Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Hum Mol Genet ; 25(3): 448-58, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26612203

RESUMO

Epilepsy or seizure disorder is among the least understood chronic medical conditions affecting over 65 million people worldwide. Here, we show that disruption of the polycystic kidney disease 2-like 1 (Pkd2l1 or Pkdl), encoding polycystin-L (PCL), a non-selective cation channel, increases neuronal excitability and the susceptibility to pentylenetetrazol-induced seizure in mice. PCL interacts with ß2-adrenergic receptor (ß2AR) and co-localizes with ß2AR on the primary cilia of neurons in the brain. Pkdl deficiency leads to the loss of ß2AR on neuronal cilia, which is accompanied with a remarkable reduction in cAMP levels in the central nervous system (CNS). The reduction of cAMP levels is associated with a reduction in the activation of cAMP response element-binding protein, but not the activation of Ca(2+)/calmodulin-dependent protein kinase II, Akt or mitogen-activated protein kinases. Our data, thus, indicate for the first time that a ciliary protein complex is required for the control of neuronal excitability in the CNS.


Assuntos
Canais de Cálcio/genética , Córtex Cerebral/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Epilepsia/genética , Hipocampo/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores de Superfície Celular/genética , Tálamo/metabolismo , Animais , Canais de Cálcio/deficiência , Córtex Cerebral/patologia , Cílios/metabolismo , Cílios/patologia , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Epilepsia/induzido quimicamente , Epilepsia/metabolismo , Epilepsia/patologia , Potenciais Pós-Sinápticos Excitadores , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/patologia , Humanos , Transporte de Íons , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Pentilenotetrazol , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Superfície Celular/deficiência , Transdução de Sinais , Tálamo/patologia
2.
Exp Cell Res ; 322(2): 355-64, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24534551

RESUMO

The small intestine consists of two histological compartments composed of the crypts and the villi. The function of the adult small intestinal epithelium is mediated by four different types of mature cells: enterocytes, goblet, enteroendocrine and Paneth. Undifferentiated cells reside in the crypts and produce these four types of mature cells. The niche-related Wnt and Bmp signaling pathways have been suggested to be involved in the regulation and maintenance of the stem cell microenvironment. In our laboratory, we isolated the first normal human intestinal epithelial crypt (HIEC) cell model from the human fetal intestine and in this study we investigated the expression of a panel of intestinal stem cell markers in HIEC cells under normal culture parameters as well as under conditions that mimic the stem cell microenvironment. The results showed that short term stimulation of HIEC cells with R-spondin 1 and Wnt-3a±SB-216763, a glycogen synthase kinase 3ß (GSK3ß) inhibitor, induced ß-catenin/TCF activity and expression of the WNT target genes, cyclin D2 and LGR5. Treatment of HIEC cells with noggin, an antagonist of BMP signaling, abolished SMAD2/5/8 phosphorylation. Inducing a switch from inactive WNT/active BMP toward active WNT/inactive BMP pathways was sufficient to trigger a robust intestinal primordial stem-like cell signature with predominant LGR5, PHLDA1, PROM1, SMOC2 and OLFM4 expression. These findings demonstrate that even fully established cultures of intestinal cells can be prompted toward a CBC stem cell-like phenotype. This model should be useful for studying the regulation of human intestinal stem cell self-renewal and differentiation.


Assuntos
Diferenciação Celular , Neoplasias Colorretais/patologia , Células Epiteliais/citologia , Intestinos/citologia , Células-Tronco/citologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia , Adulto , Western Blotting , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proliferação de Células , Células Cultivadas , Neoplasias Colorretais/metabolismo , Células Epiteliais/metabolismo , Feto/citologia , Feto/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/metabolismo , Luciferases/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Regeneração/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo , Proteínas Wnt/genética
3.
Carcinogenesis ; 35(6): 1217-27, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24403311

RESUMO

The integrin α6 subunit pre-messenger RNA undergoes alternative splicing to generate two different splice variants, named α6A and α6B, having distinct cytoplasmic domains. In the human colonic gland, these splice variants display different patterns of expression suggesting specific functions for each variant. We have previously found an up-regulation of the α6ß4 integrin in colon adenocarcinomas as well as an increase in the α6A/α6B ratio, but little is known about the involvement of α6Aß4 versus α6Bß4 in this context. The aim of this study was to elucidate the function of the α6Aß4 integrin in human colorectal cancer (CRC) cells. Expression studies on a panel of primary CRCs confirmed that the up-regulation of the α6 subunit in CRC is a direct consequence of the increase of the α6A variant. To investigate the functional significance of an α6A up-regulation in CRC, we specifically knocked down its expression in well-established CRC cell lines using a small-hairpin RNA approach. Results showed a growth rate reduction in all α6A knockdown CRC cell lines studied. The α6A silencing was also found to be associated with a significant repression of a number of Wnt/ß-catenin pathway end points. Moreover, it was accompanied by a reduction in the capacity of these cells to develop tumours in xenografts. Taken together, these results demonstrate that the α6A variant is a pro-proliferative form of the α6 integrin subunit in CRC cells and appears to mediate its effects through the Wnt/ß-catenin pathway.


Assuntos
Processamento Alternativo , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Integrina alfa6/genética , Via de Sinalização Wnt , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Proteínas Desgrenhadas , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Xenoenxertos , Humanos , Integrina alfa6/metabolismo , Espaço Intracelular/metabolismo , Camundongos , Fosfoproteínas/metabolismo , Subunidades Proteicas/genética , Transporte Proteico , Carga Tumoral
4.
J Cell Sci ; 125(Pt 14): 3454-63, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22467857

RESUMO

The crypt-villus axis constitutes the functional unit of the small intestine, where mature absorptive cells are confined to the villi, and stem cells and transit amplifying and differentiating cells are restricted to the crypts. The polycomb group (PcG) proteins repress differentiation and promote self-renewal in embryonic stem cells. PcGs prevent transcriptional activity by catalysing epigenetic modifications, such as the covalent addition of methyl groups on histone tails, through the action of the polycomb repressive complex 2 (PRC2). Although a role for PcGs in the preservation of stemness characteristics is now well established, recent evidence suggests that they may also be involved in the regulation of differentiation. Using intestinal epithelial cell models that recapitulate the enterocytic differentiation programme, we generated a RNAi-mediated stable knockdown of SUZ12, which constitutes a cornerstone for PRC2 assembly and functionality, in order to analyse intestinal cell proliferation and differentiation. Expression of SUZ12 was also investigated in human intestinal tissues, revealing the presence of SUZ12 in most proliferative epithelial cells of the crypt and an increase in its expression in colorectal cancers. Moreover, PRC2 disruption led to a significant precocious expression of a number of terminal differentiation markers in intestinal cell models. Taken together, our data identified a mechanism whereby PcG proteins participate in the repression of the enterocytic differentiation program, and suggest that a similar mechanism exists in situ to slow down terminal differentiation in the transit amplifying cell population.


Assuntos
Trato Gastrointestinal/citologia , Complexo Repressor Polycomb 2/fisiologia , Células CACO-2 , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Histonas/genética , Histonas/metabolismo , Humanos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Regiões Promotoras Genéticas , Transcrição Gênica
5.
Biochem Biophys Res Commun ; 430(4): 1195-200, 2013 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-23274493

RESUMO

The proto-oncogene Src is an important protein tyrosine kinase involved in signaling pathways that control cell adhesion, growth, migration and survival. Here, we investigated the involvement of Src family kinases (SFKs) in human intestinal cell differentiation. We first observed that Src activity peaked in early stages of Caco-2/15 cell differentiation. Inhibition of SFKs with PP2, a selective SFK inhibitor, accelerated the overall differentiation program. Interestingly, all polarization and terminal differentiation markers tested, including sucrase-isomaltase, lactase-phlorizin hydrolase and E and Li-cadherins were found to be significantly up-regulated after only 3 days of treatment in the newly differentiating cells. Further investigation of the effects of PP2 revealed a significant up-regulation of the two main intestinal epithelial cell-specific transcription factors Cdx2 and HNF1α and a reduction of polycomb PRC2-related epigenetic repressing activity as measured by a decrease in H3K27me3, two events closely related to the control of cell terminal differentiation in the intestine. Taken together, these data suggest that SFKs play a key role in the control of intestinal epithelial cell terminal differentiation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Pirimidinas/farmacologia , Quinases da Família src/antagonistas & inibidores , Células CACO-2 , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/enzimologia , Intestinos/citologia , Intestinos/enzimologia , Proto-Oncogene Mas , Fatores de Transcrição/metabolismo
6.
Cells ; 12(7)2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-37048132

RESUMO

Primary cilia are sensory antennae located at the cell surface which mediate a variety of extracellular signals involved in development, tissue homeostasis, stem cells and cancer. Primary cilia are found in an extensive array of vertebrae cells but can only be generated when cells become quiescent. The small intestinal epithelium is a rapidly self-renewing tissue organized into a functional unit called the crypt-villus axis, containing progenitor and differentiated cells, respectively. Terminally differentiated villus cells are notoriously devoid of primary cilia. We sought to determine if intestinal crypts contain a quiescent cell population that could be identified by the presence of primary cilia. Here we show that primary cilia are detected in a subset of cells located deep in the crypts slightly above a Paneth cell population. Using a normal epithelial proliferative crypt cell model, we show that primary cilia assembly and activity correlate with a quiescent state. These results provide further evidence for the existence of a quiescent cell population in the human small intestine and suggest the potential for new modes of regulation in stem cell dynamics.


Assuntos
Cílios , Intestino Delgado , Humanos , Duodeno , Divisão Celular , Celulas de Paneth
7.
Exp Cell Res ; 316(9): 1454-64, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20211617

RESUMO

In this study, we have analyzed the expression and localization of polycystin-1 in intestinal epithelial cells, a system lacking primary cilia. Polycystin-1 was found to be expressed in the epithelium of the small intestine during development and levels remained elevated in the adult. Dual-labelling indirect immunofluorescence revealed polycystin-1 at sites of cell-cell contact co-localizing with the desmosomes both in situ as well as in polarized Caco-2/15 cells. In unpolarized cultures of Caco-2/15 cells, polycystin-1 was recruited to the cell surface early during initiation of cell junction assembly. In isolated Caco-2/15 cells and HIEC-6 cell cultures, where junctional complexes are absent, polycystin-1 was found predominantly associated with the cytoskeletal elements of the intermediate filaments and microtubule networks. More precisely, polycystin-1 was seen as brightly labelled puncta decorating the keratin-18 positive filaments as well as the beta-tubulin positive microtubules, which was particularly obvious in the lamellipodia. Treatment with the microtubule-disrupting agent, nocodazole, eliminated the microtubule association of polycystin-1 but did not seem to affect its association with keratin or the desmosomes. Taken together these data suggest that polycystin-1 is involved with the establishment of cell-cell junctions in absorptive intestinal epithelial cells and exploits the microtubule-based machinery in order to be transported to the plasma membrane.


Assuntos
Polaridade Celular , Desmossomos/metabolismo , Células Epiteliais/metabolismo , Feto/metabolismo , Microtúbulos/metabolismo , Canais de Cátion TRPP/metabolismo , Adulto , Western Blotting , Células CACO-2 , Cálcio/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Queratina-18/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Cátion TRPP/genética
8.
J Cell Physiol ; 222(2): 387-400, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19885839

RESUMO

Integrin-linked kinase (ILK) plays a role in integrin signaling-mediated extracellular matrix (ECM)-cell interactions and also acts as a scaffold protein in functional focal adhesion points. In the present study, we investigated the expression and roles of ILK in human intestinal epithelial cells (IECs) in vivo and in vitro. Herein, we report that ILK and its scaffold-function interacting partners, PINCH-1, alpha-parvin, and beta-parvin, are expressed according to a decreasing gradient from the bottom of the crypt (proliferative/undifferentiated) compartment to the tip of the villus (non-proliferative/differentiated) compartment, closely following the expression pattern of the ECM/basement membrane component fibronectin. The siRNA knockdown of ILK in human IECs caused a loss of PINCH-1, alpha-parvin, and beta-parvin expression, along with a significant decrease in cell proliferation via a loss of cyclin D1 and an increase in p27 and hypophosphorylated pRb expression levels. ILK knockdown severely affected cell spreading, migration, and restitution abilities, which were shown to be directly related to a decrease in fibronectin deposition. All ILK knockdown-induced defects were rescued with exogenously deposited fibronectin. Altogether, our results indicate that ILK performs crucial roles in the control of human intestinal cell and crypt-villus axis homeostasis-especially with regard to basement membrane fibronectin deposition-as well as cell proliferation, spreading, and migration.


Assuntos
Movimento Celular , Proliferação de Células , Enterócitos/enzimologia , Fibronectinas/metabolismo , Intestinos/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Actinina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Células CACO-2 , Diferenciação Celular , Forma Celular , Proteínas de Ligação a DNA/metabolismo , Genótipo , Humanos , Intestinos/citologia , Intestinos/embriologia , Proteínas com Domínio LIM , Proteínas de Membrana , Proteínas dos Microfilamentos , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , Fatores de Tempo , Transdução Genética , Transfecção
9.
Wound Repair Regen ; 18(1): 114-22, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20082684

RESUMO

Rapid and efficient healing of epithelial damage is critical to the functional integrity of the small intestine. Epithelial repair is a complex process that has largely been studied in cultured epithelium but to a much lesser extent in mucosa. We describe a novel method for the study of wound healing using a co-culture system that combined an intestinal epithelial Caco-2/15 cell monolayer cultured on top of human intestinal myofibroblasts, which together formed a basement membrane-like structure that contained many of the major components found at the epithelial-mesenchymal interface in the human intestine. To investigate the mechanism of restitution, small lesions were generated in epithelial cell monolayers on plastic or in co-cultures without disturbing the underlying mesenchymal layer. Monitoring of wound healing showed that repair was more efficient in Caco-2/15-myofibroblast co-cultures than in Caco-2/15 monolayers and involved the deposition of basement membrane components. Functional experiments showed that the addition of type I collagen or human fibronectin to the culture medium significantly accelerated wound closure on epithelial cell co-cultures. This system may provide a new tool to investigate the mechanisms that regulate wound healing in the intestinal epithelium.


Assuntos
Fibroblastos/fisiologia , Mucosa Intestinal/lesões , Mucosa Intestinal/fisiologia , Cicatrização/fisiologia , Células CACO-2 , Movimento Celular , Proliferação de Células , Técnicas de Cocultura , Fibroblastos/citologia , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Músculo Liso/citologia
10.
Biol Cell ; 101(12): 695-708, 2009 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-19527220

RESUMO

BACKGROUND: Integrins are transmembrane alphabeta heterodimer receptors that function as structural and functional bridges between the cytoskeleton and ECM (extracellular matrix) molecules. The RGD (arginine-glycine-aspartate tripeptide motif)-dependent integrin alpha8beta1 has been shown to be involved in various cell functions in neuronal and mesenchymal-derived cell types. Its role in epithelial cells remains unknown. RESULTS: Integrin alpha8beta1 was found to be expressed in the crypt cell population of the human intestine but was absent from differentiating and mature epithelial cells of the villus. The function of alpha8beta1 in epithelial crypt cells was investigated at the cellular level using normal HIECs (human intestinal epithelial cells). Specific knockdown of alpha8 subunit expression using an shRNA (small-hairpin RNA) approach showed that alpha8beta1 plays important roles in RGD-dependent cell adhesion, migration and proliferation via a RhoA/ROCK (Rho-associated kinase)-dependent mechanism as demonstrated by active RhoA quantification and pharmacological inhibition of ROCK. Moreover, loss of alpha8beta1, through RhoA/ROCK, impairs FA (focal adhesion) complex integrity as demonstrated by faulty vinculin recruitment. CONCLUSIONS: Integrin alpha8beta1 is expressed in epithelial cells. In intestinal crypt cells, alpha8beta1 is closely involved in the regulation of adhesion, migration and cell proliferation via a predominant RhoA/ROCK-dependent mechanism. These results suggest an important role for this integrin in intestinal crypt cell homoeostasis.


Assuntos
Movimento Celular , Proliferação de Células , Integrinas/metabolismo , Intestinos/citologia , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Adesão Celular , Linhagem Celular , Células Cultivadas , Feto , Expressão Gênica , Humanos , Integrinas/genética , Mucosa Intestinal/citologia , Mucosa Intestinal/embriologia , Mucosa Intestinal/metabolismo , Intestinos/embriologia , Quinases Associadas a rho/genética , Proteína rhoA de Ligação ao GTP/genética
11.
Histochem Cell Biol ; 131(4): 531-6, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19107504

RESUMO

The integrin alpha6 subunit exists as two different variants, termed alpha6A and alpha6B. These two variants have been shown to harbor potentially distinct biochemical properties but little is known about their cellular function. The aim of this work was to characterize the expression of the integrin alpha6A and B variants in relation to cell proliferation and differentiation in the human small intestinal epithelium. The results showed distinct expression patterns for the two variants along the crypt-villus axis. Indeed, proliferative cells of the crypt were found to predominantly express alpha6A, while differentiated enterocytes and Paneth cells expressed the alpha6B variant. A similar relationship was observed in intestinal cell models by competitive RT-PCR. Further studies in the Caco-2 cell model showed that manipulating the cellular balance of the two alpha6 variants can influence transcriptional activities related to cell proliferation but not differentiation. This suggests that differential expression of the alpha6 subunits is involved in the intestinal epithelial cell renewal process. Further studies will be needed to substantiate this hypothesis.


Assuntos
Diferenciação Celular/fisiologia , Integrina alfa6/metabolismo , Integrina beta4/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Células CACO-2 , Proliferação de Células , Humanos , Mucosa Intestinal/citologia , Intestino Delgado/citologia , Isoformas de Proteínas/metabolismo
12.
BMC Cancer ; 9: 223, 2009 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-19586553

RESUMO

BACKGROUND: Integrins are known to be important contributors to cancer progression. We have previously shown that the integrin beta4 subunit is up-regulated in primary colon cancer. Its partner, the integrin alpha6 subunit, exists as two different mRNA splice variants, alpha6A and alpha6B, that differ in their cytoplasmic domains but evidence for distinct biological functions of these alpha6 splice variants is still lacking. METHODS: In this work, we first analyzed the expression of integrin alpha6A and alpha6B at the protein and transcript levels in normal human colonic cells as well as colorectal adenocarcinoma cells from both primary tumors and established cell lines. Then, using forced expression experiments, we investigated the effect of alpha6A and alpha6B on the regulation of cell proliferation in a colon cancer cell line. RESULTS: Using variant-specific antibodies, we observed that alpha6A and alpha6B are differentially expressed both within the normal adult colonic epithelium and between normal and diseased colonic tissues. Proliferative cells located in the lower half of the glands were found to predominantly express alpha6A, while the differentiated and quiescent colonocytes in the upper half of the glands and surface epithelium expressed alpha6B. A relative decrease of alpha6B expression was also identified in primary colon tumors and adenocarcinoma cell lines suggesting that the alpha6A/alpha6B ratios may be linked to the proliferative status of colonic cells. Additional studies in colon cancer cells showed that experimentally restoring the alpha6A/alpha6B balance in favor of alpha6B caused a decrease in cellular S-phase entry and repressed the activity of c-Myc. CONCLUSION: The findings that the alpha6Bbeta4 integrin is expressed in quiescent normal colonic cells and is significantly down-regulated in colon cancer cells relative to its alpha6Abeta4 counterpart are consistent with the anti-proliferative influence and inhibitory effect on c-Myc activity identified for this alpha6Bbeta4 integrin. Taken together, these findings point out the importance of integrin variant expression in colon cancer cell biology.


Assuntos
Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Integrina alfa6beta4/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/terapia , Células Epiteliais/citologia , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Integrina alfa6beta4/metabolismo , Pessoa de Meia-Idade , Modelos Biológicos , RNA Mensageiro/metabolismo , Fase S
13.
Oncogene ; 24(45): 6820-9, 2005 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16007143

RESUMO

The integrin beta4 subunit has been shown to be involved in various aspects of cancer progression. The aim of the present work was to evaluate the expression of beta4 in primary colon cancers and to investigate the occurrence of a previously identified intestinal nonfunctional variant of beta4 (beta4ctd-) for adhesion to laminin. Immunodetection of beta4 using a panel of antibodies and RT-PCR analyses were performed on series of paired primary colon tumors and corresponding resection margins. The beta4 subunit was found to be significantly overexpressed in cancer specimens at both the protein and transcript levels. Surprisingly, beta4 levels of expression were closely correlated with those of the oncogene c-Myc in individual specimens. In vitro studies of c-Myc overexpression showed an upregulation of beta4 promoter activity. Finally, the beta4ctd- form was identified in the normal proliferative colonic cells but was found to be predominantly absent in colon cancer cells, both in situ and in vitro. We concluded that the beta4ctd- form is lost from colon cancer cells, while the level of the wild-type form of beta4, which is functional for adhesion to laminin, is increased in primary tumors in relation with the expression of c-Myc.


Assuntos
Neoplasias Colorretais/metabolismo , Expressão Gênica , Genes myc , Integrina beta4/metabolismo , Regulação para Cima/genética , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Humanos , Integrina beta4/genética , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Endocrinology ; 147(5): 2325-37, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16497811

RESUMO

The aim of this study was to investigate the short-term regulation of the ACTH receptor human (h) melanocortin receptor 2 (MC2R) by transfection of a c-Myc-tagged hMC2R in the M3 cell line and assess its membrane expression by indirect immunofluorescence. Stimulation with ACTH induced production of cAMP with EC(50) values ranging from 7.6-11.9 nM in transient and stable transfectants, respectively. Pretreatment with ACTH induced a dose-dependent loss of cAMP production, from 1 pm up to 10 nM. Desensitization was also time dependent, with 70% loss of maximal responsiveness occurring after 15-min pretreatment with 10 nM ACTH, followed by a plateau up to 60 min. The decrease in hMC2R responsiveness was abrogated by individual treatment with protein kinase A (PKA) or protein kinase C inhibitors, H-89 and GF109203X. However, when added simultaneously, receptor responsiveness was raised over the maximal hMC2R activity observed in control cells. ACTH-induced loss of cAMP production was accompanied by receptor sequestration into intracellular vesicles (maximum after 30-min exposure). Cotransfection of M3 cells with the c-Myc-tagged hMC2R and beta-arrestin-2-green fluorescence protein along with sucrose treatment revealed that beta-arrestin-2-green fluorescence protein and c-Myc-hMC2R were redistributed in similar intracellular vesicles through a clathrin-dependent, but caveolae-independent, process. Sucrose pretreatment blocked receptor desensitization, indicating that hMC2R desensitization and internalization are interrelated. Moreover, preincubation with H-89 abrogated hMC2R internalization, whereas GF109203X had no effect. In conclusion, the present results indicate that PKA and protein kinase C act synergistically to induce hMC2R desensitization, but only PKA is essential for receptor internalization, highlighting the complex nature of the short-term regulatory pattern of this receptor.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Regulação da Expressão Gênica , Proteína Quinase C/fisiologia , Receptor Tipo 2 de Melanocortina/biossíntese , Arrestinas/biossíntese , Western Blotting , Linhagem Celular Tumoral , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Epitopos/química , Técnica Indireta de Fluorescência para Anticorpo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sacarose/farmacologia , Transfecção , beta-Arrestina 2 , beta-Arrestinas
15.
J Mol Biol ; 325(5): 949-62, 2003 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-12527301

RESUMO

Polycystin-2 (PC2) is the product of the second cloned gene (PKD2) responsible for autosomal dominant polycystic kidney disease and has recently been shown to be a calcium-permeable cation channel. PC2 has been shown to connect indirectly with the actin microfilament. Here, we report a direct association between PC2 and the actin microfilament. Using a yeast two-hybrid screen, we identified a specific interaction between the PC2 cytoplasmic C-terminal domain and tropomyosin-1 (TM-1), a component of the actin microfilament complex. Tropomyosins constitute a protein family of more than 20 isoforms arising mainly from alternative splicing and are present in muscle as well as non-muscle cells. We identified a new TM-1 splicing isoform in kidney and heart (TM-1a) that differs from TM-1 in the C terminus and interacted with PC2. In vitro biochemical methods, including GST pull-down, blot overlay and microtiter binding assays, confirmed the interaction between PC2 and the two TM-1 isoforms. Further experiments targeted the interacting domains to G821-R878 of PC2 and A152-E196, a common segment of TM-1 and TM-1a. Indirect double immunofluorescence experiments showed partial co-localization of PC2 and TM-1 in transfected mouse fibroblast NIH 3T3 cells. Co-immunoprecipitation (co-IP) studies using 3T3 cells and Xenopus oocytes co-expressing PC2 and TM-1 (or TM-1a) revealed in vivo association between the protein pairs. Furthermore, the in vivo interaction between the endogenous PC2 and TM-1 was demonstrated also by reciprocal co-IP using native human embryonic kidney cells and human adult kidney. Considering previous reports that TM-1 acts as a suppressor of neoplastic growth of transformed cells, it is possible that TM-1 contributes to cyst formation/growth when the anchorage of PC2 to the actin microfilament via TM-1 is altered.


Assuntos
Proteínas de Drosophila , Proteínas de Membrana/metabolismo , Tropomiosina/metabolismo , Células 3T3 , Citoesqueleto de Actina/química , Actinas/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Canais de Cálcio/metabolismo , Feminino , Glutationa Transferase/metabolismo , Histidina/metabolismo , Humanos , Técnicas In Vitro , Rim/metabolismo , Proteínas de Membrana/genética , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Oócitos/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Testes de Precipitina , Transporte Proteico , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Canais de Cátion TRPP , Tropomiosina/genética , Técnicas do Sistema de Duplo-Híbrido , Xenopus laevis/metabolismo
16.
PLoS One ; 8(8): e74337, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23991217

RESUMO

Interactions between the cell basal membrane domain and the basement membrane are involved in several cell functions including proliferation, migration and differentiation. Intestinal epithelial cells can interact with laminin, a major intestinal basement membrane glycoprotein, via several cell-surface laminin-binding proteins including integrin and non-integrin receptors. The 37/67kDa laminin receptor (37/67LR) is one of these but its role in normal epithelial cells is still unknown. The aim of this study was to characterise the expression pattern and determine the main function of 37/67LR in the normal human small intestinal epithelium. Immunolocalization studies revealed that 37/67LR was predominantly present in the undifferentiated/proliferative region of the human intestinal crypt in both the immature and adult intestine. Using a human intestinal epithelial crypt (HIEC) cell line as experimental model, we determined that 37/67LR was expressed in proliferative cells in both the cytoplasmic and membrane compartments. Small-interfering RNA-mediated reduction of 37/67LR expression led to HIEC cell-cycle reduction and loss of the ability to adhere to laminin-related peptides under conditions not altering ribosomal function. Taken together, these findings indicate that 37/67LR regulates proliferation and adhesion in normal intestinal epithelial cells independently of its known association with ribosomal function.


Assuntos
Adesão Celular/fisiologia , Proliferação de Células , Mucosa Intestinal/citologia , Receptores de Laminina/fisiologia , Adulto , Sequência de Bases , Western Blotting , Células Cultivadas , Primers do DNA , Técnicas de Silenciamento de Genes , Humanos , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Laminina/genética
17.
Autophagy ; 8(6): 893-902, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22652752

RESUMO

Recently, autophagy has been found to be strongly activated in colon cancer cells, but few studies have addressed the normal colon mucosa. The aim of this study was to characterize autophagy in normal human intestinal cells. We used the expression of LC3-II and BECN1 as well as SQSTM1 as markers of autophagy activity. Using the normal human intestinal epithelial crypt (HIEC) cell experimental model, we found that autophagy was much more active in undifferentiated cells than in differentiated cells. In the normal adult colonic mucosa, BECN1 was found in the proliferative epithelial cells of the lower part of the gland while SQSTM1 was predominantly found in the differentiated cells of the upper part of the gland and surface epithelium. Interestingly, the weak punctate pattern of SQSTM1 expression in the lower gland colocalized with BECN1-labeled autophagosomes. The usefulness of SQSTM1 as an active autophagy marker was confirmed in colon cancer specimens at the protein and transcript levels. In conclusion, our results show that autophagy is active in the colonic gland and is associated with the intestinal proliferative/undifferentiated and progenitor cell populations.


Assuntos
Autofagia , Colo/citologia , Mucosa Intestinal/citologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Colo/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Regulação para Baixo , Enterócitos/citologia , Enterócitos/metabolismo , Imunofluorescência , Humanos , Mucosa Intestinal/metabolismo , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Modelos Biológicos , Proteína Sequestossoma-1
18.
Matrix Biol ; 30(3): 195-206, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21406227

RESUMO

Collagen VI is a heterotrimer composed of three α chains (α1, α2, α3) widely expressed throughout various interstitial matrices. Collagen VI is also found near the basement membranes of many tissues where it serves as an anchoring meshwork. The aim of this study was to investigate the distribution and role of collagen VI at the epithelial-stromal interface in the intestine. Results showed that collagen VI is a bona fide epithelial basal lamina component and constitutes the major collagen type of epithelial origin in this organ. In vitro, collagen VI co-distributes with fibronectin. Targeted knockdown of collagen VI expression in intestinal epithelial cells was used to investigate its function. Depletion of collagen VI from the matrix led to a significant increase in cell spreading and fibrillar adhesion formation coinciding with an upregulation of fibronectin expression, deposition and organization as well as activation of myosin light chain phosphorylation by the myosin light chain kinase and Rho kinase dependent mechanisms. Plating cells deficient for collagen VI on collagen VI rescued the phenotype. Taken together, these data demonstrate that collagen VI is an important basal lamina component involved in the regulation of epithelial cell behavior most notably as a regulator of epithelial cell-fibronectin interactions.


Assuntos
Membrana Basal/metabolismo , Colágeno Tipo VI/metabolismo , Células Epiteliais/metabolismo , Fibronectinas/metabolismo , Movimento Celular , Forma Celular/genética , Células Cultivadas , Colágeno Tipo VI/genética , Adesões Focais/metabolismo , Humanos , Integrina beta1/metabolismo , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Proteínas dos Microfilamentos/metabolismo , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/metabolismo , Fosforilação , Interferência de RNA , Tensinas , Transcrição Gênica
19.
J Mol Histol ; 41(2-3): 143-52, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20532811

RESUMO

The expression of apoptotic factors Bcl-2 and Bax were studied in the conjunctiva of diabetic patients with and without retinopathy. All patients underwent a complete ophthalmic examination including ocular fundus and retinal fluorescein angiography. The indirect immunoperoxidase method was performed on 15 normal conjunctiva taken during cataract surgery (group 1), on 40 eyes of 40 patients with type 2 diabetes without diabetic retinopathy (group 2) and 13 eyes of 13 patients with diabetic retinopathy (group 3). In normal human conjunctiva, Bax showed positive expression in epithelial, vascular and stromal cells whereas Bcl-2 staining was negative. In the conjunctiva of diabetic patients without diabetic retinopathy, Bax was widely, and strongly, expressed in epithelial cells, vascular endothelial cells, fibroblasts and infiltrating cells such as macrophages. For patients with diabetic retinopathy, Bax was consistently strong to very strong. Bcl-2 protein expression became weak to negative for diabetic patients both with and without diabetic retinopathy. Immunoreactivity was not correlated between Bcl-2 and Bax in the conjunctiva of diabetic patients. Bax was always localized in tissues characterized by a high rate of apoptosis, whereas, Bcl-2 was absent. Our results suggest that diabetic human conjunctiva, with its inflammatory phenomena, is considered as a privileged target for programmed cell death.


Assuntos
Apoptose , Diabetes Mellitus Tipo 2/metabolismo , Retinopatia Diabética/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteína X Associada a bcl-2/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Estudos de Casos e Controles , Túnica Conjuntiva/metabolismo , Túnica Conjuntiva/patologia , Diabetes Mellitus Tipo 2/patologia , Retinopatia Diabética/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade
20.
Am J Physiol Renal Physiol ; 291(2): F395-406, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16609150

RESUMO

Polycystin-1 and polycystin-2 are involved in autosomal dominant polycystic kidney disease by unknown mechanisms. These two proteins are located in primary cilia where they mediate mechanosensation, suggesting a link between cilia function and renal disease. In this study, we sought to characterize the subcellular localization of polycystin-L, a closely related member of polycystin-2, in epithelial renal cell lines. We have shown that endogenous polycystin-l subcellular distribution is different in proliferative and nonproliferative cultures. Polycystin-L is found mostly in the endoplasmic reticulum in subconfluent cell cultures, while in confluent cells it is redistributed to sites of cell-cell contact and to the primary cilium as is polycystin-1. Subcellular fractionation confirmed a common distribution of polycystin-L and polycystin-1 in the fractions corresponding to those containing the plasma membrane of postconfluent cells. Reciprocal coimmunoprecipitation experiments showed that polycystin-L was associated with polycystin-1 in a common complex in both subconfluent and confluent cell cultures. Interestingly, we also identified a novel site for a polycystin member (polycystin-L) in unciliated cells, the centrosome, which allowed us to reveal an involvement of polycystin-l in cell proliferation.


Assuntos
Centrossomo/química , Canais de Cátion TRPP/análise , Animais , Canais de Cálcio , Linhagem Celular , Membrana Celular/química , Membrana Celular/fisiologia , Proliferação de Células , Centrossomo/fisiologia , Cílios/química , Cílios/fisiologia , Cães , Retículo Endoplasmático/química , Retículo Endoplasmático/fisiologia , Células Epiteliais/química , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Imuno-Histoquímica , Rim/química , Rim/citologia , Rim/fisiologia , Túbulos Renais Coletores/química , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/fisiologia , Glicoproteínas de Membrana/análise , Camundongos , Fosfoproteínas/análise , Ratos , Receptores de Superfície Celular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA