Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Pharmacol ; 188: 114574, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33887258

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Fator de Crescimento de Hepatócito/deficiência , Hiperglicemia/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Proto-Oncogênicas/deficiência , Sulfonamidas/uso terapêutico , Animais , Linhagem Celular , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/metabolismo , Relação Dose-Resposta a Droga , Fator de Crescimento de Hepatócito/genética , Hiperglicemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas/genética , Ratos , Sulfonamidas/farmacologia , Resultado do Tratamento
2.
Biochem Biophys Res Commun ; 489(1): 56-62, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28527887

RESUMO

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.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Fator de Crescimento de Hepatócito/imunologia , Hipergamaglobulinemia/imunologia , Interleucina-4/imunologia , Proteínas Proto-Oncogênicas/imunologia , Animais , Fator de Crescimento de Hepatócito/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas/deficiência
3.
Stem Cell Res ; 20: 42-49, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28257933

RESUMO

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.


Assuntos
Reprogramação Celular , Fator de Crescimento de Hepatócito/genética , Células-Tronco Pluripotentes Induzidas/citologia , Proteínas Proto-Oncogênicas/genética , Animais , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Fator de Crescimento de Hepatócito/deficiência , Via de Sinalização Hippo , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
J Mol Cell Cardiol ; 98: 108-16, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27496379

RESUMO

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.


Assuntos
Apoptose/genética , Aterosclerose/etiologia , Aterosclerose/metabolismo , Autofagia/genética , Fator de Crescimento de Hepatócito/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas Proto-Oncogênicas/genética , Animais , Apolipoproteínas E/deficiência , Aterosclerose/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Genótipo , Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/metabolismo , Lipoproteínas LDL/administração & dosagem , Lipoproteínas LDL/metabolismo , Camundongos , Camundongos Knockout , Placa Aterosclerótica/genética , Placa Aterosclerótica/patologia , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo
6.
Zebrafish ; 11(6): 542-50, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25353089

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Fator de Crescimento de Hepatócito/deficiência , Inflamação/genética , Inflamação/patologia , Mucosa Intestinal/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Transdução de Sinais/genética , Peixe-Zebra , Animais , Citocinas , Primers do DNA/genética , Células Caliciformes/metabolismo , Fator de Crescimento de Hepatócito/genética , Técnicas Histológicas , Imuno-Histoquímica , Mucosa Intestinal/patologia , Metaloproteinase 9 da Matriz/metabolismo , Mutação/genética , Proteínas Proto-Oncogênicas/genética , Reação em Cadeia da Polimerase em Tempo Real , Estatísticas não Paramétricas
7.
Mech Dev ; 133: 11-22, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25049204

RESUMO

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.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/genética , Macrófagos/citologia , Macrófagos/metabolismo , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
8.
Oncotarget ; 5(14): 5547-58, 2014 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-24980820

RESUMO

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.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas/metabolismo , Animais , Proliferação de Células/fisiologia , Transformação Celular Neoplásica , Modelos Animais de Doenças , Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética
9.
Hepatology ; 59(4): 1435-47, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24242874

RESUMO

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.


Assuntos
Antígeno CD11b/metabolismo , Carcinoma Hepatocelular/metabolismo , Quimiocina CXCL12/metabolismo , Cirrose Hepática/metabolismo , Fígado/metabolismo , Células Mieloides/patologia , Niacinamida/análogos & derivados , Compostos de Fenilureia/farmacologia , Receptores CXCR4/metabolismo , Receptores de Quimiocinas/metabolismo , Animais , Tetracloreto de Carbono/efeitos adversos , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular , Modelos Animais de Doenças , Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/genética , Fígado/efeitos dos fármacos , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Células Mieloides/metabolismo , Niacinamida/farmacologia , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sorafenibe
10.
Nat Med ; 19(11): 1478-88, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24141421

RESUMO

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.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/fisiologia , Fator de Crescimento de Hepatócito/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Adulto , Sequência de Aminoácidos , Animais , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/genética , Proteína Beclina-1 , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Classe III de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Feminino , Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/genética , Humanos , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Modelos Moleculares , Dados de Sequência Molecular , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fosforilação , Multimerização Proteica , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Ratos , Adulto Jovem , Proteína X Associada a bcl-2/metabolismo
11.
PLoS One ; 8(9): e73312, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24023859

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/patologia , Resistencia a Medicamentos Antineoplásicos , Células Estreladas do Fígado/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Neoplasias Hepáticas/patologia , Animais , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Cisplatino/farmacologia , Meios de Cultura/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/patologia , Fator de Crescimento de Hepatócito/deficiência , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Fenótipo
12.
Cell Rep ; 3(5): 1663-77, 2013 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-23684612

RESUMO

The transcriptional coactivator Yes-associated protein (YAP) plays an important role in organ-size control and tumorigenesis. However, how Yap gene expression is regulated remains unknown. This study shows that the Ets family member GABP binds to the Yap promoter and activates YAP transcription. The depletion of GABP downregulates YAP, resulting in a G1/S cell-cycle block and increased cell death, both of which are substantially rescued by reconstituting YAP. GABP can be inactivated by oxidative mechanisms, and acetaminophen-induced glutathione depletion inhibits GABP transcriptional activity and depletes YAP. In contrast, activating YAP by deleting Mst1/Mst2 strongly protects against acetaminophen-induced liver injury. Similar to its effects on YAP, Hippo signaling inhibits GABP transcriptional activity through several mechanisms. In human liver cancers, enhanced YAP expression is correlated with increased nuclear expression of GABP. Therefore, we conclude that GABP is an activator of Yap gene expression and a potential therapeutic target for cancers driven by YAP.


Assuntos
Antioxidantes/farmacologia , Fator de Transcrição de Proteínas de Ligação GA/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Acetaminofen/farmacologia , Animais , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fator de Transcrição de Proteínas de Ligação GA/antagonistas & inibidores , Fator de Transcrição de Proteínas de Ligação GA/genética , Células HEK293 , Células HeLa , Células Hep G2 , Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Serina-Treonina Quinase 3 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
PLoS One ; 8(3): e59836, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23527275

RESUMO

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.


Assuntos
Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/metabolismo , Regeneração Hepática/fisiologia , Animais , Western Blotting , Tetracloreto de Carbono , Primers do DNA/genética , Hepatectomia , Células Estreladas do Fígado/metabolismo , Fator de Crescimento de Hepatócito/genética , Imuno-Histoquímica , Imunoprecipitação , Regeneração Hepática/genética , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Recombinação Genética/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Mol Cell Biol ; 32(24): 5116-28, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23071096

RESUMO

The mammalian pancreas is required for normal metabolism, with defects in this vital organ commonly observed in cancer and diabetes. Development must therefore be tightly controlled in order to produce a pancreas of correct size, cell type composition, and physiologic function. Through negative regulation of Yap-dependent proliferation, the Hippo kinase cascade is a critical regulator of organ growth. To investigate the role of Hippo signaling in pancreas biology, we deleted Hippo pathway components in the developing mouse pancreas. Unexpectedly, the pancreas from Hippo-deficient offspring was reduced in size, with defects evident throughout the organ. Increases in the dephosphorylated nuclear form of Yap are apparent throughout the exocrine compartment and correlate with increases in levels of cell proliferation. However, the mutant exocrine tissue displays extensive disorganization leading to pancreatitis-like autodigestion. Interestingly, our results suggest that Hippo signaling does not directly regulate the pancreas endocrine compartment as Yap expression is lost following endocrine specification through a Hippo-independent mechanism. Altogether, our results demonstrate that Hippo signaling plays a crucial role in pancreas development and provide novel routes to a better understanding of pathological conditions that affect this organ.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pâncreas/embriologia , Pâncreas/metabolismo , Fosfoproteínas/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Proliferação de Células , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Knockout , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Gravidez , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Estabilidade Proteica , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Serina-Treonina Quinase 3 , Transdução de Sinais , Proteínas de Sinalização YAP
15.
PLoS One ; 7(8): e44010, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952854

RESUMO

Epidermodysplasia verruciformis (EV) is characterized by persistent cutaneous lesions caused by a specific group of related human papillomavirus genotypes (EV-HPVs) in otherwise healthy individuals. Autosomal recessive (AR) EVER1 and EVER2 deficiencies account for two thirds of known cases of EV. AR RHOH deficiency has recently been described in two siblings with EV-HPV infections as well as other infectious and tumoral manifestations. We report here the whole-exome based discovery of AR MST1 deficiency in a 19-year-old patient with a T-cell deficiency associated with EV-HPV, bacterial and fungal infections. MST1 deficiency has recently been described in seven patients from three unrelated kindreds with profound T-cell deficiency and various viral and bacterial infections. The patient was also homozygous for a rare ERCC3 variation. Our findings broaden the clinical range of infections seen in MST1 deficiency and provide a new genetic etiology of susceptibility to EV-HPV infections. Together with the recent discovery of RHOH deficiency, they suggest that T cells are involved in the control of EV-HPVs, at least in some individuals.


Assuntos
Predisposição Genética para Doença , Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/genética , Padrões de Herança/genética , Papillomaviridae/fisiologia , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Adolescente , Sequência de Aminoácidos , Antígenos Virais/imunologia , Sequência de Bases , Proliferação de Células/efeitos dos fármacos , Criança , Códon sem Sentido/genética , Epidermodisplasia Verruciforme/genética , Epidermodisplasia Verruciforme/imunologia , Epidermodisplasia Verruciforme/microbiologia , Epidermodisplasia Verruciforme/virologia , Exoma/genética , Fator de Crescimento de Hepatócito/química , Homozigoto , Humanos , Imunofenotipagem , Lactente , Masculino , Mitógenos/farmacologia , Dados de Sequência Molecular , Papillomaviridae/efeitos dos fármacos , Proteínas Proto-Oncogênicas/química , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Adulto Jovem
16.
Hepatology ; 55(4): 1215-26, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22095660

RESUMO

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.


Assuntos
Fator de Crescimento de Hepatócito/fisiologia , Regeneração Hepática/fisiologia , Proteínas Proto-Oncogênicas c-met/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/terapia , Quimiocina CXCL12/metabolismo , Modelos Animais de Doenças , Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/genética , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-met/deficiência , Proteínas Proto-Oncogênicas c-met/genética , Piridinas/efeitos adversos , Transplante de Células-Tronco , Células-Tronco/citologia
17.
EMBO J ; 30(12): 2465-76, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21572393

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Transporte/fisiologia , Fator de Crescimento de Hepatócito/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Microglia/citologia , Microglia/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais , Animais , Apoptose/genética , Células COS , Proteínas de Transporte/genética , Morte Celular/genética , Sobrevivência Celular/genética , Células Cultivadas , Chlorocebus aethiops , Proteínas Correpressoras , Fibroblastos/citologia , Fibroblastos/fisiologia , Células HEK293 , Células HeLa , Fator de Crescimento de Hepatócito/deficiência , Fator de Crescimento de Hepatócito/genética , Humanos , Mediadores da Inflamação , Interferon gama/administração & dosagem , Interferon gama/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout , Chaperonas Moleculares , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética
18.
Mol Cell ; 38(4): 512-23, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20513427

RESUMO

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.


Assuntos
Apoptose , Fator de Crescimento de Hepatócito/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Crescimento de Hepatócito/deficiência , Humanos , Camundongos , Fosforilação , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
19.
J Immunol ; 183(6): 3865-72, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19692642

RESUMO

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.


Assuntos
Quimiotaxia , Fator de Crescimento de Hepatócito/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Timo/citologia , Animais , Quimiocinas , Fator de Crescimento de Hepatócito/deficiência , Linfonodos , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/deficiência , Linfócitos T/citologia
20.
Front Biosci ; 13: 7072-86, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18508717

RESUMO

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.


Assuntos
Terapia Genética/métodos , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/uso terapêutico , Falência Renal Crônica/tratamento farmacológico , Falência Renal Crônica/patologia , Animais , Fibrose , Glomerulosclerose Segmentar e Focal/fisiopatologia , Glomerulosclerose Segmentar e Focal/prevenção & controle , Fator de Crescimento de Hepatócito/deficiência , Humanos , Falência Renal Crônica/genética , Túbulos Renais/patologia , Túbulos Renais/fisiologia , Camundongos , Fator de Crescimento Derivado de Plaquetas/fisiologia , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/fisiologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...