pyroptosis

焦亡
  • 文章类型: Journal Article
    PANoptosis是由先天性免疫传感器启动并由胱天蛋白酶和RIPK驱动的独特的先天性免疫炎性溶解细胞死亡途径。作为一条独特的途径,PANoptosis的执行不能通过靶向其他细胞死亡途径来阻碍,比如焦亡,凋亡,或坏死。相反,靶向关键的PANoposome组件可以作为防止这种形式的细胞死亡的策略。考虑到几种疾病的生理相关性,PANoptosis是一个关键的治疗靶点。值得注意的是,以前的研究主要集中在PANoptosis在癌症和感染性和炎症性疾病中的作用。相比之下,其在心血管疾病中的作用尚未得到全面讨论。这里,我们回顾了关于心血管疾病中PANoptosis的现有证据,包括心肌病,动脉粥样硬化,心肌梗塞,心肌炎,主动脉瘤和夹层,并探索各种靶向性全景的药物,其总体目标是提供一种新的补充方法来对抗心血管疾病。
    PANoptosis is a unique innate immune inflammatory lytic cell death pathway initiated by an innate immune sensor and driven by caspases and RIPKs. As a distinct pathway, the execution of PANoptosis cannot be hindered by targeting other cell death pathways, such as pyroptosis, apoptosis, or necroptosis. Instead, targeting key PANoptosome components can serve as a strategy to prevent this form of cell death. Given the physiological relevance in several diseases, PANoptosis is a pivotal therapeutic target. Notably, previous research has primarily focused on the role of PANoptosis in cancer and infectious and inflammatory diseases. By contrast, its role in cardiovascular diseases has not been comprehensively discussed. Here, we review the available evidence on PANoptosis in cardiovascular diseases, including cardiomyopathy, atherosclerosis, myocardial infarction, myocarditis, and aortic aneurysm and dissection, and explore a variety of agents that target PANoptosis, with the overarching goal of providing a novel complementary approach to combatting cardiovascular 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
    许多炎症性疾病,包括糖尿病肾病(DKD),与焦亡有关,一种炎症调节的细胞死亡。这项工作的目的是确定阿帕贝酮的效果,它靶向BRD4,溴结构域(BRD)和靶向溴结构域2的末端外(BET)蛋白的特异性抑制剂,对DKD的肾损伤。这项研究利用药理学和遗传学方法来研究阿帕贝酮对db/db小鼠和人肾小管上皮细胞(HK-2)的焦亡的影响。BRD4水平在暴露于高葡萄糖的HK-2细胞中和在db/db小鼠中升高。调节BRD4水平导致HK-2细胞和db/db小鼠中与NLRP3炎性体相关的炎性细胞因子的产生和细胞焦亡的变化。同样,阿帕贝酮通过抑制BRD4缓解了这些细胞过程.阿帕贝酮或BRD4siRNA通过PLK1基因启动子上的P300依赖性H3K27乙酰化抑制高糖下HK-2细胞中PLK1的表达,通过染色质免疫沉淀和免疫沉淀试验证明。总结一下,阿帕贝酮可缓解DKD的肾脏突出和纤维化。BRD4调节P300/H3K27ac/PLK1轴,导致NLRP3炎性体的激活和随后的细胞焦亡,炎症,和纤维化。这些结果可能为DKD治疗提供新的视角。
    Many inflammatory disorders, including diabetic kidney disease (DKD), are associated with pyroptosis, a type of inflammation-regulated cell death. The purpose of this work was to ascertain the effects of apabetalone, which targets BRD4, a specific inhibitor of the bromodomain (BRD) and extra-terminal (BET) proteins that target bromodomain 2, on kidney injury in DKD. This study utilized pharmacological and genetic approaches to investigate the effects of apabetalone on pyroptosis in db/db mice and human tubular epithelial cells (HK-2). BRD4 levels were elevated in HK-2 cells exposed to high glucose and in db/db mice. Modulating BRD4 levels led to changes in the generation of inflammatory cytokines and cell pyroptosis linked to NLRP3 inflammasome in HK-2 cells and db/db mice. Likewise, these cellular processes were mitigated by apabetalone through inhibition BRD4. Apabetalone or BRD4 siRNA suppressed PLK1 expression in HK-2 cells under high glucose by P300-dependent H3K27 acetylation on the PLK1 gene promoter, as demonstrated through chromatin immunoprecipitation and immunoprecipitation assays. To summarize, apabetalone relieves renal proptosis and fibrosis in DKD. BRD4 regulates the P300/H3K27ac/PLK1 axis, leading to the activation of the NLRP3 inflammasome and subsequent cell pyroptosis, inflammation, and fibrosis. These results may provide new perspectives on DKD 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
    焦亡,被称为程序性细胞死亡的一种典型模式,通常的特征是裂解的gasdermin家族(GSDMs)在细胞膜上形成孔并诱导细胞破裂,在此过程中还发现了天冬氨酸特异性蛋白酶(caspases)的激活。糖尿病肾病(DKD)是由肾脏糖尿病的并发症引起的,肾脏最重要的功能,肾小球滤过率(GFR),发生下降到低于90%的通常,甚至导致肾功能衰竭在严重的情况下。高血糖引起的持续炎症状态暗示了DKD的关键病理,越来越多的证据表明,焦亡是这种慢性免疫介导的炎症性疾病的重要原因。目前,GSDM的扩展发现,焦亡,它与先天免疫的联系更具吸引力,需要大量的研究来理清焦亡在DKD病理学中的意义。在这次审查中,我们梳理了关于焦亡的经典研究和新发现,刺破了DKD中新型的焦亡觉醒,并集中在DKD治疗中焦亡的重要性,旨在为DKD提供新的研究靶点和治疗策略。
    Pyroptosis, known as one typical mode of programmed cell death, is generally characterized by the cleaved gasdermin family (GSDMs) forming pores in the cell membrane and inducing cell rupture, and the activation of aspartate-specific proteases (caspases) has also been found during this process. Diabetic Kidney Disease (DKD) is caused by the complication of diabetes in the kidney, and the most important kidney\'s function, Glomerular Filtration Rate (GFR), happens to drop to less than 90% of its usual and even lead to kidney failure in severe cases. The persistent inflammatory state induced by high blood glucose implies the key pathology of DKD, and growing evidence shows that pyroptosis serves as a significant contributor to this chronic immune-mediated inflammatory disorder. Currently, the expanded discovery of GSDMs, pyroptosis, and its association with innate immunity has been more attractive, and overwhelming research is needed to sort out the implication of pyroptosis in DKD pathology. In this review, we comb both classical studies and newly founds on pyroptosis, prick off the novel awakening of pyroptosis in DKD, and center on the significance of pyroptosis in DKD treatment, aiming to provide new research targets and treatment strategies on DKD.
    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
    心脏骤停(CA)和心肺复苏(CPR)后的脑损伤是导致神经功能障碍和死亡的主要原因。本研究旨在探讨组蛋白去乙酰化酶6(HDAC6)在大鼠CA/CPR后神经功能恢复中的作用机制。通过CA/CPR治疗建立大鼠模型。将腺病毒包装的sh-HDAC6注射到尾静脉中。评价大鼠的神经功能,生存时间,神经功能评分,血清NSE/S100B,测定脑含水量,进行Morris水迷宫试验。HDAC6,microRNA(miR)-138-5p,Nod样受体蛋白3(NLRP3),并通过实时定量聚合酶链反应或Westernblot测定促性腺激素水平。通过染色质免疫沉淀检查miR-138-5p启动子上的HDAC6和H3K9ac富集。通过双荧光素酶报告基因测定分析miR-138-5p-NLRP3结合。NLRP3炎性体用尼日利亚霉素钠盐活化。CPR治疗后,HDAC6高表达,而miR-138-5p下调。HDAC6下调改善了神经功能并减少了焦亡。HDAC6在miR-138-5p启动子脱乙酰H3K9ac上的富集,抑制miR-138-5p,并促进NLRP3介导的焦亡。下调miR-138-5p部分逆转了CPR后HDAC6抑制的保护作用。在结论中,HDAC6在miR-138-5p启动子脱乙酰H3K9ac上的富集,抑制miR-138-5p表达并促进NLRP3介导的焦亡,心肺复苏后大鼠神经功能障碍恶化。
    Brain injury after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) is the leading cause of neurological dysfunction and death. This study aimed to explore the mechanism of histone deacetylase 6 (HDAC6) in neurofunctional recovery following CA/CPR in rats. A rat model was established by CA/CPR treatment. Adenovirus-packaged sh-HDAC6 was injected into the tail vein. To evaluate the neurofunction of rats, survival time, neurofunctional scores, serum NSE/S100B, and brain water content were measured and Morris water maze test was performed. HDAC6, microRNA (miR)-138-5p, Nod-like receptor protein 3 (NLRP3), and pyroptotic factors levels were determined by real-time quantitative polymerase chain reaction or Western blot assay. HDAC6 and H3K9ac enrichment on miR-138-5p promoter were examined by chromatin immunoprecipitation. miR-138-5p-NLRP3 binding was analyzed by dual-luciferase reporter assay. NLRP3 inflammasome was activated with nigericin sodium salt. After CPR treatment, HDAC6 was highly expressed, while miR-138-5p was downregulated. HDAC6 downregulation improved neurofunction and reduced pyroptosis. HDAC6 enrichment on the miR-138-5p promoter deacetylated H3K9ac, inhibiting miR-138-5p, and promoting NLRP3-mediated pyroptosis. Downregulating miR-138-5p partially reversed the protective effect of HDAC6 inhibition after CPR. In Conclusion, HDAC6 enrichment on miR-138-5p promoter deacetylated H3K9ac, inhibiting miR-138-5p expression and promoting NLRP3-mediated pyroptosis, worsening neurological dysfunction in rats after CPR.
    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:小关节骨关节炎(FJOA)是一种常见的腰椎骨关节炎,其特征是小关节软骨的退变。补肾活血汤(BSHXD)对OA有较好的治疗作用。我们的工作旨在进一步探讨含BSHXD的血清(BSHXD-CS)对FJOA的药理作用,并确定潜在的机制。
    方法:为了建立FJOA细胞模型,用LPS处理原代大鼠软骨细胞。采用qRT-PCR和westernblot检测mRNA和蛋白的表达,分别。通过ELISA测量促炎细胞因子的分泌水平。通过CCK8测定确定细胞活力。试剂盒检测到全球m6A水平,和NLRP3mRNAm6A水平通过Me-RIP测定法测定。通过RIP和RNA下拉法分析分子相互作用。
    结果:BSHXD-CS治疗减轻了LPS诱导的细胞损伤,炎症,NLRP3炎性小体和软骨细胞的焦亡(均p<0.05)。LPS诱导的NLRP3在软骨细胞中的上调与其高m6A修饰水平有关(p<0.05)。还观察到BSHXD-CS通过抑制STAT3减少了LPS诱导的软骨细胞中的m6A修饰(所有p<0.05),提示BSHXD-CS可以通过m6A依赖性方式抑制NLRP3表达。此外,DAA,一种M6A特异性抑制剂,被证明可以增强BSHXD-CS对LPS攻击的脓毒性的保护作用(均p<0.05)。
    结论:BSHXD-CS通过减少RNAm6A修饰,抑制软骨细胞中NLRP3炎性体活化和焦亡,从而抑制OA进展。
    BACKGROUND: Facet joint osteoarthritis (FJOA) is a common lumbar osteoarthritis characterized by degeneration of small joint cartilage. Bushen Huoxue decotion (BSHXD) has good therapeutic effects on OA. Our work aimed to further probe the pharmacological effects of BSHXD-containing serum (BSHXD-CS) on FJOA and define underlying the mechanisms invovled.
    METHODS: To establish a FJOA cell model, primary rat chondrocytes were treated with LPS. The mRNA and protein expressions were assessed using qRT-PCR and western blot, respectively. The secretion levels of pro-inflammatory cytokines were measured by ELISA. Cell viability was determined by CCK8 assay. The global m6A level was detected by the kit, and NLRP3 mRNA m6A level was determined by Me-RIP assay. The molecular interactions were analyzed by RIP and RNA pull-down assays.
    RESULTS: BSHXD-CS treatment relieved LPS-induced cell injury, inflammation, NLRP3 inflammasome and pyroptosis in chondrocytes (all p < 0.05). LPS-induced NLRP3 upregulation in chondrocytes was related to its high m6A modification level (p < 0.05). It was also observed that BSHXD-CS reduced LPS-induced m6A modification in chondrocytes via repressing STAT3 (all p < 0.05), suggesting BSHXD-CS could repress NLRP3 expression via m6A-dependent manner. Moreover, DAA, a m6A specific inhibitor, was proved to strengthen the protectively roles of BSHXD-CS on LPS-challenged pytoptosis (all p < 0.05).
    CONCLUSIONS: BSHXD-CS inhibited NLRP3 inflammasome activation and pyroptosis in chondrocytes to repress OA progression by reducing RNA m6A modification.
    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:我们先前报道了HMGB1/TLR4轴在脑出血急性期促进炎症。鉴于这一阶段已知涉及神经元焦亡和神经炎症,在此,我们探讨HMGB1/TLR信号是否激活脑出血后的炎性小体和焦亡.
    方法:Sprague-Dawley大鼠注射自体血诱发脑出血。使用改良的神经系统严重程度评分评估神经系统缺陷。这些NLRP1和NLRP3炎性体的表达和定位,以及使用Westernblot或免疫细胞化学评估焦亡和焦亡相关蛋白的水平.在接受针对NLRP1或NLRP3的短干扰RNA、HMGB1抑制剂丙酮酸乙酯或TLR4抑制剂TAK-242治疗的动物中重复这些实验。
    结果:脑出血上调同侧纹状体中的NLRP1和NLRP3,并增加这些细胞的比例。此外,caspase蛋白家族的水平(例如,pro-caspase-1和caspase-1),凋亡相关斑点样蛋白(ASC),前白细胞介素-1β(IL-1β),IL-1β也升高。这些对焦亡和相关神经功能缺损的影响,通过敲低NLRP1或NLRP3或通过抑制HMGB1或TLR4而部分逆转。HMGB1或TLR4的抑制导致NLRP3而非NLRP1的下调。
    结论:HMGB1/TLR4信号可能在脑出血急性期激活NLRP3炎性体,导致称为焦亡的炎症过程。这些见解提出了减轻出血性中风后组织损伤和相关神经功能缺损的潜在治疗目标。
    BACKGROUND: We previously reported that the HMGB1/TLR4 axis promoted inflammation during the acute phase of intracerebral hemorrhage. Given that this phase is known to involve neuronal pyroptosis and neuroinflammation, here we explore whether HMGB1/TLR signaling activate inflammasome and pyroptosis after intracerebral hemorrhage.
    METHODS: Autologous blood was injected into Sprague-Dawley rats to induce intracerebral hemorrhage. Neurological deficits were assessed using a modified neurological severity score. These expression and localization of NLRP1 and NLRP3 inflammasomes, as well as the levels of pyroptosis and pyroptosis-associated proteins were assessed using Western blot or immunocytochemistry. These experiments were repeated in animals that received treatment with short interfering RNAs against NLRP1 or NLRP3, with HMGB1 inhibitor ethyl pyruvate or TLR4 inhibitor TAK-242.
    RESULTS: Intracerebral hemorrhage upregulated NLRP1 and NLRP3 in the ipsilateral striatum and increased the proportions of these cells that were pyroptosis-positive. Additionally, the levels of caspase protein family (e.g., pro-caspase-1 and caspase-1), apoptosis-associated speck-like protein (ASC), pro-interleukin-1β (IL-1β), and IL-1β were also elevated. These effects on pyroptosis and associated neurological deficit, were partially reversed by knockdown of NLRP1 or NLRP3, or by inhibition of HMGB1 or TLR4. Inhibition of HMGB1 or TLR4 resulted in the downregulation NLRP3 but not NLRP1.
    CONCLUSIONS: The HMGB1/TLR4 signaling may activate the NLRP3 inflammasome during the acute phase of intracerebral hemorrhage, resulting in the inflammatory process known as pyroptosis. These insights suggest potential therapeutic targets for the mitigation tissue injury and associated neurological deficits following hemorrhagic stroke.
    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
    恢复和维持内皮细胞的功能对于急性呼吸窘迫综合征(ARDS)至关重要。鸟苷酸结合蛋白1(GBP1)被证明在ARDS患者中升高,但其作用和机制尚不清楚。本研究旨在探讨GBP1在肺损伤中的作用机制。我们的研究表明,当建立LPS和IFN-γ诱导的人肺微血管内皮细胞(HPMECs)损伤模型时,细胞活力显著降低,GBP1水平和炎症因子水平明显升高。si-GBP1转染时,GBP1的低表达促进细胞增殖和迁移,并降低下游炎症因子的表达。此外,GBP1的抑制显著降低了细胞焦亡的发生和NLRP3和STAT1的表达。我们的研究表明,GBP1通过STAT1/NLRP3/GSDMD信号通路减轻内皮细胞的焦化和炎症,GBP1可能是未来肺损伤治疗的新靶点。
    Restoring and maintaining the function of endothelial cells is critical for acute respiratory distress syndrome (ARDS). Guanylate binding protein 1(GBP1) is proved to elevated in ARDS patients, but its role and mechanism remains unclear. The objective of this study is to investigate the internal mechanism of GBP1 in lung injury. Our study showed that when the LPS and IFN-γ induced human Pulmonary Microvascular Endothelial Cells (HPMECs) injury model was established, cell viability was significantly reduced, and the levels of GBP1 levels and inflammatory factors were significantly increased. When transfection with si-GBP1, low expression of GBP1 promoted cell proliferation and migration, and decreased the expression of downstream inflammatory factors. Furthermore, the inhibition of GBP1 significantly reduced the occurrence of cell pyroptosis and the expression of NLRP3 and STAT1. Our study indicated that GBP1 alleviates endothelial pyroptosis and inflammation through STAT1 / NLRP3/GSDMD signaling pathway, and GBP1 may be a new target in the treatment of lung injury in the future.
    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:肾病综合征(NS)已成为全球公共卫生问题。肾纤维化是从NS到终末期肾衰竭最常见的病理变化,严重影响肾脏疾病的预后。尽管已经做出了巨大的努力来治疗NS,延迟NS进展为终末期肾衰竭的特定药物治疗是有限的.淫羊藿一般用于治疗肾脏病的中药。淫羊藿苷是淫羊藿的主要活性成分。
    方法:采用SD大鼠尾静脉注射阿霉素建立NS模型。然后灌胃淫羊藿苷和泼尼松。通过自动生化分析仪检查肾功能。分别通过苏木精-伊红和Masson染色检测肾脏的病理学。此外,RT-PCR,酶联免疫吸附测定,免疫组织化学,Western印迹和末端脱氧核苷酸转移酶介导的尼克末端标记染色用于检测与焦亡和EMT相关的蛋白。用淫羊藿苷处理暴露于阿霉素的HK-2细胞,并且使用MTT评估细胞活力。使用酶联免疫吸附测定和Western印迹评估EMT。
    结果:研究表明淫羊藿苷可明显改善大鼠肾功能和肾纤维化。此外,淫羊藿苷有效降低NOD样受体热蛋白结构域相关蛋白3、Caspase-1、GasderminD、Ly6C,和白细胞介素(IL)-1β。值得注意的是,淫羊藿苷治疗也抑制了TGF-β的水平,α-SMA和E-钙粘蛋白。
    结论:淫羊藿苷可通过抑制细胞凋亡改善肾功能,减轻肾纤维化,其机制可能与上皮间质转化有关。淫羊藿苷治疗可能被推荐为NS的新方法。
    BACKGROUND: Nephrotic syndrome(NS) has emerged as a worldwide public health problem. Renal fibrosis is the most common pathological change from NS to end-stage renal failure, seriously affecting the prognosis of renal disease. Although tremendous efforts have been made to treat NS, specific drug therapies to delay the progression of NS toward end-stage renal failure are limited. Epimedium is generally used to treat kidney disease in traditional Chinese medicine. Icariin is a principal active component of Epimedium.
    METHODS: We used Sprague Dawley rats to establish NS models by injecting doxorubicin through the tail vein. Then icariin and prednisone were intragastric administration. Renal function was examined by an automatic biochemical analyzer. Pathology of the kidney was detected by Hematoxylin-Eosin and Masson staining respectively. Furthermore, RT-PCR, Enzyme-Linked Immunosorbent Assay, Immunohistochemistry, Western Blot and Terminal-deoxynucleotidyl Transferase Mediated Nick End Labeling staining were employed to detect the proteins related to pyroptosis and EMT. HK-2 cells exposed to doxorubicin were treated with icariin, and cell viability was assessed using the MTT. EMT was assessed using Enzyme-Linked Immunosorbent Assay and Western Blot.
    RESULTS: The study showed that icariin significantly improved renal function and renal fibrosis in rats. In addition, icariin effectively decreased NOD-like receptor thermal protein domain associated protein 3,Caspase-1, Gasdermin D, Ly6C, and interleukin (IL)-1β. Notably, treatment with icariin also inhibited the levels of TGF-β, α-SMA and E-cadherin.
    CONCLUSIONS: It is confirmed that icariin can improve renal function and alleviate renal fibrosis by inhibiting pyroptosis and the mechanism may be related to epithelial-to-mesenchymal transition. Icariin treatment might be recommended as a new approach for NS.
    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
    产气荚膜梭菌(C.产气荚膜),革兰氏阳性细菌,产生各种毒素和胞外酶,可导致人类和动物的疾病。常见症状包括腹部肿胀,腹泻,和肠道炎症。严重的病例会导致肠出血等并发症,水肿,甚至死亡。导致产气荚膜梭菌感染肠道发病的主要毒素是CPA,CPB,CPB2、CPE、和PFO。其中,CPB,CPB2和CPE参与细胞凋亡的发展,虽然注册会计师与细胞死亡有关,细胞内ROS水平增加,炎症因子IL-18的释放。然而,PFO毒素在受感染的肠道中发挥作用的确切机制尚不清楚。该研究表明,产气荚膜梭菌PFO毒素感染通过体外和体内模型破坏肠上皮屏障功能。这项研究强调了在产气荚膜梭菌感染的背景下,PFO毒素对肠屏障完整性的显着影响。它揭示了PFO毒素通过引起线粒体损伤来增加ROS的产生,触发IPEC-J2细胞的焦亡,从而导致肠屏障功能受损。这些结果为开发针对产气荚膜梭菌感染的预防和治疗方法提供了科学依据。
    Clostridium perfringens (C. perfringens), a Gram-positive bacterium, produces a variety of toxins and extracellular enzymes that can lead to disease in both humans and animals. Common symptoms include abdominal swelling, diarrhea, and intestinal inflammation. Severe cases can result in complications like intestinal hemorrhage, edema, and even death. The primary toxins contributing to morbidity in C. perfringens-infected intestines are CPA, CPB, CPB2, CPE, and PFO. Amongst these, CPB, CPB2, and CPE are implicated in apoptosis development, while CPA is associated with cell death, increased intracellular ROS levels, and the release of the inflammatory factor IL-18. However, the exact mechanism by which PFO toxins exert their effects in the infected gut is still unidentified. This study demonstrates that a C. perfringens PFO toxin infection disrupts the intestinal epithelial barrier function through in vitro and in vivo models. This study emphasizes the notable influence of PFO toxins on intestinal barrier integrity in the context of C. perfringens infections. It reveals that PFO toxins increase ROS production by causing mitochondrial damage, triggering pyroptosis in IPEC-J2 cells, and consequently resulting in compromised intestinal barrier function. These results offer a scientific foundation for developing preventive and therapeutic approaches against C. perfringens infections.
    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:甲状腺癌是世界范围内罕见但日益普遍的癌症。甲状腺癌的发生发展与线粒体不稳定有关,指的是结构的改变,函数,和线粒体的能量状态。这些改变导致线粒体代谢失衡,引起细胞损伤和凋亡。然而,线粒体不稳定和甲状腺癌的分子机制仍然知之甚少.
    目的:本研究旨在探讨延胡索酸1介导PGC-1α体外调控线粒体稳态延缓甲状腺癌进展的分子机制。
    方法:本研究培养人甲状腺乳头状癌细胞系(TPC-1和K-1)和正常甲状腺细胞系(Nthy-ori3-1)。分别用oveRNA-FH1和oveRNA-NC转染TPC-1细胞和K-1细胞,指定为OveRNA-FH1组,OveRNA-NC组,TPC-1组,还有Nthy-ori3-1组.进行各种测定以评估细胞活力,增殖能力,入侵和迁移能力,以及线粒体形态变化和相关因子的表达。采用qRT-PCR和Westernblot分析PGC-1α的表达变化,线粒体动力学相关因子,和焦亡基因。这些实验的目的是评估FH1对线粒体不稳定性的影响,并阐明甲状腺癌和线粒体不稳定性的具体机制。
    结果:这项研究的结果表明,与正常甲状腺细胞系相比,FH1在甲状腺乳头状癌细胞系中的表达显着下调。在TPC-1细胞中,FH1的过表达降低细胞活力并抑制细胞增殖速率。此外,FH1过表达抑制细胞侵袭和迁移能力。TPC-1和K-1细胞线粒体形态异常改变,而FH1过表达导致相对正常的线粒体。FH1过表达也影响融合和裂变基因的表达,促进甲状腺癌细胞的裂变和抑制融合。此外,FH1过表达导致炎症和焦亡增加。这些结论通过体外成瘤实验得到进一步验证。
    结论:FH1通过PGC-1α依赖性途径调节线粒体稳态促进甲状腺癌进展,影响焦亡和细胞凋亡。
    BACKGROUND: Thyroid cancer is a rare but increasingly prevalent form of cancer worldwide. The development and progression of thyroid cancer are associated with mitochondrial instability, which refers to alterations in the structure, function, and energy status of mitochondria. These alterations lead to an imbalance in mitochondrial metabolism, causing cellular damage and apoptosis. However, the molecular mechanisms underlying mitochondrial instability and thyroid cancer remain poorly understood.
    OBJECTIVE: This study aimed to explore the molecular mechanism of delaying the progression of thyroid cancer by regulating mitochondrial homeostasis through fumarate 1-mediated PGC-1α in vitro.
    METHODS: Human papillary thyroid carcinoma cell lines (TPC-1 and K-1) and a normal thyroid cell line (Nthy-ori 3-1) were cultured in this study. TPC-1 cells and K-1 cells were separately transfected with oveRNA-FH1 and oveRNA-NC, designated as the oveRNA-FH1 group, oveRNA- NC group, TPC-1 group, and Nthy-ori 3-1 group. Various assays were performed to assess cell viability, proliferation capacity, invasion and migration abilities, as well as mitochondrial morphology changes and the expression of relevant factors. qRT-PCR and Western blot analysis were carried out to analyze the expression changes of PGC-1α, mitochondrial dynamics-related factors, and pyroptosis genes. The goal of these experiments was to evaluate the impact of FH1 on mitochondrial instability and elucidate the specific mechanisms underlying thyroid cancer and mitochondrial instability.
    RESULTS: The results of this study demonstrated that FH1 expression was significantly downregulated in thyroid papillary carcinoma cell lines compared to the normal thyroid cell line. Overexpression of FH1 reduced cell viability and inhibited cell proliferation rate in TPC-1 cells. Furthermore, FH1 overexpression suppressed cell invasion and migration abilities. Abnormal mitochondrial morphological changes were observed in TPC-1 and K-1 cells, whereas FH1 overexpression resulted in relatively normal mitochondria. FH1 overexpression also affected the expression of fusion and fission genes, promoting fission and inhibiting fusion in thyroid cancer cells. Moreover, FH1 overexpression led to increased inflammation and pyroptosis. These conclusions were further verified by in vitro tumor formation experiments.
    CONCLUSIONS: FH1 promoted thyroid cancer progression by regulating mitochondrial homeostasis via the PGC-1α-dependent pathway, which affected pyroptosis and apoptosis.
    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