Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 13(9): 814, 2022 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-36138026

RESUMEN

Tyrosine kinase inhibitors (TKIs) are the most commonly used targeted therapeutics in clear-cell renal cell carcinoma (ccRCC); however, drug resistance limits their utility and can lead to tumor "flare-up" and progression. In this study, we show that RCC resistance to sunitinib and sorafenib involves different mechanisms and leads to increased malignancy. Sunitinib decreased tumor growth and cell motility along with increased E-cadherin expression and secretion of the proangiogenic cytokines IL6 and IL8, which activated senescence in ccRCC cells and led to VE-cadherin phosphorylation, enhancing tumor angiogenesis. Sorafenib resistance increased the levels of mesenchymal markers and the secretion of MMP9, which cleaved VE-cadherin and disrupted endothelial cell integrity. Both sunitinib resistance and sorafenib resistance led to activation of the c-Met receptor IRAK1 and downregulation of the tumor suppressor MCPIP1, resulting in an increase in the metastasis of resistant cells, possibly due in part to enhanced vascularization of ccRCC. MCPIP1 overexpression partially overcame resistance to these drugs by decreasing micrometastasis and decreasing the expression of factors involved in tumorigenesis. In tumor samples from ccRCC patients, we observed a significant increase in the level of the c-Met receptor, IRAK1 and a decrease in MCPIP1 with respect to normal kidney tissue. Our results indicate separate novel mechanisms for sunitinib and sorafenib resistance, which both lead to MCPIP1 inhibition and ccRCC progression. The presented study suggests caution in the treatment of RCC with TKIs, which may lead to the unintended outcome of tumor progression.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Proteínas Proto-Oncogénicas c-met , Ribonucleasas , Factores de Transcripción , Cadherinas/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Resistencia a Antineoplásicos/genética , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Riñón/patología , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Neovascularización Patológica/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Ribonucleasas/genética , Ribonucleasas/metabolismo , Sorafenib/farmacología , Sorafenib/uso terapéutico , Sunitinib/farmacología , Sunitinib/uso terapéutico , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Eur J Pharmacol ; 922: 174804, 2022 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-35257717

RESUMEN

Metastasis is responsible for as many as 90% of cancer-associated deaths in patients. The metastatic process is a result of tumor cell migration and invasion associated with morphological changes and increased expression of several genes involved in cell migration. We have already shown that monocyte chemotactic protein-1-induced protein-1 (MCPIP1), a negative regulator of inflammatory processes, influences cell morphology, prevents the epithelial to mesenchymal transition program, and regulates metastasis in clear cell renal cell carcinoma (ccRCC). However, the mechanism by which MCPIP1 influences cell migratory potential is unknown. In this study, we investigated how MCPIP1 affects ccRCC cell migration. We showed that MCPIP1 prevents morphological transformation and drastically reduces the migration of ccRCC cells. MCPIP1 decreases the levels of Rho GTPases and reduces the phosphorylation of FAK at Tyr-397 and Tyr-861 and Src at Tyr-418. The loss of MCPIP1 RNase activity results in actin remodeling, an increase in the levels of Rho proteins and the phosphorylation of FAK on Tyr-397, which leads to Tyr-418 Src phosphorylation and an increase in migration activity. Moreover, we observed increased expression of IL-1ß in ccRCC cells and tumors lacking MCPIP1 RNase activity. Additionally, microarray analysis of tissues from patients with ccRCC revealed changes in the expression of several genes correlated with migration as tumor progression occurred. This study indicates an important role of MCPIP1 as a regulator of migratory potential in ccRCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Movimiento Celular , Quimiocina CCL2/metabolismo , Transición Epitelial-Mesenquimal/genética , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Neoplasias Renales/patología , Ribonucleasas/genética , Proteínas de Unión al GTP rho/metabolismo
3.
Oncogene ; 40(50): 6720-6735, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34657130

RESUMEN

Epithelial-mesenchymal transition (EMT) refers to the acquisition of mesenchymal properties in cells participating in tumor progression. One hallmark of EMT is the increased level of active ß-catenin, which can trigger the transcription of Wnt-specific genes responsible for the control of cell fate. We investigated how Monocyte Chemotactic Protein-1-Induced Protein-1 (MCPIP1), a negative regulator of inflammatory processes, affects EMT in a clear cell renal cell carcinoma (ccRCC) cell line, patient tumor tissues and a xenotransplant model. We showed that MCPIP1 degrades miRNAs via its RNase activity and thus protects the mRNA transcripts of negative regulators of the Wnt/ß-catenin pathway from degradation, which in turn prevents EMT. Mechanistically, the loss of MCPIP1 RNase activity led to the upregulation of miRNA-519a-3p, miRNA-519b-3p, and miRNA-520c-3p, which inhibited the expression of Wnt pathway inhibitors (SFRP4, KREMEN1, CXXC4, CSNK1A1 and ZNFR3). Thus, the level of active nuclear ß-catenin was increased, leading to increased levels of EMT inducers (SNAI1, SNAI2, ZEB1 and TWIST) and, consequently, decreased expression of E-cadherin, increased expression of mesenchymal markers, and acquisition of the mesenchymal phenotype. This study revealed that MCPIP1 may act as a tumor suppressor that prevents EMT by stabilizing Wnt inhibitors and decreasing the levels of active ß-catenin and EMT inducers.


Asunto(s)
Carcinoma de Células Renales/patología , Transición Epitelial-Mesenquimal , Factores de Transcripción Forkhead/fisiología , MicroARNs/antagonistas & inhibidores , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Proteína Wnt1/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Animales , Apoptosis , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Ratones , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Ribonucleasas/genética , Factores de Transcripción/genética , Células Tumorales Cultivadas , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
4.
Int J Mol Sci ; 22(3)2021 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525359

RESUMEN

Nonalcoholic fatty liver disease is defined as the accumulation of excessive fat in the liver in the absence of excessive alcohol consumption or any secondary cause. Although the disease generally remains asymptomatic, chronic liver inflammation leads to fibrosis, liver cirrhosis, and even to the development of hepatocellular carcinoma (HCC). Fibrosis results from epithelial-mesenchymal transition (EMT), which leads to dedifferentiation of epithelial cells into cells with a mesenchymal-like phenotype. During EMT, epithelial cells with high expression of E-cadherin, influenced by growth factors, cytokines, and inflammatory processes, undergo morphological changes via enhanced expression of, e.g., vimentin, fibronectin, and N-cadherin. An inducer of EMT and, consequently, of fibrosis development is transforming growth factor beta (TGFß), a pleiotropic cytokine associated with the progression of hepatocarcinogenesis. However, the understanding of the molecular events that direct the development of steatosis into steatohepatitis and liver fibrosis remains incomplete. Our study revealed that both prolonged exposure of hepatocarcinoma cells to fatty acids in vitro and high-fat diet in mice (20 weeks) result in inflammation. Prolonged treatment with fatty acids increased the levels of TGFß, MMP9, and ß-catenin, important EMT inducers. Moreover, the livers of mice fed a high-fat diet exhibited features of liver fibrosis with increased TGFß and IL-1 levels. Increased expression of IL-1 correlated with a decrease in monocyte chemoattractant protein-induced protein 1 (MCPIP1), a negative regulator of the inflammatory response that regulates the stability of proinflammatory transcripts encoding IL-1. Our study showed that a high-fat diet induced EMT by increasing the levels of EMT-activating transcription factors, including Zeb1, Zeb2, and Snail and changed the protein profile to a profile characteristic of the mesenchymal phenotype.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/genética , Factor de Crecimiento Transformador beta/genética , beta Catenina/genética , Animales , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/genética , Fibronectinas/genética , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Interleucina-1/genética , Interleucina-1/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Ácido Oléico/farmacología , Ribonucleasas/genética , Ribonucleasas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/metabolismo , Vimentina/genética , Vimentina/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , beta Catenina/agonistas , beta Catenina/metabolismo
5.
Eur J Pharmacol ; 888: 173591, 2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-32971087

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is the most common subtype of kidney cancer. It is highly vascularized and largely resistant to traditional chemo- and radiotherapy. Decreases in tumour suppressors and low levels of the anti-inflammatory Monocyte Chemoattractant Protein-Induced Protein 1 (MCPIP1) play important roles in the development and progression of ccRCC. MCPIP1, also called Regnase-1, possesses endonuclease activity and degrades the mRNA of proinflammatory cytokines such as IL-6, IL-1ß, IL-12 and IL-2. We previously showed that the level of MCPIP1 decreases with ccRCC progression. In this study, we explored the role of MCPIP1 in regulating the levels of tumour suppressors. We found low levels of the suppressors PTEN, RECK and TIMP3 and high levels of MMPs in patients with ccRCC who had already been shown to have low MCPIP1 expression. We demonstrated that MCPIP1 regulates the expression levels of PTEN, RECK and TIMP3 in ccRCC cell lines as well as in vivo models of ccRCC. MCPIP1 overexpression increased the expression of tumour suppressors. Moreover, we observed that the RNase activity of MCPIP1 is responsible for the modulation of apoptosis and activation of prometastatic signalling pathways. Furthermore, we found a negative correlation between high levels of IL6, a direct target of MCPIP1 RNase activity, and TIMP3 in patients, indicating that MCPIP1 and TIMP3 might collectively cause the high levels of IL6 in ccRCC patients. Taken together, our results show the importance of MCPIP1 in regulating the level of tumour suppressors and, consequently, in ccRCC development and progression.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/prevención & control , Ribonucleasas/biosíntesis , Factores de Transcripción/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Anciano , Anciano de 80 o más Años , Animales , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Ribonucleasas/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
6.
Cells ; 8(3)2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30909397

RESUMEN

C-Met tyrosine kinase receptor plays an important role under normal and pathological conditions. In tumor cells' overexpression or incorrect activation of c-Met, this leads to stimulation of proliferation, survival and increase of motile activity. This receptor is also described as a marker of cancer initiating cells. The latest research shows that the c-Met receptor has an influence on the development of resistance to targeted cancer treatment. High c-Met expression and activation in renal cell carcinomas is associated with the progression of the disease and poor survival of patients. C-Met receptor has become a therapeutic target in kidney cancer. However, the therapies used so far using c-Met tyrosine kinase inhibitors demonstrate resistance to treatment. On the other hand, the c-Met pathway may act as an alternative target pathway in tumors that are resistant to other therapies. Combination treatment together with c-Met inhibitor reduces tumor growth, vascularization and pro-metastatic behavior and results in suppressed mesenchymal phenotype and vascular endothelial growth factor (VEGF) secretion. Recently, it has been shown that the acquirement of mesenchymal phenotype or lack of cell differentiation might be related to the presence of the c-Met receptor and is consequently responsible for therapy resistance. This review presents the results from recent studies identifying c-Met as an important factor in renal carcinomas being responsible for tumor growth, progression and metastasis, indicating the role of c-Met in resistance to antitumor therapy and demonstrating the pivotal role of c-Met in supporting mesenchymal cell phenotype.


Asunto(s)
Diferenciación Celular , Resistencia a Antineoplásicos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Transición Epitelial-Mesenquimal , Humanos , Neoplasias Renales/metabolismo , Fenotipo
7.
Cancer Res ; 77(18): 4905-4920, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28716897

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is the most common type of kidney cancer and it forms highly vascularized tumors. The monocyte endoribonuclease MCPIP1 negatively regulates inflammation by degrading mRNA encoding proinflammatory cytokines, such as IL6, IL1, and IL12. MCPIP1 is also a negative regulator of NFκB and AP1 activity and it influences a broad range of miRNA activities. Here we report that MCPIP1 protein levels are decreased during renal cancer progression. In patient-derived tumors and xenografts established in NOD-SCID or nude mice, low MCPIP1 levels correlated strongly with increased proliferation, tumor outgrowth, and vascularity. MCPIP1 activity regulated secretion of VEGF, IL8, and CXCL12 leading to chemotaxis of microvascular endothelial cells, phosphorylation of VE-cadherin, and increased vascular permeability. Mechanistic investigations showed that MCPIP1 regulated ccRCC cell motility, lung metastasis, and mesenchymal phenotype by regulating key elements in the EMT signaling axis. Overall, our results illuminate how MCPIP1 serves as a key nodal point in coordinating tumor growth, angiogenesis, and metastatic spread in ccRCC. Cancer Res; 77(18); 4905-20. ©2017 AACR.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/secundario , Movimiento Celular , Neoplasias Renales/patología , Neovascularización Patológica/patología , Ribonucleasas/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Apoptosis , Carcinoma de Células Renales/irrigación sanguínea , Carcinoma de Células Renales/metabolismo , Ciclo Celular , Proliferación Celular , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Humanos , Inflamación , Neoplasias Renales/irrigación sanguínea , Neoplasias Renales/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Clasificación del Tumor , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neovascularización Patológica/metabolismo , Fosforilación , Pronóstico , Ribonucleasas/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA