Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Mol Gastroenterol Hepatol ; 17(4): 639-656, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38199279

RESUMEN

BACKGROUND & AIMS: Chronic inflammatory illnesses are debilitating and recurrent conditions associated with significant comorbidities, including an increased risk of developing cancer. Extensive tissue remodeling is a hallmark of such illnesses, and is both a consequence and a mediator of disease progression. Despite previous characterization of epithelial and stromal remodeling during inflammatory bowel disease, a complete understanding of its impact on disease progression is lacking. METHODS: A comprehensive proteomic pipeline using data-independent acquisition was applied to decellularized colon samples from the Muc2 knockout (Muc2KO) mouse model of colitis for an in-depth characterization of extracellular matrix remodeling. Unique proteomic profiles of the matrisomal landscape were extracted from prepathologic and overt colitis. Integration of proteomics and transcriptomics data sets extracted from the same murine model produced network maps describing the orchestrating role of matrisomal proteins in tissue remodeling during the progression of colitis. RESULTS: The in-depth proteomic workflow used here allowed the addition of 34 proteins to the known colon matrisomal signature. Protein signatures of prepathologic and pathologic colitic states were extracted, differentiating the 2 states by expression of small leucine-rich proteoglycans. We outlined the role of this class and other matrisomal proteins in tissue remodeling during colitis, as well as the potential for coordinated regulation of cell types by matrisomal ligands. CONCLUSIONS: Our work highlights a central role for matrisomal proteins in tissue remodeling during colitis and defines orchestrating nodes that can be exploited in the selection of therapeutic targets.


Asunto(s)
Colitis , Proteómica , Ratones , Animales , Matriz Extracelular/metabolismo , Colitis/patología , Enfermedad Crónica , Progresión de la Enfermedad
2.
Nat Genet ; 54(12): 1827-1838, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36175792

RESUMEN

We identify the sodium leak channel non-selective protein (NALCN) as a key regulator of cancer metastasis and nonmalignant cell dissemination. Among 10,022 human cancers, NALCN loss-of-function mutations were enriched in gastric and colorectal cancers. Deletion of Nalcn from gastric, intestinal or pancreatic adenocarcinomas in mice did not alter tumor incidence, but markedly increased the number of circulating tumor cells (CTCs) and metastases. Treatment of these mice with gadolinium-a NALCN channel blocker-similarly increased CTCs and metastases. Deletion of Nalcn from mice that lacked oncogenic mutations and never developed cancer caused shedding of epithelial cells into the blood at levels equivalent to those seen in tumor-bearing animals. These cells trafficked to distant organs to form normal structures including lung epithelium, and kidney glomeruli and tubules. Thus, NALCN regulates cell shedding from solid tissues independent of cancer, divorcing this process from tumorigenesis and unmasking a potential new target for antimetastatic therapies.


Asunto(s)
Neoplasias , Humanos , Ratones , Animales , Canales Iónicos/genética , Proteínas de la Membrana/genética
3.
Dis Model Mech ; 14(1)2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33093165

RESUMEN

Somatic models of tissue pathology commonly use induction of gene-specific mutations in mice mediated by spatiotemporal regulation of Cre recombinase. Subsequent investigation of the onset and development of disease can be limited by the inability to track changing cellular behaviours over time. Here, a lineage-tracing approach based on ligand-dependent activation of Dre recombinase that can be employed independently of Cre is described. The clonal biology of the intestinal epithelium following Cre-mediated stabilisation of ß-catenin reveals that, within tumours, many new clones rapidly become extinct. Surviving clones show accelerated population of tumour glands compared to normal intestinal crypts but in a non-uniform manner, indicating that intra-tumour glands follow heterogeneous dynamics. In tumour-adjacent epithelia, clone sizes are smaller than in the background epithelia, as a whole. This suggests a zone of ∼seven crypt diameters within which clone expansion is inhibited by tumours and that may facilitate their growth.


Asunto(s)
Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Mutación , Animales , Anticuerpos Monoclonales/química , Linaje de la Célula , Colon/metabolismo , Células Epiteliales/metabolismo , Epitelio/metabolismo , Proteínas de Escherichia coli/metabolismo , Femenino , Integrasas/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Intestinos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/metabolismo , Probabilidad , Recombinasas/metabolismo , Células Madre/citología , beta Catenina/metabolismo
4.
Genetics ; 212(3): 655-665, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31126976

RESUMEN

Microsatellite sequences have an enhanced susceptibility to mutation, and can act as sentinels indicating elevated mutation rates and increased risk of cancer. The probability of mutant fixation within the intestinal epithelium is dictated by a combination of stem cell dynamics and mutation rate. Here, we exploit this relationship to infer microsatellite mutation rates. First a sensitive, multiplexed, and quantitative method for detecting somatic changes in microsatellite length was developed that allowed the parallel detection of mutant [CA]n sequences from hundreds of low-input tissue samples at up to 14 loci. The method was applied to colonic crypts in Mus musculus, and enabled detection of mutant subclones down to 20% of the cellularity of the crypt (∼50 of 250 cells). By quantifying age-related increases in clone frequencies for multiple loci, microsatellite mutation rates in wild-type and Msh2-deficient epithelium were established. An average 388-fold increase in mutation per mitosis rate was observed in Msh2-deficient epithelium (2.4 × 10-2) compared to wild-type epithelium (6.2 × 10-5).


Asunto(s)
Células Madre Adultas/metabolismo , Mucosa Intestinal/citología , Repeticiones de Microsatélite , Proteína 2 Homóloga a MutS/genética , Tasa de Mutación , Células Madre Adultas/citología , Animales , Femenino , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitosis , Proteína 2 Homóloga a MutS/deficiencia
5.
Nat Cell Biol ; 20(10): 1193-1202, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30177776

RESUMEN

Solid malignancies have been speculated to depend on cancer stem cells (CSCs) for expansion and relapse after therapy. Here we report on quantitative analyses of lineage tracing data from primary colon cancer xenograft tissue to assess CSC functionality in a human solid malignancy. The temporally obtained clone size distribution data support a model in which stem cell function in established cancers is not intrinsically, but is entirely spatiotemporally orchestrated. Functional stem cells that drive tumour expansion predominantly reside at the tumour edge, close to cancer-associated fibroblasts. Hence, stem cell properties change in time depending on the cell location. Furthermore, although chemotherapy enriches for cells with a CSC phenotype, in this context functional stem cell properties are also fully defined by the microenvironment. To conclude, we identified osteopontin as a key cancer-associated fibroblast-produced factor that drives in situ clonogenicity in colon cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Proliferación Celular/genética , Células Cultivadas , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Oxaliplatino/administración & dosificación , Tamoxifeno/administración & dosificación , Microambiente Tumoral/genética
6.
J Exp Med ; 215(3): 725-727, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29440363

RESUMEN

In this issue of JEM, Balbinot et al. (https://doi.org/10.1084/jem.20170934) describe an original mechanism where Cdx2 inactivation regulates intestinal metaplastic to neoplastic transition in a paracrine fashion. Surprisingly, the target cells are neighboring "normal" Cdx2-positive cells.


Asunto(s)
Genes Homeobox , Proteínas de Homeodominio/genética , Animales , Factor de Transcripción CDX2 , Carcinogénesis , Ratones , Ratones Transgénicos
7.
Breast Cancer Res ; 18(1): 127, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27964754

RESUMEN

BACKGROUND: High-resolution 3D imaging of intact tissue facilitates cellular and subcellular analyses of complex structures within their native environment. However, difficulties associated with immunolabelling and imaging fluorescent proteins deep within whole organs have restricted their applications to thin sections or processed tissue preparations, precluding comprehensive and rapid 3D visualisation. Several tissue clearing methods have been established to circumvent issues associated with depth of imaging in opaque specimens. The application of these techniques to study the elaborate architecture of the mouse mammary gland has yet to be investigated. METHODS: Multiple tissue clearing methods were applied to intact virgin and lactating mammary glands, namely 3D imaging of solvent-cleared organs, see deep brain (seeDB), clear unobstructed brain imaging cocktails (CUBIC) and passive clarity technique. Using confocal, two-photon and light sheet microscopy, their compatibility with whole-mount immunofluorescent labelling and 3D imaging of mammary tissue was examined. In addition, their suitability for the analysis of mouse mammary tumours was also assessed. RESULTS: Varying degrees of optical transparency, tissue preservation and fluorescent signal conservation were observed between the different clearing methods. SeeDB and CUBIC protocols were considered superior for volumetric fluorescence imaging and whole-mount histochemical staining, respectively. Techniques were compatible with 3D imaging on a variety of platforms, enabling visualisation of mammary ductal and lobulo-alveolar structures at vastly improved depths in cleared tissue. CONCLUSIONS: The utility of whole-organ tissue clearing protocols was assessed in the mouse mammary gland. Most methods utilised affordable and widely available reagents, and were compatible with standard confocal microscopy. These techniques enable high-resolution, 3D imaging and phenotyping of mammary cells and tumours in situ, and will significantly enhance our understanding of both normal and pathological mammary gland development.


Asunto(s)
Imagenología Tridimensional , Glándulas Mamarias Animales/diagnóstico por imagen , Neoplasias Mamarias Animales/diagnóstico por imagen , Neoplasias Mamarias Animales/patología , Animales , Femenino , Técnica del Anticuerpo Fluorescente , Imagenología Tridimensional/métodos , Ratones , Microscopía Confocal , Imagen Óptica/métodos
8.
Oncotarget ; 6(36): 38469-86, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26540348

RESUMEN

The actin and microtubule cytoskeletons are critically important for cancer cell proliferation, and drugs that target microtubules are widely-used cancer therapies. However, their utility is compromised by toxicities due to dose and exposure. To overcome these issues, we characterized how inhibition of the actin and microtubule cytoskeleton regulatory LIM kinases could be used in drug combinations to increase efficacy. A previously-described LIMK inhibitor (LIMKi) induced dose-dependent microtubule alterations that resulted in significant mitotic defects, and increased the cytotoxic potency of microtubule polymerization inhibitors. By combining LIMKi with 366 compounds from the GSK Published Kinase Inhibitor Set, effective combinations were identified with kinase inhibitors including EGFR, p38 and Raf. These findings encouraged a drug discovery effort that led to development of CRT0105446 and CRT0105950, which potently block LIMK1 and LIMK2 activity in vitro, and inhibit cofilin phosphorylation and increase αTubulin acetylation in cells. CRT0105446 and CRT0105950 were screened against 656 cancer cell lines, and rhabdomyosarcoma, neuroblastoma and kidney cancer cells were identified as significantly sensitive to both LIMK inhibitors. These large-scale screens have identified effective LIMK inhibitor drug combinations and sensitive cancer types. In addition, the LIMK inhibitory compounds CRT0105446 and CRT0105950 will enable further development of LIMK-targeted cancer therapy.


Asunto(s)
Quinasas Lim/antagonistas & inhibidores , Mitosis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Células MCF-7 , Microtúbulos/metabolismo , Mitosis/fisiología , Neoplasias/enzimología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/enzimología , Neuroblastoma/patología
9.
Gut ; 63(3): 480-93, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23585469

RESUMEN

OBJECTIVE: Colorectal cancer (CRC) is a major contributor to cancer mortality and morbidity. LIM kinase 2 (LIMK2) promotes tumour cell invasion and metastasis. The objectives of this study were to determine how LIMK2 expression is associated with CRC progression and patient outcome, and to use genetically modified Drosophila and mice to determine how LIMK2 deletion affects gastrointestinal stem cell regulation and tumour development. DESIGN: LIMK2 expression and activity were measured by immunostaining tumours from CRC-prone mice, human CRC cell lines and 650 human tumours. LIMK knockdown in Drosophila or Limk2 deletion in mice allowed for assessment of their contributions to gastrointestinal stem cell homeostasis and tumour development. RESULTS: LIMK2 expression was reduced in intestinal tumours of cancer-prone mice, as well as in human CRC cell lines and tumours. Reduced LIMK2 expression and substrate phosphorylation were associated with shorter patient survival. Genetic analysis in Drosophila midgut and intestinal epithelial cells isolated from genetically modified mice revealed a conserved role for LIMK2 in constraining gastrointestinal stem cell proliferation. Limk2 deletion increased colon tumour size in a colitis-associated colorectal mouse cancer model. CONCLUSIONS: This study revealed that LIMK2 expression and activity progressively decrease with advancing stage, and supports the hypothesis that there is selective pressure for reduced LIMK2 expression in CRC to relieve negative constraints imposed upon gastrointestinal stem cells.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Colon/enzimología , Neoplasias Colorrectales/enzimología , Mucosa Intestinal/enzimología , Quinasas Lim/metabolismo , Células Madre Neoplásicas/enzimología , Animales , Biomarcadores de Tumor/deficiencia , Línea Celular Tumoral , Proliferación Celular , Colon/patología , Colon/fisiopatología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/fisiopatología , Metilación de ADN , Progresión de la Enfermedad , Regulación hacia Abajo , Drosophila melanogaster , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Quinasas Lim/deficiencia , Ratones , Ratones Noqueados , Células Madre Neoplásicas/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares
10.
PLoS One ; 6(2): e17143, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21358804

RESUMEN

A common goal for potential cancer therapies is the identification of differences in protein expression or activity that would allow for the selective targeting of tumor vs. normal cells. The Ras proto-oncogene family (K-Ras, H-Ras and N-Ras) are amongst the most frequently mutated genes in human cancers. As a result, there has been substantial effort dedicated to determining which pathways are activated by Ras signaling and, more importantly, which of these contribute to cancer. Although the most widely studied Ras-regulated signaling pathway is the Raf/mitogen-activated protein kinase cascade, previous research in model systems has revealed that the Rac1 GTP-binding protein is also required for Ras-induced biological responses. However, what have been lacking are rigorous in vivo Rac1 target validation data and a clear demonstration that in Ras-driven hyperplastic lesions, Rac1 activity is increased. Using a combination of genetically-modified mouse models that allow for the tissue-selective activation or deletion of signaling molecules and an activation-state sensitive Rac1 antibody that detects GTP-bound Rac1, we found that Rac1 contributes to K-Ras induced epidermal papilloma initiation and growth and that Rac1 activity is elevated by oncogenic K-Ras in vivo. Previously, it was not practical to assess Rac1 activation status in the most commonly used format for clinical tumor specimens, formalin-fixed paraffin embedded (FFPE) tissues samples. However, this study clearly demonstrates that Rac1 is essential for K-Ras driven epithelial cell hyperproliferation and that Rac1 activity is elevated in tissues expressing mutant oncogenic K-Ras, while also characterizing the activation-state specific Rac1-GTP antibody as a probe to examine Rac1 activation status in FFPE samples. Our findings will facilitate further research on the status of Rac1 activity in human tumors and will help to define the tumor types of the patient population that could potentially benefit from therapies targeting Rac activation or downstream effector signaling pathways.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Epidermis/patología , Genes ras/fisiología , Proteína de Unión al GTP rac1/fisiología , Animales , Proliferación Celular , Activación Enzimática/fisiología , Epidermis/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Genes ras/genética , Humanos , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Transgénicos , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Papiloma/genética , Papiloma/metabolismo , Papiloma/patología , Proto-Oncogenes Mas , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Transgenes , Proteína de Unión al GTP rac1/metabolismo
11.
Cell Res ; 21(4): 666-82, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21079653

RESUMEN

The central arbiter of cell fate in response to DNA damage is p53, which regulates the expression of genes involved in cell cycle arrest, survival and apoptosis. Although many responses initiated by DNA damage have been characterized, the role of actin cytoskeleton regulators is largely unknown. We now show that RhoC and LIM kinase 2 (LIMK2) are direct p53 target genes induced by genotoxic agents. Although RhoC and LIMK2 have well-established roles in actin cytoskeleton regulation, our results indicate that activation of LIMK2 also has a pro-survival function following DNA damage. LIMK inhibition by siRNA-mediated knockdown or selective pharmacological blockade sensitized cells to radio- or chemotherapy, such that treatments that were sub-lethal when administered singly resulted in cell death when combined with LIMK inhibition. Our findings suggest that combining LIMK inhibitors with genotoxic therapies could be more efficacious than single-agent administration, and highlight a novel connection between actin cytoskeleton regulators and DNA damage-induced cell survival mechanisms.


Asunto(s)
Actinas/metabolismo , Regulación de la Expresión Génica , Quinasas Lim/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Citoesqueleto , Daño del ADN , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Immunoblotting , Ratones , Análisis por Micromatrices , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos , Transcripción Genética , Quinasas Asociadas a rho/metabolismo , Proteína rhoC de Unión a GTP
12.
Mol Cell Biol ; 26(16): 6209-22, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16880530

RESUMEN

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase, initially discovered as part of the NPM-ALK fusion protein, resulting from the t(2;5) translocation that is frequently associated with anaplastic large-cell lymphomas. The native ALK protein is normally expressed in the developing and, at a weaker level, adult nervous system. We recently demonstrated that the oncogenic, constitutively kinase-activated NPM-ALK protein was antiapoptotic when expressed in Jurkat lymphoblastic cells treated with cytotoxic drugs. In contrast, we now show that Jurkat cells overexpressing the wild-type ALK receptor are more sensitive to doxorubicin-induced apoptosis than parental cells. Moreover, the ALK protein is cleaved during apoptosis in a caspase-dependent manner. Mutation of aspartic residues to asparagine allowed us to map the caspase cleavage site in the juxtamembrane region of ALK. In order to assess the role of ALK in neural cell-derived tissue, we transiently expressed ALK in the 13.S.1.24 rat neuroblast immortalized cell line. ALK expression led to apoptotic cell death of the neuroblasts. ALK ligation by specific activating antibodies decreased ALK-facilitated apoptosis in both lymphoid and neuronal cell lines. Moreover, ALK transfection reduced the survival of primary cultures of cortical neurons. Thus, ALK has a proapoptotic activity in the absence of ligand, whereas it is antiapoptotic in the presence of its ligand and when the kinase is intrinsically activated. These properties place ALK in the growing family of dependence receptors.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores de Superficie Celular/metabolismo , Quinasa de Linfoma Anaplásico , Animales , Anticuerpos/inmunología , Apoptosis/efectos de los fármacos , Ácido Aspártico/genética , Caspasa 3 , Línea Celular Tumoral , Membrana Celular/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/enzimología , Doxorrubicina/farmacología , Activación Enzimática , Expresión Génica , Humanos , Células Jurkat , Ratones , Mutación/genética , Neuronas/citología , Neuronas/enzimología , Procesamiento Proteico-Postraduccional , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras , Transfección
13.
EMBO J ; 24(6): 1192-201, 2005 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-15729359

RESUMEN

Netrin-1 receptors UNC5H (UNC5H1-4) were originally proposed to mediate the chemorepulsive activity of netrin-1 during axonal guidance processes. However, UNC5H receptors were more recently described as dependence receptors and, as such, able to trigger apoptosis in the absence of netrin-1. They were also proposed as putative tumor suppressors. Here, we show that UNC5H2 physically interacts with the serine/threonine kinase death-associated protein kinase (DAP-kinase) both in cell culture and in embryonic mouse brains. This interaction occurs in part through the respective death domains of UNC5H2 and DAP-kinase. Moreover, part of UNC5H2 proapoptotic activity occurs through this interaction because UNC5H2-induced cell death is partly impaired in the presence of dominant-negative mutants of DAP-kinase or in DAP-kinase mutant murine embryonic fibroblast cells. In the absence of netrin-1, UNC5H2 reduces DAP-kinase autophosphorylation on Ser308 and increases the catalytic activity of the kinase while netrin-1 blocks UNC5H2-dependent DAP-kinase activation. Thus, the pair netrin-1/UNC5H2 may regulate cell fate by controlling the proapoptotic kinase activity of DAP-kinase.


Asunto(s)
Apoptosis/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Factores de Crecimiento Nervioso/fisiología , Receptores de Superficie Celular/fisiología , Proteínas Supresoras de Tumor/fisiología , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis , Encéfalo/citología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Caspasas/metabolismo , Catálisis , Células Cultivadas , Proteínas Quinasas Asociadas a Muerte Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Inmunoprecipitación , Ratones , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Receptores de Netrina , Netrina-1 , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Activación Transcripcional , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/farmacología
14.
Mol Cell Biol ; 24(23): 10328-39, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15542841

RESUMEN

The MET tyrosine kinase, the receptor of hepatocyte growth factor-scatter factor (HGF/SF), is known to be essential for normal development and cell survival. We report that stress stimuli induce the caspase-mediated cleavage of MET in physiological cellular targets, such as epithelial cells, embryonic hepatocytes, and cortical neurons. Cleavage occurs at aspartic residue 1000 within the SVD site of the juxtamembrane region, independently of the crucial docking tyrosine residues Y1001 or Y1347 and Y1354. This cleavage generates an intracellular 40-kDa MET fragment containing the kinase domain. The p40 MET fragment itself causes apoptosis of MDCK epithelial cells and embryonic cortical neurons, whereas its kinase-dead version is impaired in proapoptotic activity. Finally, HGF/SF treatment does not favor MET cleavage and apoptosis, confirming the known survival role of ligand-activated MET. Our results show that stress stimuli convert the MET survival receptor into a proapoptotic factor.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Proteínas Proto-Oncogénicas c-met/fisiología , Secuencia de Aminoácidos , Animales , Ácido Aspártico/química , Ácido Aspártico/metabolismo , Western Blotting , Caspasa 3 , Supervivencia Celular , Células Cultivadas , Citocinas/metabolismo , Perros , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Hepatocitos/metabolismo , Etiquetado Corte-Fin in Situ , Ligandos , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Neuronas/metabolismo , Plásmidos/metabolismo , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Recombinantes/química , Transfección , Tirosina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA