Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Free Radic Biol Med ; 141: 205-219, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31207288

RESUMO

The transcriptional regulator YAP plays an important role in cancer progression and is negatively controlled by the Hippo pathway. YAP is frequently overexpressed in human cancers, including bladder cancer. Interestingly, YAP expression and activity can be inhibited by pro-oxidant conditions; moreover, YAP itself can also affect the cellular redox status through multiple mechanisms. 4-Hydroxynonenal (HNE), the most intensively studied end product of lipid peroxidation, is a pro-oxidant agent able to deplete GSH and has an anti-tumoral effect by affecting multiple signal pathways, including the down-regulation of oncogene expressions. These observations prompted us to investigate the effect of HNE on YAP expression and activity. We demonstrated that HNE inhibited YAP expression and its target genes in bladder cancer cells through a redox-dependent mechanism. Moreover, the YAP down-regulation was accompanied by an inhibition of proliferation, migration, invasion, and angiogenesis, as well as by an accumulation of cells in the G2/M phase of cell cycle and by an induction of apoptosis. We also established the YAP role in inhibiting cell viability and inducing apoptosis in HNE-treated cells by using an expression vector for YAP. Furthermore, we identified a post-translational mechanism for the HNE-induced YAP expression inhibition, involving an increase of YAP phosphorylation and ubiquitination, leading to proteasomal degradation. Our data established that HNE can post-translationally down-regulate YAP through a redox-dependent mechanism and that this modulation can contribute to determining the specific anti-cancer effects of HNE.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aldeídos/farmacologia , Regulação Neoplásica da Expressão Gênica , Processamento de Proteína Pós-Traducional , Fatores de Transcrição/metabolismo , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/metabolismo , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Células Endoteliais da Veia Umbilical Humana , Humanos , Invasividade Neoplásica , Neovascularização Patológica , Oncogenes , Oxirredução , Fosfoproteínas/metabolismo , Transdução de Sinais , Neoplasias da Bexiga Urinária/genética , Proteínas de Sinalização YAP
2.
Free Radic Biol Med ; 115: 447-457, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29248722

RESUMO

Redox adaptation plays an important role in cancer cells drug resistance. The antioxidant response is principally mediated by the transcription factor Nrf2, that induces the transcriptional activation of several genes involved in GSH synthesis, chemoresistance, and cytoprotection. YAP is emerging as a key mediator of chemoresistance in a variety of cancers, but its role in controlling the antioxidant status of the cells is yet elusive. Here, we show that impairing YAP protein expression reduced GSH content and Nrf2 protein and mRNA expression in bladder cancer cells. Moreover, in YAP knocked down cells the expression of FOXM1, a transcription factor involved in Nrf2 transcription, was down-regulated and the silencing of FOXM1 reduced Nrf2 expression. On the other hand, the silencing of Nrf2, as well as the depletion of GSH by BSO treatment, inhibited YAP expression, suggesting that cross-talk exists between YAP and Nrf2 proteins. Importantly, we found that silencing either YAP or Nrf2 enhanced sensitivity of bladder cancer cells to cytotoxic agents and reduced their migration. Furthermore, the inhibition of both YAP and Nrf2 expressions significantly increased cytotoxic drug sensitivity and synergistically reduced the migration of chemoresistant bladder cancer cells. These findings provide a rationale for targeting these transcriptional regulators in patients with chemoresistant bladder cancer, expressing high YAP and bearing a proficient antioxidant system.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antineoplásicos/farmacologia , Cisplatino/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Fosfoproteínas/metabolismo , Neoplasias da Bexiga Urinária/tratamento farmacológico , Proteínas Adaptadoras de Transdução de Sinal/genética , Antioxidantes/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Fator 2 Relacionado a NF-E2/genética , Oxirredução , Estresse Oxidativo , Fosfoproteínas/genética , RNA Interferente Pequeno/genética , Receptor Cross-Talk , Fatores de Transcrição , Neoplasias da Bexiga Urinária/genética , Proteínas de Sinalização YAP
3.
J Pathol ; 226(4): 588-97, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21959987

RESUMO

Liver fibrogenesis is sustained by pro-fibrogenic myofibroblast-like cells (MFs), mainly originating from activated hepatic stellate cells (HSC/MFs) or portal (myo)fibroblasts, and is favoured by hypoxia-dependent angiogenesis. Human HSC/MFs were reported to express vascular-endothelial growth factor (VEGF) and VEGF-receptor type 2 and to migrate under hypoxic conditions. This study was designed to investigate early and delayed signalling mechanisms involved in hypoxia-induced migration of human HSC/MFs. Signal transduction pathways and intracellular generation of reactive oxygen species (ROS) were evaluated by integrating morphological, cell, and molecular biology techniques. Non-oriented and oriented migration were evaluated by using wound healing assay and the modified Boyden's chamber assay, respectively. The data indicate that hypoxia-induced migration of HSC/MFs is a biphasic process characterized by the following sequence of events: (a) an early (15 min) and mitochondria-related increased generation of intracellular ROS which (b) was sufficient to switch on activation of ERK1/2 and JNK1/2 that were responsible for the early phase of oriented migration; (c) a delayed and HIF-1α-dependent increase in VEGF expression (facilitated by ROS) and its progressive, time-dependent release in the extracellular medium that (d) was mainly responsible for sustained migration of HSC/MFs. Finally, immunohistochemistry performed on HCV-related fibrotic/cirrhotic livers revealed HIF-2α and haem-oxygenase-1 positivity in hepatocytes and α-SMA-positive MFs, indicating that MFs were likely to be exposed in vivo to both hypoxia and oxidative stress. In conclusion, hypoxia-induced migration of HSC/MFs involves an early, mitochondrial-dependent ROS-mediated activation of ERK and JNK, followed by a delayed- and HIF-1α-dependent up-regulation and release of VEGF.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Movimento Celular/fisiologia , Células Estreladas do Fígado/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Hipóxia Celular , Células Cultivadas , Cultura em Câmaras de Difusão , Inativação Gênica , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Células Estreladas do Fígado/citologia , Hepatite C/metabolismo , Hepatite C/patologia , Hepatite C/virologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Mitocôndrias/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização/fisiologia
4.
Amino Acids ; 42(2-3): 783-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21901470

RESUMO

Epithelial-to-mesenchymal transition and mesenchymal-to-epithelial transition are biologic processes responsible for conversion of epithelial cells into a mesenchymal phenotype or viceversa, respectively. They occur during embryo- and foetal-development and, in adult organisms, are involved in wound healing, in the genesis and progression of organ fibrosis as well as in the invasiveness of epithelial cancer cells. The key event of epithelial-to-mesenchymal transition is the loss of E-cadherin expression due to repressor activity of the transcriptional factor Snai1. Intracellular Snai1 levels are controlled through translational and post-translational events such as phosphorylation and de-phosphorylation, potentially modulated by polyamine content. Epithelial MDCK cells exposed to TGF-ß(1) acquired a fibroblastoid phenotype and expressed mesenchymal markers. These changes were emphasized in cells that were also exposed to DFMO in order to decrease the intracellular levels of polyamines. Addition of exogenous polyamines almost completely abolished the combined action of DFMO and TGF-ß(1) and rapidly reverted to epithelial phenotype MDCK cells previously undergone to mesenchymal phenotype. Nuclear extracts of cells treated with DFMO + TGF-ß(1) revealed the presence of Snai1 immunopositive bands in a range of molecular weight between 55 and 72 kDa, with additional positive bands detected at MW greater than 170 kDa. Same bands resulted positive to anti-Sumo 2/3 antibody, suggesting that an intracellular low level of polyamines favours Snai1 nuclear accumulation under the form of polysumoylated proteins.


Assuntos
Poliaminas Biogênicas/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Animais , Western Blotting , Linhagem Celular , Cães , Humanos , Fosforilação
5.
Amino Acids ; 42(2-3): 769-74, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21901471

RESUMO

During our study of the cytostatic effect of agmatine, we were able to isolate an agmatine resistant clone from a parental hepatoma cell line, HTC. These cells, called Agres, had slower growth rate than the parental cells when cultured in normal medium. The modification in polyamine content induced by agmatine was much lower in these cells and ornithine decarboxylase, S-adenosylmethionine decarboxylase and spermidine/spermine acetyltransferase activities were much less affected. By investigating the mechanism responsible for these modifications, it was shown that agmatine and polyamines were not taken up by Agres cells. Their resistance to the antiproliferative effects of agmatine may thus arise from a lack of the polyamine transport system. Moreover, Agres cells were able to take up both glutamic acid and arginine at a rate significantly higher than that detected for HTC cells, most likely to provide components for compensatory increase of PA synthesis. These results emphasize the importance of polyamine transport for cell growth.


Assuntos
Agmatina/farmacologia , Neoplasias Hepáticas Experimentais/patologia , Animais , Western Blotting , Linhagem Celular Tumoral , Imunofluorescência , Ratos
6.
Int J Cancer ; 130(3): 694-704, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21400508

RESUMO

Anaplastic thyroid carcinoma (ATC) has a rapidly fatal clinical course, being resistant to multimodal treatments. Microtubules, α/ß tubulin heterodimers, are crucial in cell signaling, division and mitosis and are among the most successful targets for anticancer therapy. Panobinostat (LBH589) is a potent deacetylase inhibitor acting both on histones and nonhistonic proteins, including α-tubulin. In vitro LBH589, evaluated in three ATC cell lines (BHT-101, CAL-62 and 8305C), resulted in impairment of cell viability, inhibition of colony formation, cell cycle arrest and apoptosis induction. Mechanistically, we showed that LBH589 not only affected the expression of p21 and cyclin D1, but markedly determined microtubule stabilization as evidenced by tubulin acetylation and increased tubulin polymerization. In a SCID xenograft model implanted with CAL-62 cells, the cytotoxic properties of LBH589 were confirmed. The drug at the dose of 20 mg/kg significantly impaired tumor growth (final tumor volume 2.5-fold smaller than in untreated animals); at this dose, no relevant side effects were observed. In tumors of treated animals, a significant reduction of Ki67, which was negatively correlated with tubulin acetylation, was observed. Moreover, acetyl-tubulin levels negatively correlated with tumor volume at sacrifice, reinforcing the opinion that tubulin acetylation has a role in the inhibition of tumor growth. In conclusion, LBH589, acting on both histones and nonhistonic proteins in anaplastic thyroid cancer, appears to be a promising therapeutic agent for the treatment of this kind of cancer which is known not to respond to conventional therapy.


Assuntos
Antineoplásicos/toxicidade , Inibidores de Histona Desacetilases/toxicidade , Ácidos Hidroxâmicos/toxicidade , Neoplasias da Glândula Tireoide/metabolismo , Acetilação/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Ácidos Hidroxâmicos/uso terapêutico , Indóis , Camundongos , Camundongos Nus , Panobinostat , Carcinoma Anaplásico da Tireoide , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/genética , Tubulina (Proteína)/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Stem Cells ; 29(6): 952-63, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21563276

RESUMO

Hypoxic conditions have been reported to facilitate preservation of undifferentiated mesenchymal stem cell (MSC) phenotype and positively affect their colony-forming potential, proliferation, and migration/mobilization. In this study, designed to dissect mechanisms underlying hypoxia-dependent migration of bone marrow-derived human MSC (hMSC), signal transduction, and molecular mechanisms were evaluated by integrating morphological, molecular, and cell biology techniques, including the wound healing assay (WHA) and modified Boyden's chamber assay (BCA) to monitor migration. Exposure of hMSCs to moderate hypoxia resulted in a significant increase of migration of hMSCs in both WHA (from 6 to 20 hours) and BCA (within 6 hours). Mechanistic experiments outlined the following sequence of hypoxia-dependent events: (a) very early (15 minutes) increased generation of intracellular reactive oxygen species (ROS), which (b) was sufficient to switch on activation of extracellular regulated kinase 1/2 and c-Jun N-terminal protein kinase 1/2, found to be relevant for the early phase of hMSC migration; (c) hypoxia inducible factor-1 (HIF-1)-dependent increased expression of vascular endothelial growth factor (VEGF) (facilitated by ROS) and its progressive release that was responsible for (d) a delayed and sustained migration of hMSCs. These results suggest that hypoxia-dependent migration relies on a previously unrecognized biphasic scenario involving an early phase, requiring generation of ROS, and a delayed phase sustained by HIF-1-dependent expression and release of VEGF.


Assuntos
Células da Medula Óssea/fisiologia , Movimento Celular , Células-Tronco Mesenquimais/fisiologia , Antígenos CD/metabolismo , Células da Medula Óssea/citologia , Hipóxia Celular , Células Cultivadas , Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estabilidade Proteica , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
J Hepatol ; 54(5): 964-74, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21145826

RESUMO

BACKGROUND & AIMS: Liver fibrogenesis is sustained by myofibroblast-like cells originating from hepatic stellate cells (HSC/MFs), portal fibroblasts or bone marrow-derived cells, including mesenchymal stem cells (MSCs). Herein, we investigated the mechanistic role of intracellular generation of reactive oxygen species (ROS) and redox-sensitive signal transduction pathways in mediating chemotaxis, a critical profibrogenic response for human HSC/MFs and for MSC potentially engrafting chronically injured liver. METHODS: Intracellular generation of ROS and signal transduction pathways were evaluated by integrating morphological and molecular biology techniques. Chemokinesis and chemotaxis were evaluated by wound healing assay and modified Boyden's chamber assay, respectively. Additional in vivo evidence was obtained in human specimens from HCV-related cirrhosis. RESULTS: Human MSCs and HSC/MFs migrate in response to a panel of polypeptide chemoattractants and extracellularly generated superoxide anion. All polypeptides induced a NADPH-oxidase-dependent intracellular rise in ROS, resulting in activation of ERK1/2 and JNK1/2. Moreover, menadione or 2,3-dimethoxy-1,4-naphthoquinone, which generate intracellular superoxide anion or hydrogen peroxide, respectively, induced ERK1/2 and JNK1/2 activation and migration. JNK1 activation was predominant for migration as shown by specific silencing. Finally, activation of ERK1/2 and JNK1/2 was found in extracts obtained from HSC/MFs during the course of an oxidative stress-mediated model of liver injury and phosphorylated JNK1/2 isoforms were detected in α-smooth muscle actin-positive myofibroblasts lining fibrotic septa in human cirrhotic livers. CONCLUSIONS: Intracellular generation of ROS, through activation of specific signaling pathways, is a critical event for directional migration of HSC/MFs and MSCs.


Assuntos
Células da Medula Óssea/citologia , Células Estreladas do Fígado/citologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Células-Tronco Mesenquimais/citologia , Espécies Reativas de Oxigênio/metabolismo , Células da Medula Óssea/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Fatores Quimiotáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células Estreladas do Fígado/metabolismo , Hepatite C Crônica/metabolismo , Hepatite C Crônica/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Células-Tronco Mesenquimais/metabolismo
9.
Lab Invest ; 90(6): 929-39, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20212449

RESUMO

Epithelial-to-mesenchymal transition (EMT) is involved in embryonic development as well as in several pathological conditions. Literature indicates that polyamine availability may affect transcription of c-myc, matrix metalloproteinase (MMP)1, MMP2, TGFbeta(1), and collagen type I mRNA. The aim of this study was to elucidate polyamines role in EMT in vitro. Madin-Darby canine kidney (MDCK) cells were subjected to experimental manipulation of intracellular levels of polyamines. Acquisition of mesenchymal phenotype was evaluated by means of immunofluorescence, western blots, and zymograms. MDCK cells were then subjected to 2D gel proteomic study and incorporation of a biotinilated polyamine (BPA). Polyamine endocellular availability modulated EMT process. Polyamine-depleted cells treated with TGFbeta(1) showed enhanced EMT with a marked decrease of E-cadherin expression at plasma membrane level and an increased expression of mesenchymal markers such as fibronectin and alpha-smooth muscle actin. Polyamine-depleted cells showed a twofold increased expression of the rough endoplasmic reticulum (ER)-stress proteins GRP78, GRP94, and HSP90 alpha/beta in 2D gels. The latter data were confirmed by western blot analysis. Administration of BPA showed that polyamines are covalently linked, within the cell, to ER-stress proteins. Intracellular polyamine availability affects EMT in MDCK cells possibly through the modulation of ER-stress protein homeostasis.


Assuntos
Rim/citologia , Rim/fisiologia , Mesoderma/fisiologia , Animais , Comunicação Celular/fisiologia , Cães , Regulação para Baixo , Desenvolvimento Embrionário , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Metaloproteinases da Matriz/metabolismo , Mesoderma/efeitos dos fármacos , Poliaminas/metabolismo , Desnaturação Proteica , RNA Mensageiro/genética , Espermidina Sintase/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia
10.
Int J Biochem Cell Biol ; 41(3): 586-94, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18708157

RESUMO

It is well established that cobalt mediates the occurrence of oxidative stress which contributes to cell toxicity and death. However, the mechanisms of these effects are not fully understood. This investigation aimed at establishing if cobalt acts as an inducer of mitochondrial-mediated apoptosis and at clarifying the mechanism of this process. Cobalt, in the ionized species Co(2+), is able to induce the phenomenon of mitochondrial permeability transition (MPT) in rat liver mitochondria (RLM) with the opening of the transition pore. In fact, Co(2+) induces mitochondrial swelling, which is prevented by cyclosporin A and other typical MPT inhibitors such as Ca(2+) transport inhibitors and bongkrekic acid, as well as anti-oxidant agents. In parallel with mitochondrial swelling, Co(2+) also induces the collapse of electrical membrane potential. However in this case, cyclosporine A and the other MPT inhibitors (except ruthenium red and EGTA) only partially prevent DeltaPsi drop, suggesting that Co(2+) also has a proton leakage effect on the inner mitochondrial membrane. MPT induction is due to oxidative stress, as a result of generation by Co(2+) of the highly damaging hydroxyl radical, with the oxidation of sulfhydryl groups, glutathione and pyridine nucleotides. Co(2+) also induces the release of the pro-apoptotic factors, cytochrome c and AIF. Incubation of rat hepatocyte primary cultures with Co(2+) results in apoptosis induction with caspase activation and increased level of expression of HIF-1alpha. All these observations allow us to state that, in the presence of calcium, Co(2+) is an inducer of apoptosis triggered by mitochondrial oxidative stress.


Assuntos
Cobalto/toxicidade , Hepatócitos/fisiologia , Mitocôndrias Hepáticas/fisiologia , Estresse Oxidativo/fisiologia , Animais , Apoptose/fisiologia , Fator de Indução de Apoptose/genética , Fator de Indução de Apoptose/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Fracionamento Celular , Células Cultivadas , Cobalto/análise , Citocromos c/análise , Hepatócitos/ultraestrutura , Peróxido de Hidrogênio/análise , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Membranas Intracelulares , Masculino , Potencial da Membrana Mitocondrial , Mitocôndrias Hepáticas/química , Permeabilidade , Ratos , Ratos Wistar
11.
Carcinogenesis ; 29(12): 2267-78, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18791199

RESUMO

Epithelial-mesenchymal transition (EMT) and hypoxia are considered as crucial events favouring invasion and metastasis of many cancer cells. In this study, different human neoplastic cell lines of epithelial origin were exposed to hypoxic conditions in order to investigate whether hypoxia per se may trigger EMT programme as well as to mechanistically elucidate signal transduction mechanisms involved. The following human cancer cell lines were used: HepG2 (from human hepatoblastoma), PANC-1 (from pancreatic carcinoma), HT-29 (from colon carcinoma) and MCF-7 (from breast carcinoma). Cancer cells were exposed to carefully controlled hypoxic conditions and investigated for EMT changes and signal transduction by using morphological, cell and molecular biology techniques. All cancer cells responded to hypoxia within 72 h by classic EMT changes (fibroblastoid phenotype, SNAIL and beta-catenin nuclear translocation and changes in E-cadherin) and by increased migration and invasiveness. This was involving very early inhibition of glycogen synthase kinase-3beta (GSK-3beta), early SNAIL translocation as well as later and long-lasting activation of Wnt/beta-catenin-signalling machinery. Experimental manipulation, including silencing of hypoxia-inducible factor (HIF)-1alpha and the specific inhibition of mitochondrial generation of reactive oxygen species (ROS), revealed that early EMT-related events induced by hypoxia (GSK-3beta inhibition and SNAIL translocation) were dependent on transient intracellular increased generation of ROS whereas late migration and invasiveness were sustained by HIF-1alpha- and vascular endothelial growth factor (VEGF)-dependent mechanisms. These findings indicate that in cancer cells, early redox mechanisms can switch on hypoxia-dependent EMT programme whereas increased invasiveness is sustained by late and HIF-1alpha-dependent release of VEGF.


Assuntos
Hipóxia Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Transdução de Sinais/fisiologia , Western Blotting , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Epitélio/metabolismo , Epitélio/patologia , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mesoderma/metabolismo , Mesoderma/patologia , Invasividade Neoplásica/genética , Invasividade Neoplásica/fisiopatologia , Oxirredução , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
12.
Int J Biochem Cell Biol ; 40(9): 1861-71, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18343708

RESUMO

Disruption of cell-to-cell contacts, as observed in many pathophysiological conditions, prime hepatocytes for compensatory hyperplastic response that involves induction of several genes, including proto-oncogenes and other gene targets of beta-catenin signaling pathway. By using cultured hepatocytes and experimental models of adherens junction disruption we have investigated changes in beta-catenin subcellular localization and their relationships with inducible nitric oxide synthase (iNOS) expression. Two experimental models were employed: (a) rat hepatocytes obtained by collagenase liver perfusion within the first 48 h of culture; (b) 48-h old cultured hepatocytes, transiently transfected or not with a plasmid encoding for dominant/negative inhibitory kappa B-alpha, exposed to ethylene glycol-bis-(2-aminoethylether)-N,N,N',N'-tetraacetic acid/LiCl treatment. beta-Catenin signaling and cellular localization, iNOS expression and nuclear factor kappaB involvement, were investigated using morphological, cell and molecular biology techniques. E-cadherin-mediated disruption of cell-to-cell contacts induces early beta-catenin translocation from membrane to cytoplasm and nuclear compartments, events that are followed by up-regulation of c-myc, cyclin D1 and beta-transducin repeat-containing protein expression. This, in turn, resulted eventually in iNOS induction that was mechanistically related to nuclear factor kappaB activation, as unequivocally shown in cells expressing dominant negative inhibitory kappa B-alpha. Our data indicate that E-cadherin disassembly and concomitant inactivation of glycogen synthase kinase-3beta result in nuclear factor kappaB-dependent induction of iNOS in hepatocytes.


Assuntos
Hepatócitos/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Regulação para Cima , beta Catenina/metabolismo , Animais , Caderinas/metabolismo , Bovinos , Comunicação Celular , Células Cultivadas , Ácido Egtázico/farmacologia , Indução Enzimática/efeitos dos fármacos , Hepatócitos/enzimologia , Cloreto de Lítio/farmacologia , Masculino , Óxido Nítrico Sintase Tipo II/metabolismo , Transporte Proteico , Ratos , Reprodutibilidade dos Testes , Regulação para Cima/efeitos dos fármacos , Proteínas Contendo Repetições de beta-Transducina/metabolismo
13.
Am J Pathol ; 170(6): 1942-53, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17525262

RESUMO

Pathological angiogenesis is associated with the fibrogenic progression of chronic liver diseases. Experimental data suggest that hypoxia and vascular endothelial growth factor (VEGF) may stimulate proliferation and synthesis of type I collagen in activated, myofibroblast-like rat hepatic stellate cells (HSC/MFs). In this study, we investigated whether hypoxia, recombinant VEGF, or angiopoietin 1 (Ang-1) may affect other crucial profibrogenic features. In human HSC/MFs, which constitutively express VEGF receptor-1 and -2 (VEGFR-1, VEGFR-2) and the Ang-1 receptor Tie-2, exposure to hypoxia, VEGF, or Ang-1 resulted in a Ras/Erk-dependent stimulation of chemokinesis and chemotaxis. Migration of human HSC/MFs under hypoxic conditions involved up-regulation of VEGF-A, Ang-1, and related receptors and was mainly dependent on VEGFR-2 (Flk-1). In specimens from either cirrhotic rat livers or from patients with hepatitis C virus-related cirrhosis, HSC/MFs expressed proangiogenic factors and related receptors in areas of active fibrogenesis (ie, at the leading or lateral edge of developing incomplete fibrotic septa). Data presented herein suggest that VEGF and Ang-1 may contribute to fibrogenesis by acting as hypoxia-inducible, autocrine, and paracrine factors able to recruit myofibroblast-like cells. Moreover, HSC/MFs, in addition to their established profibrogenic role, may also contribute to neoangiogenesis during chronic hepatic wound healing.


Assuntos
Angiopoietina-1/metabolismo , Movimento Celular/fisiologia , Hipóxia , Fígado/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Meios de Cultura/metabolismo , Fibrose/metabolismo , Fibrose/patologia , Hepatite C/patologia , Humanos , Fígado/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , Ratos , Ratos Wistar , Receptor TIE-2/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Cicatrização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA