Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Biol Chem ; 285(48): 37895-908, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-20876529

RESUMEN

Reversible ubiquitination orchestrated by the opposition of ubiquitin ligases and deubiquitinating enzymes mediates endocytic trafficking of cell surface receptors for lysosomal degradation. Ubiquitin-specific protease 8 (USP8) has previously been implicated in endocytosis of several receptors by virtue of their deubiquitination. The present study explores an indirect role for USP8 in cargo trafficking through its regulation of the chemokine receptor 4 (CXCR4). Contrary to the effects of USP8 loss on enhanced green fluorescent protein, we find that USP8 depletion stabilizes CXCR4 on the cell surface and attenuates receptor degradation without affecting its ubiquitination status. In the presence of ligand, diminished CXCR4 turnover is accompanied by receptor accumulation on enlarged early endosomes and leads to enhancement of phospho-ERK signaling. Perturbation in CXCR4 trafficking, resulting from USP8 inactivation, occurs at the ESCRT-0 checkpoint, and catalytic mutation of USP8 specifically targeted to the ESCRT-0 complex impairs the spatial and temporal organization of the sorting endosome. USP8 functionally opposes the ubiquitin ligase AIP4 with respect to ESCRT-0 ubiquitination, thereby promoting trafficking of CXCR4. Collectively, our findings demonstrate a functional cooperation between USP8, AIP4, and the ESCRT-0 machinery at the early sorting phase of CXCR4 and underscore the versatility of USP8 in shaping trafficking events at the early-to-late endosome transition.


Asunto(s)
Endopeptidasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endosomas/metabolismo , Receptores CXCR4/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Endopeptidasas/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Endosomas/genética , Células HEK293 , Células HeLa , Humanos , Ratones , Transporte de Proteínas , Receptores CXCR4/genética , Ubiquitina Tiolesterasa/genética , Ubiquitinación
2.
Am J Physiol Gastrointest Liver Physiol ; 301(2): G368-76, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21617115

RESUMEN

Upon ligand binding, epidermal growth factor (EGF) receptor (R) autophosphorylates on COOH-terminal tyrosines, generating docking sites for signaling partners that stimulate proliferation, restitution, and chemotaxis. Specificity for individual EGFR tyrosines in cellular responses has been hypothesized but not well documented. Here we tested the requirement for particular tyrosines, and associated downstream pathways, in mouse colon epithelial cell chemotactic migration. We compared these requirements to those for the phenotypically distinct restitution (wound healing) migration. Wild-type, Y992/1173F, Y1045F, Y1068F, and Y1086F EGFR constructs were expressed in EGFR(-/-) cells; EGF-induced chemotaxis or restitution were determined by Boyden chamber or modified scratch wound assay, respectively. Pharmacological inhibitors of p38, phospholipase C (PLC), Src, MEK, JNK/SAPK, phosphatidylinositol 3-kinase (PI 3-kinase), and protein kinase C (PKC) were used to block EGF-stimulated signaling. Pathway activation was determined by immunoblot analysis. Unlike wild-type EGFR, Y992/1173F and Y1086F EGFR did not stimulate colon epithelial cell chemotaxis toward EGF; Y1045F and Y1068F EGFR partially stimulated chemotaxis. Only wild-type EGFR promoted colonocyte restitution. Inhibition of p38, PLC, and Src, or Grb2 knockdown, blocked chemotaxis; JNK, PI 3-kinase, and PKC inhibitors or c-Cbl knockdown blocked restitution but not chemotaxis. All four EGFR mutants stimulated downstream signaling in response to EGF, but Y992/1173F EGFR was partially defective in PLCγ activation whereas both Y1068F and Y1086F EGFR failed to activate Src. We conclude that specific EGFR tyrosines play key roles in determining cellular responses to ligand. Chemotaxis and restitution, which have different migration phenotypes and physiological consequences, have overlapping but not identical EGFR signaling requirements.


Asunto(s)
Quimiotaxis/fisiología , Factor de Crecimiento Epidérmico/fisiología , Células Epiteliales/fisiología , Receptores ErbB/metabolismo , Transducción de Señal/fisiología , Tirosina/fisiología , Animales , Sitios de Unión/fisiología , Línea Celular , Quimiotaxis/efectos de los fármacos , Colon/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/metabolismo , Receptores ErbB/deficiencia , Receptores ErbB/genética , Receptores ErbB/fisiología , Proteína Adaptadora GRB2/deficiencia , Proteína Adaptadora GRB2/fisiología , Ratones , Mutación , Fosfolipasa C gamma/fisiología , Fosforilación , Proteínas Proto-Oncogénicas c-cbl/deficiencia , Proteínas Proto-Oncogénicas c-cbl/fisiología , ARN Interferente Pequeño , Transfección , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Dominios Homologos src/fisiología , Familia-src Quinasas
3.
Proc Natl Acad Sci U S A ; 105(33): 11772-7, 2008 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-18701712

RESUMEN

TNF is a pleiotropic cytokine that activates both anti- and proapoptotic signaling pathways, with cell fate determined by the balance between these two pathways. Activation of ErbB family members, including EGF receptor (EGFR/ErbB1), promotes cell survival and regulates several signals that overlap with those stimulated by TNF. This study was undertaken to determine the effects of TNF on EGFR and ErbB2 activation and intestinal epithelial cell survival. Mice, young adult mouse colon epithelial cells, and EGFR knockout mouse colon epithelial cells were treated with TNF. Activation of EGFR, ErbB2, Akt, Src, and apoptosis were determined in vivo and in vitro. TNF stimulated EGFR phosphorylation in young adult mouse colon epithelial cells, and loss of EGFR expression or inhibition of kinase activity increased TNF-induced apoptosis, which was prevented in WT but not by kinase-inactive EGFR expression. Similarly, TNF injection stimulated apoptosis in EGFR-kinase-defective mice (EGFR(wa2)) compared with WT mice. TNF also activated ErbB2, and loss of ErbB2 expression increased TNF-induced apoptosis. Furthermore, Src-kinase activity and the expression of both EGFR and ErbB2 were required for TNF-induced cell survival. Akt was shown to be a downstream target of TNF-activated EGFR and ErbB2. These findings demonstrate that EGFR and ErbB2 are critical mediators of TNF-regulated antiapoptotic signals in intestinal epithelial cells. Given evidence for TNF signaling in the development of colitis-associated carcinoma, this observation has significant implications for understanding the role of EGFR in maintaining intestinal epithelial cell homeostasis during cytokine-mediated inflammatory responses.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Epiteliales/metabolismo , Receptores ErbB/metabolismo , Mucosa Intestinal/metabolismo , Receptor ErbB-2/metabolismo , Activación Transcripcional/genética , Factor de Necrosis Tumoral alfa/farmacología , Animales , Activación Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Receptores ErbB/deficiencia , Receptores ErbB/genética , Regulación de la Expresión Génica/efectos de los fármacos , Intestinos/citología , Intestinos/efectos de los fármacos , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/genética , Activación Transcripcional/efectos de los fármacos , Familia-src Quinasas/metabolismo
4.
Mol Cell Biol ; 27(9): 3313-26, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17339338

RESUMEN

Integrins control many cell functions, including generation of reactive oxygen species (ROS) and regulation of collagen synthesis. Mesangial cells, found in the glomerulus of the kidney, are able to produce large amounts of ROS via the NADPH oxidase. We previously demonstrated that integrin alpha1-null mice develop worse fibrosis than wild-type mice following glomerular injury and this is due, in part, to excessive ROS production by alpha1-null mesangial cells. In the present studies, we describe the mechanism whereby integrin alpha1-null mesangial cells produce excessive ROS. Integrin alpha1-null mesangial cells have constitutively increased basal levels of activated Rac1, which result in its increased translocation to the cell membrane, excessive ROS production, and consequent collagen IV deposition. Basal Rac1 activation is a direct consequence of ligand-independent increased epidermal growth factor receptor (EGFR) phosphorylation in alpha1-null mesangial cells. Thus, our study demonstrates that integrin alpha1beta1-EGFR cross talk is a key step in negatively regulating Rac1 activation, ROS production, and excessive collagen synthesis, which is a hallmark of diseases characterized by irreversible fibrosis.


Asunto(s)
Receptores ErbB/metabolismo , Integrina alfa1beta1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Membrana Celular/metabolismo , Forma de la Célula , Células Cultivadas , Colágeno/biosíntesis , Regulación hacia Abajo , Activación Enzimática , Humanos , Integrina alfa1beta1/deficiencia , Integrina alfa1beta1/genética , Ligandos , Células Mesangiales/citología , Células Mesangiales/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-vav/metabolismo , Receptores de Colágeno/metabolismo , Proteínas de Unión al GTP rac/genética
5.
Am J Physiol Gastrointest Liver Physiol ; 294(1): G276-85, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17991704

RESUMEN

Regulated intestinal epithelial cell migration plays a key role in wound healing and maintenance of a healthy gastrointestinal tract. Epidermal growth factor (EGF) stimulates cell migration and wound closure in intestinal epithelial cells through incompletely understood mechanisms. In this study we investigated the role of the small GTPase Rac in EGF-induced cell migration using an in vitro wound-healing assay. In mouse colonic epithelial (MCE) cell lines, EGF-stimulated wound closure was accompanied by a doubling of the number of cells containing lamellipodial extensions at the wound margin, increased Rac membrane translocation in cells at the wound margin, and rapid Rac activation. Either Rac1 small interfering (si)RNA or a Rac1 inhibitor completely blocked EGF-stimulated wound closure. Whereas EGF failed to activate Rac in colon cells from EGF receptor (EGFR) knockout mice, stable expression of wild-type EGFR restored EGF-stimulated Rac activation and migration. Pharmacological inhibition of either phosphatidylinositol 3-kinase (PI3K) or Src family kinases reduced EGF-stimulated Rac activation. Cotreatment of cells with both inhibitors completely blocked EGF-stimulated Rac activation and localization to the leading edge of cells and lamellipodial extension. Our results present a novel mechanism by which the PI3K and Src signaling cascades cooperate to activate Rac and promote intestinal epithelial cell migration downstream of EGFR.


Asunto(s)
Movimiento Celular , Colon/metabolismo , Células Epiteliales/metabolismo , Receptores ErbB/metabolismo , Neuropéptidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Cicatrización de Heridas , Proteínas de Unión al GTP rac/metabolismo , Familia-src Quinasas/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Colon/efectos de los fármacos , Colon/enzimología , Activación Enzimática , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/enzimología , Receptores ErbB/deficiencia , Receptores ErbB/genética , Ratones , Ratones Noqueados , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Seudópodos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Transfección , Cicatrización de Heridas/efectos de los fármacos , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1 , Familia-src Quinasas/antagonistas & inhibidores
6.
EMBO J ; 25(24): 5683-92, 2006 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-17139251

RESUMEN

Internalization and proteolytic degradation of epidermal growth factor (EGF) receptor (R) following ligand binding is an important mechanism for regulating EGF-stimulated signals. Using pharmacological and RNA interference inhibition of p38 mitogen-activated protein kinase, we show that p38 is required for efficient EGF-induced EGFR destruction but not internalization. In the absence of p38 activity, EGF fails to stimulate the ubiquitin ligase Cbl or ubiquitinylation of EGFR, and internalized EGFR accumulates in intracellular vesicles containing caveolin-1. These effects are accompanied by loss of EGFR phosphorylation on Y1045, a phosphorylation site required for Cbl activation. Furthermore, similar to cells treated with p38 inhibitors, intestinal epithelial cells expressing Y1045F EGFR mutants show increased proliferation but not migration in response to EGF, thus uncoupling these biological responses. Together these data position p38 as a modulator of ligand-stimulated EGFR processing and demonstrate that this processing has a profound impact on the cellular outcome of EGFR signaling.


Asunto(s)
Movimiento Celular/fisiología , Regulación hacia Abajo , Receptores ErbB/metabolismo , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Animales , Células COS , Movimiento Celular/efectos de los fármacos , Chlorocebus aethiops , Colon/citología , Colon/efectos de los fármacos , Perros , Regulación hacia Abajo/efectos de los fármacos , Endocitosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Receptores ErbB/deficiencia , Humanos , Ratones , Proteína Quinasa 11 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 11 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Ubiquitina/metabolismo
7.
J Biol Chem ; 277(32): 29028-35, 2002 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-12006559

RESUMEN

Nephrocystin is the protein product of the gene mutated in juvenile nephronophthisis, an autosomal recessive cystic kidney disease afflicting children and young adults. Because the normal cellular function of nephrocystin is largely unknown, the molecular defects underlying disease pathogenesis remain obscure. Analysis of nephrocystin amino acid sequences from human and other species revealed three distinct conserved domains including Src homology 3 and coil-coil domains in the N-terminal region, as well as a large highly conserved C-terminal region bearing no obvious homology to other proteins and hence referred to as the "nephrocystin homology domain" (NHD). The objective of this study was to gain insight into nephrocystin function by defining functional properties of the conserved domains. We analyzed a series of nephrocystin deletion mutants expressed in Madin-Darby canine kidney and COS-7 cells. This analysis revealed previously unrecognized functional attributes of the NHD, including abilities to promote both self-association and epithelial cell-cell junctional targeting. We further observed that Madin-Darby canine kidney cell lines stably expressing a nephrocystin mutant with a deletion of the Src homology 3 domain have reduced ability to establish tight junctions as measured by transepithelial electrical resistance. Finally, from a two-hybrid screen and coimmunoprecipitation studies we identified members of the filamin family of actin-binding proteins as having the capacity to interact with the NHD. These findings support a functional role for nephrocystin as a docking protein involved in organizing a protein complex to regulate the actin cytoskeleton at sites of epithelial cell-cell adhesion and further suggest that these properties are important for establishing epithelial cell polarity.


Asunto(s)
Proteínas Contráctiles/química , Células Epiteliales/metabolismo , Proteínas de Microfilamentos/química , Proteínas/química , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células COS , Adhesión Celular , Polaridad Celular , Proteínas del Citoesqueleto , Perros , Epítopos , Filaminas , Proteínas de la Membrana , Microscopía Fluorescente , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Tiempo , Técnicas del Sistema de Dos Híbridos , Dominios Homologos src
8.
J Biol Chem ; 279(37): 38331-7, 2004 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-15247284

RESUMEN

Crk-associated substrate (p130(CAS) or CAS) is a major integrin-associated Src substrate that undergoes tyrosine phosphorylation at multiple YXXP motifs in its substrate domain (SD) to create docking sites for SH2-containing signaling effectors. Notably, recruitment of Crk adaptor proteins to the CAS SD sites is implicated in promoting cell migration. However, it is unclear which or how many of the 15 CAS SD YXXP tyrosines are critically involved. To gain a better understanding of CAS SD function, we assessed the signaling capacity of individual YXXP motifs. Using site-directed mutagenesis combined with tryptic phosphopeptide mapping, we determined that the ten tyrosines in YXXP motifs 6-15 are the major sites of CAS SD phosphorylation by Src. Phosphopeptide binding assays showed that all of these sites are capable of binding the Crk SH2 domain. To evaluate the requirement for CAS YXXP sites in stimulating cell migration, a series of phenylalanine substitution variants were expressed in CAS -/- mouse embryo fibroblasts. CAS expression enhanced the rate of cell migration into a monolayer wound in a manner dependent on the major sites of Src phosphorylation. Effective wound healing was achieved by CAS variants containing as few as four of the major sites, indicating sufficiency of partial SD signaling function in this cell migration response.


Asunto(s)
Proteínas/química , Tirosina/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células COS , Movimiento Celular , Proteína Sustrato Asociada a CrK , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Mapeo Peptídico , Péptidos/química , Fenilalanina/química , Fosforilación , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas/metabolismo , Proteína p130 Similar a la del Retinoblastoma , Transducción de Señal , Tripsina/química , Cicatrización de Heridas , Dominios Homologos src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA