PPARδ

PPAR δ
  • 文章类型: Journal Article
    易损动脉粥样硬化斑块破裂,致命的动脉粥样硬化血栓事件的主要原因,与全球死亡风险增加有关。过氧化物酶体增殖物激活受体δ(PPARδ)已被证明可以调节血管平滑肌细胞(SMC)表型转换,and,因此,动脉粥样硬化斑块稳定性。据报道,褪黑素在心血管疾病中起着有益的作用;然而,动脉粥样硬化斑块易损性改善的潜在机制尚不清楚.在这项研究中,我们评估了褪黑素在调节SMC表型转换中的作用及其对改善动脉粥样硬化斑块易损性的作用,并探讨了这一过程的潜在机制.我们分析了高胆固醇饮食(HCD)喂养的载脂蛋白E敲除(ApoE-/-)小鼠和人主动脉SMC(HASMC)的动脉粥样硬化斑块易损性特征和SMC表型转变标志物。褪黑素减少动脉粥样硬化斑块大小和坏死核心区域,同时增加胶原含量,纤维帽厚度,平滑肌α-肌动蛋白阳性细胞覆盖在斑块上,这些都是已知的易损斑块的表型特征。在动脉粥样硬化病变中,褪黑素显著降低合成SMC表型和KLF4的表达,增加PPARδ的表达,但不是PPARα和PPARγ,在HCD喂养的ApoE-/-小鼠中。这些结果随后在褪黑激素处理的HASMC中得到证实。使用PPARδ沉默和免疫沉淀测定的进一步分析表明,PPARδ在褪黑激素诱导的SMC表型从合成转换为收缩转换中起作用。总的来说,我们提供了第一个证据表明,褪黑激素通过增强PPARδ介导的SMC表型转换来介导其对斑块失稳的保护作用,从而表明褪黑素治疗动脉粥样硬化的潜力。
    Vulnerable atherosclerotic plaque rupture, the leading cause of fatal atherothrombotic events, is associated with an increased risk of mortality worldwide. Peroxisome proliferator-activated receptor delta (PPARδ) has been shown to modulate vascular smooth muscle cell (SMC) phenotypic switching, and, hence, atherosclerotic plaque stability. Melatonin reportedly plays a beneficial role in cardiovascular diseases; however, the mechanisms underlying improvements in atherosclerotic plaque vulnerability remain unknown. In this study, we assessed the role of melatonin in regulating SMC phenotypic switching and its consequential contribution to the amelioration of atherosclerotic plaque vulnerability and explored the mechanisms underlying this process. We analyzed features of atherosclerotic plaque vulnerability and markers of SMC phenotypic transition in high-cholesterol diet (HCD)-fed apolipoprotein E knockout (ApoE-/-) mice and human aortic SMCs (HASMCs). Melatonin reduced atherosclerotic plaque size and necrotic core area while enhancing collagen content, fibrous cap thickness, and smooth muscle alpha-actin positive cell coverage on the plaque cap, which are all known phenotypic characteristics of vulnerable plaques. In atherosclerotic lesions, melatonin significantly decreased the synthetic SMC phenotype and KLF4 expression and increased the expression of PPARδ, but not PPARα and PPARγ, in HCD-fed ApoE-/- mice. These results were subsequently confirmed in the melatonin-treated HASMCs. Further analysis using PPARδ silencing and immunoprecipitation assays revealed that PPARδ plays a role in the melatonin-induced SMC phenotype switching from synthetic to contractile. Collectively, we provided the first evidence that melatonin mediates its protective effect against plaque destabilization by enhancing PPARδ-mediated SMC phenotypic switching, thereby indicating the potential of melatonin in treating atherosclerosis.
    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
    细胞因子白细胞介素-38(IL-38),最近发现的IL-1家族成员,已被证明可以调节炎症并改善肥胖个体的肝内质网应激和脂质代谢。然而,其对骨骼肌细胞胰岛素信号的影响及其潜在机制尚不清楚.使用棕榈酸盐治疗建立体外肥胖模型,进行蛋白质印迹分析以评估靶蛋白。商业试剂盒用于测量培养的肌细胞中的葡萄糖摄取。我们的研究表明,IL-38治疗减轻了胰岛素信号的损害,包括IRS-1和Akt磷酸化,并且在棕榈酸盐处理的C2C12肌细胞中葡萄糖摄取增加。在棕榈酸盐处理后,在这些细胞中观察到STAT3介导的信号传导和氧化应激水平增加。IL-38治疗逆转了这些作用。此外,IL-38处理上调PPARδ的表达,SIRT1和抗氧化剂。使用适当的siRNA敲低PPARδ或SIRT1消除了IL-38对胰岛素信号的影响,氧化应激,和STAT3依赖性途径。这些结果表明IL-38通过PPARδ/SIRT1抑制骨骼肌细胞中STAT3介导的信号传导和氧化应激来减轻胰岛素抵抗。这项研究提供了基本证据来支持IL-38作为治疗胰岛素抵抗和2型糖尿病的安全治疗剂的潜在用途。
    The cytokine interleukin-38 (IL-38), a recently discovered member of the IL-1 family, has been shown to regulate inflammation and improve hepatic endoplasmic reticulum stress and lipid metabolism in individuals with obesity. However, its impact on insulin signaling in skeletal muscle cells and the underlying mechanisms remain unclear. In vitro obesity models were established using palmitate treatment, and Western blot analysis was performed to assess target proteins. Commercial kits were used to measure glucose uptake in cultured myocytes. Our study showed that IL-38 treatment alleviated the impairment of insulin signaling, including IRS-1 and Akt phosphorylation, and increased glucose uptake in palmitate-treated C2C12 myocytes. Increased levels of STAT3-mediated signaling and oxidative stress were observed in these cells following palmitate treatment, and these effects were reversed by IL-38 treatment. In addition, IL-38 treatment upregulated the expression of PPARδ, SIRT1 and antioxidants. Knockdown of PPARδ or SIRT1 using appropriate siRNAs abrogated the effects of IL-38 on insulin signaling, oxidative stress, and the STAT3-dependent pathway. These results suggest that IL-38 alleviates insulin resistance by inhibiting STAT3-mediated signaling and oxidative stress in skeletal muscle cells through PPARδ/SIRT1. This study provides fundamental evidence to support the potential use of IL-38 as a safe therapeutic agent for the treatment of insulin resistance and type 2 diabetes.
    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
    具有高水平活性氧(ROS)的功能异常的骨髓(BM)内皮祖细胞(EPCs)是导致急性白血病或同种异体移植后骨髓增生异常肿瘤的移植物功能不良(PGF)患者造血功能缺陷的原因。然而,BMEPCs调节其细胞内ROS水平和支持造血能力的潜在机制尚未得到很好的阐明。在这里,我们证明了过氧化物酶体增殖物激活受体δ(PPARδ)的水平降低,脂质激活的核受体,在PGF患者的BMEPCs中,与具有良好移植物功能(GGF)的患者进行了比较。体外测定进一步确定PPARδ敲低有助于减少和功能失调的BMEPCs,以支持造血能力受损为特征,通过PPARδ过表达恢复。此外,GW501516,PPARδ的激动剂,在体外和体内修复了5-氟尿嘧啶(5FU)引发的受损BMEPCs。临床上,GW501516对PPARδ的激活使化疗后完全缓解(CR)的PGF患者或急性白血病患者受损的BMEPCs受益。机械上,我们发现NADPH氧化酶(NOXs)的表达增加,主要的ROS生成酶,可能导致BMEPC中ROS水平升高,PPARδ不足可能通过ROS/p53途径引发BMEPC损伤。总的来说,我们发现PPARδ缺陷有助于BMEPC功能障碍,而骨髓EPCs中PPARδ的激活可改善骨髓抑制治疗后的造血支持能力,这可能不仅为白血病患者而且为其他癌症患者提供潜在的治疗靶点。
    Dysfunctional bone marrow (BM) endothelial progenitor cells (EPCs) with high levels of reactive oxygen species (ROS) are responsible for defective hematopoiesis in poor graft function (PGF) patients with acute leukemia or myelodysplastic neoplasms post-allotransplant. However, the underlying mechanism by which BM EPCs regulate their intracellular ROS levels and the capacity to support hematopoiesis have not been well clarified. Herein, we demonstrated decreased levels of peroxisome proliferator-activated receptor delta (PPARδ), a lipid-activated nuclear receptor, in BM EPCs of PGF patients compared with those with good graft function (GGF). In vitro assays further identified that PPARδ knockdown contributed to reduced and dysfunctional BM EPCs, characterized by the impaired ability to support hematopoiesis, which were restored by PPARδ overexpression. Moreover, GW501516, an agonist of PPARδ, repaired the damaged BM EPCs triggered by 5-fluorouracil (5FU) in vitro and in vivo. Clinically, activation of PPARδ by GW501516 benefited the damaged BM EPCs from PGF patients or acute leukemia patients in complete remission (CR) post-chemotherapy. Mechanistically, we found that increased expression of NADPH oxidases (NOXs), the main ROS-generating enzymes, may lead to elevated ROS level in BM EPCs, and insufficient PPARδ may trigger BM EPC damage via ROS/p53 pathway. Collectively, we found that defective PPARδ contributes to BM EPC dysfunction, whereas activation of PPARδ in BM EPCs improves their hematopoiesis-supporting ability after myelosuppressive therapy, which may provide a potential therapeutic target not only for patients with leukemia but also for those with other cancers.
    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
    骨质疏松症在绝经后妇女和老年人中尤为普遍。在本研究中,我们研究了新型小分子E0924G(N-(4-甲氧基-吡啶-2-基)-5-甲基呋喃-2-甲酰胺)对骨质疏松的影响.E0924G显著增加骨保护素(OPG)和runt相关转录因子2(RUNX2)的蛋白表达水平,从而显著促进MC3T3-E1细胞成骨。E0924G还显着降低了破骨细胞的分化,并抑制了NF-κB配体(RANKL)诱导的RAW264.7巨噬细胞的破骨细胞的骨吸收和F-肌动蛋白环的形成。重要的是,与OVX对照组或SAMR1小鼠相比,在卵巢切除(OVX)大鼠和SAMP6老年小鼠中口服E0924G显着增加了骨矿物质密度并减少了骨丢失。进一步的机理研究表明,E0924G可以结合并激活过氧化物酶体增殖物激活受体δ(PPARδ),当PPARδ被敲除或抑制时,E0924G诱导的促成骨细胞作用和对破骨细胞分化的抑制作用明显消失。总之,这些数据强烈表明,E0924G有可能通过激活PPARδ信号通路对骨形成和骨吸收进行双重调节,从而预防OVX诱导的和年龄相关的骨质疏松症.
    Osteoporosis is particularly prevalent among postmenopausal women and the elderly. In the present study, we investigated the effect of the novel small molecule E0924G (N-(4-methoxy-pyridine-2-yl)-5-methylfuran-2-formamide) on osteoporosis. E0924G significantly increased the protein expression levels of osteoprotegerin (OPG) and runt-related transcription factor 2 (RUNX2), and thus significantly promoted osteogenesis in MC3T3-E1 cells. E0924G also significantly decreased osteoclast differentiation and inhibited bone resorption and F-actin ring formation in receptor activator of NF-κB ligand (RANKL)-induced osteoclasts from RAW264.7 macrophages. Importantly, oral administration of E0924G in both ovariectomized (OVX) rats and SAMP6 senile mice significantly increased bone mineral density and decreased bone loss compared to OVX controls or SAMR1 mice. Further mechanistic studies showed that E0924G could bind to and then activate peroxisome proliferator-activated receptor delta (PPARδ), and the pro-osteoblast effect and the inhibition of osteoclast differentiation induced by E0924G were significantly abolished when PPARδ was knocked down or inhibited. In conclusion, these data strongly suggest that E0924G has the potential to prevent OVX-induced and age-related osteoporosis by dual regulation of bone formation and bone resorption through activation of the PPARδ signaling pathway.
    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
    骨关节炎(OA)是最常见的退行性关节疾病,和PPARs参与其发病机制;然而,PPARδ变化影响OA发病机制的具体机制尚待发现。这项研究的目的是确定PPARδ如何影响OA的发生和发展。体外,我们发现PPARδ激活可改善IL-1β刺激的OA软骨细胞的凋亡和细胞外基质(ECM)降解。此外,PPARδ激活可能通过调节AKT/mTOR信号传导来部分调节软骨细胞自噬和凋亡。在体内,向关节腔内注射PPARδ激动剂可改善ECM降解,去稳定内侧半月板(DMM)产生的大鼠OA模型的凋亡和自噬,最终关节软骨延迟退变。因此,靶向PPARδ治疗OA可能是一种可能性。
    Osteoarthritis (OA) is the most prevalent degenerative joint disease, and PPARs are involved in its pathogenesis; however, the specific mechanisms by which changes in PPARδ impact the OA pathogenesis yet to be discovered. The purpose of this study was to ascertain how PPARδ affects the onset and development of OA. In vitro, we found that PPARδ activation ameliorated apoptosis and extracellular matrix (ECM) degradation in OA chondrocytes stimulated by IL-1β. In addition, PPARδ activation may modulate AKT/mTOR signaling to partially regulate chondrocyte autophagy and apoptosis. In vivo, injection of PPARδ agonist into the articular cavity improved ECM degradation, apoptosis and autophagy in rats OA models generated by destabilization medial meniscus (DMM), eventually delayed degeneration of articular cartilage. Thus, targeting PPARδ for OA treatment may be a possibility.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    神经炎症涉及小胶质细胞表型的不同改变,含有恶性促炎M1表型和神经保护性抗炎M表型。目前,没有有效的治疗方法来调节这种改变。通过qPCR检测受褪黑素影响的原代小胶质细胞的M1/M2标记。通过西方印迹探索功能活动,荧光素酶活性,EMSA,和ChIP分析。通过分子对接和LIGPLOT分析评估结构相互作用。通过超微结构TEM检查ER应力检测,calapin活动,和ERSE测定。功能性神经行为评估用于研究褪黑素对体内神经炎症的影响。褪黑素靶向过氧化物酶体增殖物激活受体δ(PPARδ)活性,增强LPS刺激的从M1到M2表型的极化改变,从而抑制原发性小胶质细胞中NFκB-IKKβ的活化。PPARδ激动剂L-165,041或过表达的PPARδ质粒(ov-PPARδ)显示相似的结果。分子对接筛选,动态仿真方法,褪黑素的生物学研究表明,激活位点位于PPARδ(磷酸-Thr256-PPARδ)。活化的小胶质细胞通过增强ER应激降低了PPARδ活性以及下游SIRT1的形成。褪黑激素,PPARδ激动剂和ov-PPARδ均有效逆转了上述作用。褪黑素通过调节LPS激活的小胶质细胞中的calapin活性和表达来阻断ER应激。此外,褪黑素或L-165,041通过阻断小胶质细胞活动改善了LPS加重的神经炎小鼠的神经行为缺陷,并促进M2型小胶质细胞的表型改变。褪黑素通过ER-应激依赖性PPARδ/SIRT1信号级联调节小胶质细胞活化,在体外和体内抑制神经炎症。该治疗策略是用于治疗神经炎症相关病症的令人鼓舞的药理学方法。
    Neuro-inflammation involves distinct alterations of microglial phenotypes, containing nocuous pro-inflammatory M1-phenotype and neuroprotective anti-inflammatory M-phenotype. Currently, there is no effective treatment for modulating such alterations. M1/M2 marker of primary microglia influenced by Melatonin were detected via qPCR. Functional activities were explored by western blotting, luciferase activity, EMSA, and ChIP assay. Structure interaction was assessed by molecular docking and LIGPLOT analysis. ER-stress detection was examined by ultrastructure TEM, calapin activity, and ERSE assay. The functional neurobehavioral evaluations were used for investigation of Melatonin on the neuroinflammation in vivo. Melatonin had targeted on Peroxisome Proliferator Activated Receptor Delta (PPARδ) activity, boosted LPS-stimulated alterations in polarization from the M1 to the M2 phenotype, and thereby inhibited NFκB-IKKβ activation in primary microglia. The PPARδ agonist L-165,041 or over-expression of PPARδ plasmid (ov-PPARδ) showed similar results. Molecular docking screening, dynamic simulation approaches, and biological studies of Melatonin showed that the activated site was located at PPARδ (phospho-Thr256-PPARδ). Activated microglia had lowered PPARδ activity as well as the downstream SIRT1 formation via enhancing ER-stress. Melatonin, PPARδ agonist and ov-PPARδ all effectively reversed the above-mentioned effects. Melatonin blocked ER-stress by regulating calapin activity and expression in LPS-activated microglia. Additionally, Melatonin or L-165,041 ameliorated the neurobehavioral deficits in LPS-aggravated neuroinflammatory mice through blocking microglia activities, and also promoted phenotype changes to M2-predominant microglia. Melatonin suppressed neuro-inflammation in vitro and in vivo by tuning microglial activation through the ER-stress-dependent PPARδ/SIRT1 signaling cascade. This treatment strategy is an encouraging pharmacological approach for the remedy of neuro-inflammation associated disorders.
    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
    全氟辛烷磺酸(PFOS)作为全氟烷基和多氟烷基物质(PFAS)的典型代表广泛分布在环境中,并在人体中广泛检测。尽管越来越多的证据表明全氟辛烷磺酸对男性生殖的有害影响,全氟辛烷磺酸对生精细胞的直接毒性和相关机制仍然知之甚少。本研究旨在探讨全氟辛烷磺酸对精子发生的直接影响及其分子机制。通过整合动物研究,转录组分析,在硅毒理学方法中,和体外验证研究,我们确定了导致PFOS诱导的生精损伤的分子启动事件和关键事件.小鼠实验表明,精母细胞与PFOS诱导的生精障碍有关,过氧化物酶体增殖物激活受体δ(PPARδ)的激活与PFOS给药小鼠的精母细胞丢失有关。用增加的PFOS梯度处理GC-2spd(ts)细胞,这与人群中全氟辛烷磺酸的环境和职业暴露水平有关。治疗72小时后,收获细胞用于RNA测序。转录组分析和基准剂量(BMD)建模确定内质网(ER)应激是PFOS介导的精母细胞凋亡的关键事件,并确定了内质网应激信号扰动的起点(PoD)。根据计算出的PoD值,进一步的生物信息学分析结合体外和体内验证表明,全氟辛烷磺酸通过激活小鼠精母细胞中的PPARδ引起代谢应激,导致Beclin1参与的肌醇1,4,5-三磷酸受体(IP3R)致敏。IP3R介导的内质网钙稳态的破坏引发内质网钙耗竭,导致暴露于PFOS的小鼠精母细胞内质网应激和凋亡。本研究系统研究了全氟辛烷磺酸对精子发生的直接影响,揭示了全氟辛烷磺酸致精子发生障碍的相关分子机制。为PFAS相关的男性生殖毒性提供新的见解和潜在的预防/治疗靶点。
    Perfluorooctane sulfonic acid (PFOS) as an archetypal representative of per- and polyfluoroalkyl substances (PFAS) is ubiquitously distributed in the environment and extensively detected in human bodies. Although accumulating evidence is suggestive of the deleterious effects of PFOS on male reproduction, the direct toxicity of PFOS towards spermatogenic cells and the relevant mechanisms remain poorly understood. The aims of the present study were to explore the direct effects and underlying molecular mechanisms of PFOS on spermatogenesis. Through integrating animal study, transcriptome profiling, in silico toxicological approaches, and in vitro validation study, we identified the molecular initiating event and key events contributing to PFOS-induced spermatogenic impairments. The mouse experiments revealed that spermatocytes were involved in PFOS-induced spermatogenic disorders and the activation of peroxisome proliferator-activated receptor delta (PPARδ) was linked to spermatocyte loss in PFOS-administrated mice. GC-2spd(ts) cells were treated with an increased gradient of PFOS, which was relevant to environmental and occupational exposure levels of PFOS in populations. Following 72-h treatment, cells was harvested for RNA sequencing. The transcriptome profiling and benchmark dose (BMD) modeling identified endoplasmic reticulum (ER) stress as the key event for PFOS-mediated spermatocyte apoptosis and determined the point-of-departure (PoD) for perturbations of ER stress signaling. Based on the calculated PoD value, further bioinformatics analyses combined with in vitro and in vivo validations showed that PFOS caused metabolic stress by activating PPARδ in mouse spermatocytes, which was responsible for Beclin 1-involved inositol 1,4,5-trisphosphate receptor (IP3R) sensitization. The disruption of IP3R-mediated ER calcium homeostasis triggered ER calcium depletion, leading to ER stress and apoptosis in mouse spermatocytes exposed to PFOS. This study systematically investigated the direct impacts of PFOS on spermatogenesis and unveiled the relevant molecular mechanism of PFOS-induced spermatogenic disorders, providing novel insights and potential preventive/therapeutic targets for PFAS-associated male reproductive toxicity.
    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
    Seladelpar,选择性PPARδ激动剂,改善人类肝脏疾病中肝损伤的标志物,但非酒精性脂肪性肝炎(NASH)和肝纤维化的组织学改善一直是任何单一药物的挑战。要了解免费代理如何与seladelpar合作以实现最佳结果,这项研究评估了NASH小鼠模型中的多种治疗方法(单独和组合)。用seladelpar治疗高脂肪淀粉素肝脏NASH(AMLN)饮食的小鼠12周,GLP-1-R(胰高血糖素样肽-1受体)激动剂利拉鲁肽,凋亡信号调节激酶1(ASK1)抑制剂selonsertib,法尼醇X受体(FXR)激动剂奥贝胆酸,并与seladelpar联合利拉鲁肽或selonsertib。Seladelpar治疗显着改善了肝功能的血浆标志物。Seladelpar单独或联合使用可明显减少肝纤维化(羟脯氨酸,新胶原蛋白合成率,纤维化和纤维化染色的mRNA指数)与媒介物和其他单一药剂相比。还观察到肝脏脂肪变性的稳健减少。Seladelpar产生了代谢基因表达的重组,特别是对于那些促进过氧化物酶体和线粒体脂质氧化的基因。总之,在该模型中,单独使用seladelpar和联合使用利拉鲁肽观察到NASH和NASH诱导的纤维化有显著改善.广泛的基因表达分析表明,seladelpar应与多种作用机制协同作用。
    Seladelpar, a selective peroxisome proliferator-activated receptor δ (PPARδ) agonist, improves markers of hepatic injury in human liver diseases, but histological improvement of nonalcoholic steatohepatitis (NASH) and liver fibrosis has been challenging with any single agent. To discover how complementary agents could work with seladelpar to achieve optimal outcomes, this study evaluated a variety of therapeutics (alone and in combination) in a mouse model of NASH. Mice on a high-fat amylin liver NASH (AMLN) diet were treated for 12 wk with seladelpar, GLP-1-R (glucagon-like peptide-1 receptor) agonist liraglutide, apoptosis signal-regulating kinase 1 (ASK1) inhibitor selonsertib, farnesoid X receptor (FXR) agonist obeticholic acid, and with seladelpar in combination with liraglutide or selonsertib. Seladelpar treatment markedly improved plasma markers of liver function. Seladelpar alone or in combination resulted in stark reductions in liver fibrosis (hydroxyproline, new collagen synthesis rate, mRNA indices of fibrosis, and fibrosis staining) compared with vehicle and the other single agents. Robust reductions in liver steatosis were also observed. Seladelpar produced a reorganization of metabolic gene expression, particularly for those genes promoting peroxisomal and mitochondrial lipid oxidation. In summary, substantial improvements in NASH and NASH-induced fibrosis were observed with seladelpar alone and in combination with liraglutide in this model. Broad gene expression analysis suggests seladelpar should be effective in concert with diverse mechanisms of action.NEW & NOTEWORTHY NASH is a chronic, progressive, and increasingly problematic liver disease that has been resistant to treatment with individual therapeutics. In this study using a diet-induced mouse model of NASH, we found that the PPARδ agonist seladelpar reduced fibrosis and NASH pathology alone and in combinations with a GLP-1-R agonist (liraglutide) or an ASK1 inhibitor (selonsertib). Liver transcriptome analysis comparing each agent and coadministration suggests seladelpar should be effective in combination with a variety of therapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    耐力运动是一种重要的健康调节剂。我们使用单核(sn)multiome测序技术,在四名受试者以70%VO2max运动前和运动后40分钟后3.5小时收集的人股外侧肌样本中,研究了人骨骼肌对急性耐力运动的细胞类型特异性适应。以及来自两个仰卧静息昼夜节律对照的匹配时间样本。从包括14种细胞类型的37,154个细胞核获得高质量的相同细胞RNA-seq和ATAC-seq数据。在肌肉纤维类型中,确定了共享和纤维类型特定的监管计划.单细胞电路分析在快速、慢纤维和中间纤维以及表达LUM的FAP细胞,涉及总共328个转录因子(TFs),作用于调节2,025个基因的改变的可接近性位点。这些数据和电路映射提供了对运动的潜在组织和代谢重塑反应过程的单细胞洞察。
    Endurance exercise is an important health modifier. We studied cell-type specific adaptations of human skeletal muscle to acute endurance exercise using single-nucleus (sn) multiome sequencing in human vastus lateralis samples collected before and 3.5 hours after 40 min exercise at 70% VO2max in four subjects, as well as in matched time of day samples from two supine resting circadian controls. High quality same-cell RNA-seq and ATAC-seq data were obtained from 37,154 nuclei comprising 14 cell types. Among muscle fiber types, both shared and fiber-type specific regulatory programs were identified. Single-cell circuit analysis identified distinct adaptations in fast, slow and intermediate fibers as well as LUM-expressing FAP cells, involving a total of 328 transcription factors (TFs) acting at altered accessibility sites regulating 2,025 genes. These data and circuit mapping provide single-cell insight into the processes underlying tissue and metabolic remodeling responses to exercise.
    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
    肥胖在中医中被定义为湿热证。黄连是一种用于清热和消除肥胖及其并发症的湿气的草药。小檗碱(BBR),黄连的主要活性化合物,显示抗肥胖效果。过氧化物酶体增殖物激活受体(PPARs)是一组调节能量代谢相关基因表达的核受体蛋白,脂质代谢,炎症,和脂肪生成。然而,PPAR是否参与BBR的抗肥胖作用尚不清楚.因此,本研究的目的是阐明PPARs在BBR治疗肥胖中的作用以及潜在的分子机制.我们的数据表明,BBR对PPARγ和PPARδ而不是PPARα的水平产生剂量依赖性调节。基因沉默和特异性拮抗剂治疗的结果表明,PPARδ是BBR作用的关键。在3T3L1前脂肪细胞中,BBR减少脂质积累;在高脂饮食(HFD)诱导的肥胖小鼠中,BBR减少了体重增加和白色脂肪组织质量,并校正了受干扰的生化参数,包括脂质水平和炎症和氧化标志物。BBR的体外和体内功效均被PPARδ特异性拮抗剂的存在所逆转。一项机制研究的结果表明,BBR可以激活3T3L1细胞和HFD小鼠的PPARδ,PPARδ内源性下游基因的显著上调证明了这一点。BBR激活后,PPARδ的转录功能被调用,表现出CCAAT/增强子结合蛋白α(Cebpα)和Pparγ启动子的负调节和血红素加氧酶-1(Ho-1)启动子的正介导。总之,这是关于BBR的新型抗肥胖机制的首次报道,这是通过PPARδ依赖性的脂质积累减少来实现的。
    Obesity is defined as a dampness-heat syndrome in traditional Chinese medicine. Coptidis Rhizoma is an herb used to clear heat and eliminate dampness in obesity and its complications. Berberine (BBR), the main active compound in Coptidis Rhizoma, shows anti-obesity effects. Peroxisome proliferator-activated receptors (PPARs) are a group of nuclear receptor proteins that regulate the expression of genes involved in energy metabolism, lipid metabolism, inflammation, and adipogenesis. However, whether PPARs are involved in the anti-obesity effect of BBR remains unclear. As such, the aim of this study was to elucidate the role of PPARs in BBR treatment on obesity and the underlying molecular mechanisms. Our data showed that BBR produced a dose-dependent regulation of the levels of PPARγ and PPARδ but not PPARα. The results of gene silencing and specific antagonist treatment demonstrated that PPARδ is key to the effect of BBR. In 3T3L1 preadipocytes, BBR reduced lipid accumulation; in high-fat-diet (HFD)-induced obese mice, BBR reduced weight gain and white adipose tissue mass and corrected the disturbed biochemical parameters, including lipid levels and inflammatory and oxidative markers. Both the in vitro and in vivo efficacies of BBR were reversed by the presence of a specific antagonist of PPARδ. The results of a mechanistic study revealed that BBR could activate PPARδ in both 3T3L1 cells and HFD mice, as evidenced by the significant upregulation of PPARδ endogenous downstream genes. After activating by BBR, the transcriptional functions of PPARδ were invoked, exhibiting negative regulation of CCAAT/enhancer-binding protein α (Cebpα) and Pparγ promoters and positive mediation of heme oxygenase-1 (Ho-1) promoter. In summary, this is the first report of a novel anti-obesity mechanism of BBR, which was achieved through the PPARδ-dependent reduction in lipid accumulation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号