Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 35(13): 1671-8, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-26096932

RESUMO

The α1ß1 collagen receptor is only present in a few epithelial cell types. In the intestine, it is specifically expressed in proliferating crypt cells. This integrin has been reported to be involved in various cancers where it mediates the downstream activation of the Ras/ERK proliferative pathway. We have recently shown that integrin α1ß1 is present in two-thirds of colon adenocarcinomas, but the mechanism by which ITGA1 expression is regulated is not known. DNA methylation, involved in ITGA1 repression during megakaryocyte differentiation, is not the mechanism of ITGA1 regulation in colorectal cancer cells. Our in silico analysis of the ITGA1 promoter revealed two response elements for MYC, an oncogenic factor known to regulate cancer cell proliferation, invasion and migration. In situ, the expressions of both MYC and ITGA1 are localized in the lower crypt of the normal colon and correlate in 72% of the 65 analyzed colorectal cancers. MYC pharmacological inhibition or downregulation of expression with short hairpin RNA in HT29, T84 and SW480 cells resulted in reduced ITGA1 expression at both the transcript and protein levels. Chromatin immunoprecipitation assays revealed that MYC was bound to the chromatin region of the ITGA1 proximal promoter, whereas MYC overexpression enhanced ITGA1 promoter activity that was reduced with MAD co-transfection or by the disruption of the response elements. We concluded that MYC is a key regulating factor for the control of ITGA1 expression.


Assuntos
Neoplasias Colorretais/genética , Integrina alfa1beta1/genética , Proteínas Proto-Oncogênicas c-myc/fisiologia , Células CACO-2 , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HT29 , Humanos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/metabolismo
2.
Genes Immun ; 13(7): 556-65, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22914433

RESUMO

Hepatocyte nuclear 4 alpha (HNF4α), involved in glucose and lipid metabolism, has been linked to intestinal inflammation and abnormal mucosal permeability. Moreover, in a genome-wide association study, the HNF4A locus has been associated with ulcerative colitis. The objective of our study was to evaluate the association between HNF4α genetic variants and Crohn's disease (CD) in two distinct Canadian pediatric cohorts. The sequencing of the HNF4A gene in 40 French Canadian patients led to the identification of 27 single nucleotide polymorphism (SNP)s with a minor allele frequency >5%. To assess the impact of these SNPs on disease susceptibility, we first conducted a case-control discovery study on 358 subjects with CD and 542 controls. We then carried out a replication study in a separate cohort of 416 cases and 1208 controls. In the discovery cohort, the genotyping of the identified SNPs revealed that six were significantly associated with CD. Among them, rs1884613 was replicated in the second CD cohort (odds ratio (OR): 1.33; P<0.012) and this association remained significant when both cohorts were combined and after correction for multiple testing (OR: 1.39; P<0.004). An 8-marker P2 promoter haplotype containing rs1884613 was also found associated with CD (P<2.09 × 10(-4) for combined cohorts). This is the first report showing that the HNF4A locus may be a common genetic determinant of childhood-onset CD. These findings highlight the importance of the intestinal epithelium and oxidative protection in the pathogenesis of CD.


Assuntos
Doença de Crohn/genética , Fator 4 Nuclear de Hepatócito/genética , Polimorfismo de Nucleotídeo Único , Adolescente , Canadá/epidemiologia , Estudos de Casos e Controles , Criança , Frequência do Gene , Estudos de Associação Genética , Haplótipos , Humanos , Regiões Promotoras Genéticas
3.
J Cell Physiol ; 209(2): 241-52, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16897777

RESUMO

With the discovery of CARD15 as susceptibility gene for Crohn's disease (CD) a first link to a potential defect in the innate immune system was made. In this work we aimed to analyze enterocyte NOD2/CARD15 expression and regulation in response to bacterial motifs and the consequences of the most common CD-specific CARD15 mutation on antibacterial responses of normal intestinal epithelial cells (IEC). Under normal conditions, IEC lines and ileal enterocytes did not express NOD2/CARD15 mRNA or protein, contrary to IEC derived from inflammatory CD sections. In vitro analyses revealed that the simple contact with non-pathogenic commensal E. Coli K12 was sufficient to induced NOD2/CARD15 mRNA and protein in human IEC (HIEC). We identified bacterial flagellin interacting with TLR5 as major motif in this regulation of NOD2/CARD15. E. Coli mutants not expressing flagellin (DeltaFliC) failed to induce CARD15. Similarly, in HIEC transfected with a plasmid encoding dominant negative TLR5, no CARD15 induction was observed after K12 contact. Isolated TLR2 or TLR4 stimulation had no or only a marginal effect on NOD2/CARD15 expression. NOD2/CARD15 negative HIEC were unresponsive to muramyl dipeptide (MDP), but once NOD2/CARD15 was induced, HIEC and Caco2 cells responded to intra or extracellular MDP presentation with the activation of the NFkB pathway. IEC transfected with the Crohn-specific CARD15 mutant (F3020insC, FS) failed to activate NFkB after MDP-challenge, in contrast to CARD15WT IEC. In response to MDP, IEC induced a massive antibacterial peptide (ABP) response, seen in the apical release of CCL20. This was completely abolished in IEC carrying CARD15FS. These data suggest a critical role of NOD2/CARD15 in the bacterial clearance of the intestinal epithelium while CD-specific mutated NOD2/CARD15 causes an impaired epithelial barrier.


Assuntos
Enterócitos/metabolismo , Enterócitos/microbiologia , Escherichia coli/metabolismo , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptor 5 Toll-Like/metabolismo , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Motivos de Aminoácidos , Peptídeos Catiônicos Antimicrobianos/metabolismo , Células CACO-2 , Células Cultivadas , Enterócitos/citologia , Células HT29 , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação/genética , Proteína Adaptadora de Sinalização NOD2 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
Biochem Biophys Res Commun ; 337(2): 505-9, 2005 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-16198312

RESUMO

Upregulation of the T-cell derived cytokine interleukin (IL-17) was reported in the inflamed intestinal mucosa of patients with inflammatory bowel disorders. In this study, we analyzed the effect of IL-17 on human intestinal epithelial cell (HIEC) turnover and functions. Proliferation and apoptosis in response to IL-17 was monitored in HIEC (cell counts, [(3)H]thymidine incorporation method, and annexinV-PI-apoptosis assay). Signalling pathways were analyzed by Western blots, electromobility shift assay, and immunofluorescence studies. IL-17 proved to be a potent inhibitor of HIEC proliferation without any pro-apoptotic/necrotic effect. The growth inhibitory effect of IL-17 was mediated via the p38 stress kinase. Consequently, the p38-SAPkinase-inhibitor SB203580 abrogated this anti-mitotic effect. In parallel, IL-17 provoked the degradation of IkappaBalpha, allowing nuclear translocation of the p65 NF-kappaB subunit and induction of the NF-kappaB-controlled genes IL-6 and -8. IL-17 potently blocks epithelial cell turnover while at the same time amplifying an inflammatory response in a positive feedback manner.


Assuntos
Apoptose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Interleucina-17/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Mitose/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Células Epiteliais/metabolismo , Humanos , Mediadores da Inflamação/farmacologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , NF-kappa B/metabolismo , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
5.
J Cell Physiol ; 203(1): 15-26, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15389642

RESUMO

Claudins, and particularly claudin-2, are important regulatory components of tight junction permeability. A better understanding of the involvement of claudin-2 in intestinal barrier functions requires the characterization of its distribution and regulation in the intestine. Interestingly, the claudin-2 gene promoter harbors a number of similarities to that of sucrase-isomaltase, a marker of enterocyte differentiation. We thus investigated the expression of claudin-2 in relation to the transcription factors CDX2, HNF-1alpha, and GATA-4 in the human intestine. The characterization of claudin-2 and the expression of the above transcription factors were performed by immunofluorescence, Western blot, and RT-PCR in the developing human intestinal epithelium. The functional role of CDX2, HNF-1alpha, and GATA-4 on claudin-2 regulation was also examined by ectopic expression studies in intestinal cell models. Claudin-2 was detected in both crypt and villus cells of the small intestine but restricted to undifferentiated crypt cells in the colon. CDX2 and HNF-1alpha were expressed along the entire intestine whereas GATA-4 was undetectable in the colon. Accordingly, in the colonic Caco-2 cell model, claudin-2 was found to be present only in undifferentiated cells. Like in the colonic epithelium, GATA-4 was found to be also lacking in Caco-2 cells while CDX2 and HNF-1alpha were present at significant levels. Cotransfection experiments showed that the claudin-2 promoter was activated by CDX2, HNF-1alpha, and GATA-4 in a cooperative manner. Furthermore, forced GATA-4 expression in Caco-2 cells enhances maintenance of claudin-2 expression during differentiation. These observations suggest that optimal claudin-2 expression in the gut relies on the presence of GATA-4, suggesting a role for this factor in intestinal regionalization.


Assuntos
Proteínas de Ligação a DNA/genética , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Proteínas de Membrana/metabolismo , Fatores de Transcrição/genética , Fator de Transcrição CDX2 , Células CACO-2 , Diferenciação Celular/fisiologia , Claudinas , Colo/citologia , Colo/fisiologia , Proteínas de Ligação a DNA/metabolismo , Fator de Transcrição GATA4 , Regulação da Expressão Gênica/fisiologia , Fator 1 Nuclear de Hepatócito , Fator 1-alfa Nuclear de Hepatócito , Proteínas de Homeodomínio/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Intestino Delgado/citologia , Intestino Delgado/fisiologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/fisiologia , Junções Íntimas/fisiologia , Fatores de Transcrição/metabolismo
6.
Gut ; 51(6): 842-8, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12427787

RESUMO

BACKGROUND: Circulating levels of endotoxin (or lipopolysaccharide (LPS)) and anti-endotoxin antibodies are increased in patients with inflammatory bowel disease, supporting the hypothesis of a role for endogenous bacterial products in the pathogenesis of these disorders. AIM: The aim of this study was to analyse the direct effects of LPS on intestinal epithelial cell turnover. METHODS AND RESULTS: LPS significantly inhibited growth of the human non-transformed immature crypt cell line (HIEC), whereas IEC-6 cell proliferation was stimulated by LPS. As LPS is a physiological inducer of tumour necrosis factor alpha (TNFalpha) in various cell systems and this cytokine exerted similar anti-proliferative (HIEC) or growth stimulatory (IEC-6 cells) effects, the study thus tested the hypothesis that endogenously produced TNFalpha in response to LPS mediates this growth modulatory effect in an autoparacrine/paracrine way. Therefore, during LPS stimulation, the biological activity of TNFalpha was blocked using neutralising anti-TNFalpha antibodies, as well as inhibitory, antagonistic antibodies directed against the p55 TNF receptor, signalling the antimitotic TNFalpha effect in HIEC. Both experimental approaches completely abolished the growth modulatory effects of LPS in HIEC/IEC-6 cells. Production and secretion of TNFalpha by HIEC/IEC-6 cells in response to LPS was confirmed on mRNA and protein level by reverse transcription polymerase chain reaction (RT-PCR) and enzyme linked immunosorbent assay. LPS signalling was independent of CD14 in HIEC, as these cells lack this receptor. However, HIEC expressed TLR4 and MD2 resulting in a fully functional signalling complex as demonstrated by RT-PCR, western blot, and immunofluorescence analyses. CONCLUSION: These results support the hypothesis that LPS induced changes of intestinal epithelial cell turnover may directly contribute to the pathogenesis of inflammatory epithelial cell lesions by endogenous TNFalpha production by enterocytes.


Assuntos
Proteínas de Drosophila , Enterócitos/imunologia , Doenças Inflamatórias Intestinais/imunologia , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/imunologia , Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Enterócitos/efeitos dos fármacos , Enterócitos/patologia , Expressão Gênica/efeitos dos fármacos , Humanos , Doenças Inflamatórias Intestinais/patologia , Intestino Delgado , Receptores de Lipopolissacarídeos/análise , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Glicoproteínas de Membrana/análise , RNA Mensageiro/análise , Receptores de Superfície Celular/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 4 Toll-Like , Receptores Toll-Like , Fator de Necrose Tumoral alfa/genética
7.
Biochem Biophys Res Commun ; 290(4): 1308-14, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11812006

RESUMO

Various members of the tumor necrosis factor receptor superfamily are implicated in the regulation of enterocyte apoptosis. Previously, we observed that human intestinal epithelial cells are rather unsusceptible to Fas-induced apoptosis. In the present study we analyzed these protective mechanisms in the human intestinal epithelial cell line HIEC, focusing on antiapoptotic molecules of the IAP family which block apoptosis at the level of the caspase cascade. HIEC expressed all key molecules required to trigger Fas-induced apoptosis. However, no apoptosis occurred after activation of Fas, whereas an upregulation of antiapoptotic cIAP1 and 2 was observed. Suppression of this upregulation with the proteasome inhibitor MG132 or the protein synthesis inhibitor cycloheximide highly sensitized HIEC toward Fas-induced apoptosis. Western blot analyses revealed that both inhibitors potently suppressed endogenously produced cIAP1 and 2. No effect was observed on XIAP expression. These data indicate that enterocytes are particularly protected against Fas-induced apoptosis on the level of executionary caspases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose/fisiologia , Enterócitos/citologia , Enterócitos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas/metabolismo , Receptor fas/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/genética , Caspases/metabolismo , Linhagem Celular , Cicloeximida/farmacologia , Regulação para Baixo , Proteína de Domínio de Morte Associada a Fas , Humanos , Proteínas Inibidoras de Apoptose , Leupeptinas/farmacologia , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ubiquitina-Proteína Ligases , Regulação para Cima , Receptor fas/genética
8.
J Cell Biochem ; 81(3): 463-76, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11255229

RESUMO

SPARC is a glycoprotein of the extracellular matrix that exhibits a number of biological functions such as disruption of cell adhesion and modulation of matrix metalloprotease expression. These properties, in concert with the expression of the molecule during development, repair, and neoplastic progression, suggest that SPARC has an important role in remodeling in a variety of tissues. However, the role of SPARC in the intestine is unclear since the development expression and tissular origin of SPARC in this organ appears to be species-dependent. As a first step to investigate the function of SPARC in the tissues of the intestine, we have analyzed its expression at the protein and mRNA levels in the human fetal and adult small intestinal and colonic mucosa as well as in intestinal cell models. Our results show that SPARC expression is differentially regulated during development and along the length of the human intestine. In the colon, SPARC was predominantly found at the epithelial-mesenchymal interface at the fetal stage, below detection levels in the normal adult, but re-expressed in the stroma of colonic tumors. In the small intestine, low levels of SPARC expression were observed at an early stage of morphogenesis (between 9 and 11 weeks) but expression was not detected at subsequent developmental stages nor was it induced in the mucosa of Crohn's disease. While SPARC appeared to be produced mainly by mesenchymal and stromal cells in the intact intestine it was not detected in colon cancer cells. Taken together, these results indicate that SPARC is subject to an onco-fetal pattern of expression in the stroma of the colonic mucosa while its expression is much more restricted in the small intestine, suggesting a differential involvement of this molecule in the extracellular matrix remodeling occurring along the length of the developing and diseased human intestinal mucosa.


Assuntos
Mucosa Intestinal/metabolismo , Osteonectina/metabolismo , Células Estromais/fisiologia , Sequência de Bases , Linhagem Celular , Primers do DNA , Imunofluorescência , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/citologia , Osteonectina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tenascina/metabolismo
9.
Gastroenterology ; 120(2): 423-38, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11159883

RESUMO

BACKGROUND & AIMS: Growth arrest and differentiation are generally considered to be temporally and functionally linked phenomena in the intestinal epithelium. METHODS: To delineate the mechanism(s) responsible for the loss of proliferative potential as committed intestinal cells start to differentiate, we have analyzed the regulation of G(1)-phase regulatory proteins in relation to differentiation in the intact epithelium as well as in well-established intestinal cell models that allow the recapitulation of the crypt-villus axis in vitro. RESULTS: With intestinal cell differentiation, we have observed an induction of the cell cycle inhibitors p21(Cip), p27(Kip1), and p57(Kip2) expression with an increased association of p27(Kip1) and p57(Kip2) with cyclin-dependent kinase 2 (Cdk2). At the same time, there was an accumulation of the hypophosphorylated form of the pRb proteins and a strong decline in Cdk2 activity. Stable expression of a p27(Kip1) antisense complementary DNA in Caco-2/15 cells did not prevent growth arrest induced by confluence, but repressed villin, sucrase-isomaltase, and alkaline phosphatase expression. CONCLUSIONS: Our results indicate that the growth arrest that precedes differentiation involves the activation of Rb proteins and the inhibition of Cdk2. Furthermore, intestinal cell differentiation apparently requires a function of p27(Kip1) other than that which leads to inhibition of Cdks.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Mucosa Intestinal/citologia , Mucosa Intestinal/enzimologia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Proto-Oncogênicas , Proteínas Supressoras de Tumor , Elementos Antissenso (Genética) , Células CACO-2 , Diferenciação Celular/fisiologia , Ciclina D , Ciclina D1/metabolismo , Ciclina E/genética , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Inibidor de Quinase Dependente de Ciclina p57 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Feto/citologia , Fase G1/fisiologia , Humanos , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteína do Retinoblastoma/metabolismo , Complexo Sacarase-Isomaltase/genética
10.
Expert Rev Mol Med ; 3(24): 1-18, 2001 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-14585148

RESUMO

Laminins are a multigene family of extracellular matrix molecules. Quantitatively, they are one of the most abundant glycoproteins present in basement membranes. Functionally, they can modulate several key biological activities, including cell adhesion and migration, gene expression and cell survival. Variability in the spatial and temporal expression of laminins, as well as of their specific receptors of the integrin family, in various tissues and organs, suggests that different laminins perform distinct functions. This article focuses on the human intestinal epithelium as a paradigm to illustrate the potential relationship between laminin-cell interactions and the cell state. This rapidly renewing epithelium consists of spatially separated proliferative and differentiated cell populations located in the crypts and on the villi, respectively. Differential distributions of the various laminins and laminin-binding integrins have been observed along the crypt-villus axis in both the developing and the adult intestine, and important alterations in the pattern of laminin expression have been reported in various intestinal pathologies, such as tufting enteropathy, Crohn's disease and ulcerative colitis, and colorectal cancer. More-direct approaches, including experimentation with in vitro and in vivo models, have provided evidence in support of a role for laminins in intestinal cell functions. Although further work is still needed, laminins emerge more and more as key regulators of specific cell functions important in both intestinal health and intestinal disease.

11.
Microsc Res Tech ; 51(2): 169-78, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11054867

RESUMO

The intestinal epithelium is a highly dynamic tissue, which depends on a variety of factors for the regulation of its rapid renewal and expression of digestive functions. Over the last 10 years, it has become evident that among these factors are cell interactions with the extracellular matrix, more specifically with the underlying basement membrane, through a series of specific cell membrane receptors, many of which are integrins. Integrins regulate the assembly of adhesive junctions as well as the activation of various signaling pathways, leading to the modulation of gene expression. The analysis of the integrin repertoire along the crypt-villus axis in the human small intestinal epithelium identifies a number of beta1 and beta4 integrins, showing differential patterns of expression relative to its two functional compartments. Among them are the integrins alpha3beta1, alpha7Bbeta1 and the functional form of alpha6beta4 that appear to be related, in concert with the distribution of their ligands, to the process of intestinal cell differentiation, and the integrins alpha2beta1, alpha1beta1, alpha5beta1, and the non-functional form of alpha6beta4 that seem to be coupled with the undifferentiated/proliferative status of crypt cells. These observations delineate the potential complexity of the organization of epithelial cell-matrix interactions involved in the maintenance of the human intestinal crypt-villus axis.


Assuntos
Integrinas/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Membrana Basal/metabolismo , Matriz Extracelular/metabolismo , Humanos , Imuno-Histoquímica , Integrinas/análise
12.
Anat Embryol (Berl) ; 202(3): 223-33, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10994995

RESUMO

Laminins may play important roles in gastric gland development due to the differential localization of their alpha chains in human fetal fundic (oxyntic) mucosa. To extend this hypothesis, the current investigation was undertaken to compare the anatomical and cellular distribution of epithelial integrin subunits with those of laminin alpha chains in the human stomach at different ages (8-22 weeks of gestation) using indirect immunofluorescence. In the body, fundus and antrum regions, the beta1 and alpha6 subunits were associated with the entire epithelium at all developmental stages in the same way as laminin chains (alpha1/alpha5) detected with 4C7 monoclonal antibody. By contrast, the alpha3 and beta4 subunits of alpha3beta1 and alpha6beta4 integrins together with the laminin alpha3-chain were concentrated in the surface and foveolus compartments composed of differentiating mucous cells. Most importantly, the alpha2 integrin subunit was expressed in a rather complex pattern: (1) it was located at the base and at cell-cell boundaries of surface/foveolar epithelium, (2) was specifically repressed in differentiating parietal cells, and (3) its expression increased in maturing glands, where it became concentrated at the basal pole of epithelial cells simultaneously exhibiting a strong reactivity for laminin-2 (alpha2-chain). Taken altogether, our observations provide new evidence for the implication of different laminins and their receptors in the development of all human gastric epithelial lineages, surface mucous cells or glandular cells. The coordinated expression of alpha2 and alpha3 integrin subunits as well as the cellular re-distribution of alpha2beta1 integrin likely represent key events for the differentiation of glandular secretory cell types, especially maturing chief cells responsible for the synthesis/secretion of gastric digestive enzymes in fundic-type glands.


Assuntos
Diferenciação Celular , Células Epiteliais/química , Células Epiteliais/citologia , Mucosa Gástrica/embriologia , Cadeias alfa de Integrinas , Integrinas/análise , Antígenos CD/análise , Técnica Indireta de Fluorescência para Anticorpo , Mucosa Gástrica/química , Mucosa Gástrica/citologia , Idade Gestacional , Humanos , Integrina alfa2 , Integrina alfa3 , Integrina alfa5 , Integrina alfaV , Integrina beta1/análise , Integrina beta4 , Laminina/análise
13.
Microsc Res Tech ; 49(4): 394-406, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10820523

RESUMO

The intestinal epithelium is continuously and rapidly renewed by a process involving cell generation, migration, and differentiation, from the stem cell population located at the bottom of the crypt to the extrusion of the terminally differentiated cells at the tip of the villus. Because of the lack of normal human intestinal cell models, most of our knowledge about the regulation of human intestinal cell functions has been derived from studies conducted on cell cultures generated from experimental animals and human colon cancers. However, important advances have been achieved over recent years in the generation of normal human intestinal cell models. These models include (a) intestinal cell lines with typical crypt cell proliferative noncommitted characteristics, (b) conditionally immortalized intestinal cell lines that can be induced to differentiate, and (c) primary cultures of differentiated villuslike cells that can be maintained in culture for up to 10 days. Each of these models should help in the investigation of the specific aspects of human intestinal function and regulation. Furthermore, taken together, these models provide an integrated system that allows an in vitro recapitulation of the entire crypt-villus axis of the normal human small intestine.


Assuntos
Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Intestino Delgado/citologia , Intestino Delgado/fisiologia , Diferenciação Celular , Linhagem Celular , Humanos , Microscopia Eletrônica , Microvilosidades/ultraestrutura
14.
Histol Histopathol ; 15(2): 577-85, 2000 04.
Artigo em Inglês | MEDLINE | ID: mdl-10809379

RESUMO

The tenascins are a family of multifunctional extracellular matrix glycoproteins subject to complex spatial and temporal patterns of expression in the course of various organogenetic processes, namely those involving epithelial-mesenchymal interactions. In the intestine, the tenascins, in particular tenascin-C, have been found to be differentially expressed in the developing and adult small intestinal and colonic mucosa as well as in neoplasm. While tenascin-C emerges as a key player likely to be involved in intestinal mucosa development, maintenance and disease, its exact role in the regulation of fundamental intestinal cell function(s) such as proliferation, migration and tissue-specific gene expression remains however to be established.


Assuntos
Intestinos/crescimento & desenvolvimento , Tenascina/fisiologia , Adulto , Animais , Humanos , Tenascina/genética , Tenascina/metabolismo , Distribuição Tecidual
15.
J Lipid Res ; 41(1): 12-22, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10627497

RESUMO

The recent availability of spontaneously proliferating, non-transformed human crypt intestinal epithelial cells (HIEC) affords an opportunity to investigate lipid metabolism in undifferentiated enterocytes. The major purpose of this study was to explore the capability of undifferentiated crypt cells to synthesize, assemble, and secrete lipids and apolipoproteins. HIEC were cultured in medium with 5% fetal bovine serum for 5 to 21 d. The cells were clearly able to incorporate [(14)C]oleic acid (dpm/mg protein) into triglycerides (128,279 +/- 16,988), phospholipids (30, 278 +/- 2,107), and cholesteryl esters (2,180 +/- 207). Although improvement in lipid secretion was noted with prolongation of cell culture periods, low efficiency of lipid export (10.3 +/- 2.2% of intracellular content) characterized the HIEC. All phospholipid classes were elaborated, with phosphatidylcholine accounting for 79. 3 +/- 1.3% of cellular phospholipids. Chylomicrons were the dominant (46.4%) lipoproteins secreted, followed by high, low, and very low density lipoproteins (HDL, LDL, and VLDL) comprising 22.5, 20.2, and 10.8% of the total, respectively. HIEC elaborated most of the major apolipoprotein (apo) classes (A-I, A-IV, B-100, C, and E), but were less efficient in producing apoB-48. In contrast to the production of apoA-I and C as early as 5 days after confluence, apoA-I and A-IV were maximally expressed at 11 d. Culture media accumulated much more apoB-100 than apoB-48 (B-48/B-100 ratio 0.21 +/- 0.03), reflecting limited apoB mRNA editing. HIEC demonstrated both endogenous cholesterol synthesis and LDL receptor expression. Cholesterol synthesis was sensitive to 25-hydroxycholesterol and mevinolin, but unresponsive to LDL treatment, suggesting independent regulation pathways. In contrast, LDL inhibited receptor activity. The present findings provide the first solid evidence that immature HIEC are capable of key fat absorptive functions of well-differentiated enterocytes. The intracellular mechanisms required for lipid and apolipoprotein synthesis as well as for lipoprotein assembly are already present in intestinal crypt cells. These cells also retain the capacity for sterol enzyme and receptor expression. However, certain limitations, especially apoB-48 production and lipoprotein secretion as well as unresponsiveness of cholesterol synthesis to LDL, may be ascribed to the lack of differentiation.


Assuntos
Apolipoproteínas/biossíntese , Mucosa Intestinal/metabolismo , Lipídeos/biossíntese , Animais , Transporte Biológico , Radioisótopos de Carbono , Bovinos , Colesterol/biossíntese , Cromatografia Líquida , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Mucosa Intestinal/citologia , Microscopia Eletrônica , Ácido Oleico/metabolismo , Receptores de LDL/metabolismo
16.
Am J Pathol ; 156(1): 45-50, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10623652

RESUMO

Laminins are a large family of heterotrimeric basement membrane molecules that mediate crucial cell functions such as adhesion, proliferation, migration, and differentiation. Up to now, three distinct laminins have been identified in the normal human small intestinal epithelium. Laminin-1 (alpha1beta1gamma1) and laminin-5 (alpha3beta3gamma2) are mainly expressed at the base of villus cells, whereas laminin-2 (alpha2beta1gamma1) is restricted to the bottom of the crypts. The expression of these molecules has not yet been studied in Crohn's disease (CD), but it could be altered, in light of the important changes occurring in the architecture of the crypt-villus axis under the active state of the disease. To test this hypothesis, the expression of laminin alpha1, alpha2, and alpha3 subunits was analyzed in control, inflamed, and corresponding uninflamed CD small intestinal specimens by indirect immunofluorescence and reverse transcriptase-polymerase chain reaction. Surprisingly, alpha1 and alpha3 remained strongly expressed by all villus cells, whereas alpha2, normally expressed in the bottom of the crypts in control and uninflamed CD specimens, was lacking in inflamed CD specimens. However, this loss of alpha2 expression was associated with a significant up-regulation of both alpha1 and alpha3 expression in the crypts of inflamed CD specimens. A significant up-regulation of the alpha1 subunit was also observed in the crypts of uninflamed CD specimens. At the transcript levels, alpha1 was found significantly higher in inflamed than uninflamed CD specimens. Taken together, these observations identify important alterations in laminin expression in the small intestine with CD and suggest that compositional changes in the epithelial basement membrane may play a role in this disease.


Assuntos
Doença de Crohn/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Laminina/metabolismo , Adulto , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Laminina/genética , Masculino , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual
17.
Exp Cell Res ; 253(2): 493-502, 1999 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-10585272

RESUMO

The gastric pit-gland unit is a highly dynamic and compartimentalized structure which assumes important key functions such as acid secretion, digestion of dietary proteins and triglycerides, protection, and epithelial restitution following injury. However, in vitro models representative of the intact gastric epithelium are still lacking. The current study was undertaken to investigate the possibility of generating such primary cultures from human fetal stomach. The use of Matrisperse, a nonenzymatic solution, allowed complete dissociation of the epithelial layer and the maintenance for at least 7 days of all gastric epithelial cell types in primary culture on plastic. Indirect immunofluorescence and Western blot analyses confirmed the purity of epithelial cultures, composed of 60% mucus-secreting cells, 25% zymogenic chief cells, 5% parietal cells, and a small proportion of mitotic precursors. Their functionality was demonstrated by the presence of zonulae occludens and adherens at cell to cell contacts, [(3)H]thymidine incorporation, Periodic acid Schiff staining, and expression of growth factor receptors (EGF/TGFalpha, IGF1, HGF, KGF), gastric H(+)/K(+)-ATPase, pepsinogen (Pg5), and human gastric lipase (HGL). Chief cells were able to produce and secrete both Pg5 and HGL and to respond to EGF treatment. In conclusion, we developed a new primary culture system of human gastric epithelium characterized for the first time by the absence of added matrix and the maintenance of functional chief cells. It represents an experimental breakthrough that will serve applications in investigating the actions of hormones, mesenchymal growth factors, and basement membrane proteins on human gastric functions in vitro.


Assuntos
Técnicas de Cultura de Células/métodos , Celulas Principais Gástricas/citologia , Celulas Principais Gástricas/enzimologia , Adulto , Biomarcadores , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Células Cultivadas , Celulas Principais Gástricas/química , Fator de Crescimento Epidérmico/farmacologia , Feto/citologia , Regulação Enzimológica da Expressão Gênica , Humanos , Cinética , Lipase/análise , Lipase/genética , Mesoderma/citologia , Pepsinogênio A/análise , Pepsinogênio A/genética , RNA Mensageiro/análise , Receptores de Fatores de Crescimento/análise
18.
J Biol Chem ; 274(42): 29819-25, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10514460

RESUMO

Integrins are important mediators of cell-laminin interactions. In the small intestinal epithelium, which consists of spatially separated proliferative and differentiated cell populations located, respectively, in the crypt and on the villus, laminins and laminin-binding integrins are differentially expressed along the crypt-villus axis. One exception to this is the integrin alpha(6)beta(4), which is thought to be ubiquitously expressed by intestinal cells. However, in this study, a re-evaluation of the beta(4) subunit expression with different antibodies revealed that two forms of beta(4) exist in the human intestinal epithelium. Furthermore, we show that differentiated enterocytes express a full-length 205-kDa beta(4)A subunit, whereas undifferentiated crypt cells express a novel beta(4)A subunit that does not contain the COOH-terminal segment of the cytoplasmic domain (beta(4)A(ctd-)). This new form was not found to arise from alternative beta(4) mRNA splicing. Moreover, we found that these two beta(4)A forms can associate into alpha(6)beta(4)A complexes; however, the beta(4)A(ctd-) integrin expressed by the undifferentiated crypt cells is not functional for adhesion to laminin-5. Hence, these studies identify a novel alpha(6)beta(4)A(ctd-) integrin expressed in undifferentiated intestinal crypt cells that is functionally distinct.


Assuntos
Antígenos CD/metabolismo , Diferenciação Celular , Mucosa Intestinal/metabolismo , Adulto , Antígenos CD/química , Sequência de Bases , Primers do DNA , Humanos , Integrina beta4 , Mucosa Intestinal/citologia , Células Tumorais Cultivadas
19.
Am J Physiol ; 277(3): G631-41, 1999 09.
Artigo em Inglês | MEDLINE | ID: mdl-10484389

RESUMO

The intracellular signaling pathways responsible for cell cycle arrest and establishment of differentiated cells along the gut axis remain largely unknown. In the present study, we analyzed the regulation of p42/p44 mitogen-activated protein kinase (MAPK) in the process of proliferation and differentiation of human intestinal cells. In vitro studies were done in Caco-2/15 cells, a human colon cancer cell line that spontaneously differentiates into an enterocyte phenotype. In vivo studies were performed on cryostat sections of human fetal intestinal epithelium by indirect immunofluorescence. We found that inhibition of the p42/p44 MAPK signaling by the PD-98059 compound or by ectopic expression of the MAPK phosphatase-1 strongly attenuated E2F-dependent transcriptional activity in Caco-2/15 cells. p42/p44 MAPK activities dramatically decreased as soon as Caco-2/15 cells reached confluence. However, significant levels of activated p42 MAPK were detected in differentiated Caco-2/15 cells. Addition of PD-98059 during differentiation interfered with sustained activation of p42 MAPK and sucrase-isomaltase expression. Although p42/p44 MAPKs were expressed in both the villus tip and crypt cells, their phosphorylated and active forms were detected in the undifferentiated crypt cells. Our results indicate that elevated p42/p44 MAPK activities stimulate cell proliferation of intestinal cells, whereas low sustained levels of MAPK activities correlated with G1 arrest and increased expression of sucrase-isomaltase.


Assuntos
Mucosa Intestinal/citologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Células CACO-2 , Ciclo Celular/fisiologia , Diferenciação Celular , Linhagem Celular , DNA/biossíntese , Feto/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Expressão Gênica/fisiologia , Humanos , Mucosa Intestinal/embriologia , Mucosa Intestinal/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Oligo-1,6-Glucosidase/genética , Oligo-1,6-Glucosidase/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Ratos , Sacarase/genética , Sacarase/metabolismo
20.
Front Biosci ; 4: D310-21, 1999 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10077538

RESUMO

Integrins are a large family of cell-surface receptors involved in cell adhesion to the extracellular matrix. In epithelia, it is mainly the integrins belonging to the beta1 and beta4 classes that bind to basement membrane molecules such as the laminins and the type IV collagens. Beta1 and beta4 integrins regulate the assembly of adhesive junctions as well as the activation of various signaling pathways leading to the modulation of gene expression. In this review, I will discuss what is currently known about integrins in human intestinal epithelial cells. The interest in the intestinal cell model to analyze cell-matrix interactions will be delineated and the recent experimental evidence showing that these interactions can regulate cell proliferation and differentiation will be presented.


Assuntos
Matriz Extracelular/fisiologia , Integrinas/fisiologia , Intestinos/fisiologia , Comunicação Celular , Diferenciação Celular , Divisão Celular , Colágeno/metabolismo , Colágeno/fisiologia , Células Epiteliais/fisiologia , Humanos , Integrinas/metabolismo , Mucosa Intestinal/metabolismo , Laminina/metabolismo , Laminina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...