Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hepatology ; 61(3): 930-41, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25251599

RESUMO

UNLABELLED: In hepatocellular carcinoma (HCC), intrahepatic metastasis frequently correlates with epithelial to mesenchymal transition (EMT) of malignant hepatocytes. Several mechanisms have been identified to be essentially involved in hepatocellular EMT, among them transforming growth factor (TGF)-ß signaling. Here we show the up-regulation and activation of the receptor tyrosine kinase Axl in EMT-transformed hepatoma cells. Knockdown of Axl expression resulted in abrogation of invasive and transendothelial migratory abilities of mesenchymal HCC cells in vitro and Axl overexpression-induced metastatic colonization of epithelial hepatoma cells in vivo. Importantly, Axl knockdown severely impaired resistance to TGF-ß-mediated growth inhibition. Analysis of the Axl interactome revealed binding of Axl to 14-3-3ζ, which is essentially required for Axl-mediated cell invasion, transendothelial migration, and resistance against TGF-ß. Axl/14-3-3ζ signaling caused phosphorylation of Smad3 linker region (Smad3L) at Ser213, resulting in the up-regulation of tumor-progressive TGF-ß target genes such as PAI1, MMP9, and Snail as well as augmented TGF-ß1 secretion in mesenchymal HCC cells. Accordingly, high Axl expression in HCC patient samples correlated with elevated vessel invasion of HCC cells, higher risk of tumor recurrence after liver transplantation, strong phosphorylation of Smad3L, and lower survival. In addition, elevated expression of both Axl and 14-3-3ζ showed strongly reduced survival of HCC patients. CONCLUSION: Our data suggest that Axl/14-3-3ζ signaling is central for TGF-ß-mediated HCC progression and a promising target for HCC therapy.


Assuntos
Comunicação Autócrina , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Transdução de Sinais , Fator de Crescimento Transformador beta/fisiologia , Proteínas 14-3-3/fisiologia , Carcinoma Hepatocelular/mortalidade , Movimento Celular , Transição Epitelial-Mesenquimal , Feminino , Humanos , Neoplasias Hepáticas/mortalidade , Masculino , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/fisiologia , Receptor Tirosina Quinase Axl
2.
Mutat Res ; 728(1-2): 23-34, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21605699

RESUMO

Metastasis is the leading cause of cancer mortality. The metastatic cascade represents a multi-step process which includes local tumor cell invasion, entry into the vasculature followed by the exit of carcinoma cells from the circulation and colonization at the distal sites. At the earliest stage of successful cancer cell dissemination, the primary cancer adapts the secondary site of tumor colonization involving the tumor-stroma crosstalk. The migration and plasticity of cancer cells as well as the surrounding environment such as stromal and endothelial cells are mandatory. Consequently, the mechanisms of cell movement are of utmost relevance for targeted intervention of which three different types have been reported. Tumor cells can migrate either collectively, in a mesenchymal or in an amoeboid type of movement and intravasate the blood or lymph vasculature. Intravasation by the interaction of tumor cells with the vascular endothelium is mechanistically poorly understood. Changes in the epithelial plasticity enable carcinoma cells to switch between these types of motility. The types of migration may change depending on the intervention thereby increasing the velocity and aggressiveness of invading cancer cells. Interference with collective or mesenchymal cell invasion by targeting integrin expression or metalloproteinase activity, respectively, resulted in an amoeboid cell phenotype as the ultimate exit strategy of cancer cells. There are little mechanistic details reported in vivo showing that the amoeboid behavior can be either reversed or efficiently inhibited. Future concepts of metastasis intervention must simultaneously address the collective, mesenchymal and amoeboid mechanisms of cell invasion in order to advance in anti-metastatic strategies as these different types of movement can coexist and cooperate. Beyond the targeting of cell movements, the adhesion of cancer cells to the stroma in heterotypic circulating tumor cell emboli is of paramount relevance for anti-metastatic therapy.


Assuntos
Invasividade Neoplásica , Metástase Neoplásica/fisiopatologia , Migração Transendotelial e Transepitelial , Movimento Celular , Humanos , Mesoderma/citologia , Modelos Biológicos , Microambiente Tumoral
3.
Hepatology ; 54(1): 164-72, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21452288

RESUMO

UNLABELLED: Signal transducer and activator of transcription 3 (Stat3) is activated in a variety of malignancies, including hepatocellular carcinoma (HCC). Activation of Ras occurs frequently at advanced stages of HCC by aberrant signaling through growth factor receptors or inactivation of effectors negatively regulating Ras signaling. Here, we addressed the role of Stat3 in Ras-dependent HCC progression in the presence and absence of p19(ARF) /p14(ARF) . We show that constitutive active (ca) Stat3 is tumor suppressive in Ras-transformed p19(ARF-/-) hepatocytes, whereas the expression of Stat3 lacking Tyr(705) phosphorylation (U-Stat3) enhances tumor formation. Accordingly, Ras-transformed Stat3(Δhc) /p19(ARF-/-) hepatocytes (lacking Stat3 and p19(ARF) ) showed increased tumor growth, compared to those expressing Stat3, demonstrating a tumor-suppressor activity of Stat3 in cells lacking p19(ARF) . Notably, endogenous expression of p19(ARF) in Ras-transformed hepatocytes conveyed oncogenic Stat3 functions, resulting in augmented or reduced HCC progression after the expression of caStat3 or U-Stat3, respectively. In accord with these data, the knockdown of p14(ARF) (the human homolog of p19(ARF) ) in Hep3B cells was associated with reduced pY-Stat3 levels during tumor growth to circumvent the tumor-suppressive effect of Stat3. Inhibition of Janus kinases (Jaks) revealed that Jak causes pY-Stat3 activation independently of p14(ARF) levels, indicating that p14(ARF) controls the oncogenic function of pY-Stat3 downstream of Jak. CONCLUSION: These data show evidence that p19(ARF) /p14(ARF) determines the pro- or anti-oncogenic activity of U-Stat3 and pY-Stat3 in Ras-dependent HCC progression.


Assuntos
Carcinoma Hepatocelular/fisiopatologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Neoplasias Hepáticas/fisiopatologia , Fator de Transcrição STAT3/fisiologia , Animais , Carcinoma Hepatocelular/patologia , Proliferação de Células , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Inibidor p16 de Quinase Dependente de Ciclina/genética , Modelos Animais de Doenças , Progressão da Doença , Hepatócitos/patologia , Janus Quinases/fisiologia , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Knockout , Transdução de Sinais/fisiologia
4.
Mol Cancer Ther ; 10(5): 850-60, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21364009

RESUMO

The epithelial to mesenchymal transition (EMT) of malignant hepatocytes is a crucial event in hepatocellular carcinoma (HCC) progression and recurrence. We aimed to establish a human model of EMT to examine drug efficacy and specificity in HCC progression. Human HCC cell populations were characterized by immunofluorescence analysis, migration and invasion assays, array comparative genomic hybridization, whole-genome expression profiling, and promoter methylation. Therapeutic agents clinically used against HCC were examined for efficacy by determination of IC(50) values. We show that liver cancer cell lines exhibited either an epithelial or mesenchymal phenotype of which the latter showed strong migratory and invasive abilities in vitro. The common cellular origin of both cell types indicated that mesenchymal HCC cells have been derived from epithelial hepatocytes through EMT in the HCC patient. Drug exposure of mesenchymal HCC cells showed higher resistance to the targeted therapeutic agents sorafenib and erlotinib as compared to epithelial HCC cells, which were slightly more resistant to cytostatic drugs. Most remarkably, combined treatment with doxorubicin and sorafenib caused increased susceptibility of both HCC cell types resulting in enhanced drug efficacy. Taken together, this EMT model of human HCC allows the identification of molecular mechanisms and the assessment of therapeutic drug efficacy during liver cancer progression in preclinical studies.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Hepatocelular/fisiopatologia , Transformação Celular Neoplásica/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Células Epiteliais/patologia , Neoplasias Hepáticas/fisiopatologia , Mesoderma/patologia , Linhagem Celular Tumoral , Movimento Celular , Transformação Celular Neoplásica/metabolismo , Ilhas de CpG/genética , Metilação de DNA/genética , Progressão da Doença , Células Epiteliais/efeitos dos fármacos , Perfilação da Expressão Gênica , Humanos , Masculino , Mesoderma/efeitos dos fármacos , Repetições de Microssatélites/genética , Pessoa de Meia-Idade , Osteonectina/genética , Osteonectina/metabolismo , Fenótipo
5.
World J Hepatol ; 2(1): 1-7, 2010 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21160950

RESUMO

Studying physiological and pathophysiological mechanisms in the liver on a molecular basis is a challenging task. During two dimensional (2D) culture conditions hepatocytes dedifferentiate rapidly by losing metabolic functions and structural integrity. Hence, inappropriate 2D hepatocellular models hamper studies on the xenobiotic metabolism of the liver which strongly influences drug potency. Also, the lack of effective therapies against hepatocellular carcinoma shows the urgent need for robust models to investigate liver functions in a defined hepatic microenvironment. Here, we summarize and discuss three-dimensional cultures of hepatocytes, herein referred to as hepatospheres, which provide versatile tools to investigate hepatic metabolism, stemness and cancer development.

6.
Hepatology ; 51(4): 1319-26, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20162728

RESUMO

UNLABELLED: Growth hormone (GH) resistance and low serum levels of insulinlike growth factor 1 (IGF-1) are common features in human liver fibrosis and cirrhosis. Signal transducer and activator of transcription 5 (STAT5) controls several vital functions in the liver, including GH-mediated transcription of IGF-1. To investigate the role of STAT5 in liver fibrogenesis, we specifically deleted the Stat5a/b locus both in hepatocytes and cholangiocytes in the multidrug resistance gene 2 knockout (Mdr2(-/-)) mouse model of cholestasis. Double knockout mice develop an early and severe liver fibrosis phenotype, accompanied by perturbed expression of key regulators of bile acid homeostasis. Deletion of Stat5 resulted in GH resistance, and IGF-1 levels in serum were undetectable. We could observe reduced expression of important hepatoprotective genes, such as epidermal growth factor receptor (Egfr), hepatocyte nuclear factor 6 (Hnf6), prolactin receptor (Prlr), and leukemia inhibitory factor receptor (Lifr) as well as increased numbers of apoptotic hepatocytes. CONCLUSION: Our data suggest that loss of STAT5 sensitizes hepatocytes to bile acid-induced damage and apoptosis caused by disruption of GH-induced transcription of Igf-1 and down-regulation of hepatoprotective genes. These findings could contribute to the understanding of liver fibrosis and future treatment strategies for liver fibrosis.


Assuntos
Colestase/complicações , Hormônio do Crescimento/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Cirrose Hepática Experimental/etiologia , Fator de Transcrição STAT5/fisiologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/fisiologia , Animais , Apoptose , Modelos Animais de Doenças , Receptores ErbB/genética , Fator 6 Nuclear de Hepatócito/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Transdução de Sinais , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
7.
Am J Pathol ; 176(1): 472-81, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20008139

RESUMO

Transforming growth factor-beta cooperates with oncogenic Ras to activate nuclear beta-catenin during the epithelial to mesenchymal transition of hepatocytes, a process relevant in the progression of hepatocellular carcinoma (HCC). In this study we investigated the role of beta-catenin in the differentiation of murine, oncogene-targeted hepatocytes and in 133 human HCC patients scheduled for orthotopic liver transplantation. Transforming growth factor-beta caused dissociation of plasma membrane E-cadherin/beta-catenin complexes and accumulation of nuclear beta-catenin in Ras-transformed, but otherwise normal hepatocytes in p19(ARF)-/- mice. Both processes were inhibited by Smad7-mediated disruption of transforming growth factor-beta signaling. Overexpression of constitutively active beta-catenin resulted in high levels of CK19 and M2-PK, whereas ablation of beta-catenin by axin overexpression caused strong expression of CK8 and CK18. Therefore, nuclear beta-catenin resulted in dedifferentiation of neoplastic hepatocytes to immature progenitor cells, whereas loss of nuclear beta-catenin led to a differentiated HCC phenotype. Poorly differentiated human HCC showed cytoplasmic redistribution or even loss of E-cadherin, suggesting epithelial to mesenchymal transition. Analysis of 133 HCC patient samples revealed that 58.6% of human HCC exhibited strong nuclear beta-catenin accumulation, which correlated with clinical features such as vascular invasion and recurrence of disease after orthotopic liver transplantation. These data suggest that activation of beta-catenin signaling causes dedifferentiation to malignant, immature hepatocyte progenitors and facilitates recurrence of human HCC after orthotopic liver transplantation.


Assuntos
Carcinoma Hepatocelular/patologia , Núcleo Celular/metabolismo , Neoplasias Hepáticas/patologia , Fígado/patologia , Recidiva Local de Neoplasia/metabolismo , Células-Tronco/patologia , beta Catenina/metabolismo , Animais , Caderinas/metabolismo , Carcinoma Hepatocelular/irrigação sanguínea , Carcinoma Hepatocelular/metabolismo , Diferenciação Celular , Membrana Celular/metabolismo , Epitélio/metabolismo , Epitélio/patologia , Feminino , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/metabolismo , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/metabolismo , Transplante de Fígado , Masculino , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Neovascularização Patológica/complicações , Fenótipo , Transporte Proteico , Transdução de Sinais , Proteína Smad7/metabolismo , Fator de Crescimento Transformador beta/metabolismo
8.
Future Oncol ; 5(8): 1169-79, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19852728

RESUMO

The transition of epithelial cells to a mesenchymal phenotype is of paramount relevance for embryonic development and adult wound healing. During the past decade, the epithelial-mesenchymal transition (EMT) has been increasingly recognized to occur during the progression of various carcinomas such as hepatocellular carcinoma (HCC). Here, we focus on EMT in both experimental liver models and human HCC, emphasizing the underlying molecular mechanisms which show partial recurrence of embryonic programs such as TGF-beta and Wnt/ beta-catenin signaling, including collaboration with hepatitis viruses. We further discuss the differentiation repertoire of malignant hepatocytes with respect to the potential acquisition of stemness, and the involvement of the mesenchymal to epithelial transition, the reversal of EMT, in cancer dissemination and metastatic colonization. The strong evidence for EMT in HCC patients demands novel strategies in pathological assessments and therapeutic concepts to efficiently combat HCC progression.


Assuntos
Carcinoma Hepatocelular/patologia , Diferenciação Celular/fisiologia , Transformação Celular Neoplásica/patologia , Epitélio/patologia , Neoplasias Hepáticas/patologia , Animais , Carcinoma Hepatocelular/genética , Desdiferenciação Celular/fisiologia , Transformação Celular Neoplásica/genética , Humanos , Neoplasias Hepáticas/genética , Mesoderma/patologia , Metaplasia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...