Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Antioxidants (Basel) ; 13(1)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-38247453

RESUMEN

Cholangiocarcinoma (CCA) is a primary liver tumor that accounts for 2% of all cancer-related deaths worldwide yearly. It can arise from cholangiocytes of biliary tracts, peribiliary glands, and possibly from progenitor cells or even hepatocytes. CCA is characterized by high chemoresistance, aggressiveness, and poor prognosis. Potentially curative surgical therapy is restricted to a small number of patients with early-stage disease (up to 35%). Accumulating evidence indicates that CCA is an oxidative stress-driven carcinoma resulting from chronic inflammation. Oxidative stress, due to enhanced reactive oxygen species (ROS) production and/or decreased antioxidants, has been recently suggested as a key factor in cholangiocyte oncogenesis through gene expression alterations and molecular damage. However, due to different experimental models and conditions, contradictory results regarding oxidative stress in cholangiocarcinoma have been reported. The role of ROS and antioxidants in cancer is controversial due to their context-dependent ability to stimulate tumorigenesis and support cancer cell proliferation or promote cell death. On these bases, the present narrative review is focused on illustrating the role of oxidative stress in cholangiocarcinoma and the main ROS-driven intracellular pathways. Heterogeneous data about antioxidant effects on cancer development are also discussed.

2.
Cancers (Basel) ; 14(17)2022 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-36077744

RESUMEN

Primary liver cancers represent the third-most-common cause of cancer-related mortality worldwide, with an incidence of 80-90% for hepatocellular carcinoma (HCC) and 10-15% for cholangiocarcinoma (CCA), and an increasing morbidity and mortality rate. Although HCC and CCA originate from independent cell populations (hepatocytes and biliary epithelial cells, respectively), they develop in chronically inflamed livers. Evidence obtained in the last decade has revealed a role for cytokines of the IL-6 family in the development of primary liver cancers. These cytokines operate through the receptor subunit gp130 and the downstream Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathways. Oncostatin M (OSM), a member of the IL-6 family, plays a significant role in inflammation, autoimmunity, and cancer, including liver tumors. Although, in recent years, therapeutic approaches for the treatment of HCC and CCA have been implemented, limited treatment options with marginal clinical benefits are available. We discuss how OSM-related pathways can be selectively inhibited and therapeutically exploited for the treatment of liver malignancies.

3.
Cells ; 10(10)2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34685739

RESUMEN

Chronic liver injury of different etiologies may result in hepatic fibrosis, a scar formation process consisting in altered deposition of extracellular matrix. Progression of fibrosis can lead to impaired liver architecture and function, resulting in cirrhosis and organ failure. Although fibrosis was previous thought to be an irreversible process, recent evidence convincingly demonstrated resolution of fibrosis in different organs when the cause of injury is removed. In the liver, due to its high regenerative ability, the extent of fibrosis regression and reversion to normal architecture is higher than in other tissues, even in advanced disease. The mechanisms of liver fibrosis resolution can be recapitulated in the following main points: removal of injurious factors causing chronic hepatic damage, elimination, or inactivation of myofibroblasts (through various cell fates, including apoptosis, senescence, and reprogramming), inactivation of inflammatory response and induction of anti-inflammatory/restorative pathways, and degradation of extracellular matrix. In this review, we will discuss the major cellular and molecular mechanisms underlying the regression of fibrosis/cirrhosis and the potential therapeutic approaches aimed at reversing the fibrogenic process.


Asunto(s)
Cirrosis Hepática/genética , Cirrosis Hepática/patología , Animales , Matriz Extracelular/metabolismo , Humanos , Inflamación/patología , Cirrosis Hepática/fisiopatología , Miofibroblastos/patología , Inducción de Remisión , Remodelación Vascular
4.
Hepatology ; 74(4): 2007-2020, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33959996

RESUMEN

BACKGROUND AND AIMS: Cholangiocarcinoma (CCA) is characterized by high resistance to chemotherapy and poor prognosis. Several oncogenic pathways converge on activation of extracellular signal-regulated kinase 5 (ERK5), whose role in CCA has not been explored. The aim of this study was to investigate the role of ERK5 in the biology of CCA. APPROACH AND RESULTS: ERK5 expression was detected in two established (HuCCT-1 and CCLP-1) and two primary human intrahepatic CCA cell lines (iCCA58 and iCCA60). ERK5 phosphorylation was increased in CCA cells exposed to soluble mediators. In both HuCCT-1 and CCLP-1 cells, ERK5 was localized in the nucleus, and exposure to fetal bovine serum (FBS) further increased the amount of nuclear ERK5. In human CCA specimens, ERK5 mRNA expression was increased in tumor cells and positively correlated with portal invasion. ERK5 protein levels were significantly associated with tumor grade. Growth, migration, and invasion of CCA cells were decreased when ERK5 was silenced using specific short hairpin RNA (shRNA). The inhibitory effects on CCA cell proliferation, migration and invasion were recapitulated by treatment with small molecule inhibitors targeting ERK5. In addition, expression of the angiogenic factors VEGF and angiopoietin 1 was reduced after ERK5 silencing. Conditioned medium from ERK5-silenced cells had a lower ability to induce tube formation by human umbilical vein endothelial cells and to induce migration of myofibroblasts and monocytes/macrophages. In mice, subcutaneous injection of CCLP-1 cells silenced for ERK5 resulted in less frequent tumor development and smaller size of xenografts compared with cells transfected with nontargeting shRNA. CONCLUSIONS: ERK5 is a key mediator of growth and migration of CCA cells and supports a protumorigenic crosstalk between the tumor and the microenvironment.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos , Colangiocarcinoma/genética , Proteína Quinasa 7 Activada por Mitógenos/genética , Animales , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Medios de Cultivo Condicionados , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Macrófagos , Ratones , Monocitos , Miofibroblastos , Clasificación del Tumor , Invasividad Neoplásica , Trasplante de Neoplasias , Neovascularización Patológica/genética , Fenotipo , ARN Mensajero/metabolismo
5.
Cancers (Basel) ; 12(8)2020 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-32784743

RESUMEN

Cholangiocarcinoma (CCA), a heterogeneous tumor with poor prognosis, can arise at any level in the biliary tree. It may derive from epithelial cells in the biliary tracts and peribiliary glands and possibly from progenitor cells or even hepatocytes. Several risk factors are responsible for CCA onset, however an inflammatory milieu nearby the biliary tree represents the most common condition favoring CCA development. Chemokines play a key role in driving the immunological response upon liver injury and may sustain tumor initiation and development. Chemokine receptor-dependent pathways influence the interplay among various cellular components, resulting in remodeling of the hepatic microenvironment towards a pro-inflammatory, pro-fibrogenic, pro-angiogenic and pre-neoplastic setting. Moreover, once tumor develops, chemokine signaling may influence its progression. Here we review the role of chemokines in the regulation of CCA development and progression, and the modulation of angiogenesis, metastasis and immune control. The potential role of chemokines and their receptors as possible biomarkers and/or therapeutic targets for hepatobiliary cancer is also discussed.

6.
Cells ; 9(3)2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32138158

RESUMEN

Cholangiocarcinoma (CCA) is a deadly tumor without an effective therapy. Unique metabolic and bioenergetics features are important hallmarks of tumor cells. Metabolic plasticity allows cancer cells to survive in poor nutrient environments and maximize cell growth by sustaining survival, proliferation, and metastasis. In recent years, an increasing number of studies have shown that specific signaling networks contribute to malignant tumor onset by reprogramming metabolic traits. Several evidences demonstrate that numerous metabolic mediators represent key-players of CCA progression by regulating many signaling pathways. Besides the well-known Warburg effect, several other different pathways involving carbohydrates, proteins, lipids, and nucleic acids metabolism are altered in CCA. The goal of this review is to highlight the main metabolic processes involved in the cholangio-carcinogeneis that might be considered as potential novel druggable candidates for this disease.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Plasticidad de la Célula/genética , Colangiocarcinoma/genética , Neoplasias de los Conductos Biliares/fisiopatología , Proliferación Celular , Colangiocarcinoma/fisiopatología , Humanos , Mitocondrias/metabolismo
7.
Oncogenesis ; 8(11): 61, 2019 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-31641101

RESUMEN

Cholangiocarcinoma (CCA) is a particularly aggressive hepatobiliary malignancy, for which the molecular mechanisms underlying the malignant phenotype are still poorly understood, and novel and effective therapeutic strategies are limited. The pro-survival protein kinase CK2 is frequently overexpressed in cancer and is receiving increasing interest as an anti-tumor drug target. Its precise role in CCA biology is still largely unknown. Here we show that expression of the CK2α and α' catalytic subunits and of the ß regulatory subunit is increased in human CCA samples. Increased expression of CK2 subunits was shown in CCA cell lines compared to non-transformed cholangiocytes. We used chemical inhibition of CK2 and genetic modification by CRISPR/Cas9 to explore the contribution of CK2 to the malignant phenotype of CCA cells. Disruption of CK2 activity results in cell death through apoptosis, reduced invasion and migration potential, and G0/G1 cell cycle arrest. Importantly, CCA cells with a reduced CK2 activity are more sensitive to chemotherapy. Altogether, our results demonstrate that CK2 significantly contributes to increased proliferative potential and augmented growth of CCA cells and indicate the rationale for its targeting as a promising pharmacologic strategy for cholangiocarcinoma.

8.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2246-2256, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31059778

RESUMEN

Development of cholangiocarcinoma (CCA) is dependent on a cross-talk with stromal cells, which release different chemokines including CXCL12, that interacts with two different receptors, CXCR4 and CXCR7. The aim of the present study was to investigate the role of CXCR7 in CCA cells. CXCR7 is overexpressed by different CCA cell lines and in human CCA specimens. Knock-down of CXCR7 in HuCCT-1 cells reduced migration, invasion, and CXCL12-induced adhesion to collagen I. Survival of CCA was also reduced in CXCR7-silenced cells. The ability of CXCL12 to induce cell migration and survival was also blocked by CCX733, a CXCR7 antagonist. Similar effects of CXCR7 activation were observed in CCLP-1 cells and in primary iCCA cells. Enrichment of tumor stem-like cells by a 3D culture system resulted in increased CXCR7 expression compared to cells grown in monolayers, and genetic knockdown of CXCR7 robustly reduced sphere formation both in HuCCT-1 and in CCLP-1 cells. In HuCCT-1 cells CXCR7 was found to interact with ß-arrestin 2, which was necessary to mediate CXCL12-induced migration, but not survival. In conclusion, CXCR7 is widely expressed in CCA, and contributes to the aggressive phenotype of CCA cells, inducing cell migration, invasion, adhesion, survival, growth and stem cell-like features. Cell migration induced by CXCR7 requires interaction with ß-arrestin 2.


Asunto(s)
Neoplasias de los Conductos Biliares/patología , Colangiocarcinoma/patología , Receptores CXCR/metabolismo , Neoplasias de los Conductos Biliares/metabolismo , Movimiento Celular , Supervivencia Celular , Quimiocina CXCL12/metabolismo , Colangiocarcinoma/metabolismo , Femenino , Humanos , Hígado/metabolismo , Hígado/patología , Masculino , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores CXCR/antagonistas & inhibidores , Receptores CXCR/genética , Células Tumorales Cultivadas , Arrestina beta 2/metabolismo
9.
Int J Mol Sci ; 19(10)2018 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-30249019

RESUMEN

Cholangiocarcinoma (CCA) is a severe and mostly intractable adenocarcinoma of biliary epithelial cells. A typical feature of CCA is its highly desmoplastic microenvironment containing fibrogenic connective tissue and an abundance of immune cells (T lymphocytes, Natural Killer (NK) cells, and macrophages) infiltrating tumor epithelium. This strong desmoplasia is orchestrated by various soluble factors and signals, suggesting a critical role in shaping a tumor growth-permissive microenvironment that is responsible for CCA poor clinical outcome. Indeed stroma not only provides an abundance of factors that facilitate CCA initiation, growth and progression, but also a prejudicial impact on therapeutic outcome. This review will give an overview of tumor-stroma signaling in a microenvironment critically regulating CCA development and progression. Identification of CCA secreted factors by both the fibroblast component and immune cell subsets might provide ample opportunities for pharmacological targeting of this type of cancer.


Asunto(s)
Neoplasias de los Conductos Biliares/fisiopatología , Colangiocarcinoma/fisiopatología , Epitelio/inmunología , Fibroblastos/inmunología , Subgrupos de Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Animales , Epitelio/patología , Fibroblastos/patología , Humanos , Subgrupos de Linfocitos T/patología
10.
Int J Mol Sci ; 17(9)2016 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-27657051

RESUMEN

Nonalcoholic steatohepatitis (NASH) is the main cause of chronic liver disease in the Western world and a major health problem, owing to its close association with obesity, diabetes, and the metabolic syndrome. NASH progression results from numerous events originating within the liver, as well as from signals derived from the adipose tissue and the gastrointestinal tract. In a fraction of NASH patients, disease may progress, eventually leading to advanced fibrosis, cirrhosis and hepatocellular carcinoma. Understanding the mechanisms leading to NASH and its evolution to cirrhosis is critical to identifying effective approaches for the treatment of this condition. In this review, we focus on some of the most recent data reported on the pathogenesis of NASH and its fibrogenic progression, highlighting potential targets for treatment or identification of biomarkers of disease progression.

11.
J Hepatol ; 57(4): 813-20, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22727731

RESUMEN

BACKGROUNDS & AIMS: Cholangiocarcinoma (CCA) is highly fatal because of early invasion, widespread metastasis, and lack of an effective therapy. Migration, invasion, and metastasis of CCA cells are modulated by signals received from stromal cells. The SDF-1-CXCR4 axis emerges as a pivotal regulator of migration and survival of different tumor cells. The aim of the present study was to characterize the interaction between CCA cells and human hepatic stellate cells (hHSC) focusing on the role of SDF-1. METHODS: The intrahepatic CCA cell line HuCCT-1 and primary hHSC were used for this study. RNA expression was examined by RTQ-PCR and protein expression by Western blotting. Immunofluorescence microscopy and immunohistochemistry were also employed. Migration of CCA cells was assessed using modified Boyden chambers. RESULTS: CXCR4 was clearly expressed in CCA cells of human CCA liver specimens. SDF-1 and hHSC conditioned medium (CM) promoted HuCCT-1 cell migration, which was abrogated by pre-incubation with AMD3100, a non-peptide antagonist of the CXCR4 receptor. In addition, HuCCT-1 cells silenced for CXCR4 did not migrate in presence of SDF-1. Both P-ERK and p-AKT were implicated in HuCCT-1 migration and showed a biphasic trend under stimulation of SDF-1. Finally, SDF-1 induced apoptotic rescue of HuCCT-1 cells by binding to CXCR4. CONCLUSIONS: Our study demonstrates that CCA cells migration and survival are modulated by the crosstalk between SDF-1, released by hHSC, and HuCCT-1 cells bearing CXCR4.


Asunto(s)
Neoplasias de los Conductos Biliares/metabolismo , Conductos Biliares Intrahepáticos , Comunicación Celular , Quimiocina CXCL12/metabolismo , Colangiocarcinoma/metabolismo , Células Estrelladas Hepáticas/metabolismo , Receptores CXCR4/metabolismo , Adulto , Anciano , Apoptosis/efectos de los fármacos , Bencilaminas , Línea Celular Tumoral , Quimiocina CXCL12/farmacología , Quimiotaxis/efectos de los fármacos , Quimiotaxis/genética , Ciclamas , Femenino , Silenciador del Gen , Compuestos Heterocíclicos/farmacología , Humanos , Hígado/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/farmacología , Masculino , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , ARN/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/genética
12.
Fibrogenesis Tissue Repair ; 2(1): 1, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19183465

RESUMEN

BACKGROUND: Human hepatic stellate cells have been shown to be resistant to apoptotic stimuli. This is likely dependent on the activation of anti-apoptotic pathways upon transition of these cells to myofibroblast-like cells. In particular, previous studies have demonstrated an increased expression of the anti-apoptotic protein Bcl-2 and a decreased expression of the pro-apoptotic protein Bax during the transition of the hepatic stellate cell phenotype from quiescent to myofibroblast-like cells. However, the role and expression of other key anti-apoptotic and survival pathways elicited by polypeptide growth factors involved in the chronic wound healing process remain to be elucidated. In particular, insulin growth factor-I promotes chemotactic and mitogenic effects in activated human hepatic stellate cells and these effects are mediated by the activation of PI 3-K. The role of insulin growth factor-I as a survival factor in human hepatic stellate cells needs to be substantiated. The aim of this study was to evaluate the involvement of other key anti-apoptotic pathways such as PI-3K/Akt/p-Bad in response to insulin growth factor-I. RESULTS: Insulin growth factor-I induced activation of Akt followed by Bad phosphorylation after 15 minutes of incubation. These effects were PI-3k dependent since selective inhibitors of this molecule, wortmannin and LY294002, inhibited both Akt and Bad phosphorylation. The effect of insulin growth factor-I on the activation of two downstream targets of Akt activation, that is, GSK3 and FHKR, both implicated in the promotion of cell survival was also investigated. Both targets became phosphorylated after 15 minutes of incubation, and these effects were also PI-3K-dependent. Despite the activation of this survival pathway insulin growth factor-I did not have a remarkable biological effect, probably because other insulin growth factor-I-independent survival pathways were already maximally activated in the process of hepatic stellate cell activation. However, after incubation of the cells with a strong apoptotic stimuli such as Fas ligand+cycloheximide, a small percentage of hepatic stellate cells underwent programmed cell death that was partially rescued by insulin growth factor-I. CONCLUSION: In addition to Bcl-2, several other anti-apoptotic pathways are responsible for human hepatic stellate cell resistance to apoptosis. These features are relevant for the progression and limited reversibility of liver fibrosis in humans.

13.
J Hepatol ; 40(1): 60-8, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14672615

RESUMEN

BACKGROUND/AIMS: 4-Hydroxynonenal (HNE) is a putative pro-fibrogenic product of oxidative stress able to elicit apoptosis and cytotoxicity in several cell types. This study has been performed to evaluate its 'in vivo' levels in injured liver and whether HNE may induce apoptosis and/or affect selected phenotypic responses in activated human hepatic stellate cells (HSC/MF). METHODS/RESULTS: During the development of acute liver injury induced by CCl(4), liver tissue HNE levels were in the range 0.5-10 microM, as shown by high performance liquid chromatography analysis. Cultured human HSC/MF, developed cytotoxicity only if exposed to very high HNE concentrations (25-50 microM) without any sign of induction of classic, caspase-dependent apoptosis, as assessed by evaluating morphology and biochemical parameters of cell death. HNE, at non-cytotoxic doses, up-regulated procollagen type I and tissue inhibitor of metalloproteinases-1 gene expression and/or protein synthesis without significantly affecting chemotaxis (wound healing and haptotaxis assay), matrix metalloproteinases 1 and 2 mRNA expression and activity as well as basal DNA synthesis. CONCLUSIONS: HNE, at concentrations compatible with those detected in vivo, does not elicit HSC/MF classic apoptosis but, rather, may act as a potent pro-fibrogenic stimulus for the expression of genes involved in excess extracellular matrix deposition and proposed as survival signals for HSC/MF.


Asunto(s)
Aldehídos/farmacología , Cirrosis Hepática/inducido químicamente , Hígado/efectos de los fármacos , Actinas/metabolismo , Enfermedad Aguda , Aldehídos/administración & dosificación , Aldehídos/metabolismo , Animales , Apoptosis , Tetracloruro de Carbono , Muerte Celular , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas , Citoesqueleto/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/etiología , Hepatopatías/metabolismo , Hepatopatías/patología , Hepatopatías/fisiopatología , Masculino , Concentración Osmolar , Fenotipo , Ratas , Ratas Wistar , Transducción de Señal
14.
Gastroenterology ; 124(2): 504-20, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12557155

RESUMEN

BACKGROUND & AIMS: Several lines of evidence indicate that aldosterone antagonists may exert direct antifibrogenic effects. The aim of this study was to evaluate the possible direct antifibrogenic effects of canrenone, the active metabolite of spironolactone, in activated human hepatic stellate cells. METHODS: The effects of canrenone were assessed on platelet-derived growth factor-induced mitogenic and chemotactic effects and the increased de novo synthesis of different extracellular matrix components induced by transforming growth factor-beta1. RESULTS: Canrenone dose-dependently reduced platelet-derived growth factor-induced cell proliferation and motility. This effect was not associated with either changes in the phosphorylation of platelet-derived growth factor receptor and phospholipase C gamma or in the activation of the Ras/extracellular signal-regulated kinase pathway, whereas it was accompanied by a dose-dependent inhibition of platelet-derived growth factor-induced phosphatidylinositol 3-kinase activity. In addition, canrenone inhibited the activity of the Na(+)/H(+) exchanger 1 induced by platelet-derived growth factor. The effect of canrenone on Na(+)/H(+) exchanger 1 activity was reproduced by phosphatidylinositol 3-kinase inhibitors, thus supporting an inhibitory action of canrenone on phosphatidylinositol 3-kinase activity. To further address this possibility, the action of canrenone was compared with that of 2 established Na(+)/H(+) exchanger 1 inhibitors: ethylisopropylamiloride and cariporide. Whereas ethylisopropylamiloride was able to inhibit platelet-derived growth factor-induced phosphatidylinositol 3-kinase activity, cariporide was without any effect. Both compounds reproduced the effects of canrenone on platelet-derived growth factor-induced mitogenesis and chemotaxis. Finally, canrenone was able to reduce transforming growth factor-beta1-induced de novo synthesis of procollagen type I/IV and fibronectin and thrombin-induced hepatic stellate cell contraction. CONCLUSIONS: These results indicate that canrenone may be active as an antifibrogenic drug.


Asunto(s)
Amilorida/análogos & derivados , Canrenona/farmacología , Cirrosis Hepática/prevención & control , Hígado/efectos de los fármacos , Antagonistas de Receptores de Mineralocorticoides/farmacología , Aldosterona/metabolismo , Amilorida/farmacología , Calcio/metabolismo , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Proteínas de la Matriz Extracelular/biosíntesis , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Guanidinas/farmacología , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Membranas Intracelulares/metabolismo , Hígado/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Sulfonas/farmacología , Trombina/farmacología
15.
Hepatology ; 36(3): 582-91, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12198650

RESUMEN

The biologic effects of growth factors are dependent on cell adhesion, and a cross talk occurs between growth factors and adhesion complexes. The aim of the present study was to evaluate the influence of cell adhesion on the major intracellular signaling pathways elicited by platelet-derived growth factor (PDGF) in hepatic stellate cells (HSC). PDGF signaling was investigated in an experimental condition characterized by lack of cell adhesion for different intervals of time. Basal and PDGF-induced focal adhesion kinase (FAK) tyrosine phosphorylation was maintained in a condition of cell suspension for 2, 4, and 6 hours, whereas it was completely lost after 12 and 24 hours. We examined MAP kinase activity at 2 and 24 hours, corresponding to the higher and lower levels of FAK phosphorylation. In these experiments, MAP kinase activity correlated with FAK phosphorylation. Stimulation with PDGF was able to cause Ras-GTP loading only in adherent cells. The ability of PDGF to induce phosphatidylinositol 3-kinase (PI 3-K) activity was abrogated in cells maintained in suspension. The Ser473 phosphorylation of Akt was only marginally affected by the lack of cell adhesion. We then evaluated the association of FAK with c-Src. This association was found to be cell adhesion dependent, and it did not appear to be dependent from phosphorylated FAK. These changes in PDGF-induced intracellular signaling were associated with a remarkable reduction of PDGF-proliferative potential in nonadherent cells, although no marked differences in the apoptotic rate were observed. In conclusion, these results suggest that cell adhesion differentially regulates major signaling pathways activated by PDGF in HSC.


Asunto(s)
Hepatocitos/enzimología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Serina-Treonina Quinasas , Proteína Tirosina Quinasa CSK , Calcio/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos , Isoenzimas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfolipasa C gamma , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Fosfolipasas de Tipo C/metabolismo , Tirosina/metabolismo , Proteínas ras/metabolismo , Familia-src Quinasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA