Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Development ; 147(4)2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31988184

RESUMEN

Integrin dimers α3/ß1, α6/ß1 and α6/ß4 are the mammary epithelial cell receptors for laminins, which are major components of the mammary basement membrane. The roles of specific basement membrane components and their integrin receptors in the regulation of functional gland development have not been analyzed in detail. To investigate the functions of laminin-binding integrins, we obtained mutant mice with mammary luminal cell-specific deficiencies of the α3 and α6 integrin chains generated using the Cre-Lox approach. During pregnancy, mutant mice displayed decreased luminal progenitor activity and retarded lobulo-alveolar development. Mammary glands appeared functional at the onset of lactation in mutant mice; however, myoepithelial cell morphology was markedly altered, suggesting cellular compensation mechanisms involving cytoskeleton reorganization. Notably, lactation was not sustained in mutant females, and the glands underwent precocious involution. Inactivation of the p53 gene rescued the growth defects but did not restore lactogenesis in mutant mice. These results suggest that the p53 pathway is involved in the control of mammary cell proliferation and survival downstream of laminin-binding integrins, and underline an essential role of cell interactions with laminin for lactogenic differentiation.


Asunto(s)
Integrinas/fisiología , Lactancia , Glándulas Mamarias Animales/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Supervivencia Celular , Citoesqueleto/fisiología , Progresión de la Enfermedad , Femenino , Eliminación de Gen , Hormonas/fisiología , Integrina alfa3/fisiología , Integrina alfa6/fisiología , Integrina beta1/fisiología , Integrina beta4/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Mutantes , Mutación , Células Madre Neoplásicas/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovario/fisiología , Fenotipo , Embarazo , Preñez , Pronóstico , Unión Proteica , Multimerización de Proteína
2.
Cell Death Differ ; 24(12): 2054-2065, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28777373

RESUMEN

MicroRNAs are important regulators of gene expression and are involved in cellular processes such as proliferation or differentiation, particularly during development of numerous organs including the inner ear. However, it remains unknown if miRNAs are required during the earliest stages of otocyst and cochlear duct development. Here, we report that a conditional loss of Dicer expression in the otocyst impairs the early development of the inner ear as a result of the accumulation of DNA damage that trigger p53-mediated apoptosis. Moreover, cochlear progenitors in the prosensory domain do not exit the cell cycle. Our unbiased approach identified ItgA3 as a target of miR-183, which are both enriched in the otic vesicle. We observed that the repression of integrin alpha 3 by miR-183 controls cell proliferation in the developing cochlea. Collectively, our results reveal that Dicer and miRNAs play essential roles in the regulation of early inner ear development.


Asunto(s)
Oído Interno/embriología , Integrina alfa3/fisiología , MicroARNs/fisiología , Animales , Diferenciación Celular/fisiología , Línea Celular , Cóclea/citología , Cóclea/embriología , ARN Helicasas DEAD-box/genética , Femenino , Ratones , Ratones Noqueados , Embarazo , Ribonucleasa III/genética , Transducción de Señal
3.
Hepatology ; 64(6): 2103-2117, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27639064

RESUMEN

In patients with hepatocellular carcinoma (HCC) receiving sorafenib, drug resistance is common. HCC develops in a microenvironment enriched with extracellular matrix proteins including laminin (Ln)-332, produced by hepatic stellate cells (HSCs). Ln-332 is the ligand of α3ß1 and α6ß4 integrins, differently expressed on the HCC cell surface, that deliver intracellular pathways. The aim of this study was to investigate the effect of Ln-332 on sorafenib's effectiveness. HCC cells were challenged with sorafenib in the presence of Ln-332 and of HSC conditioned medium (CM). Sorafenib impaired HCC cell proliferation and induced apoptosis. HSC-CM or Ln-332 inhibited sorafenib's effectiveness in HCC cells expressing both α3ß1 and α6ß4. Inhibiting α3 but not α6 integrin subunit using blocking antibodies or small interfering RNA abrogated the protection induced by Ln-332 and HSC-CM. Hep3B cells expressing α6ß4 but lacking the α3 integrin were insensitive to Ln-332 and HSC-CM protective effects. Hep3B α3-positive, but not wild-type and scramble transfected, cells acquired protection by sorafenib when plated on Ln-332-CM or HSCs. Sorafenib dephosphorylated focal adhesion kinase (FAK) and extracellular signal-regulated kinases 1/2, whereas Ln-332 and HSC-CM partially restored the pathways. Silencing FAK, but not extracellular signal-regulated kinases 1/2, abrogated the protection induced by Ln-332 and HSC-CM, suggesting a specific role for FAK. Sorafenib down-regulated total FAK, inducing its proteasomal degradation, while Ln-332 and HSC-CM promoted the escape of FAK from ubiquitination, probably inducing a preferential membrane localization. CONCLUSION: This study unveils a novel mechanism of sorafenib resistance depending on the α3ß1/Ln-332 axis and requiring FAK ubiquitination, providing new insights into personalizing therapy for patients with HCC. (Hepatology 2016;64:2103-2117).


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Resistencia a Antineoplásicos , Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Células Estrelladas Hepáticas/fisiología , Integrina alfa3/fisiología , Laminina/fisiología , Neoplasias Hepáticas/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Ubiquitinación , Humanos , Niacinamida/uso terapéutico , Sorafenib , Células Tumorales Cultivadas
4.
Br J Cancer ; 108(12): 2516-24, 2013 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-23652300

RESUMEN

BACKGROUND: Glioma stem-like cell (GSC) properties are responsible for gliomagenesis and recurrence. GSCs are invasive but its mechanism remains to be elucidated. Here, we attempted to identify the molecules that promote invasion in GSCs. METHODS: Neurospheres and CD133⁺ cells were collected from glioblastoma (GBM) specimens and glioma cell lines by sphere-formation method and magnetic affinity cell sorting, respectively. Differential expression of gene candidates, its role in invasion and its signaling pathway were evaluated in glioma cell lines. RESULTS: Neurospheres from surgical specimens attached to fibronectin and laminin, the receptors of which belong to the integrin family. Integrin α3 was overexpressed in CD133⁺ cells compared with CD133⁻ cells in all the glioma cell lines (4 out of 4). Immunohistochemistry demonstrated the localisation of integrin α3 in GBM cells, including invading cells, and in the tumour cells around the vessels, which is believed to be a stem cell niche. The expression of integrin α3 was correlated with migration and invasion. The invasion activity of glioma cells was linked to the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2. CONCLUSION: Our results suggest that integrin α3 contributes to the invasive nature of GSCs via ERK1/2, which renders integrin α3 a prime candidate for anti-invasion therapy for GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Integrina alfa3/genética , Integrina alfa3/fisiología , Células Madre Neoplásicas/metabolismo , Neoplasias Encefálicas/genética , Adhesión Celular/genética , Movimiento Celular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibronectinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Humanos , Integrina alfa3/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Invasividad Neoplásica , Células Madre Neoplásicas/patología , Fosforilación , Células Tumorales Cultivadas , Regulación hacia Arriba/genética
5.
J Invest Dermatol ; 132(2): 448-57, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21956124

RESUMEN

Mouse keratinocytes migrate significantly slower than their human counterparts in vitro on uncoated surfaces. We tested the hypothesis that this is a consequence of differences in the extracellular matrix (ECM) that cells deposit. In support of this, human keratinocyte motility was markedly reduced when plated onto the ECM of mouse skin cells, whereas the latter cells migrated faster when plated onto human keratinocyte ECM. The ECM of mouse and human keratinocytes contained similar levels of the α3 laminin subunit of laminin-332. However, mouse skin cells expressed significantly more fibronectin (FN) than human cells. To assess whether FN is a motility regulator, we used small interfering RNA (siRNA) to reduce the expression of FN in mouse keratinocytes. The treated mouse keratinocytes moved significantly more rapidly than wild-type mouse skin cells. Moreover, the FN-depleted mouse cell ECM supported increased migration of both mouse and human keratinocytes. Furthermore, the motility of human keratinocytes was slowed when plated onto FN-coated substrates or human keratinocyte ECM supplemented with FN in a dose-dependent manner. Consistent with these findings, the ECM of α3 integrin-null keratinocytes, which also migrated faster than wild-type cells, was FN deficient. Our results provide evidence that FN is a brake to skin cell migration supported by laminin-332-rich matrices.


Asunto(s)
Movimiento Celular , Fibronectinas/fisiología , Queratinocitos/fisiología , Animales , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/fisiología , Línea Celular , Humanos , Integrina alfa3/fisiología , Ratones , Ratones Endogámicos C57BL , Kalinina
6.
Osteoarthritis Cartilage ; 18(10): 1300-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20633668

RESUMEN

OBJECTIVE: We previously reported that more than 60% of synovial mesenchymal stem cells (MSCs) placed on osteochondral defects adhered to the defect within 10 min and promoted cartilage regeneration. The efficiency of adherence is considered to depend on the interaction between cells and extracellular matrix (ECM), in which integrins may play some important roles. Divalent cations such as calcium, magnesium, and manganese may affect functions of integrins, and the integrins may be involved in differentiation of MSCs. Among divalent cations, magnesium is used in clinical practice as a therapeutic agent and increases the affinity of integrin to ECM. In this study, we investigated whether magnesium enhanced adherence and chondrogenesis of synovial MSC through integrins. METHODS: We performed assays for adherence of human synovial MSCs to collagen-coated slides, in vitro chondrogenesis, ex vivo assays for adherence of human synovial MSCs to osteochondral defect, and in vivo assays for adherence and cartilage formation of synovial MSCs in a rabbit osteochondral defect model. RESULTS: Magnesium increased adhesion of human synovial MSCs to collagen, and this effect was inhibited by neutralizing antibodies for integrin α3 and ß1. Magnesium also promoted synthesis of cartilage matrix during in vitro chondrogenesis of synovial MSCs, which was diminished by neutralizing antibodies for integrin ß1 but not for integrin α3. Ex vivo analyses demonstrated that magnesium enhanced adherence of human synovial MSCs to osteochondral defects. In vivo studies in rabbits showed that magnesium promoted adherence at 1 day and cartilage formation of synovial MSCs at 2 weeks. CONCLUSION: Magnesium enhanced adherence of synovial MSCs through integrins, which promoted synthesis of cartilage matrix at an early phase.


Asunto(s)
Condrogénesis/efectos de los fármacos , Integrinas/fisiología , Magnesio/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Membrana Sinovial/efectos de los fármacos , Adulto , Animales , Cartílago Articular/fisiología , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Humanos , Integrina alfa3/fisiología , Integrina beta1/fisiología , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Conejos , Regeneración , Membrana Sinovial/citología
7.
J Periodontal Res ; 45(2): 284-91, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20470260

RESUMEN

BACKGROUND AND OBJECTIVE: It remains controversial whether or not the junctional epithelium cells that are directly attached to teeth migrate on the enamel surface, as those cells are able to adhere firmly to the enamel. The aim of this study was to investigate the expression patterns of laminin gamma(2), integrin beta(4) and integrin alpha(3), and to examine their potential function in cell migration. MATERIAL AND METHODS: Oral epithelium cells obtained from Sprague-Dawley rats were established in primary culture. We employed a wound-healing assay to characterize the direction of cell extension at the start of cell migration, and observed different localizations of laminin and integrins using immunofluorescence. For functional analyses of integrins, we employed a phosphatidylinositol-3-kinase (PI3K) activator to promote integrin beta(4) function and used P1B5 to inhibit integrin alpha(3) function, and we analyzed the percentage of re-epithelialization as the migration function. RESULTS: Marked accumulation of laminin gamma(2) was detected in the peripheral cytoplasm of cells adjacent to the wound area, as shown by the results of the migration assay. Integrin beta(4) was detected in the distal cell processes of actively migrating cells, while integrin alpha(3) was found in cell membranes of cells adjacent to the wound area. In the functional analyses, the percentage of re-epithelialization was significantly lower in the PI3K-activator group and in the P1B5-treated group (2.5% and 7.2%, respectively) than in the control group (39.0%) (p < 0.01). CONCLUSION: The results suggest that laminin gamma(2) is secreted as a foothold for cell migration, that integrin beta(4) participates in cell adhesion and that integrin alpha(3) is involved in cell migration in the primary culture cells.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Inserción Epitelial/citología , Integrina alfa3/fisiología , Integrina beta4/fisiología , Animales , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/análisis , Membrana Celular/ultraestructura , Movimiento Celular/fisiología , Núcleo Celular/ultraestructura , Extensiones de la Superficie Celular/ultraestructura , Células Cultivadas , Colorantes , Citoplasma/ultraestructura , Activación Enzimática , Inserción Epitelial/fisiología , Células Epiteliales/fisiología , Células Epiteliales/ultraestructura , Técnica del Anticuerpo Fluorescente , Integrina alfa3/análisis , Integrina alfa3/efectos de los fármacos , Integrina beta4/análisis , Integrina beta4/efectos de los fármacos , Microscopía Confocal , Fosfatidilinositol 3-Quinasas/farmacología , Ratas , Ratas Sprague-Dawley , Cicatrización de Heridas/fisiología , Kalinina
8.
Cancer Res ; 69(11): 4835-42, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19458077

RESUMEN

The WW domain-containing oxidoreductase (WWOX) gene is located at FRA16D, a common fragile site involved in human cancer. Targeted deletion of Wwox in mice causes increased spontaneous tumor incidence, confirming that WWOX is a bona fide tumor suppressor gene. We show that stable transfection of WWOX into human PEO1 ovarian cancer cells, containing homozygous WWOX deletion, abolishes in vivo tumorigenicity, but this does not correlate with alteration of in vitro growth. Rather, WWOX restoration in PEO1, or WWOX overexpression in SKOV3 ovarian cancer cells, results in reduced attachment and migration on fibronectin, an extracellular matrix component linked to peritoneal metastasis. Conversely, siRNA-mediated knockdown of endogenous WWOX in A2780 ovarian cancer cells increases adhesion to fibronectin. In addition, whereas there is no WWOX-dependent difference in cell death in adherent cells, WWOX-transfected cells in suspension culture display a proapoptotic phenotype. We further show that WWOX expression reduces membranous integrin alpha(3) protein but not integrin alpha(3) mRNA levels, and that adhesion of PEO1 cells to fibronectin is predominantly mediated through integrin alpha(3). We therefore propose that WWOX acts as an ovarian tumor suppressor by modulating the interaction between tumor cells and the extracellular matrix and by inducing apoptosis in detached cells. Consistent with this, the suppression of PEO1 tumorigenicity by WWOX can be partially overcome by implanting these tumor cells in Matrigel. These data suggest a possible role for the loss of WWOX in the peritoneal dissemination of human ovarian cancer cells.


Asunto(s)
Carcinoma/genética , Fibronectinas/metabolismo , Integrina alfa3/metabolismo , Neoplasias Ováricas/genética , Oxidorreductasas/genética , Proteínas Supresoras de Tumor/genética , Animales , Apoptosis/genética , Carcinoma/metabolismo , Carcinoma/patología , Adhesión Celular/genética , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Integrina alfa3/fisiología , Ratones , Ratones Desnudos , Invasividad Neoplásica , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/secundario , Unión Proteica/genética , Transfección , Oxidorreductasa que Contiene Dominios WW , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Learn Mem ; 14(9): 606-15, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17848500

RESUMEN

Integrins comprise a large family of heterodimeric, transmembrane cell adhesion receptors that mediate diverse neuronal functions in the developing and adult CNS. Recent pharmacological and genetic studies have suggested that beta1-integrins are critical in synaptic plasticity and memory formation. To further define the role of integrins in these processes, we generated a postnatal forebrain and excitatory neuron-specific knockout of alpha3-integrin, one of several binding partners for beta1 subunit. At hippocampal Schaffer collateral-CA1 synapses, deletion of alpha3-integrin resulted in impaired long-term potentiation (LTP). Basal synaptic transmission and paired-pulse facilitation were normal in the absence of alpha3-integrin. Behavioral studies demonstrated that the mutant mice were selectively defective in a hippocampus-dependent, nonmatch-to-place working memory task, but were normal in other hippocampus-dependent spatial tasks. The impairment in LTP and working memory is similar to that observed in beta1-integrin conditional knockout mice, suggesting that alpha3-integrin is the functional binding partner for beta1 for these processes in the forebrain.


Asunto(s)
Hipocampo/fisiología , Integrina alfa3/fisiología , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Animales , Electrofisiología , Eliminación de Gen , Hipocampo/ultraestructura , Técnicas In Vitro , Integrina alfa3/genética , Aprendizaje por Laberinto , Trastornos de la Memoria/genética , Trastornos de la Memoria/fisiopatología , Trastornos de la Memoria/psicología , Ratones , Ratones Noqueados , Microscopía Electrónica , Destreza Motora , Prosencéfalo/metabolismo , Desempeño Psicomotor , Natación , Sinapsis/metabolismo
10.
Dev Biol ; 308(1): 15-29, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17601529

RESUMEN

Laminin alpha chains have unique spatiotemporal expression patterns during development and defining their function is necessary to understand the regulation of epithelial morphogenesis. We investigated the function of laminin alpha5 in mouse submandibular glands (SMGs). Lama5(-/-) SMGs have a striking phenotype: epithelial clefting is delayed, although proliferation occurs; there is decreased FGFR1b and FGFR2b, but no difference in Lama1 expression; later in development, epithelial cell organization and lumen formation are disrupted. In wild-type SMGs alpha5 and alpha1 are present in epithelial clefts but as branching begins alpha5 expression increases while alpha1 decreases. Lama5 siRNA decreased branching, p42 MAPK phosphorylation, and FGFR expression, and branching was rescued by FGF10. FGFR siRNA decreased Lama5 suggesting that FGFR signaling provides positive feedback for Lama5 expression. Anti-beta1 integrin antibodies decreased FGFR and Lama5 expression, suggesting that beta1 integrin signaling provides positive feedback for Lama5 and FGFR expression. Interestingly, the Itga3(-/-):Itga6(-/-) SMGs have a similar phenotype to Lama5(-/-). Our findings suggest that laminin alpha5 controls SMG epithelial morphogenesis through beta1 integrin signaling by regulating FGFR expression, which also reciprocally regulates the expression of Lama5. These data link changes in basement membrane composition during branching morphogenesis with FGFR expression and signaling.


Asunto(s)
Integrina beta1/fisiología , Laminina/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/genética , Glándula Submandibular/embriología , Animales , Proliferación Celular , Epitelio/embriología , Retroalimentación , Factor 1 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Integrina alfa3/genética , Integrina alfa3/fisiología , Integrina alfa6/genética , Integrina alfa6/fisiología , Laminina/deficiencia , Laminina/genética , Ratones , Ratones Noqueados , Modelos Biológicos , Morfogénesis , Fenotipo , ARN Interferente Pequeño/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Transducción de Señal , Glándula Submandibular/fisiología
11.
Dev Cell ; 11(3): 325-37, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16950124

RESUMEN

Shh-Gli signaling controls cell fates in the developing ventral neural tube by regulating the patterned expression of transcription factors in neural progenitors. However, the molecular mechanisms that limit target gene responses to specific domains are unclear. Here, we show that Wnt pathway inhibitors regulate the threshold response of a ventral Shh target gene, Nkx2.2, to establish its restricted expression in the ventral spinal cord. Identification and characterization of an Nkx2.2 enhancer reveals that expression is directly regulated by positive Shh-Gli signaling and negative Tcf repressor activity. Our data indicate that the dorsal limit of Nkx2.2 is controlled by Tcf4-mediated transcriptional repression, and not by a direct requirement for high-level Shh-Gli signaling, arguing against a simple model based on differential Gli factor affinities in target genes. These results identify a transcriptional mechanism that integrates graded Shh and Wnt signaling to define progenitor gene expression domains and cell fates in the neural tube.


Asunto(s)
Sistema Nervioso Central/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Transcripción Genética , Proteínas Wnt/antagonistas & inhibidores , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Biomarcadores/análisis , Pollos , Secuencia Conservada , Elementos de Facilitación Genéticos , Proteínas del Ojo/genética , Proteínas Hedgehog , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Integrina alfa3/fisiología , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Transgénicos , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Neuronas/química , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Proteínas Represoras/genética , Factores de Transcripción TCF/genética , Factor de Transcripción 4 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Proteínas Wnt/metabolismo , Proteínas de Pez Cebra , Proteína con Dedos de Zinc GLI1
12.
Differentiation ; 74(2-3): 105-18, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16533309

RESUMEN

Satellite cells are myogenic precursor cells, participating in growth, and regeneration of skeletal muscles. The proteins that play a role in myogenesis are integrins. In this report, we show that the integrin alpha3 subunit is expressed in quiescent satellite cells and activated myoblasts. We also find that in myoblasts the integrin alpha3 subunit is localized at cell-cell and cell-extracellular matrix contacts. We notice that increase in protein and mRNA encoding the integrin alpha3 subunit accompanies myoblast differentiation. Using double immunofluorescence and immunoprecipitation experiments, we demonstrate that the integrin alpha3 subunit co-localizes with actin, and binds the integrin beta1 subunit and ADAM12, suggesting that the complex alpha3beta1/ADAM12 is probably involved in myoblast fusion. Importantly, overexpression of the full-length integrin alpha3 subunit increases myoblast fusion whereas an antibody against its extracellular domain inhibits fusion. These data demonstrate that the integrin alpha3 subunit may contribute to satellite cell activation and then myoblast adhesion and fusion.


Asunto(s)
Integrina alfa3/fisiología , Mioblastos Esqueléticos/citología , Células Satélite del Músculo Esquelético/citología , Proteínas ADAM/metabolismo , Proteína ADAM12 , Actinas/metabolismo , Animales , Anticuerpos/farmacología , Adhesión Celular , Fusión Celular , Células Cultivadas , Integrina alfa3/análisis , Integrina alfa3/genética , Integrina beta1/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Wistar , Células Satélite del Músculo Esquelético/metabolismo
13.
Am J Physiol Lung Cell Mol Physiol ; 290(2): L343-50, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16169900

RESUMEN

Mechanical forces regulate lung maturation in the fetus by promoting type II epithelial differentiation. However, the cell surface receptors that transduce these mechanical cues into cellular responses remain largely unknown. When distal lung type II epithelial cells isolated from embryonic day 19 rat fetuses were cultured on flexible plates coated with laminin, fibronectin, vitronectin, collagen, or elastin and exposed to a level of mechanical strain (5%) similar to that observed in utero, transmembrane signaling responses were induced under all conditions, as measured by ERK activation. However, mechanical stress maximally increased expression of the type II cell differentiation marker surfactant protein C when cells were cultured on laminin substrates. Strain-induced alveolar epithelial differentiation was inhibited by interfering with cell binding to laminin using soluble laminin peptides (IKVIV or YIGSR) or blocking antibodies against integrin beta1, alpha3, or alpha6. Additional studies were carried out with substrates coated directly with different nonactivating anti-integrin antibodies. Blocking integrin beta1 and alpha6 binding sites inhibited both cell adhesion and differentiation, whereas inhibition of alpha3 prevented differentiation without altering cell attachment. These data demonstrate that various integrins contribute to mechanical control of type II lung epithelial cell differentiation on laminin substrates. However, they may act via distinct mechanisms, including some that are independent of their cell anchoring role.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/citología , Integrina alfa3/fisiología , Integrina alfa6/fisiología , Integrina beta1/fisiología , Pulmón/citología , Animales , Adhesión Celular/efectos de los fármacos , Activación Enzimática , Matriz Extracelular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Feto/citología , Laminina/fisiología , Péptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Estrés Mecánico
14.
Clin Exp Metastasis ; 22(5): 391-402, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16283482

RESUMEN

Integrins play an important role in cellular matrix interactions requisite for cancer cell adhesion, growth, migration and invasion. In this study, we have investigated the expression of integrin subunits alpha3, alpha6, alphav and beta1 in normal ovaries, benign ovarian tumors and ovarian carcinomas of different pathological grades. The expression of these integrins in ovarian cancer cell lines was also investigated, and their role in sustaining proliferation, adhesion, migration and invasion in cohort with the activation of signaling pathways in response to extracellular matrices (ECM) was evaluated. We demonstrate a differential expression pattern of alpha3, alpha6, alphav and beta1 integrin subunits in ovarian carcinomas compared to normal ovaries and benign ovarian tumors. Ovarian cancer cell lines (Hey, Ovcar3 and Peo.36) demonstrated significantly high expression of alpha3, alpha6, alphav and beta1 integrin subunits. A significant increase in proliferation and adhesion (P<0.05) in response to collagen 1 (Coll) and laminin (LM), ligands for integrin receptor alpha3beta1 and alpha6beta1 was observed in ovarian cancer cell lines. On the other hand, fibronectin (FN), a receptor for alphavbeta1 integrin, increased proliferation in all ovarian cancer cell lines studied but only enhanced adhesion in Hey cell line (P<0.05). Neutralizing antibodies against alpha3, alpha6, alphav and beta1 integrin subunits inhibited ECM-induced proliferation, but increased adhesion to ECM was inhibited by beta1 integrin subunit antibody. No suppression of Coll, LM and FN-induced (Hey cells only) adhesion was observed in the presence of alpha3 or alphav subunit antibodies but LM-induced adhesion was inhibited by blocking alpha6 subunit functions. LM, FN and Coll enhanced chemotactic migration in Hey cells, but direct invasion across ECM was observed only in the presence of LM and Coll. Blocking antibodies against alpha3, alpha6 and beta1 integrin subunits inhibited both chemotactic migration and invasion of Hey cells in response to respective ECM. Adhesion of ovarian cancer cells to FN, Coll and LM activated Ras, Erk and Akt pathways. Neutralizing alphav and beta1 functions did not inhibit FN-induced activation of Ras and Erk pathways but inhibited the Akt pathway. On the other hand, antibodies against alpha6 and beta1 subunits, but not alpha3 subunit, inhibited LM-induced activation of Ras but did not inhibit the downstream Akt pathway. Neutralizing beta1 subunit function however, inhibited LM-induced Erk activation. Coll-induced activation of Ras, Erk and Akt pathways was inhibited by alpha3 and beta1 integrin subunit antibodies. These results indicate that alpha3beta1, alphavbeta1 and alpha6beta1 integrin mediate proliferation, adhesion, migration and invasion of ovarian cancer cells in response to ECM and targeting these integrins to modulate integrin-ECM interactions in tumor cells may be a promising tool to reduce the dissemination of ovarian carcinoma in vivo.


Asunto(s)
Carcinoma/fisiopatología , Integrina alfa3/fisiología , Integrina alfa6/fisiología , Integrina alfaV/fisiología , Integrina beta1/fisiología , Neoplasias Ováricas/fisiopatología , Adhesión Celular , Movimiento Celular , Proliferación Celular , Colágeno/fisiología , Matriz Extracelular/fisiología , Femenino , Fibronectinas/fisiología , Humanos , Integrina alfa3/biosíntesis , Integrina alfa6/biosíntesis , Integrina alfaV/biosíntesis , Integrina beta1/biosíntesis , Laminina/fisiología , Ligandos , Células Tumorales Cultivadas
15.
Diabetes ; 54(7): 2080-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15983209

RESUMEN

The integrin receptors play a major role in tissue morphogenesis and homeostasis by regulating cell interactions with extracellular matrix proteins. We have examined the expression pattern of integrin subunits in the human fetal pancreas (8-20 weeks fetal age) and the relevance of beta1 integrin function for insulin gene expression and islet cell survival. Its subunits alpha3, alpha5, and alpha6 beta1 integrins are expressed in ductal cells at 8 weeks, before glucagon- and insulin-immunoreactive cells bud off; their levels gradually increase in both ductal cells and islet clusters up to 20 weeks. Colocalization of alpha3, alpha5 and alpha6 beta1 integrins with endocrine cell markers was frequently observed in 8- to 20-week fetal pancreatic cells. When the beta1 integrin receptor was functionally blocked in cultured islet-epithelial clusters with a beta1 immunoneutralizing antibody or following transient beta1 integrin small interfering RNA treatment, there was inhibition of cell adhesion to extracellular matrices, decreased expression of insulin, and increased cell apoptosis. These data offer evidence for dynamic and cell-specific changes in integrin expression during human pancreatic islet neogenesis. They also provide an initial insight into a molecular basis for cell-matrix interactions during islet development and suggest that beta1 integrin plays a vital role in regulating islet cell adhesion, gene expression, and survival.


Asunto(s)
Integrina beta1/fisiología , Páncreas/embriología , Apoptosis , Adhesión Celular , Células Epiteliales/inmunología , Células Epiteliales/fisiología , Femenino , Desarrollo Fetal , Regulación del Desarrollo de la Expresión Génica/inmunología , Edad Gestacional , Humanos , Insulina/biosíntesis , Integrina alfa3/genética , Integrina alfa3/fisiología , Integrina alfa5/genética , Integrina alfa5/fisiología , Integrina alfa6/genética , Integrina alfa6/fisiología , Integrina beta1/genética , Páncreas/efectos de los fármacos , Páncreas/inmunología , Embarazo , ARN Interferente Pequeño
16.
Cancer Res ; 64(17): 6065-70, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15342388

RESUMEN

The interaction between gastric carcinoma cells and the peritoneal lining is a key step in peritoneal dissemination. In this study, we examined the roles of the beta1 family of integrin receptors in the adhesion of such cells to the peritoneum. The adhesion of several gastric carcinoma cell lines to peritonea excised from mice was inhibited most by an anti-alpha3 integrin antibody and to a lesser extent by an anti-alpha2 integrin antibody. In the peritoneal implantation of NUGC-4 human gastric carcinoma cells in athymic mice, treatment of the cells with anti-alpha2 or anti-alpha3 integrin antibody reduced the number of disseminated nodules; suppression by the anti-alpha3 integrin antibody was stronger than that by the anti-alpha2 integrin antibody. The cDNAs to human alpha2 and alpha3 integrins were introduced into K562 leukemic cells, which were positive for the integrin beta1 subunit but negative for the alpha2 or alpha3 subunit. The alpha3 integrin-transfected cells adhered to excised peritoneum and to a monolayer of peritoneal mesothelial cells more firmly than did the alpha2 integrin-transfected cells or the mock transfectant. Reverse transcription-PCR was used to analyze the expression of laminin-5 and laminin-10/11, which have been reported to serve as high-affinity ligands for alpha3beta1 integrin. mRNA for these laminin isoforms was found in mesothelial cells from the diaphragm and parietal peritoneum. These results strongly suggest that alpha3beta1 integrin plays an essential role in mediating the initial attachment of cancer cells to the peritoneum, leading to the formation of peritoneal metastasis.


Asunto(s)
Integrina alfa3beta1/fisiología , Peritoneo/patología , Neoplasias Gástricas/patología , Anticuerpos/farmacología , Adhesión Celular/fisiología , Línea Celular Tumoral , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Integrina alfa2/biosíntesis , Integrina alfa2/fisiología , Integrina alfa3/biosíntesis , Integrina alfa3/fisiología , Integrina alfa3beta1/antagonistas & inhibidores , Integrina alfa3beta1/inmunología , Integrina alfa3beta1/metabolismo , Ligandos
17.
Biochem J ; 379(Pt 1): 141-50, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-14720123

RESUMEN

Signalling by integrin-mediated cell anchorage to extracellular matrix proteins is co-operative with other receptor-mediated signalling pathways to regulate cell adhesion, spreading, proliferation, survival, migration, differentiation and gene expression. It was observed that an anchorage-independent gastric carcinoma cell line (SNU16) became adherent on TGF-beta1 (transforming growth factor beta1) treatment. To understand how a signal cross-talk between integrin and TGF-beta1 pathways forms the basis for TGF-beta1 effects, cell adhesion and signalling activities were studied using an adherent subline (SNU16Ad, an adherent variant cell line derived from SNU16) derived from the SNU16 cells. SNU16 and SNU16Ad cells, but not integrin alpha5-expressing SNU16 cells, showed an increase in adhesion on extracellular matrix proteins after TGF-beta1 treatment. This increase was shown to be mediated by an integrin alpha3 subunit, which was up-regulated in adherent SNU16Ad cells and in TGF-beta1-treated SNU16 cells, compared with the parental SNU16 cells. After TGF-beta1 treatment of SNU16Ad cells on fibronectin, Tyr-416 phosphorylation of c-Src was increased, but Ras-GTP loading and ERK1/ERK2 (extracellular-signal-regulated kinases 1 and 2) activity were decreased, which showed a dependence on c-Src family kinase activity. Studies on adhesion and signalling activities using pharmacological inhibitors or by transient-transfection approaches showed that inhibition of ERK1/ERK2 activity increased TGF-beta1-mediated cell adhesion slightly, but not the basal cell adhesion significantly, and that c-Src family kinase activity and decrease in Ras/ERKs cascade activity were required for the TGF-beta1 effects. Altogether, the present study indicates that TGF-beta1 treatment causes anchorage-independent gastric carcinoma cells to adhere by an increase in integrin alpha3 level and a c-Src family kinase activity-dependent decrease in Ras/ERKs cascade activity.


Asunto(s)
Carcinoma/patología , Adhesión Celular/fisiología , Integrina alfa3/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Neoplasias/fisiología , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta/farmacología , Familia-src Quinasas/fisiología , Animales , Carcinoma/metabolismo , Adhesión Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero , Metilación de ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Matriz Extracelular , Fibronectinas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Guanosina Trifosfato/fisiología , Humanos , Integrina alfa3/biosíntesis , Integrina alfa3/genética , Integrinas/análisis , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Subunidades de Proteína , Ratas , Neoplasias Gástricas/metabolismo , Factor de Crecimiento Transformador beta1 , Proteínas Activadoras de ras GTPasa/fisiología , Familia-src Quinasas/antagonistas & inhibidores
18.
Dev Biol ; 254(2): 226-37, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12591243

RESUMEN

Antibody inhibition and alpha6beta1 ligand binding experiments indicate that the egg integrin alpha6beta1 functions as a receptor for sperm during gamete fusion; yet, eggs null for the alpha6 integrin exhibit normal fertilization. Alternative integrins may be involved in sperm-egg binding and fusion and could compensate for the absence of alpha6beta1. Various beta1 integrins and alphav integrins are present on mouse eggs. Some of these integrins are also reported to be receptors for ADAMs, which are expressed on sperm. Using alpha3 integrin null eggs, we found that the alpha3beta1 integrin was not essential for sperm-egg binding and fusion. Oocyte-specific, beta1 integrin conditional knockout mice allowed us to obtain mature eggs lacking all beta1 integrins. We found that the beta1 integrin null eggs were fully functional in fertilization both in vivo and in vitro. Furthermore, neither anti-mouse beta3 integrin function-blocking monoclonal antibody (mAb) nor alphav integrin function-blocking mAb inhibited sperm binding to or fusion with beta1 integrin null eggs. Thus, function of beta3 or alphav integrins does not seem to be involved in compensating for the absence of beta1 integrins. These results indicate that none of the integrins known to be present on mouse eggs or to be ADAM receptors are essential for sperm-egg binding/fusion, and thus, egg integrins may not play the role in gamete fusion previously attributed to them.


Asunto(s)
Integrinas/fisiología , Fusión de Membrana , Glicoproteínas de Membrana/fisiología , Metaloendopeptidasas/fisiología , Óvulo/fisiología , Interacciones Espermatozoide-Óvulo , Proteínas ADAM , Animales , Antígenos CD/fisiología , Femenino , Fertilinas , Integrina alfa3/fisiología , Integrina alfaV/fisiología , Integrina beta1/fisiología , Integrina beta3/fisiología , Masculino , Ratones , Óvulo/química , Subunidades de Proteína , Tetraspanina 29
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...