Phosphoglycerate Kinase

磷酸甘油酸激酶
  • 文章类型: Journal Article
    蛋白质翻译后修饰(PTM)对于癌细胞适应缺氧至关重要;然而,赖氨酸巴豆化(Kcr)在缺氧中的功能意义尚不清楚。在这里,我们报告了在常氧和缺氧下全球巴豆的定量蛋白质组学分析,并证明MDA-MB231细胞中101种蛋白质的128个Kcr位点改变。具体来说,我们观察到K131cr显著下降,缺氧时磷酸甘油酸激酶1(PGK1)的K156cr和K220cr。烯酰辅酶A水合酶1(ECHS1)上调并与PGK1相互作用,导致低氧下PGK1Kcr下调。PGK1Kcr的缺失通过激活丙酮酸脱氢酶激酶1(PDHK1)促进糖酵解并抑制线粒体丙酮酸代谢。低PGK1K131cr水平与乳腺癌的恶性程度和不良预后相关。我们的发现表明,PGK1Kcr是协调糖酵解和三羧酸(TCA)循环的信号,可以作为乳腺癌的诊断指标。
    Protein post-translational modifications (PTMs) are crucial for cancer cells to adapt to hypoxia; however, the functional significance of lysine crotonylation (Kcr) in hypoxia remains unclear. Herein we report a quantitative proteomics analysis of global crotonylome under normoxia and hypoxia, and demonstrate 128 Kcr site alterations across 101 proteins in MDA-MB231 cells. Specifically, we observe a significant decrease in K131cr, K156cr and K220cr of phosphoglycerate kinase 1 (PGK1) upon hypoxia. Enoyl-CoA hydratase 1 (ECHS1) is upregulated and interacts with PGK1, leading to the downregulation of PGK1 Kcr under hypoxia. Abolishment of PGK1 Kcr promotes glycolysis and suppresses mitochondrial pyruvate metabolism by activating pyruvate dehydrogenase kinase 1 (PDHK1). A low PGK1 K131cr level is correlated with malignancy and poor prognosis of breast cancer. Our findings show that PGK1 Kcr is a signal in coordinating glycolysis and the tricarboxylic acid (TCA) cycle and may serve as a diagnostic indicator for breast cancer.
    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
    急性肺损伤(ALI)的特征是炎症反应不受调节,通常导致严重的发病率和最终死亡。由M1巨噬细胞极化和焦亡引起的过度炎症已被揭示在ALI中具有关键作用。最近的研究表明,糖酵解重编程在调节巨噬细胞极化和焦亡中很重要。然而,ALI背后的特定过程尚未确定。在这项研究中,我们建立了脂多糖(LPS)诱导的ALI模型,并证明通过使用2-脱氧-D-葡萄糖(2-DG)阻断糖酵解显着下调M1巨噬细胞标记和焦凋亡相关基因的表达,这与体外结果一致。此外,我们的研究表明,磷酸甘油酸激酶1(PGK1),糖酵解途径中必不可少的酶,与NOD交互-,含有LRR和pyrin结构域的蛋白3(NLRP3)。我们发现LPS刺激可在体内和体外改善PGK1和NLRP3的组合。有趣的是,PGK1的缺失降低了NLRP3的磷酸化水平。基于小鼠骨髓源性巨噬细胞(BMDMs)的体外研究,我们进一步证实siPGK1通过抑制巨噬细胞的焦亡和M1巨噬细胞的极化发挥保护作用。PGK1抑制剂NG52抑制ALI中过度炎症的发生。总的来说,我们有理由考虑采用以调节PGK1和NLRP3之间的关系为重点的治疗策略,以此作为减轻ALI中炎性巨噬细胞活化的手段.
    Acute lung injury (ALI) is characterized by an unregulated inflammatory reaction, often leading to severe morbidity and ultimately death. Excessive inflammation caused by M1 macrophage polarization and pyroptosis has been revealed to have a critical role in ALI. Recent study suggests that glycolytic reprogramming is important in the regulation of macrophage polarization and pyroptosis. However, the particular processes underlying ALI have yet to be identified. In this study, we established a Lipopolysaccharide(LPS)-induced ALI model and demonstrated that blocking glycolysis by using 2-Deoxy-D-glucose(2-DG) significantly downregulated the expression of M1 macrophage markers and pyroptosis-related genes, which was consistent with the in vitro results. Furthermore, our research has revealed that Phosphoglycerate Kinase 1(PGK1), an essential enzyme in the glycolysis pathway, interacts with NOD-, LRR- and pyrin domain-containing protein 3(NLRP3). We discovered that LPS stimulation improves the combination of PGK1 and NLRP3 both in vivo and in vitro. Interestingly, the absence of PGK1 reduces the phosphorylation level of NLRP3. Based on in vitro studies with mice bone marrow-derived macrophages (BMDMs), we further confirmed that siPGK1 plays a protective role by inhibiting macrophage pyroptosis and M1 macrophage polarization. The PGK1 inhibitor NG52 suppresses the occurrence of excessive inflammation in ALI. In general, it is plausible to consider a therapeutic strategy that focuses on modulating the relationship between PGK1 and NLRP3 as a means to mitigate the activation of inflammatory macrophages in ALI.
    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:急性低压缺氧引起的脑损伤一直是登山者健康管理的挑战;因此,迫切需要新的神经保护剂。Meldonium,一种众所周知的心脏保护药物,据报道具有神经保护作用。然而,相关机制尚未阐明。我们假设meldonium可能在低压缺氧脑损伤中起潜在的新作用。
    方法:我们初步评估了meldonium对小鼠和原代海马神经元急性缺氧的神经保护功效。使用药物靶标结合Huprot™微阵列芯片和质谱分析筛选了meldonium的潜在分子靶标,然后用表面等离子体共振(SPR)对其进行了验证。分子对接,和下拉分析。通过基因敲低和过表达探索这种结合的功能效应。
    结果:该研究清楚地表明,用美曲多胺预处理可迅速减轻神经元病理损伤,脑血流变化,和线粒体损伤及其对氧化应激损伤的级联反应,从而提高小鼠大脑和原代海马神经元的存活率,揭示了meldonium在急性高原脑损伤中的显着药理功效。一方面,我们证实meldonium直接与磷酸甘油酸激酶1(PGK1)相互作用以促进其活性,改善糖酵解和丙酮酸代谢以促进ATP的产生。另一方面,meldonium还通过在急性缺氧下PGK1易位到线粒体以调节TNF受体相关蛋白1(TRAP1)分子伴侣的活性来改善线粒体损伤。
    结论:这些结果进一步解释了meldonium作为能量优化剂的机制,并为预防高海拔地区急性低压缺氧脑损伤提供了策略。
    BACKGROUND: Acute hypobaric hypoxia-induced brain injury has been a challenge in the health management of mountaineers; therefore, new neuroprotective agents are urgently required. Meldonium, a well-known cardioprotective drug, has been reported to have neuroprotective effects. However, the relevant mechanisms have not been elucidated. We hypothesized that meldonium may play a potentially novel role in hypobaric hypoxia cerebral injury.
    METHODS: We initially evaluated the neuroprotection efficacy of meldonium against acute hypoxia in mice and primary hippocampal neurons. The potential molecular targets of meldonium were screened using drug-target binding Huprot™ microarray chip and mass spectrometry analyses after which they were validated with surface plasmon resonance (SPR), molecular docking, and pull-down assay. The functional effects of such binding were explored through gene knockdown and overexpression.
    RESULTS: The study clearly shows that pretreatment with meldonium rapidly attenuates neuronal pathological damage, cerebral blood flow changes, and mitochondrial damage and its cascade response to oxidative stress injury, thereby improving survival rates in mice brain and primary hippocampal neurons, revealing the remarkable pharmacological efficacy of meldonium in acute high-altitude brain injury. On the one hand, we confirmed that meldonium directly interacts with phosphoglycerate kinase 1 (PGK1) to promote its activity, which improved glycolysis and pyruvate metabolism to promote ATP production. On the other hand, meldonium also ameliorates mitochondrial damage by PGK1 translocating to mitochondria under acute hypoxia to regulate the activity of TNF receptor-associated protein 1 (TRAP1) molecular chaperones.
    CONCLUSIONS: These results further explain the mechanism of meldonium as an energy optimizer and provide a strategy for preventing acute hypobaric hypoxia brain injury at high altitudes.
    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
    胃溃疡是一种在世界范围内高度流行的消化道疾病,这是经常性的,很难治愈,如果不及时治疗,有时会转化为胃癌,对人类健康造成巨大威胁。开发治疗胃溃疡的新药,我们在GES-1细胞中使用乙醇应激模型进行了一系列筛选,我们发现拉米夫定从乙醇毒性中拯救细胞。然后,我们证实了这一发现使用良好建立的乙醇诱导的小鼠胃溃疡模型,我们的发现表明,拉米夫定可以直接激活磷酸甘油酸激酶1(PGK1,EC2.7.2.3),它结合并刺激超氧化物歧化酶1(SOD1,EC1.15.1.1)以抑制铁性凋亡并最终改善胃溃疡。此外,AAV-PGK1表现出与拉米夫定相当的胃保护作用。这些发现有望为胃溃疡提供新的治疗策略。包括拉米夫定和AAV-PGK1。
    Gastric ulcer is a highly prevalent digestive tract disease across the world, which is recurrent and hard to cure, sometimes transforming into gastric cancer if left untreated, posing great threat to human health. To develop new medicines for gastric ulcer, we ran a series of screens with ethanol stress model in GES-1 cells, and we uncovered that lamivudine rescued cells from ethanol toxicity. Then, we confirmed this discovery using the well-established ethanol-induced gastric ulcer model in mice and our findings suggest that lamivudine can directly activate phosphoglycerate kinase 1 (PGK1, EC 2.7.2.3), which binds and stimulates superoxide dismutase 1 (SOD1, EC 1.15.1.1) to inhibit ferroptosis and ultimately improve gastric ulcer. Moreover, AAV-PGK1 exhibited comparable gastroprotective effects to lamivudine. The findings are expected to offer novel therapeutic strategies for gastric ulcer, encompassing both lamivudine and AAV-PGK1.
    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
    泛素化在肿瘤进展中起着关键的调节作用。在泛素-蛋白酶体系统(UPS)的组成部分中,泛素蛋白连接酶E3已成为关键分子。然而,E3泛素连接酶的生物学功能及其在胃癌(GC)中协调糖酵解的潜在机制仍有待阐明。在这项研究中,我们进行了全面的转录组学分析,以确定GC中的核心E3泛素连接酶,随后在体外和体内广泛验证了含三方基序50(TRIM50)的表达模式和临床意义。值得注意的是,我们发现TRIM50在GC组织中下调,与恶性进展和患者生存率低相关。功能上,TRIM50的过表达通过抑制肿瘤相关巨噬细胞(TAMs)的M2极化,抑制GC细胞增殖,间接减轻GC细胞的侵袭和迁移。机械上,TRIM50通过泛素化磷酸甘油酸激酶1(PGK1)抑制糖酵解途径,从而直接抑制GC细胞增殖。同时,乳酸的减少导致TAMs的M2极化减少,间接抑制GC细胞的侵袭和迁移。值得注意的是,GC中TRIM50的下调由METTL3/YTHDF2轴以m6A依赖性方式介导.在我们的研究中,我们确定TRIM50是一种肿瘤抑制基因(TSG),它通过泛素化PGK1有效抑制糖酵解和GC的恶性进展,从而为GC的诊断和治疗提供了新的见解和有希望的靶点.
    Ubiquitination plays a pivotal regulatory role in tumor progression. Among the components of the ubiquitin-proteasome system (UPS), ubiquitin-protein ligase E3 has emerged as a key molecule. Nevertheless, the biological functions of E3 ubiquitin ligases and their potential mechanisms orchestrating glycolysis in gastric cancer (GC) remain to be elucidated. In this study, we conducted a comprehensive transcriptomic analysis to identify the core E3 ubiquitin ligases in GC, followed by extensive validation of the expression patterns and clinical significance of Tripartite motif-containing 50 (TRIM50) both in vitro and in vivo. Remarkably, we found that TRIM50 was downregulated in GC tissues, associated with malignant progression and poor patient survival. Functionally, overexpression of TRIM50 suppressed GC cell proliferation and indirectly mitigated the invasion and migration of GC cells by inhibiting the M2 polarization of tumor-associated macrophages (TAMs). Mechanistically, TRIM50 inhibited the glycolytic pathway by ubiquitinating Phosphoglycerate Kinase 1 (PGK1), thereby directly suppressing GC cell proliferation. Simultaneously, the reduction in lactate led to diminished M2 polarization of TAMs, indirectly inhibiting the invasion and migration of GC cells. Notably, the downregulation of TRIM50 in GC was mediated by the METTL3/YTHDF2 axis in an m6A-dependent manner. In our study, we definitively identified TRIM50 as a tumor suppressor gene (TSG) that effectively inhibits glycolysis and the malignant progression of GC by ubiquitinating PGK1, thus offering novel insights and promising targets for the diagnosis and treatment of GC.
    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
    缺血性卒中被认为是导致死亡和残疾的最常见原因之一。其中神经炎症起着关键作用。新的证据支持PGK1/Nrf2/HO-1信号传导可以调节炎症和氧化损伤。Albiflorin(ALB),白芍的主要成分,具有抗炎和抗氧化特性。然而,它如何发挥保护作用仍需进一步探索。在我们的研究中,建立大脑中动脉闭塞(MCAO)模型,并应用Longa评分调查神经功能缺损程度。采用二氢乙啶(DHE)染色和丙二醛(MDA)检测脂质过氧化水平。使用2,3,5-氯化三苯基四唑(TTC)染色来测量梗死面积。采用伊文思蓝染色观察血脑屏障(BBB)的完整性。HE染色观察各组脑组织损伤情况。免疫荧光染色,采用酶联免疫吸附试验(ELISA)和免疫印迹法检测炎症因子和蛋白质水平。我们最终观察到ALB缓解了脑梗死症状,减轻脑组织中的氧化损伤,减轻MCAO大鼠的神经炎症和细胞损伤。PGK1的过表达消除了实验性脑梗死后ALB的保护作用。ALB促进PGK1降解并诱导Nrf2信号级联激活,用于随后的抗炎和抗氧化损伤。一般来说,ALB在治疗脑缺血中发挥保护作用,并且它可能以PGK1/Nrf2/HO-1信令为目标。因此,ALB可能是减轻脑梗死后神经炎症和保护脑细胞的潜在治疗剂。
    Ischemic stroke is acknowledged as one of the most frequent causes of death and disability, in which neuroinflammation plays a critical role. Emerging evidence supports that the PGK1/Nrf2/HO-1 signaling can modulate inflammation and oxidative injury. Albiflorin (ALB), a main component of Radix paeoniae Alba, possesses anti-inflammatory and antioxidative properties. However, how it exerts a protective role still needs further exploration. In our study, the middle cerebral artery occlusion (MCAO) model was established, and the Longa score was applied to investigate the degree of neurological impairment. Dihydroethidium (DHE) staining and Malondialdehyde (MDA) assay were used to detect the level of lipid peroxidation. 2, 3, 5-Triphenyltetrazolium chloride (TTC) staining was used to measure the infarct area. Evans blue staining was employed to observe the integrality of the blood-brain barrier (BBB). The injury of brain tissue in each group was observed via HE staining. Immunofluorescence staining, enzyme-linked immunosorbent assay (ELISA) and western blot assay were used for the measurement of inflammatory factors and protein levels. We finally observed that ALB relieved cerebral infarction symptoms, attenuated oxidative damage in brain tissues, and reduced neuroinflammation and cell injury in MCAO rats. The overexpression of PGK1 abrogated the protective effect of ALB after experimental cerebral infarction. ALB promoted PGK1 degradation and induced Nrf2 signaling cascade activation for subsequent anti-inflammatory and antioxidant damage. Generally speaking, ALB exerted a protective role in treating cerebral ischemia, and it might target at PGK1/Nrf2/HO-1 signaling. Thus, ALB might be a potential therapeutic agent to alleviate neuroinflammation and protect brain cells after cerebral infarction.
    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
    依托咪酯(ETO),一种用于麻醉诱导的催眠药,已被证明会导致长期的认知缺陷。在本研究中,我们研究了ETO是否可以激活HIF1A/PGK1通路来拮抗术后认知功能障碍(POCD)小鼠的氧化损伤.建立ETO介导POCD的小鼠模型,和病理变化,凋亡,HE染色分析小鼠海马组织炎症因子,TUNEL检测,和ELISA。ETO被发现会导致小鼠的认知功能障碍。进行综合数据库挖掘以筛选出与ETO和POCD均相关的转录因子。缺氧诱导因子1-α(HIF1A)在POCD小鼠中过度表达,HIF1A的下调减轻了小鼠的认知功能障碍。HIF1A下调抑制磷酸甘油酸激酶1(PGK1)的转录。PGK1的过表达减轻了HIF1A敲低对POCD小鼠氧化应激的缓解作用。此外,PGK1的HIF1A激活诱导HT-22细胞的氧化应激和凋亡,同时抑制细胞活力。一起来看,我们证明了PGK1的HIF1A激活在ETO介导的POCD中诱导氧化应激。
    Etomidate (ETO), a hypnotic agent used for anesthesia induction, has been shown to induce long-lasting cognitive deficits. In the present study, we investigated whether ETO could activate the HIF1A/PGK1 pathway to antagonize oxidative damage in mice with postoperative cognitive dysfunction (POCD). A mouse model of ETO-mediated POCD was established, and pathological changes, apoptosis, and inflammatory factors in mouse hippocampal tissues were analyzed by HE staining, TUNEL assay, and ELISA. ETO was revealed to cause cognitive dysfunction in mice. Integrated database mining was conducted to screen out transcription factors that are both related to ETO and POCD. Hypoxia-inducible factor 1-alpha (HIF1A) was overexpressed in mice with POCD, and downregulation of HIF1A alleviated cognitive dysfunction in mice. HIF1A downregulation inhibited the transcription of phosphoglycerate kinase 1 (PGK1). Overexpression of PGK1 abated the alleviating effects of HIF1A knockdown on oxidative stress in mice with POCD. In addition, HIF1A activation of PGK1 induced oxidative stress and apoptosis in HT-22 cells while inhibiting cell viability. Taken together, we demonstrated that HIF1A activation of PGK1 induced oxidative stress in ETO-mediated POCD.
    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
    HOXC6(同源异型框C6)和甲基转移酶样3(METTL3)已显示与前列腺癌(PCa)的进展有关。然而,尚未报道HOXC6是否通过METTL3介导的N6-甲基腺苷(m6A)修饰在PCa中产生致癌作用.细胞计数套件-8(CCK-8),5-乙炔基-2'-脱氧尿苷(EdU),流式细胞术,transwell,划痕,球体形成试验用于细胞生长,入侵,迁移和干性分析。通过测量葡萄糖消耗来评估糖酵解,乳酸生成和ATP/ADP比。通过RNA免疫沉淀(Me-RIP)测定确定N6-甲基腺嘌呤(m6A)修饰谱。与PGK1(磷酸甘油酸激酶1)相互作用的蛋白质通过免疫共沉淀测定进行确认。在小鼠中进行肿瘤形成实验用于体内测定。PCa组织和细胞显示高表达HOXC6和METTL3。功能上,HOXC6或METTL3的沉默抑制PCa细胞增殖,入侵,迁移,stemness,和糖酵解。此外,METTL3诱导HOXC6m6A修饰以稳定其表达。此外,m6A阅读器IGF2BP2直接识别并结合HOXC6mRNA,并保持其稳定性,并参与METTL3对HOXC6表达的调控。此外,IGF2BP2敲低受损PCa细胞增殖,入侵,迁移,stemness,和通过调节HOXC6的糖酵解。此外,HOXC6还与PCa细胞中的糖化酶PGK1相互作用。体内测定进一步显示METTL3沉默降低HOXC6和PGK1的表达,并阻碍PCa生长。METTL3通过在m6A-IGF2BP2依赖性机制中维持HOXC6表达促进PCa进展。
    HOXC6 (Homeobox C6) and methyltransferase-like 3 (METTL3) have been shown to be involved in the progression of prostate cancer (PCa). However, whether HOXC6 performs oncogenic effects in PCa via METTL3-mediated N6-methyladenosine (m6A) modification is not yet reported. The Cell Counting Kit-8 (CCK-8), 5-ethynyl-2\'-deoxyuridine (EdU), flow cytometry, transwell, scratch, sphere formation assays were applied for cell growth, invasion, migration and stemness analyses. Glycolysis was evaluated by measuring glucose consumption, lactate generation and ATP/ADP ratio. The N6-methyladenine (m6A) modification profile was determined by RNA immunoprecipitation (Me-RIP) assay. The proteins that interact with PGK1 (phosphoglycerate kinase 1) were confirmed by Co-immunoprecipitation assay. Tumor formation experiments in mice were conducted for in vivo assay. PCa tissues and cells showed highly expressed HOXC6 and METTL3. Functionally, the silencing of HOXC6 or METTL3 suppresses PCa cell proliferation, invasion, migration, stemness, and glycolysis. Moreover, METTL3-induced HOXC6 m6A modification to stabilize its expression. In addition, the m6A reader IGF2BP2 directly recognized and bound to HOXC6 mRNA, and maintained its stability, and was involved in the regulation of HOXC6 expression by METTL3. Furthermore, IGF2BP2 knockdown impaired PCa cell proliferation, invasion, migration, stemness, and glycolysis by regulating HOXC6. Besides that HOXC6 interacted with the glycoytic enzyme PGK1 in PCa cells. In vivo assays further showed that METTL3 silencing reduced the expression of HOXC6 and PGK1, and impeded PCa growth. METTL3 promoted PCa progression by maintaining HOXC6 expression in an m6A-IGF2BP2-dependent mechanism.
    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
    对念珠菌进行正确诊断的延迟引起了医疗保健系统设置的关注,和免疫蛋白质组学研究对于鉴定免疫反应蛋白以获得新的诊断策略很重要。在这项研究中,进行了由金黄色葡萄球菌的非聚集和聚集表型以及白色念珠菌和假丝酵母引起的免疫活性小鼠全身感染,并将获得的血清用于研究其对C.auris蛋白的免疫反应性。结果显示毒力较高,就感染迹象而言,减肥,和组织病理学损伤,非聚合隔离的。此外,C.auris的毒力比白色念珠菌小,但比C.haemuronii强。关于免疫蛋白质组学研究,13个斑点由感染两种金丝酵母表型的小鼠的血清识别,并通过质谱分析对应于烯醇化酶,磷酸甘油酸激酶,甘油醛-3-磷酸脱氢酶,和磷酸甘油酸变位酶.这四种蛋白质也被获自播散性金丝酵母感染的人类患者的血清识别,但不被获自感染白色念珠菌或烟曲霉的小鼠的血清识别。点识别数据可通过ProteomeXchange与标识符PXD049077获得。总之,这项研究表明,鉴定出的蛋白质可能是潜在的候选物,可作为C.auris的新诊断甚至治疗靶点进行研究。
    The delay in making a correct diagnosis of Candida auris causes concern in the healthcare system setting, and immunoproteomics studies are important to identify immunoreactive proteins for new diagnostic strategies. In this study, immunocompetent murine systemic infections caused by non-aggregative and aggregative phenotypes of C. auris and by Candida albicans and Candida haemulonii were carried out, and the obtained sera were used to study their immunoreactivity against C. auris proteins. The results showed higher virulence, in terms of infection signs, weight loss, and histopathological damage, of the non-aggregative isolate. Moreover, C. auris was less virulent than C. albicans but more than C. haemulonii. Regarding the immunoproteomics study, 13 spots recognized by sera from mice infected with both C. auris phenotypes and analyzed by mass spectrometry corresponded to enolase, phosphoglycerate kinase, glyceraldehyde-3-phosphate dehydrogenase, and phosphoglycerate mutase. These four proteins were also recognized by sera obtained from human patients with disseminated C. auris infection but not by sera obtained from mice infected with C. albicans or Aspergillus fumigatus. Spot identification data are available via ProteomeXchange with the identifier PXD049077. In conclusion, this study showed that the identified proteins could be potential candidates to be studied as new diagnostic or even therapeutic targets for C. auris.
    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
    神经元能量代谢失调与缺血再灌注(I/R)的各种病理有关,然而,RGMA在神经元代谢重编程中的作用尚未见报道.在这项研究中,我们发现I/R后RGMA表达显著增加,与对照小鼠相比,MCAO/R小鼠糖酵解代谢产物和糖酵解途径蛋白表达增加。此外,RGMA水平与神经元能量代谢密切相关。我们发现RGMA的敲低可以将神经元能量代谢向氧化磷酸化和戊糖磷酸途径转移,从而保护小鼠免受缺血再灌注损伤。机械上,敲低RGMA可以下调PGK1的表达,减少缺血再灌注后糖酵解通量的增加。此外,我们发现敲低RGMA可以减少USP10与PGK1之间的相互作用,从而影响PGK1的泛素化降解。总之,我们的数据表明,RGMA可能通过抑制USP10介导的PGK1去泛素化来调节神经元能量代谢,从而保护其免受I/R损伤.本研究为阐明I/R后神经元损伤的内在机制提供了新思路。
    Neuronal energy metabolism dysregulation is involved in various pathologies of Ischemia-reperfusion (I/R), yet the role of RGMA in neuronal metabolic reprogramming has not been reported. In this study, we found that RGMA expression significantly increased after I/R, and compared to control mice, mice with MCAO/R showed an increase in glycolytic metabolic products and the expression of glycolytic pathway proteins. Furthermore, RGMA levels are closely related to neuronal energy metabolism. We discovered that knockdown of RGMA can shift neuronal energy metabolism towards oxidative phosphorylation and the pentose phosphate pathway, thereby protecting mice from ischemic reperfusion injury. Mechanistically, knockdown of RGMA can downregulate PGK1 expression, reducing the increase in glycolytic flux following ischemia reperfusion. Moreover, we found that knockdown of RGMA can reduce the interaction between USP10 and PGK1, thus affecting the ubiquitination degradation of PGK1. In summary, our data suggest that RGMA may regulate neuronal energy metabolism by inhibiting the USP10-mediated deubiquitination of PGK1, thus protecting it from I/R injury. This study provides new ideas for clarifying the intrinsic mechanism of neuronal damage after I/R.
    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