Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Biol Cell ; 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29907957

RESUMEN

BACKGROUND INFORMATION: Tumor stroma remodeling is a key feature of malignant tumors and can promote cancer progression. Laminins are major constituents of basement membranes that physically separate the epithelium from the underlying stroma. RESULTS: By employing mouse models expressing high and low levels of the laminin α1 chain (LMα1), we highlighted its implication in a tumor-stroma crosstalk, thus leading to increased colon tumor incidence, angiogenesis and tumor growth. The underlying mechanism involves attraction of carcinoma-associated fibroblasts by LMα1, VEGFA expression triggered by the complex integrin α2ß1-CXCR4 and binding of VEGFA to LM-111, which in turn promotes angiogenesis, tumor cell survival and proliferation. A gene signature comprising LAMA1, ITGB1, ITGA2, CXCR4 and VEGFA has negative predictive value in colon cancer. CONCLUSIONS: Together, we have identified VEGFA, CXCR4 and α2ß1 integrin downstream of LMα1 in colon cancer as of bad prognostic value for patient survival. SIGNIFICANCE: This information opens novel opportunities for diagnosis and treatment of colon cancer.

3.
Gut ; 66(10): 1748-1760, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-27371534

RESUMEN

OBJECTIVE: Epidemiological and clinical data indicate that patients suffering from IBD with long-standing colitis display a higher risk to develop colorectal high-grade dysplasia. Whereas carcinoma invasion and metastasis rely on basement membrane (BM) disruption, experimental evidence is lacking regarding the potential contribution of epithelial cell/BM anchorage on inflammation onset and subsequent neoplastic transformation of inflammatory lesions. Herein, we analyse the role of the α6ß4 integrin receptor found in hemidesmosomes that attach intestinal epithelial cells (IECs) to the laminin-containing BM. DESIGN: We developed new mouse models inducing IEC-specific ablation of α6 integrin either during development (α6ΔIEC) or in adults (α6ΔIEC-TAM). RESULTS: Strikingly, all α6ΔIEC mutant mice spontaneously developed long-standing colitis, which degenerated overtime into infiltrating adenocarcinoma. The sequence of events leading to disease onset entails hemidesmosome disruption, BM detachment, IL-18 overproduction by IECs, hyperplasia and enhanced intestinal permeability. Likewise, IEC-specific ablation of α6 integrin induced in adult mice (α6ΔIEC-TAM) resulted in fully penetrant colitis and tumour progression. Whereas broad-spectrum antibiotic treatment lowered tissue pathology and IL-1ß secretion from infiltrating myeloid cells, it failed to reduce Th1 and Th17 response. Interestingly, while the initial intestinal inflammation occurred independently of the adaptive immune system, tumourigenesis required B and T lymphocyte activation. CONCLUSIONS: We provide for the first time evidence that loss of IECs/BM interactions triggered by hemidesmosome disruption initiates the development of inflammatory lesions that progress into high-grade dysplasia and carcinoma. Colorectal neoplasia in our mouse models resemble that seen in patients with IBD, making them highly attractive for discovering more efficient therapies.


Asunto(s)
Adenocarcinoma/fisiopatología , Colitis/fisiopatología , Neoplasias Colorrectales/fisiopatología , Citocinas/metabolismo , Hemidesmosomas/fisiología , Integrina alfa6/genética , Integrina alfa6beta4/metabolismo , Mucosa Intestinal/metabolismo , Inmunidad Adaptativa , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Linfocitos B , Membrana Basal/fisiopatología , Caspasa 1/metabolismo , Colitis/genética , Colitis/metabolismo , Colitis/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Citocinas/genética , Células Epiteliales/metabolismo , Hemidesmosomas/genética , Homeostasis/genética , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Queratina-18/metabolismo , Queratina-8/metabolismo , Activación de Linfocitos , Ratones , Moco/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Permeabilidad , Índice de Severidad de la Enfermedad , Transducción de Señal , Linfocitos T
4.
Methods ; 94: 85-100, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26439175

RESUMEN

Mechanical interaction between cells and their surrounding extracellular matrix (ECM) controls key processes such as proliferation, differentiation and motility. For many years, two-dimensional (2D) models were used to better understand the interactions between cells and their surrounding ECM. More recently, variation of the mechanical properties of tissues has been reported to play a major role in physiological and pathological scenarios such as cancer progression. The 3D architecture of the ECM finely tunes cellular behavior to perform physiologically relevant tasks. Technical limitations prevented scientists from obtaining accurate assessment of the mechanical properties of physiologically realistic matrices. There is therefore a need for combining the production of high-quality cell-derived 3D matrices (CDMs) and the characterization of their topographical and mechanical properties. Here, we describe methods that allow to accurately measure the young modulus of matrices produced by various cellular types. In the first part, we will describe and review several protocols for generating CDMs matrices from endothelial, epithelial, fibroblastic, muscle and mesenchymal stem cells. We will discuss tools allowing the characterization of the topographical details as well as of the protein content of such CDMs. In a second part, we will report the methodologies that can be used, based on atomic force microscopy, to accurately evaluate the stiffness properties of the CDMs through the quantification of their young modulus. Altogether, such methodologies allow characterizing the stiffness and topography of matrices deposited by the cells, which is key for the understanding of cellular behavior in physiological conditions.


Asunto(s)
Matriz Extracelular/fisiología , Animales , Bovinos , Módulo de Elasticidad , Matriz Extracelular/química , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Células Madre Mesenquimatosas/fisiología , Ratones , Microscopía de Fuerza Atómica , Miocitos del Músculo Liso/fisiología , Células 3T3 NIH
5.
Cell Adh Migr ; 9(1-2): 4-13, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25611571

RESUMEN

The extracellular matrix (ECM) molecule tenascin-C (TNC) promotes tumor progression. This has recently been demonstrated in the stochastic murine RIP1-Tag2 insulinoma model, engineered to either express TNC abundantly or to be devoid of TNC. However, our knowledge about organization of the TNC microenvironment is scant. Here we determined the spatial distribution of TNC together with other ECM molecules in murine RIP1-Tag2 insulinoma and human cancer tissue (insulinoma and colorectal carcinoma). We found that TNC is organized in matrix tracks together with other ECM molecules of the AngioMatrix signature, a previously described gene expression profile that characterizes the angiogenic switch. Moreover, stromal cells including endothelial cells, fibroblasts and leukocytes were enriched in the TNC tracks. Thus, TNC tracks may provide niches for stromal cells and regulate their behavior. Given similarities of TNC rich niches for stromal cells in human insulinoma and colon cancer, we propose that the RIP1-Tag2 model may be useful for providing insights into the contribution of the tumor stroma specific ECM as promoter of cancer progression.


Asunto(s)
Movimiento Celular/fisiología , Neoplasias Colorrectales/metabolismo , Matriz Extracelular/metabolismo , Células del Estroma/patología , Tenascina/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Humanos , Ratones Transgénicos
6.
PLoS One ; 9(10): e111336, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25347196

RESUMEN

Laminins (LM), basement membrane molecules and mediators of epithelial-stromal communication, are crucial in tissue homeostasis. Inflammatory Bowel Diseases (IBD) are multifactorial pathologies where the microenvironment and in particular LM play an important yet poorly understood role in tissue maintenance, and in cancer progression which represents an inherent risk of IBD. Here we showed first that in human IBD colonic samples and in murine colitis the LMα1 and LMα5 chains are specifically and ectopically overexpressed with a concomitant nuclear p53 accumulation. Linked to this observation, we provided a mechanism showing that p53 induces LMα1 expression at the promoter level by ChIP analysis and this was confirmed by knockdown in cell transfection experiments. To mimic the human disease, we induced colitis and colitis-associated cancer by chemical treatment (DSS) combined or not with a carcinogen (AOM) in transgenic mice overexpressing LMα1 or LMα5 specifically in the intestine. We demonstrated that high LMα1 or LMα5 expression decreased susceptibility towards experimentally DSS-induced colon inflammation as assessed by histological scoring and decrease of pro-inflammatory cytokines. Yet in a pro-oncogenic context, we showed that LM would favor tumorigenesis as revealed by enhanced tumor lesion formation in both LM transgenic mice. Altogether, our results showed that nuclear p53 and associated overexpression of LMα1 and LMα5 protect tissue from inflammation. But in a mutation setting, the same LM molecules favor progression of IBD into colitis-associated cancer. Our transgenic mice represent attractive new models to acquire knowledge about the paradoxical effect of LM that mediate either tissue reparation or cancer according to the microenvironment. In the early phases of IBD, reinforcing basement membrane stability/organization could be a promising therapeutic approach.


Asunto(s)
Carcinoma/metabolismo , Neoplasias del Colon/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Laminina/metabolismo , Animales , Células CACO-2 , Citocinas/metabolismo , Células HCT116 , Células HT29 , Humanos , Laminina/genética , Ratones , Proteína p53 Supresora de Tumor/metabolismo
7.
Cell Rep ; 5(2): 482-92, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-24139798

RESUMEN

The extracellular matrix molecule tenascin-C (TNC) is a major component of the cancer-specific matrix, and high TNC expression is linked to poor prognosis in several cancers. To provide a comprehensive understanding of TNC's functions in cancer, we established an immune-competent transgenic mouse model of pancreatic ß-cell carcinogenesis with varying levels of TNC expression and compared stochastic neuroendocrine tumor formation in abundance or absence of TNC. We show that TNC promotes tumor cell survival, the angiogenic switch, more and leaky vessels, carcinoma progression, and lung micrometastasis. TNC downregulates Dickkopf-1 (DKK1) promoter activity through the blocking of actin stress fiber formation, activates Wnt signaling, and induces Wnt target genes in tumor and endothelial cells. Our results implicate DKK1 downregulation as an important mechanism underlying TNC-enhanced tumor progression through the provision of a proangiogenic tumor microenvironment.


Asunto(s)
Regulación hacia Abajo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Tenascina/metabolismo , Proteínas Wnt/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Transducción de Señal , Tenascina/deficiencia , Tenascina/genética , Proteínas Wnt/antagonistas & inhibidores
9.
PLoS One ; 7(5): e37710, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22666383

RESUMEN

Laminins are major constituents of basement membranes and are essential for tissue homeostasis. Laminin-511 is highly expressed in the intestine and its absence causes severe malformation of the intestine and embryonic lethality. To understand the mechanistic role of laminin-511 in tissue homeostasis, we used RNA profiling of embryonic intestinal tissue of lama5 knockout mice and identified a lama5 specific gene expression signature. By combining cell culture experiments with mediated knockdown approaches, we provide a mechanistic link between laminin α5 gene deficiency and the physiological phenotype. We show that laminin α5 plays a crucial role in both epithelial and mesenchymal cell behavior by inhibiting Wnt and activating PI3K signaling. We conclude that conflicting signals are elicited in the absence of lama5, which alter cell adhesion, migration as well as epithelial and muscle differentiation. Conversely, adhesion to laminin-511 may serve as a potent regulator of known interconnected PI3K/Akt and Wnt signaling pathways. Thus deregulated adhesion to laminin-511 may be instrumental in diseases such as human pathologies of the gut where laminin-511 is abnormally expressed as it is shown here.


Asunto(s)
Mucosa Intestinal/metabolismo , Intestinos/citología , Laminina/deficiencia , Laminina/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Proteínas Wnt/metabolismo , Animales , Biomarcadores/metabolismo , Adhesión Celular/genética , Diferenciación Celular/genética , Técnicas de Inactivación de Genes , Humanos , Mucosa Intestinal/citología , Intestinos/embriología , Ratones , Células Musculares/citología , Organogénesis/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Transcriptoma/genética
10.
Mol Cancer ; 9(1): 65, 2010 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-20307265

RESUMEN

BACKGROUND: Functional loss of the tumor suppressor Smad4 is involved in pancreatic and colorectal carcinogenesis and has been associated with the acquisition of invasiveness. We have previously demonstrated that the heterotrimeric basement membrane protein laminin-332 is a Smad4 target. Namely, Smad4 functions as a positive transcriptional regulator of all three genes encoding laminin-332; its loss is thus implicated in the reduced or discontinuous deposition of the heterotrimeric basement membrane molecule as evident in carcinomas. Uncoupled expression of laminin genes, on the other hand, namely overexpression of the laminin-gamma2 chain is an impressive marker at invasive edges of carcinomas where tumor cells are maximally exposed to signals from stromal cell types like macrophages. As Smad4 is characterized as an integrator of multiple extracellular stimuli in a strongly contextual manner, we asked if loss of Smad4 may also be involved in uncoupled expression of laminin genes in response to altered environmental stimuli. Here, we address Smad4 dependent effects of the prominent inflammatory cytokine TNFalpha on tumor cells. RESULTS: Smad4-reconstituted colon carcinoma cells like adenoma cells respond to TNFalpha with an increased expression of all three chains encoding laminin-332; coincubation with TGFbeta and TNFalpha leads to synergistic induction and to the secretion of large amounts of the heterotrimer. In contrast, in Smad4-deficient cells TNFalpha can induce expression of the gamma2 and beta3 but not the alpha3 chain. Surprisingly, this uncoupled induction of laminin-332 chains in Smad4-negative cells rather than causing intracellular accumulation is followed by the release of gamma2 into the medium, either in a monomeric form or in complexes with as yet unknown proteins. Soluble gamma2 is associated with increased cell migration. CONCLUSIONS: Loss of Smad4 may lead to uncoupled induction of laminin-gamma2 in response to TNFalpha and may therefore represent one of the mechanisms which underlie accumulation of laminin-gamma2 at the invasive margin of a tumor. The finding, that gamma2 is secreted from tumor cells in significant amounts and is associated with increased cell migration may pave the way for further investigation to better understand its functional relevance for tumor progression.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Laminina/metabolismo , Proteína Smad4/deficiencia , Factor de Necrosis Tumoral alfa/farmacología , Adenoma/metabolismo , Secuencia de Aminoácidos , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Sinergismo Farmacológico , Técnicas de Silenciamiento del Gen , Humanos , Laminina/química , Espectrometría de Masas , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Conformación Proteica , Proteína Smad4/genética , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Kalinina
11.
J Biol Chem ; 285(10): 7697-711, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20048158

RESUMEN

The Neuromutagenesis Facility at the Jackson Laboratory generated a mouse model of retinal vasculopathy, nmf223, which is characterized clinically by vitreal fibroplasia and vessel tortuosity. nmf223 homozygotes also have reduced electroretinogram responses, which are coupled histologically with a thinning of the inner nuclear layer. The nmf223 locus was mapped to chromosome 17, and a missense mutation was identified in Lama1 that leads to the substitution of cysteine for a tyrosine at amino acid 265 of laminin alpha1, a basement membrane protein. Despite normal localization of laminin alpha1 and other components of the inner limiting membrane, a reduced integrity of this structure was suggested by ectopic cells and blood vessels within the vitreous. Immunohistochemical characterization of nmf223 homozygous retinas demonstrated the abnormal migration of retinal astrocytes into the vitreous along with the persistence of hyaloid vasculature. The Y265C mutation significantly reduced laminin N-terminal domain (LN) interactions in a bacterial two-hybrid system. Therefore, this mutation could affect interactions between laminin alpha1 and other laminin chains. To expand upon these findings, a Lama1 null mutant, Lama1(tm1.1Olf), was generated that exhibits a similar but more severe retinal phenotype than that seen in nmf223 homozygotes. The increased severity of the Lama1 null mutant phenotype is probably due to the complete loss of the inner limiting membrane in these mice. This first report of viable Lama1 mouse mutants emphasizes the importance of this gene in retinal development. The data presented herein suggest that hypomorphic mutations in human LAMA1 could lead to retinal disease.


Asunto(s)
Laminina , Mutación Missense , Isoformas de Proteínas , Retina , Enfermedades de la Retina , Vasos Retinianos , Adulto , Secuencia de Aminoácidos , Animales , Astrocitos/citología , Astrocitos/metabolismo , Membrana Basal/citología , Membrana Basal/metabolismo , Electrorretinografía , Femenino , Prueba de Complementación Genética , Humanos , Laminina/genética , Laminina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Fenotipo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Retina/anomalías , Retina/anatomía & histología , Retina/fisiología , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología , Vasos Retinianos/anomalías , Vasos Retinianos/anatomía & histología , Vasos Retinianos/fisiología , Alineación de Secuencia , Transgenes
12.
Physiol Genomics ; 27(2): 141-55, 2006 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-16868071

RESUMEN

DNA-binding transcription factors bind to promoters that carry their binding sites. Transcription factors therefore function as nodes in gene regulatory networks. In the present work we used a bioinformatic approach to search for transcription factors that might function as nodes in gene regulatory networks during the differentiation of the small intestinal epithelial cell. In addition we have searched for connections between transcription factors and the villus metabolome. Transcriptome data were generated from mouse small intestinal villus, crypt, and fetal intestinal epithelial cells. Metabolome data were generated from crypt and villus cells. Our results show that genes that are upregulated during fetal to adult and crypt to villus differentiation have an overrepresentation of potential hepatocyte nuclear factor (HNF)-4 binding sites in their promoters. Moreover, metabolome analyses by magic angle spinning (1)H nuclear magnetic resonance spectroscopy showed that the villus epithelial cells contain higher concentrations of lipid carbon chains than the crypt cells. These findings suggest a model where the HNF-4 transcription factor influences the villus metabolome by regulating genes that are involved in lipid metabolism. Our approach also identifies transcription factors of importance for crypt functions such as DNA replication (E2F) and stem cell maintenance (c-Myc).


Asunto(s)
Enterocitos/metabolismo , Regulación de la Expresión Génica/fisiología , Factor Nuclear 4 del Hepatocito/fisiología , Transcripción Genética/fisiología , Algoritmos , Animales , Sitios de Unión , Diferenciación Celular/genética , Clonación Molecular , Replicación del ADN/genética , ADN Recombinante/genética , ADN Recombinante/metabolismo , Factores de Transcripción E2F/genética , Factores de Transcripción E2F/fisiología , Endodermo/citología , Enterocitos/citología , Enterocitos/ultraestructura , Genes myc , Genómica/métodos , Íleon/citología , Mucosa Intestinal/citología , Mucosa Intestinal/embriología , Mucosa Intestinal/crecimiento & desarrollo , Metabolismo de los Lípidos/genética , Espectroscopía de Resonancia Magnética , Metaloendopeptidasas/genética , Ratones , Ratones Endogámicos C57BL , Microvellosidades/metabolismo , Modelos Genéticos , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-myc/fisiología , Células Madre/citología , Biología de Sistemas/métodos
13.
Eur J Cell Biol ; 85(8): 769-83, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16762451

RESUMEN

The deleted in colorectal cancer (DCC) gene encodes a 170- to 190-kDa protein of the Immunoglobulin superfamily. Firstly identified as a tumor suppressor gene in human colorectal carcinomas, the main function for DCC has been described in the nervous system as part of a receptor complex for netrin-1. Moreover, roles in mucosecretory cell differentiation and as inducer of apoptosis have also been reported. DCC knockout mice supported a crucial role for this gene in axonal migration, yet questioned its implication in tumor suppression and mucosecretory differentiation. The work presented here demonstrates that a DCC-transfected HT-29 colonic human cell line (HT-29/DCC) displays an increase in cell-cell adhesion to the detriment of cell-matrix interactions: HT-29/DCC cells exhibit more and better-structured desmosomes while focal adhesions and hemidesmosomes are disrupted. HT-29/DCC cells show no changes in adherent junctions but upon treatment with TPA, HT-29/DCC cells show resistance to scattering, and maintain E-cadherin in the membrane. In addition, the actin cytoskeleton is affected in HT-29/DCC cells: stress fibers are disrupted while cortical actin remains intact. We identified a putative ERM-M (ezrin/radixin/moesin and merlin) binding domain in the juxtamembrane region of the DCC protein. In vitro pull-down assays demonstrate the interaction of the DCC cytoplasmic domain with the N-terminal region of ezrin and merlin, and co-immunoprecipitation assays in transiently DCC-transfected COS-1 cells showed that the interaction between DCC and ezrin also takes place in vivo. Altogether, our results suggest that DCC could regulate cell adhesion and migration through its association with ERM-M proteins.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Western Blotting , Adhesión Celular/fisiología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Receptor DCC , Desmosomas/metabolismo , Desmosomas/ultraestructura , Matriz Extracelular/metabolismo , Células HT29 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Proteínas de Microfilamentos/metabolismo , Microscopía Electrónica , Modelos Genéticos , Datos de Secuencia Molecular , Neurofibromina 2/metabolismo , Unión Proteica , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/fisiología , Transfección/métodos , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología
14.
J Cell Physiol ; 206(2): 545-55, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16245305

RESUMEN

Intestinal epithelial cells are characterized by continuous renewal and differentiation events, which may be influenced by the basement membrane, and in particular laminins, which are major components of this specialized extracellular matrix. The function and signaling pathways of laminins in these processes are still poorly documented. In this study, we investigated the possible role and the subcellular localization of nucleolin, a nuclear shuttling protein, in relation to differentiation of human intestinal epithelial Caco2/TC7 cells triggered by exogenous laminin-1. Immunofluorescence and Western blot analysis indicated that laminin-1 induced early differentiation of the cells concomitantly to a decrease in nuclear nucleolin and its a cell surface location. We also showed that both effects of laminin-1 on Caco2/TC7 cells--induction of the differentiation marker sucrase-isomaltase and redistribution of nucleolin--could be mediated by a beta1-integrin dependent cascade that implicated activation of the p38 MAPK pathway. In addition, knock-down of nucleolin expression by the small interfering RNA strategy mimicked the effect of laminin-1 as it resulted in the induction of cell polarization and differentiation. Thus, our study suggests that changes in the subcellular distribution and expression level of nucleolin play an important role in intestinal cell differentiation and relay the signaling pathway induced by laminin-1.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Laminina/farmacología , Laminina/fisiología , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/metabolismo , Fracciones Subcelulares/metabolismo , Secuencia de Bases , Transporte Biológico , Células CACO-2 , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Cadenas beta de Integrinas/metabolismo , Laminina/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Transducción de Señal , Fracciones Subcelulares/efectos de los fármacos , Nucleolina
15.
Exp Cell Res ; 303(2): 494-503, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15652360

RESUMEN

Laminins are structurally and functionally major components of the extracellular matrix. Four isoforms of laminins (laminin-1, -2, -5 and -10) are expressed in a specific pattern along the crypt-villus axis of the intestine. Previous works indicated that expression of these isoforms is developmentally regulated and that laminins could modulate the behaviour of intestinal cells, but the exact role of each isoform remained unclear. Here, we report the first systematic analysis of the cellular functions of the four isoforms using the human colon adenocarcinoma Caco2/TC7 cell line as a model. We compared the respective abilities of each isoform to modulate adhesion, proliferation and differentiation of intestinal epithelial cells. We found that the isoforms were functionally distinct, with laminin-10 being the most adhesive substratum, laminin-2, laminin-5 and laminin-10 enhancing cellular proliferation and at the opposite, laminin-1 stimulating intestinal cell differentiation. To begin to characterise the molecular events induced by the different isoforms, we examined by immunofluorescence the intracellular distribution of several nuclear proteins, recently highlighted by a nuclear proteomic approach. We observed clear nucleocytoplasmic redistribution of these proteins, which depended on the laminin isoform. These results provide evidence for a distinct functional role of laminins in intestinal cell functions characterised by specific localisation of nuclear proteins.


Asunto(s)
Mucosa Intestinal/metabolismo , Laminina/metabolismo , Proteínas Nucleares/metabolismo , Secuencia de Bases , Células CACO-2 , Adhesión Celular , Diferenciación Celular , Proliferación Celular , Citoplasma/metabolismo , ADN Complementario/genética , Células Epiteliales/metabolismo , Células HT29 , Humanos , Inmunohistoquímica , Laminina/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
16.
Genes Dev ; 18(12): 1385-90, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15198980

RESUMEN

Although Apc is well characterized as a tumor-suppressor gene in the intestine, the precise mechanism of this suppression remains to be defined. Using a novel inducible Ahcre transgenic line in conjunction with a loxP-flanked Apc allele we, show that loss of Apc acutely activates Wnt signaling through the nuclear accumulation of beta-catenin. Coincidentally, it perturbs differentiation, migration, proliferation, and apoptosis, such that Apc-deficient cells maintain a "crypt progenitor-like" phenotype. Critically, for the first time we confirm a series of Wnt target molecules in an in vivo setting and also identify a series of new candidate targets within the same setting.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Proteínas Proto-Oncogénicas/fisiología , Animales , Diferenciación Celular , Movimiento Celular , Proteínas del Citoesqueleto/metabolismo , Inmunohistoquímica , Mucosa Intestinal/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Fenotipo , ARN Mensajero/análisis , ARN Mensajero/aislamiento & purificación , Transducción de Señal , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Wnt , beta Catenina
17.
Cancer Res ; 64(1): 180-8, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-14729622

RESUMEN

Clear cell renal carcinoma (CCRC) is responsible for 2% of cancer-related deaths worldwide and is resistant to virtually all therapies, indicating the importance of a search for new therapeutic targets. Parathyroid hormone-related protein (PTHrP) is a polyprotein derived from normal and malignant cells that regulates cell growth. In the current study, we show that blocking PTHrP with antibodies or antagonizing the common parathyroid hormone (PTH)/PTHrP receptor, the PTH1 receptor, dramatically blunts the expansion of human CCRC in vitro by promoting cell death. Importantly, in nude mice, anti-PTHrP antibodies induced complete regression of 70% of the implanted tumors by inducing cell death. In addition, we demonstrate that the von Hippel-Lindau tumor suppressor protein, which functions as a gatekeeper for CCRC, negatively regulates PTHrP expression at the post-transcriptional level. These studies indicate that PTHrP is an essential growth factor for CCRC and is a novel target for the von Hippel-Lindau tumor suppressor protein. Taken together, these results strongly suggest that targeting the PTHrP/PTH1 receptor system may provide a new avenue for the treatment of this aggressive cancer in humans.


Asunto(s)
Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Apoptosis , Carcinoma de Células Renales/genética , División Celular , Sustancias de Crecimiento/genética , Humanos , Neoplasias Renales/genética , Ratones , Ratones Desnudos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau , Enfermedad de von Hippel-Lindau/genética
18.
Proteomics ; 4(1): 93-105, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14730675

RESUMEN

Self-renewing tissues such as the intestine contain progenitor proliferating cells which subsequently differentiate. Cell proliferation and differentiation involve gene regulation processes which take place in the nucleus. A human intestinal epithelial cell line model (Caco2/TC7) which reproduces these dynamic processes has been used to perform proteomic studies on nuclear proteins. Nuclei from Caco2/TC7 cells at proliferative and differentiated stages were purified by subcellular fractionation. After two-dimensional gel electrophoresis separation and ruthenium staining, 400 protein spots were detected by image analysis. Eighty-five spots corresponding to 60 different proteins were identified by matrix-assisted laser desorption/ionization mass spectrometry in nuclei from proliferative cells. Comparison of nuclear proteomes from proliferative or differentiated cells by differential display resulted in the identification of differentially expressed proteins such as nucleolin, hnRNP A2/B1 and hnRNP A1. By using Western blot analysis, we found that the expression and number of specific isoforms of these nuclear proteins decreased in differentiated cells. Immunocytochemistry experiments also showed that in proliferative cells nucleolin was distributed in nucleoli-like bodies. In contrast, hnRNPs A2/B1 and A1 were dispersed throughout the nucleus. This study of the nuclear proteome from intestinal epithelial cells represents the first step towards the establishment of a protein database which will be a valuable resource in future studies on the differential expression of nuclear proteins in response to physiological, pharmacological and pathological modulations.


Asunto(s)
Núcleo Celular/química , Colon/química , Proteoma/química , Células CACO-2 , División Celular/fisiología , Núcleo Celular/fisiología , Colon/citología , Colon/fisiología , Neoplasias del Colon/química , Electroforesis en Gel Bidimensional , Epitelio/química , Epitelio/fisiología , Humanos , Inmunohistoquímica , Proteoma/fisiología , Tinción con Nitrato de Plata , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
19.
J Biol Chem ; 279(10): 9103-14, 2004 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-14634001

RESUMEN

Laminin-1 (alpha1beta1gamma1), a basement membrane (BM) constituent, has been associated with differentiation processes and also with malignant progression. In the intestinal tissue, the alpha1 chain is expressed and secreted in the subepithelial BM during the developmental period; in the adult rodent tissue, it is restricted to the BM of the dividing cells. To understand how laminin alpha1 chain expression is regulated, we cloned and characterized a 2-kb promoter region of the Lama1 mouse gene. Analysis of the promoter was conducted in the Caco2-TC7 intestinal epithelial cells by transient transfection of serially deleted and site-directed mutated promoter constructs, by electrophoretic mobility shift assays, and expression of selected transcription factors. We determined that a proximal region, which includes an Sp1-binding GC box and a Krüppel-like element, was important for the promoter activity. This region is conserved between the human and mouse genes. Interestingly, two Krüppel-like factors KLF4 and KLF5 exhibit opposing effects on the Lama1 promoter activity that are decreased and increased, respectively, in the intestinal epithelial cells. These data corroborate the complementary expression of KLF4 and KLF5 along the intestinal crypt-villus axis and the parallel expression of KLF5 and laminin alpha1 chain in the crypt region. Finally, we showed that glucocorticoids stimulate the promoter activity. This study is the first characterization of the Lama1 promoter; we identified regulatory elements that may account for the expression pattern of the endogenous protein in the mouse intestine.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Laminina/genética , Transactivadores/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Línea Celular , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel , Laminina/metabolismo , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/efectos de los fármacos , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
20.
Biochem J ; 371(Pt 1): 211-21, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12519076

RESUMEN

The trimeric extracellular matrix molecule laminin-5 and its constituent chains (alpha 3, beta 3, gamma 2) are normally not detectable intracellularly in intestinal epithelial cells but the laminin gamma 2 chain can be detected in cancer cells at the invasive front of a subset of colon carcinomas. These cells are subjected to cytokines such as transforming growth factor beta 1 (TGF-beta 1) and hepatocyte growth factor (HGF), produced by the tumour cells or by the surrounding stromal cells. The purpose of the present work was to investigate whether TGF-beta 1 and HGF, known to stimulate the LAMC2 gene encoding the laminin gamma 2 chain, might synergize to activate the LAMC2 promoter, and to identify the promoter elements involved. We find evidence for synergy between TGF-beta and HGF with respect to laminin gamma 2 chain expression and promoter activation and demonstrate that this requires the 5' activator protein-1 (AP-1) element of the promoter and an additional upstream element which is also responsive to co-expression of the Smad3 protein from the TGF-beta signalling pathway. The transcripts encoding the other laminin-5 chains are not synergistically activated by HGF and TGF-beta. Thus the synergistic activation of the LAMC2 gene is mediated via different cis-elements and results in an overproduction of the laminin gamma 2 chain relative to the other laminin-5 constituent chains. This difference may explain why laminin gamma 2 chains accumulate in the cells at the invasive front of colon carcinomas.


Asunto(s)
Carcinoma/metabolismo , Neoplasias del Colon/metabolismo , Laminina/genética , Laminina/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal/fisiología , Carcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Sinergismo Farmacológico , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Secuencias Reguladoras de Ácidos Nucleicos , Elementos de Respuesta , Transducción de Señal/genética , Proteína smad3 , Proteína Smad4 , Transactivadores/genética , Transactivadores/metabolismo , Factor de Transcripción AP-1/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA