PPARα

PPAR α
  • 文章类型: Journal Article
    肝癌是癌症相关死亡的第四大原因,全球发病率稳步上升。作为肝癌的一个众所周知的标志,代谢改变与脂质体变化有关,根据最近的脂质组学研究,原发性肝癌的一个共同特征。过氧化物酶体增殖物激活受体α(PPARα)是一种具有重要脂质稳态功能的配体激活转录因子,因此,我们旨在了解使用PPAR-α激动剂WY-14643后激活PPARα的分子机制和途径,并鉴定与PPARα活性相关的候选生物标志物,评估其在肝癌中的作用.通过DESeq2评估来自单独肥胖受试者的肝癌组织和治疗后的肝组织之间的不同表达基因(DEGs)的数据,并使用加权基因共表达网络分析(WGCNA)分析模块基因。最终的候选基因通过在高度排名的DEG和棕色模块之间的交叉基因来鉴定,与药物治疗呈显著负相关。我们进行了蛋白质-蛋白质相互作用网络,和KEGG富集分析,使用cyto-hubba插件鉴定了核心hub基因(CD40,CXCL9,CXCL10,TNFSF14,GBP2,GBP3,APOL3,CLDN1),其中,我们重点研究了在肿瘤发生中起关键作用的GBP2,并根据临床结局评估其表达.总之,基于WGCNA的共表达网络将GBP2确定为与PPARα激动剂治疗负相关的hub基因之一。GBP2的高表达与HCC的进展密切相关。因此,GBP2可能是肝癌PPARα活性研究的潜在候选者。
    Liver cancer is the fourth leading cause of cancer-related deaths with a steadily increasing rate worldwide, as a well-known hallmark of liver cancer, metabolic alterations are related to liposomal changes, a common characteristic of primary liver cancers based on recent lipidomics studies. Peroxisome proliferator-activated receptor α (PPARα) is a ligand-activated transcription factor with important lipid homeostasis function, therefore we aimed to understand the molecular mechanisms and pathways that activate PPARα after using PPAR-α agonist WY-14643 and identify candidate biomarkers related to PPARα activity and evaluate their effects in liver cancer. The data from differently expressed genes (DEGs) between liver cancer tissue from obese subjects alone and liver tissue after treatment were evaluated by DESeq2 and module genes were analyzed using weighted gene co-expression network analysis (WGCNA). Final candidate genes were identified by intersecting genes among highly ranked DEGs and the brown module, which demonstrated a significant negative correlation with drug treatments. We conducted a protein-protein interaction network, and KEGG enrichment analysis, and core hub genes (CD40, CXCL9, CXCL10, TNFSF14, GBP2, GBP3, APOL3, CLDN1) were identified using the cyto-hubba plugin, among them we focused on GBP2 that plays key roles in oncogenesis and evaluate its expressional with clinical outcomes. In conclusion, the WGCNA-based co-expression network identified GBP2 as one of the hub genes with a negative relation with PPARα agonist treatments. higher expression of GBP2 was closely associated with HCC progression. Therefore, GBP2 might be a potential candidate for the study of PPARα activity in HCC.
    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
    虽然线粒体中的脂肪酸氧化(FAO)是静止淋巴细胞的主要能量来源,在经历代谢重编程的活化淋巴细胞中促进FAO的影响尚不清楚.这里,我们证明了贝贝特,一种临床上用于治疗高甘油三酯血症的选择性PPARα调节剂,改变T细胞的代谢系统,缓解多种自身免疫性疾病。培马贝特抑制Th17细胞,但不抑制Th1细胞,通过抑制增强的FAO引发的谷氨酰胺分解和糖酵解。相比之下,常规PPARα激动剂非诺贝特通过抑制总体代谢甚至在不足以诱导脂肪酸氧化的剂量下显著抑制细胞生长。临床上,接受培美贝特治疗的患者外周血Th17/Treg比值显著降低.我们的结果表明,通过培美贝特介导的PPARα选择性激活来增强FAO可以抑制Th17细胞的代谢程序,并且可能是治疗自身免疫性疾病的可行选择。
    While fatty acid oxidation (FAO) in mitochondria is a primary energy source for quiescent lymphocytes, the impact of promoting FAO in activated lymphocytes undergoing metabolic reprogramming remains unclear. Here, we demonstrate that pemafibrate, a selective PPARα modulator used clinically for the treatment of hypertriglyceridemia, transforms metabolic system of T-cells and alleviates several autoimmune diseases. Pemafibrate suppresses Th17 cells but not Th1 cells, through the inhibition of glutaminolysis and glycolysis initiated by enhanced FAO. In contrast, a conventional PPARα agonist fenofibrate significantly inhibits cell growth by restraining overall metabolisms even at a dose insufficient to induce fatty acid oxidation. Clinically, patients receiving pemafibrate showed a significant decrease of Th17/Treg ratio in peripheral blood. Our results suggest that augmented FAO by pemafibrate-mediated selective activation of PPARα restrains metabolic programs of Th17 cells and could be a viable option for the treatment of autoimmune diseases.
    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
    男性患代谢功能障碍相关脂肪性肝炎(MASH)的风险高于女性;然而,MASH发育中介导性二态性的机制尚不完全清楚。基于营养的小鼠模型表明,失调的脂肪酸生物合成促进MASH。药物概括了MASH,没有饮食变化。这份简短的报告利用药物诱导的MASH模型研究了性二态性与男性对MASH易感性的关联,并着重于非常长链的脂肪酸生物合成途径。我们在MASH诱导后5周和15周时通过免疫和使用蛋白质印迹的年龄匹配的未免疫对照来评估雄性和雌性小鼠肝脏。我们的结果表明,PPARα和CYP4a12a保护女性,而CYP4v2不能保护男性免受MASH发育。我们的结果对理解MASH发病机制中的性二态性具有重要意义。
    Males are at higher risk for developing metabolic dysfunction-associated steatohepatitis (MASH) than females; however, mechanisms mediating sexual dimorphism in MASH development are not completely understood. Nutrition-based mouse models suggest that dysregulated fatty acid biosynthesis promotes MASH. Drugs recapitulate MASH without diet variabilities. This brief report investigates associations of sexual dimorphism with male susceptibility to MASH utilizing a drug-induced MASH model and focuses on very-long-chain fatty acid biosynthesis pathways. We assessed male and female mouse livers at 5 and 15 weeks following MASH induction by immunizations and age-matched un-immunized controls utilizing Western blot. Our results suggest that PPAR alpha and CYP4a12a protect females, while CYP4v2 does not protect males from MASH development. Our results have important implications for understanding sexual dimorphism in the pathogenesis of MASH.
    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
    背景:原代人肝细胞(PHHs)对于药物代谢评估非常有价值,肝脏疾病建模和肝细胞移植。然而,由于捐助来源有限,它们的供应受到很大限制,在体外培养时,它们的增殖能力受到限制,功能降低。为了应对这一挑战,我们的目的是开发一种新的方法,在体外有效扩增PHHs而不丧失功能。
    方法:通过模拟体内肝脏再生途径,我们开发了一个两步策略,涉及去分化/扩增和随后成熟的PHHs,以在体外产生丰富的功能性肝细胞。最初,我们应用了SiPer,一种预测算法,鉴定能够激活肝脏再生转录因子的候选小分子,从而配制新的肝扩增培养基以将PHH去分化为增殖性人肝祖细胞样细胞(ProHPLC)。然后使用新的肝细胞成熟条件将这些ProHPLC再分化成功能成熟的肝细胞。此外,我们研究了在新条件下PHHs扩展的潜在机制。
    结果:含有氢化可的松的新型肝扩增培养基促进PHHs去分化为ProHPLC,与在先前建立的扩增条件下培养的细胞相比,其表现出关键的肝祖细胞特征,并证明增殖能力显着增加。值得注意的是,这些随后成熟的肝细胞在转录组谱方面与PHH相当,药物代谢活性和体内植入能力。重要的是,我们的发现提示氢化可的松增强PHHs的扩增可能是通过PPARα信号通路和再生转录因子介导的。
    结论:本研究提出了一个两步策略,该策略最初将PHH诱导为增殖状态(ProHPLC),以确保足够的细胞数量,随后ProHPLC成熟为功能齐全的肝细胞,以保证最佳的细胞质量。这种方法为基于肝细胞的应用提供了产生大量接种细胞的有希望的手段。
    BACKGROUND: Primary human hepatocytes (PHHs) are highly valuable for drug-metabolism evaluation, liver disease modeling and hepatocyte transplantation. However, their availability is significantly restricted due to limited donor sources, alongside their constrained proliferation capabilities and reduced functionality when cultured in vitro. To address this challenge, we aimed to develop a novel method to efficiently expand PHHs in vitro without a loss of function.
    METHODS: By mimicking the in vivo liver regeneration route, we developed a two-step strategy involving the de-differentiation/expansion and subsequent maturation of PHHs to generate abundant functional hepatocytes in vitro. Initially, we applied SiPer, a prediction algorithm, to identify candidate small molecules capable of activating liver regenerative transcription factors, thereby formulating a novel hepatic expansion medium to de-differentiate PHHs into proliferative human hepatic progenitor-like cells (ProHPLCs). These ProHPLCs were then re-differentiated into functionally mature hepatocytes using a new hepatocyte maturation condition. Additionally, we investigated the underlying mechanism of PHHs expansion under our new conditions.
    RESULTS: The novel hepatic expansion medium containing hydrocortisone facilitated the de-differentiation of PHHs into ProHPLCs, which exhibited key hepatic progenitor characteristics and demonstrated a marked increase in proliferation capacity compared to cells cultivated in previously established expansion conditions. Remarkably, these subsequent matured hepatocytes rivaled PHHs in terms of transcriptome profiles, drug metabolizing activities and in vivo engraftment capabilities. Importantly, our findings suggest that the enhanced expansion of PHHs by hydrocortisone may be mediated through the PPARα signaling pathway and regenerative transcription factors.
    CONCLUSIONS: This study presents a two-step strategy that initially induces PHHs into a proliferative state (ProHPLCs) to ensure sufficient cell quantity, followed by the maturation of ProHPLCs into fully functional hepatocytes to guarantee optimal cell quality. This approach offers a promising means of producing large numbers of seeding cells for hepatocyte-based applications.
    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
    动脉粥样硬化是动脉粥样硬化性心血管疾病(ASCVD)的病理原因,在细胞衰老过程中迅速发展。钠-葡萄糖协同转运蛋白2抑制剂(SGLT2is)可减少ASCVD患者的主要心血管事件,并具有潜在的抗衰老作用。这里,我们研究了SGLT2抑制剂dapagliflozin对动脉粥样硬化小鼠细胞衰老的影响.与生理盐水处理的ApoE-/-对照小鼠相比,ApoE-/-dapagliflozin组的人,接受胃内达格列净(0.1mgkg-1d-1)14周,表现出主动脉斑块总面积的减少(48.8%±6.6%vs.74.6%±8.0%,P<0.05),脂质核心面积的减少((0.019±0.0037)mm2vs。(0.032±0.0062)mm2,P<0.05)和斑块内衰老细胞的百分比(16.4%±3.7%vs.30.7%±2.0%,P<0.01),而纤维帽厚度的增加((21.6±2.1)µm与(14.6±1.5)µm,P<0.01)。小鼠主动脉弓的转录组测序揭示了PPARα和脂肪酸代谢信号通路参与达格列净改善细胞老化和斑块进展的机制。体外,dapagliflozin抑制PPARα及其下游信号FABP4的表达,从而在高脂条件下减少了人主动脉平滑肌细胞(HASMCs)中衰老细胞的积累。这种作用伴随着细胞内脂质含量的降低和氧化应激的减轻。然而,达格列净的这些有益作用可以通过PPARα过表达而逆转。生物信息学分析和分子对接模拟显示达格列净可能通过与RXRA蛋白直接相互作用发挥其作用,从而影响PPARα信号通路的表达。总之,达格列净可能通过抑制RXRA-PPARα-FABP4信号通路改变主动脉平滑肌细胞的衰老,导致动脉粥样硬化进展的减速。
    Atherosclerosis is the pathological cause of atherosclerotic cardiovascular disease (ASCVD), which rapidly progresses during the cellular senescence. Sodium-glucose cotransporter 2 inhibitors (SGLT2is) reduce major cardiovascular events in patients with ASCVD and have potential antisenescence effects. Here, we investigate the effects of the SGLT2 inhibitor dapagliflozin on cellular senescence in atherosclerotic mice. Compared with ApoE-/- control mice treated with normal saline, those in the ApoE-/- dapagliflozin group, receiving intragastric dapagliflozin (0.1 mg kg-1 d-1) for 14 weeks, exhibited the reduction in the total aortic plaque area (48.8%±6.6% vs. 74.6%±8.0%, P<0.05), the decrease in the lipid core area ((0.019±0.0037) mm2vs. (0.032±0.0062) mm2, P<0.05) and in the percentage of senescent cells within the plaques (16.4%±3.7% vs. 30.7%±2.0%, P<0.01), while the increase in the thickness of the fibrous cap ((21.6±2.1) µm vs. (14.6±1.5) µm, P<0.01). Transcriptome sequencing of the aortic arch in the mice revealed the involvement of the PPARα and the fatty acid metabolic signaling pathways in dapagliflozin\'s mechanism of ameliorating cellular aging and plaque progression. In vitro, dapagliflozin inhibited the expression of PPARα and its downstream signal FABP4, by which the accumulation of senescent cells in human aortic smooth muscle cells (HASMCs) was reduced under high-fat conditions. This effect was accompanied by a reduction in the intracellular lipid content and alleviation of oxidative stress. However, these beneficial effects of dapagliflozin could be reversed by the PPARα overexpression. Bioinformatics analysis and molecular docking simulations revealed that dapagliflozin might exert its effects by directly interacting with the RXRA protein, thereby influencing the expression of the PPARα signaling pathway. In conclusion, the cellular senescence of aortic smooth muscle cells is potentially altered by dapagliflozin through the suppression of the RXRA-PPARα-FABP4 signaling pathway, resulting in a deceleration of atherosclerotic progression.
    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
    目的:代谢相关脂肪性肝病(MAFLD)是全球最常见的慢性肝病之一,但其确切的发病机制仍不清楚。本研究试图阐明多核糖核苷酸核苷酸转移酶1(PNPT1)在MAFLD进展中的参与和分子机制。
    方法:该研究采用蛋白质印迹和qRT-PCR来评估来自诊断为MAFLD的个体的肝脏样本和接受高脂肪饮食的小鼠模型中的PNPT1水平。细胞研究调查了PNPT1对脂质代谢的影响,凋亡,和肝细胞的线粒体稳定性。免疫荧光用于跟踪PNPT1在高脂条件下的亚细胞运动。进行RNA免疫沉淀和功能测定以鉴定PNPT1和Mcl-1mRNA之间的相互作用。研究了PPARα作为PNPT1上游转录调节因子的作用。重组腺病毒载体用于调节体内PNPT1表达。
    结果:发现PNPT1在MAFLD患者和HFD小鼠的肝组织中明显减少。体外,PNPT1直接调节肝脏脂质代谢,凋亡,和线粒体稳定性。在脂质升高的条件下,PNPT1从线粒体迁移到细胞质,改变其生理功能。RNA免疫沉淀显示PNPT1的KH和S1结构域结合并降解Mcl-1mRNA,进而影响线粒体通透性。转录调节因子PPARα被鉴定为PNPT1的重要影响者,影响其表达和随后的细胞功能。PNPT1表达的改变与小鼠MAFLD的进展直接相关。
    结论:该研究通过与Mcl-1的相互作用及其对脂质代谢和线粒体稳定性的调节作用,证实了PNPT1在MAFLD发展中的关键作用。这些见解凸显了PNPT1和MAFLD之间的复杂关联,阐明其分子途径,并为MAFLD管理提供潜在的新的治疗途径。
    OBJECTIVE: Metabolic-associated fatty liver disease (MAFLD) represents one of the most prevalent chronic liver conditions worldwide, but its precise pathogenesis remains unclear. This research endeavors to elucidate the involvement and molecular mechanisms of polyribonucleotide nucleotidyltransferase 1 (PNPT1) in the progression of MAFLD.
    METHODS: The study employed western blot and qRT-PCR to evaluate PNPT1 levels in liver specimens from individuals diagnosed with MAFLD and in mouse models subjected to a high-fat diet. Cellular studies investigated the effects of PNPT1 on lipid metabolism, apoptosis, and mitochondrial stability in hepatocytes. Immunofluorescence was utilized to track the subcellular movement of PNPT1 under high lipid conditions. RNA immunoprecipitation and functional assays were conducted to identify interactions between PNPT1 and Mcl-1 mRNA. The role of PPARα as an upstream transcriptional regulator of PNPT1 was investigated. Recombinant adenoviral vectors were utilized to modulate PNPT1 expression in vivo.
    RESULTS: PNPT1 was found to be markedly reduced in liver tissues from MAFLD patients and HFD mice. In vitro, PNPT1 directly regulated hepatic lipid metabolism, apoptosis, and mitochondrial stability. Under conditions of elevated lipids, PNPT1 relocated from mitochondria to cytoplasm, modifying its physiological functions. RNA immunoprecipitation revealed that the KH and S1 domains of PNPT1 bind to and degrade Mcl-1 mRNA, which in turn affects mitochondrial permeability. The transcriptional regulator PPARα was identified as a significant influencer of PNPT1, impacting both its expression and subsequent cellular functions. Alterations in PNPT1 expression were directly correlated with the progression of MAFLD in mice.
    CONCLUSIONS: The study confirms the pivotal function of PNPT1 in the development of MAFLD through its interactions with Mcl-1 and its regulatory effects on lipid metabolism and mitochondrial stability. These insights highlight the intricate association between PNPT1 and MAFLD, shedding light on its molecular pathways and presenting a potential new therapeutic avenue for MAFLD management.
    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
    能量代谢紊乱,主要表现为抑制脂肪酸降解和脂质积累,与衰老加速高度相关。然而,干预措施不足。这里,我们报道了Omega-3多不饱和脂肪酸(Omega-3PUFA),尤其是EPA,对维持能量代谢和脂质稳态对减缓器官衰老具有有益作用。作为过氧化物酶体增殖物激活受体α(PPARα)的内源性激动剂,Omega-3PUFA显着促进了多个衰老器官中脂肪酸β-氧化和ATP的产生。因此,Omega-3PUFAs有效抑制与年龄相关的病理变化,保存的器官功能,延缓衰老过程。在mfat-1转基因小鼠中也证明了Omega-3PUFA的有益作用,自发产生丰富的内源性Omega-3PUFA。总之,我们的研究创新性地证明了Omega-3PUFAs在饮食中通过促进能量代谢来减缓衰老。补充Omega-3PUFA或fat-1转基因提供了一种有希望的治疗方法,以促进老年人的健康衰老。
    Energy metabolism disorder, mainly exhibiting the inhibition of fatty acid degradation and lipid accumulation, is highly related with aging acceleration. However, the intervention measures are deficient. Here, we reported Omega-3 polyunsaturated fatty acids (Omega-3 PUFAs), especially EPA, exerted beneficial effects on maintaining energy metabolism and lipid homeostasis to slow organ aging. As the endogenous agonist of peroxisome proliferator-activated receptor α (PPARα), Omega-3 PUFAs significantly boosted fatty acid β-oxidation and ATP production in multiple aged organs. Consequently, Omega-3 PUFAs effectively inhibited age-related pathological changes, preserved organ function, and retarded aging process. The beneficial effects of Omega-3 PUFAs were also testified in mfat-1 transgenic mice, which spontaneously generate abundant endogenous Omega-3 PUFAs. In conclusion, our study innovatively demonstrated Omega-3 PUFAs administration in diet slow aging through promoting energy metabolism. The supplement of Omega-3 PUFAs or fat-1 transgene provides a promising therapeutic approach to promote healthy aging in the elderly.
    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
    背景:非酒精性脂肪性肝病(NAFLD)是肝脏相关发病率和死亡率的主要原因,肝脏脂肪变性是标志性症状。丹参(Smil,Dan-Shen)和纹状体DC(Lstr,川雄)常用于治疗心血管疾病,具有调节脂质代谢的潜能。然而,Smil/Lstr组合是否可用于治疗NAFLD及其调脂特性的潜在机制尚不清楚.
    目的:评估短期高脂饮食(HFD)诱导的斑马鱼模型评估肝脏脂肪变性表型的可行性和可靠性,并研究Smil/Lstr的肝脏降脂作用。以及它的活性成分。
    方法:在HFD斑马鱼模型中使用荧光成像和组织化学检查肝脏和多个其他器官系统的表型改变。内源性评估Smil/Lstr组合的肝脏特异性降脂作用。进一步探索了斑马鱼的活性分子和功能机制,人类肝细胞,和仓鼠模型。
    结果:在5天的HFD斑马鱼中,在血管和肝脏中检测到明显的脂质积累,正如用油红O和荧光脂质探针染色增加所证明的。在模型中观察到肝肥大,伴有大泡性脂肪变性。Smil/Lstr组合给药可有效恢复HFD斑马鱼的脂质分布并减轻肝肥大。在油酸刺激的肝细胞中,Smil/Lstr组合显着减少脂质积累和细胞损伤。随后,基于斑马鱼的表型筛选,天然的苯酞senkyunolideI(SEI)被确定为介导肝脏中Smil/Lstr组合的降脂活性的主要分子。此外,SEI上调脂质代谢调节剂PPARα的表达和下调的脂肪酸转位酶CD36,而PPARα拮抗剂充分阻断SEI对肝性脂肪变性的调节作用。最后,在仓鼠模型中进一步验证了SEI对肝脏脂质积累和PPARα信号传导的作用.
    结论:我们提出了一种基于斑马鱼的肝脂肪变性调节剂筛选策略,并发现Smil/Lstr组合及其成分SEI对肝脏脂质积累和PPARα信号传导的调节作用,提示其作为NAFLD治疗新候选药物的潜在价值。
    BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is the leading cause of liver-related morbidity and mortality, with hepatic steatosis being the hallmark symptom. Salvia miltiorrhiza Bunge (Smil, Dan-Shen) and Ligusticum striatum DC (Lstr, Chuan-Xiong) are commonly used to treat cardiovascular diseases and have the potential to regulate lipid metabolism. However, whether Smil/Lstr combo can be used to treat NAFLD and the mechanisms underlying its lipid-regulating properties remain unclear.
    OBJECTIVE: To assess the feasibility and reliability of a short-term high-fat diet (HFD) induced zebrafish model for evaluating hepatic steatosis phenotype and to investigate the liver lipid-lowering effects of Smil/Lstr, as well as its active components.
    METHODS: The phenotypic alterations of liver and multiple other organ systems were examined in the HFD zebrafish model using fluorescence imaging and histochemistry. The liver-specific lipid-lowering effects of Smil/Lstr combo were evaluated endogenously. The active molecules and functional mechanisms were further explored in zebrafish, human hepatocytes, and hamster models.
    RESULTS: In 5-day HFD zebrafish, significant lipid accumulation was detected in the blood vessels and the liver, as evidenced by increased staining with Oil Red O and fluorescent lipid probes. Hepatic hypertrophy was observed in the model, along with macrovesicular steatosis. Smil/Lstr combo administration effectively restored the lipid profile and alleviated hepatic hypertrophy in the HFD zebrafish. In oleic-acid stimulated hepatocytes, Smil/Lstr combo markedly reduced lipid accumulation and cell damage. Subsequently, based on zebrafish phenotypic screening, the natural phthalide senkyunolide I (SEI) was identified as a major molecule mediating the lipid-lowering activities of Smil/Lstr combo in the liver. Moreover, SEI upregulated the expression of the lipid metabolism regulator PPARα and downregulated fatty acid translocase CD36, while a PPARα antagonist sufficiently blocked the regulatory effect of SEI on hepatic steatosis. Finally, the roles of SEI on hepatic lipid accumulation and PPARα signaling were further verified in the hamster model.
    CONCLUSIONS: We proposed a zebrafish-based screening strategy for modulators of hepatic steatosis and discovered the regulatory roles of Smil/Lstr combo and its component SEI on liver lipid accumulation and PPARα signaling, suggesting their potential value as novel candidates for NAFLD treatment.
    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
    牦牛(Bosgrunniens)是一种特殊的牲畜品种,主要分布在中国的青藏高原。肉牛肌内脂肪(IMF)含量是肉质的重要指标。在这项研究中,RNA-Seq和Protein-Seq分别用于对4岁牛的背最长肌(LD)组织的转录组和蛋白质组进行测序,在相同的育肥条件下,IMF含量存在显着差异。五个重叠基因(MYL3、ACADS、L2HGDH,使用组合分析筛选IGFN1和ENSBGRG00000000-926)。功能验证试验表明,关键基因ACADS抑制牦牛肌内前脂肪细胞(YIMA)的分化和增殖,促进线粒体生物发生基因表达,并增加线粒体膜电位(MMP)。此外,共转染实验进一步证明,干扰ACADS逆转了PPARα激动剂促进脂质分化的作用。总之,ACADS可能通过调节PPARα信号通路抑制YIAM中的脂质沉积。这些发现为牦牛肉品质的分子机制提供了见解。
    The Yak (Bos grunniens) is a special breed of livestock predominantly distributed in the Qinghai-Tibet Plateau of China. Intramuscular fat (IMF) content in beef cattle is a vital indicator of meat quality. In this study, RNA-Seq and Protein-Seq were respectively employed to sequence the transcriptome and proteome of the longissimus dorsi (LD) tissue from 4-year-old yaks with significant differences in IMF content under the same fattening conditions. Five overlapping genes (MYL3, ACADS, L2HGDH, IGFN1, and ENSBGRG00000000-926) were screened using combined analysis. Functional verification tests demonstrated that the key gene ACADS inhibited yak intramuscular preadipocyte (YIMA) differentiation and proliferation, promoted mitochondrial biogenesis gene expression, and increased the mitochondrial membrane potential (MMP). Furthermore, co-transfection experiments further demonstrated that interfering with ACADS reversed the effect of PPARα agonists in promoting lipid differentiation. In conclusion, ACADS potentially inhibits lipid deposition in YIAMs by regulating the PPARα signalling pathway. These findings offer insights into the molecular mechanisms underlying yak meat quality.
    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
    内质网(ER)应激是通过增加从头脂肪生成引起肝脂肪变性的主要原因。叉头盒O6(FoxO6)是介导胰岛素信号传导至葡萄糖和脂质代谢的转录因子。因此,失调的FoxO6参与肝脏脂肪生成。这项研究阐明了FoxO6在体内和体外ER应激诱导的肝脂肪变性中的作用。在组成型活性FoxO6等位基因过表达的小鼠和FoxO6-null小鼠中监测肝ER应激反应和β-氧化。对于体外研究,高葡萄糖处理过表达组成型活性FoxO6和FoxO6-siRNA的肝细胞,和测量脂质代谢变化。ER应激诱导的FoxO6活化在体内抑制肝脏β-氧化。过氧化物酶体增殖物激活受体α(PPARα)在组成型活性FoxO6等位基因中的表达和转录活性显著降低。否则,抑制β-氧化基因在FoxO6-siRNA和FoxO6-KO小鼠中减少。我们的数据显示FoxO6诱导的肝脏脂质积累受到胰岛素信号的负调控。高糖治疗作为高血糖症引起内质网应激诱导基因的表达,肝细胞中的FoxO6活化使其恶化。然而,高糖介导的ER应激通过PPARα和FoxO6之间的相互作用抑制β-氧化基因的表达,这与体内研究中的发现一致-脂质分解代谢也受FoxO6调节。此外,胰岛素抵抗通过FoxO6和PPARα之间的相互作用抑制b-氧化促进肝脏脂肪变性,which,由于高血糖引起的内质网应激,损害胰岛素信号。主要信息:我们最初的目的是在分子水平上描述PPARα调节和转录因子FoxO6通路与脂质代谢的相互关系。高糖促进FoxO6激活诱导肝细胞脂质积累的证据。PPARα激活胰岛素信号的作用。FoxO6在FoxO6过表达小鼠中通过PPARα失活在肝脏脂质积累中起关键作用。
    Endoplasmic reticulum (ER) stress is a major cause of hepatic steatosis through increasing de novo lipogenesis. Forkhead box O6 (FoxO6) is a transcription factor mediating insulin signaling to glucose and lipid metabolism. Therefore, dysregulated FoxO6 is involved in hepatic lipogenesis. This study elucidated the role of FoxO6 in ER stress-induced hepatic steatosis in vivo and in vitro. Hepatic ER stress responses and β-oxidation were monitored in mice overexpressed with constitutively active FoxO6 allele and FoxO6-null mice. For the in vitro study, liver cells overexpressing constitutively active FoxO6 and FoxO6-siRNA were treated with high glucose, and lipid metabolism alterations were measured. ER stress-induced FoxO6 activation suppressed hepatic β-oxidation in vivo. The expression and transcriptional activity of peroxisome proliferator-activated receptor α (PPARα) were significantly decreased in the constitutively active FoxO6 allele. Otherwise, inhibiting β-oxidation genes were reduced in the FoxO6-siRNA and FoxO6-KO mice. Our data showed that the FoxO6-induced hepatic lipid accumulation was negatively regulated by insulin signaling. High glucose treatment as a hyperglycemia condition caused the expression of ER stress-inducible genes, which was deteriorated by FoxO6 activation in liver cells. However, high glucose-mediated ER stress suppressed β-oxidation gene expression through interactions between PPARα and FoxO6 corresponding to findings in the in vivo study-lipid catabolism is also regulated by FoxO6. Furthermore, insulin resistance suppressed b-oxidation through the interaction between FoxO6 and PPARα promotes hepatic steatosis, which, due to hyperglycemia-induced ER stress, impairs insulin signaling. KEY MESSAGES: Our original aims were to delineate the interrelation between the regulation of PPARα and the transcription factor FoxO6 pathway in relation to lipid metabolism at molecular levels. Evidence on high glucose promoted FoxO6 activation induced lipid accumulation in liver cells. The effect of PPARα activation of the insulin signaling. FoxO6 plays a pivotal role in hepatic lipid accumulation through inactivation of PPARα in FoxO6-overexpression mice.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号