Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Int J Mol Sci ; 23(21)2022 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-36362246

RESUMO

Interleukin-33 (IL-33) is an immunomodulatory cytokine which plays critical roles in tissue function and immune-mediated diseases. IL-33 is abundant within the brain and spinal cord tissues where it acts as a key cytokine to coordinate the exchange between the immune and central nervous system (CNS). In this review, we report the recent advances to our knowledge regarding the role of IL-33 and of its receptor ST2 in cerebral malaria, and in particular, we highlight the pivotal role that IL-33/ST2 signaling pathway could play in brain and cerebrospinal barriers permeability. IL-33 serum levels are significantly higher in children with severe Plasmodium falciparum malaria than children without complications or noninfected children. IL-33 levels are correlated with parasite load and strongly decrease with parasite clearance. We postulate that sequestration of infected erythrocytes or merozoites liberation from schizonts could amplify IL-33 production in endothelial cells, contributing either to malaria pathogenesis or recovery.


Assuntos
Malária Cerebral , Malária Falciparum , Criança , Humanos , Interleucina-33/metabolismo , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Células Endoteliais/metabolismo , Malária Falciparum/parasitologia , Citocinas , Plasmodium falciparum/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 40(11): 2619-2631, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32907372

RESUMO

OBJECTIVE: The occurrence of new blood vessel formation in the lungs of asthmatic patients suggests a critical role for airway endothelial cells (ECs) in the disease. IL-33 (Interleukin-33)-a cytokine abundantly expressed in human lung ECs-recently emerged as a key factor in the development of allergic diseases, including asthma. In the present study, we evaluated whether mouse and human ECs exposed to the common Dermatophagoides farinae allergen produce IL-33 and characterized the activated signaling pathways. Approach and Results: Mouse primary lung ECs were exposed in vitro to D farinae extract or rmIL-33 (recombinant murine IL-33). Both D farinae and rmIL-33 induced Il-33 transcription without increasing the IL-33 production and upregulated the expression of its receptor, as well as genes involved in angiogenesis and the regulation of immune responses. In particular, D farinae and rmIL-33 upregulated Fas/Cd95 transcript level, yet without promoting apoptosis. Inhibition of caspases involved in the Fas signaling pathway, increased IL-33 protein level in ECs, suggesting that Fas may decrease IL-33 level through caspase-8-dependent mechanisms. Our data also showed that the NF-κB (nuclear factor-κB), PI3K/Akt, and Wnt/ß-catenin pathways regulate Il-33 transcription in both mouse and human primary ECs. CONCLUSIONS: Herein, we described a new mechanism involved in the control of IL-33 production in lung ECs exposed to allergens.


Assuntos
Antígenos de Dermatophagoides/farmacologia , Células Endoteliais/efeitos dos fármacos , Interleucina-33/farmacologia , Pulmão/irrigação sanguínea , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor fas/metabolismo , Animais , Caspase 8/metabolismo , Linhagem Celular , Células Endoteliais/enzimologia , Células Endoteliais/imunologia , Ativação Enzimática , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Humanos , Interleucina-33/genética , Interleucina-33/metabolismo , Camundongos , Regulação para Cima , Via de Sinalização Wnt , Receptor fas/genética
3.
Arterioscler Thromb Vasc Biol ; 33(3): 621-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23329136

RESUMO

OBJECTIVE: Dyslipidemia contributes to endothelial dysfunction in type 2 diabetes mellitus. Fenofibrate (FF), a ligand of the peroxisome proliferator-activated receptor-α (PPARα), has beneficial effects on microvascular complications. FF may act on the endothelium by regulating vasoactive factors, including endothelin-1 (ET-1). In vitro, FF decreases ET-1 expression in human microvascular endothelial cells. We investigated the molecular mechanisms involved in the effect of FF treatment on plasma levels of ET-1 in type 2 diabetes mellitus patients. METHODS AND RESULTS: FF impaired the capacity of transforming growth factor-ß to induce ET-1 gene expression. PPARα activation by FF increased expression of the transcriptional repressor Krüppel-like factor 11 and its binding to the ET-1 gene promoter. Knockdown of Krüppel-like factor 11 expression potentiated basal and transforming growth factor-ß-stimulated ET-1 expression, suggesting that Krüppel-like factor 11 downregulates ET-1 expression. FF, in a PPARα-independent manner, and insulin enhanced glycogen synthase kinase-3ß phosphorylation thus reducing glycogen synthase kinase-3 activity that contributes to the FF-mediated reduction of ET-1 gene expression. In type 2 diabetes mellitus, improvement of flow-mediated dilatation of the brachial artery by FF was associated with a decrease in plasma ET-1. CONCLUSIONS: FF decreases ET-1 expression by a PPARα-dependent mechanism, via transcriptional induction of the Krüppel-like factor 11 repressor and by PPARα-independent actions via inhibition of glycogen synthase kinase-3 activity.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Dislipidemias/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Endotelina-1/metabolismo , Fenofibrato/uso terapêutico , Hipolipemiantes/uso terapêutico , PPAR alfa/agonistas , Proteínas Reguladoras de Apoptose , Sítios de Ligação , Artéria Braquial/efeitos dos fármacos , Artéria Braquial/fisiopatologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Regulação para Baixo , Dislipidemias/sangue , Dislipidemias/metabolismo , Dislipidemias/fisiopatologia , Células Endoteliais/metabolismo , Endotelina-1/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , PPAR alfa/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Vasodilatação/efeitos dos fármacos
4.
J Biol Chem ; 285(9): 5983-92, 2010 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-19955185

RESUMO

The nuclear receptor peroxisome proliferator-activated receptor alpha (PPARalpha) is a key regulator of genes implicated in lipid homeostasis and inflammation. PPARalpha trans-activity is enhanced by recruitment of coactivators such as SRC1 and CBP/p300 and is inhibited by binding of corepressors such as NCoR and SMRT. In addition to ligand binding, PPARalpha activity is regulated by post-translational modifications such as phosphorylation and ubiquitination. In this report, we demonstrate that hPPARalpha is SUMOylated by SUMO-1 on lysine 185 in the hinge region. The E2-conjugating enzyme Ubc9 and the SUMO E3- ligase PIASy are implicated in this process. In addition, ligand treatment decreases the SUMOylation rate of hPPARalpha. Finally, our results demonstrate that SUMO-1 modification of hPPARalpha down-regulates its trans-activity through the specific recruitment of corepressor NCoR but not SMRT leading to the differential expression of a subset of PPARalpha target genes. In conclusion, hPPARalpha SUMOylation on lysine 185 down-regulates its trans-activity through the selective recruitment of NCoR.


Assuntos
Correpressor 1 de Receptor Nuclear/metabolismo , PPAR alfa/metabolismo , Proteína SUMO-1/metabolismo , Sítios de Ligação , Linhagem Celular , Proteínas Correpressoras/metabolismo , Regulação da Expressão Gênica , Humanos , Cinética , Lisina/metabolismo , PPAR alfa/fisiologia , Transporte Proteico
5.
J Clin Invest ; 115(11): 3228-38, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16239970

RESUMO

Vascular SMC proliferation is a crucial event in occlusive cardiovascular diseases. PPARalpha is a nuclear receptor controlling lipid metabolism and inflammation, but its role in the regulation of SMC growth remains to be established. Here, we show that PPARalpha controls SMC cell-cycle progression at the G1/S transition by targeting the cyclin-dependent kinase inhibitor and tumor suppressor p16(INK4a) (p16), resulting in an inhibition of retinoblastoma protein phosphorylation. PPARalpha activates p16 gene transcription by both binding to a canonical PPAR-response element and interacting with the transcription factor Sp1 at specific proximal Sp1-binding sites of the p16 promoter. In a carotid arterial-injury mouse model, p16 deficiency results in an enhanced SMC proliferation underlying intimal hyperplasia. Moreover, PPARalpha activation inhibits SMC growth in vivo, and this effect requires p16 expression. These results identify an unexpected role for p16 in SMC cell-cycle control and demonstrate that PPARalpha inhibits SMC proliferation through p16. Thus, the PPARalpha/p16 pathway may be a potential pharmacological target for the prevention of cardiovascular occlusive complications of atherosclerosis.


Assuntos
Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidores do Crescimento/fisiologia , Músculo Liso Vascular/patologia , PPAR alfa/fisiologia , Túnica Íntima/patologia , Regulação para Cima , Animais , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Células Cultivadas , Quinase 4 Dependente de Ciclina/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Regulação para Baixo/genética , Fase G1/genética , Inibidores do Crescimento/deficiência , Inibidores do Crescimento/genética , Células HeLa , Humanos , Hiperplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , PPAR alfa/deficiência , PPAR alfa/genética , Fosforilação , Proteína do Retinoblastoma/metabolismo , Fase S/genética , Transcrição Gênica/fisiologia , Túnica Íntima/citologia , Regulação para Cima/genética
6.
Circ Res ; 98(3): 361-9, 2006 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-16397146

RESUMO

Statins are inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase used in the prevention of cardiovascular disease (CVD). In addition to their cholesterol-lowering activities, statins exert pleiotropic antiinflammatory effects, which might contribute to their beneficial effects not only on CVD but also on lipid-unrelated immune and inflammatory diseases, such as rheumatoid arthritis, asthma, stroke, and transplant rejection. However, the molecular mechanisms involved in these antiinflammatory properties of statins are unresolved. Here we show that the peroxisome proliferator-activated receptor (PPAR) alpha mediates antiinflammatory effects of simvastatin in vivo in models of acute inflammation. The inhibitory effects of statins on lipopolysaccharide-induced inflammatory response genes were abolished in PPARalpha-deficient macrophages and neutrophils. Moreover, simvastatin inhibited PPARalpha phosphorylation by lipopolysaccharide-activated protein kinase C (PKC) alpha. A constitutive active form of PKCalpha inhibited nuclear factor kappaB transrepression by PPARalpha whereas simvastatin enhanced transrepression activity of wild-type PPARalpha, but not of PPARalpha mutated in its PKC phosphorylation sites. These data indicate that the acute antiinflammatory effect of simvastatin occurs via PPARalpha by a mechanism involving inhibition of PKCalpha inactivation of PPARalpha transrepression activity.


Assuntos
Edema/tratamento farmacológico , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , PPAR alfa/fisiologia , Proteína Quinase C/antagonistas & inibidores , Transdução de Sinais/fisiologia , Animais , Extremidades/irrigação sanguínea , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR alfa/deficiência , PPAR alfa/genética , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/farmacologia
7.
Expert Opin Emerg Drugs ; 11(3): 379-401, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16939380

RESUMO

More than 70% of patients with Type 2 diabetes mellitus (T2DM) die because of cardiovascular diseases. Current therapeutic strategies are based on separate treatment of insulin resistance and dyslipidaemia. Development of drugs with multimodal activities should improve management of the global cardiovascular risk of T2DM patients and result in better patient compliance. New therapeutic strategies are aimed at targeting the entire spectrum of dysfunctioning organs, cells and regulatory pathways implicated in the pathogenesis of T2DM, dyslipidaemia and atherosclerosis. PPAR family members play major roles in the regulation of lipid metabolism, glucose homeostasis and inflammatory processes, making these transcription factors ideal targets for therapeutic strategies against these diseases. This review discusses why PPARs and development of novel selective PPAR modulators, dual and pan PPAR agonists constitute promising approaches for the treatment of diabetes, dyslipidaemia and atherosclerosis.


Assuntos
Aterosclerose/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Animais , Aterosclerose/sangue , Aterosclerose/complicações , Fármacos Cardiovasculares/farmacologia , Fármacos Cardiovasculares/uso terapêutico , Terapia Combinada/métodos , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Quimioterapia Combinada , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Receptores Ativados por Proliferador de Peroxissomo/sangue
8.
Mol Endocrinol ; 18(8): 1906-18, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15131257

RESUMO

Peroxisome proliferator-activated receptor (PPAR) alpha is a nuclear receptor implicated in several physiological processes such as lipid and lipoprotein metabolism, glucose homeostasis, and the inflammatory response. PPARalpha is activated by natural fatty acids and synthetic compounds like fibrates. PPARalpha activity has been shown to be modulated by its phosphorylation status. PPARalpha is phosphorylated by kinases such as the MAPKs and cAMP-activated protein kinase A. In this report, we show that protein kinase C (PKC) inhibition impairs ligand-activated PPARalpha transcriptional activity. Furthermore, PKC inhibition decreases PPARalpha ligand-induction of its target genes including PPARalpha itself and carnitine palmitoyltransferase I. By contrast, PKC inhibition enhances PPARalpha transrepression properties as demonstrated using the fibrinogen-beta gene as model system. Finally, PKC inhibition decreases PPARalpha phosphorylation activity of hepatocyte cell extracts. In addition, PPARalpha purified protein is phosphorylated in vitro by recombinant PKCalpha and betaII. The replacement of serines 179 and 230 by alanine residues reduces the phosphorylation of the PPARalpha protein. The PPARalpha S179A-S230A protein displays an impaired ligand-induced transactivation activity and an enhanced trans-repression activity. Altogether, our data indicate that the PKC signaling pathway acts as a molecular switch dissociating the transactivation and transrepression functions of PPARalpha, which involved phosphorylation of serines 179 and 230.


Assuntos
PPAR alfa/metabolismo , Proteína Quinase C/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Ativação Transcricional , Alanina/genética , Alanina/metabolismo , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Fibrinogênio/genética , Expressão Gênica , Humanos , Ligantes , Mutagênese Sítio-Dirigida/genética , PPAR alfa/química , PPAR alfa/genética , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Serina/genética , Serina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
9.
J Steroid Biochem Mol Biol ; 85(2-5): 267-73, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12943712

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors belonging to the nuclear receptor superfamily. Three PPARs isoforms have been characterized: PPARalpha, beta/delta and gamma. As other nuclear receptors, the PPARs are organized in distinct functional domains: A/B, C or DNA binding domain (DBD), D, E or ligand binding domain (LBD) and F. The A/B domain contains the activation function 1 (AF-1) which is transcriptionally active in absence of ligands. The DBD and the LBD of the PPARs determine the specificity of promoter DNA sequence recognition and ligand recognition, respectively. An activation function 2 (AF-2) is contained in the E domain, which mediates the ligand-dependent activation of the receptor. The transcriptional activity of the PPARs is regulated by post-translational modifications, such as phosphorylation and ubiquitination. Phosphorylation of PPARs is controlled by environmental factors activating different kinase pathways leading to the modulation of their activities. PPARs degradation by the ubiquitin-proteasome system modulates the intensity of the ligand response by controlling the level of PPAR proteins in the cells. PPARs also control the expression of genes implicated in the inflammatory response via negative interference with different inflammatory pathways, such as NFkappaB, AP-1, C/EBP beta, STAT-1 and NFAT. As such, PPARs influence inflammatory cytokine production and cell recruitment to the inflammatory sites. A better understanding of the mechanism of action of PPARs could improve the design of more specific and more efficient novel drugs. Molecules with dissociated effects could be useful for the treatment of lipid disorders or inflammation.


Assuntos
Regulação da Expressão Gênica , Inflamação/fisiopatologia , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Animais , Regulação da Expressão Gênica/fisiologia , Humanos , Fosforilação , Processamento de Proteína Pós-Traducional , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo
10.
Curr Opin Allergy Clin Immunol ; 13(5): 495-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23945177

RESUMO

PURPOSE OF REVIEW: To examine the recent, most relevant genetic and epigenetic modifications of the epithelial barrier in response to the environmental factors, including allergens, viruses, and pollutants, susceptible to participate to asthma. RECENT FINDINGS: IL-33 and TSLP gene polymorphisms are found in almost all asthma studies. Recent data have highlighted a new population of innate lymphoid cells, activated by these two cytokines, and mediating type 2 innate immunity dependent asthma. Gene variants of innate pattern recognition receptors associated with asthma have been evidenced in early viral infected high-risk birth cohorts, as well as polymorphisms in pathways involved in type I interferon (IFN) production, giving further insight into the role of viruses in asthma development. Novel epigenetic mechanisms have been evidenced in asthma and in response to the environmental pollutants, and point out genes like TSLP, which may link environmental pollution and asthma. SUMMARY: Genetic data support the role of a specific set of epithelial-derived proTh2 cytokines, including IL-33 and TSLP, as well as the role of decreased type I IFN in virus-induced impaired epithelial barrier. Epigenetic modifications of epithelial genes are promising mechanisms that warrant further investigation.


Assuntos
Asma/genética , Citocinas/genética , Interleucinas/genética , Receptores de Reconhecimento de Padrão/genética , Mucosa Respiratória/imunologia , Alérgenos/imunologia , Animais , Asma/etiologia , Exposição Ambiental/efeitos adversos , Epigênese Genética , Interação Gene-Ambiente , Humanos , Imunidade Inata/genética , Interferon Tipo I/imunologia , Interleucina-33 , Polimorfismo Genético , Equilíbrio Th1-Th2 , Linfopoietina do Estroma do Timo
11.
Methods Mol Biol ; 952: 145-61, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23100230

RESUMO

Studies on the regulation of nuclear receptors, such as the peroxisome proliferator-activated receptors (PPARs), are important to enhance our understanding of their molecular, cellular, and physiological behavior. A decade ago, it was shown that the SUMOylation pathway plays a very important role in the regulation of transcription factor activity. The SUMOylation process involves the covalent binding of SUMO protein to the target protein. However, experimental procedures to demonstrate that low-expressed proteins, such as PPARs, are SUMOylated, remain tricky, and require specific optimization for each protein.Here, we provide a simple and useful experimental method to investigate the SUMOylation of PPARs in a cellular context. The procedure for studying SUMOylation in living cells is based on the purification under denaturating conditions of total SUMOylated proteins followed by the specific detection of the PPAR proteins. For that purpose, cells are transfected with both 6xHistidine-tagged SUMO and PPAR expression vectors. Since the polyHistidine tag binds to nickel cationic ion-linked agarose matrix (Ni-NTA matrix), His-tagged SUMO proteins covalently linked to the protein substrate can be specifically precipitated and separated from the unSUMOylated proteins. The SUMO-modified PPAR proteins can subsequently be visualized by western blotting using anti-PPAR antibodies. Many questions relative to the regulation of PPAR SUMOylation can be appropriately addressed by adapting this protocol.


Assuntos
Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Sumoilação , Western Blotting , Linhagem Celular Tumoral , Biologia Computacional , Humanos , Receptores Ativados por Proliferador de Peroxissomo/genética , Transfecção
12.
Biochem Biophys Res Commun ; 319(2): 663-70, 2004 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-15178457

RESUMO

Peroxisome proliferator-activated receptor alpha (PPARalpha) is a ligand-activated transcription factor. PPARalpha regulates lipid and glucose metabolism and controls the inflammatory response. Recently, we have shown that PPARalpha is a short-lived protein degraded by the ubiquitin-proteasome system. In this study, we have analysed the effects of interaction with RXRalpha, CBP, and N-CoR and also the implication of phosphorylation on ubiquitination and stability of PPARalpha. Our results show that interaction of PPARalpha with RXRalpha or CBP leads to an increase in the turnover of the protein. In contrast, interaction with the corepressor N-CoR, which inhibits its transcriptional activity, leads to a stabilization of the protein. Interestingly, treatment of cells with an inhibitor of Ser/Thr phosphatases known to lead to hyperphosphorylation of PPARalpha induces its transcriptional activity which is accompanied by a stabilization of the protein. These data indicate that heterodimerization, recruitment of cofactors, and post-translational modifications can modulate the stability of PPARalpha.


Assuntos
Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Animais , Células COS , Dimerização , Ligantes , Fosforilação , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores do Ácido Retinoico/metabolismo , Receptores X de Retinoides , Fatores de Transcrição/metabolismo , Ubiquitina/metabolismo
13.
J Biol Chem ; 277(40): 37254-9, 2002 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-12118000

RESUMO

Peroxisome proliferator activated-receptor alpha (PPARalpha) is a ligand-activated transcription factor belonging to the nuclear receptor family. PPARalpha is implicated in the regulation of lipid and glucose metabolism and in the control of inflammatory response. Recently, it has been demonstrated that a number of nuclear receptors are degraded by the ubiquitin-proteasome pathway. Since PPARalpha exhibits a circadian expression rhythm and since PPARalpha is rapidly regulated under certain pathophysiological conditions such as the acute phase inflammatory response, we hypothesized that PPARalpha protein levels must be under tight control. Here, we studied the mechanisms controlling PPARalpha protein levels and their consequences on the transcriptional control of PPARalpha target genes. Using pulse-chase experiments, it is shown that PPARalpha is a short-lived protein and that addition of its ligands stabilizes this nuclear receptor. By transient cotransfection experiments using expression vectors for PPARalpha and hemagglutinin-tagged ubiquitin, it is demonstrated that PPARalpha protein is ubiquitinated and that its ligands decrease the ubiquitination of this nuclear receptor, thus providing a mechanism for the ligand-dependent stabilization observed in pulse-chase experiments. In addition, treatment with MG132, a selective proteasome inhibitor, increases the level of ubiquitinated PPARalpha and inhibits its degradation in transfected cells. Furthermore, MG132 treatment enhances the level of endogenous PPARalpha in HepG2 cells. Finally, transient transfection and quantitative reverse transcription-PCR show that inhibition of PPARalpha degradation increases its transcriptional activation and expression of target genes such as apoA-II and fatty acid transport protein (FATP). Taken together, these data demonstrate that PPARalpha is degraded by the ubiquitin-proteasome system in a ligand-dependent manner. Regulation of its degradation provides a novel regulatory mechanism of transcriptional activity of this nuclear receptor.


Assuntos
Cisteína Endopeptidases/metabolismo , Regulação da Expressão Gênica , Complexos Multienzimáticos/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ubiquitina/metabolismo , Animais , Células COS , Núcleo Celular/fisiologia , Chlorocebus aethiops , Humanos , Ligantes , Complexo de Endopeptidases do Proteassoma , Biossíntese de Proteínas , RNA Mensageiro/genética , Proteínas Recombinantes/metabolismo , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA