RESUMO
Selenium-binding protein 1 (Selenbp1) is a 2,3,7,8-tetrechlorodibenzo-p-dioxin inducible protein whose function is yet to be comprehensively elucidated. As the highly homologous isoform, Selenbp2, is expressed at low levels in the kidney, it is worthwhile comparing wild-type C57BL mice and Selenbp1-deficient mice under dioxin-free conditions. Accordingly, we conducted a mouse metabolomics analysis under non-dioxin-treated conditions. DNA microarray analysis was performed based on observed changes in lipid metabolism-related factors. The results showed fluctuations in the expression of numerous genes. Real-time RT-PCR confirmed the decreased expression levels of the cytochrome P450 4a (Cyp4a) subfamily, known to be involved in fatty acid ω- and ω-1 hydroxylation. Furthermore, peroxisome proliferator-activated receptor-α (Pparα) and retinoid-X-receptor-α (Rxrα), which form a heterodimer with Pparα to promote gene expression, were simultaneously reduced. This indicated that reduced Cyp4a expression was mediated via decreased Pparα and Rxrα. In line with this finding, increased levels of leukotrienes and prostaglandins were detected. Conversely, decreased hydrogen peroxide levels and reduced superoxide dismutase (SOD) activity supported the suppression of the renal expression of Sod1 and Sod2 in Selenbp1-deficient mice. Therefore, we infer that ablation of Selenbp1 elicits oxidative stress caused by increased levels of superoxide anions, which alters lipid metabolism via the Pparα pathway.
Assuntos
Metabolismo dos Lipídeos/genética , Proteínas de Ligação a Selênio/metabolismo , Animais , Citocromo P-450 CYP4A/metabolismo , Expressão Gênica , Rim/patologia , Lipídeos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/genética , PPAR alfa/metabolismo , PPAR alfa/fisiologia , RNA Mensageiro/genética , Receptor X Retinoide alfa/metabolismo , Receptor X Retinoide alfa/fisiologia , Proteínas de Ligação a Selênio/genética , Fatores de Transcrição/metabolismoRESUMO
Understanding the molecular mechanisms of ultraviolet (UV) induced melanoma formation is becoming crucial with more reported cases each year. Expression of type II nuclear receptor Retinoid-X-Receptor α (RXRα) is lost during melanoma progression in humans. Here, we observed that in mice with melanocyte-specific ablation of RXRα and RXRß, melanocytes attract fewer IFN-γ secreting immune cells than in wild-type mice following acute UVR exposure, via altered expression of several chemoattractive and chemorepulsive chemokines/cytokines. Reduced IFN-γ in the microenvironment alters UVR-induced apoptosis, and due to this, the survival of surrounding dermal fibroblasts is significantly decreased in mice lacking RXRα/ß. Interestingly, post-UVR survival of the melanocytes themselves is enhanced in the absence of RXRα/ß. Loss of RXRs α/ß specifically in the melanocytes results in an endogenous shift in homeostasis of pro- and anti-apoptotic genes in these cells and enhances their survival compared to the wild type melanocytes. Therefore, RXRs modulate post-UVR survival of dermal fibroblasts in a "non-cell autonomous" manner, underscoring their role in immune surveillance, while independently mediating post-UVR melanocyte survival in a "cell autonomous" manner. Our results emphasize a novel immunomodulatory role of melanocytes in controlling survival of neighboring cell types besides controlling their own, and identifies RXRs as potential targets for therapy against UV induced melanoma.
Assuntos
Ciclo Celular/efeitos da radiação , Imunidade Inata/fisiologia , Melanócitos/fisiologia , Receptor X Retinoide alfa/fisiologia , Receptor X Retinoide beta/fisiologia , Raios Ultravioleta , Animais , Melanócitos/efeitos da radiação , Camundongos , Camundongos Transgênicos , Receptor X Retinoide alfa/genética , Receptor X Retinoide beta/genéticaRESUMO
BACKGROUND: Pulmonary arterial hypertension is a chronic lung disease associated with severe pulmonary vascular changes. A pathogenic role of oxidized lipids such as hydroxyeicosatetraenoic and hydroxyoctadecadienoic acids is well established in vascular disease. Apolipoprotein A-I mimetic peptides, including 4F, have been reported to reduce levels of these oxidized lipids and improve vascular disease. However, the role of oxidized lipids in the progression of pulmonary arterial hypertension and the therapeutic action of 4F in pulmonary arterial hypertension are not well established. METHODS AND RESULTS: We studied 2 different rodent models of pulmonary hypertension (PH): a monocrotaline rat model and a hypoxia mouse model. Plasma levels of hydroxyeicosatetraenoic and hydroxyoctadecadienoic acids were significantly elevated in PH. 4F treatment reduced these levels and rescued preexisting PH in both models. MicroRNA analysis revealed that microRNA-193-3p (miR193) was significantly downregulated in the lung tissue and serum from both patients with pulmonary arterial hypertension and rodents with PH. In vivo miR193 overexpression in the lungs rescued preexisting PH and resulted in downregulation of lipoxygenases and insulin-like growth factor-1 receptor. 4F restored PH-induced miR193 expression via transcription factor retinoid X receptor α. CONCLUSIONS: These studies establish the importance of microRNAs as downstream effectors of an apolipoprotein A-I mimetic peptide in the rescue of PH and suggest that treatment with apolipoprotein A-I mimetic peptides or miR193 may have therapeutic value.
Assuntos
Hipertensão Pulmonar/tratamento farmacológico , MicroRNAs/fisiologia , Peptídeos/uso terapêutico , Animais , Proliferação de Células , Células Cultivadas , Humanos , Ácidos Hidroxieicosatetraenoicos/administração & dosagem , Hipertensão Pulmonar/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor IGF Tipo 1/genética , Receptor X Retinoide alfa/fisiologiaRESUMO
While formation of higher-order oncogenic transcriptional complexes is critical for RARalpha fusion proteins in acute promyelocytic leukemia, the essential components and their roles in mediating transformation are still largely unknown. To this end, the present study demonstrates that homodimerization is not sufficient for RARalpha fusion-mediated transformation, which requires higher-order homotetramerization. Surprisingly, intrinsic homo-oligomeric DNA binding by the fusion proteins is also dispensable. Importantly, higher-order RXR/RARalpha fusion hetero-oligomeric complexes that aberrantly recruit transcriptional corepressors to downstream targets are essential for transformation. Intervention of RXR-dependent pathways by panRXR-agonists or RXRalpha shRNAs suppresses RARalpha fusion-mediated transformation. Taken together, these results define the oncogenic threshold for self-association and reveal the pathological significance of higher-order RARalpha fusion/RXR hetero-oligomeric complexes and their potential value as a therapeutic target.
Assuntos
Transformação Celular Neoplásica , Receptores do Ácido Retinoico/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Receptor X Retinoide alfa/fisiologia , Biopolímeros , Receptores do Ácido Retinoico/química , Receptor alfa de Ácido Retinoico , Fator de Transcrição STAT5/fisiologiaRESUMO
Mononuclear cell migration into the vascular subendothelium constitutes an early event of the atherogenic process. Because the effect of retinoid X receptor (RXR)α on arterial mononuclear leukocyte recruitment is poorly understood, this study investigated whether RXR agonists can affect this response and the underlying mechanisms involved. Decreased RXRα expression was detected after 4 h stimulation of human umbilical arterial endothelial cells with TNF-α. Interestingly, under physiological flow conditions, TNF-α-induced endothelial adhesion of human mononuclear cells was concentration-dependently inhibited by preincubation of the human umbilical arterial endothelial cells with RXR agonists such as bexarotene or 9-cis-retinoid acid. RXR agonists also prevented TNF-α-induced VCAM-1 and ICAM-1 expression, as well as endothelial growth-related oncogene-α and MCP-1 release. Suppression of RXRα expression with a small interfering RNA abrogated these responses. Furthermore, inhibition of MAPKs and NF-κB pathways were involved in these events. RXR agonist-induced antileukocyte adhesive effects seemed to be mediated via RXRα/peroxisome proliferator-activated receptor (PPAR)γ interaction, since endothelial PPARγ silencing abolished their inhibitory responses. Furthermore, RXR agonists increased RXR/PPARγ interaction, and combinations of suboptimal concentrations of both nuclear receptor ligands inhibited TNF-α-induced mononuclear leukocyte arrest by 60-65%. In vivo, bexarotene dose-dependently inhibited TNF-α-induced leukocyte adhesion to the murine cremasteric arterioles and decreased VCAM-1 and ICAM-1 expression. Therefore, these results reveal that RXR agonists can inhibit the initial inflammatory response that precedes the atherogenic process by targeting different steps of the mononuclear recruitment cascade. Thus, RXR agonists may constitute a new therapeutic tool in the control of the inflammatory process associated with cardiovascular disease.
Assuntos
Inibição de Migração Celular/imunologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , PPAR gama/metabolismo , Receptor X Retinoide alfa/agonistas , Bexaroteno , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/imunologia , Linhagem Celular , Inibição de Migração Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Endotélio Vascular/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/sangue , Leucócitos Mononucleares/efeitos dos fármacos , Microcirculação/efeitos dos fármacos , Microcirculação/imunologia , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/imunologia , PPAR gama/fisiologia , Receptor X Retinoide alfa/biossíntese , Receptor X Retinoide alfa/fisiologia , Tetra-Hidronaftalenos/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia , Artérias Umbilicais/efeitos dos fármacos , Artérias Umbilicais/imunologia , Artérias Umbilicais/patologia , Molécula 1 de Adesão de Célula Vascular/sangueRESUMO
BACKGROUND: In order for vitamin D to signal and regulate inflammatory pathways, it must bind to its receptor (VDR) which must heterodimerize with the retinoid X receptor alpha (RXRα). Although the role that vitamin D signaling plays in the development and progression of colitis, a disease characterized by excessive inflammation, has been suggested, little research has been done on determining the role that RXRα plays in acute colitis development. AIMS: This study sought to determine the effects that reduced availability of RXRα would have on the development of acute murine colitis. Expression of inflammatory markers, VDR and RXRα were investigated to determine if the reduction in expression of RXRα in RXRα(+/-) mice would result in increased inflammatory signaling and receptor downregulation as compared to their wild-type littermates. METHODS: An acute murine model of colitis, the axozymethane (AOM) and dextran sulfate sodium (DSS) model was utilized in wild-type and RXRα(+/-) mice. Gross manifestations of colitis measured included weight loss and colitis score. Immunblots and real-time PCR were performed for inflammatory markers and receptor expression. RESULTS: RXRα(+/-) mice induced with AOM/DSS colitis demonstrated increased gene expression of Snail and Snail2, transcription factors downstream of inflammatory mediators, as compared to their wild-type littermates. CONCLUSIONS: This demonstrates the importance of RXRα in regulating inflammation in acute colitis and also identifies RXRα expression as a new consideration when developing successful interventions for acute colitis due to the requirement of numerous receptors for RXRα.
Assuntos
Colite/fisiopatologia , Receptor X Retinoide alfa/fisiologia , Animais , Azoximetano/toxicidade , Carcinógenos/toxicidade , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Regulação para Baixo/fisiologia , Heterozigoto , Immunoblotting , Inflamação/fisiopatologia , Camundongos , Receptores de Calcitriol/fisiologiaRESUMO
Autoimmune glomerulonephritis is a common manifestation of systemic lupus erythematosus (SLE). In this study, we show that mice lacking macrophage expression of the heterodimeric nuclear receptors PPARγ or RXRα develop glomerulonephritis and autoantibodies to nuclear Ags, resembling the nephritis seen in SLE. These mice show deficiencies in phagocytosis and clearance of apoptotic cells, and they are unable to acquire an anti-inflammatory phenotype upon feeding of apoptotic cells, which is critical for the maintenance of self-tolerance. These results demonstrate that stimulation of PPARγ and RXRα in macrophages facilitates apoptotic cell engulfment, and they provide a potential strategy to avoid autoimmunity against dying cells and to attenuate SLE.
Assuntos
Apoptose/imunologia , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Macrófagos/imunologia , Macrófagos/patologia , PPAR gama/deficiência , Fagocitose/imunologia , Receptor X Retinoide alfa/deficiência , Animais , Anticorpos Antinucleares/biossíntese , Anticorpos Antinucleares/metabolismo , Anticorpos Antinucleares/fisiologia , Apoptose/genética , Feminino , Nefrite Lúpica/genética , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , PPAR gama/genética , PPAR gama/fisiologia , Fagocitose/genética , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/fisiologia , Tolerância a Antígenos Próprios/genética , Tolerância a Antígenos Próprios/imunologiaRESUMO
PRKAR1A (R1A)-retinoic acid receptor-alpha (R1A-RARalpha) is the sixth RARalpha-containing fusion protein in acute promyelocytic leukemia (APL). Using the murine bone-marrow retroviral transduction/transformation assay, we showed that R1A-RARalpha fusion protein could transform bone-marrow progenitor/stem cells. In gel-shift assays, R1A-RARalpha was able to bind to a panel of retinoic acid response elements both as a homodimer and as a heterodimer with RXRalpha, and demonstrated distinct DNA-binding characteristics compared with wild-type RARalpha/RXRalpha or other X-RARalpha chimeric proteins. The ratio of R1A-RARalpha to RXRalpha proteins affected the retinoic acid response element interaction pattern of R1A-RARalpha/RXRalpha complexes. Studies comparing R1A-RARalpha with R1A-RARalpha(DeltaRIIa) demonstrated that the RIIa protein interaction domain located within R1A was responsible for R1A-RARalpha homodimeric DNA binding and interaction with wild-type R1A protein. However, the RIIa domain was not required for R1A-RARalpha-mediated transformation because its deletion in R1A-RARalpha(DeltaRIIa) did not compromise its transformation capability. In contrast, introduction of point mutations within the RARalpha portion of either R1A-RARalpha or R1A-RARalpha(DeltaRIIa), previously demonstrated to eliminate RXRalpha interaction or treatment of transduced cells with RXRalpha shRNA or a RXRalpha agonist, reduced transformation capability. Thus, leukemic transformation by APL fusion protein PRKAR1A-RARalpha is critically dependent on RXRalpha, which suggests RXRalpha is a promising target for APL.
Assuntos
Transformação Celular Neoplásica/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Receptores do Ácido Retinoico/metabolismo , Receptor X Retinoide alfa/metabolismo , Animais , Proliferação de Células , Transformação Celular Neoplásica/genética , Células Cultivadas , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Regulação Leucêmica da Expressão Gênica , Células HeLa , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Ligação Proteica/fisiologia , Receptores do Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Receptor X Retinoide alfa/fisiologiaRESUMO
Naturally occurring splice variants of human constitutive androstane receptor (hCAR) exist, including hCAR-SV23 (insertion of amino acids SPTV), hCAR-SV24 (APYLT), and hCAR-SV25 (SPTV and APYLT). An extract of Ginkgo biloba was reported to activate hCAR-SV24 and the wild type (hCAR-WT). However, it is not known whether it selectively affects hCAR splice variants, how it activates hCAR isoforms, and which chemical is responsible for the effects of the extract. Therefore, we evaluated the impact of G. biloba extract on the functionality of hCAR-SV23, hCAR-SV24, hCAR-SV25, and hCAR-WT and compared it with that of phenobarbital, di-(2-ethylhexyl)phthalate (DEHP), 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime (CITCO), and 1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) in cell-based reporter gene assays. Among the hCAR splice variants investigated, only hCAR-SV23 was activated by G. biloba extract, and this required cotransfection of a retinoid X receptor α (RXRα) expression plasmid. The extract activated hCAR-SV23 to a lesser extent than hCAR-WT, but ginkgolide A, ginkgolide B, ginkgolide C, ginkgolide J, and bilobalide were not responsible for the effects of the extract. CITCO activated hCAR-SV23, hCAR-SV24, and hCAR-WT. By comparison, phenobarbital activated hCAR-WT, whereas DEHP activated hCAR-SV23, hCAR-SV24 (with exogenous RXRα supplementation), and hCAR-WT. TCPOBOP did not affect the activity of any of the isoforms. G. biloba extract and phenobarbital did not bind or recruit coactivators to the ligand-binding domains of hCAR-WT and hCAR-SV23, whereas positive results were obtained with the controls (CITCO for hCAR-WT and DEHP for hCAR-SV23). In conclusion, G. biloba extract activates hCAR in an isoform-selective manner, and hCAR-SV23, hCAR-SV24, and hCAR-WT have overlapping, but distinct, sets of ligands.
Assuntos
Ginkgo biloba/química , Ginkgolídeos/farmacologia , Extratos Vegetais/farmacologia , Receptores Citoplasmáticos e Nucleares/agonistas , Receptor Constitutivo de Androstano , Genes Reporter , Ginkgolídeos/química , Células Hep G2 , Humanos , Ligantes , Extratos Vegetais/química , Plasmídeos , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptor X Retinoide alfa/agonistas , Receptor X Retinoide alfa/fisiologia , Transfecção , Técnicas do Sistema de Duplo-HíbridoRESUMO
The preservation of body proteins is essential to guarantee their functions in organisms. Therefore, the utilization of amino acids as energy substrates is regulated by a precise fine-tuned mechanism. Recent evidence suggests that the transcription factors peroxisome proliferator-activated receptor alpha (PPARα) and hepatocyte nuclear factor 4 alpha (HNF4α) are involved in this regulatory mechanism. Thus, the aim of this study was to determine how these transcription factors interact to regulate the expression of amino acid catabolism genes. In vivo studies using PPARα-knockout mice (Pparα-null) fed different amounts of dietary protein showed that in the absence of PPARα, there was a significant increase in HNF4α abundance in the liver, which corresponded with an increase in amino acid catabolizing enzyme (AACE) expression and the generation of increased amounts of postprandial urea. Moreover, this effect was proportional to the increase in dietary protein consumed. Chromatin immunoprecipitation assays showed that HNF4α can bind to the promoter of AACE serine dehydratase (SDS), an effect that was potentiated by dietary protein in the Pparα-null mice. The mechanistic studies revealed that the presence of retinoid X receptor alpha (RXRα) is essential to repress HNF4α activity in the presence of PPARα, and this interaction accelerates HNF4α degradation via the proteasome pathway. These results showed that PPARα can downregulate liver amino acid catabolism in the presence of RXRα by inhibiting HNF4α activity.
Assuntos
Aminoácidos/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Fígado/metabolismo , PPAR alfa/fisiologia , Receptor X Retinoide alfa/fisiologia , Animais , Regulação para Baixo/genética , Células HEK293 , Células Hep G2 , Humanos , Masculino , Metabolismo/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR alfa/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteólise , Receptor X Retinoide alfa/genéticaRESUMO
Although hereditary or acquired defects in hepatobiliary transporter systems cause or predispose to cholestasis, adaptive bile acid transporter changes can counteract cholestasis by reducing hepatocellular and systemic concentrations of retained cholephiles. An important level in the regulation of adaptive bile acid transporters and overflow pathways is mediated at the transcriptional level by nuclear hormone receptors. Moreover, therapeutic approaches targeting nuclear receptors in cholestasis may stimulate these adaptive changes and open a new perspective for the treatment of cholestatic liver diseases. This review gives a comprehensive overview on bile acid transporters in the enterohepatic circulation and their adaptive changes in response to cholestasis as well as the regulatory networks underlying these adaptive mechanisms.
Assuntos
Proteínas de Transporte/fisiologia , Colestase/fisiopatologia , Glicoproteínas de Membrana/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/fisiologia , Adaptação Fisiológica , Animais , Ácidos e Sais Biliares/metabolismo , Receptor Constitutivo de Androstano , Circulação Êntero-Hepática/fisiologia , Hepatócitos/fisiologia , Humanos , Fator 2 Relacionado a NF-E2/fisiologia , PPAR alfa/agonistas , PPAR alfa/fisiologia , Receptor de Pregnano X , Receptores de Calcitriol/fisiologia , Receptores Citoplasmáticos e Nucleares/imunologia , Receptores de Glucocorticoides/fisiologia , Receptores do Ácido Retinoico/fisiologia , Receptores de Esteroides/fisiologia , Receptor alfa de Ácido Retinoico , Receptor X Retinoide alfa/fisiologiaRESUMO
Macrophages play a central role in the development of atherosclerosis through the accumulation of oxidized LDL (oxLDL). AIM (Spalpha/Api6) has previously been shown to promote macrophage survival; however, its function in atherogenesis is unknown. Here we identify AIM as a critical factor that protects macrophages from the apoptotic effects of oxidized lipids. AIM protein is induced in response to oxLDL loading and is highly expressed in foam cells within atherosclerotic lesions. Interestingly, both expression of AIM in lesions and its induction by oxidized lipids require the action of LXR/RXR heterodimers. AIM-/- macrophages are highly susceptible to oxLDL-induced apoptosis in vitro and undergo accelerated apoptosis in atherosclerotic lesions in vivo. Moreover, early atherosclerotic lesions in AIM-/-LDLR-/- double knockout mice are dramatically reduced when compared to AIM+/+LDLR-/- controls. We conclude that AIM production facilitates macrophage survival within atherosclerotic lesions and that loss of AIM decreases early lesion development by increasing macrophage apoptosis.
Assuntos
Arteriosclerose/etiologia , Macrófagos/patologia , Receptores Imunológicos/fisiologia , Animais , Apoptose , Linhagem Celular , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Lipoproteínas LDL/metabolismo , Receptores X do Fígado , Camundongos , Camundongos Knockout , Receptores Nucleares Órfãos , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores Imunológicos/deficiência , Receptores de LDL/deficiência , Receptor X Retinoide alfa/fisiologiaRESUMO
UNLABELLED: The farnesoid X receptor/retinoid X receptor-alpha (FXR/RXRalpha) complex regulates bile salt homeostasis, in part by modulating transcription of the bile salt export pump (BSEP/ABCB11) and small heterodimer partner (SHP/NR0B2). FXR is activated by bile salts, RXRalpha by the vitamin A derivative 9-cis retinoic acid (9cRA). Cholestasis is associated with vitamin A malabsorption. Therefore, we evaluated the role of vitamin A/9cRA in the expression of human and mouse bile salt export pump (hBSEP/mBsep), small heterodimer partner (hSHP/mShp), and mouse sodium-dependent taurocholate co-transporting polypeptide (mNtcp). HBSEP and hSHP transcription were analyzed in FXR/RXRalpha-transfected HepG2 cells exposed to chenodeoxycholic acid (CDCA) and/or 9cRA. BSEP promoter activity was determined by luciferase reporter assays, DNA-binding of FXR and RXRalpha by pull-down assays. Serum bile salt levels and hepatic expression of Bsep, Shp, and Ntcp were determined in vitamin A-deficient (VAD)/cholic acid (CA)-fed C57BL/6J mice. Results indicated that 9cRA strongly repressed the CDCA-induced BSEP transcription in HepG2 cells, whereas it super-induced SHP transcription; 9cRA reduced DNA-binding of FXR and RXRalpha. The 9cRA repressed the CDCA-induced BSEP promoter activity irrespective of the exact sequence of the FXR-binding site. In vivo, highest Bsep messenger RNA (mRNA), and protein expression was observed in CA-fed VAD mice. Shp transcription was highest in CA-fed vitamin A-sufficient mice. Ntcp protein expression was strongly reduced in CA-fed VAD mice, whereas mRNA levels were normal. CA-fed control and VAD mice had similarly increased serum bile salt levels. CONCLUSION: We showed that 9cRA has opposite effects on bile salt-activated transcription of FXR/RXRalpha target genes. Vitamin A deficiency in CA-fed mice leads to high BSEP expression. Clearance of serum bile salts may, however, be limited because of post-transcriptional reduction of Ntcp. The molecular effects of vitamin A supplementation during cholestasis need further analysis to predict a therapeutic effect.
Assuntos
Transportadores de Cassetes de Ligação de ATP/biossíntese , Ácido Quenodesoxicólico/farmacologia , Proteínas de Ligação a DNA/fisiologia , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptor X Retinoide alfa/fisiologia , Fatores de Transcrição/fisiologia , Tretinoína/farmacologia , Vitamina A/farmacologia , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Alitretinoína , Animais , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Ácido Cólico/farmacologia , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transportadores de Ânions Orgânicos Dependentes de Sódio/biossíntese , Receptores Citoplasmáticos e Nucleares/genética , Elementos de Resposta , Simportadores/biossíntese , Fatores de Transcrição/genética , Vitamina A/administração & dosagem , Deficiência de Vitamina A/fisiopatologiaRESUMO
The basic helix-loop-helix proteins differentiated embryo chondrocyte 1 (DEC1) and DEC2 are involved in circadian rhythm control. Because the metabolism of dietary nutrients has been linked to circadian regulation, we examined the effect of DEC1 and DEC2 on the function of the metabolite-sensing nuclear receptors, ligand-dependent transcription factors, including retinoid X receptor (RXR) and liver X receptor (LXR). Transfection assays showed that DEC1 and DEC2 repressed ligand-dependent transactivation by RXR. Knockdown of endogenous DEC1 and DEC2 expression with small interfering RNAs augmented ligand-dependent RXRalpha transactivation. DEC1 and DEC2 interacted directly with RXRalpha, and ligand addition enhanced their association. DEC1 and DEC2 modified interaction of RXRalpha with cofactor proteins. Transfection assays using DEC1 and DEC2 mutants revealed that the C-terminal region of DEC2 is required for repression and that an LXXLL motif in DEC1 and DEC2 is necessary for RXRalpha repression. DEC1 and DEC2 repressed the induction of LXR target genes, associated with the promoter of an LXR target gene, and dissociated from the promoter with ligand treatment. Knockdown of endogenous DEC1 and DEC2 enhanced the LXR target gene expression in hepatocytes. Expression of Dec1, Dec2, and Srebp-1c showed a circadian rhythm in the liver of mice, whereas that of Lxralpha, Lxrbeta, and Rxralpha was not rhythmic. DEC1 and DEC2 also repressed the transactivation of other RXR heterodimers, such as farnesoid X receptor, vitamin D receptor, and retinoic acid receptor. Thus, the repressor function of DEC1 and DEC2 may be extended to other RXR heterodimer nuclear receptors.
Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Condrócitos/fisiologia , Receptores X de Retinoides/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Linhagem Celular , Regulação para Baixo , Glutationa Transferase/biossíntese , Histona Desacetilases/fisiologia , Proteínas de Homeodomínio/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor X Retinoide alfa/biossíntese , Receptor X Retinoide alfa/fisiologia , Receptores X de Retinoides/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/fisiologia , Ativação Transcricional/fisiologia , TransfecçãoRESUMO
Variants of the human intestinal fatty acid binding protein 2 gene (FABP2) are associated with traits of the metabolic syndrome. Relevant FABP2 promoter polymorphisms c.-80_-79insT, c.-136_-132delAGTAG, c.-168_-166delAAGinsT, c.-260G>A, c.-471G>A, and c.-778G>T result in two haplotypes A and B. Activation of haplotypes by rosiglitazone stimulated PPARgamma/RXRalpha leads to 2-fold higher activity of haplotype B than A. As shown by chimeric FABP2 promoter constructs, the higher responsiveness of FABP2 haplotype B is mainly but not solely determined by polymorphism c.-471G>A. As shown by EMSA and promoter-reporter assays, Oct-1 interacts with the -471 region of FABP2 promoters, induces the activities of both FABP2 promoter haplotypes and abolishes the different activities of haplotypes induced by rosiglitazone activated PPARgamma/RXRalpha. In conclusion, our findings suggest a functional role of PPARgamma/RXRalpha and Oct-1 in the regulation of the FABP2 gene.
Assuntos
Proteínas de Ligação a Ácido Graxo/genética , Regulação da Expressão Gênica , Fator 1 de Transcrição de Octâmero/fisiologia , PPAR gama/fisiologia , Regiões Promotoras Genéticas , Receptor X Retinoide alfa/fisiologia , Sítios de Ligação , Células CACO-2 , Regulação da Expressão Gênica/efeitos dos fármacos , Haplótipos , Células HeLa , Humanos , Fator 1 de Transcrição de Octâmero/metabolismo , PPAR gama/metabolismo , Polimorfismo Genético , Ligação Proteica , Isoformas de Proteínas/genética , Receptor X Retinoide alfa/metabolismo , Rosiglitazona , Tiazolidinedionas/farmacologia , Transcrição Gênica , TransfecçãoRESUMO
Indirect regulation of transforming growth factor (TGF)-beta signaling by retinoids occurs on a long-term timescale, secondary to transcriptional events. Studies by our group show loss of retinoid X receptor (RXR) alpha results in increased TGFbeta2 in the midgestational heart, which may play a role in the cardiac defects seen in this model [S.W. Kubalak, D.R. Hutson, K.K. Scott and R.A. Shannon, Elevated transforming growth factor beta2 enhances apoptosis and contributes to abnormal outflow tract and aortic sac development in retinoic X receptor alpha knockout embryos, Development 129 (2002) 733-746.]. Acute and direct interactions between retinoid and TGFbeta signaling, however, are not clearly understood. Treatment of dispersed hearts and NIH3T3 cells for 1 h with TGFbeta and retinoids (dual treatment) resulted in increased phosphorylated Smad2 and Smad3 when compared to treatment with TGFbeta alone. Of all dual treatments, those with the RXR agonist Bexarotene, resulted in the highest level of phosphorylated Smad2, a 7-fold increase over TGFbeta2 alone. Additionally, during dual treatment phosphorylation of Smad2 occurs via the TGFbeta type I receptor but not by increased activation of the receptor. As loss of RXRalpha results in increased levels of Smad2 phosphorylation in response to TGFbeta treatment and since nuclear accumulation of phosphorylated Smad2 is decreased during dual treatment, we propose that RXRalpha directly regulates the activities of Smad2. These data show retinoid signaling influences the TGFbeta pathway in an acute and direct manner that has been unappreciated until now.
Assuntos
Núcleo Celular/metabolismo , Fibroblastos/efeitos dos fármacos , Retinoides/farmacologia , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Bexaroteno , Fibroblastos/metabolismo , Coração/fisiologia , Immunoblotting , Técnicas Imunoenzimáticas , Imunoprecipitação , Luciferases/metabolismo , Camundongos , Camundongos Knockout , Vison , Células NIH 3T3 , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Receptor X Retinoide alfa/agonistas , Receptor X Retinoide alfa/fisiologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Frações Subcelulares , Tetra-Hidronaftalenos/farmacologia , Transcrição GênicaRESUMO
To determine if changes in endometrial expression of the enzymes and receptors involved in prostaglandin (PG) synthesis and action might provide insights into the PGs involved in the initiation of decidualization, ovariectomized steroid-treated rats at the equivalent of day 5 of pseudopregnancy were given a deciduogenic stimulus and killed at various times up to 32 h thereafter. The expression of PG-endoperoxide synthases (PTGS1 and PTGS2), microsomal PGE synthases (PTGES and PTGES2), cytosolic PGE synthase (PTGES3), prostacyclin synthase (PTGIS), prostacyclin receptor, peroxisome proliferator-activated receptor delta (PPARD) and retinoid x receptor alpha (RXRA) in endometrium was assessed by semiquantitative RT-PCR, western blot analyses and immunohistochemistry. In addition, to determine which PG is involved in mediating decidualization, we compared the ability of PGE(2), stable analogues of PGI(2), L165041 (an agonist of PPARD), and docasahexanoic acid (an agonist of RXRA) to increase endometrial vascular permeability (EVP, an early event in decidualization), and decidualization when infused into the uterine horns of rats sensitized for the decidual cell reaction (DCR). EVP was assessed by uterine concentrations of Evans blue 10 h after initiation of infusions. DCR was assessed by the uterine mass 5 days after the initiation of the infusions. Because enzymes associated with the synthesis of PGE(2), including PTGS2, are up-regulated in response to a deciduogenic stimulus and because PGE(2) was more effective than the PGI(2) analogues and PPARD and RXRA agonists in increasing EVP and inducing decidualization, we suggest that PGE(2) is most likely the PG involved in the initiation of decidualization in the rat.
Assuntos
Sistema Enzimático do Citocromo P-450/genética , Implantação do Embrião/fisiologia , Endométrio/metabolismo , Oxirredutases Intramoleculares/genética , Prostaglandina-Endoperóxido Sintases/genética , Animais , Sequência de Bases , Western Blotting , Sistema Enzimático do Citocromo P-450/fisiologia , Primers do DNA/genética , Dinoprosta/farmacologia , Dinoprostona/fisiologia , Feminino , Expressão Gênica , Imuno-Histoquímica , Oxirredutases Intramoleculares/fisiologia , Dados de Sequência Molecular , PPAR delta/genética , PPAR delta/fisiologia , Gravidez , Prostaglandina-E Sintases , Prostaglandina-Endoperóxido Sintases/fisiologia , Pseudogravidez/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/fisiologia , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase ReversaRESUMO
Human ATP-binding cassette (ABC) transporter ABCC2 (cMOAT/MRP2) plays a crucial role in the hepatobiliary transport of sulfate-, glucuronide-, and glutathione-conjugated metabolites as well as a variety of amphiphilic organic anions derived from hepatic metabolism. Molecular mechanisms underlying the induction of this hepatic ABC transporter are of great interest to understand the transport-metabolism interplay in vivo. In the present study, to gain insight into the mechanism of ABCC2 induction, we tested a total of 46 structurally diverse compounds, including nuclear receptor ligands, antibiotics, bile salts, phytochemicals, and anticancer drugs. Among them, we found that LXRalpha ligands, i.e., T0901317, paxilline, and 22(R)-hydroxycholesterol, acted potently to induce the expression of ABCC2 at both mRNA and protein levels in human hepatocellular carcinoma HepG2 cells. The ABCC2 induction by T0901317 was dose- and time-dependent, where the induction pattern of ABCC2 was very similar to that of ABCG1, one of the target genes of LXRalpha. The ABCC2 induction by T0901317 was more strongly elicited when the LXRalpha gene was transiently transfected into HepG2 cells. In contrast, ABCC2 induction by T0901317 was attenuated by transient transfection of a dominant negative LXRalpha variant, suggesting that LXRalpha is involved in ABCC2 induction. Interestingly, RXR, a heterodimer partner of LXRalpha, affected the mRNA levels of ABCC2 and ABCG1 differently. ABCC2 induction by T0901317 was enhanced by RXR siRNA treatment, whereas ABCG1 induction was suppressed by the same treatment. This is the first report demonstrating that LXRalpha is potentially involved in ABCC2 induction.
Assuntos
Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Receptores Nucleares Órfãos/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Anti-Infecciosos/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Ciclosporina/farmacologia , Doxorrubicina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hidrocarbonetos Fluorados/farmacologia , Hidroxicolesteróis/farmacologia , Immunoblotting , Indóis/farmacologia , Receptores X do Fígado , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Receptores Nucleares Órfãos/agonistas , Receptores Nucleares Órfãos/antagonistas & inibidores , Receptores Nucleares Órfãos/genética , RNA Interferente Pequeno , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/fisiologia , Sirolimo/farmacologia , Sulfonamidas/farmacologiaRESUMO
Retinoic acid (RA) promotes differentiation in multiple neurogenic cell types by promoting gene reprogramming through retinoid receptors and also by inducing cytosolic signaling events. The nuclear RXR receptors are one of the main mediators of RA cellular effects, classically by joining the direct receptors of RA, the nuclear RAR receptors, in RAR/RXR dimers which act as transcription factors. Distinct RXR genes lead to RXRα, RXRß and RXRγ subtypes, but their specific roles in neuronal differentiation remain unclear. We firstly investigated both RXRs and RARs expression profiles during RA-mediated neuronal differentiation of human neuroblastoma cell line SH-SY5Y, and found varying levels of retinoid receptors transcript and protein contents along the process. In order to understand the roles of the expression of distinct RXR subtypes to RA signal transduction, we performed siRNA-mediated silencing of RXRα and RXRß during the first stages of SH-SY5Y differentiation. Our results showed that RXRα is required for RA-induced neuronal differentiation of SH-SY5Y cells, since its silencing compromised cell cycle arrest and prevented the upregulation of neuronal markers and the adoption of neuronal morphology. Besides, silencing of RXRα affected the phosphorylation of ERK1/2. By contrast, silencing of RXRß improved neurite extension and led to increased expression of tau and synaptophysin, suggesting that RXRß may negatively regulate neuronal parameters related to neurite outgrowth and function. Our results indicate distinct functions for RXR subtypes during RA-dependent neuronal differentiation and reveal new perspectives for studying such receptors as clinical targets in therapies aiming at restoring neuronal function.