Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Dev Cell ; 5(4): 657-63, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14536066

RESUMEN

Obesity-associated diabetes is epidemic in industrialized societies. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) is highly expressed in adipose tissue and the presumed molecular target for antidiabetic thiazolidinedione drugs that reverse insulin resistance but also promote weight gain. Phosphorylation reduces the activity of PPARgamma in vitro, but physiological relevance has not been demonstrated. We have studied mice homozygous for a mutation (S112A) that prevents PPARgamma phosphorylation. Surprisingly, the weights and adipose mass of PPARgamma-S112A mice are not greater than wild-type. Remarkably, however, genetic prevention of PPARgamma phosphorylation preserves insulin sensitivity in the setting of diet-induced obesity. Underlying this protection are smaller fat cells, elevated serum adiponectin, and reduced free fatty acid levels. Thus, the phosphorylation state of PPARgamma modulates insulin sensitivity. Compounds that prevent PPARgamma phosphorylation or ligands that induce the conformation of nonphosphorylated PPARgamma may selectively enhance insulin sensitivity without increasing body weight.


Asunto(s)
Resistencia a la Insulina , Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología , Adiponectina , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo , Alanina/genética , Sustitución de Aminoácidos , Animales , Glucemia/metabolismo , Southern Blotting , Peso Corporal , Tamaño de la Célula , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Ácidos Grasos no Esterificados/sangre , Femenino , Expresión Génica , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Immunoblotting , Insulina/metabolismo , Insulina/farmacología , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/sangre , Ratones Transgénicos/genética , Mutación , Obesidad/sangre , Obesidad/inducido químicamente , Fosforilación , Proteínas/genética , Proteínas/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Serina/genética , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Mol Cell Biol ; 26(11): 4185-200, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16705170

RESUMEN

The genetic basis of pancreatic ductal adenocarcinoma, which constitutes the most common type of pancreatic malignancy, involves the sequential activation of oncogenes and inactivation of tumor suppressor genes. Among the pivotal genetic alterations are Ki-RAS oncogene activation and p53 tumor suppressor gene inactivation. We explain that the combination of these genetic events facilitates pancreatic carcinogenesis as revealed in novel three-dimensional cell (spheroid cyst) culture and in vivo subcutaneous and orthotopic xenotransplantation models. N-cadherin, a member of the classic cadherins important in the regulation of cell-cell adhesion, is induced in the presence of Ki-RAS mutation but subsequently downregulated with the acquisition of p53 mutation as revealed by gene microarrays and corroborated by reverse transcription-PCR and Western blotting. N-cadherin modulates the capacity of pancreatic ductal cells to migrate and invade, in part via complex formation with keratinocyte growth factor receptor and neural cell adhesion molecule and in part via interaction with p120-catenin. However, modulation of these complexes by Ki-RAS and p53 leads to enhanced cell migration and invasion. This preferentially induces the downstream effector AKT over mitogen-activated protein kinase to execute changes in cellular behavior. Thus, we are able to define molecules that in part are directly affected by Ki-RAS and p53 during pancreatic ductal carcinogenesis, and this provides a platform for potential new molecularly based therapeutic interventions.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Proteína Oncogénica p21(ras)/metabolismo , Conductos Pancreáticos/citología , Conductos Pancreáticos/patología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Uniones Adherentes/patología , Animales , Carcinoma Ductal Pancreático/patología , Técnicas de Cultivo de Célula , Transformación Celular Neoplásica , Quistes/patología , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Ratones , Mutación/genética , Invasividad Neoplásica/patología , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Proteína Oncogénica p21(ras)/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína p53 Supresora de Tumor/genética
3.
Stem Cell Reports ; 10(6): 1947-1958, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29805107

RESUMEN

Two principal stem cell pools orchestrate the rapid cell turnover in the intestinal epithelium. Rapidly cycling Lgr5+ stem cells are intercalated between the Paneth cells at the crypt base (CBCs) and injury-resistant reserve stem cells reside above the crypt base. The intermediate filament Keratin 15 (Krt15) marks either stem cells or long-lived progenitor cells that contribute to tissue repair in the hair follicle or the esophageal epithelium. Herein, we demonstrate that Krt15 labels long-lived and multipotent cells in the small intestinal crypt by lineage tracing. Krt15+ crypt cells display self-renewal potential in vivo and in 3D organoid cultures. Krt15+ crypt cells are resistant to high-dose radiation and contribute to epithelial regeneration following injury. Notably, loss of the tumor suppressor Apc in Krt15+ cells leads to adenoma and adenocarcinoma formation. These results indicate that Krt15 marks long-lived, multipotent, and injury-resistant crypt cells that may function as a cell of origin in intestinal cancer.


Asunto(s)
Transformación Celular Neoplásica , Mucosa Intestinal/citología , Queratina-15/metabolismo , Tolerancia a Radiación , Células Madre/metabolismo , Células Madre/efectos de la radiación , Animales , Biomarcadores , Diferenciación Celular , Proliferación Celular , Autorrenovación de las Células , Transformación Celular Neoplásica/metabolismo , Relación Dosis-Respuesta en la Radiación , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Ratones , Células de Paneth/citología , Células de Paneth/metabolismo , Células de Paneth/efectos de la radiación , Células Madre/citología
4.
J Clin Invest ; 110(6): 761-9, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12235107

RESUMEN

Squamous cancers of the oral cavity and esophagus are common worldwide, but no good genetically based animal model exists. A number of environmental factors as well as genetic alterations have been identified in these cancers, yet the specific combination of genetic events required for cancer progression remains unknown. The Epstein-Barr virus ED-L2 promoter (L2) can be used to target genes in a specific fashion to the oral-esophageal squamous epithelium. To that end, we generated L2-cyclin D1 (L2D1(+)) mice and crossbred these with p53-deficient mice. Whereas L2D1(+) mice exhibit a histologic phenotype of oral-esophageal dysplasia, the combination of cyclin D1 expression and p53 deficiency results in invasive oral-esophageal cancer. The development of the precancerous lesions was significantly reversed by the application of sulindac in the drinking water of the L2D1(+)/p53(+/-) mice. Furthermore, cell lines derived from oral epithelia of L2D1(+)/p53(+/-) and L2D1(+)/p53(-/-) mice, but not control mice, formed tumors in athymic nude mice. These data demonstrate that L2D1(+)/p53(+/-) mice provide a well-defined, novel, and faithful model of oral-esophageal cancer, which allows for the testing of novel chemopreventive, diagnostic, and therapeutic approaches.


Asunto(s)
Modelos Animales de Enfermedad , Neoplasias Esofágicas/genética , Neoplasias de la Boca/genética , Neoplasias de Células Escamosas/genética , Animales , Antineoplásicos/uso terapéutico , Ciclina D1/genética , Ciclina D1/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/patología , Genotipo , Herpesvirus Humano 4/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/patología , Neoplasias de Células Escamosas/tratamiento farmacológico , Neoplasias de Células Escamosas/patología , Regiones Promotoras Genéticas , Sulindac/uso terapéutico , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
5.
Cancer Biol Ther ; 6(4): 534-40, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17457048

RESUMEN

Insulin-like growth factor binding protein (IGFBP)-3 exerts either proapoptotic or growth stimulatory effects depending upon the cellular context. IGFBP-3 is overexpressed frequently in esophageal cancer. Yet, the role of IGFBP-3 in esophageal tumor biology remains elusive. To delineate the functional consequences of IGFBP-3 overexpression, we stably transduced Ha-Ras(V12)-transformed human esophageal cells with either wild-type or mutant IGFBP-3, the latter incapable of binding Insulin-like growth factor (IGFs) as a result of substitution of amino-terminal Ile56, Leu80, and Leu81 residues with Glycine residues. Wild-type, but not mutant, IGFBP-3 prevented IGF-1 from activating the IGF-1 receptor and AKT, and suppressed anchorage-independent cell growth. When xenografted in nude mice, in vivo bioluminescence imaging demonstrated that wild-type, but not mutant IGFBP-3, abrogated tumor formation by the Ras-transformed cells with concurrent induction of apoptosis, implying a prosurvival effect of IGF in cancer cell adaptation to the microenvironment. Moreover, there was more aggressive tumor growth by mutant IGFBP-3 overexpressing cells than control cell tumors, without detectable caspase-3 cleavage in tumor tissues, indicating an IGF-independent growth stimulatory effect of mutant IGFBP-3. In aggregate, these data suggest that IGFBP-3 contributes to esophageal tumor development and progression through IGF-dependent and independent mechanisms.


Asunto(s)
Neoplasias Esofágicas/patología , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Neoplasias Esofágicas/metabolismo , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Ratones , Ratones Desnudos , Mutación , Trasplante de Neoplasias , Receptor IGF Tipo 1/agonistas , Receptor IGF Tipo 1/metabolismo
6.
J Clin Invest ; 127(6): 2378-2391, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28481227

RESUMEN

The esophageal lumen is lined by a stratified squamous epithelium comprised of proliferative basal cells that differentiate while migrating toward the luminal surface and eventually desquamate. Rapid epithelial renewal occurs, but the specific cell of origin that supports this high proliferative demand remains unknown. Herein, we have described a long-lived progenitor cell population in the mouse esophageal epithelium that is characterized by expression of keratin 15 (Krt15). Genetic in vivo lineage tracing revealed that the Krt15 promoter marks a long-lived basal cell population able to self-renew, proliferate, and generate differentiated cells, consistent with a progenitor/stem cell population. Transcriptional profiling demonstrated that Krt15+ basal cells are molecularly distinct from Krt15- basal cells. Depletion of Krt15-derived cells resulted in decreased proliferation, thereby leading to atrophy of the esophageal epithelium. Further, Krt15+ cells were radioresistant and contributed to esophageal epithelial regeneration following radiation-induced injury. These results establish the presence of a long-lived and indispensable Krt15+ progenitor cell population that provides additional perspective on esophageal epithelial biology and the widely prevalent diseases that afflict this epithelium.


Asunto(s)
Esófago/citología , Queratina-15/metabolismo , Células Madre/fisiología , Animales , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Supervivencia Celular/efectos de la radiación , Esófago/fisiología , Esófago/efectos de la radiación , Homeostasis , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/citología , Regiones Promotoras Genéticas , Traumatismos Experimentales por Radiación/fisiopatología , Regeneración , Células Madre/efectos de la radiación , Activación Transcripcional
7.
Cancer Res ; 63(9): 2005-9, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12727809

RESUMEN

A frequent genetic alteration found in premalignant stages of pancreatic adenocarcinoma is K-ras oncogene point mutation. The mechanistic basis for the inability of K-ras mutation to transform pancreatic ductal cells is unclear, although cooperating events with p16 inactivation, p53 mutation, and SMAD 4 mutation are recognized to be necessary. We have generated a novel mouse model in which the cytokeratin 19 promoter, specifically active in pancreatic ductal cells but not other cell types of the pancreas, is fused to mutant K-ras. This is of direct relevance to human pancreatic cancer because premalignant lesions are found specifically in ductal cells. There is dramatic periductal lymphocytic infiltration in the pancreata of transgenic mice, predominantly CD4+ T lymphocytes, which may act as an adaptive immune response to activated ras-mediated signaling. In addition, gene array analysis reveals an induction of N-cadherin in transgenic mice pancreatic ductal cells, the significance of which relates to promotion of cell adhesion and deterrence of cell migration. Apart from these important biological considerations, there is parallel activity of the cytokeratin 19 promoter in the stem cell region of the gastric epithelium, namely in mucous neck cells. Activated K-ras in this context causes mucous neck cell hyperplasia, a precursor to gastric adenocarcinoma. There is concomitant parietal cell decrease, which is a key step toward gastric adenocarcinoma. Taken together, we have defined how mutant K-ras signaling modulates important molecular events in the initiating events of pancreatic and gastric carcinogenesis.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Transformación Celular Neoplásica/genética , Mucosa Gástrica/patología , Genes ras/genética , Linfocitos Infiltrantes de Tumor/patología , Neoplasias Pancreáticas/genética , Lesiones Precancerosas/genética , Neoplasias Gástricas/genética , Adenocarcinoma/genética , Animales , Humanos , Hiperplasia , Queratinas/genética , Ratones , Ratones Transgénicos , Mutación , Regiones Promotoras Genéticas , Transfección , Células Tumorales Cultivadas
8.
Oncogene ; 23(40): 6760-8, 2004 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-15273725

RESUMEN

Oncogenic Ras induces premature senescence in primary cells. Such an oncogene-induced senescence involves activation of tumor suppressor genes that provide a checkpoint mechanism against malignant transformation. In mouse, the ARF-p53 pathway mediates Ha-Ras(G12V)-induced senescence, and p19(ARF-/-) and p53(-/-) cells undergo transformation upon Ras activation. In addition, mouse cells, unlike human cells, express constitutively active telomerase and have long telomeres. However, it is unclear how Ras activation affects human cells of epithelial origin with p53 mutation and/or telomerase activation. In order to address this question, Ha-Ras(G12V) was expressed ectopically in primary as well as hTERT-immortalized human esophageal keratinocytes stably expressing dominant-negative p53 mutants. In human esophageal keratinocytes, we found that Ha-Ras(G12V) induced senescence regardless of p53 status and telomerase activation. Ras activation resulted in changes of cellular morphology, activation of senescence-associated beta-galactosidase, and suppression of cell proliferation, all coupled with reduction in the hyperphosphorylated form of the retinoblastoma protein (pRb). Furthermore, Ha-Ras(G12V) upregulated p16(INK4a) and downregulated cyclin-dependent kinase Cdk4 in human esophageal keratinocytes. Thus, Ras-mediated senescence may involve distinct mechanisms between human and mouse cells. Inactivation of the pRb pathway may be necessary for Ras to overcome senescence and transform human esophageal epithelial cells.


Asunto(s)
Senescencia Celular/fisiología , Genes ras/genética , Queratinocitos/fisiología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteínas ras/genética , Sustitución de Aminoácidos , Línea Celular , Senescencia Celular/genética , Proteínas de Unión al ADN , Esófago , Vectores Genéticos , Humanos , Retroviridae/genética , Telomerasa/genética , Telomerasa/metabolismo , Proteína p53 Supresora de Tumor/fisiología
9.
Diabetes ; 53(8): 1937-41, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15189975

RESUMEN

Resistin is an adipocyte-secreted protein that circulates at increased levels in obesity. Acute administration of resistin impairs glucose tolerance, but the effects of chronic hyperresistinemia have not been established. Here we describe the generation and characterization of transgenic mice that have high circulating levels of resistin in the setting of normal weight. Fasted blood glucose was higher in resistin-transgenic mice than in their nontransgenic littermates, and glucose tolerance was impaired in the hyperresistinemic mice. Metabolic studies in the setting of a hyperinsulinemic-euglycemic clamp protocol revealed that chronically hyperresistinemic mice have elevated glucose production. This increase in glucose production may be partly explained by increased expression of hepatic phosphoenolpyruvate carboxykinase. Thus, chronic hyperresistinemia impairs normal glucose metabolism.


Asunto(s)
Glucemia/metabolismo , Hormonas Ectópicas/sangre , Animales , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Homeostasis , Hiperinsulinismo , Insulina/administración & dosificación , Insulina/farmacología , Hígado/enzimología , Ratones , Ratones Transgénicos , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Prealbúmina/genética , Resistina
10.
Am J Cancer Res ; 2(4): 459-75, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22860235

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive forms of squamous cell carcinomas. Common genetic lesions in ESCC include p53 mutations and EGFR overexpression, both of which have been implicated in negative regulation of Notch signaling. In addition, cyclin D1 is overexpressed in ESCC and can be activated via EGFR, Notch and Wnt signaling. To elucidate how these genetic lesions may interact during the development and progression of ESCC, we tested a panel of genetically engineered human esophageal cells (keratinocytes) in organotypic 3D culture (OTC), a form of human tissue engineering. Notch signaling was suppressed in culture and mice by dominant negative Mastermind-like1 (DNMAML1), a genetic pan-Notch inhibitor. DNMAML1 mice were subjected to 4-Nitroquinoline 1-oxide-induced oral-esophageal carcinogenesis. Highly invasive characteristics of primary human ESCC were recapitulated in OTC as well as DNMAML1 mice. In OTC, cyclin D1 overexpression induced squamous hyperplasia. Concurrent EGFR overexpression and mutant p53 resulted in transformation and invasive growth. Interestingly, cell proliferation appeared to be regulated differentially between those committed to squamous-cell differentiation and those invading into the stroma. Invasive cells exhibited Notch-independent activation of cyclin D1 and Wnt signaling. Within the oral-esophageal squamous epithelia, Notch signaling regulated squamous-cell differentiation to maintain epithelial integrity, and thus may act as a tumor suppressor by preventing the development of a tumor-promoting inflammatory microenvironment.

11.
Cancer Cell ; 19(4): 470-83, 2011 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-21481789

RESUMEN

p120-catenin (p120ctn) interacts with E-cadherin, but to our knowledge, no formal proof that p120ctn functions as a bona fide tumor suppressor gene has emerged to date. We report herein that p120ctn loss leads to tumor development in mice. We have generated a conditional knockout model of p120ctn whereby mice develop preneoplastic and neoplastic lesions in the oral cavity, esophagus, and squamous forestomach. Tumor-derived cells secrete granulocyte macrophage colony-stimulating factor (GM-CSF), macrophage colony-stimulating factor (M-CSF), monocyte chemotactic protein-1 (MCP-1), and tumor necrosis factor-α (TNFα). The tumors contain significant desmoplasia and immune cell infiltration. Immature myeloid cells comprise a significant percentage of the immune cells present and likely participate in fostering a favorable tumor microenvironment, including the activation of fibroblasts.


Asunto(s)
Carcinoma de Células Escamosas/etiología , Cateninas/genética , Neoplasias Esofágicas/etiología , Genes Supresores de Tumor , Inflamación/etiología , Neoplasias de la Boca/etiología , Animales , Cadherinas/análisis , Cateninas/análisis , Cateninas/deficiencia , Cateninas/fisiología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Fibroblastos/fisiología , Humanos , Ratones , Células Mieloides/fisiología , FN-kappa B/fisiología , Catenina delta
12.
Science ; 303(5661): 1195-8, 2004 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-14976316

RESUMEN

The association between obesity and diabetes supports an endocrine role for the adipocyte in maintaining glucose homeostasis. Here we report that mice lacking the adipocyte hormone resistin exhibit low blood glucose levels after fasting, due to reduced hepatic glucose production. This is partly mediated by activation of adenosine monophosphate-activated protein kinase and decreased expression of gluconeogenic enzymes in the liver. The data thus support a physiological function for resistin in the maintenance of blood glucose during fasting. Remarkably, lack of resistin diminishes the increase in post-fast blood glucose normally associated with increased weight, suggesting a role for resistin in mediating hyperglycemia associated with obesity.


Asunto(s)
Glucemia/metabolismo , Ayuno , Hormonas Ectópicas/fisiología , Proteínas Quinasas Activadas por AMP , Adipocitos/metabolismo , Animales , Peso Corporal , Dieta , Grasas de la Dieta/administración & dosificación , Marcación de Gen , Gluconeogénesis , Prueba de Tolerancia a la Glucosa , Glucosa-6-Fosfatasa/metabolismo , Homeostasis , Hormonas Ectópicas/administración & dosificación , Hormonas Ectópicas/sangre , Hormonas Ectópicas/genética , Insulina/sangre , Hígado/metabolismo , Masculino , Ratones , Complejos Multienzimáticos/metabolismo , Obesidad/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes/administración & dosificación , Resistina , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA