首页 > 最新文献

Neurochemical Research最新文献

英文 中文
Gut Microbiota-Mediated Alterations of Hippocampal CB1R Regulating the Diurnal Variation of Cognitive Impairment Induced by Hepatic Ischemia-Reperfusion Injury in Mice. 肠道微生物群介导的海马CB1R变化调节小鼠肝缺血再灌注损伤所致认知障碍的昼夜变化
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-02 DOI: 10.1007/s11064-024-04182-0
Zhigang He, Yanbo Liu, Zhen Li, Tianning Sun, Zhixiao Li, Cheng Liu, Hongbing Xiang

Patients suffering from hepatic ischemia-reperfusion injury (HIRI) frequently exhibit postoperative cognitive deficits. Our previous observations have emphasized the diurnal variation in hepatic ischemia-reperfusion injury-induced cognitive impairment, in which gut microbiota-associated hippocampal lipid metabolism plays an important role. Herein, we further investigated the molecular mechanisms involved in the process. Hepatic ischemia-reperfusion surgery was performed under morning (ZT0, 08:00) and evening (ZT12, 20:00). Fecal microbiota transplantation was used to associate HIRI model with pseudo-germ-free mice. The novel object recognition test and Y-maze test were used to assess cognitive function. 16S rRNA gene sequencing and analysis were used for microbial analysis. Western blotting was used for hippocampal protein analysis. Compared with the ZT0-HIRI group, ZT12-HIRI mice showed learning and short term memory impairment, accompanied by down-regulated expression of hippocampal CB1R, but not CB2R. Both gut microbiota composition and microbiota metabolites were significantly different in ZT12-HIRI mice compared with ZT0-HIRI. Fecal microbiota transplantation from the ZT12-HIRI was demonstrated to induce cognitive impairment behavior and down-regulated hippocampal CB1R and β-arrestin1. Intraperitoneal administration of CB1R inhibitor AM251 (1 mg/kg) down-regulated hippocampal CB1R and caused cognitive impairment in ZT0-HIRI mice. And intraperitoneal administration of CB1R agonist WIN 55,212-2 (1 mg/kg) up-regulated hippocampal CB1R and improved cognitive impairment in ZT12-HIRI mice. In summary, the results suggest that gut microbiota may regulate the diurnal variation of HIRI-induced cognitive function by interfering with hippocampal CB1R.

肝缺血再灌注损伤(HIRI)患者经常表现出术后认知障碍。我们之前的观察强调了肝缺血再灌注损伤诱发认知障碍的昼夜变化,而肠道微生物群相关的海马脂质代谢在其中发挥了重要作用。在此,我们进一步研究了这一过程的分子机制。肝缺血再灌注手术分别在早上(ZT0,08:00)和晚上(ZT12,20:00)进行。利用粪便微生物群移植将肝缺血再灌注模型与假无精子小鼠联系起来。新物体识别测试和Y-迷宫测试用于评估认知功能。16S rRNA基因测序和分析用于微生物分析。海马蛋白分析采用了 Western 印迹法。与ZT0-HIRI组相比,ZT12-HIRI组小鼠表现出学习和短期记忆障碍,并伴有海马CB1R表达下调,但CB2R表达没有下调。与 ZT0-HIRI 小鼠相比,ZT12-HIRI 小鼠的肠道微生物群组成和微生物群代谢物均有显著差异。实验证明,ZT12-HIRI小鼠的粪便微生物群移植会诱发认知障碍行为,并下调海马CB1R和β-arrestin1。腹腔注射 CB1R 抑制剂 AM251(1 mg/kg)可下调 ZT0-HIRI 小鼠海马 CB1R 并导致认知障碍。而腹腔注射 CB1R 激动剂 WIN 55,212-2 (1 毫克/千克)可上调海马 CB1R 并改善 ZT12-HIRI 小鼠的认知障碍。总之,研究结果表明,肠道微生物群可能通过干扰海马CB1R来调节HIRI诱导的认知功能的昼夜变化。
{"title":"Gut Microbiota-Mediated Alterations of Hippocampal CB1R Regulating the Diurnal Variation of Cognitive Impairment Induced by Hepatic Ischemia-Reperfusion Injury in Mice.","authors":"Zhigang He, Yanbo Liu, Zhen Li, Tianning Sun, Zhixiao Li, Cheng Liu, Hongbing Xiang","doi":"10.1007/s11064-024-04182-0","DOIUrl":"10.1007/s11064-024-04182-0","url":null,"abstract":"<p><p>Patients suffering from hepatic ischemia-reperfusion injury (HIRI) frequently exhibit postoperative cognitive deficits. Our previous observations have emphasized the diurnal variation in hepatic ischemia-reperfusion injury-induced cognitive impairment, in which gut microbiota-associated hippocampal lipid metabolism plays an important role. Herein, we further investigated the molecular mechanisms involved in the process. Hepatic ischemia-reperfusion surgery was performed under morning (ZT0, 08:00) and evening (ZT12, 20:00). Fecal microbiota transplantation was used to associate HIRI model with pseudo-germ-free mice. The novel object recognition test and Y-maze test were used to assess cognitive function. 16S rRNA gene sequencing and analysis were used for microbial analysis. Western blotting was used for hippocampal protein analysis. Compared with the ZT0-HIRI group, ZT12-HIRI mice showed learning and short term memory impairment, accompanied by down-regulated expression of hippocampal CB1R, but not CB2R. Both gut microbiota composition and microbiota metabolites were significantly different in ZT12-HIRI mice compared with ZT0-HIRI. Fecal microbiota transplantation from the ZT12-HIRI was demonstrated to induce cognitive impairment behavior and down-regulated hippocampal CB1R and β-arrestin1. Intraperitoneal administration of CB1R inhibitor AM251 (1 mg/kg) down-regulated hippocampal CB1R and caused cognitive impairment in ZT0-HIRI mice. And intraperitoneal administration of CB1R agonist WIN 55,212-2 (1 mg/kg) up-regulated hippocampal CB1R and improved cognitive impairment in ZT12-HIRI mice. In summary, the results suggest that gut microbiota may regulate the diurnal variation of HIRI-induced cognitive function by interfering with hippocampal CB1R.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141185959","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Combination of Zhuli Decoction and N-butylphthalide Inhibits Cell Apoptosis Induced by CO Poisoning through the PI3K/AKT/GSK-3β Signaling Pathway. 竹沥煎剂和N-丁基苯酞联合应用可通过PI3K/AKT/GSK-3β信号通路抑制一氧化碳中毒诱导的细胞凋亡
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-01 DOI: 10.1007/s11064-024-04179-9
Huiping Song, Aochun Yue, Xudong Zhou, Weiwei Zhao, Wei Han, Qin Li

Carbon monoxide poisoning (COP) represents a significant global health burden, characterized by its morbidity and high mortality rates. The pathogenesis of COP-induced brain injury is complex, and effective treatment modalities are currently lacking. In this study, we employed network pharmacology to identify therapeutic targets and associated signaling pathways of Zhuli Decoction (ZLD) for COP. Subsequently, we conducted both in vitro and in vivo experiments to validate the therapeutic efficacy of ZLD in combination with N-butylphthalide (NBP) for acute COP-induced injury. Our network pharmacology analysis revealed that the primary components of ZLD exerted therapeutic effects through the modulation of multiple targets and pathways. The in vitro and in vivo experiments demonstrated that the combination of NBP and ZLD effectively inhibited apoptosis and up-regulated the activities of P-PI3K (Tyr458), P-AKT (Ser473), P-GSK-3β (Ser9), and Bcl-2, thus leading to the protection of neuronal cells and improvement in cognitive function in rats following COP, which was better than the effects observed with NBP or ZLD alone. The rescue experiment further showed that LY294002, a PI3K inhibitor, significantly attenuated the therapeutic efficacy of NBP + ZLD. The neuroprotection effects of NBP and ZLD against COP-induced brain injury are closely linked to the activation of the PI3K/AKT/GSK-3β signaling pathway.

一氧化碳中毒(COP)是全球重大的健康负担,其特点是发病率和死亡率高。一氧化碳中毒诱发脑损伤的发病机制十分复杂,目前尚缺乏有效的治疗方法。在本研究中,我们采用网络药理学方法确定了竹沥煎剂(Zhuli Decoction,ZLD)对 COP 的治疗靶点和相关信号通路。随后,我们进行了体外和体内实验,验证了ZLD与N-丁基苯酞(NBP)联合治疗急性COP诱导损伤的疗效。我们的网络药理学分析表明,ZLD 的主要成分通过调节多个靶点和途径发挥治疗作用。体外和体内实验表明,NBP和ZLD联合使用可有效抑制细胞凋亡,并上调P-PI3K(Tyr458)、P-AKT(Ser473)、P-GSK-3β(Ser9)和Bcl-2的活性,从而保护COP后大鼠的神经细胞并改善其认知功能,效果优于单独使用NBP或ZLD。抢救实验进一步表明,PI3K 抑制剂 LY294002 明显减弱了 NBP + ZLD 的疗效。NBP和ZLD对COP诱导的脑损伤的神经保护作用与PI3K/AKT/GSK-3β信号通路的激活密切相关。
{"title":"The Combination of Zhuli Decoction and N-butylphthalide Inhibits Cell Apoptosis Induced by CO Poisoning through the PI3K/AKT/GSK-3β Signaling Pathway.","authors":"Huiping Song, Aochun Yue, Xudong Zhou, Weiwei Zhao, Wei Han, Qin Li","doi":"10.1007/s11064-024-04179-9","DOIUrl":"10.1007/s11064-024-04179-9","url":null,"abstract":"<p><p>Carbon monoxide poisoning (COP) represents a significant global health burden, characterized by its morbidity and high mortality rates. The pathogenesis of COP-induced brain injury is complex, and effective treatment modalities are currently lacking. In this study, we employed network pharmacology to identify therapeutic targets and associated signaling pathways of Zhuli Decoction (ZLD) for COP. Subsequently, we conducted both in vitro and in vivo experiments to validate the therapeutic efficacy of ZLD in combination with N-butylphthalide (NBP) for acute COP-induced injury. Our network pharmacology analysis revealed that the primary components of ZLD exerted therapeutic effects through the modulation of multiple targets and pathways. The in vitro and in vivo experiments demonstrated that the combination of NBP and ZLD effectively inhibited apoptosis and up-regulated the activities of P-PI3K (Tyr458), P-AKT (Ser473), P-GSK-3β (Ser9), and Bcl-2, thus leading to the protection of neuronal cells and improvement in cognitive function in rats following COP, which was better than the effects observed with NBP or ZLD alone. The rescue experiment further showed that LY294002, a PI3K inhibitor, significantly attenuated the therapeutic efficacy of NBP + ZLD. The neuroprotection effects of NBP and ZLD against COP-induced brain injury are closely linked to the activation of the PI3K/AKT/GSK-3β signaling pathway.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141185975","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cytosolic Escape of Mitochondrial DNA Triggers cGAS-STING Pathway-Dependent Neuronal PANoptosis in Response to Intermittent Hypoxia. 线粒体 DNA 的胞浆逸出触发 cGAS-STING 通路依赖性神经元泛凋亡以应对间歇性缺氧
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-04 DOI: 10.1007/s11064-024-04151-7
Shuying Wang, Jin Tan, Qiang Zhang

Intermittent hypoxia (IH) is the predominant pathophysiological disturbance in obstructive sleep apnea (OSA), characterized by neuronal cell death and neurocognitive impairment. We focus on the accumulated mitochondrial DNA (mtDNA) in the cytosol, which acts as a damage-associated molecular pattern (DAMP) and activates the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, a known trigger for immune responses and neuronal death in degenerative diseases. However, the specific role and mechanism of the mtDNA-cGAS-STING axis in IH-induced neural damage remain largely unexplored. Here, we investigated the involvement of PANoptosis, a novel type of programmed cell death linked to cytosolic mtDNA accumulation and the cGAS-STING pathway activation, in neuronal cell death induced by IH. Our study found that PANoptosis occurred in primary cultures of hippocampal neurons and HT22 cell lines exposed to IH. In addition, we discovered that during IH, mtDNA released into the cytoplasm via the mitochondrial permeability transition pore (mPTP) activates the cGAS-STING pathway, exacerbating PANoptosis-associated neuronal death. Pharmacologically inhibiting mPTP opening or depleting mtDNA significantly reduced cGAS-STING pathway activation and PANoptosis in HT22 cells under IH. Moreover, our findings indicated that the cGAS-STING pathway primarily promotes PANoptosis by modulating endoplasmic reticulum (ER) stress. Inhibiting or silencing the cGAS-STING pathway substantially reduced ER stress-mediated neuronal death and PANoptosis, while lentivirus-mediated STING overexpression exacerbated these effects. In summary, our study elucidates that cytosolic escape of mtDNA triggers cGAS-STING pathway-dependent neuronal PANoptosis in response to IH, mainly through regulating ER stress. The discovery of the novel mechanism provides theoretical support for the prevention and treatment of neuronal damage and cognitive impairment in patients with OSA.

间歇性缺氧(IH)是阻塞性睡眠呼吸暂停(OSA)的主要病理生理紊乱,其特点是神经细胞死亡和神经认知功能受损。我们的研究重点是细胞质中积累的线粒体DNA(mtDNA),它作为损伤相关分子模式(DAMP),激活环GMP-AMP合成酶(cGAS)-干扰素基因刺激器(STING)通路,这是已知的退行性疾病中免疫反应和神经元死亡的触发器。然而,mtDNA-cGAS-STING 轴在 IH 诱导的神经损伤中的具体作用和机制在很大程度上仍未得到探讨。在这里,我们研究了 PANoptosis(一种新型的程序性细胞死亡,与细胞膜 mtDNA 积累和 cGAS-STING 通路激活有关)在 IH 诱导的神经细胞死亡中的参与情况。我们的研究发现,在暴露于 IH 的海马神经元原代培养物和 HT22 细胞系中发生了泛凋亡。此外,我们还发现,在 IH 过程中,通过线粒体通透性转换孔(mPTP)释放到细胞质中的 mtDNA 激活了 cGAS-STING 通路,加剧了与 PANoptosis 相关的神经元死亡。药物抑制 mPTP 开放或耗尽 mtDNA 可显著减少 IH 条件下 HT22 细胞中 cGAS-STING 通路的激活和 PANoptosis 的发生。此外,我们的研究结果表明,cGAS-STING通路主要通过调节内质网(ER)应激促进PAN凋亡。抑制或沉默 cGAS-STING 通路可大大减少 ER 应激介导的神经元死亡和 PAN 凋亡,而慢病毒介导的 STING 过表达会加剧这些效应。总之,我们的研究阐明了mtDNA的胞浆逸出主要通过调节ER应激,在IH反应中触发cGAS-STING通路依赖的神经元PAN凋亡。这一新机制的发现为预防和治疗 OSA 患者的神经元损伤和认知障碍提供了理论支持。
{"title":"Cytosolic Escape of Mitochondrial DNA Triggers cGAS-STING Pathway-Dependent Neuronal PANoptosis in Response to Intermittent Hypoxia.","authors":"Shuying Wang, Jin Tan, Qiang Zhang","doi":"10.1007/s11064-024-04151-7","DOIUrl":"10.1007/s11064-024-04151-7","url":null,"abstract":"<p><p>Intermittent hypoxia (IH) is the predominant pathophysiological disturbance in obstructive sleep apnea (OSA), characterized by neuronal cell death and neurocognitive impairment. We focus on the accumulated mitochondrial DNA (mtDNA) in the cytosol, which acts as a damage-associated molecular pattern (DAMP) and activates the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, a known trigger for immune responses and neuronal death in degenerative diseases. However, the specific role and mechanism of the mtDNA-cGAS-STING axis in IH-induced neural damage remain largely unexplored. Here, we investigated the involvement of PANoptosis, a novel type of programmed cell death linked to cytosolic mtDNA accumulation and the cGAS-STING pathway activation, in neuronal cell death induced by IH. Our study found that PANoptosis occurred in primary cultures of hippocampal neurons and HT22 cell lines exposed to IH. In addition, we discovered that during IH, mtDNA released into the cytoplasm via the mitochondrial permeability transition pore (mPTP) activates the cGAS-STING pathway, exacerbating PANoptosis-associated neuronal death. Pharmacologically inhibiting mPTP opening or depleting mtDNA significantly reduced cGAS-STING pathway activation and PANoptosis in HT22 cells under IH. Moreover, our findings indicated that the cGAS-STING pathway primarily promotes PANoptosis by modulating endoplasmic reticulum (ER) stress. Inhibiting or silencing the cGAS-STING pathway substantially reduced ER stress-mediated neuronal death and PANoptosis, while lentivirus-mediated STING overexpression exacerbated these effects. In summary, our study elucidates that cytosolic escape of mtDNA triggers cGAS-STING pathway-dependent neuronal PANoptosis in response to IH, mainly through regulating ER stress. The discovery of the novel mechanism provides theoretical support for the prevention and treatment of neuronal damage and cognitive impairment in patients with OSA.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141236259","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
miR-206-3p Targets Brain-Derived Neurotrophic Factor and Affects Postoperative Cognitive Function in Aged Mice. miR-206-3p 靶向脑源性神经营养因子并影响老年小鼠的术后认知功能
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-05-30 DOI: 10.1007/s11064-024-04174-0
Shentong Wang, Jia Zhao, Chengran Wang, Yuhan Yao, Zhiyao Song, Longyun Li, Jinlan Jiang

Postoperative cognitive dysfunction (POCD) occurs after surgery and severely impairs patients' quality of life. Finding POCD-associated variables can aid in its diagnosis and prognostication. POCD is associated with noncoding RNAs, such as microRNAs (miRNAs), involved in metabolic function, immune response alteration, and cognitive ability impairment; however, the underlying mechanisms remain unclear. The aim of this study was to investigate hub miRNAs (i.e., miRNAs that have an important regulatory role in diseases) regulating postoperative cognitive function and the associated mechanisms. Hub miRNAs were identified by bioinformatics, and their expression in mouse hippocampus tissues was determined using real-time quantitative polymerase chain reaction. Hub miRNAs were overexpressed or knocked down in cell and animal models to test their effects on neuroinflammation and postoperative cognitive function. Six differentially expressed hub miRNAs were identified. miR-206-3p was the only broadly conserved miRNA, and it was used in follow-up studies and animal experiments. Its inhibitors reduced the release of proinflammatory cytokines in BV-2 microglia by regulating its target gene, brain-derived neurotrophic factor (BDNF), and the downstream signaling pathways. miR-206-3p inhibition suppressed microglial activation in the hippocampi of mice and improved learning and cognitive decline. Therefore, miR-206-3p significantly affects POCD, implying its potential as a therapeutic target.

手术后会出现认知功能障碍(POCD),严重影响患者的生活质量。找到与 POCD 相关的变量有助于诊断和预后。POCD 与参与代谢功能、免疫反应改变和认知能力损伤的非编码 RNAs(如 microRNAs,miRNAs)有关,但其潜在机制仍不清楚。本研究旨在研究调控术后认知功能的枢纽 miRNA(即在疾病中具有重要调控作用的 miRNA)及其相关机制。研究人员通过生物信息学方法鉴定了枢纽miRNA,并利用实时定量聚合酶链反应测定了它们在小鼠海马组织中的表达。在细胞和动物模型中过表达或敲除枢纽miRNA,以检测它们对神经炎症和术后认知功能的影响。研究发现了六种不同表达的中枢 miRNA,其中 miR-206-3p 是唯一一种广泛保守的 miRNA,它被用于后续研究和动物实验。其抑制剂通过调节其靶基因脑源性神经营养因子(BDNF)和下游信号通路,减少了 BV-2 小胶质细胞中促炎细胞因子的释放。抑制 miR-206-3p 可抑制小鼠海马中的小胶质细胞活化,改善学习和认知能力下降。因此,miR-206-3p 对 POCD 有重大影响,意味着它有可能成为治疗靶点。
{"title":"miR-206-3p Targets Brain-Derived Neurotrophic Factor and Affects Postoperative Cognitive Function in Aged Mice.","authors":"Shentong Wang, Jia Zhao, Chengran Wang, Yuhan Yao, Zhiyao Song, Longyun Li, Jinlan Jiang","doi":"10.1007/s11064-024-04174-0","DOIUrl":"10.1007/s11064-024-04174-0","url":null,"abstract":"<p><p>Postoperative cognitive dysfunction (POCD) occurs after surgery and severely impairs patients' quality of life. Finding POCD-associated variables can aid in its diagnosis and prognostication. POCD is associated with noncoding RNAs, such as microRNAs (miRNAs), involved in metabolic function, immune response alteration, and cognitive ability impairment; however, the underlying mechanisms remain unclear. The aim of this study was to investigate hub miRNAs (i.e., miRNAs that have an important regulatory role in diseases) regulating postoperative cognitive function and the associated mechanisms. Hub miRNAs were identified by bioinformatics, and their expression in mouse hippocampus tissues was determined using real-time quantitative polymerase chain reaction. Hub miRNAs were overexpressed or knocked down in cell and animal models to test their effects on neuroinflammation and postoperative cognitive function. Six differentially expressed hub miRNAs were identified. miR-206-3p was the only broadly conserved miRNA, and it was used in follow-up studies and animal experiments. Its inhibitors reduced the release of proinflammatory cytokines in BV-2 microglia by regulating its target gene, brain-derived neurotrophic factor (BDNF), and the downstream signaling pathways. miR-206-3p inhibition suppressed microglial activation in the hippocampi of mice and improved learning and cognitive decline. Therefore, miR-206-3p significantly affects POCD, implying its potential as a therapeutic target.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141174025","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
AdipoRon Ameliorates Synaptic Dysfunction and Inhibits tau Hyperphosphorylation through the AdipoR/AMPK/mTOR Pathway in T2DM Mice. AdipoRon 可通过 AdipoR/AMPK/mTOR 途径改善 T2DM 小鼠的突触功能障碍并抑制 tau 过度磷酸化。
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-05-31 DOI: 10.1007/s11064-024-04162-4
Wenyan Zhao, Wei Zhang, Yingying Hu, Yuliang Zhou, Jinying Zhao, Yahong Li, Zhipeng Xu

There is growing evidence showing that adiponectin (APN) can improve Alzheimer's disease(AD)-like pathological changes by improving insulin resistance. However, the role of AdipoRon (an Adiponectin receptor agonist) on synaptic plasticity and cognitive dysfunction in the early stages of type 2 diabetes mellitus(T2DM) remains unknown. In this study, we investigated the neuroprotective effect and the molecular mechanism underlying the effect of AdipoRon in T2DM mice. We found that AdipoRon significantly restored the cognitive deficits in T2DM mice, including shorter escape latency, more crossing times, increased distances, and percentage of time in the target quadrant. In addition, AdipoRon treatment up-regulated synaptic proteins (PSD95, SYN, GAP43, and SYP), increased the number of hippocampal synapses and attenuated synaptic damage, including the length, the number and the density of dendritic spines in CA1 and DG regions. Furthermore, AdipoRon attenuated Tau phosphorylation at multiple AD-related sites (p-tau 205, p-tau 396, p-tau 404) by promoting AdipoR expression and activating the AMPK/mTOR pathway. Our data suggests that AdipoRon exerts neuroprotective effects on the T2DM mice, which may be mediated by the activation of the AdipoR/AMPK/mTOR signaling pathway.

越来越多的证据表明,脂肪连接素(APN)可以通过改善胰岛素抵抗来改善阿尔茨海默病(AD)样病理变化。然而,在2型糖尿病(T2DM)的早期阶段,AdipoRon(一种脂肪连接素受体激动剂)对突触可塑性和认知功能障碍的作用仍然未知。本研究探讨了 AdipoRon 对 T2DM 小鼠神经的保护作用及其分子机制。我们发现,AdipoRon能显著恢复T2DM小鼠的认知缺陷,包括更短的逃逸潜伏期、更多的穿越时间、更长的距离以及在目标象限的时间百分比。此外,AdipoRon还能上调突触蛋白(PSD95、SYN、GAP43和SYP),增加海马突触数量,减轻突触损伤,包括CA1和DG区树突棘的长度、数量和密度。此外,AdipoRon 还通过促进 AdipoR 的表达和激活 AMPK/mTOR 通路,减轻了多个 AD 相关位点(p-tau 205、p-tau 396、p-tau 404)的 Tau 磷酸化。我们的数据表明,AdipoRon对T2DM小鼠具有神经保护作用,这可能是通过激活AdipoR/AMPK/mTOR信号通路介导的。
{"title":"AdipoRon Ameliorates Synaptic Dysfunction and Inhibits tau Hyperphosphorylation through the AdipoR/AMPK/mTOR Pathway in T2DM Mice.","authors":"Wenyan Zhao, Wei Zhang, Yingying Hu, Yuliang Zhou, Jinying Zhao, Yahong Li, Zhipeng Xu","doi":"10.1007/s11064-024-04162-4","DOIUrl":"10.1007/s11064-024-04162-4","url":null,"abstract":"<p><p>There is growing evidence showing that adiponectin (APN) can improve Alzheimer's disease(AD)-like pathological changes by improving insulin resistance. However, the role of AdipoRon (an Adiponectin receptor agonist) on synaptic plasticity and cognitive dysfunction in the early stages of type 2 diabetes mellitus(T2DM) remains unknown. In this study, we investigated the neuroprotective effect and the molecular mechanism underlying the effect of AdipoRon in T2DM mice. We found that AdipoRon significantly restored the cognitive deficits in T2DM mice, including shorter escape latency, more crossing times, increased distances, and percentage of time in the target quadrant. In addition, AdipoRon treatment up-regulated synaptic proteins (PSD95, SYN, GAP43, and SYP), increased the number of hippocampal synapses and attenuated synaptic damage, including the length, the number and the density of dendritic spines in CA1 and DG regions. Furthermore, AdipoRon attenuated Tau phosphorylation at multiple AD-related sites (p-tau 205, p-tau 396, p-tau 404) by promoting AdipoR expression and activating the AMPK/mTOR pathway. Our data suggests that AdipoRon exerts neuroprotective effects on the T2DM mice, which may be mediated by the activation of the AdipoR/AMPK/mTOR signaling pathway.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141178621","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effect of Capsaicin on 3-NP-Induced Neurotoxicity: A Pre-Clinical Study. 辣椒素对 3-NP 诱导的神经毒性的影响:临床前研究
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-05-30 DOI: 10.1007/s11064-024-04158-0
Sakshi Tyagi, Ajit Kumar Thakur

The study objectives are to investigate the ability of capsaicin to revert the toxic effects in glutamate and lipopolysaccharide (LPS)-induced neurotoxicity in Neuro2a (N2a) cells as well as thwarting cognitive impairments, mitochondrial deficits, and oxidative insults induced by 3-nitropropanoic acid (3-NP) in a rodent model of Huntington's disease. In-vitro study with N2a cells was performed through MTT and LDH assay and their biochemical examinations were also performed. 3-NP-administered mice (n = 6) were treated with capsaicin (5, 10, and 20 mg/kg) through the per-oral (p.o.) route for 7 consecutive days. Physiological and behavioral studies were performed in drug-treated mice. After behavioral studies, biochemical parameters were performed for cytokines levels, various oxidative stress parameters, and mitochondrial enzyme complex activities with mitochondrial permeability. N2a cells treated with capsaicin demonstrated neuroprotective effects and reduced neurotoxicity. Based on experimental observation, in an in-vitro study, the effective dose of CAP was 50 µM. Moreover, a 100 µM dose of capsaicin had toxic effects on neuronal cells (N2a cells). On the other hand, the effective dose of 3-NP was 20 mg/kg, (p.o.) in animals (in-vivo). All tested doses of capsaicin upturned the cognitive impairment and motor in-coordination effects induced by 3-NP. 3-NP-injected mice demonstrated substantially increased pro-inflammatory cytokine concentrations, defective mitochondrial complex activity, and augmented oxidative insult. However, capsaicin at different doses reduced oxidative damage and cytokines levels and improved mitochondrial complex activity along with mitochondrial permeability. Furthermore, capsaicin (10 and 20 mg/kg) improved the TNF-α concentration. These findings suggested because of the anti-inflammatory and antioxidant effect, capsaicin can be considered a novel treatment for the management of neurodegenerative disorders by reverting the antioxidant enzyme activity, pro-inflammatory cytokines concentration, and mitochondrial functions.

研究的目的是探讨辣椒素能否逆转谷氨酸和脂多糖(LPS)诱导的神经毒性对神经2a(N2a)细胞的影响,以及能否挫败3-硝基丙酸(3-NP)在亨廷顿氏病啮齿动物模型中诱导的认知障碍、线粒体缺陷和氧化损伤。通过 MTT 和 LDH 试验对 N2a 细胞进行了体外研究,并对其进行了生化检查。连续 7 天通过口服(p.o.)途径用辣椒素(5、10 和 20 毫克/千克)治疗服用 3-NP 的小鼠(n = 6)。对用药小鼠进行了生理和行为研究。行为研究后,对细胞因子水平、各种氧化应激参数、线粒体酶复合物活性和线粒体通透性等生化参数进行了检测。经辣椒素处理的 N2a 细胞具有神经保护作用,并降低了神经毒性。根据实验观察,在体外研究中,辣椒素的有效剂量为 50 µM。此外,100 µM剂量的辣椒素会对神经细胞(N2a细胞)产生毒性作用。另一方面,3-NP 对动物(体内)的有效剂量为 20 毫克/千克(口服)。所有测试剂量的辣椒素都能逆转 3-NP 引起的认知障碍和运动不协调效应。注射 3-NP 的小鼠表现出促炎细胞因子浓度大幅增加、线粒体复合物活性缺陷和氧化损伤加剧。然而,不同剂量的辣椒素可降低氧化损伤和细胞因子水平,改善线粒体复合物活性和线粒体通透性。此外,辣椒素(10 毫克/千克和 20 毫克/千克)还能改善 TNF-α 的浓度。这些研究结果表明,由于辣椒素具有抗炎和抗氧化作用,它可以通过恢复抗氧化酶活性、促炎细胞因子浓度和线粒体功能来治疗神经退行性疾病。
{"title":"Effect of Capsaicin on 3-NP-Induced Neurotoxicity: A Pre-Clinical Study.","authors":"Sakshi Tyagi, Ajit Kumar Thakur","doi":"10.1007/s11064-024-04158-0","DOIUrl":"10.1007/s11064-024-04158-0","url":null,"abstract":"<p><p>The study objectives are to investigate the ability of capsaicin to revert the toxic effects in glutamate and lipopolysaccharide (LPS)-induced neurotoxicity in Neuro2a (N2a) cells as well as thwarting cognitive impairments, mitochondrial deficits, and oxidative insults induced by 3-nitropropanoic acid (3-NP) in a rodent model of Huntington's disease. In-vitro study with N2a cells was performed through MTT and LDH assay and their biochemical examinations were also performed. 3-NP-administered mice (n = 6) were treated with capsaicin (5, 10, and 20 mg/kg) through the per-oral (p.o.) route for 7 consecutive days. Physiological and behavioral studies were performed in drug-treated mice. After behavioral studies, biochemical parameters were performed for cytokines levels, various oxidative stress parameters, and mitochondrial enzyme complex activities with mitochondrial permeability. N2a cells treated with capsaicin demonstrated neuroprotective effects and reduced neurotoxicity. Based on experimental observation, in an in-vitro study, the effective dose of CAP was 50 µM. Moreover, a 100 µM dose of capsaicin had toxic effects on neuronal cells (N2a cells). On the other hand, the effective dose of 3-NP was 20 mg/kg, (p.o.) in animals (in-vivo). All tested doses of capsaicin upturned the cognitive impairment and motor in-coordination effects induced by 3-NP. 3-NP-injected mice demonstrated substantially increased pro-inflammatory cytokine concentrations, defective mitochondrial complex activity, and augmented oxidative insult. However, capsaicin at different doses reduced oxidative damage and cytokines levels and improved mitochondrial complex activity along with mitochondrial permeability. Furthermore, capsaicin (10 and 20 mg/kg) improved the TNF-α concentration. These findings suggested because of the anti-inflammatory and antioxidant effect, capsaicin can be considered a novel treatment for the management of neurodegenerative disorders by reverting the antioxidant enzyme activity, pro-inflammatory cytokines concentration, and mitochondrial functions.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141174083","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DL-3-n-Butylphthalide Ameliorates Post-stroke Emotional Disorders by Suppressing Neuroinflammation and PANoptosis. DL-3-正丁基苯酞通过抑制神经炎症和泛酸凋亡改善中风后的情感障碍
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-04 DOI: 10.1007/s11064-024-04171-3
Yanhui Cui, Zhaolan Hu, Laifa Wang, Bi Zhu, Ling Deng, Hui Zhang, Xueqin Wang

Post-stroke emotional disorders such as post-stroke anxiety and post-stroke depression are typical symptoms in patients with stroke. They are closely associated with poor prognosis and low quality of life. The State Food and Drug Administration of China has approved DL-3-n-butylphthalide (NBP) as a treatment for ischemic stroke (IS). Clinical research has shown that NBP alleviates anxiety and depressive symptoms in patients with IS. Therefore, this study explored the role and molecular mechanisms of NBP in cases of post-stroke emotional disorders using network pharmacology and experimental validation. The results showed that NBP treatment significantly increased the percentage of time spent in the center of the middle cerebral artery occlusion (MCAO) rats in the open field test and the percentage of sucrose consumption in the sucrose preference test. Network pharmacology results suggest that NBP may regulate neuroinflammation and cell death. Further experiments revealed that NBP inhibited the toll-like receptor 4/nuclear factor kappa B signaling pathway, decreased the level of pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, and interleukin-6, and M1-type microglia markers (CD68, inducible nitric oxide synthase), and reduced the expression of PANoptosis-related molecules including caspase-1, caspase-3, caspase-8, gasdermin D, and mixed lineage kinase domain-like protein in the hippocampus of the MACO rats. These findings demonstrate that the mechanisms through which NBP ameliorates post-stroke emotional disorders in rats are associated with inhibiting neuroinflammation and PANoptosis, providing a new strategy and experimental basis for treating post-stroke emotional disorders.

中风后焦虑和中风后抑郁等中风后情绪障碍是中风患者的典型症状。它们与预后不良和生活质量低下密切相关。中国国家食品药品监督管理局已批准 DL-3-n-butylphthalide (NBP) 作为缺血性脑卒中(IS)的治疗药物。临床研究表明,NBP 可减轻 IS 患者的焦虑和抑郁症状。因此,本研究利用网络药理学和实验验证,探讨了 NBP 在中风后情绪障碍病例中的作用和分子机制。结果表明,NBP治疗能显著增加大脑中动脉闭塞(MCAO)大鼠在开阔地试验中中心停留时间的百分比和蔗糖偏好试验中蔗糖消耗的百分比。网络药理学结果表明,NBP 可调节神经炎症和细胞死亡。进一步的实验发现,NBP 可抑制收费样受体 4/ 核因子卡巴 B 信号通路,降低促炎细胞因子的水平,包括肿瘤坏死因子-α、白细胞介素-1β 和白细胞介素-6、在 MACO 大鼠的海马中,M1 型小胶质细胞标志物(CD68、诱导型一氧化氮合酶)和 PAN 细胞凋亡相关分子(包括 caspase-1、caspase-3、caspase-8、gasdermin D 和混合系激酶域样蛋白)的表达均有所减少。这些研究结果表明,NBP改善大鼠中风后情感障碍的机制与抑制神经炎症和PAN凋亡有关,为治疗中风后情感障碍提供了新的策略和实验依据。
{"title":"DL-3-n-Butylphthalide Ameliorates Post-stroke Emotional Disorders by Suppressing Neuroinflammation and PANoptosis.","authors":"Yanhui Cui, Zhaolan Hu, Laifa Wang, Bi Zhu, Ling Deng, Hui Zhang, Xueqin Wang","doi":"10.1007/s11064-024-04171-3","DOIUrl":"10.1007/s11064-024-04171-3","url":null,"abstract":"<p><p>Post-stroke emotional disorders such as post-stroke anxiety and post-stroke depression are typical symptoms in patients with stroke. They are closely associated with poor prognosis and low quality of life. The State Food and Drug Administration of China has approved DL-3-n-butylphthalide (NBP) as a treatment for ischemic stroke (IS). Clinical research has shown that NBP alleviates anxiety and depressive symptoms in patients with IS. Therefore, this study explored the role and molecular mechanisms of NBP in cases of post-stroke emotional disorders using network pharmacology and experimental validation. The results showed that NBP treatment significantly increased the percentage of time spent in the center of the middle cerebral artery occlusion (MCAO) rats in the open field test and the percentage of sucrose consumption in the sucrose preference test. Network pharmacology results suggest that NBP may regulate neuroinflammation and cell death. Further experiments revealed that NBP inhibited the toll-like receptor 4/nuclear factor kappa B signaling pathway, decreased the level of pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, and interleukin-6, and M1-type microglia markers (CD68, inducible nitric oxide synthase), and reduced the expression of PANoptosis-related molecules including caspase-1, caspase-3, caspase-8, gasdermin D, and mixed lineage kinase domain-like protein in the hippocampus of the MACO rats. These findings demonstrate that the mechanisms through which NBP ameliorates post-stroke emotional disorders in rats are associated with inhibiting neuroinflammation and PANoptosis, providing a new strategy and experimental basis for treating post-stroke emotional disorders.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141247117","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Propofol Mitigates Sepsis-Induced Brain Injury by Inhibiting Ferroptosis Via Activation of the Nrf2/HO-1axis. 丙泊酚通过激活Nrf2/HO-1轴抑制铁凋亡减轻败血症诱发的脑损伤
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-01 DOI: 10.1007/s11064-024-04163-3
Ye Zhou, Yangliang Yang, Liang Yi, Mengzhi Pan, Weiqing Tang, Hongwei Duan

Background: Sepsis-associated encephalopathy (SAE) develops in 30-70% of hospitalized patients with sepsis. In intensive care units (ICUs), propofol is often administered to ensure an appropriate level of sedation in mechanically ventilated patients. Ferroptosis is a newly identified mode of cellular death characterized by the peroxidation of membrane lipids and excessive iron. This study was conducted to explore the interplay between propofol, sepsis, and ferroptosis.

Methods: An acute systemic inflammatory model was constructed via the intraperitoneal administration of lipopolysaccharide (LPS). Nissl and Fluoro-Jade C (FJC) staining were employed to display neuronal damage and degeneration. Western blotting and immunofluorescence (IF) staining of Bax and Bcl-2 were used to confirm the neural apoptosis. QPCR of cytokines and DHE staining were used to indicate neuroinflammation. To validate ferroptosis, we assessed the content of malondialdehyde (MDA), GSH, and tissue iron, accompanied by transcription level of CHAC1, PTGS2 and GPX4. Additionally, we examined the content of acyl-CoA synthetase long-chain family member 4 (ACSL4), xCT (SLC7A11, solute carrier family 7 member 11), and glutathione peroxidase 4 (GPX4). The IF staining of Iba1-labeled microglia and GFAP-marked astrocytes were used to measure the gliosis. Erastin was pre-pretreated to confirm the anti-ferroptotic capability of propofol. ML385 was preconditioned to explore the role of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) in propofol-repressed ferroptosis.

Results: Propofol dose-dependently inhibited the decrease of Nissl-positive neurons and the increase of FJC-stained neurons in septic hippocampus and cortex. Neural cytokines, oxidative stress, apoptosis and gliosis were reduced by propofol. Propofol repressed the level of MDA, iron, CHAC1, PTGS2, ACLS4 and restored the content of GSH, GPX4, xCT, Nrf2 and HO-1, thus inhibiting sepsis-induced ferroptosis. All protections from propofol could be reversed by eratsin and ML385 pretreatment.

Conclusion: Propofol protected against sepsis-induced brain damage, neuroinflammation, neuronal apoptosis and gliosis through the activation of the Nrf2/HO-1 axis to combat ferroptosis.

背景:脓毒症相关脑病(SAE)在 30-70% 的脓毒症住院患者中发病。在重症监护病房(ICU)中,为确保机械通气患者达到适当的镇静水平,通常会使用异丙酚。铁中毒是一种新发现的细胞死亡模式,其特点是膜脂过氧化和铁过量。本研究旨在探讨异丙酚、败血症和铁中毒之间的相互作用:方法:通过腹腔注射脂多糖(LPS)构建了急性全身炎症模型。方法:通过腹腔注射脂多糖(LPS)构建了一个急性全身炎症模型,采用Nissl和Fluoro-Jade C(FJC)染色显示神经元损伤和变性。用 Western 印迹和免疫荧光 (IF) 染色 Bax 和 Bcl-2 来证实神经细胞凋亡。细胞因子的 QPCR 和 DHE 染色用于显示神经炎症。为了验证铁变态反应,我们评估了丙二醛(MDA)、GSH和组织铁的含量,以及CHAC1、PTGS2和GPX4的转录水平。此外,我们还检测了酰基-CoA 合成酶长链家族成员 4(ACSL4)、xCT(SLC7A11,溶质运载家族 7 成员 11)和谷胱甘肽过氧化物酶 4(GPX4)的含量。用 Iba1 标记的小胶质细胞和 GFAP 标记的星形胶质细胞的 IF 染色来测量胶质细胞增生。对 Erastin 进行预处理以确认异丙酚的抗铁血病能力。对ML385进行预处理,以探讨核因子红细胞2相关因子2(Nrf2)和血红素加氧酶1(HO-1)在异丙酚抑制铁突变中的作用:结果:丙泊酚剂量依赖性地抑制了败血症海马和皮层中Nissl阳性神经元的减少和FJC染色神经元的增加。神经细胞因子、氧化应激、细胞凋亡和胶质细胞增生在异丙酚作用下均有所减少。丙泊酚抑制了MDA、铁、CHAC1、PTGS2和ACLS4的水平,恢复了GSH、GPX4、xCT、Nrf2和HO-1的含量,从而抑制了败血症诱导的铁变态反应。丙泊酚的所有保护作用均可被麦拉嗪和ML385预处理逆转:结论:丙泊酚通过激活Nrf2/HO-1轴来对抗铁卟啉沉积,从而保护脓毒症诱导的脑损伤、神经炎症、神经细胞凋亡和胶质细胞沉积。
{"title":"Propofol Mitigates Sepsis-Induced Brain Injury by Inhibiting Ferroptosis Via Activation of the Nrf2/HO-1axis.","authors":"Ye Zhou, Yangliang Yang, Liang Yi, Mengzhi Pan, Weiqing Tang, Hongwei Duan","doi":"10.1007/s11064-024-04163-3","DOIUrl":"10.1007/s11064-024-04163-3","url":null,"abstract":"<p><strong>Background: </strong>Sepsis-associated encephalopathy (SAE) develops in 30-70% of hospitalized patients with sepsis. In intensive care units (ICUs), propofol is often administered to ensure an appropriate level of sedation in mechanically ventilated patients. Ferroptosis is a newly identified mode of cellular death characterized by the peroxidation of membrane lipids and excessive iron. This study was conducted to explore the interplay between propofol, sepsis, and ferroptosis.</p><p><strong>Methods: </strong>An acute systemic inflammatory model was constructed via the intraperitoneal administration of lipopolysaccharide (LPS). Nissl and Fluoro-Jade C (FJC) staining were employed to display neuronal damage and degeneration. Western blotting and immunofluorescence (IF) staining of Bax and Bcl-2 were used to confirm the neural apoptosis. QPCR of cytokines and DHE staining were used to indicate neuroinflammation. To validate ferroptosis, we assessed the content of malondialdehyde (MDA), GSH, and tissue iron, accompanied by transcription level of CHAC1, PTGS2 and GPX4. Additionally, we examined the content of acyl-CoA synthetase long-chain family member 4 (ACSL4), xCT (SLC7A11, solute carrier family 7 member 11), and glutathione peroxidase 4 (GPX4). The IF staining of Iba1-labeled microglia and GFAP-marked astrocytes were used to measure the gliosis. Erastin was pre-pretreated to confirm the anti-ferroptotic capability of propofol. ML385 was preconditioned to explore the role of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) in propofol-repressed ferroptosis.</p><p><strong>Results: </strong>Propofol dose-dependently inhibited the decrease of Nissl-positive neurons and the increase of FJC-stained neurons in septic hippocampus and cortex. Neural cytokines, oxidative stress, apoptosis and gliosis were reduced by propofol. Propofol repressed the level of MDA, iron, CHAC1, PTGS2, ACLS4 and restored the content of GSH, GPX4, xCT, Nrf2 and HO-1, thus inhibiting sepsis-induced ferroptosis. All protections from propofol could be reversed by eratsin and ML385 pretreatment.</p><p><strong>Conclusion: </strong>Propofol protected against sepsis-induced brain damage, neuroinflammation, neuronal apoptosis and gliosis through the activation of the Nrf2/HO-1 axis to combat ferroptosis.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141185971","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ferritinophagy and Ferroptosis in Cerebral Ischemia Reperfusion Injury. 脑缺血再灌注损伤中的噬铁蛋白和铁变态反应
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-04 DOI: 10.1007/s11064-024-04161-5
Xiaoyue Liu, Canming Xie, Yao Wang, Jing Xiang, Litong Chen, Jia Yuan, Chutao Chen, Haomei Tian

Cerebral ischemia-reperfusion injury (CIRI) is the second leading cause of death worldwide, posing a huge risk to human life and health. Therefore, investigating the pathogenesis underlying CIRI and developing effective treatments are essential. Ferroptosis is an iron-dependent mode of cell death, which is caused by disorders in iron metabolism and lipid peroxidation. Previous studies demonstrated that ferroptosis is also a form of autophagic cell death, and nuclear receptor coactivator 4(NCOA4) mediated ferritinophagy was found to regulate ferroptosis by interfering with iron metabolism. Ferritinophagy and ferroptosis are important pathogenic mechanisms in CIRI. This review mainly summarizes the link and regulation between ferritinophagy and ferroptosis and further discusses their mechanisms in CIRI. In addition, the potential treatment methods targeting ferritinophagy and ferroptosis for CIRI are presented, providing new ideas for the prevention and treatment of clinical CIRI in the future.

脑缺血再灌注损伤(CIRI)是全球第二大死亡原因,对人类的生命和健康构成巨大威胁。因此,研究 CIRI 的发病机制并开发有效的治疗方法至关重要。铁变态反应是一种铁依赖性细胞死亡模式,由铁代谢紊乱和脂质过氧化引起。先前的研究表明,铁嗜酸也是细胞自噬死亡的一种形式,核受体辅激活子4(NCOA4)介导的铁蛋白吞噬被发现可通过干扰铁代谢来调节铁嗜酸。噬铁蛋白和铁突变是 CIRI 的重要致病机制。本综述主要总结了噬铁蛋白和嗜铁细胞之间的联系和调控,并进一步探讨了它们在 CIRI 中的作用机制。此外,还介绍了针对 CIRI 的嗜铁蛋白和嗜铁细胞的潜在治疗方法,为今后临床 CIRI 的预防和治疗提供了新思路。
{"title":"Ferritinophagy and Ferroptosis in Cerebral Ischemia Reperfusion Injury.","authors":"Xiaoyue Liu, Canming Xie, Yao Wang, Jing Xiang, Litong Chen, Jia Yuan, Chutao Chen, Haomei Tian","doi":"10.1007/s11064-024-04161-5","DOIUrl":"10.1007/s11064-024-04161-5","url":null,"abstract":"<p><p>Cerebral ischemia-reperfusion injury (CIRI) is the second leading cause of death worldwide, posing a huge risk to human life and health. Therefore, investigating the pathogenesis underlying CIRI and developing effective treatments are essential. Ferroptosis is an iron-dependent mode of cell death, which is caused by disorders in iron metabolism and lipid peroxidation. Previous studies demonstrated that ferroptosis is also a form of autophagic cell death, and nuclear receptor coactivator 4(NCOA4) mediated ferritinophagy was found to regulate ferroptosis by interfering with iron metabolism. Ferritinophagy and ferroptosis are important pathogenic mechanisms in CIRI. This review mainly summarizes the link and regulation between ferritinophagy and ferroptosis and further discusses their mechanisms in CIRI. In addition, the potential treatment methods targeting ferritinophagy and ferroptosis for CIRI are presented, providing new ideas for the prevention and treatment of clinical CIRI in the future.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11233298/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141247087","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Neuroprotective and Neurite Outgrowth Stimulating Effects of New Low-Basicity 5-HT7 Receptor Agonists: In Vitro Study in Human Neuroblastoma SH-SY5Y Cells. 新型低碱度 5-HT7 受体激动剂的神经保护和神经元生长刺激作用:人神经母细胞瘤 SH-SY5Y 细胞体外研究。
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-04 DOI: 10.1007/s11064-024-04159-z
Klaudia Jakubowska, Adam S Hogendorf, Sławomir Gołda, Danuta Jantas

There is some evidence that the serotonin receptor subtype 7 (5-HT7) could be new therapeutic target for neuroprotection. The aim of this study was to compare the neuroprotective and neurite outgrowth potential of new 5-HT7 receptor agonists (AH-494, AGH-238, AGH-194) with 5-CT (5-carboxyamidotryptamine) in human neuroblastoma SH-SY5Y cells. The results revealed that 5-HT7 mRNA expression was significantly higher in retinoic acid (RA)-differentiated cells when compared to undifferentiated ones and it was higher in cell cultured in neuroblastoma experimental medium (DMEM) compared to those placed in neuronal (NB) medium. Furthermore, the safety profile of compounds was favorable for all tested compounds at concentration used for neuroprotection evaluation (up to 1 μM), whereas at higher concentrations (above 10 μM) the one of the tested compounds, AGH-194 appeared to be cytotoxic. While we observed relatively modest protective effects of 5-CT and AH-494 in UN-SH-SY5Y cells cultured in DMEM, in UN-SH-SY5Y cells cultured in NB medium we found a significant reduction of H2O2-evoked cell damage by all tested 5-HT7 agonists. However, 5-HT7-mediated neuroprotection was not associated with inhibition of caspase-3 activity and was not observed in RA-SH-SY5Y cells exposed to H2O2. Furthermore, none of the tested 5-HT7 agonists altered the damage induced by 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenylpyridinium ion (MPP +) and doxorubicin (Dox) in UN- and RA-SH-SY5Y cells cultured in NB. Finally we showed a stimulating effect of AH-494 and AGH-194 on neurite outgrowth. The obtained results provide insight into neuroprotective and neurite outgrowth potential of new 5-HT7 agonists.

有证据表明,5-羟色胺受体亚型 7(5-HT7)可能是神经保护的新治疗靶点。本研究旨在比较新型 5-HT7 受体激动剂(AH-494、AGH-238、AGH-194)与 5-CT(5-羧基酰胺色胺)在人神经母细胞瘤 SH-SY5Y 细胞中的神经保护和神经元生长潜力。结果显示,与未分化细胞相比,5-HT7 mRNA 在维甲酸(RA)分化细胞中的表达量明显更高;与在神经元(NB)培养基中培养的细胞相比,在神经母细胞瘤实验培养基(DMEM)中培养的细胞中,5-HT7 mRNA 的表达量更高。此外,在用于神经保护评估的浓度下(最高为 1 μM),所有受测化合物的安全性都很好,而在较高浓度下(超过 10 μM),其中一种受测化合物 AGH-194 似乎具有细胞毒性。在 DMEM 培养的 UN-SH-SY5Y 细胞中,我们观察到 5-CT 和 AH-494 的保护作用相对较弱;而在 NB 培养基培养的 UN-SH-SY5Y 细胞中,我们发现所有测试的 5-HT7 激动剂都能显著减少 H2O2 诱导的细胞损伤。然而,5-HT7 介导的神经保护与抑制 caspase-3 活性无关,在暴露于 H2O2 的 RA-SH-SY5Y 细胞中也未观察到。此外,在 UN- 和 RA-SH-SY5Y 细胞培养的 NB 中,测试的 5-HT7 激动剂都不会改变 6-羟基多巴胺(6-OHDA)、1-甲基-4-苯基吡啶鎓离子(MPP +)和多柔比星(Dox)诱导的损伤。最后,我们还发现了 AH-494 和 AGH-194 对神经元生长的刺激作用。这些结果使我们对新型 5-HT7 激动剂的神经保护和神经元生长潜力有了更深入的了解。
{"title":"Neuroprotective and Neurite Outgrowth Stimulating Effects of New Low-Basicity 5-HT<sub>7</sub> Receptor Agonists: In Vitro Study in Human Neuroblastoma SH-SY5Y Cells.","authors":"Klaudia Jakubowska, Adam S Hogendorf, Sławomir Gołda, Danuta Jantas","doi":"10.1007/s11064-024-04159-z","DOIUrl":"10.1007/s11064-024-04159-z","url":null,"abstract":"<p><p>There is some evidence that the serotonin receptor subtype 7 (5-HT<sub>7</sub>) could be new therapeutic target for neuroprotection. The aim of this study was to compare the neuroprotective and neurite outgrowth potential of new 5-HT<sub>7</sub> receptor agonists (AH-494, AGH-238, AGH-194) with 5-CT (5-carboxyamidotryptamine) in human neuroblastoma SH-SY5Y cells. The results revealed that 5-HT<sub>7</sub> mRNA expression was significantly higher in retinoic acid (RA)-differentiated cells when compared to undifferentiated ones and it was higher in cell cultured in neuroblastoma experimental medium (DMEM) compared to those placed in neuronal (NB) medium. Furthermore, the safety profile of compounds was favorable for all tested compounds at concentration used for neuroprotection evaluation (up to 1 μM), whereas at higher concentrations (above 10 μM) the one of the tested compounds, AGH-194 appeared to be cytotoxic. While we observed relatively modest protective effects of 5-CT and AH-494 in UN-SH-SY5Y cells cultured in DMEM, in UN-SH-SY5Y cells cultured in NB medium we found a significant reduction of H<sub>2</sub>O<sub>2</sub>-evoked cell damage by all tested 5-HT<sub>7</sub> agonists. However, 5-HT<sub>7</sub>-mediated neuroprotection was not associated with inhibition of caspase-3 activity and was not observed in RA-SH-SY5Y cells exposed to H<sub>2</sub>O<sub>2</sub>. Furthermore, none of the tested 5-HT<sub>7</sub> agonists altered the damage induced by 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenylpyridinium ion (MPP +) and doxorubicin (Dox) in UN- and RA-SH-SY5Y cells cultured in NB. Finally we showed a stimulating effect of AH-494 and AGH-194 on neurite outgrowth. The obtained results provide insight into neuroprotective and neurite outgrowth potential of new 5-HT<sub>7</sub> agonists.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11233329/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141247137","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Neurochemical Research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1