mitofusin-2

Mitofusin - 2
  • 文章类型: Journal Article
    利达霉素(LDM)已被证实具有很强的抗胰腺癌作用,并可影响胰腺癌细胞的线粒体功能。Mitofusin-2(Mfn2)位于线粒体的外膜,Mfn2目前被认为在胰腺癌的癌症抑制中起作用。为了探讨LDM的抗胰腺癌作用是否与Mfn2介导的线粒体自噬有关,生物信息学和体外细胞实验用于实验研究。实验结果表明,Mfn2与胰腺癌线粒体自噬相关。Lidamycin可以上调胰腺癌中Mfn2的表达,影响EMT的进程,影响活性氧和线粒体膜电位的水平,并增加线粒体自噬标记蛋白BNIP3L和Beclin1的表达。这些结果证明Mfn2通过调节Mfn2的表达影响胰腺癌细胞的线粒体自噬。
    Lidamycin (LDM) has been confirmed to have a strong anti-pancreatic cancer effect and can affect the mitochondrial function of pancreatic cancer cells. Mitofusin-2 (Mfn2) is located in the outer membrane of mitochondria, and Mfn2 is currently believed to play a role in cancer inhibition in pancreatic cancer. In order to explore whether the anti-pancreatic cancer effect of LDM is related to Mfn2-mediated mitophagy, Bioinformatics and in vitro cell experiments are used for experimental research. The experimental results demonstrated that Mfn2 is correlated with mitochondrial autophagy in pancreatic cancer. Lidamycin can increase the expression of Mfn2 in pancreatic cancer and affect the process of EMT, affect the level of reactive oxygen species and mitochondrial membrane potential, and increase the expression of mitochondrial autophagy marker proteins BNIP3L and Beclin1. These results demonstrate that Mfn2 affects mitophagy in pancreatic cancer cells by regulating the expression of Mfn2.
    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
    目的:Mitofusin-2(MFN2)是一种线粒体膜蛋白,在调节线粒体融合和细胞代谢中起关键作用。为了进一步阐明MFN2的影响,本研究旨在探讨其对肝细胞癌(HCC)细胞功能的意义及其在介导化疗敏感性中的潜在作用。
    方法:本研究调查了沉默和过表达MFN2对存活的影响,扩散,入侵和迁移能力,和索拉非尼对MHCC97-L肝癌细胞的耐药性。使用XAV939(β-连环蛋白抑制剂)和HLY78(β-连环蛋白激活剂)进行另外的实验以进一步验证这些发现。
    结果:沉默MFN2可显著促进MHCC97-L细胞的存活和增殖,增强了他们的入侵和迁移能力,增加了索拉非尼的IC50,减少了TUNEL阳性细胞的百分比,并降低促凋亡蛋白的表达。此外,沉默MFN2显著诱导β-catenin的核易位,增加β-连环蛋白乙酰化水平,增强下游调节蛋白Snail1和波形蛋白的表达,同时抑制E-cadherin表达。相反,过表达MFN2逆转了上述MHCC97-L细胞中观察到的效应。结果证实沉默MFN2激活了β-catenin/上皮间质转化(EMT)通路,降低了细胞对索拉非尼的敏感性,可以通过XAV939治疗逆转。相反,MFN2过表达抑制β-catenin/EMT通路,增加细胞对索拉非尼的敏感性,这可以被HLY78改变。
    结论:肝癌细胞MFN2低表达促进β-catenin的核转位,从而激活EMT途径并介导对索拉非尼的抗性。
    OBJECTIVE: Mitofusin-2 (MFN2) is a mitochondrial membrane protein that plays a critical role in regulating mitochondrial fusion and cellular metabolism. To further elucidate the impact of MFN2, this study aimed to investigate its significance on hepatocellular carcinoma (HCC) cell function and its potential role in mediating chemosensitivity.
    METHODS: This study investigated the effects of silencing and overexpressing MFN2 on the survival, proliferation, invasion and migration abilities, and sorafenib resistance of MHCC97-L HCC cells. Additional experiments were conducted using XAV939 (a β-catenin inhibitor) and HLY78 (a β-catenin activator) to further validate these findings.
    RESULTS: Silencing MFN2 significantly promoted the survival and proliferation of MHCC97-L cells, enhanced their invasion and migration capacities, increased the IC50 of sorafenib, reduced the percentage of TUNEL-positive cells, and decreased the expression of proapoptotic proteins. Additionally, silencing MFN2 markedly induced the nuclear translocation of β-catenin, increased β-catenin acetylation levels and enhanced the expression of the downstream regulatory proteins Snail1 and Vimentin while inhibiting E-cadherin expression. Conversely, overexpressing MFN2 reversed the effects observed in MHCC97-L cells mentioned above. The results confirmed that silencing MFN2 activated the β-catenin/epithelial-mesenchymal transition (EMT) pathway and reduced the sensitivity of cells to sorafenib, which could be reversed by XAV939 treatment. Conversely, overexpression of MFN2 inhibited the β-catenin/EMT pathway and increased the sensitivity of cells to sorafenib, which could be altered by HLY78.
    CONCLUSIONS: Low expression of MFN2 in HCC cells promotes the nuclear translocation of β-catenin, thereby activating the EMT pathway and mediating resistance to sorafenib.
    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
    锌稳态对于维持氧化还原平衡至关重要,细胞增殖,和凋亡。然而,过量的锌暴露是有毒的,会导致线粒体功能障碍。在这项研究中,用不同浓度的Zn2+处理大鼠心肌细胞H9c2细胞,建立锌超负荷模型。我们的结果表明,锌过载增加LDH和活性氧(ROS)水平,导致细胞死亡,线粒体膜电位降低,线粒体功能和动力学受损。此外,锌过载激活PINK1/Parkin信号通路并通过ROS诱导线粒体自噬,而NAC抑制线粒体自噬并减弱PINK1/Parkin通路的激活,从而保留线粒体生物发生。此外,我们的数据还表明,Mfn2缺失增加了ROS的产生,并加剧了锌过载引起的细胞毒性。因此,我们的研究结果表明,Zn2+诱导的ROS产生导致线粒体自噬和线粒体功能障碍,损伤H9c2心肌细胞。此外,Mfn2可能在锌离子介导的内质网和线粒体相互作用中起关键作用。我们的结果为锌诱导的毒理学提供了新的视角。
    Zinc homeostasis is essential for maintaining redox balance, cell proliferation, and apoptosis. However, excessive zinc exposure is toxic and leads to mitochondrial dysfunction. In this study, we established a zinc overload model by treating rat cardiomyocyte H9c2 cells with Zn2+ at different concentrations. Our results showed that zinc overload increased LDH and reactive oxygen species (ROS) levels, leading to cell death, mitochondrial membrane potential decrease and impaired mitochondrial function and dynamics. Furthermore, zinc overload activated the PINK1/Parkin signaling pathway and induced mitochondrial autophagy via ROS, while NAC inhibited mitophagy and weakened the activation of PINK1/Parkin pathway, thereby preserving mitochondrial biogenesis. In addition, our data also showed that Mfn2 deletion increased ROS production and exacerbated cytotoxicity induced by zinc overload. Our results therefore suggest that Zn2+-induced ROS generation causes mitochondrial autophagy and mitochondrial dysfunction, damaging H9c2 cardiomyocytes. Additionally, Mfn2 may play a key role in zinc ion-mediated endoplasmic reticulum and mitochondrial interactions. Our results provide a new perspective on zinc-induced toxicology.
    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
    高脂肪消费促进肥胖的发展,这与各种慢性病有关。线粒体是真核细胞的能量工厂,通过微调的质量控制网络保持自我稳定。在本研究中,我们评估了高脂饮食(HFD)诱导的多器官线粒体超微结构和动力学蛋白表达的变化。C57BL/6J雄性小鼠饲喂HFD或正常饮食(ND)24周。与ND喂养的小鼠相比,HFD喂养的小鼠表现出体重增加,心肌细胞增大,肺纤维化,肝脂肪变性,肾脏和脾脏结构异常。心脏的细胞凋亡,肝脏,肾脏增加。观察到细胞脂滴沉积和线粒体变形。与线粒体生物发生(TFAM)相关的蛋白质,裂变(DRP1),自噬(LC3和LC3-II:LC3-I比率),线粒体自噬(PINK1)在不同器官表现出不同的变化。线粒体融合调节因子mitofusin-2(MFN2)和视神经萎缩1(OPA1)在多个器官中始终下调,甚至是脾脏.TOMM20和ATP5A蛋白在心脏中增强,骨骼肌,和脾脏,并在肾脏中减弱。这些结果表明,高脂肪喂养引起多个器官的病理变化,伴有线粒体超微结构损伤,以及MFN2和OPA1下调。线粒体融合蛋白可以成为治疗代谢疾病的有希望的靶标和/或标志物。
    High-fat consumption promotes the development of obesity, which is associated with various chronic illnesses. Mitochondria are the energy factories of eukaryotic cells, maintaining self-stability through a fine-tuned quality-control network. In the present study, we evaluated high-fat diet (HFD)-induced changes in mitochondrial ultrastructure and dynamics protein expression in multiple organs. C57BL/6J male mice were fed HFD or normal diet (ND) for 24 weeks. Compared with ND-fed mice, HFD-fed mice exhibited increased body weight, cardiomyocyte enlargement, pulmonary fibrosis, hepatic steatosis, renal and splenic structural abnormalities. The cellular apoptosis of the heart, liver, and kidney increased. Cellular lipid droplet deposition and mitochondrial deformations were observed. The proteins related to mitochondrial biogenesis (TFAM), fission (DRP1), autophagy (LC3 and LC3-II: LC3-I ratio), and mitophagy (PINK1) presented different changes in different organs. The mitochondrial fusion regulators mitofusin-2 (MFN2) and optic atrophy-1 (OPA1) were consistently downregulated in multiple organs, even the spleen. TOMM20 and ATP5A protein were enhanced in the heart, skeletal muscle, and spleen, and attenuated in the kidney. These results indicated that high-fat feeding caused pathological changes in multiple organs, accompanied by mitochondrial ultrastructural damage, and MFN2 and OPA1 downregulation. The mitochondrial fusion proteins may become promising targets and/or markers for treating metabolic disease.
    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
    肺动脉高压(PAH)主要是由于肺动脉平滑肌细胞(PASMCs)的异常增殖和凋亡抵抗所致。内皮祖细胞(EPC)衍生的外泌体(Exos)(EPC-Exos)缓解PAH。然而,关于EPC-Exos是否有助于PAH的病理过程的知识仍然不足,特别是PASMC的维修。本研究旨在确定EPC-Exos对增殖的影响,迁移,通过生物信息学分析和体外检测,探讨PASMCs细胞凋亡的可能分子机制。生物信息学分析表明,Ras信号通路和Exos在PAH中起着至关重要的作用。将PAH差异微小RNA(miRNA)和EPC-Exos中鉴定的miRNA相交以获得miR-21-5p。靶基因预测程序预测mitofusin-2(Mfn2)是miR-21-5p的潜在靶标。细胞实验表明,EPC-Exos减弱了生存力,扩散,迁移,缺氧条件下PASMCs的抗凋亡性。机械上,EPC-Exos显著上调Mfn2表达并减弱Ras-Raf-ERK1/2信号通路活性。总之,EPC-Exos抑制细胞活力,扩散,缺氧条件下PASMCs的迁移和促进细胞凋亡。有可能通过转运miR-21-5p来提高Mfn2的表达,直接或通过靶向Mfn2的表达来抑制Ras-Raf-ERK1/2信号通路。EPC-Exos是治疗PAH的潜在治疗候选物。
    Pulmonary arterial hypertension (PAH) mainly occurs as a result of abnormal proliferation and apoptosis resistance of pulmonary artery smooth muscle cells (PASMCs). Endothelial progenitor cell (EPC)-derived exosomes (Exos) (EPC-Exos) relieve PAH. However, there is still insufficient knowledge of whether EPC-Exos contribute to the pathological process of PAH, especially for PASMC repair. This study aimed to determine the effects of EPC-Exos on the proliferation, migration, and apoptosis of PASMCs and explore the possible underlying molecular mechanisms through bioinformatics analysis and in vitro testing. Bioinformatics analysis showed that the Ras signaling pathway and Exos were crucial in PAH. The PAH differential microRNAs (miRNAs) and miRNAs identified in EPC-Exos were intersected to obtain miR-21-5p. A target gene prediction program predicted mitofusin-2 (Mfn2) as a potential target of miR-21-5p. Cellular experiments demonstrated that EPC-Exos attenuated the viability, proliferation, migration, and apoptosis resistance of PASMCs under hypoxia. Mechanistically, EPC-Exos significantly upregulated Mfn2 expression and attenuated Ras-Raf-ERK1/2 signaling pathway activity. In conclusion, EPC-Exos suppress cell viability, proliferation, and migration and promote apoptosis in PASMCs under hypoxic conditions. It is possible to transport miR-21-5p to improve the expression of Mfn2 and inhibit the Ras-Raf-ERK1/2 signaling pathway directly or by targeting the expression of Mfn2. EPC-Exos are a potential therapeutic candidate for the treatment of PAH.
    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
    这项研究的目的是分析线粒体相关的内质网膜(MAMs)功能障碍是否介导了砷(As)引起的肺铁性凋亡和急性肺损伤(ALI)。由于暴露导致肺泡结构受损,小鼠炎症细胞浸润和肺功能下降。铁蛋白,铁过载的标志,增加了,GPX4,脂质过氧化指标,在暴露于As的肺和肺上皮细胞(MLE-12)中减少。用Fer-1(Fer-1)预处理,铁凋亡的抑制剂,缓解诱发的阿里。此外,在Fer-1预处理的小鼠中,抑制了诱导的非血红素铁沉积。此外,在Fer-1预处理的MLE-12细胞中,As触发的线粒体损伤和铁死亡得到缓解。机械上,在暴露于As的MLE-12细胞和小鼠肺中观察到PERK磷酸化和mitofusin-2(Mfn-2)减少。此外,在暴露于As的MLE-12细胞中,PERK和Mfn-2之间的相互作用下调,并且观察到MAMs功能障碍。有趣的是,PERK抑制剂和Mfn-2过表达均减轻了MLE-12细胞中As诱导的铁凋亡。此外,CLPP和mtHSP70,线粒体应激的标志物,被上调,线粒体ROS(mtROS)升高,线粒体膜电位(MMP)和ATP在暴露于As的MLE-12细胞中降低。甲磺酸线粒体醌(MitoQ),一种新型的线粒体靶向抗氧化剂,减轻As诱导的过量mtROS,线粒体应激,肺上皮细胞中的MAMs功能障碍。同样,体内实验表明,MitoQ预处理可以抵抗As诱导的肺铁凋亡和ALI。这些数据表明mtROS引发的MAMs功能障碍是,至少部分地,与As诱发的铁中毒和ALI有关。
    The goal of this study was to analyze whether mitochondria-associated endoplasmic reticulum membrane (MAMs) dysfunction mediated arsenic (As)-evoked pulmonary ferroptosis and acute lung injury (ALI). As exposure led to alveolar structure damage, inflammatory cell infiltration and pulmonary function decline in mice. Ferritin, the marker of iron overload, was increased, GPX4, the index of lipid peroxidation, was decreased in As-exposed lungs and pulmonary epithelial cells (MLE-12). Pretreatment with ferrostatin-1 (Fer-1), the inhibitor of ferroptosis, alleviated As-evoked ALI. In addition, As-induced non-heme iron deposition was inhibited in Fer-1 pretreated-mice. Moreover, As-triggered mitochondria damage and ferroptosis were mitigated in Fer-1 pretreated-MLE-12 cells. Mechanistically, PERK phosphorylation and mitofusin-2 (Mfn-2) reduction was observed in As-exposed MLE-12 cells and mice lungs. Additionally, the interaction between PERK and Mfn-2 was downregulated and MAMs dysfunction was observed in As-exposed MLE-12 cells. Intriguingly, PERK inhibitor and Mfn-2-overexpression all mitigated As-induced ferroptosis in MLE-12 cells. Additionally, CLPP and mtHSP70, the markers of mitochondrial stress, were upregulated, mitochondrial ROS (mtROS) was elevated, mitochondrial membrane potential (MMP) and ATP were decreased in As-exposed MLE-12 cells. Mitoquinone mesylate (MitoQ), a novel mitochondrial-targeted antioxidant, alleviated As-induced excess mtROS, mitochondrial stress, MAMs dysfunction in pulmonary epithelial cells. Similarly, in vivo experiments indicated that MitoQ pretreatment countered As-induced pulmonary ferroptosis and ALI. These data indicated that mtROS-initiated MAMs dysfunction is, at least partially, implicated in As-evoked ferroptosis and ALI.
    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
    Mitofusin-2(MFN2)是一种参与线粒体融合调控的GTP酶,这与各种生理和病理过程有关,包括能量代谢,细胞分化,和胚胎发育。然而,目前尚不清楚MFN2是否参与间充质干细胞(MSCs)的代谢和成骨分化。
    通过慢病毒构建MFN2敲低(MFN2-KD)和MFN2过表达(MFN2-OE)诱导的多能干细胞来源的间充质干细胞(iPSC-MSCs)。商业试剂盒用于检测糖酵解和氧化磷酸化(OXPHOS)速率。流式细胞术,蛋白质印迹,定量实时聚合酶链反应(qRT-PCR),RNA-seq,免疫荧光,和免疫沉淀用于表型和分子机制评估。
    我们证明了MFN2和Wnt/β-catenin信号通路调节iPSC-MSCs的糖酵解。MFN2的缺失促进了iPSC-MSCs的成骨分化,在有足够氧气的情况下进行有氧糖酵解,这增加了葡萄糖的消耗和乳酸的产生,以及糖酵解酶活性和基因表达。抑制Wnt/β-catenin信号通路可使MFN2-KDiPSC-MSCs的糖酵解速率和成骨分化增强正常化。MFN2-OEiPSC-MSC表现出相反的表型。
    下调MFN2通过Wnt/β-catenin信号通路介导的有氧糖酵解促进iPSC-MSCs的成骨分化。我们的研究揭示了MFN2在调节MSCs成骨分化和能量代谢方面的新功能,为牙槽骨修复和牙周再生治疗提供新的治疗靶点和理论依据。
    Mitofusin-2 (MFN2) is a kind of GTPase that participates in the regulation of mitochondrial fusion, which is related to a variety of physiological and pathological processes, including energy metabolism, cell differentiation, and embryonic development. However, it remains unclear whether MFN2 is involved in the metabolism and osteogenic differentiation of mesenchymal stem cells (MSCs).
    MFN2 knockdown (MFN2-KD) and MFN2-overexpressing (MFN2-OE) induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs) were constructed by lentivirus. The commercial kits were utilized to detect the glycolysis and oxidative phosphorylation (OXPHOS) rate. Flow cytometry, Western blot, quantitative real-time polymerase chain reaction (qRT-PCR), RNA-seq, immunofluorescence, and immunoprecipitation were employed for phenotype and molecular mechanism assessment.
    We demonstrated that MFN2 and Wnt/β-catenin signaling pathway regulated glycolysis of iPSC-MSCs. The lack of MFN2 promoted the osteogenic differentiation of iPSC-MSCs, and aerobic glycolysis in the presence of sufficient oxygen, which increased glucose consumption and lactic acid production, as well as the glycolytic enzyme activity and gene expression. Inhibiting the Wnt/β-catenin signaling pathway normalized the enhanced glycolytic rate and osteogenic differentiation of MFN2-KD iPSC-MSCs. MFN2-OE iPSC-MSCs displayed the opposite phenotype.
    Downregulating MFN2 promotes osteogenic differentiation of iPSC-MSCs through aerobic glycolysis mediated by the Wnt/β-catenin signaling pathway. Our research reveals the new function of MFN2 in regulating the osteogenic differentiation and energy metabolism of MSCs, which will provide a new therapeutic target and theoretical basis for alveolar bone repair and periodontal regenerative 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
    即使在糖尿病性心肌病中没有显著心肌细胞损失的情况下,心脏纤维化也是明显的,并且高葡萄糖(HG)水平独立地激活心脏成纤维细胞(CFs)并促进细胞增殖。线粒体呼吸和糖酵解,这是细胞增殖和线粒体相关膜(MAMs)的关键,严重参与了这个过程。然而,MAMs在HG诱导的CFs增殖中的作用和潜在机制尚不清楚。评价响应HG处理的CFs的增殖和凋亡。对MAM进行了量化,使用海马XF分析仪测定线粒体呼吸和细胞糖酵解水平。还确定了信号转导和转录激活因子3(STAT3)和mitofusin-2(MFN2)响应HG的变化,其对细胞增殖的影响,MAMs,和线粒体呼吸进行了评估。通过双荧光素酶报告基因测定(DLRA)和染色质免疫沉淀(CHIP)测定STAT3对MFN2转录的影响。HG诱导的CFs增殖增加了糖酵解水平和三磷酸腺苷(ATP)的产生,线粒体呼吸受到抑制。HG处理后MAMs和MFN2表达显著降低,MFN2表达的恢复抵消了HG对细胞增殖的影响,MAMs的线粒体呼吸,糖酵解水平,和ATP生产。线粒体STAT3含量未被HG改变,但磷酸化STAT3和细胞核STAT3水平升高。STAT3的抑制逆转了HG诱导的MFN2水平的降低。DLRA和CHIP通过与位于从转录起始位点计数的约-400bp处的MFN2启动子区域中的序列相互作用,在转录水平上直接证明了STAT3对MFN2的负调控。本研究表明,HG通过促进STAT3转位到细胞核独立诱导CFs增殖,通过以MFN2抑制的方式抑制MAMs,将线粒体呼吸转换为糖酵解以产生ATP。
    Cardiac fibrosis is evident even in the situation without a significant cardiomyocyte loss in diabetic cardiomyopathy and a high glucose (HG) level independently activates the cardiac fibroblasts (CFs) and promotes cell proliferation. Mitochondrial respiration and glycolysis, which are key for cell proliferation and the mitochondria-associated membranes (MAMs), are critically involved in this process. However, the roles and the underlying mechanism of MAMs in the proliferation of HG-induced CFs are largely unknown. The proliferation and apoptosis of CFs responding to HG treatment were evaluated. The MAMs were quantified, and the mitochondrial respiration and cellular glycolytic levels were determined using the Seahorse XF analyzer. The changes of signal transducer and activator of transcription 3 (STAT3) and mitofusin-2 (MFN2) in responding to HG were also determined, the effects of which on cell proliferation, MAMs, and mitochondrial respiration were assessed. The effects of STAT3 on MFN2 transcription was determined by the dual-luciferase reporter assay (DLRA) and chromatin immunoprecipitation (CHIP). HG-induced CFs proliferation increased the glycolytic levels and adenosine triphosphate (ATP) production, while mitochondrial respiration was inhibited. The MAMs and MFN2 expressions were significantly reduced on the HG treatment, and the restoration of MFN2 expression counteracted the effects of HG on cell proliferation, mitochondrial respiration of the MAMs, glycolytic levels, and ATP production. The mitochondrial STAT3 contents were not changed by HG, but the levels of phosphorylated STAT3 and nuclear STAT3 were increased. The inhibition of STAT3 reversed the reduction of MFN2 levels induced by HG. The DLRA and CHIP directly demonstrated the negative regulation of MFN2 by STAT3 at the transcription levels via interacting with the sequences in the MFN2 promoter region locating at about -400 bp counting from the start site of transcription. The present study demonstrated that the HG independently induced CFs proliferation via promoting STAT3 translocation to the nucleus, which switched the mitochondrial respiration to glycolysis to produce ATP by inhibiting MAMs in an MFN2-depression manner.
    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
    已有研究探讨了microRNAs(miRNAs)在脑缺血/再灌注损伤(CI/RI)中的作用。但miR-326-5pinCI/RI的具体机制尚不清楚。因此,本研究旨在揭示miR-326-5p/信号转导和转录激活因子-3(STAT3)轴在CI/RI中的作用机制。建立了两种模型(原代大鼠皮质神经元的氧和葡萄糖剥夺[OGD]和Sprague-Dawley大鼠的大脑中动脉阻塞[MCAO]),以在体外和体内模拟CI/RI,分别。用OGD处理的神经元和MCAO大鼠进行功能损失和获得测定。之后,生存能力,凋亡,OGD处理的神经元的氧化应激和线粒体膜电位进行了测试,以及病理变化,MCAO大鼠脑组织细胞凋亡和线粒体膜电位。Mitofusin-2(Mfn2),检测MCAO大鼠OGD处理的神经元和脑组织中miR-326-5p和STAT3的表达。Mfn2和miR-326-5p减少,STAT3在OGD处理的MCAO大鼠的神经元和脑组织中升高。miR-326-5p靶向并负调控STAT3表达。恢复miR-326-5p或降低STAT3增强活力,抑制细胞凋亡和氧化应激,在OGD处理的神经元中线粒体膜电位增加和Mfn2表达增加。上调miR-326-5p或下调STAT3缓解病理变化,抑制MCAO大鼠脑组织细胞凋亡,线粒体膜电位和Mfn2表达升高。本研究阐明上调的miR-326-5p或下调的STAT3通过升高Mfn2表达来保护抗CI/RI。
    Studies have greatly explored the role of microRNAs (miRNAs) in cerebral ischemia/reperfusion injury (CI/RI). But the specific mechanism of miR-326-5p in CI/RI is still elusive. Hence, this study was to unmask the mechanism of miR-326-5p/signal transducer and activator of transcription-3 (STAT3) axis in CI/RI. Two models (oxygen and glucose deprivation [OGD] in primary rat cortical neurons and middle cerebral artery occlusion [MCAO] in Sprague-Dawley rats) were established to mimic CI/RI in vitro and in vivo, respectively. Loss- and gain-of function assays were performed with OGD-treated neurons and with MCAO rats. Afterward, viability, apoptosis, oxidative stress and mitochondrial membrane potential in OGD-treated neurons were tested, as well as pathological changes, apoptosis and mitochondrial membrane potential in brain tissues of MCAO rats. Mitofusin-2 (Mfn2), miR-326-5p and STAT3 expression in OGD-treated neurons and in brain tissues of MCAO rats were detected. Mfn2 and miR-326-5p were reduced, and STAT3 was elevated in OGD-treated neurons and brain tissues of MCAO rats. miR-326-5p targeted and negatively regulated STAT3 expression. Restoring miR-326-5p or reducing STAT3 reinforced viability, inhibited apoptosis and oxidative stress, increased mitochondrial membrane potential and increased Mfn2 expression in OGD-treated neurons. Up-regulating miR-326-5p or down-regulating STAT3 relieved pathological changes, inhibited apoptosis and elevated mitochondrial membrane potential and Mfn2 expression in brain tissues of rats with MCAO. This study elucidates that up-regulated miR-326-5p or down-regulated STAT3 protects against CI/RI by elevating Mfn2 expression.
    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
    Obesity has become a global health issue, which can cause metabolic abnormalities systemically leading to increased morbidity of series diseases. At present, researches have presented obesity is a high-risk factor for colitis, and berberine shows positive therapeutic effect on colitis. Thus, we explored the beneficial effects and potential mechanisms of berberine on obesity-exacerbated colitis in this article. High-fat diet (HFD) exacerbated dextran sulfate sodium (DSS) induced colitis mice model was applied, the results showed that HFD promoted DSS-induced weight loss and inflammatory manifestations in intestine. The results of cytokines in serum and mRNA expression of inflammatory indicators in colon showed that HFD increased all their levels evidently, and the outcomes of Western blot analyses presented that HFD downregulated the MFN2 expression, inhibited the phosphorylation of AMPK as well as upregulated the BIP/Grp78 expression, while berberine could significantly reverse all these situations. In vitro, we stimulated Caco-2 cells with palmitic acid (PA) to replicate the lipotoxicity damage in the intestine, and the results presented that intervention therapy of berberine effectively enhanced the MFN2 expression, inhibited the mRNA levels of inflammatory factors, and reversed the PA induced protein level changes of AMPK and BIP/Grp78. In general, we proposed that berberine could regulate MFN2 to alleviate obesity exacerbated colitis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号