Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Mol Cell ; 38(4): 512-23, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20513427

RESUMEN

PHLPP1 and PHLPP2 phosphatases exert their tumor-suppressing functions by dephosphorylation and inactivation of Akt in several breast cancer and glioblastoma cells. However, Akt, or other known targets of PHLPPs that include PKC and ERK, may not fully elucidate the physiological role of the multifunctional phosphatases, especially their powerful apoptosis induction function. Here, we show that PHLPPs induce apoptosis in cancer cells independent of the known targets of PHLPPs. We identified Mst1 as a binding partner that interacts with PHLPPs both in vivo and in vitro. PHLPPs dephosphorylate Mst1 on the T387 inhibitory site, which activate Mst1 and its downstream effectors p38 and JNK to induce apoptosis. The same T387 site can be phosphorylated by Akt. Thus, PHLPP, Akt, and Mst1 constitute an autoinhibitory triangle that controls the fine balance of apoptosis and proliferation that is cell type and context dependent.


Asunto(s)
Apoptosis , Factor de Crecimiento de Hepatocito/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor de Crecimiento de Hepatocito/deficiencia , Humanos , Ratones , Fosforilación , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
2.
Biochem Biophys Res Commun ; 489(1): 56-62, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28527887

RESUMEN

MST1 deficiency causes T and B cell lymphopenia, resulting in combined immunodeficiency. However, MST1-deficient patients also exhibit autoimmune-like symptoms such as hypergammaglobulinemia and autoantibody production. Recent studies have shown that the autoimmune responses observed in MST1-deficient patients were most likely attributable to defective regulatory T (Treg) cells instead of intrinsic signals in MST1-lacking B cells. Nevertheless, it is not determined how MST1 deficiency in T cells breaks B cell tolerance and causes systemic autoimmune-like phenotypes. In this study, we confirmed that Mst1-/- mice developed hypergammaglobulinemia associated with increased levels of IgG, IgA, and IgE. We also showed that uncontrolled B cell responses were resulted from the IL-4-rich environment created by CD4+ T cells. Defective MST1-FOXO1 signaling down-regulated Treg cells, resulting in the collapse of immune tolerance where the populations of Th2 and T follicular helper cells expanded. In conclusion, we suggest that MST1 acts as a molecular brake to maintain immune tolerance by regulating T cell-mediated B cell activation.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Factor de Crecimiento de Hepatocito/inmunología , Hipergammaglobulinemia/inmunología , Interleucina-4/inmunología , Proteínas Proto-Oncogénicas/inmunología , Animales , Factor de Crecimiento de Hepatocito/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/deficiencia
3.
J Mol Cell Cardiol ; 98: 108-16, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27496379

RESUMEN

Emerging evidence favors the notion that macrophage autophagy plays a prominent role in the pathogenesis of vulnerable plaque, suggesting the therapeutic potential of targeting autophagy in atherosclerosis. Here ApoE(-/-) mice were crossed with Mst1 knockout or Mst1 Tg mice to generate ApoE(-/-):Mst1(-/-) and ApoE(-/-):Mst1Tg mice. All animals were fed high-fat-diet for 4months to induce arterial atherosclerosis. Murine macrophage RAW264.7 cells were subjected to ox-LDL (50µg/mL) in an effort to examine the cellular mechanisms. A significant increase in the levels of Mst1 and p-Mst1 was observed in the aorta of ApoE(-/-) mice. Mst1 knockout significantly reduced atherosclerotic area, decreased lipid core area and macrophage accumulation as compared with ApoE(-/-) mice. Along the same line, Mst1 overexpression increased plaque area, lipid core and macrophage accumulation as compared with ApoE(-/-) mice. Mst1 deficiency significantly increased levels of Beclin1 and LC3II, while decreased that of p62 in aortic atherosclerosis. Moreover, in vitro data indicated that Mst1 knockdown prompted more typical autophagosomes upon ox-LDL challenge. Mst1 knockdown also enhanced autophagic flux as evidenced by GFP-mRFP-LC3 staining, increased LC3-II expression and decreased p62 expression in the presence of bafilomycin A1. Mst1 knockdown decreased, while Mst1 overexpression increased macrophage apoptosis upon ox-LDL exposure. In conclusion, Mst1 deficiency diminishes atherosclerosis and stabilizes atherosclerotic plaques in ApoE(-/-) mice. Mst1 may participate in atherosclerosis progression through inhibition of macrophage autophagy and promotion of macrophage apoptosis.


Asunto(s)
Apoptosis/genética , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Autofagia/genética , Factor de Crecimiento de Hepatocito/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas Proto-Oncogénicas/genética , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Genotipo , Factor de Crecimiento de Hepatocito/deficiencia , Factor de Crecimiento de Hepatocito/metabolismo , Lipoproteínas LDL/administración & dosificación , Lipoproteínas LDL/metabolismo , Ratones , Ratones Noqueados , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/metabolismo
4.
EMBO J ; 30(12): 2465-76, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21572393

RESUMEN

Microglia, the resident macrophages of the mammalian central nervous system, migrate to sites of tissue damage or infection and become activated. Although the persistent secretion of inflammatory mediators by the activated cells contributes to the pathogenesis of various neurological disorders, most activated microglia eventually undergo apoptosis through the process of activation-induced cell death (AICD). The molecular mechanism of AICD, however, has remained unclear. Here, we show that Daxx and mammalian Ste20-like kinase-1 (MST1) mediate apoptosis elicited by interferon-γ (IFN-γ) in microglia. IFN-γ upregulated the expression of Daxx, which in turn mediated the homodimerization, activation, and nuclear translocation of MST1 and apoptosis in microglial cells. Depletion of Daxx or MST1 by RNA interference also attenuated IFN-γ-induced cell death in primary rat microglia. Furthermore, the extent of IFN-γ-induced death of microglia in the brain of MST1-null mice was significantly reduced compared with that apparent in wild-type mice. Our results thus highlight new functions of Daxx and MST1 that they are the key mediators of microglial cell death initiated by the proinflammatory cytokine IFN-γ.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Portadoras/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Microglía/citología , Microglía/fisiología , Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal , Animales , Apoptosis/genética , Células COS , Proteínas Portadoras/genética , Muerte Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Chlorocebus aethiops , Proteínas Co-Represoras , Fibroblastos/citología , Fibroblastos/fisiología , Células HEK293 , Células HeLa , Factor de Crecimiento de Hepatocito/deficiencia , Factor de Crecimiento de Hepatocito/genética , Humanos , Mediadores de Inflamación , Interferón gamma/administración & dosificación , Interferón gamma/fisiología , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Chaperonas Moleculares , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética
5.
Hepatology ; 59(4): 1435-47, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24242874

RESUMEN

UNLABELLED: Sorafenib--a broad kinase inhibitor--is a standard therapy for advanced hepatocellular carcinoma (HCC) and has been shown to exert antifibrotic effects in liver cirrhosis, a precursor of HCC. However, the effects of sorafenib on tumor desmoplasia--and its consequences on treatment resistance--remain unknown. We demonstrate that sorafenib has differential effects on tumor fibrosis versus liver fibrosis in orthotopic models of HCC in mice. Sorafenib intensifies tumor hypoxia, which increases stromal-derived factor 1 alpha (SDF-1α) expression in cancer and stromal cells and, subsequently, myeloid differentiation antigen-positive (Gr-1(+)) myeloid cell infiltration. The SDF-1α/C-X-C receptor type 4 (CXCR4) pathway directly promotes hepatic stellate cell (HSC) differentiation and activation through the mitogen-activated protein kinase pathway. This is consistent with the association between SDF-1α expression with fibrotic septa in cirrhotic liver tissues as well as with desmoplastic regions of human HCC samples. We demonstrate that after treatment with sorafenib, SDF-1α increased the survival of HSCs and their alpha-smooth muscle actin and collagen I expression, thus increasing tumor fibrosis. Finally, we show that Gr-1(+) myeloid cells mediate HSC differentiation and activation in a paracrine manner. CXCR4 inhibition, using AMD3100 in combination with sorafenib treatment, prevents the increase in tumor fibrosis--despite persistently elevated hypoxia--in part by reducing Gr-1(+) myeloid cell infiltration and inhibits HCC growth. Similarly, antibody blockade of Gr-1 reduces tumor fibrosis and inhibits HCC growth when combined with sorafenib treatment. CONCLUSION: Blocking SDF-1α/CXCR4 or Gr-1(+) myeloid cell infiltration may reduce hypoxia-mediated HCC desmoplasia and increase the efficacy of sorafenib treatment.


Asunto(s)
Antígeno CD11b/metabolismo , Carcinoma Hepatocelular/metabolismo , Quimiocina CXCL12/metabolismo , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Células Mieloides/patología , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Receptores CXCR4/metabolismo , Receptores de Quimiocina/metabolismo , Animales , Tetracloruro de Carbono/efectos adversos , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Factor de Crecimiento de Hepatocito/deficiencia , Factor de Crecimiento de Hepatocito/genética , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Células Mieloides/metabolismo , Niacinamida/farmacología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Transducción de Señal/fisiología , Sorafenib
6.
Hepatology ; 55(4): 1215-26, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22095660

RESUMEN

UNLABELLED: Hepatocyte growth factor (HGF)/c-Met supports a pleiotrophic signal transduction pathway that controls stem cell homeostasis. Here, we directly addressed the role of c-Met in stem-cell-mediated liver regeneration by utilizing mice harboring c-met floxed alleles and Alb-Cre or Mx1-Cre transgenes. To activate oval cells, the hepatic stem cell (HSC) progeny, we used a model of liver injury induced by diet containing the porphyrinogenic agent, 3,5-diethocarbonyl-1,4-dihydrocollidine (DDC). Deletion of c-met in oval cells was confirmed in both models by polymerase chain reaction analysis of fluorescence-activated cell-sorted epithelial cell adhesion molecule (EpCam)-positive cells. Loss of c-Met receptor decreased the sphere-forming capacity of oval cells in vitro as well as reduced oval cell pool, impaired migration, and decreased hepatocytic differentiation in vivo, as demonstrated by double immunofluorescence using oval- (A6 and EpCam) and hepatocyte-specific (i.e. hepatocyte nuclear factor 4-alpha) antibodies. Furthermore, lack of c-Met had a profound effect on tissue remodeling and overall composition of HSC niche, which was associated with greatly reduced matrix metalloproteinase (MMP)9 activity and decreased expression of stromal-cell-derived factor 1. Using a combination of double immunofluorescence of cell-type-specific markers with MMP9 and gelatin zymography on the isolated cell populations, we identified macrophages as a major source of MMP9 in DDC-treated livers. The Mx1-Cre-driven c-met deletion caused the greatest phenotypic impact on HSCs response, as compared to the selective inactivation in the epithelial cell lineages achieved in c-Met(fl/fl); Alb-Cre(+/-) mice. However, in both models, genetic loss of c-met triggered a similar cascade of events, leading to the failure of HSC mobilization and death of the mice. CONCLUSION: These results establish a direct contribution of c-Met in the regulation of HSC response and support a unique role for HGF/c-Met as an essential growth-factor-signaling pathway for regeneration of diseased liver.


Asunto(s)
Factor de Crecimiento de Hepatocito/fisiología , Regeneración Hepática/fisiología , Proteínas Proto-Oncogénicas c-met/fisiología , Transducción de Señal/fisiología , Células Madre/fisiología , Animales , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/terapia , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Factor de Crecimiento de Hepatocito/deficiencia , Factor de Crecimiento de Hepatocito/genética , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Mutantes , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-met/deficiencia , Proteínas Proto-Oncogénicas c-met/genética , Piridinas/efectos adversos , Trasplante de Células Madre , Células Madre/citología
7.
J Immunol ; 183(6): 3865-72, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19692642

RESUMEN

The MST1 kinase was recently identified as playing an essential role in the promotion of lymphocyte polarization and adhesion stimulated by chemokines and TCR signaling. However, the physiological relevance of the Mst1 pathway in thymocyte development is not completely understood. In this study, we analyzed the effect of Mst1 disruption on thymocyte development and migration. Mst1-deficient (Mst1(-/-)) mice displayed an accumulation of mature thymocytes in the thymus, a dramatic reduction of lymphocytes in blood and peripheral lymphoid tissues, and a decrease of homing ability to peripheral lymph nodes. Mst1(-/-) thymocytes were impaired in chemotactic response to chemokines, such as CCL19, but not to sphingosine-1-phosphate. Further analyses of Mst1(-/-) mice revealed a severe impairment in the egress of mature T cells from the thymus. T lineage-specific knockout of the Mst1 gene demonstrates a cell-intrinsic role for Mst1 in regulating T cell development. Our study indicates that Mst1 is crucial in controlling lymphocyte chemotaxis and thymocyte emigration.


Asunto(s)
Quimiotaxis , Factor de Crecimiento de Hepatocito/fisiología , Proteínas Proto-Oncogénicas/fisiología , Timo/citología , Animales , Quimiocinas , Factor de Crecimiento de Hepatocito/deficiencia , Ganglios Linfáticos , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/deficiencia , Linfocitos T/citología
8.
Biochem Pharmacol ; 188: 114574, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33887258

RESUMEN

Hyperglycaemic memory refers to the damages occurred under early hyperglycaemic environment in organs of diabetic patients persisting after intensive glycaemic control. Mammalian sterile 20-like kinase 1 (Mst1) contributes to the development of diabetic cardiomyopathy. Here, we investigated the role of Mst1 in hyperglycaemic memory and test the effect of XMU-MP-1, a Mst1 inhibitor, on hyperglycaemic memory in hearts. Eight weeks after induction of type 1 diabetes by injection with streptozotocin (STZ) in mice, glycaemic control was obtained by means of insulin treatment and maintained for 4 additional weeks. In the diabetic mice, insulin treatment alone did not reduce phosphorylation of Mst1 or improve cardiac function. Treatment with XMU-MP-1 alone immediately after induction of diabetes for 12 weeks did not improve myocardial function in mice. But treatment with XMU-MP-1 for the later 4 weeks relieved myocardial dysfunction when glycaemic control was obtained by insulin treatment simultaneously. Mst1 deficiency and glycaemic control synergistically improved myocardial function and reduced apoptosis in myocardium of diabetic mice. Mechanistically, when Mst1 was deficient or inhibited by XMU-MP-1, AMPK was activated and mitochondrial dysfunction was attenuated. In vitro, treatment with AMPK activator reversed the detrimental effects of Mst1 overexpression in cultured cardiomyocytes. XMU-MP-1 might thus be envisaged as a complement for insulin treatment against diabetic cardiomyopathy.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Factor de Crecimiento de Hepatocito/deficiencia , Hiperglucemia/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Proteínas Proto-Oncogénicas/deficiencia , Sulfonamidas/uso terapéutico , Animales , Línea Celular , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatías Diabéticas/tratamiento farmacológico , Cardiomiopatías Diabéticas/metabolismo , Relación Dosis-Respuesta a Droga , Factor de Crecimiento de Hepatocito/genética , Hiperglucemia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas/genética , Ratas , Sulfonamidas/farmacología , Resultado del Tratamiento
9.
Cancer Res ; 67(20): 9844-51, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17942915

RESUMEN

Hepatocyte growth factor (HGF) has been reported to have both positive and negative effects on carcinogenesis. Here, we show that the loss of c-Met signaling in hepatocytes enhanced rather than suppressed the early stages of chemical hepatocarcinogenesis. c-Met conditional knockout mice (c-metfl/fl, AlbCre+/-; MetLivKO) treated with N-nitrosodiethylamine developed significantly more and bigger tumors and with a shorter latency compared with control (w/w, AlbCre+/-; Cre-Ctrl) mice. Accelerated tumor development was associated with increased rate of cell proliferation and prolonged activation of epidermal growth factor receptor (EGFR) signaling. MetLivKO livers treated with N-nitrosodiethylamine also displayed elevated lipid peroxidation, decreased ratio of reduced glutathione to oxidized glutathione, and up-regulation of superoxide dismutase 1 and heat shock protein 70, all consistent with increased oxidative stress. Likewise, gene expression profiling done at 3 and 5 months after N-nitrosodiethylamine treatment revealed up-regulation of genes associated with cell proliferation and stress responses in c-Met mutant livers. The negative effects of c-Met deficiency were reversed by chronic p.o. administration of antioxidant N-acetyl-L-cysteine. N-acetyl-L-cysteine blocked the EGFR activation and reduced the N-nitrosodiethylamine-initiated hepatocarcinogenesis to the levels of Cre-Ctrl mice. These results argue that intact HGF/c-Met signaling is essential for maintaining normal redox homeostasis in the liver and has tumor suppressor effect(s) during the early stages of N-nitrosodiethylamine-induced hepatocarcinogenesis.


Asunto(s)
Factor de Crecimiento de Hepatocito/deficiencia , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/metabolismo , Proteínas Proto-Oncogénicas c-met/deficiencia , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Cocarcinogénesis , Dietilnitrosamina , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Noqueados , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal
10.
Front Biosci ; 13: 7072-86, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508717

RESUMEN

Hepatocyte growth factor (HGF) and Met/HGF receptor play roles in dynamic growth and morphogenesis during development and regeneration of organs, including the kidney. In the kidney, HGF targets different types of cells, while its biological actions depend on a target cell type. During the earlier stages of chronic renal failure, renal HGF expression increased, but in later stages HGF expression decreased, associated with manifestation of chronic renal failure. When anti-HGF IgG was administered into mice with chronic renal failure, renal dysfunction and fibrosis were accelerated, indicating a role of endogenous HGF to suppress chronic renal failure. For myofibroblasts, a key cell type in tissue fibrosis, HGF exerted biological activities, including (i) inhibition of growth, (ii) suppression of fibrogenic cytokine expression, and (iii) enhancement of matrix metalloproteinases involved in subsequent apoptosis. In models of glomerular and tubulo-interstitial fibrosis, administration of HGF or HGF gene therapy improved renal fibrosis and dysfunction. Since insufficient production of HGF is causative for renal fibrosis, supplementation with HGF represents a new approach to inhibit or improve chronic renal failure.


Asunto(s)
Terapia Genética/métodos , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/uso terapéutico , Fallo Renal Crónico/tratamiento farmacológico , Fallo Renal Crónico/patología , Animales , Fibrosis , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Glomeruloesclerosis Focal y Segmentaria/prevención & control , Factor de Crecimiento de Hepatocito/deficiencia , Humanos , Fallo Renal Crónico/genética , Túbulos Renales/patología , Túbulos Renales/fisiología , Ratones , Factor de Crecimiento Derivado de Plaquetas/fisiología , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/fisiología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores
11.
Stem Cell Res ; 20: 42-49, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28257933

RESUMEN

Adult fibroblasts can be reprogrammed into induced pluripotent stem cells (iPSC) for use in various applications. However, there are challenges in iPSC generation including low reprogramming efficiency, yield, cell survival and viability. Since the Hippo signalling pathway is a key pathway involved in regulating cell proliferation and survival, we here test whether modification of the Hippo pathway will enhance the efficiency of iPSC generation and improve their survival. The Hippo pathway was modified by genetic ablation of the mammalian sterile-20 like kinase 1 (Mst1), a major component of the pathway. Using adult skin fibroblasts isolated from Mst1 knockout mice (Mst1-/-) as a source of iPSC we found that genetic ablation of Mst1 leads to significantly increased reprogramming efficiency by 43.8%. Moreover, Mst1-/- iPSC displayed increase proliferation by 12% as well as an increase in cell viability by 20% when treated with a chemical hypoxic inducer. Mechanistically, we found higher activity of YAP, the main downstream effector of the Hippo pathway, in iPSC lacking Mst1. In conclusion, our data suggests that Mst1 can be targeted to improve the efficiency of adult somatic cell reprogramming as well as to enhance iPSC proliferation and survival.


Asunto(s)
Reprogramación Celular , Factor de Crecimiento de Hepatocito/genética , Células Madre Pluripotentes Inducidas/citología , Proteínas Proto-Oncogénicas/genética , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Factor de Crecimiento de Hepatocito/deficiencia , Vía de Señalización Hippo , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Circulation ; 111(11): 1407-14, 2005 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-15781752

RESUMEN

BACKGROUND: Hepatocyte growth factor (HGF) is an important mitogen and morphogen that contributes to the repair process after lung injury. The goal of the present study was to characterize its role in pulmonary emphysema, which may lead to the development of new treatment strategies with HGF. METHODS AND RESULTS: HGF mRNA and protein levels in lung tissue and plasma from elastase-induced emphysema rats transiently increased, then declined significantly to below the basal level in a time-dependent manner (P<0.01). Furthermore, changes in HGF were correlated with histologically progressive emphysematous changes and deterioration in pulmonary physiology. Use of the HVJ (hemagglutinating virus of Japan) envelope method resulted in successful transfection of cDNA encoding human HGF, as demonstrated by an efficient expression of HGF in alveolar endothelial and epithelial cells. Transfection of HGF resulted in a more extensive pulmonary vasculature and inhibition of alveolar wall cell apoptosis, and those effects led to improved exercise tolerance and gas exchange (P<0.05), which persisted for more than 1 month. CONCLUSIONS: Decreased HGF expression due to a failure in sustained endogenous production after injury was associated with emphysema-related histopathologic and physiological changes in the present rat model. In addition, induction of HGF expression by a gene-transfection method resulted in improved pulmonary function via inhibition of alveolar cell apoptosis, enhancement of alveolar regeneration, and promotion of angiogenesis.


Asunto(s)
Terapia Genética , Vectores Genéticos/uso terapéutico , Factor de Crecimiento de Hepatocito/fisiología , Enfisema Pulmonar/terapia , Animales , Apoptosis , División Celular , ADN Complementario/administración & dosificación , ADN Complementario/genética , Modelos Animales de Enfermedad , Genes Reporteros , Factor de Crecimiento de Hepatocito/biosíntesis , Factor de Crecimiento de Hepatocito/deficiencia , Factor de Crecimiento de Hepatocito/genética , Humanos , Flujometría por Láser-Doppler , Masculino , Neovascularización Fisiológica , Elastasa Pancreática/toxicidad , Fenotipo , Proteínas Proto-Oncogénicas c-met/biosíntesis , Proteínas Proto-Oncogénicas c-met/genética , Circulación Pulmonar , Enfisema Pulmonar/inducido químicamente , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Pruebas de Función Respiratoria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus Sendai/genética , Transducción Genética
13.
Biochim Biophys Acta ; 1224(3): 333-41, 1994 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-7803487

RESUMEN

Addition of L-carnitine and some of its analogs to low-serum incubation medium of murine hepatocytic C2.8 cells prolonged maintenance of life and enhanced cell growth, as compared to controls. The drug acted synergistically with hepatocyte growth factor (HGF). Addition of L-carnitine to cells that had grown confluently in medium supplemented with HGF, significantly delayed the onset of cell death (apoptosis) initiated after HGF deprivation. Protection by L-carnitine was dose-dependent and stereospecific. Similar findings were obtained with three analogs of L-carnitine (i.e. isovaleryl-L-carnitine-HCl, isovaleryl-L-carnitine acid fumarate and butyryl L-carnitine taurine amide). In contrast, four different analogs (i.e. isovaleryl-L-carnitine-eptyl-ester-HCl, isovaleryl-L-carnitine-idroxy-butyric-HCl, L-threonyl-L-carnitine-HCl and L-paramethyl-cinnamoil-carnitine-HCl) were inactive. Although the mechanism of cytoprotection stimulated by L-carnitine remains unresolved, the data suggest that this compound serves as a co-factor that influences C2.8 cells to become less susceptible to damaging actions of noxious agents or conditions initiated after HGF withdrawal.


Asunto(s)
Apoptosis/efectos de los fármacos , Carnitina/farmacología , Factor de Crecimiento de Hepatocito/deficiencia , Hígado/efectos de los fármacos , Animales , Sangre , Carnitina/análogos & derivados , Línea Celular , Senescencia Celular , Medios de Cultivo , Hígado/citología , Ratones
14.
Circulation ; 110(18): 2896-902, 2004 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-15505094

RESUMEN

BACKGROUND: Pulmonary hypertension (PH) is a progressive disease characterized by raised pulmonary vascular resistance, thought to be curable only through lung transplantation. Pathophysiologically, proliferation of pulmonary artery smooth muscle cells triggers pulmonary arterial stenosis and/or regurgitation, especially in advanced PH. METHODS AND RESULTS: Using a rat model of advanced pulmonary vascular disease produced by injecting monocrotaline, we show that hepatocyte growth factor (HGF) targets pulmonary arterioles and blocks the progression of PH. In these rats, endogenous HGF production was dramatically downregulated during developing experimental PH, but c-Met/HGF receptor was abundant in the medial layers of pulmonary arterioles. HGF gene transfection 2 weeks after the monocrotaline injection resulted in milder medial hyperplasia in lung arterioles and inhibited overgrowth of pulmonary artery smooth muscle cells. Notably, exogenous HGF reduced lung expression levels of endothelin-1 and transforming growth factor-beta, which are critically involved in PH-linked fibrogenic events. Overall, medial wall thickening of pulmonary arteries was almost completely prevented by HGF, and the total collagen deposition in the lung decreased; both effects contributed to the suppression of pulmonary artery hypertension. CONCLUSIONS: Our results suggest that the loss of endogenous HGF may be a feature of the pathogenesis of PH and that HGF supplementation may minimize pathological lung conditions, even advanced PH.


Asunto(s)
Matriz Extracelular/metabolismo , Terapia Genética , Factor de Crecimiento de Hepatocito/fisiología , Hipertensión Pulmonar/terapia , Túnica Media/patología , Animales , Arteriolas/patología , Colágeno/análisis , Endotelina-1/biosíntesis , Endotelina-1/genética , Fibrosis , Factor de Crecimiento de Hepatocito/deficiencia , Factor de Crecimiento de Hepatocito/genética , Humanos , Hiperplasia , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Masculino , Monocrotalina/toxicidad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Proteínas Proto-Oncogénicas c-met/biosíntesis , Proteínas Proto-Oncogénicas c-met/genética , Ratas , Ratas Wistar , Transfección , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/genética
15.
Zebrafish ; 11(6): 542-50, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25353089

RESUMEN

Several genome-wide association studies have identified the genes encoding for macrophage-stimulating protein (MSP) and its receptor RON (Recepteur d'Origine Nantais) as possible susceptibility factors in inflammatory bowel disease. While it has been shown that the MSP-RON signaling pathway is involved in tissue injury responses, current mouse models for MSP and RON deficiency have not clearly demonstrated a role of MSP-RON signaling in the context of intestinal inflammation. In this study, we report that the recently identified zebrafish Msp mutant (msp(t34230)) develops spontaneous intestinal inflammation over time. From 14 to 28 weeks postfertilization Msp-deficient zebrafish show intestinal eosinophilia, increased intestinal expression of inflammatory marker mmp9, and activation of intestinal goblet cells. Moreover, these Msp mutant zebrafish are more susceptible toward ethanol-induced epithelial damage, which resulted in increased infiltration and proliferation of immune cells within the lamina propria and prolonged intestinal proinflammatory cytokine responses in some mutant fish. In light of the recent development of many tools to visualize, monitor, and genetically modify zebrafish, these Msp-deficient zebrafish will enable in-depth in vivo analysis of epithelial and macrophage-specific MSP-RON signaling in the context of intestinal inflammation.


Asunto(s)
Modelos Animales de Enfermedad , Factor de Crecimiento de Hepatocito/deficiencia , Inflamación/genética , Inflamación/patología , Mucosa Intestinal/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Transducción de Señal/genética , Pez Cebra , Animales , Citocinas , Cartilla de ADN/genética , Células Caliciformes/metabolismo , Factor de Crecimiento de Hepatocito/genética , Técnicas Histológicas , Inmunohistoquímica , Mucosa Intestinal/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Estadísticas no Paramétricas
16.
Oncotarget ; 5(14): 5547-58, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-24980820

RESUMEN

The Ron receptor is deregulated in a variety of cancers. Hepatocyte growth factor-like protein (HGFL) is the ligand for Ron and is constitutively secreted from hepatocytes into the circulation. While a few recent reports have emerged analyzing ectopic HGFL overexpression of in cancer cells, no studies have examined host-produced HGFL in tumorigenesis. To examine HGFL function in prostate cancer, the TRAMP mouse model, which is predisposed to develop prostate tumors, was utilized. Prostate tumors from TRAMP mice exhibit elevated levels of HGFL, which correlated with upregulation in human prostate cancer. To directly implicate HGFL in prostate tumorigenesis, TRAMP mice deficient in HGFL (HGFL-/-TRAMP+) were generated. HGFL-/- TRAMP+ mice developed significantly smaller prostate tumors compared to controls. Analysis of HGFL-/- tumors revealed reduced tumor vascularization. No differences in cancer cell proliferation were detected between HGFL-/- TRAMP+ and HGFL+/+ TRAMP+ mice. However, a significant increase in cancer cell death was detected in HGFL-/- TRAMP+ prostates which correlated with decreased pro-survival targets. In vitro analysis demonstrated robust STAT3 activation resulting in Bcl2-dependent survival following treatment of prostate cancer cells with HGFL. These data document a novel function for endogenous HGFL in prostate cancer by imparting a critical survival signal to tumor cells.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proliferación Celular/fisiología , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Factor de Crecimiento de Hepatocito/deficiencia , Factor de Crecimiento de Hepatocito/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética
17.
Mech Dev ; 133: 11-22, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25049204

RESUMEN

The Ron receptor tyrosine kinase regulates multiple cellular processes and is important during mammary gland development and tumor progression. Hepatocyte growth factor-like protein [HGFL] is the only known ligand for the Ron receptor and recent studies have identified major roles for HGFL during breast cancer metastasis. Understanding the functional importance HGFL during mammary gland development will provide significant insights onto its contribution during tumor development and metastasis. In this study, we assessed the role of HGFL during postnatal mammary gland development using mice that were either proficient [HGFL +/+] or deficient [HGFL-/-] for HGFL. Postnatal ductal morphology and stromal cell associations were analyzed at multiple time points through puberty until adulthood. HGFL deficiency resulted in several mammary gland developmental defects including smaller terminal end buds [TEBs], significantly fewer TEBs, and delayed ductal outgrowth during early puberty. Additionally, HGFL deficient animals exhibited significantly altered TEB epithelial cell turnover with decreased proliferation and increased apoptosis coupled with decreased TEB diameter. Macrophage recruitment to the TEBs was also significantly decreased in the HGFL-/- mice compared to controls. Moreover, the levels of STAT3 mRNA as well as the phosphorylation status of this protein were lower in the HGFL-/- mammary glands compared to controls. Taken together, our data provide the first evidence for HGFL as a positive regulator of mammary gland ductal morphogenesis by controlling overall epithelial cell turnover, macrophage recruitment, and STAT3 activation in the developing mammary gland. With a function in early mammary gland development, HGFL represents a potential target for the development of novel breast cancer therapies.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Factor de Crecimiento de Hepatocito/deficiencia , Factor de Crecimiento de Hepatocito/genética , Macrófagos/citología , Macrófagos/metabolismo , Glándulas Mamarias Animales/citología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
18.
PLoS One ; 8(3): e59836, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527275

RESUMEN

Hepatocyte growth factor (HGF) has been shown to be indispensable for liver regeneration because it serves as a main mitogenic stimulus driving hepatocytes toward proliferation. We hypothesized that ablating HGF in adult mice would have a negative effect on the ability of hepatocytes to regenerate. Deletion of the HGF gene was achieved by inducing systemic recombination in mice lacking exon 5 of HGF and carrying the Mx1-cre or Cre-ER(T) transgene. Analysis of liver genomic DNA from animals 10 days after treatment showed that a majority (70-80%) of alleles underwent cre-induced genetic recombination. Intriguingly, however, analysis by RT-PCR showed the continued presence of both unrecombined and recombined forms of HGF mRNA after treatment. Separation of liver cell populations into hepatocytes and non-parenchymal cells showed equal recombination of genomic HGF in both cell types. The presence of the unrecombined form of HGF mRNA persisted in the liver in significant amounts even after partial hepatectomy (PH), which correlated with insignificant changes in HGF protein and hepatocyte proliferation. The amount of HGF produced by stellate cells in culture was indirectly proportional to the concentration of HGF, suggesting that a decrease in HGF may induce de novo synthesis of HGF from cells with residual unrecombined alleles. Carbon tetrachloride (CCl4)-induced regeneration resulted in a substantial decrease in preexisting HGF mRNA and protein, and subsequent PH led to a delayed regenerative response. Thus, HGF mRNA persists in the liver even after genetic recombination affecting most cells; however, PH subsequent to CCl4 treatment is associated with a decrease in both HGF mRNA and protein and results in compromised liver regeneration, validating an important role of this mitogen in hepatic growth.


Asunto(s)
Factor de Crecimiento de Hepatocito/deficiencia , Factor de Crecimiento de Hepatocito/metabolismo , Regeneración Hepática/fisiología , Animales , Western Blotting , Tetracloruro de Carbono , Cartilla de ADN/genética , Hepatectomía , Células Estrelladas Hepáticas/metabolismo , Factor de Crecimiento de Hepatocito/genética , Inmunohistoquímica , Inmunoprecipitación , Regeneración Hepática/genética , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Recombinación Genética/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
PLoS One ; 8(9): e73312, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24023859

RESUMEN

As the main source of extracellular matrix proteins in tumor stroma, hepatic stellate cells (HSCs) have a great impact on biological behaviors of hepatocellular carcinoma (HCC). In the present study, we have investigated a mechanism whereby HSCs modulate the chemoresistance of hepatoma cells. We used human HSC line lx-2 and chemotherapeutic agent cisplatin to investigate their effects on human HCC cell line Hep3B. The results showed that cisplatin resistance in Hep3B cells was enhanced with LX-2 CM (cultured medium) exposure in vitro as well as co-injection with LX-2 cells in null mice. Meanwhile, in presence of LX-2 CM, Hep3B cells underwent epithelial to mesenchymal transition (EMT) and upregulation of cancer stem cell (CSC) -like properties. Besides, LX-2 cells synthesized and secreted hepatic growth factor (HGF) into the CM. HGF receptor tyrosine kinase mesenchymal-epithelial transition factor (Met) was activated in Hep3B cells after LX-2 CM exposure. The HGF level of LX-2 CM could be effectively reduced by using HGF neutralizing antibody. Furthermore, depletion of HGF in LX-2 CM abolished its effects on activation of Met as well as promotion of the EMT, CSC-like features and cisplatin resistance in Hep3B cells. Collectively, secreting HGF into tumor milieu, HSCs may decrease hepatoma cells sensitization to chemotherapeutic agents by promoting EMT and CSC-like features via HGF/Met signaling.


Asunto(s)
Carcinoma Hepatocelular/patología , Resistencia a Antineoplásicos , Células Estrelladas Hepáticas/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias Hepáticas/patología , Animales , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Cisplatino/farmacología , Medios de Cultivo/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Factor de Crecimiento de Hepatocito/deficiencia , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Fenotipo
20.
Nat Med ; 19(11): 1478-88, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24141421

RESUMEN

Here we show that Mst1, a proapoptotic kinase, impairs protein quality control mechanisms in the heart through inhibition of autophagy. Stress-induced activation of Mst1 in cardiomyocytes promoted accumulation of p62 and aggresome formation, accompanied by the disappearance of autophagosomes. Mst1 phosphorylated the Thr108 residue in the BH3 domain of Beclin1, which enhanced the interaction between Beclin1 and Bcl-2 and/or Bcl-xL, stabilized the Beclin1 homodimer, inhibited the phosphatidylinositide 3-kinase activity of the Atg14L-Beclin1-Vps34 complex and suppressed autophagy. Furthermore, Mst1-induced sequestration of Bcl-2 and Bcl-xL by Beclin1 allows Bax to become active, thereby stimulating apoptosis. Mst1 promoted cardiac dysfunction in mice subjected to myocardial infarction by inhibiting autophagy, associated with increased levels of Thr108-phosphorylated Beclin1. Moreover, dilated cardiomyopathy in humans was associated with increased levels of Thr108-phosphorylated Beclin1 and signs of autophagic suppression. These results suggest that Mst1 coordinately regulates autophagy and apoptosis by phosphorylating Beclin1 and consequently modulating a three-way interaction among Bcl-2 proteins, Beclin1 and Bax.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Factor de Crecimiento de Hepatocito/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Beclina-1 , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Fosfatidilinositol 3-Quinasas Clase III/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Femenino , Factor de Crecimiento de Hepatocito/deficiencia , Factor de Crecimiento de Hepatocito/genética , Humanos , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fosforilación , Multimerización de Proteína , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Ratas , Adulto Joven , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA