PPRE

PPRE
  • 文章类型: Journal Article
    在哺乳动物细胞中,两个细胞器,线粒体和过氧化物酶体,分享降解脂肪酸链的能力。尽管每个细胞器都有自己的脂肪酸β-氧化途径,一个独特的线粒体系统供给ATP合成的氧化磷酸化途径。同时,过氧化物酶体β-氧化途径参与细胞产热。1965年的一个科学里程碑帮助发现了氯贝特在大鼠肝脏中的肝肿大效应,随后被鉴定为啮齿动物的过氧化物酶体增殖物和过氧化物酶体脂肪酸β-氧化途径的激活剂。这些过氧化物酶体增殖物后来被鉴定为过氧化物酶体增殖物激活受体α(PPARα)的激活配体,1990年克隆。配体激活的异二聚体PPARα/RXRα识别DNA序列,称为PPRE(过氧化物酶体增殖反应元件),对应于两个半共有六核苷酸基序,AGGTCA,由一个核苷酸分开。因此,含有PPRE/PPARα/RXRα/配体/共调节剂的组装复合物控制与肝脏过氧化物酶体脂肪酸β-氧化有关的基因的表达。这篇综述调动了大量讨论杂项轴的发现,涵盖了PPARα在物种和组织中的详细表达模式,几种PPARαKO小鼠模型的经验教训以及膳食微量营养素对PPARα功能的调节。
    In mammalian cells, two cellular organelles, mitochondria and peroxisomes, share the ability to degrade fatty acid chains. Although each organelle harbors its own fatty acid β-oxidation pathway, a distinct mitochondrial system feeds the oxidative phosphorylation pathway for ATP synthesis. At the same time, the peroxisomal β-oxidation pathway participates in cellular thermogenesis. A scientific milestone in 1965 helped discover the hepatomegaly effect in rat liver by clofibrate, subsequently identified as a peroxisome proliferator in rodents and an activator of the peroxisomal fatty acid β-oxidation pathway. These peroxisome proliferators were later identified as activating ligands of Peroxisome Proliferator-Activated Receptor α (PPARα), cloned in 1990. The ligand-activated heterodimer PPARα/RXRα recognizes a DNA sequence, called PPRE (Peroxisome Proliferator Response Element), corresponding to two half-consensus hexanucleotide motifs, AGGTCA, separated by one nucleotide. Accordingly, the assembled complex containing PPRE/PPARα/RXRα/ligands/Coregulators controls the expression of the genes involved in liver peroxisomal fatty acid β-oxidation. This review mobilizes a considerable number of findings that discuss miscellaneous axes, covering the detailed expression pattern of PPARα in species and tissues, the lessons from several PPARα KO mouse models and the modulation of PPARα function by dietary micronutrients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    过氧化物酶体增殖物激活受体γ(PPARG)核受体调节能量代谢和胰岛素敏感性。在这项研究中,我们提出了新的证据,证明PPARG在调节骨细胞功能中的重要作用,并支持能量代谢调节与骨量之间结合的新兴概念。我们报告说,PPARG是必不可少的硬化素生产,最近批准的治疗骨质疏松症的目标。我们的骨细胞特异性PPARG缺失小鼠模型(Dmp1CrePparγflfl或γOTKO)的特征是骨量增加和骨髓肥胖减少,这与WNT信号的上调和内骨成骨细胞的骨形成活性增加一致。对来自γOTKO和对照小鼠的骨细胞的分析显示,在转录本和蛋白质水平上,PPARG与SOST/硬化蛋白之间具有极好的相关性。Sost基因转录起始位点上游的8kb序列具有多个PPARG结合元件(PPRE),其中至少两个结合PPARG,其动力学反映出其被完全激动剂罗格列酮激活,并与Sost转录本和硬化蛋白表达水平升高相关(Pearson'sr=0.991,p=0.001)。年龄较大的γOTKO雌性小鼠在很大程度上受到TZD诱导的骨丢失的保护,这提供了可以在药理学上靶向骨细胞中的PPARG的概念证据。这些发现表明,PPARG的转录活性对于骨细胞中硬化蛋白的表达至关重要,并支持考虑用选择性调节剂靶向PPARG活性来治疗骨质疏松症。
    The peroxisome proliferator activated receptor gamma (PPARG) nuclear receptor regulates energy metabolism and insulin sensitivity. In this study, we present novel evidence for an essential role of PPARG in the regulation of osteocyte function, and support for the emerging concept of the conjunction between regulation of energy metabolism and bone mass. We report that PPARG is essential for sclerostin production, a recently approved target to treat osteoporosis. Our mouse model of osteocyte-specific PPARG deletion (Dmp1CrePparγflfl or γOTKO) is characterized with increased bone mass and reduced bone marrow adiposity, which is consistent with upregulation of WNT signaling and increased bone forming activity of endosteal osteoblasts. An analysis of osteocytes derived from γOTKO and control mice showed an excellent correlation between PPARG and SOST/sclerostin at the transcript and protein levels. The 8 kb sequence upstream of Sost gene transcription start site possesses multiple PPARG binding elements (PPREs) with at least two of them binding PPARG with dynamics reflecting its activation with full agonist rosiglitazone and correlating with increased levels of Sost transcript and sclerostin protein expression (Pearson\'s r = 0.991, p = 0.001). Older γOTKO female mice are largely protected from TZD-induced bone loss providing proof of concept that PPARG in osteocytes can be pharmacologically targeted. These findings demonstrate that transcriptional activities of PPARG are essential for sclerostin expression in osteocytes and support consideration of targeting PPARG activities with selective modulators to treat osteoporosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    人细胞色素P4501B1(CYP1B1)介导的内源性和外源性生物转化在人乳腺癌的进展中起着重要作用。在这项研究中,我们研究了过氧化物酶体增殖物激活受体α(PPARα)激动剂WY-14643的作用,CYP1B1在MCF7乳腺癌细胞中的表达及相关机制。我们进行了定量逆转录聚合酶链反应,瞬时转染,和染色质免疫沉淀来评估PPARα对过氧化物酶体增殖反应元件(PPRE)介导的转录的影响。WY-14643增加了CYP1B1的蛋白质和mRNA水平,以及启动子活性,在MCF-7细胞中。此外,WY-14643加GW6471,一种PPARα拮抗剂,显著抑制WY-14643介导的CYP1B1表达增加。通过小干扰RNA敲低PPARα显著抑制了WY-14643对CYP1B1表达的诱导,表明WY-14643通过PPARα依赖性机制诱导CYP1B1表达。生物信息学分析鉴定了CYP1B1基因启动子区域内的推定PPREs(-833/-813)。通过缺失诱变使这些推定的PPRE失活抑制了WY-14643介导的CYP1B1启动子激活的诱导。此外,WY-14643诱导PPARα呈现能够特异性结合CYP1B1启动子中的PPRE结合位点的形式。我们的发现表明WY-14643通过激活PPARα诱导CYP1B1的表达。
    Human cytochrome P450 1B1 (CYP1B1)-mediated biotransformation of endobiotics and xenobiotics plays an important role in the progression of human breast cancer. In this study, we investigated the effects of WY-14643, a peroxisome proliferator-activated receptor α (PPARα) agonist, on CYP1B1 expression and the related mechanism in MCF7 breast cancer cells. We performed quantitative reverse transcription-polymerase chain reaction, transient transfection, and chromatin immunoprecipitation to evaluate the effects of PPARα on peroxisome proliferator response element (PPRE)-mediated transcription. WY-14643 increased the protein and mRNA levels of CYP1B1, as well as promoter activity, in MCF-7 cells. Moreover, WY-14643 plus GW6471, a PPARα antagonist, significantly inhibited the WY-14643-mediated increase in CYP1B1 expression. PPARα knockdown by a small interfering RNA markedly suppressed the induction of CYP1B1 expression by WY-14643, suggesting that WY-14643 induces CYP1B1 expression via a PPARα-dependent mechanism. Bioinformatics analysis identified putative PPREs (-833/-813) within the promoter region of the CYP1B1 gene. Inactivation of these putative PPREs by deletion mutagenesis suppressed the WY-14643-mediated induction of CYP1B1 promoter activation. Furthermore, WY-14643 induced PPARα to assume a form capable of binding specifically to the PPRE-binding site in the CYP1B1 promoter. Our findings suggest that WY-14643 induces the expression of CYP1B1 through activation of PPARα.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    丹参酮Ⅱ_A(TanⅡ_A),丹参的脂溶性成分,不仅可以改善脂质代谢和降低脂质过氧化的浓度,还能抵抗氧化损伤,清除自由基,控制炎症,对肝功能损害后的预后有保护作用。因此,对TanⅡ_A保护肝脏的确切机制的研究可为TanⅡ_A对肝损伤的防治作用提供重要的理论和实验依据。在本研究中,研究了TanⅡ_A对4-羟基壬烯醛(4-HNE)诱导的肝损伤的保护作用及其机制。正常肝组织NCTC1469细胞用于通过4-HNE处理诱导肝细胞氧化损伤。通过乳酸脱氢酶(LDH)释放量分析和hoechst染色检测TanⅡ_A对肝细胞氧化损伤的保护作用。通过Westernblot分析TanⅡ_A处理前后NCTC1469细胞中过氧化物酶体增殖物激活受体α(PPARα)和过氧化物酶体增殖物反应元件(PPRE)的蛋白表达变化。通过实时聚合酶链反应(PCR)分析脂肪醛脱氢酶(FALDH)的基因表达变化。结果表明,4-HNE增加了LDH的释放量,降低了NCTC1469细胞的细胞活力,TanⅡ_A逆转了4-HNE诱导的肝细胞损伤。Westernblot分析和RT-PCR分析结果显示,4-HNE降低PPARα和FALDH的表达,增加4-HNE的表达。然而,TanⅡ_A处理后,PPARα和FALDH的表达显着增加,4-HNE的表达明显降低。本研究证实了TanⅡ_A对肝细胞损伤的疗效明显,其机制可能与激活PPARα和FALDH表达以及清除4-HNE有关。
    Tanshinone Ⅱ_A( Tan Ⅱ_A),the liposoluble constituents of Salvia miltiorrhiza,can not only ameliorate the lipidic metabolism and decrease the concentration of lipid peroxidation,but also resist oxidation damage,scavenge free radicals and control inflammation,with a protective effect on prognosis after liver function impairment. Therefore,the studies on the exact mechanism of Tan Ⅱ_A in protecting the liver can provide important theoretical and experimental basis for the prevention and treatment effect of Tan Ⅱ_A for liver injury. In the present study,the protective effects and mechanism of Tan Ⅱ_A on 4-hydroxynonenal( 4-HNE)-induced liver injury were investigated in vitro. Normal liver tissues NCTC 1469 cells were used to induce hepatocytes oxidative damages by 4-HNE treatment. The protective effect of Tan Ⅱ_A on hepatocytes oxidative damages was detected by release amount of lactate dehydrogenase( LDH) analysis and hoechst staining. The protein expression changes of peroxisome proliferator-activated receptor α( PPARα) and peroxisome proliferator response element( PPRE) were analyzed by Western blot analysis in NCTC 1469 cells before and after Tan Ⅱ_A treatment. The gene expression changes of fatty aldehyde dehydrogenase( FALDH) were analyzed by Real-time polymerase chain reaction( PCR) analysis. The results showed that 4-HNE increased the release amount of LDH,lowered the cell viability of NCTC 1469 cells,and Tan Ⅱ_A reversed 4-HNE-induced hepatocyte damage. Western blot analysis and RT-PCR analysis results showed that 4-HNE decreased the expression of PPARα and FALDH and increased the expression of 4-HNE. However,the expression of PPARα and FALDH were increased significantly and the expression of 4-HNE was decreased obviously after Tan Ⅱ_A treatment. This study confirmed that the curative effect of Tan Ⅱ_A was obvious on hepatocytes damage,and the mechanism may be associated with activating PPARα and FALDH expression as well as scavenging 4-HNE.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肥胖是发达国家的主要公共卫生问题,其原因是食物摄入增加和能量消耗减少,并且通常与异常的脂质代谢相关。这里,我们证明DEC1是一种基本的螺旋-环-螺旋转录因子,在调节小鼠棕色脂肪组织(BAT)的脂质消耗中起重要作用,这是产热的主要部位。纯合Dec1缺失减轻高脂饮食诱导的肥胖,脂肪细胞肥大,脂肪体积和肝脂肪变性。此外,DEC1缺乏会增加白天的体温,并增强解偶联剂蛋白1的表达,解偶联剂蛋白1是产热的关键因素,和肩胛骨间BAT中各种脂解相关基因。体外实验表明,DEC1通过直接结合RXRα来抑制过氧化物酶体增殖物激活受体γ和类维生素AX受体α(RXRα)的异二聚体诱导的各种脂解相关基因的表达。这些观察结果表明,由DEC1缺乏引起的BAT的脂解作用增强导致脂质消耗增加,从而减少脂肪组织和肝脏中的脂质积累。因此,DEC1可以作为抑制脂质消耗的节能因子,这可能与控制肥胖有关。
    Obesity is a major public health problem in developed countries resulting from increased food intake and decreased energy consumption and usually associated with abnormal lipid metabolism. Here, we show that DEC1, a basic helix-loop-helix transcription factor, plays an important role in the regulation of lipid consumption in mouse brown adipose tissue (BAT), which is the major site of thermogenesis. Homozygous Dec1 deletion attenuated high-fat-diet-induced obesity, adipocyte hypertrophy, fat volume and hepatic steatosis. Furthermore, DEC1 deficiency increased body temperature during daytime and enhanced the expression of uncoupler protein 1, a key factor of thermogenesis, and various lipolysis-related genes in interscapular BAT. In vitro experiments suggested that DEC1 suppresses the expression of various lipolysis-related genes induced by the heterodimer of peroxisome proliferator-activated receptor γ and retinoid X receptor α (RXRα) through direct binding to RXRα. These observations suggest that enhanced lipolysis in BAT caused by DEC1 deficiency leads to an increase in lipid consumption, thereby decreasing lipid accumulation in adipose tissues and the liver. Thus, DEC1 may serve as an energy-saving factor that suppresses lipid consumption, which may be relevant to managing obesity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    基质衍生因子-1α(SDF-1α)及其同源受体CXCR4在介导乳腺癌细胞侵袭和转移中起关键作用。因此,能够抑制CXCR4激活的药物可能会增加减少肿瘤进展的关键工具,尤其是在最具侵袭性的乳腺癌疾病中。过氧化物酶体增殖物激活受体(PPAR)γ,核受体超家族的成员,已经发现在不同的癌细胞中下调CXCR4基因的表达,然而,这种效应的分子机制尚不完全清楚。这里,我们发现了一种新的PPARγ介导的机制,该机制在上皮和基质乳腺癌细胞中均负调节CXCR4的表达.我们发现,配体激活的PPARγ通过将类视黄醇和甲状腺激素受体(SMRT)的沉默介体募集到乳腺癌细胞系CXCR4启动子内新鉴定的PPAR反应元件(PPRE)上,下调CXCR4的转录活性。因此,PPARγ激动剂罗格列酮(BRL)显著抑制细胞迁移和侵袭,因为它在PPARγ拮抗剂GW9662存在下被逆转。根据癌症相关成纤维细胞(CAFs)的能力,乳腺癌间质中最丰富的成分,分泌高水平的SDF-1α,BRL降低了源自CAF的条件培养基(CM)诱导的迁移促进活性,并影响了CAF-CM激活的CXCR4下游信号通路。此外,暴露于BRL的CAF显示CXCR4的表达降低,运动性和侵袭性降低以及以形态改变为特征的表型。总的来说,我们的研究结果为PPARγ在抑制乳腺癌进展中的作用提供了新的见解,并进一步强调了PPARγ配体在针对乳腺癌患者的癌症和周围基质细胞的未来治疗中的实用性.
    Stromal Derived Factor-1α (SDF-1α) and its cognate receptor CXCR4 play a key role in mediating breast cancer cell invasion and metastasis. Therefore, drugs able to inhibit CXCR4 activation may add critical tools to reduce tumor progression, especially in the most aggressive form of the breast cancer disease. Peroxisome Proliferator-Activated Receptor (PPAR) γ, a member of the nuclear receptor superfamily, has been found to downregulate CXCR4 gene expression in different cancer cells, however the molecular mechanism underlying this effect is not fully understood. Here, we identified a novel PPARγ-mediated mechanism that negatively regulates CXCR4 expression in both epithelial and stromal breast cancer cells. We found that ligand-activated PPARγ downregulated CXCR4 transcriptional activity through the recruitment of the silencing mediator of retinoid and thyroid hormone receptor (SMRT) corepressor onto a newly identified PPAR response element (PPRE) within the CXCR4 promoter in breast cancer cell lines. As a consequence, the PPARγ agonist rosiglitazone (BRL) significantly inhibited cell migration and invasion and this effect was PPARγ-mediated, since it was reversed in the presence of the PPARγ antagonist GW9662. According to the ability of cancer-associated fibroblasts (CAFs), the most abundant component of breast cancer stroma, to secrete high levels of SDF-1α, BRL reduced migratory promoting activities induced by conditioned media (CM) derived from CAFs and affected CXCR4 downstream signaling pathways activated by CAF-CM. In addition, CAFs exposed to BRL showed a decreased expression of CXCR4, a reduced motility and invasion along with a phenotype characterized by an altered morphology. Collectively, our findings provide novel insights into the role of PPARγ in inhibiting breast cancer progression and further highlight the utility of PPARγ ligands for future therapies aimed at targeting both cancer and surrounding stromal cells in breast cancer patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Cachexia, the most severe paraneoplastic syndrome, occurs in about 80% of patients with advanced cancer; it cannot be reverted by conventional, enteral, or parenteral nutrition. For this reason, nutritional interventions must be based on the use of substances possessing, alongside nutritional and energetic properties, the ability to modulate production of the pro-inflammatory factors responsible for the metabolic changes characterising cancer cachexia. In light of their nutritional and anti-inflammatory properties, polyunsaturated fatty acids (PUFAs), and in particular n-3, have been investigated for treating cachexia; however, the results have been contradictory. Since both n-3 and n-6 PUFAs can affect cell functions in several ways, this research investigated the possibility that the effects of both n-3 and n-6 PUFAs could be mediated by their major aldehydic products of lipid peroxidation, 4-hydroxyhexenal (HHE) and 4-hydroxynonenal (HNE), and by their anti-inflammatory properties. An \"in vitro\" cancer cachexia model, consisting of human lung cancer cells (A427) and murine myoblasts (C2C12), was used. The results showed that: 1) both n-3 and n-6 PUFAs reduced the growth of lung cancer cells without causing cell death, increased lipid peroxidation and Peroxisome Proliferator-Activated Receptor (PPAR)α, and decreased TNFα; 2) culture medium conditioned by A427 cells grown in the absence of PUFAs blocked myosin production and the differentiation of C2C12 muscle cells; conversely, muscle cells grown in culture medium conditioned by the same cells in the presence of PUFAs showed myosin expression and formed myotubes; 3) adding HHE or HNE directly to C2C12 cells maintained in culture medium conditioned by A427 cells in the absence of PUFAs stimulated myosin production and myotube formation; 4) putative consensus sequences for (PPARs) have been found in genes encoding fast isoforms of myosin heavy chain, by a bioinformatics approach. The overall results show, first, the ability of both n-3 and n-6 PUFAs and their lipid peroxidation products to prevent the blocking of myosin expression and myotube formation caused in C2C12 cells by medium conditioned by human lung tumour cells. The C2C12 cell differentiation can be due to direct effect of lipid peroxidation products, as evidenced by treating C2C12 cells with HHE and HNE, and to the decrease of pro-inflammatory TNFα in A427 cell culture medium. The presence of consensus sequences for PPARs in genes encoding the fast isoforms of myosin heavy chain suggests that the effects of PUFAs, HHE, and HNE are PPAR-mediated.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    In previous work, it is suggested that the excessive amount of fatty acids transported by FABP5 may facilitate the malignant progression of prostate cancer cells through a FABP5-PPARγ-VEGF signal transduction axis to increase angiogenesis. To further functionally characterise the FABP5-PPARγ-VEGF signal transduction pathway, we have, in this work, investigated the molecular mechanisms involved in its tumorigenicity promoting role in prostate cancer. Suppression of PPARγ in highly malignant prostate cancer cells produced a significant reduction (up to 53%) in their proliferation rate, invasiveness (up to 89%) and anchorage-independent growth (up to 94%) in vitro. Knockdown of PPARγ gene in PC3-M cells by siRNA significantly reduced the average size of tumours formed in nude mice by 99% and tumour incidence by 90%, and significantly prolonged the latent period by 3.5 fold. Results in this study combined with some previous results suggested that FABP5 promoted VEGF expression and angiogenesis through PPARγ which was activated by fatty acids transported by FABP5. Further investigations showed that PPARγ up-regulated VEGF expression through acting with the PPAR-responsive elements in the promoter region of VEGF gene in prostate cancer cells. Although androgen can modulate VEGF expression through Sp1/Sp3 binding site on VEGF promoter in androgen-dependent prostate cancer cells, this route, disappeared as the cells gradually lost their androgen dependency; was replaced by the FABP5-PPARγ-VEGF signalling pathway. These results suggested that the FABP5-PPARγ-VEGF signal transduction axis, rather than androgen modulated route, may be a more important novel therapeutic target for angiogenesis-suppression treatment of castration resistant prostate cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    目的:脂肪组织是一种高度通用的系统,其中脂肪细胞中的线粒体在主动组织重塑过程中发生显着变化。BCL2/腺病毒E1B19kDa蛋白相互作用蛋白3(BNIP3)是线粒体蛋白和已知的线粒体质量调节剂。在这项研究中,我们研究了BNIP3在脂肪细胞中的作用,特别是在过氧化物酶体增殖物激活受体γ(PPARγ)诱导的脂肪组织重塑条件下。
    方法:体外评价BNIP3在3T3-L1脂肪细胞中的表达,饲喂高脂肪饮食的C57BL/6小鼠和db/db小鼠体内。罗格列酮治疗后,在BNIP3敲低脂肪细胞中研究了线粒体生物能量学。通过启动子测定和电泳迁移率变化测定(EMSA)表征了推定的过氧化物酶体增殖激素反应元件(PPRE)。
    结果:蛋白BNIP3在棕色脂肪组织中的含量高于白色脂肪组织。此外,BNIP3表达通过3T3-L1前脂肪细胞分化上调,饥饿和罗格列酮治疗。相反,在与胰岛素抵抗相关的各种条件下,脂肪细胞中的BNIP3表达降低。通过罗格列酮处理恢复了BNIP3的这种下调。BNIP3在脂肪细胞中的敲除抑制罗格列酮诱导的线粒体生物合成和功能,部分由5'AMP激活蛋白激酶(AMPK)-过氧化物酶体增殖物激活受体γ介导,共激活因子1α(PGC1α)信号通路。罗格列酮处理增加了报告基因测定中Bnip3的转录水平,并且通过EMSA证明了Bnip3启动子中PPRE位点的存在。
    结论:蛋白质BNIP3有助于改善暴露于罗格列酮时发生的线粒体生物能学。它可能是在胰岛素抵抗条件下恢复线粒体功能障碍的新治疗靶标。
    OBJECTIVE: Adipose tissue is a highly versatile system in which mitochondria in adipocytes undergo significant changes during active tissue remodelling. BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3) is a mitochondrial protein and a known mitochondrial quality regulator. In this study, we investigated the role of BNIP3 in adipocytes, specifically under conditions of peroxisome proliferator-activated receptor-γ (PPARγ)-induced adipose tissue remodelling.
    METHODS: The expression of BNIP3 was evaluated in 3T3-L1 adipocytes in vitro, C57BL/6 mice fed a high-fat diet and db/db mice in vivo. Mitochondrial bioenergetics was investigated in BNIP3-knockdown adipocytes after rosiglitazone treatment. A putative peroxisome proliferator hormone responsive element (PPRE) was characterised by promoter assay and electrophoretic mobility shift assay (EMSA).
    RESULTS: The protein BNIP3 was more abundant in brown adipose tissue than white adipose tissue. Furthermore, BNIP3 expression was upregulated by 3T3-L1 pre-adipocyte differentiation, starvation and rosiglitazone treatment. Conversely, BNIP3 expression in adipocytes decreased under various conditions associated with insulin resistance. This downregulation of BNIP3 was restored by rosiglitazone treatment. Knockdown of BNIP3 in adipocytes inhibited rosiglitazone-induced mitochondrial biogenesis and function, partially mediated by the 5\' AMP-activated protein kinase (AMPK)-peroxisome proliferator-activated receptor γ, co-activator 1 α (PGC1α) signalling pathway. Rosiglitazone treatment increased the transcription level of Bnip3 in the reporter assay and the presence of the PPRE site in the Bnip3 promoter was demonstrated by EMSA.
    CONCLUSIONS: The protein BNIP3 contributes to the improvement of mitochondrial bioenergetics that occurs on exposure to rosiglitazone. It may be a novel therapeutic target for restoring mitochondrial dysfunction under insulin-resistant conditions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    过氧化物酶体增殖物激活受体(PPAR)通过控制参与脂质摄取和脂肪酸氧化等过程的许多基因,在调节肝脂质代谢中起着重要作用。在这里,我们将缺氧诱导的脂滴相关(Hilpda/Hig2)鉴定为新的PPAR靶基因,并证明其参与肝脂质代谢。微阵列分析显示,Hilpda是PPARα激动剂Wy14643在小鼠精确切割的肝切片中诱导最多的基因之一。在小鼠和人肝细胞中证实了Wy14643对HilpdamRNA的诱导。口服给药Wy14643类似地诱导野生型小鼠而不是Ppara(-/-)小鼠的肝脏中的HilpdamRNA水平。反激活研究和染色质免疫沉淀表明,Hilpda是通过位于转录起始位点上游1200个碱基对的保守PPAR响应元件的直接PPARα靶基因。通过腺相关病毒在小鼠肝脏中过度表达HILPDA导致肝脏甘油三酯储存增加4倍,在从头脂肪生成中涉及的关键基因没有任何变化,β-氧化,或者脂解。此外,细胞内脂肪酶活性不受HILPDA过表达的影响。引人注目的是,HILPDA过表达显著损害肝甘油三酯分泌。一起来看,我们的数据揭示了HILPDA是一种新的PPAR靶标,它通过调节甘油三酯分泌来提高肝脏甘油三酯的储存.
    Peroxisome proliferator-activated receptors (PPARs) play major roles in the regulation of hepatic lipid metabolism through the control of numerous genes involved in processes such as lipid uptake and fatty acid oxidation. Here we identify hypoxia-inducible lipid droplet-associated (Hilpda/Hig2) as a novel PPAR target gene and demonstrate its involvement in hepatic lipid metabolism. Microarray analysis revealed that Hilpda is one of the most highly induced genes by the PPARα agonist Wy14643 in mouse precision cut liver slices. Induction of Hilpda mRNA by Wy14643 was confirmed in mouse and human hepatocytes. Oral dosing with Wy14643 similarly induced Hilpda mRNA levels in livers of wild-type mice but not Ppara(-/-) mice. Transactivation studies and chromatin immunoprecipitation showed that Hilpda is a direct PPARα target gene via a conserved PPAR response element located 1200 base pairs upstream of the transcription start site. Hepatic overexpression of HILPDA in mice via adeno-associated virus led to a 4-fold increase in liver triglyceride storage, without any changes in key genes involved in de novo lipogenesis, β-oxidation, or lipolysis. Moreover, intracellular lipase activity was not affected by HILPDA overexpression. Strikingly, HILPDA overexpression significantly impaired hepatic triglyceride secretion. Taken together, our data uncover HILPDA as a novel PPAR target that raises hepatic triglyceride storage via regulation of triglyceride secretion.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号