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










Base de dados
Intervalo de ano de publicação
1.
Front Oncol ; 9: 54, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30828563

RESUMO

Colorectal cancer is the third major cause of cancer-related mortality worldwide. The upward trend in incidence and mortality rates, poor sensitivity to conventional therapies and a dearth of early diagnostic parameters pose a huge challenge in the management of colorectal cancer in India. Due to the high level of genetic diversity present in the Indian population, unraveling the genetic contributions toward pathogenesis is key for understanding the etiology of colorectal cancer and in reversing this trend. We have established a novel cell line, MBC02, from an Indian colorectal cancer patient and have carried out extensive molecular characterization to unravel the pathological alterations in this cell line. In-depth molecular analysis of MBC02 revealed suppression of E-cadherin expression, concomitant with overexpression of EMT related molecules, which manifested in the form of highly migratory and invasive cells. Loss of membrane-tethered E-cadherin released ß-catenin from the adherens junction resulting in its cytoplasmic and nuclear accumulation and consequently, upregulation of c-Myc. MBC02 also showed dramatic transcriptional upregulation of ß-catenin. Remarkably, we observed significantly elevated proteasome activity that perhaps co-evolved to compensate for the unnaturally high mRNA level of ß-catenin to regulate the increased protein load. In addition, there was substantial misregulation of other clinically relevant signaling pathways that have clinical relevance in the pathogenesis of colorectal cancer. Our findings pave the way toward understanding the molecular differences that could define pathogenesis in cancers originating in the Indian population.

2.
Am J Physiol Cell Physiol ; 314(6): C732-C740, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29513568

RESUMO

Coronary microvascular rarefaction, due to endothelial cell (EC) dysfunction, is one of the causes of increased morbidity and mortality in diabetes. Coronary ECs in diabetes are more apoptotic due partly to mitochondrial calcium overload. This study was designed to investigate the role of hexokinase 2 (HK2, an endogenous inhibitor of voltage-dependent anion channel) in coronary endothelial dysfunction in type 2 diabetes. We used mouse coronary ECs (MCECs) isolated from type 2 diabetic mice and human coronary ECs (HCECs) from type 2 diabetic patients to examine protein levels and mitochondrial function. ECs were more apoptotic and capillary density was lower in the left ventricle of diabetic mice than the control. MCECs from diabetic mice exhibited significant increase in mitochondrial Ca2+ concentration ([Ca2+]mito) compared with the control. Among several regulatory proteins for [Ca2+]mito, hexokinase 1 (HK1) and HK2 were significantly lower in MCECs from diabetic mice than control MCECs. We also found that the level of HK2 ubiquitination was higher in MCECs from diabetic mice than in control MCECs. In line with the data from MCECs, HCECs from diabetic patients showed lower HK2 protein levels than HCECs from nondiabetic patients. High-glucose treatment, but not high-fat treatment, significantly decreased HK2 protein levels in MCECs. HK2 overexpression in MCECs of diabetic mice not only lowered the level of [Ca2+]mito, but also reduced mitochondrial reactive oxygen species production toward the level seen in control MCECs. These data suggest that HK2 is a potential therapeutic target for coronary microvascular disease in diabetes by restoring mitochondrial function in coronary ECs.


Assuntos
Cálcio/metabolismo , Vasos Coronários/enzimologia , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Tipo 2/enzimologia , Angiopatias Diabéticas/enzimologia , Células Endoteliais/enzimologia , Hexoquinase/metabolismo , Mitocôndrias/enzimologia , Animais , Apoptose , Glicemia/metabolismo , Vasos Coronários/patologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/patologia , Células Endoteliais/patologia , Hexoquinase/genética , Humanos , Masculino , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Ubiquitinação , Regulação para Cima
3.
Oncotarget ; 7(41): 66892-66905, 2016 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-27602497

RESUMO

Glycogen Synthase Kinase-3ß (GSK3ß) is a serine/threonine kinase, known to regulate various cellular processes including proliferation, differentiation, survival, apoptosis as well as TRAIL-resistance. Thus pathways that can modulate GSK3ß axis are important targets for cancer drug development. Our earlier studies have shown that combinatorial treatment with Troglitazone (TZD) and TRAIL can induce apoptosis in TRAIL-resistant cancer cells. The current studies were undertaken to investigate whether GSK3ß pathway was modulated during this apoptosis. Our results indicated an increase in inhibitory GSK3ßSer9 phosphorylation during apoptosis, mediated via AKT. At a later time, however, TZD alone and TRAIL-TZD combination produced a dramatic reduction of GSK3ß expression, which was abolished by cycloheximide. Luciferase assays with GSK3ß-luc promoter reporter showed that TZD can effectively antagonize GSK3ß promoter activity. Since TZD is a ligand for transcription factor PPARγ and can activate AMPK, we determined their roles on antagonism of GSK3ß. Knockdown of PPARγ was unable to restore GSK3ß expression or antagonize GSK3ßSer9 phosphorylation. Although pretreatment with Compound C (pharmacological inhibitor of AMPK) partially rescued GSK3ß expression, knockdown of AMPKα1 or α2 alone or in combination were ineffective. These studies suggested a novel PPARγ-AMPK-independent mechanism of targeting GSK3ß by TZD, elucidation of which might provide newer insights to improve our understanding of TRAIL-resistance.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Neoplasias/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cromanos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Tiazolidinedionas/farmacologia , Troglitazona
4.
Am J Physiol Cell Physiol ; 305(10): C1033-40, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23986204

RESUMO

Endothelial cell (EC) dysfunction is implicated in cardiovascular diseases, including diabetes. The decrease in nitric oxide (NO) bioavailability is the hallmark of endothelial dysfunction, and it leads to attenuated vascular relaxation and atherosclerosis followed by a decrease in blood flow. In the heart, decreased coronary blood flow is responsible for insufficient oxygen supply to cardiomyocytes and, subsequently, increases the incidence of cardiac ischemia. In this study we investigate whether and how reactive oxygen species (ROS) in mitochondria contribute to coronary endothelial dysfunction in type 2 diabetic (T2D) mice. T2D was induced in mice by a high-fat diet combined with a single injection of low-dose streptozotocin. ACh-induced vascular relaxation was significantly attenuated in coronary arteries (CAs) from T2D mice compared with controls. The pharmacological approach reveals that NO-dependent, but not hyperpolarization- or prostacyclin-dependent, relaxation was decreased in CAs from T2D mice. Attenuated ACh-induced relaxation in CAs from T2D mice was restored toward control level by treatment with mitoTempol (a mitochondria-specific O2(-) scavenger). Coronary ECs isolated from T2D mice exhibited a significant increase in mitochondrial ROS concentration and decrease in SOD2 protein expression compared with coronary ECs isolated from control mice. Furthermore, protein ubiquitination of SOD2 was significantly increased in coronary ECs isolated from T2D mice. These results suggest that augmented SOD2 ubiquitination leads to the increase in mitochondrial ROS concentration in coronary ECs from T2D mice and attenuates coronary vascular relaxation in T2D mice.


Assuntos
Vasos Coronários/fisiopatologia , Diabetes Mellitus Experimental/metabolismo , Endotélio Vascular/fisiopatologia , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Acetilcolina , Animais , Meios de Cultura , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Glucose/farmacologia , Humanos , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/genética , Óxido Nítrico , Ácido Palmítico/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Superóxido Dismutase
5.
Cancer Lett ; 338(2): 292-9, 2013 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-23752066

RESUMO

Although calcineurin inhibitors (CNIs) are very useful in preventing allograft rejection, they can mediate a rapid progression of post-transplantation malignancies. The CNI cyclosporine A (CsA) can promote renal tumor growth through activation of the proto-oncogene ras and over-expression of the angiogenic cytokine VEGF; the ras activation also induces over-expression of the cytoprotective enzyme HO-1, which promotes survival of renal cancer cells. Here, we show that the natural product honokiol significantly inhibited CsA-induced and Ras-mediated survival of renal cancer cells through the down-regulations of VEGF and HO-1. Thus, honokiol treatment may help to prevent tumor-promoting effects of CsA in transplant patients.


Assuntos
Compostos de Bifenilo/farmacologia , Inibidores de Calcineurina , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/patologia , Lignanas/farmacologia , Proteínas ras/metabolismo , Apoptose/efeitos dos fármacos , Calcineurina/genética , Calcineurina/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclosporina/antagonistas & inibidores , Ciclosporina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Genes ras , Heme Oxigenase-1/biossíntese , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Fosforilação/efeitos dos fármacos , Proto-Oncogene Mas , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Transfecção , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Quinases raf/genética , Quinases raf/metabolismo , Proteínas ras/genética
6.
J Biol Chem ; 287(38): 32113-23, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22843690

RESUMO

The cytoprotective enzyme heme oxygenase-1 (HO-1) is often overexpressed in different types of cancers and promotes cancer progression. We have recently shown that the Ras-Raf-ERK pathway induces HO-1 to promote survival of renal cancer cells. Here, we examined the possible mechanisms underlying HO-1-mediated cell survival. Considering the growing evidence about the significance of apoptosis and autophagy in cancer, we tried to investigate how HO-1 controls these events to regulate survival of cancer cells. Rapamycin (RAPA) and sorafenib, two commonly used drugs for renal cancer treatment, were found to induce HO-1 expression in renal cancer cells Caki-1 and 786-O; and the apoptotic effect of these drugs was markedly enhanced upon HO-1 knockdown. Overexpression of HO-1 protected the cells from RAPA- and sorafenib-induced apoptosis and also averted drug-mediated inhibition of cell proliferation. HO-1 induced the expression of anti-apoptotic Bcl-xL and decreased the expression of autophagic proteins Beclin-1 and LC3B-II; while knockdown of HO-1 down-regulated Bcl-xL and markedly increased LC3B-II. Moreover, HO-1 promoted the association of Beclin-1 with Bcl-xL and Rubicon, a novel negative regulator of autophagy. Drug-induced dissociation of Beclin-1 from Rubicon and the induction of autophagy were also inhibited by HO-1. Together, our data signify that HO-1 is up-regulated in renal cancer cells as a survival strategy against chemotherapeutic drugs and promotes growth of tumor cells by inhibiting both apoptosis and autophagy. Thus, application of chemotherapeutic drugs along with HO-1 inhibitor may elevate therapeutic efficiency by reducing the cytoprotective effects of HO-1 and by simultaneous induction of both apoptosis and autophagy.


Assuntos
Apoptose , Autofagia , Regulação Neoplásica da Expressão Gênica , Heme Oxigenase-1/metabolismo , Neoplasias Renais/enzimologia , Proteínas Reguladoras de Apoptose/biossíntese , Proteínas Relacionadas à Autofagia , Proteína Beclina-1 , Benzenossulfonatos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Neoplasias Renais/patologia , Proteínas de Membrana/biossíntese , Proteínas Associadas aos Microtúbulos/biossíntese , Niacinamida/análogos & derivados , Compostos de Fenilureia , Protoporfirinas/química , Piridinas/farmacologia , Sirolimo/farmacologia , Sorafenibe
7.
Cancer Lett ; 321(2): 179-86, 2012 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-22343319

RESUMO

Calcineurin inhibitors (CNIs) may promote post-transplantation cancer through altered expression of cytokines and chemokines in tumor cells. We found that there is a potential cross-talk among CNI-induced signaling molecules and mTOR. Here, we utilized a murine model of post-transplantation cancer to examine the effect of a combination therapy (CNI + mTOR-inhibitor rapamycin) on allograft survival and renal cancer progression. The therapy prolonged allograft survival; and significantly attenuated CNI-induced post-transplantation cancer progression, with down-regulation of mTOR and S6-kinase phosphorylation. Also, rapamycin inhibited CNI-induced over-expression of the angiogenic cytokine VEGF, and the chemokine receptor CXCR3 and its ligands in post-transplantation tumor tissues.


Assuntos
Inibidores de Calcineurina , Quimiocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/efeitos dos fármacos , Imunossupressores/farmacologia , Neoplasias Renais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptores CXCR3/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Calcineurina/farmacologia , Linhagem Celular Tumoral , Quimioterapia Combinada , Transplante de Rim , Camundongos , Transplante , Tolerância ao Transplante , Transplante Homólogo
9.
PLoS One ; 6(8): e23919, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21886838

RESUMO

Malignancy is a major problem in patients treated with immunosuppressive agents. We have demonstrated that treatment with calcineurin inhibitors (CNIs) can induce the activation of proto-oncogenic Ras, and may promote a rapid progression of human renal cancer through the overexpression of vascular endothelial growth factor (VEGF). Interestingly, we found that CNI-induced VEGF overexpression and cancer cell proliferation was inhibited by rapamycin treatment, indicating potential involvement of the mammalian target of rapamycin (mTOR) pathway in this tumorigenic process. Here, we examined the role of mTOR pathway in mediating CNI- and Ras-induced overexpression of VEGF in human renal cancer cells (786-0 and Caki-1). We found that the knockdown of raptor (using siRNA) significantly decreased CNI-induced VEGF promoter activity as observed by promoter-luciferase assay, suggesting the role of mTOR complex1 (mTORC1) in CNI-induced VEGF transcription. It is known that mTOR becomes activated following phosphorylation of its negative regulator PRAS40, which is a part of mTORC1. We observed that CNI treatment and activation of H-Ras (through transfection of an active H-Ras plasmid) markedly increased the phosphorylation of PRAS40, and the transfection of cells using a dominant-negative plasmid of Ras, significantly decreased PRAS40 phosphorylation. Protein kinase C (PKC)-ζ and PKC-δ, which are critical intermediary signaling molecules for CNI-induced tumorigenic pathway, formed complex with PRAS40; and we found that the CNI treatment increased the complex formation between PRAS40 and PKC, particularly (PKC)-ζ. Inhibition of PKC activity using pharmacological inhibitor markedly decreased H-Ras-induced phosphorylation of PRAS40. The overexpression of PRAS40 in renal cancer cells significantly down-regulated CNI- and H-Ras-induced VEGF transcriptional activation. Finally, it was observed that CNI treatment increased the expression of phosho-PRAS40 in renal tumor tissues in vivo. Together, the phosphorylation of PRAS40 is critical for the activation of mTOR in CNI-induced VEGF overexpression and renal cancer progression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Inibidores de Calcineurina , Neoplasias Renais/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Linhagem Celular Tumoral , Humanos , Neoplasias Renais/patologia , Fosforilação , Sirolimo/farmacologia , Transcrição Gênica , Proteínas ras
10.
J Biol Chem ; 286(38): 33580-90, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21808062

RESUMO

The stress-inducible cytoprotective enzyme heme oxygenase-1 (HO-1) may play a critical role in the growth and metastasis of tumors. We demonstrated that overexpressed HO-1 promotes the survival of renal cancer cells by inhibiting cellular apoptosis; we also showed that the proto-oncogene H-Ras becomes activated in these cells under stress following treatment with immunosuppressive agents. However, it is not known if there is an association between Ras activation and HO-1 overexpression. Here, we examined if the activation of H-Ras pathway could induce HO-1, and promote the survival of renal cancer cells (786-0 and Caki-1). In co-transfection assays, using HO-1 promoter-luciferase construct, we found that the activated H-Ras, H-Ras(12V), promoted HO-1 transcriptional activation. The inhibition of endogenous H-Ras by specific dominant-negative mutant/siRNA markedly ablated the HO-1 promoter activity. Active H-Ras increased HO-1 mRNA and protein expression. Moreover, transfection with effector domain mutant constructs of active H-Ras showed that H-Ras-induced HO-1 overexpression was primarily mediated through the Raf signaling pathway. Using pharmacological inhibitor, we observed that ERK is a critical intermediary molecule for Ras-Raf-induced HO-1 expression. Activation of H-Ras and ERK promoted nuclear translocation of the transcription factor Nrf2 for its binding to the specific sequence of HO-1 promoter. The knockdown of Nrf2 significantly inhibited H-Ras-induced HO-1 transcription. Finally, by FACS analysis using Annexin-V staining, we demonstrated that the H-Ras-ERK-induced and HO-1-mediated pathway could protect renal cancer cells from apoptosis. Thus, targeting the Ras-Raf-ERK pathway for HO-1 overexpression may serve as novel therapeutics for the treatment of renal cancer.


Assuntos
Apoptose , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Heme Oxigenase-1/metabolismo , Neoplasias Renais/enzimologia , Neoplasias Renais/patologia , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Heme Oxigenase-1/genética , Humanos , Neoplasias Renais/genética , Fator 2 Relacionado a NF-E2/metabolismo , Regiões Promotoras Genéticas/genética , Transporte Proteico , Proto-Oncogene Mas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Transcrição Gênica , Ativação Transcricional
11.
Blood ; 116(11): 1980-9, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20538805

RESUMO

In these studies, we find that the vascular endothelial growth factor (VEGF) receptor KDR is expressed on subsets of mitogen-activated CD4(+) and CD8(+) T cells in vitro. We also found that KDR colocalizes with CD3 on mitogen-activated T cells in vitro and on infiltrates within rejecting human allografts in vivo. To evaluate whether VEGF and KDR mediate lymphocyte migration across endothelial cells (ECs), we used an in vitro live-time transmigration model and observed that both anti-VEGF and anti-KDR antibodies inhibit the transmigration of both CD4(+) and CD8(+) T cells across tumor necrosis factor α (TNFα)-activated, but not unactivated ECs. In addition, we found that interactions among CD4(+) or CD8(+) T cells and TNFα-activated ECs result in the induction of KDR on each T cell subset, and that KDR-expressing lymphocytes preferentially transmigrate across TNFα-activated ECs. Finally, using a humanized severe combined immunodeficient mouse model of lymphocyte trafficking, we found that KDR-expressing lymphocytes migrate into human skin in vivo, and that migration is reduced in mice treated with a blocking anti-VEGF antibody. These observations demonstrate that induced expression of KDR on subsets of T cells, and locally expressed VEGF, facilitate EC-dependent lymphocyte chemotaxis, and thus, the localization of T cells at sites of inflammation.


Assuntos
Movimento Celular/fisiologia , Linfócitos T/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Western Blotting , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Citometria de Fluxo , Prepúcio do Pênis/metabolismo , Prepúcio do Pênis/transplante , Humanos , Recém-Nascido , Masculino , Camundongos , Camundongos SCID , Microscopia de Fluorescência , Interferência de RNA , Transplante de Pele , Linfócitos T/citologia , Transplante Heterólogo , Fator de Necrose Tumoral alfa/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
12.
J Biol Chem ; 285(33): 25196-202, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20554520

RESUMO

The high incidence of cancer and its aggressive progression is a common and major problem in patients receiving immunosuppressive therapy. The calcineurin inhibitors (CNIs) may have protumorigenic effects and can promote the overexpression of several molecules inducing tumor growth. We have recently demonstrated that CNIs can mediate the transcriptional activation of the angiogenic cytokine vascular endothelial growth factor (VEGF) and promote a rapid progression of human renal cancer. Here, we investigated whether the CNI cyclosporine (CsA) and the mTOR inhibitor rapamycin (RAPA) could alter the mRNA stability of VEGF in 786-0 and Caki-1 renal cancer cells. Following actinomycin D treatment, we observed that CsA increased, whereas RAPA decreased the VEGF mRNA stability as observed by real time PCR. It is established that the mRNA-binding protein HuR may play a critical role in VEGF mRNA stability. By using HuR-siRNA, we found that the knockdown of HuR significantly decreased the CNI-induced VEGF mRNA stability. By Western blot analysis, it has been observed that CNI treatment induced the translocation of HuR from the nucleus to the cytoplasm; CNIs also induced the association between HuR and PKC-delta and promoted the phosphorylation of HuR. Finally, we found that the inhibition of PKC-delta using a dominant negative plasmid significantly decreased the CsA-induced cytoplasmic translocation of HuR and VEGF mRNA stability. Together, targeting the pathways that promote CNI-induced transcription as well as the mRNA stability of VEGF might serve as novel therapeutics for the prevention and treatment of cancer in immunosuppressed patients.


Assuntos
Imunossupressores/farmacologia , Neoplasias Renais/genética , Estabilidade de RNA/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Antígenos de Superfície/metabolismo , Western Blotting , Linhagem Celular Tumoral , Ciclosporina/farmacologia , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Humanos , Imunoprecipitação , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteína Quinase C-delta/metabolismo , Transporte Proteico/efeitos dos fármacos , Estabilidade de RNA/genética , Proteínas de Ligação a RNA/metabolismo , Sirolimo/farmacologia
13.
J Immunol ; 184(2): 545-9, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20008289

RESUMO

In this study, we find that CD45RO+ memory populations of CD4+ T lymphocytes express the vascular endothelial growth factor (VEGF) receptors KDR and Flt-1 at both the mRNA and protein levels. Furthermore, by Western blot analysis, we find that VEGF increases the phosphorylation and activation of ERK and Akt within CD4+CD45RO+ T cells. These VEGF-mediated signaling responses were inhibited by a KDR-specific small interfering RNA in a VEGF receptor-expressing Jurkat T cell line and by SU5416, a pharmacological KDR inhibitor, in CD4+CD45RO+ T cells. We also find that VEGF augments mitogen-induced production of IFN-gamma in a dose-dependent manner (p < 0.001) and significantly (p < 0.05) increases directed chemotaxis of this T cell subset. Collectively, our results for the first time define a novel function for VEGF and KDR in CD45RO+ memory T cell responses that are likely of great pathophysiological importance in immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Memória Imunológica , Interferon gama/biossíntese , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/análise , Células Cultivadas , Quimiotaxia , Humanos , Células Jurkat , Antígenos Comuns de Leucócito , RNA Mensageiro/análise , Transdução de Sinais/imunologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/análise , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia
14.
Cancer Res ; 69(23): 8902-9, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19903851

RESUMO

The development of cancer is a major problem in immunosuppressed patients, particularly after solid organ transplantation. We have recently shown that calcineurin inhibitors (CNI) used to treat transplant patients may play a critical role in the rapid progression of renal cancer. To examine the intracellular signaling events for CNI-mediated direct tumorigenic pathway(s), we studied the effect of CNI on the activation of proto-oncogenic Ras in human normal renal epithelial cells (REC) and renal cancer cells (786-0 and Caki-1). We found that CNI treatment significantly increased the level of activated GTP-bound form of Ras in these cells. In addition, CNI induced the association of Ras with one of its effector molecules, Raf, but not with Rho and phosphatidylinositol 3-kinase; CNI treatment also promoted the phosphorylation of the Raf kinase inhibitory protein and the downregulation of carabin, all of which may lead to the activation of the Ras-Raf pathway. Blockade of this pathway through either pharmacologic inhibitors or gene-specific small interfering RNA significantly inhibited CNI-mediated augmented proliferation of renal cancer cells. Finally, it was observed that CNI treatment increased the growth of human renal tumors in vivo, and the Ras-Raf pathway is significantly activated in the tumor tissues of CNI-treated mice. Together, targeting the Ras-Raf pathway may prevent the development/progression of renal cancer in CNI-treated patients.


Assuntos
Inibidores de Calcineurina , Neoplasias Renais/enzimologia , Proteínas ras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Animais , Calcineurina/metabolismo , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Ciclosporina/farmacologia , Regulação para Baixo , Proteínas Ativadoras de GTPase , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Camundongos , Camundongos Nus , Camundongos SCID , Proto-Oncogene Mas , Transdução de Sinais , Tacrolimo/farmacologia , Quinases raf/metabolismo
15.
BMC Biochem ; 10: 36, 2009 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-20043841

RESUMO

BACKGROUND: Estrogen receptor alpha (ERalpha) phosphorylation is important for estrogen-dependent transcription of ER-dependent genes, ligand-independent receptor activation and endocrine therapy response in breast cancer. However ERalpha phosphorylation at the previously identified sites does not fully account for these receptor functions. To determine if additional ERalpha phosphorylation sites exist, COS-1 cells expressing human ERalpha were labeled with [32P]H3PO4 in vivo and ERalpha tryptic phosphopeptides were isolated to identify phosphorylation sites. RESULTS: Previously uncharacterized phosphorylation sites at serines 46/47, 282, 294, and 559 were identified by manual Edman degradation and phosphoamino acid analysis and confirmed by mutagenesis and phospho-specific antibodies. Antibodies detected phosphorylation of endogenous ERalpha in MCF-7, MCF-7-LCC2, and Ishikawa cancer cell lines by immunoblot. Mutation of Ser-282 and Ser-559 to alanine (S282A, S559A) resulted in ligand independent activation of ERalpha as determined by both ERE-driven reporter gene assays and endogenous pS2 gene expression in transiently transfected HeLa cells. Mutation of Ser-46/47 or Ser-294 to alanine markedly reduced estradiol dependent reporter activation. Additionally protein kinase CK2 was identified as a kinase that phosphorylated ERalpha at S282 and S559 using motif analysis, in vitro kinase assays, and incubation of cells with CK2 kinase inhibitor. CONCLUSION: These novel ERalpha phosphorylation sites represent new means for modulation of ERalpha activity. S559 represents the first phosphorylation site identified in the extreme C-terminus (F domain) of a steroid receptor.


Assuntos
Caseína Quinase II/metabolismo , Receptor alfa de Estrogênio/metabolismo , Substituição de Aminoácidos , Animais , Anticorpos Fosfo-Específicos/imunologia , Anticorpos Fosfo-Específicos/metabolismo , Células COS , Caseína Quinase II/química , Linhagem Celular Tumoral , Chlorocebus aethiops , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/genética , Regulação da Expressão Gênica , Células HeLa , Humanos , Mutagênese Sítio-Dirigida , Fosforilação
16.
Cancer Res ; 68(14): 5689-98, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18632621

RESUMO

Cancer is an increasing and major problem after solid organ transplantation. In part, the increased cancer risk is associated with the use of immunosuppressive agents, especially calcineurin inhibitors. We propose that the effect of calcineurin inhibitors on the expression of vascular endothelial growth factor (VEGF) leads to an angiogenic milieu that favors tumor growth. Here, we used 786-0 human renal cancer cells to investigate the effect of cyclosporine (CsA) on VEGF expression. Using a full-length VEGF promoter-luciferase construct, we found that CsA markedly induced VEGF transcriptional activation through the protein kinase C (PKC) signaling pathway, specifically involving PKC zeta and PKC delta isoforms. Moreover, CsA promoted the association of PKC zeta and PKC delta with the transcription factor Sp1 as observed by immunoprecipitation assays. Using promoter deletion constructs, we found that CsA-mediated VEGF transcription was primarily Sp1 dependent. Furthermore, CsA-induced and PKC-Sp1-mediated VEGF transcriptional activation was partially inhibited by von Hippel-Lindau protein. CsA also promoted the progression of human renal tumors in vivo, wherein VEGF is overexpressed. Finally, to evaluate the in vivo significance of CsA-induced VEGF overexpression in terms of post-transplantation tumor development, we injected CT26 murine carcinoma cells (known to form angiogenic tumors) into mice with fully MHC mismatched cardiac transplants. We observed that therapeutic doses of CsA increased tumor size and VEGF mRNA expression and also enhanced tumor angiogenesis. However, coadministration of a blocking anti-VEGF antibody inhibited this CsA-mediated tumor growth. Collectively, these findings define PKC-mediated VEGF transcriptional activation as a key component in the progression of CsA-induced post-transplantation cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/induzido quimicamente , Transplante de Órgãos/efeitos adversos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Inibidores de Calcineurina , Linhagem Celular Tumoral , Ciclosporina/farmacologia , Humanos , Imunossupressores/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Transplante de Neoplasias , Neovascularização Patológica
17.
Am J Physiol Renal Physiol ; 293(4): F1222-30, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17652371

RESUMO

The turnover and repair of peritubular capillaries is essential for the maintenance of normal renal tubular structure and function. Following injury, ineffective capillary repair/angiogenesis may result in chronic disease, whereas effective repair attenuates the injury process. Thus the process of healing in the kidney is likely dependent on an intricate balance between angiogenic and anti-angiogenic factors to maintain the renal microvasculature. We investigated the role of cytoprotective heme oxygenase-1 (HO-1) in the regulation of chemokines in human renal proximal tubular epithelial cells (RPTEC). Transfection of RPTEC with a HO-1 overexpression plasmid promoted a marked induction in the mRNA expression of the anti-angiogenic chemokine CXCL-10, along with angiogenic chemokines CXCL-8 and CCL-2. Utilizing a CXCL-10 promoter luciferase construct, we observed that HO-1-induced CXCL-10 expression is regulated at the transcriptional level. However, with increases in concentrations and time intervals of HO-1 induction, there was a marked decrease in CXCL-10 expression. Using pharmacological inhibitors, we found that HO-1-induced early robust CXCL-10 transcription is mediated through the PKC signaling pathway. To evaluate the functional significance of HO-1-induced CXCL-10 release, we cultured human vascular endothelial cells in the absence and presence of culture supernatants of the HO-1 plasmid-transfected RPTEC. We found that early (24 h) supernatants of the HO-1 plasmid-transfected cells (RPTEC) inhibited endothelial cell proliferation, and this effect was blocked by addition of a CXCL-10 neutralizing antibody. Thus HO-1 can regulate the expression of the anti-angiogenic CXCL-10 and may alter a critical balance between angiogenic vs. anti-angiogenic factors that are important to maintain renal microvasculature during injury.


Assuntos
Quimiocina CXCL10/metabolismo , Células Epiteliais/metabolismo , Heme Oxigenase-1/metabolismo , Túbulos Renais Proximais/metabolismo , Proliferação de Células , Células Cultivadas , Quimiocina CXCL2/metabolismo , Células Epiteliais/citologia , Regulação da Expressão Gênica , Heme Oxigenase-1/genética , Humanos , Interleucina-8/metabolismo , Túbulos Renais Proximais/citologia , Proteína Quinase C/metabolismo , Transdução de Sinais/fisiologia , Transfecção
18.
Mol Endocrinol ; 20(5): 971-83, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16410316

RESUMO

Nuclear hormone receptors, such as the estrogen receptors (ERs), are regulated by specific kinase signaling pathways. Here, we demonstrate that the p38 MAPK stimulates both ERalpha- and ERbeta-mediated transcription in MCF-7 breast carcinoma, Ishikawa endometrial adenocarcinoma, and human embryonic kidney 293 cells. Inhibition of this potentiation using the p38 inhibitor, RWJ67657, blocked estrogen-mediated transcription and proliferation. Activated ERs promote gene expression in part through the recruitment of the p160 class of coactivators. Because no direct p38 phosphorylation sites have been determined on either ERalpha or beta, we hypothesized that p38 could target the p160 class of coactivators. We show for the first time using pharmacological and molecular techniques that the p160 coactivator glucocorticoid receptor-interacting protein 1 (GRIP1) is phosphorylated and potentiated by the p38 MAPK signaling cascade in vitro and in vivo. S736 was identified as a necessary site for p38 induction of GRIP1 transcriptional activation. The C terminus of GRIP1 was also demonstrated to contain a p38-responsive region. Taken together, these results indicate that p38 stimulates ER-mediated transcription by targeting the GRIP1 coactivator.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Coativador 2 de Receptor Nuclear/metabolismo , Transcrição Gênica , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Estrogênios/farmacologia , Humanos , Imidazóis/farmacologia , MAP Quinase Quinase 6/metabolismo , Coativador 2 de Receptor Nuclear/genética , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
19.
Front Biosci ; 10: 2346-72, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15970500

RESUMO

Estrogen is an important steroid hormone with diverse functions in different parts of the human body. The developmental and physiological role of estrogen is mediated by estrogen receptor alpha (ER-alpha) and newly identified ER-beta. Regulation of expression of various important cellular oncogenes and tumor suppressor genes is a key component of estrogen and ER action. The expression of these genes is crucial in various processes such as cell cycle progression, mammary gland development, growth factor pathways and apoptosis. A very precise and accurate control of these genes is required for normal growth and functioning of cells. Aberrant expression of these genes through elevated expression, gene amplification or mutation may lead to induction and/or progression of different cancers including estrogen-dependent breast cancers. This review briefly describes the role of different genes that are regulated by estrogen in female reproductive tissues and breast cancer.


Assuntos
Neoplasias da Mama/fisiopatologia , Estrogênios/fisiologia , Reprodução/fisiologia , Proteína BRCA1/fisiologia , Catepsina D/fisiologia , Ciclina D1/fisiologia , Feminino , Genes myc/fisiologia , Humanos , Proteínas de Membrana/fisiologia , Presenilina-2 , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Proteínas Proto-Oncogênicas c-fos/fisiologia , Receptores de Estrogênio/fisiologia , Receptores de Progesterona/fisiologia
20.
Int J Cancer ; 105(3): 377-83, 2003 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12704672

RESUMO

A male breast tumor associated antigen (MBTAA) was purified and partially characterized from human male breast tumor. Three protein peaks were obtained by DEAE-cellulose column chromatography of a crude extract of human male breast tumor tissues. Circulating antibodies against one of these peaks, MF1, which contained MBTAA, were observed in male breast cancer patients but not in normal male or male patients with carcinoma of other organs (stomach, colon, lung). The MBTAA was partially purified from MF1 by subjecting the fraction to SDS-PAGE and eluting the protein from band 3 (MB-3) and by subjecting MF1 to size exclusion-high performance liquid chromatography (SE-HPLC). The MBTAA was characterized as a glycoprotein with MW of approximately 72 kDa. It showed no immunological relatedness with TAG-72, a tumor associated antigen expressed in breast epithelial cells. A 72 kDa protein, immunologically related to MBTAA, was detected and partially purified from female breast tumor. The female breast cancer patients did not have circulating antibodies against this 72 kDa protein or MBTAA. Presence of 72 kDa glycoprotein MBTAA in MF1 and specificity of the anti-MBTAA antibodies in the sera of male breast cancer patients were further confirmed by Western blot analysis. Absence of anti-MBTAA antibodies in healthy men and in patients with other cancers suggested that expression of MBTAA may be malignancy-associated and is highly overexpressed in male breast cancer.


Assuntos
Antígenos de Neoplasias/química , Antígenos de Neoplasias/isolamento & purificação , Neoplasias da Mama Masculina/metabolismo , Anticorpos Monoclonais/metabolismo , Western Blotting , Neoplasias da Mama Masculina/imunologia , Cromatografia DEAE-Celulose , Cromatografia Líquida de Alta Pressão , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Glicoproteínas/química , Humanos , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...