanos , Linfoma de Células B/patología , Biblioteca de Péptidos , Péptidos/metabolismo , Mapas de Interacción de Proteínas , Proteína de Unión al GTP rac1/metabolismo
4.
Inflammation ; 42(1): 91-102, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30121880

RESUMEN

IL-17-producing Th17 cells and IFN-γ and IL-17 double-producing Th1/17 cells have been identified as the pathogenic cells in inflammatory bowel disease (IBD). Retinoic acid-related orphan receptor γt (RORγt) is a master regulator for the differentiation and activation of Th17 and Th1/17 cells. We discovered a novel orally available TAK-828F, a strong and selective RORγt inverse agonist. To assess the potential of RORγt blockade in the therapy for IBD, the efficacy of TAK-828F in activated T cell transfer mouse colitis model was investigated. This model was highly sensitive to the prophylactic treatment of anti-TNF-α monoclonal antibody but partially susceptible to sulfasalazine, tacrolimus, and prednisolone. Oral administration of TAK-828F, at doses of 1 and 3 mg/kg, b.i.d, strongly protected the progression of colitis. TAK-828F decreased the population of Th17 and Th1/17 cells in a dose-dependent manner in the mesenteric lymph node. Moreover, expression of mRNA that are characteristic of the Th17 signature, such as IL-17A and IL-17F in the colon, were inhibited by TAK-828F, while the expression of IL-10, an anti-inflammatory cytokine, was increased. In the therapeutic treatment, TAK-828F lessened disease severity compared to the vehicle control mice. Interestingly, gene expression of zonula occludens-1 (ZO-1) and mucin 2 (Muc2), which play an important role in barrier function of the intestinal mucosa, was recovered by TAK-828F. These results indicate that blocking RORγt has promising pharmacological profile in the colitis model. RORγt blockade may provide a novel therapeutic paradigm for treatment of IBD with unique mechanism by which improves imbalance of the immune system.


Asunto(s)
Acetatos/farmacología , Colitis/tratamiento farmacológico , Agonismo Inverso de Drogas , Naftiridinas/farmacología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Mucosa Intestinal/metabolismo , Ganglios Linfáticos/citología , Ratones , Células Th17/citología , Células Th17/efectos de los fármacos
5.
Int Immunopharmacol ; 60: 160-169, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29730559

RESUMEN

C-C chemokine receptor 9 (CCR9) is the homing receptor for C-C motif chemokine ligand 25 (CCL25), and contributes to the maintenance of mucosal immunity and pathogenesis of inflammatory bowel disease (IBD) through the recruitment of T cells into the gut mucosa. Recent reports suggest that the interaction of CCR9 and CCL25 in the large intestine correlate with disease severity of colonic IBD. MLN3126 is an orally available small molecular compound with potent and selective CCR9 antagonist activity. MLN3126 inhibited CCL25-induced calcium mobilization in human CCR9 transfected cells and CCL25-induced chemotaxis of mouse primary thymocytes in a dose-dependent manner. The potential effect of MLN3126 in an activated T cell transfer mouse colitis model was compared with that of an anti-tumor necrosis factor (TNF)-α antibody. CCL25 protein was detected in the colon of mucosal epithelial cells and CCR9+ CD4+ T cells were observed in the lamina propria of the colon of mice with colitis. Dietary administration of MLN3126 to the mice maintained sufficient concentration of the compound in the plasma and dose-dependently inhibited progression of colitis compared to the vehicle control group. Anti-TNF-α antibody, a surrogate for a standard of care for IBD treatment, was also efficacious in the colitis model. These results suggest that MLN3126 would be a promising orally available CCR9 antagonist to treat colonic IBD.


Asunto(s)
Antiinflamatorios/uso terapéutico , Colitis/tratamiento farmacológico , Receptores CCR/antagonistas & inhibidores , Animales , Células CHO , Movimiento Celular/efectos de los fármacos , Quimiocinas CC/inmunología , Colitis/inmunología , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Cricetulus , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos BALB C , Ratones SCID , Linfocitos T/inmunología
6.
Dig Dis Sci ; 47(7): 1538-45, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12141814

RESUMEN

Nonsteroidal antiinflammatory drugs (NSAIDs) produce gastric damage in experimental animals, irrespective of the route of administration. However, aspirin (ASA) causes damage only when it is given orally. In the present study, we examined the gastric ulcerogenic effect of subcutaneously administered ASA in rats, in comparison with various NSAIDs, and investigated the reason why ASA does not cause damage in the stomach, in relation to its metabolite salicylic acid (SA). Since the antiinflammatory action of SA is known to be mediated, partly, by endogenous adenosine (AD), we also examined the possible involvement of AD in the protective action of SA. Various NSAIDs (indomethacin, flurbiprofen, naproxen, diclrofenac, ASA, SA) were administered subcutaneously, and the gastric mucosa was examined macroscopically 4 hr later. All NSAIDs tested, except ASA and SA, caused hemorrhagic lesions in the stomach, with a marked gastric hypermotility and a decrease of mucosal PGE2 contents. These ulcerogenic and motility responses caused by NSAIDs were blocked by pretreatment with atropine or PGE2. ASA, although inhibiting PGE2 generation, caused neither hypermotility nor damage in the stomach. On the other hand, SA alone inhibited basal gastric motility without any effect on mucosal PGE2 contents, and this agent, when given together with indomethacin, prevented gastric hypermotility and lesion formation in response to indomethacin, without affecting the reduced PGE2 contents. Likewise, ASA inhibited these responses to indomethacin, yet the effects appeared later than those of SA. Following administration of ASA, the blood SA levels reached a peak within 30 min and remained elevated for 4 hr. In addition, the protective effect of SA was not significantly influenced by either the AD deaminase or the AD-receptor antagonists. These results suggest that the failure of parenteral ASA to induce gastric damage may be explained by a protective action of SA metabolized from ASA. SA has a cytoprotective action against NSAID-induced gastric lesions, and this action is not mediated by endogenous AD but may be functionally associated with inhibition of the gastric motility response.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/efectos adversos , Aspirina/administración & dosificación , Aspirina/efectos adversos , Mucosa Gástrica/efectos de los fármacos , Ácido Salicílico/farmacología , Adenosina/fisiología , Animales , Mucosa Gástrica/patología , Motilidad Gastrointestinal/efectos de los fármacos , Indometacina/administración & dosificación , Indometacina/efectos adversos , Inyecciones Subcutáneas , Masculino , Ratas , Ratas Sprague-Dawley
7.
Digestion ; 66(3): 145-53, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12481160

RESUMEN

BACKGROUND/AIM: Studies have demonstrated an important role for endogenous PG and NO in the healing of chronic gastric ulcers. We investigated the effects of COX and NOS inhibitors on the healing of duodenal ulcers, in comparison with gastric ulcers, in rats. METHODS: Gastric and duodenal ulcers were induced by serosal application of acetic acid (0.1 ml of 100% acetic acid) for 60 and 20 s, respectively. Indomethacin (a nonselective COX inhibitor) or rofecoxib (a selective COX-2 inhibitor) was given p.o. once daily for 14 days from 3 days after ulcer induction, while N(G)-nitro-L-arginine methyl ester (L-NAME: a nonselective NOS inhibitor) or aminoguanidine (a relatively selective iNOS inhibitor) was given s.c. twice daily during this period. RESULTS: Both gastric and duodenal ulcers induced by acetic acid healed spontaneously within 17 days to a minimal size. Daily administration of indomethacin or rofecoxib significantly delayed the healing of gastric but not duodenal ulcers. In contrast, the healing of both gastric and duodenal ulcers was delayed by repeated administration of either L-NAME or aminoguanidine. Ulceration markedly increased the PGE(2) content of the ulcerated mucosa in both the stomach and duodenum, and the increased PG biosynthetic response was inhibited by either indomethacin or rofecoxib in both tissues. The expression of both COX-2 and iNOS mRNAs was upregulated in the ulcerated mucosa of the stomach and duodenum. CONCLUSION: These results suggest that COX-2/PG is actively involved in the healing of gastric but not duodenal ulcers, although the mRNA for COX-2 is expressed in the duodenal mucosa after ulceration, as potently as in the gastric mucosa. In contrast, NO produced by both cNOS and iNOS plays a role in the healing of both gastric and duodenal ulcers.


Asunto(s)
Inhibidores de la Ciclooxigenasa/farmacología , Úlcera Duodenal/fisiopatología , Indometacina/farmacología , Lactonas/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Úlcera Gástrica/fisiopatología , Sulfonas , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda