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











Base de dados
Intervalo de ano de publicação
1.
Chem Biol Interact ; 351: 109712, 2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-34699766

RESUMO

Irinotecan (CPT-11) is one of the main agents used to treat colorectal cancer; unfortunately, it is associated with increased intestinal mucositis developing. Luteolin has been shown to prevent damage induced by this chemotherapeutic in mice; thus, in this research, we have investigated luteolin's action mechanism in human intestinal epithelial cells. The potential of luteolin in reducing inflammation and oxidative stress induced by irinotecan in Caco-2 cells was evaluated by PCR through mRNA expression of inflammatory and oxidative genes and by ELISA at the protein level. To assess whether luteolin's ability to control irinotecan-induced damage occurs in a PPARγ dependent manner, experiments were performed on PPARγ downregulated cells. Irinotecan downregulated PPARγ expression and upregulated inflammatory and oxidative genes, while luteolin upregulated PPARγ, HO-1, SOD and decreased expression of IL-1ß and iNOS. Interestingly, when the cells were co-stimulated with luteolin and irinotecan, the flavonoid reversed the inflammation and oxidative imbalance evoked by the chemotherapeutic. However, when these experiments were performed in cells downregulated for PPARγ, luteolin lost the capacity to increase PPARγ and reverse the effect of irinotecan in all tested genes, except by IL-1ß. The present study showed that the protective effect of luteolin against irinotecan is PPARγ dependent.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Irinotecano/toxicidade , Luteolina/farmacologia , PPAR gama/metabolismo , Células CACO-2 , Regulação para Baixo/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Humanos , Interleucina-1beta/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Superóxido Dismutase/metabolismo , Regulação para Cima/efeitos dos fármacos
2.
Int J Mol Sci ; 23(1)2021 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-35008525

RESUMO

Oxaliplatin, the first-line chemotherapeutic agent against colorectal cancer (CRC), induces peripheral neuropathies, which can lead to dose limitation and treatment discontinuation. Downregulation of potassium channels, which involves histone deacetylase (HDAC) activity, has been identified as an important tuner of acute oxaliplatin-induced hypersensitivity. MS-275, a class I histone deacetylase inhibitor (HDACi), prevents acute oxaliplatin-induced peripheral neuropathy (OIPN). Moreover, MS-275 exerts anti-tumor activity in several types of cancers, including CRC. We thus hypothesized that MS-275 could exert both a preventive effect against OIPN and potentially a synergistic effect combined with oxaliplatin against CRC development. We first used RNAseq to assess transcriptional changes occurring in DRG neurons from mice treated by repeated injection of oxaliplatin. Moreover, we assessed the effects of MS-275 on chronic oxaliplatin-induced peripheral neuropathy development in vivo on APCMin/+ mice and on cancer progression when combined with oxaliplatin, both in vivo on APCMin/+ mice and in a mouse model of an orthotopic allograft of the CT26 cell line as well as in vitro in T84 and HT29 human CRC cell lines. We found 741 differentially expressed genes (DEGs) between oxaliplatin- and vehicle-treated animals. While acute OIPN is known as a channelopathy involving HDAC activity, chronic OIPN exerts weak ion channel transcriptional changes and no HDAC expression changes in peripheral neurons from OIPN mice. However, MS-275 prevents the development of sensory neuropathic symptoms induced by repeated oxaliplatin administration in APCMin/+ mice. Moreover, combined with oxaliplatin, MS-275 also exerts synergistic antiproliferative and increased survival effects in CT26-bearing mice. Consistently, combined drug associations exert synergic apoptotic and cell death effects in both T84 and HT29 human CRC cell lines. Our results strongly suggest combining oxaliplatin and MS-275 administration in CRC patients in order to potentiate the antiproliferative action of chemotherapy, while preventing its neurotoxic effect.


Assuntos
Benzamidas/farmacologia , Proliferação de Células/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Síndromes Neurotóxicas/tratamento farmacológico , Oxaliplatina/farmacologia , Piridinas/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Células HT29 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
3.
Cells ; 8(6)2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31212833

RESUMO

An original immuno-regulatory strategy against inflammatory bowel diseases based on the use of 28 kDa glutathione S-transferase (P28GST), a unique schistosome protein, was recently proposed. Improvement of intestinal inflammation occurs through restoration of the immunological balance between pro-inflammatory T-helper 1 (Th1) responses and both T-helper 2 (Th2) and regulatory responses. However, detailed mechanisms explaining how P28GST prevents colitis and promotes gut homeostasis remain unknown. Considering the complex interplay between the adaptive and innate immune system and the intestinal microbiota, we raised the question of the possible role of the microbial ecosystem in the anti-inflammatory effects mediated by the helminth-derived P28GST protein. We first analyzed, by 16S rRNA sequencing, the bacterial profiles of mice fecal microbiota at several time points of the P28GST-immunomodulation period prior to trinitrobenzene sulfonic acid (TNBS)-colitis. The influence of gut microbiota in the P28GST-mediated anti-inflammatory effects was then assessed by fecal microbiota transplantation experiments from P28GST-immunized mice to either conventional or microbiota depleted naïve recipient mice. Finally, the experimental data were supplemented by the temporal fecal microbiota compositions of P28GST-treated Crohn's disease patients from a pilot clinical study (NCT02281916). The P28GST administration slightly modulated the diversity and composition of mouse fecal microbiota while it significantly reduced experimental colitis in mice. Fecal microbiota transplantation experiments failed to restore the P28GST-induced anti-inflammatory effects. In Crohn's disease patients, P28GST also induced slight changes in their overall fecal bacterial composition. Collectively, these results provide key elements in both the anti-inflammatory mechanisms and the safe therapeutic use of immunomodulation with such promising helminth-derived molecules.


Assuntos
Anti-Inflamatórios/uso terapêutico , Microbioma Gastrointestinal , Glutationa Transferase/uso terapêutico , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Colite/induzido quimicamente , Colite/microbiologia , Colite/prevenção & controle , Colite/terapia , Doença de Crohn/microbiologia , Transplante de Microbiota Fecal , Fezes/microbiologia , Feminino , Humanos , Imunização , Imunomodulação , Camundongos Endogâmicos BALB C , Fenótipo , Ácido Trinitrobenzenossulfônico
4.
Inflamm Bowel Dis ; 22(2): 279-92, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26535766

RESUMO

BACKGROUND: Intestinal fibrosis is mainly associated with Crohn's disease and is defined as a progressive and excessive deposition of extracellular matrix components. No specific antifibrotic therapies are available. In this study, we evaluate the antifibrotic effect of a novel 5-ASA analog able to activate the peroxisome proliferator-activated receptor γ, named GED-0507-34 Levo. METHODS: Colonic fibrosis was induced in 110 C57BL/6 mice by 3 cycles of 2.5% (wt/vol) dextran sulfate sodium administration for 6 weeks. The preventive effects of oral daily GED (30 mg · kg(-1) · d(-1)) administration were evaluated using a macroscopic and histological score and also through biological endpoints. Expression of main markers of myofibroblasts activation was determined in transforming growth factor (TGF-ß)-stimulated intestinal fibroblasts and epithelial cells. RESULTS: GED improved macroscopic and microscopic intestinal lesions in dextran sulfate sodium-treated animals and reduced the profibrotic gene expression of Acta2, COL1a1, and Fn1 by 1.48-folds (P < 0.05), 1.93-folds (P < 0.005), and 1.03-fold (P < 0.05), respectively. It reduced protein levels of main markers of fibrosis (α-SMA and Collagen I-II) and the main TGF-ß/Smad pathway components. GED also decreased the interleukin-13 and connective tissue growth factor expression by 1.89-folds (P < 0.05) and 2.2-folds (P < 0.005), respectively. GED inhibited TGF-ß-induced activation of both fibroblast and intestinal epithelial cell lines, by regulating mRNA expression of α-SMA and fibronectin, and restoring the TGF-ß-induced loss of intestinal epithelial cell markers. GED treatment also reduced the TGF-ß and ACTA1 expression in primary human intestinal fibroblasts from ulcerative colitis patients. CONCLUSIONS: GED ameliorates intestinal fibrosis in dextran sulfate sodium-induced chronic colitis in mice and regulates major profibrotic cellular and molecular mechanisms.


Assuntos
Compostos de Anilina/farmacologia , Colite/tratamento farmacológico , Fibroblastos/efeitos dos fármacos , Fibrose/tratamento farmacológico , Inflamação/complicações , Intestinos/efeitos dos fármacos , PPAR gama/metabolismo , Fenilpropionatos/farmacologia , Animais , Western Blotting , Células Cultivadas , Colite/etiologia , Colite/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose/etiologia , Fibrose/metabolismo , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Inflamação/patologia , Mucosa Intestinal/metabolismo , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Steroids ; 103: 64-71, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25560486

RESUMO

Steroids are fundamental hormones that control a wide variety of physiological processes such as metabolism, immune functions, and sexual characteristics. Historically, steroid synthesis was considered a function restricted to the adrenals and the gonads. In the past 20 years, a significant number of studies have demonstrated that steroids could also be synthesized or metabolized by other organs. According to these studies, the intestine appears to be a major source of de novo produced glucocorticoids as well as a tissue capable of producing and metabolizing sex steroids. This finding is based on the detection of steroidogenic enzyme expression as well as the presence of bioactive steroids in both the rodent and human gut. Within the intestinal mucosa, the intestinal epithelial cell layer is one of the main cellular sources of steroids. Glucocorticoid synthesis regulation in the intestinal epithelial cells is unique in that it does not involve the classical positive regulator steroidogenic factor-1 (SF-1) but a closely related homolog, namely the liver receptor homolog-1 (LRH-1). This local production of immunoregulatory glucocorticoids contributes to intestinal homeostasis and has been linked to pathophysiology of inflammatory bowel diseases. Intestinal epithelial cells also possess the ability to metabolize sex steroids, notably estrogen; this mechanism may impact colorectal cancer development. In this review, we contextualize and discuss what is known about intestinal steroidogenesis and regulation as well as the key role these functions play both in physiological and pathological conditions.


Assuntos
Mucosa Intestinal/metabolismo , Esteroides/biossíntese , Animais , Humanos , Intestinos/citologia
6.
Gut ; 64(6): 901-10, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25053717

RESUMO

BACKGROUND AND AIMS: Immune tolerance breakdown during UC involves the peroxisome proliferator-activated receptor-γ (PPARγ), a key factor in mucosal homoeostasis and the therapeutic target of 5-aminosalycilates, which expression is impaired during UC. Here we assess the impact of glucocorticoids (GCs) on PPARγ expression, focusing especially on extra-adrenal cortisol production by colonic epithelial cells (CECs). METHODS: Activation of PPARγ in the colon was evaluated using transgenic mice for the luciferase gene under PPAR control (peroxisome proliferator response element-luciferase mice). Protein and mRNA expression of PPARγ were evaluated with colon fragments and purified CEC from mice. Cortisol production and steroidogenic factor expression were quantified in human CEC of patients with UC and those of controls. Gene expression knockdown by short hairpin RNA in Caco-2 cells was used for functional studies. RESULTS: GCs were able to raise luciferase activity in peroxisome proliferator response element-luciferase mice. In the mice colons and Caco-2 cells, PPARγ expression was increased either with GCs or with an inducer of steroidogenesis and then decreased after treatment with a steroidogenesis inhibitor. Cortisol production and steroidogenic factor expression, such as liver receptor homologue-1 (LRH-1), were decreased in CEC isolated from patients with UC, directly correlating with PPARγ impairment. Experiments on Caco-2 cells lacking LRH-1 expression confirmed that LRH-1 controls PPARγ expression by regulating GC synthesis in CEC. CONCLUSIONS: These results demonstrate cortisol control of PPARγ expression in CEC, highlighting cortisol production deficiency in colonocytes as a key molecular event in the pathophysiology of UC.


Assuntos
Colite Ulcerativa/metabolismo , Colo/metabolismo , Glucocorticoides/biossíntese , Mucosa Intestinal/metabolismo , PPAR gama/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Células CACO-2 , Colite Ulcerativa/patologia , Colo/patologia , Células Epiteliais/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Tolerância Imunológica , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Transgênicos , PPAR gama/genética , RNA Mensageiro/metabolismo
7.
Cell Rep ; 8(3): 743-53, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25066127

RESUMO

DNA methylation is a central epigenetic modification that is established by de novo DNA methyltransferases. The mechanisms underlying the generation of genomic methylation patterns are still poorly understood. Using mass spectrometry and a phosphospecific Dnmt3a antibody, we demonstrate that CK2 phosphorylates endogenous Dnmt3a at two key residues located near its PWWP domain, thereby downregulating the ability of Dnmt3a to methylate DNA. Genome-wide DNA methylation analysis shows that CK2 primarily modulates CpG methylation of several repeats, most notably of Alu SINEs. This modulation can be directly attributed to CK2-mediated phosphorylation of Dnmt3a. We also find that CK2-mediated phosphorylation is required for localization of Dnmt3a to heterochromatin. By revealing phosphorylation as a mode of regulation of de novo DNA methyltransferase function and by uncovering a mechanism for the regulation of methylation at repetitive elements, our results shed light on the origin of DNA methylation patterns.


Assuntos
Caseína Quinase II/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Processamento de Proteína Pós-Traducional , Células 3T3 , Animais , Linhagem Celular Tumoral , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/química , DNA Metiltransferase 3A , Regulação para Baixo , Heterocromatina/metabolismo , Humanos , Camundongos , Fosforilação , Estrutura Terciária de Proteína , Elementos Nucleotídeos Curtos e Dispersos
8.
J Med Chem ; 57(13): 5489-508, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-24428717

RESUMO

Since the discovery in 1995 of α-galactosylceramide 1 (α-GalCer), also known as KRN7000,1 hundreds of compounds have been synthesized in order to activate invariant natural killer T (iNKT) cells. Such keen interest for this lymphocyte cell type is due to its ability to produce different cytokines that bias the immune response toward a Th1 or Th2 profile. Thus, an understanding of the immune polarization mechanism via iNKT activation may pave the way toward new therapeutics in various domains including cancer and infectious and autoimmune diseases. In this review, we propose an up-to-date analysis of iNKT activators associated with a structure-activity relationship (SAR) study aimed at complementing available reviews by highlighting molecular bases for a selective immune response.


Assuntos
Galactosilceramidas/farmacologia , Células T Matadoras Naturais/imunologia , Animais , Antígenos CD1d/imunologia , Humanos , Ligantes , Ativação Linfocitária/imunologia , Camundongos , Modelos Moleculares , Células T Matadoras Naturais/efeitos dos fármacos , Ratos , Relação Estrutura-Atividade , Células Th1/imunologia , Células Th2/imunologia
9.
Inflamm Bowel Dis ; 19(11): 2315-25, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23949622

RESUMO

BACKGROUND: Vanin-1 is an epithelial pantetheinase, which regulates intestinal inflammation in mouse. We investigated whether human VNN1 levels could be associated to the susceptibility to inflammatory bowel diseases (IBD) and explored the participation of PPARg to these processes. METHODS: We studied VNN1 expression in colon biopsies from IBD patients. We investigated polymorphisms in the regulatory regions of the VNN1 gene and examined their genetic association with the disease. Functional relevance of these single-nucleotide polymorphisms (SNPs) was assayed, and we tested PPARg in nuclear complexes associated with specific VNN1 polymorphic sequences. In mouse, we examined Vanin-1 expression in gut and feces during dextran sulfate sodium-induced colitis and assayed the effect of PPARg on Vanin-1 regulation. RESULTS: VNN1 is expressed by enterocytes and is upregulated in IBD. Three SNPs are statistically associated to IBD. The regions containing these SNPs specifically bind nuclear complexes and are correlated with the VNN1 transcript abundance in colon in an allele-dependent manner. One rare SNP is associated to severe ulcerative colitis with strong VNN1 and dropped PPARg levels. PPARg is involved in nuclear complexes that bound to VNN1 regulatory sites. Similarly, Vanin-1 is tightly regulated in the mouse gut in normal and colitis conditions and PPARg regulates its expression. CONCLUSIONS: VNN1 is a marker for IBD. Polymorphic positions in the VNN1 locus are direct targets for nuclear factors that might regulate the level of VNN1 in colon, and this could be linked to IBD susceptibility. It is hoped that modulating locally VNN1 expression or activity can be exploited to develop future therapeutic strategies against IBD.


Assuntos
Amidoidrolases/genética , Suscetibilidade a Doenças , Doenças Inflamatórias Intestinais/genética , Polimorfismo de Nucleotídeo Único/genética , Sequências Reguladoras de Ácido Nucleico/genética , Amidoidrolases/metabolismo , Animais , Western Blotting , Estudos de Casos e Controles , Ensaio de Desvio de Mobilidade Eletroforética , Imunofluorescência , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Humanos , Técnicas Imunoenzimáticas , Doenças Inflamatórias Intestinais/patologia , Camundongos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos
10.
Carcinogenesis ; 34(11): 2580-6, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23843037

RESUMO

Epidemiological evidences suggested that 5-aminosalicylic acid (5-ASA) therapy may prevent the development of colorectal cancer in inflammatory bowel disease patients. Our aim is to investigate whether peroxisome proliferator-activated receptor-γ (PPARγ) mediates the antineoplastic effects of 5-ASA. HT-29 and Caco-2 cells were treated by 5-ASA, rosiglitazone (PPARγ ligand) or etoposide (anticarcinogenic drug). Epithelial cell growth, proliferation and apoptosis were assessed by cell count, Ki-67 staining and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, respectively. The antineoplastic effect of 5-ASA was evaluated in a xenograft tumor model in SCID mice and in azoxymethane (AOM)-induced colon carcinogenesis in A/JOlaHsd mice. The role of PPARγ was examined by administration of PPARγ antagonist, GW9662 and in PPAR knockdown cells. Compared with untreated cells, treatment of HT-29 cells by 5-ASA inhibited significantly cell growth and cell proliferation (respectively, 60% and 63%) and induced apoptosis in 75% of cells. These effects were abolished by co-treatment with GW9662 and blunted in PPAR knockdown cells. Contrarily to etoposide, similar inhibitory effects of GW9662 were obtained in HT-29 cells treated with rosiglitazone. In the xenograft model, GW9662 abolished the therapeutic effect of 5-ASA, which decreased tumor weight and volume by 80% in SCID mice compared with untreated mice. In A/JOlaHsd mice, 5-ASA suppressed colon carcinogenesis by decreasing the number of aberrant crypt foci (75%) and aberrant crypts (22%) induced by AOM treatment with an absence of 5-ASA response after GW9662 administration. In conclusion, 5-ASA exerts potent antineoplastic effects that are mediated through PPARγ. These data provide new rational for designing more effective and safe antineoplastic PPARγ ligands with topical effects.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Intestinos/efeitos dos fármacos , Mesalamina/farmacologia , PPAR gama/farmacologia , Animais , Azoximetano/toxicidade , Western Blotting , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Humanos , Técnicas Imunoenzimáticas , Mucosa Intestinal/metabolismo , Intestinos/patologia , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos SCID , PPAR gama/antagonistas & inibidores , Células Tumorais Cultivadas
11.
PLoS One ; 8(4): e62208, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23638007

RESUMO

Cigarette smoke (CS) protects against intestinal inflammation during ulcerative colitis. Immunoregulatory mechanisms sustaining this effect remain unknown. The aim of this study was to assess the effects of CS on experimental colitis and to characterize the intestinal inflammatory response at the cellular and molecular levels. Using the InExpose® System, a smoking device accurately reproducing human smoking habit, we pre-exposed C57BL/6 mice for 2 weeks to CS, and then we induced colitis by administration of dextran sodium sulfate (DSS). This system allowed us to demonstrate that CS exposure improved colonic inflammation (significant decrease in clinical score, body weight loss and weight/length colonic ratio). This improvement was associated with a significant decrease in colonic proinflammatory Th1/Th17 cytokine expression, as compared to unexposed mice (TNF (p=0.0169), IFNγ (p<0.0001), and IL-17 (p=0.0008)). Smoke exposure also induced an increased expression of IL-10 mRNA (p=0.0035) and a marked recruitment of iNKT (invariant Natural Killer T; CD45+ TCRß+ CD1d tetramer+) cells in the colon of DSS-untreated mice. Demonstration of the role of iNKT cells in CS-dependent colitis improvement was performed using two different strains of NKT cells deficient mice. Indeed, in Jα18KO and CD1dKO animals, CS exposure failed to induce significant regulation of DSS-induced colitis both at the clinical and molecular levels. Thus, our study demonstrates that iNKT cells are pivotal actors in the CS-dependent protection of the colon. These results highlight the role of intestinal iNKT lymphocytes and their responsiveness to environmental stimuli. Targeting iNKT cells would represent a new therapeutic way for inflammatory bowel diseases.


Assuntos
Colite/imunologia , Células T Matadoras Naturais/citologia , Células T Matadoras Naturais/imunologia , Nicotiana/química , Fumaça , Animais , Contagem de Células , Colite/induzido quimicamente , Colite/metabolismo , Citocinas/metabolismo , Sulfato de Dextrana/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células T Matadoras Naturais/efeitos dos fármacos
12.
Nutrition ; 28(2): 113-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22208553

RESUMO

The etiology of inflammatory bowel disease and, in particular, Crohn's disease involves a deregulated mucosal immune system under the influence of intestinal flora and environmental factors in genetically susceptible individuals. A new hypothesis has focused on mesenteric fat hypertrophy and the presence of ectopic fat surrounding inflamed bowel, the so-called creeping fat, which are hallmarks of Crohn's disease. Mesenteric adipose tissue is currently recognized as an active actor in immunity with a capacity for mediator secretion. These mediators include classic pro- and anti-inflammatory cytokines or chemokines and hormone-like adipokines with multiple effects. Mesenteric fat participates in the course of Crohn's disease and may play an active role in the regulation of intestinal inflammation. However, little is known about the origin and role of mesenteric fat in Crohn's disease, essentially because of a lack of experimental models that develop creeping fat. The purpose of this review is to present the recent data describing the immune properties of mesenteric fat and the recent advances in animal models, which have suggested a new hypothesis about the role of creeping fat in Crohn's disease.


Assuntos
Doença de Crohn/etiologia , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/imunologia , Gordura Intra-Abdominal/imunologia , Coristoma/imunologia , Doença de Crohn/imunologia , Humanos , Mesentério/imunologia
13.
Curr Opin Clin Nutr Metab Care ; 13(5): 574-80, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20625283

RESUMO

PURPOSE OF REVIEW: Increasing evidence indicates that adipose tissue is an active endocrine organ involved in metabolic syndrome and regulation of inflammation. Visceral fat accumulation is a hallmark of both obesity and Crohn's disease. Here, we present recent data describing the immune properties of intra-abdominal adipose tissue that could link the innate immune response to obesity-related disorders and gut inflammation. RECENT FINDINGS: Innate immune properties of adipocytes have become well characterized since recent studies described the Toll-like receptor (TLR) expression repertoire and specific TLR ligand responses of adipocytes. Adipokine secretion profiles have also been elucidated both in obese patients, when they may be involved in obesity-associated metabolic disease, and in Crohn's disease. Whereas mesenteric fat hypertrophy and fat wrapping of the bowel are characteristic of Crohn's disease, there exists a paucity of information concerning this important pathophysiological aspect. Our current classical animal models are of limited interest when investigating the role of mesenteric fat in gut inflammation. Recent new alternative disease paradigms could help to design more specific models for elucidating chronic transmural inflammation of the gut. SUMMARY: Obesity and Crohn's disease share common features with the development of mesenteric fat that may be involved in gut inflammation. Further studies are required to clearly assess the origin and influence of intestinal fat deposits upon gut inflammation, notably during Crohn's disease development.


Assuntos
Adipócitos/imunologia , Doença de Crohn/imunologia , Trato Gastrointestinal/imunologia , Imunidade Inata , Gordura Intra-Abdominal/imunologia , Obesidade/imunologia , Adipocinas/metabolismo , Animais , Doença de Crohn/patologia , Trato Gastrointestinal/patologia , Humanos , Gordura Intra-Abdominal/patologia , Receptores Toll-Like/metabolismo
14.
Biochem Biophys Res Commun ; 377(4): 1079-84, 2008 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18977200

RESUMO

Histone deacetylases (HDAC) form a conserved enzyme family that control gene expression via the removal of acetyl residues from histones and other proteins and are under increasing investigation as therapeutic targets, notably in cancer and parasitic diseases. To investigate the conservation of these enzymes in the platyhelminth parasite Schistosoma mansoni, we cloned and characterized three class I HDACs, orthologues of mammalian HDAC1, 3 and 8, and confirmed their identities by phylogenetic analysis. The identification of an HDAC8 orthologue showed that it is not vertebrate-specific as previously thought and insertions in its catalytic domain suggest specific enzymatic properties. SmHDAC1, 3, and 8 mRNAs are expressed at all schistosome life-cycle stages. SmHDAC1 repressed transcriptional activity in a mammalian cell line and this activity was dependent on its catalytic activity since transcription was partially restored by treatment with trichostatin A and a catalytic site mutant failed to repress transcription.


Assuntos
Regulação da Expressão Gênica , Proteínas de Helminto/classificação , Proteínas de Helminto/metabolismo , Histona Desacetilases/classificação , Histona Desacetilases/metabolismo , Schistosoma mansoni/enzimologia , Sequência de Aminoácidos , Animais , Domínio Catalítico/genética , Clonagem Molecular , Sequência Conservada , Inibidores Enzimáticos/farmacologia , Proteínas de Helminto/genética , Histona Desacetilases/genética , Ácidos Hidroxâmicos/farmacologia , Dados de Sequência Molecular , Filogenia , Schistosoma mansoni/genética , Transcrição Gênica
15.
Biochem J ; 382(Pt 1): 337-51, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15104535

RESUMO

SmFtz-F1 (Schistosoma mansoni Fushi Tarazu-Factor 1) belongs to the Ftz-F1 subfamily of nuclear receptors, but displays marked structural differences compared with its mammalian homologues SF-1 (steroidogenic factor-1) or liver receptor homologue-1. These include a long F domain (104 amino acids), an unusually large hinge region (133 amino acids) and a poorly conserved E-domain. Here, using Gal4 constructs and a mammalian two-hybrid assay, we have characterized the roles of these specific regions both in the transcriptional activity of the receptor and in its interactions with cofactors. Our results have shown that, although the AF-2 (activation function-2) region is the major activation function of the receptor, both the F and D domains are essential for AF-2-dependent activity. Modelling of SmFtz-F1 LBD (ligand-binding domain) and structure-guided mutagenesis allowed us to show the important role of helix H1 in maintaining the structural conformation of the LBD, and suggested that its autonomous transactivation activity, also observed with SF-1, is fortuitous. This strategy also allowed us to study an eventual ligand-dependence for this orphan receptor, the predicted three-dimensional models suggesting that the SmFtz-F1 LBD contains a large and well-defined ligand-binding pocket sealed by two arginine residues orientated towards the interior of the cavity. Mutation of these two residues provoked a loss of transcriptional activity of the receptor, and strongly reduced its interaction with SRC1 (steroid receptor cofactor-1), suggesting a ligand-dependent activity for SmFtz-F1. Taken together, our results argue for original and specific functional activities for this platyhelminth nuclear receptor.


Assuntos
Proteínas de Ligação a DNA/química , Peptídeos/fisiologia , Schistosoma mansoni/química , Fatores de Transcrição/química , Animais , Linhagem Celular , Chlorocebus aethiops , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Fatores de Transcrição Fushi Tarazu , Proteínas de Helminto/química , Proteínas de Helminto/metabolismo , Proteínas de Helminto/fisiologia , Histona Acetiltransferases , Humanos , Proteínas de Insetos , Rim/química , Rim/citologia , Rim/metabolismo , Ligantes , Modelos Genéticos , Modelos Moleculares , Coativador 1 de Receptor Nuclear , Peptídeos/metabolismo , Ligação Proteica/fisiologia , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Fator Esteroidogênico 1 , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Ativação Transcricional/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA