Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
3.
Breast Cancer Res ; 14(4): R109, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22812567

RESUMEN

INTRODUCTION: Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is diagnosed in approximately 15% of all human breast cancer (BrCa) patients. Currently, no targeted therapies exist for this subtype of BrCa and prognosis remains poor. Our laboratory has previously identified a proliferation/DNA repair/cell cycle gene signature (Tag signature) that is characteristic of human TNBC. We hypothesize that targeting the dysregulated biological networks in the Tag gene signature will lead to the identification of improved combination therapies for TNBC. METHODS: Cross-species genomic analysis was used to identify human breast cancer cell lines that express the Tag signature. Knock-down of the up-regulated genes in the Tag signature by siRNA identified several genes that are critical for TNBC cell growth. Small molecule inhibitors to two of these genes were analyzed, alone and in combination, for their effects on cell proliferation, cell cycle, and apoptosis in vitro and tumor growth in vivo. Synergy between the two drugs was analyzed by the Chou-Talalay method. RESULTS: A custom siRNA screen was used to identify targets within the Tag signature that are critical for growth of TNBC cells. Ribonucleotide reductase 1 and 2 (RRM1 and 2) and checkpoint kinase 1 (CHK1) were found to be critical targets for TNBC cell survival. Combination therapy, to simultaneously attenuate cell cycle checkpoint control through inhibition of CHK1 while inducing DNA damage with gemcitabine, improved therapeutic efficacy in vitro and in xenograft models of TNBC. CONCLUSIONS: This combination therapy may have translational value for patients with TNBC and improve therapeutic response for this aggressive form of breast cancer.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/genética , Ribonucleótido Reductasas/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/genética , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Análisis por Conglomerados , Daño del ADN/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Concentración 50 Inhibidora , Ratones , Proteínas Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteína de Retinoblastoma/metabolismo , Ribonucleósido Difosfato Reductasa/antagonistas & inhibidores , Ribonucleósido Difosfato Reductasa/genética , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Carga Tumoral/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
4.
Toxicol Pathol ; 38(1): 88-95, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20080934

RESUMEN

Cross-species genomic analyses have proven useful for identifying common genomic alterations that occur in human cancers and mouse models designed to recapitulate human tumor development. High-throughput molecular analyses provide a valuable tool for identifying particular animal models that may represent aspects of specific subtypes of human cancers. Corresponding alterations in gene copy number and expression in tumors from mouse and human suggest that these conserved changes may be mechanistically essential for cancer development and progression, and therefore, they may be critical targets for therapeutic intervention. Using a cross-species analysis approach, mouse models in which the functions of p53, Rb, and BRCA1 have been disrupted demonstrate molecular features of human, triple-negative (ER-, PR-, and ERBB2-), basal-type breast cancer. Using mouse tumor models based on the targeted abrogation of p53 and Rb function, we identified a large, integrated genetic network that correlates to poor outcome in several human epithelial cancers. This gene signature is highly enriched for genes involved in DNA replication and repair, chromosome maintenance, cell cycle regulation, and apoptosis. Current studies are determining whether inactivation of specific members within this signature, using drugs or siRNA, will identify potentially important new targets to inhibit triple-negative, basal-type breast cancer for which no targeted therapies currently exist.


Asunto(s)
Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Neoplasias Mamarias Experimentales/genética , Animales , Femenino , Perfilación de la Expresión Génica , Ingeniería Genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ratones
5.
Breast Cancer Res ; 10(5): 213, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18828875

RESUMEN

The application of high-throughput genomic technologies has revealed that individual breast tumors display a variety of molecular features that require more personalized approaches to treatment. Several recent studies have demonstrated that a cross-species analytic approach provides a powerful means to filter through genetic complexity by identifying evolutionarily conserved genetic networks that are fundamental to the oncogenic process. Mouse-human tumor comparisons will provide insights into cellular origins of tumor subtypes, define interactive oncogenetic networks, identify potential novel therapeutic targets, and further validate as well as guide the selection of genetically engineered mouse models for preclinical testing.


Asunto(s)
Adenocarcinoma/genética , Neoplasias de la Mama/genética , Secuencia Conservada , Evolución Molecular , Redes Reguladoras de Genes , Genes Relacionados con las Neoplasias , Neoplasias Mamarias Experimentales/genética , Adenocarcinoma/clasificación , Adenocarcinoma/prevención & control , Adenocarcinoma/secundario , Animales , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/prevención & control , ADN de Neoplasias/genética , Progresión de la Enfermedad , Sistemas de Liberación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Epigénesis Genética , Femenino , Dosificación de Gen , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Modelos Animales , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/patología , Especificidad de la Especie , Estudios de Validación como Asunto
6.
Exp Cell Res ; 314(18): 3382-91, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18674534

RESUMEN

Differentiation of marrow-derived mesenchymal progenitors to either the osteoblast or adipocyte lineage is reciprocally regulated. Factors that promote osteoblastogenesis inhibit adipogenesis, while adipogenic factors are inhibitory to osteoblast differentiation. Heparin, a soluble glycosaminoglycan, inhibits bone formation in vivo and osteoblast cell differentiation and function in vitro, and has been shown to promote adipocyte differentiation. To elucidate the role that heparin plays in the adipogenic induction of murine mesenchymal progenitors, we studied immortalized marrow stromal cells (IM-MSC), the MSC cell line, ST2, and 3T3L1 pre-adipocytes. Heparin alone was not sufficient to induce adipogenesis, but enhanced the induction under a variety of adipogenic cocktails. This effect was both dose- and time-dependent. Heparin showed a positive effect at concentrations > 0.1 microg/ml when applied before day 3 during the induction course. Heparin's effect on adipogenesis was independent of cell proliferation, cell density, and extracellular lipid. This effect is likely related to the unique structure of heparin because another polyanionic glycosaminoglycan, dextran sulfate, did not promote adipogenic differentiation. Heparin treatment altered morphology and adhesion characteristics of progenitor cells, resulting in cell rounding and aggregation. As well, heparin counteracted the known inhibitory effect of fibronectin on adipogenesis and decreased basal focal adhesion kinase and paxillin phosphorylation. We conclude that heparin-mediated disruption of cell-matrix adhesion enhances adipogenic potential.


Asunto(s)
Adipocitos/fisiología , Diferenciación Celular/fisiología , Heparina/fisiología , Células Madre Mesenquimatosas/fisiología , Adipocitos/citología , Adipocitos/efectos de los fármacos , Animales , Secuencia de Bases , Western Blotting , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Sulfato de Dextran/farmacología , Relación Dosis-Respuesta a Droga , Matriz Extracelular/metabolismo , Heparina/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
7.
J Bone Miner Res ; 22(12): 1924-32, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17708715

RESUMEN

UNLABELLED: Overexpression of Wnt10b from the osteocalcin promoter in transgenic mice increases postnatal bone mass. Increases in osteoblast perimeter, mineralizing surface, and bone formation rate without detectable changes in pre-osteoblast proliferation, osteoblast apoptosis, or osteoclast number and activity suggest that, in this animal model, Wnt10b primarily increases bone mass by stimulating osteoblastogenesis. INTRODUCTION: Wnt signaling regulates many aspects of development including postnatal accrual of bone. Potential mechanisms for how Wnt signaling increases bone mass include regulation of osteoblast and/or osteoclast number and activity. To help differentiate between these possibilities, we studied mice in which Wnt10b is expressed specifically in osteoblast lineage cells or in mice devoid of Wnt10b. MATERIALS AND METHODS: Transgenic mice, in which mouse Wnt10b is expressed from the human osteocalcin promoter (Oc-Wnt10b), were generated in C57BL/6 mice. Transgene expression was evaluated by RNase protection assay. Quantitative assessment of bone variables was done by radiography, muCT, and static and dynamic histomorphometry. Mechanisms of bone homeostasis were evaluated with assays for BrdU, TUNEL, and TRACP5b activity, as well as serum levels of C-terminal telopeptide of type I collagen (CTX). The endogenous role of Wnt10b in bone was assessed by dynamic histomorphometry in Wnt10b(-/-) mice. RESULTS: Oc-Wnt10b mice have increased mandibular bone and impaired eruption of incisors during postnatal development. Analyses of femoral distal metaphyses show significantly higher BMD, bone volume fraction, and trabecular number. Increased bone formation is caused by increases in number of osteoblasts per bone surface, rate of mineral apposition, and percent mineralizing surface. Although number of osteoclasts per bone surface is not altered, Oc-Wnt10b mice have increased total osteoclast activity because of higher bone mass. In Wnt10b(-/-) mice, changes in mineralizing variables and osteoblast perimeter in femoral distal metaphyses were not observed; however, bone formation rate is reduced because of decreased total bone volume and trabecular number. CONCLUSIONS: High bone mass in Oc-Wnt10b mice is primarily caused by increased osteoblastogenesis, with a minor contribution from elevated mineralizing activity of osteoblasts.


Asunto(s)
Diferenciación Celular , Osteoblastos/metabolismo , Osteocalcina , Osteogénesis , Células Madre/metabolismo , Proteínas Wnt/biosíntesis , Fosfatasa Ácida/biosíntesis , Fosfatasa Ácida/genética , Animales , Animales Recién Nacidos , Apoptosis/genética , Densidad Ósea/genética , Diferenciación Celular/genética , Proliferación Celular , Homeostasis/genética , Humanos , Incisivo/crecimiento & desarrollo , Incisivo/metabolismo , Incisivo/patología , Isoenzimas/biosíntesis , Isoenzimas/genética , Mandíbula/crecimiento & desarrollo , Mandíbula/metabolismo , Mandíbula/patología , Ratones , Ratones Noqueados , Modelos Biológicos , Tamaño de los Órganos/genética , Osteoblastos/patología , Osteoclastos/metabolismo , Osteoclastos/patología , Células Madre/patología , Fosfatasa Ácida Tartratorresistente , Transgenes , Proteínas Wnt/genética
8.
J Biol Chem ; 282(19): 14515-24, 2007 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-17351296

RESUMEN

Mesenchymal precursor cells have the potential to differentiate into several cell types, including adipocytes and osteoblasts. Activation of Wnt/beta-catenin signaling shifts mesenchymal cell fate toward osteoblastogenesis at the expense of adipogenesis; however, molecular mechanisms by which Wnt signaling alters mesenchymal cell fate have not been fully investigated. Our prior work indicates that multipotent precursors express adipogenic and osteoblastogenic transcription factors at physiological levels and that ectopic expression of Wnt10b in bipotential ST2 cells suppresses expression of CCAAT/enhancer-binding protein alpha (C/EBPalpha) and peroxisome proliferator-activated receptor gamma (PPARgamma) and increases expression of Runx2, Dlx5, and osterix. Here, we demonstrate that transient activation of Wnt/beta-catenin signaling rapidly suppresses C/EBPalpha and PPARgamma, followed by activation of osteoblastogenic transcription factors. Enforced expression of C/EBPalpha or PPARgamma partially rescues lipid accumulation and decreases mineralization in ST2 cells expressing Wnt10b, suggesting that suppression of C/EBPalpha and PPARgamma is required for Wnt/beta-catenin to alter cell fate. Furthermore, knocking down expression of C/EBPalpha, PPARgamma, or both greatly reduces adipogenic potential and causes spontaneous osteoblastogenesis in ST2 cells and mouse embryonic fibroblasts, suggesting that Wnt signaling alters the fate of mesenchymal precursor cells primarily by suppressing C/EBPalpha and PPARgamma.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Células Madre Mesenquimatosas/fisiología , Osteogénesis , PPAR gamma/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Proteína alfa Potenciadora de Unión a CCAAT/genética , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , PPAR gamma/genética , Factor de Transcripción Sp7 , Células del Estroma/citología , Células del Estroma/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
9.
Diabetes ; 56(2): 295-303, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17259372

RESUMEN

The Wnt family of secreted signaling molecules has profound effects on diverse developmental processes, including the fate of mesenchymal progenitors. While activation of Wnt signaling blocks adipogenesis, inhibition of endogenous Wnt/beta-catenin signaling by Wnt10b promotes spontaneous preadipocyte differentiation. Transgenic mice with expression of Wnt10b from the FABP4 promoter (FABP4-Wnt10b) have less adipose tissue when maintained on a normal chow diet and are resistant to diet-induced obesity. Here we demonstrate that FABP4-Wnt10b mice largely avert weight gain and metabolic abnormalities associated with genetic obesity. FABP4-Wnt10b mice do not gain significant body weight on the ob/ob background, and at 8 weeks of age, they have an approximately 70% reduction in visceral and subcutaneous adipose tissues compared with ob/ob mice. Similarly, on the lethal yellow agouti (A(y)) background, FABP4-Wnt10b mice have 50-70% less adipose tissue weight and circulating leptin at 5 months of age. Wnt10b-Ay mice are more glucose tolerant and insulin sensitive than A(y) controls, perhaps due to reduced expression and circulation of resistin. Reduced expression of inflammatory cytokines may also contribute to improved glucose homeostasis.


Asunto(s)
Tejido Adiposo/fisiología , Proteínas de Unión a Ácidos Grasos/fisiología , Resistencia a la Insulina/fisiología , Obesidad/fisiopatología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Wnt/fisiología , Proteína de Señalización Agouti , Animales , Glucemia/fisiología , Modelos Animales de Enfermedad , Ingestión de Energía/fisiología , Femenino , Péptidos y Proteínas de Señalización Intercelular/genética , Leptina/deficiencia , Leptina/genética , Masculino , Ratones , Ratones Transgénicos , Obesidad/genética , Consumo de Oxígeno/fisiología , Paniculitis/fisiopatología
10.
Proc Natl Acad Sci U S A ; 102(9): 3324-9, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15728361

RESUMEN

Wnts comprise a family of secreted signaling proteins that regulate diverse developmental processes. Activation of Wnt signaling by Wnt10b inhibits differentiation of preadipocytes and blocks adipose tissue development; however, the effect of Wnt10b on other mesenchymal lineages has not been defined. To explore the physiological role of Wnt signaling in bone development, we analyzed FABP4-Wnt10b mice, which express the Wnt10b transgene in marrow. Femurs from FABP4-Wnt10b mice have almost four times as much bone in the distal metaphyses and are mechanically stronger. These mice maintain elevated bone mass at least through 23 months of age. In addition, FABP4-Wnt10b mice are protected from the bone loss characteristic of estrogen deficiency. We used pharmacological and genetic approaches to demonstrate that canonical Wnt signaling stimulates osteoblastogenesis and inhibits adipogenesis of bipotential mesenchymal precursors. Wnt10b shifts cell fate toward the osteoblast lineage by induction of the osteoblastogenic transcription factors Runx2, Dlx5, and osterix and suppression of the adipogenic transcription factors C/EBPalpha and PPARgamma. One mechanism whereby Wnt10b promotes osteoblastogenesis is suppression of PPARgamma expression. Finally, Wnt10b-/- mice have decreased trabecular bone and serum osteocalcin, confirming that Wnt10b is an endogenous regulator of bone formation.


Asunto(s)
Densidad Ósea/fisiología , División Celular/fisiología , Osteoblastos/citología , Proteínas Proto-Oncogénicas/fisiología , Envejecimiento/fisiología , Animales , Femenino , Ratones , Ratones Noqueados , Ovariectomía , Proteínas Proto-Oncogénicas/genética , Proteínas Wnt
11.
Circ Res ; 94(9): 1235-41, 2004 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-15059934

RESUMEN

Hypertrophic cardiomyopathy mutations A63V and E180G in alpha-tropomyosin (alpha-Tm) have been shown to cause slow cardiac muscle relaxation. In this study, we used two complementary genetic strategies, gene transfer in isolated rat myocytes and transgenesis in mice, to ascertain whether parvalbumin (Parv), a myoplasmic calcium buffer, could correct the diastolic dysfunction caused by these mutations. Sarcomere shortening measurements in rat cardiac myocytes expressing the alpha-Tm A63V mutant revealed a slower time to 50% relengthening (T50R: 44.2+/-1.4 ms in A63V, 36.8+/-1.0 ms in controls; n=96 to 108; P<0.001) when compared with controls. Dual gene transfer of alpha-Tm A63V and Parv caused a marked decrease in T50R (29.8+/-1.0 ms). However, this increase in relaxation rate was accompanied with a decrease in shortening amplitude (114.6+/-4.4 nm in A63+Parv, 137.8+/-5.3 nm in controls). Using an asynchronous gene transfer strategy, Parv expression was reduced (from approximately 0.12 to approximately 0.016 mmol/L), slow relaxation redressed, and shortening amplitude maintained (T50R=33.9+/-1.6 ms, sarcomere shortening amplitude=132.2+/-7.0 nm in A63V+PVdelayed; n=56). Transgenic mice expressing the E180G alpha-Tm mutation and mice expressing Parv in the heart were crossed. In isolated adult myocytes, the alpha-Tm mutation alone (E180G+/PV-) had slower sarcomere relengthening kinetics than the controls (T90R: 199+/-7 ms in E180G+/PV-, 130+/-4 ms in E180G-/PV-; n=71 to 72), but when coexpressed with Parv, cellular relaxation was faster (T90R: 36+/-4 ms in E180G+/PV+). Collectively, these findings show that slow relaxation caused by alpha-Tm mutants can be corrected by modifying calcium handling with Parv.


Asunto(s)
Señalización del Calcio/fisiología , Cardiomiopatía Hipertrófica/genética , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Parvalbúminas/fisiología , Tropomiosina/genética , Citoesqueleto de Actina/ultraestructura , Sustitución de Aminoácidos , Animales , Señalización del Calcio/genética , Cruzamientos Genéticos , Estudios de Factibilidad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Mutación Missense , Parvalbúminas/genética , Mutación Puntual , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/fisiología , Sarcómeros/ultraestructura , Relación Estructura-Actividad , Factores de Tiempo , Transducción Genética , Tropomiosina/química
12.
Biochem Biophys Res Commun ; 302(1): 12-6, 2003 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-12593840

RESUMEN

This report examines the balance of positive and negative adipogenic factors in a line of immortalized 243 embryonic fibroblasts that undergo spontaneous preadipocyte differentiation. Control of adipogenesis reflects the interplay of factors that promote or inhibit expression of C/EBPalpha and PPARgamma. The 243 cells express C/EBPalpha early and at elevated levels compared to 3T3-F442A preadipocytes or adipocytes. Cell clones were derived from the heterogeneous 243 population for ability or inability to differentiate into adipocytes. Wnt10b, a secreted protein that inhibits adipogenesis, is expressed at high levels in cells with low adipogenic potential and is undetectable in preadipocytes that spontaneously differentiate. In contrast, C/EBPalpha is expressed at reduced levels in cells with low adipogenic potential, and is expressed at high levels in preadipocytes that spontaneously differentiate. These data are consistent with a model in which decreased Wnt10b, coupled with increased C/EBPalpha, results in induction of PPARgamma and spontaneous adipogenesis of 243 cells.


Asunto(s)
Adipocitos/citología , Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Proteínas Proto-Oncogénicas/fisiología , Células 3T3 , Animales , Diferenciación Celular , Línea Celular Transformada , Células Clonales , Ratones , Proteínas Wnt
13.
J Biol Chem ; 277(34): 30998-1004, 2002 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-12055200

RESUMEN

We have identified Wnt10b as a potent inhibitor of adipogenesis that must be suppressed for preadipocytes to differentiate in vitro. Here, we demonstrate that a specific inhibitor of glycogen synthase kinase 3, CHIR 99021, mimics Wnt signaling in preadipocytes. CHIR 99021 stabilizes free cytosolic beta-catenin and inhibits adipogenesis by blocking induction of CCAAT/enhancer-binding protein alpha and peroxisome proliferator-activated receptor gamma. Preadipocyte differentiation is inhibited when 3T3-L1 cells are exposed to CHIR 99021 for any 24 h period during the first 3 days of adipogenesis. Consistent with this time frame of inhibition, expression of Wnt10b mRNA is suppressed upon induction of differentiation, with a 50% decline by 6 h and complete inhibition by 36 h. Of the agents used to induce differentiation, exposure of 3T3-L1 cells to methyl-isobutylxanthine or cAMP is sufficient to suppress expression of Wnt10b mRNA. Inhibition of adipogenesis by Wnt10b is likely mediated by Wnt receptors, Frizzled 1, 2, and/or 5, and co-receptors low density lipoprotein receptor-related proteins 5 and 6. These receptors, like Wnt10b, are highly expressed in preadipocytes and stromal vascular cells. Finally, we demonstrate that disruption of extracellular Wnt signaling by expression of secreted Frizzled related proteins causes spontaneous adipocyte conversion.


Asunto(s)
Adipocitos/fisiología , Proteínas Proto-Oncogénicas/fisiología , Células 3T3 , Animales , Proteína alfa Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Diferenciación Celular , AMP Cíclico/fisiología , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3 , Glucógeno Sintasa Quinasas , Ratones , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Células Madre/fisiología , Factores de Transcripción/antagonistas & inhibidores , Proteínas Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA