Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Cell Physiol ; 233(2): 968-978, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28383766

RESUMO

TWEAK regulates multiple physio-pathological processes in fibroblasts such as fibrosis. It also induces migration and invasion in tumors and it can activate p38 MAPK in various cell types. Moreover, p38α MAPK promotes migration and invasion in several cancer cells types and in mouse embryonic fibroblasts (MEFs). However, it remains unknown if TWEAK could promote migration in fibroblasts and whether p38α MAPK might play a role. Our results reveal that TWEAK activates ERKs, Akt, and p38α/ß MAPKs and reduces secreted Fibulin 3 in MEFs. TWEAK also increases migration and invasion in wt and p38α deficient MEFs, which indicates that p38α MAPK is not required to mediate these effects. In contrast, ERKs inhibition significantly decreases TWEAK-induced migration and Fibulin 3 knock-down mimics TWEAK effect. These results indicate that both ERKs activation and Fibulin 3 down-regulation would contribute to mediate TWEAK pro-migratory effect. In fact, the additional regulation of ERKs and/or p38ß as a consequence of Fibulin 3 decrease might be also involved in the pro-migratory effect of TWEAK in MEFs. In conclusion, our studies uncover novel mechanisms by which TWEAK would favor tissue repair by promoting fibroblasts migration.


Assuntos
Movimento Celular , Citocina TWEAK/metabolismo , Proteínas da Matriz Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/enzimologia , Animais , Células Cultivadas , Citocina TWEAK/genética , Regulação para Baixo , Ativação Enzimática , Proteínas da Matriz Extracelular/genética , Camundongos , Proteína Quinase 11 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fatores de Tempo
2.
J Biol Chem ; 290(7): 4383-97, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25548290

RESUMO

p38 MAPKs regulate migration and invasion. However, the mechanisms involved are only partially known. We had previously identified fibulin 3, which plays a role in migration, invasion, and tumorigenesis, as a gene regulated by p38α. We have characterized in detail how p38 MAPK regulates fibulin 3 expression and its role. We describe here for the first time that p38α, p38γ, and p38δ down-regulate fibulin 3 expression. p38α has a stronger effect, and it does so through hypermethylation of CpG sites in the regulatory sequences of the gene. This would be mediated by the DNA methylase, DNMT3A, which is down-regulated in cells lacking p38α, but once re-introduced represses Fibulin 3 expression. p38α through HuR stabilizes dnmt3a mRNA leading to an increase in DNMT3A protein levels. Moreover, by knocking-down fibulin 3, we have found that Fibulin 3 inhibits migration and invasion in MEFs by mechanisms involving p38α/ß inhibition. Hence, p38α pro-migratory/invasive effect might be, at least in part, mediated by fibulin 3 down-regulation in MEFs. In contrast, in HCT116 cells, Fibulin 3 promotes migration and invasion through a mechanism dependent on p38α and/or p38ß activation. Furthermore, Fibulin 3 promotes in vitro and in vivo tumor growth of HCT116 cells through a mechanism dependent on p38α, which surprisingly acts as a potent inducer of tumor growth. At the same time, p38α limits fibulin 3 expression, which might represent a negative feed-back loop.


Assuntos
Movimento Celular , Neoplasias do Colo/patologia , Metilação de DNA , Embrião de Mamíferos/metabolismo , Proteínas da Matriz Extracelular/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Animais , Western Blotting , Adesão Celular , Proliferação de Células , Células Cultivadas , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Regulação para Baixo , Embrião de Mamíferos/citologia , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/citologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Nus , Invasividade Neoplásica , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Elementos de Resposta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cicatrização , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Am J Physiol Endocrinol Metab ; 305(1): E101-12, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23651848

RESUMO

Visceral fat is strongly associated with insulin resistance. Obesity-associated adipose tissue inflammation and inflammatory cytokine production are considered key mediators of insulin signaling inhibition. TWEAK is a relatively new member of the TNF cytokine superfamily, which can exist as full length membrane-associated (mTWEAK) and soluble (sTWEAK) isoforms. Although TWEAK has been shown to have important functions in chronic inflammatory diseases its physiological role in adipose tissue remains unresolved. In this study, we explore the molecular mechanisms involved in the modulation of TNF-α-induced effects on insulin sensitivity by sTWEAK in a human visceral adipose cell line and also in primary human adipocytes obtained from visceral fat depots. Our data reveal that sTWEAK ameliorates TNF-α-induced insulin resistance on glucose uptake, GLUT4 translocation and insulin signaling without affecting other metabolic effects of TNF-α such as lipolysis or apoptotis. Co-immunoprecipitation experiments in adipose cells revealed that pretreatment with sTWEAK specifically inhibits TRAF2 association with TNFR1, but not with TNFR2, which mediates insulin resistance. However, sTWEAK does not affect other downstream molecules activated by TNF-α, such as TAK1. Rather, sTWEAK abolishes the stimulatory effect of TNF-α on JNK1/2, which is directly involved in the development of insulin resistance. This is associated with an increase in PP2A activity upon sTWEAK treatment. Silencing of the PP2A catalytic subunit gene overcomes the dephosphorylation effect of sTWEAK on JNK1/2, pointing to PP2A as a relevant mediator of sTWEAK-induced JNK inactivation. Overall, our data reveal a protective role of TWEAK in glucose homeostasis and identify PP2A as a new driver in the modulation of TNF-α signaling by sTWEAK.


Assuntos
Resistência à Insulina/fisiologia , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/metabolismo , Proteína Fosfatase 2/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Necrose Tumoral/metabolismo , Linhagem Celular , Citocina TWEAK , Glucose/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Humanos , Gordura Intra-Abdominal/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Obesidade/metabolismo , Cultura Primária de Células , Solubilidade , Fator 2 Associado a Receptor de TNF/metabolismo
4.
IUBMB Life ; 65(7): 572-83, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23671028

RESUMO

Recent advances have demonstrated that the adipose tissue plays a central role in regulating overall energy balance. Obesity results from a chronic deregulation of energy balance, with energy intake exceeding energy expenditure. Recently, new mechanisms that control the obesity phenotype such as the equilibrium between white and brown adipose tissue function has been identified. In this context, it is becoming increasingly clear that in addition to cellular growth, AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) also regulate lipid metabolism and adipogenesis. Here, we review recent advances in the understanding of the molecular mechanisms involved in white and brown differentiation programs focusing on AMPK and mTOR signaling pathways, which may play differential roles in white adipose tissue and brown adipose tissue development. In view of the worldwide epidemic of obesity and its associated metabolic disorders such as insulin resistance and type 2 diabetes, targeting these kinases may represent a potential approach for reducing adiposity and improving obesity-related diseases. © 2013 IUBMB Life, 65(7):572-583, 2013.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético , Obesidade/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Adipócitos/metabolismo , Adipogenia/genética , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Diferenciação Celular/genética , Diabetes Mellitus Tipo 2/patologia , Humanos , Obesidade/patologia , Serina-Treonina Quinases TOR/genética
5.
Curr Genomics ; 10(1): 26-34, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19721808

RESUMO

The infection by mucosal human papillomavirus (HPV) is causally associated with tumor development in cervix and oropharynx. The mechanisms responsible for this oncogenic potential are mainly due to the product activities of two early viral oncogenes: E6 and E7. Although a large number of cellular targets have been described for both oncoproteins, the interaction with tumor suppressors p53 and retinoblastoma protein (pRb) emerged as the key functional activities. E6 degrades tumor suppressor p53, thus inhibiting p53-dependent functions, whereas E7 binds and degrades pRb, allowing the transcription of E2F-dependent genes. Since these two tumor suppressors exert their actions through transcriptional modulation, functional genomics has provided a large body of data that reflects the altered gene expression of HPVinfected cells or tissues. Here we will review the similarities and differences of these findings, and we also compare them with those obtained with transgenic mouse models bearing the deletion of some of the viral oncogene targets. The comparative analysis supports molecular evidences about the role of oncogenes E6 and E7 in the interference with the mentioned cellular functions, and also suggests that the mentioned transgenic mice can be used as models for HPV-associated diseases such as human cervical, oropharynx, and skin carcinomas.

6.
Oncotarget ; 7(29): 45060-45078, 2016 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-27286263

RESUMO

C3G, a Guanine nucleotide Exchange Factor (GEF) for Rap1 and R-Ras, has been shown to play important roles in development and cancer. Previous studies determined that C3G regulates cell death through down-regulation of p38α MAPK activity. Here, we found that C3G knock-down in MEFs and HCT116 cells promotes migration and invasion through Rap1-mediated p38α hyper-activation. These effects of C3G were inhibited by Rap1 knock-down or inactivation. The enhanced migration observed in C3G depleted HCT116 cells was associated with reduction in E-cadherin expression, internalization of ZO-1, actin cytoskeleton reorganization and decreased adhesion. We also found that matrix metalloproteases MMP2 and MMP9 are involved in the pro-invasive effect of C3G down-regulation. Additionally, our studies revealed that both C3G and p38α collaborate to promote growth of HCT116 cells in vitro and in vivo, possibly by enhancing cell survival. In fact, knocking-down C3G or p38α individually or together promoted cell death in vitro, although only the double C3G-p38α silencing was able to increase cell death within tumors. Notably, we found that the pro-tumorigenic function of C3G does not depend on p38α or Rap1 activation. Altogether, our studies uncover novel mechanisms by which C3G controls key aspects of tumorigenesis.


Assuntos
Carcinogênese/metabolismo , Neoplasias Colorretais/patologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Movimento Celular/fisiologia , Neoplasias Colorretais/metabolismo , Ativação Enzimática/fisiologia , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Células HCT116 , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia
7.
PLoS One ; 10(6): e0129644, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26068931

RESUMO

OBJECTIVE: Evidence from mouse models suggests that zinc-α2-glycoprotein (ZAG) is a novel anti-obesity adipokine. In humans, however, data are controversial and its physiological role in adipose tissue (AT) remains unknown. Here we explored the molecular mechanisms by which ZAG regulates carbohydrate metabolism in human adipocytes. METHODS: ZAG action on glucose uptake and insulin action was analyzed. ß1 and ß2-adrenoreceptor (AR) antagonists and siRNA targeting PP2A phosphatase were used to examine the mechanisms by which ZAG modulates insulin sensitivity. Plasma levels of ZAG were measured in a lean patient cohort stratified for HOMA-IR. RESULTS: ZAG treatment increased basal glucose uptake, correlating with an increase in GLUT expression, but induced insulin resistance in adipocytes. Pretreatment of adipocytes with propranolol and a specific ß1-AR antagonist demonstrated that ZAG effects on basal glucose uptake and GLUT4 expression are mediated via ß1-AR, whereas inhibition of insulin action is dependent on ß2-AR activation. ZAG treatment correlated with an increase in PP2A activity. Silencing of the PP2A catalytic subunit abrogated the negative effect of ZAG on insulin-stimulated AKT phosphorylation and glucose uptake but not on GLUT4 expression and basal glucose uptake. ZAG circulating levels were unchanged in a lean patient cohort stratified for HOMA-IR. Neither glucose nor insulin was associated with plasma ZAG. CONCLUSIONS: ZAG inhibits insulin-induced glucose uptake in human adipocytes by impairing insulin signaling at the level of AKT in a ß2-AR- and PP2A-dependent manner.


Assuntos
Adipócitos/metabolismo , Insulina/metabolismo , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Plasma Seminal/metabolismo , Adipócitos/efeitos dos fármacos , Adulto , Índice de Massa Corporal , Células Cultivadas , Ativação Enzimática , Feminino , Glucose/metabolismo , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Humanos , Resistência à Insulina , Masculino , Pessoa de Meia-Idade , Proteína Fosfatase 2/genética , Proteínas de Plasma Seminal/sangue , Proteínas de Plasma Seminal/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transcriptoma , Glicoproteína Zn-alfa-2
8.
J Clin Endocrinol Metab ; 98(8): E1323-33, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23783093

RESUMO

CONTEXT: Soluble TNF-like weak inducer of apoptosis (sTWEAK) is generated by the intracellular proteolytic cleavage of full-length membrane-bound TNF-like weak inducer of apoptosis (mTWEAK). sTWEAK levels are reduced in diseases with an inflammatory component. Additionally, sTWEAK hampers TNFα activity in human cells. OBJECTIVES: The objectives of the study were as follows: 1) to determine circulating sTWEAK in severe obesity and after bariatric surgery; 2) to study m/sTWEAK and its receptor fibroblast growth factor-inducible 14 (Fn14) protein expression in sc adipose tissue (SAT) of severely obese subjects, in SAT stromal vascular fraction (SVF), and isolated adipocytes and in human monocyte-derived macrophages; and 3) to explore, on human adipocytes, the sTWEAK effect on TNFα proinflammatory activity. DESIGN: sTWEAK levels were measured in cohort 1: severely obese subjects (n = 23) and a control group (n = 35); and in cohort 2: (n = 23) severely obese subjects before and after surgery. The m/sTWEAK and Fn14 expressions were determined in SAT biopsies, SVF, and isolated adipocytes from severely obese and control subjects and in human monocyte-derived macrophages. In human primary cultured adipocytes, sTWEAK pretreated and TNFα challenged, IL-6, IL-8, and adiponectin protein and gene expressions were determined and nuclear factor-κ B and MAPK signaling analyzed. RESULTS: sTWEAK levels were reduced in severely obese subjects. After surgery, sTWEAK levels rose in 69% of patients. mTWEAK protein expression was increased in SAT and SVF of severely obese subjects, whereas Fn14 was up-regulated in isolated adipocytes. M2 human monocyte-derived macrophages overexpress mTWEAK. In human adipocytes, sTWEAK down-regulates TNFα cytokine production by hampering TNFα intracellular signaling events. CONCLUSION: The decrease of sTWEAK in severely obese patients may favor the proinflammatory activity elicited by TNFα.


Assuntos
Adipócitos/imunologia , Cirurgia Bariátrica , Citocinas/biossíntese , Obesidade/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Fatores de Necrose Tumoral/sangue , Adulto , Índice de Massa Corporal , Citocina TWEAK , Ácidos Graxos não Esterificados/sangue , Feminino , Humanos , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Obesidade/cirurgia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA