首页 > 最新文献

Neurochemical Research最新文献

英文 中文
Neurochemical Anatomy of Cushing's Syndrome. 库欣综合征的神经化学解剖学。
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-04 DOI: 10.1007/s11064-024-04172-2
Robert Lalonde, Catherine Strazielle

The neurochemical anatomy underlying Cushing's syndrome is examined for regional brain metabolism as well as neurotransmitter levels and receptor binding of biogenic amines and amino acids. Preliminary studies generally indicate that glucose uptake, blood flow, and activation on fMRI scans decreased in neocortical areas and increased in subcortical areas of patients with Cushing's syndrome or disease. Glucocorticoid-mediated increases in hippocampal metabolism occurred despite in vitro evidence of glucocorticoid-induced decreases in glucose uptake or consumption, indicating that in vivo increases are the result of indirect, compensatory, or preliminary responses. In animal studies, glucocorticoid administration decreased 5HT levels and 5HT1A receptor binding in several brain regions while adrenalectomy increased such binding. Region-specific effects were also obtained in regard to the dopaminergic system, with predominant actions of glucocorticoid-induced potentiation of reuptake blockers and releasing agents. More in-depth neuroanatomical analyses are warranted of these and amino acid-related neurotransmission.

库欣综合征的神经化学解剖学研究包括区域脑代谢、神经递质水平以及生物胺和氨基酸的受体结合。初步研究普遍表明,库欣综合征或疾病患者的新皮层区域葡萄糖摄取量、血流量和 fMRI 扫描激活减少,皮层下区域增加。尽管有体外证据表明糖皮质激素诱导的葡萄糖摄取或消耗减少,但糖皮质激素介导的海马代谢增加仍然发生,这表明体内的增加是间接、代偿或初步反应的结果。在动物实验中,服用糖皮质激素会降低一些脑区的 5HT 水平和 5HT1A 受体结合率,而肾上腺切除术则会增加这种结合率。对多巴胺能系统也有特定区域的影响,主要是糖皮质激素诱导的再摄取阻断剂和释放剂的增效作用。有必要对这些和氨基酸相关的神经传递进行更深入的神经解剖学分析。
{"title":"Neurochemical Anatomy of Cushing's Syndrome.","authors":"Robert Lalonde, Catherine Strazielle","doi":"10.1007/s11064-024-04172-2","DOIUrl":"10.1007/s11064-024-04172-2","url":null,"abstract":"<p><p>The neurochemical anatomy underlying Cushing's syndrome is examined for regional brain metabolism as well as neurotransmitter levels and receptor binding of biogenic amines and amino acids. Preliminary studies generally indicate that glucose uptake, blood flow, and activation on fMRI scans decreased in neocortical areas and increased in subcortical areas of patients with Cushing's syndrome or disease. Glucocorticoid-mediated increases in hippocampal metabolism occurred despite in vitro evidence of glucocorticoid-induced decreases in glucose uptake or consumption, indicating that in vivo increases are the result of indirect, compensatory, or preliminary responses. In animal studies, glucocorticoid administration decreased 5HT levels and 5HT<sub>1A</sub> receptor binding in several brain regions while adrenalectomy increased such binding. Region-specific effects were also obtained in regard to the dopaminergic system, with predominant actions of glucocorticoid-induced potentiation of reuptake blockers and releasing agents. More in-depth neuroanatomical analyses are warranted of these and amino acid-related neurotransmission.</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":"141236260","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
Flavonoids as Potential Therapeutics Against Neurodegenerative Disorders: Unlocking the Prospects. 黄酮类化合物是治疗神经退行性疾病的潜在药物:开启前景。
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-01 DOI: 10.1007/s11064-024-04177-x
Md Faysal, Zerrouki Dehbia, Mehrukh Zehravi, Sherouk Hussein Sweilam, M Akiful Haque, Kusuma Praveen Kumar, Rita D Chakole, Satish P Shelke, Swapna Sirikonda, Mohamed H Nafady, Sharuk L Khan, Firzan Nainu, Irfan Ahmad, Talha Bin Emran

Neurodegeneration, the decline of nerve cells in the brain, is a common feature of neurodegenerative disorders (NDDs). Oxidative stress, a key factor in NDDs such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease can lead to neuronal cell death, mitochondria impairment, excitotoxicity, and Ca2+ stress. Environmental factors compromising stress response lead to cell damage, necessitating novel therapeutics for preventing or treating brain disorders in older individuals and an aging population. Synthetic medications offer symptomatic benefits but can have adverse effects. This research explores the potential of flavonoids derived from plants in treating NDDs. Flavonoids compounds, have been studied for their potential to enter the brain and treat NDDs. These compounds have diverse biological effects and are currently being explored for their potential in the treatment of central nervous system disorders. Flavonoids have various beneficial effects, including antiviral, anti-allergic, antiplatelet, anti-inflammatory, anti-tumor, anti-apoptotic, and antioxidant properties. Their potential to alleviate symptoms of NDDs is significant.

神经退行性变(脑神经细胞衰退)是神经退行性疾病(NDDs)的共同特征。氧化应激是阿尔茨海默病、帕金森病、肌萎缩性脊髓侧索硬化症和亨廷顿病等神经退行性疾病的一个关键因素,可导致神经细胞死亡、线粒体受损、兴奋毒性和 Ca2+ 应激。损害应激反应的环境因素会导致细胞损伤,因此需要新型疗法来预防或治疗老年人和老龄化人群的脑部疾病。合成药物可对症治疗,但也可能产生不良影响。这项研究探讨了从植物中提取的类黄酮在治疗 NDD 方面的潜力。黄酮类化合物因其进入大脑和治疗 NDD 的潜力而受到研究。这些化合物具有多种生物效应,目前正在探索其治疗中枢神经系统疾病的潜力。类黄酮具有多种有益作用,包括抗病毒、抗过敏、抗血小板、抗炎、抗肿瘤、抗细胞凋亡和抗氧化特性。它们在缓解非传染性疾病症状方面的潜力巨大。
{"title":"Flavonoids as Potential Therapeutics Against Neurodegenerative Disorders: Unlocking the Prospects.","authors":"Md Faysal, Zerrouki Dehbia, Mehrukh Zehravi, Sherouk Hussein Sweilam, M Akiful Haque, Kusuma Praveen Kumar, Rita D Chakole, Satish P Shelke, Swapna Sirikonda, Mohamed H Nafady, Sharuk L Khan, Firzan Nainu, Irfan Ahmad, Talha Bin Emran","doi":"10.1007/s11064-024-04177-x","DOIUrl":"10.1007/s11064-024-04177-x","url":null,"abstract":"<p><p>Neurodegeneration, the decline of nerve cells in the brain, is a common feature of neurodegenerative disorders (NDDs). Oxidative stress, a key factor in NDDs such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease can lead to neuronal cell death, mitochondria impairment, excitotoxicity, and Ca<sup>2+</sup> stress. Environmental factors compromising stress response lead to cell damage, necessitating novel therapeutics for preventing or treating brain disorders in older individuals and an aging population. Synthetic medications offer symptomatic benefits but can have adverse effects. This research explores the potential of flavonoids derived from plants in treating NDDs. Flavonoids compounds, have been studied for their potential to enter the brain and treat NDDs. These compounds have diverse biological effects and are currently being explored for their potential in the treatment of central nervous system disorders. Flavonoids have various beneficial effects, including antiviral, anti-allergic, antiplatelet, anti-inflammatory, anti-tumor, anti-apoptotic, and antioxidant properties. Their potential to alleviate symptoms of NDDs is significant.</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":"141185955","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
Phosphodiesterase 8 (PDE8): Distribution and Cellular Expression and Association with Alzheimer's Disease. 磷酸二酯酶 8 (PDE8):分布和细胞表达以及与阿尔茨海默病的关系
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-05-23 DOI: 10.1007/s11064-024-04156-2
Nian-Zhuang Qiu, Hui-Mei Hou, Tian-Yang Guo, Yu-Li Lv, Yao Zhou, Fang-Fang Zhang, Feng Zhang, Xiao-Dan Wang, Wei Chen, Yong-Feng Gao, Mei-Hua Chen, Xue-Hui Zhang, Han-Ting Zhang, Hao Wang

Phosphodiesterase 8 (PDE8), as a member of PDE superfamily, specifically promotes the hydrolysis and degradation of intracellular cyclic adenosine monophosphate (cAMP), which may be associated with pathogenesis of Alzheimer's disease (AD). However, little is currently known about potential role in the central nervous system (CNS). Here we investigated the distribution and expression of PDE8 in brain of mouse, which we believe can provide evidence for studying the role of PDE8 in CNS and the relationship between PDE8 and AD. Here, C57BL/6J mice were used to observe the distribution patterns of two subtypes of PDE8, PDE8A and PDE8B, in different sexes in vivo by western blot (WB). Meanwhile, C57BL/6J mice were also used to demonstrate the distribution pattern of PDE8 in selected brain regions and localization in neural cells by WB and multiplex immunofluorescence staining. Furthermore, the triple transgenic (3×Tg-AD) mice and wild type (WT) mice of different ages were used to investigate the changes of PDE8 expression in the hippocampus and cerebral cortex during the progression of AD. PDE8 was found to be widely expressed in multiple tissues and organs including heart, kidney, stomach, brain, and liver, spleen, intestines, and uterus, with differences in expression levels between the two subtypes of PDE8A and PDE8B, as well as two sexes. Meanwhile, PDE8 was widely distributed in the brain, especially in areas closely related to cognitive function such as cerebellum, striatum, amygdala, cerebral cortex, and hippocampus, without differences between sexes. Furthermore, PDE8A was found to be expressed in neuronal cells, microglia and astrocytes, while PDE8B is only expressed in neuronal cells and microglia. PDE8A expression in the hippocampus of both female and male 3×Tg-AD mice was gradually increased with ages and PDE8B expression was upregulated only in cerebral cortex of female 3×Tg-AD mice with ages. However, the expression of PDE8A and PDE8B was apparently increased in both cerebral cortex and hippocampus in both female and male 10-month-old 3×Tg-AD mice compared WT mice. These results suggest that PDE8 may be associated with the progression of AD and is a potential target for its prevention and treatment in the future.

磷酸二酯酶 8(PDE8)是磷酸二酯酶超家族的成员,专门促进细胞内环磷酸腺苷(cAMP)的水解和降解,这可能与阿尔茨海默病(AD)的发病机制有关。然而,目前人们对其在中枢神经系统(CNS)中的潜在作用知之甚少。在此,我们研究了 PDE8 在小鼠大脑中的分布和表达,相信这能为研究 PDE8 在中枢神经系统中的作用以及 PDE8 与 AD 的关系提供证据。本研究以C57BL/6J小鼠为研究对象,通过Western blot(WB)技术观察PDE8的两种亚型PDE8A和PDE8B在不同性别小鼠体内的分布模式。同时,还利用C57BL/6J小鼠,通过WB和多重免疫荧光染色法证明了PDE8在选定脑区的分布模式以及在神经细胞中的定位。此外,还利用三重转基因(3×Tg-AD)小鼠和不同年龄的野生型(WT)小鼠研究了PDE8在AD进展过程中在海马和大脑皮层的表达变化。结果发现,PDE8广泛表达于心、肾、胃、脑、肝、脾、肠、子宫等多个组织和器官,PDE8A和PDE8B两种亚型以及两种性别的表达水平存在差异。同时,PDE8广泛分布于大脑,尤其是与认知功能密切相关的区域,如小脑、纹状体、杏仁核、大脑皮层和海马,且无性别差异。此外,研究还发现,PDE8A 在神经元细胞、小胶质细胞和星形胶质细胞中均有表达,而 PDE8B 仅在神经元细胞和小胶质细胞中表达。随着年龄的增长,PDE8A在雌性和雄性3×Tg-AD小鼠海马中的表达逐渐增加,而PDE8B仅在雌性3×Tg-AD小鼠大脑皮层中表达上调。然而,与 WT 小鼠相比,雌性和雄性 10 月龄 3×Tg-AD 小鼠大脑皮层和海马中 PDE8A 和 PDE8B 的表达均明显增加。这些结果表明,PDE8可能与AD的进展有关,是未来预防和治疗AD的潜在靶点。
{"title":"Phosphodiesterase 8 (PDE8): Distribution and Cellular Expression and Association with Alzheimer's Disease.","authors":"Nian-Zhuang Qiu, Hui-Mei Hou, Tian-Yang Guo, Yu-Li Lv, Yao Zhou, Fang-Fang Zhang, Feng Zhang, Xiao-Dan Wang, Wei Chen, Yong-Feng Gao, Mei-Hua Chen, Xue-Hui Zhang, Han-Ting Zhang, Hao Wang","doi":"10.1007/s11064-024-04156-2","DOIUrl":"10.1007/s11064-024-04156-2","url":null,"abstract":"<p><p>Phosphodiesterase 8 (PDE8), as a member of PDE superfamily, specifically promotes the hydrolysis and degradation of intracellular cyclic adenosine monophosphate (cAMP), which may be associated with pathogenesis of Alzheimer's disease (AD). However, little is currently known about potential role in the central nervous system (CNS). Here we investigated the distribution and expression of PDE8 in brain of mouse, which we believe can provide evidence for studying the role of PDE8 in CNS and the relationship between PDE8 and AD. Here, C57BL/6J mice were used to observe the distribution patterns of two subtypes of PDE8, PDE8A and PDE8B, in different sexes in vivo by western blot (WB). Meanwhile, C57BL/6J mice were also used to demonstrate the distribution pattern of PDE8 in selected brain regions and localization in neural cells by WB and multiplex immunofluorescence staining. Furthermore, the triple transgenic (3×Tg-AD) mice and wild type (WT) mice of different ages were used to investigate the changes of PDE8 expression in the hippocampus and cerebral cortex during the progression of AD. PDE8 was found to be widely expressed in multiple tissues and organs including heart, kidney, stomach, brain, and liver, spleen, intestines, and uterus, with differences in expression levels between the two subtypes of PDE8A and PDE8B, as well as two sexes. Meanwhile, PDE8 was widely distributed in the brain, especially in areas closely related to cognitive function such as cerebellum, striatum, amygdala, cerebral cortex, and hippocampus, without differences between sexes. Furthermore, PDE8A was found to be expressed in neuronal cells, microglia and astrocytes, while PDE8B is only expressed in neuronal cells and microglia. PDE8A expression in the hippocampus of both female and male 3×Tg-AD mice was gradually increased with ages and PDE8B expression was upregulated only in cerebral cortex of female 3×Tg-AD mice with ages. However, the expression of PDE8A and PDE8B was apparently increased in both cerebral cortex and hippocampus in both female and male 10-month-old 3×Tg-AD mice compared WT mice. These results suggest that PDE8 may be associated with the progression of AD and is a potential target for its prevention and treatment 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":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141086403","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
Epigenetic Mechanisms of Aluminum-Induced Neurotoxicity and Alzheimer's Disease: A Focus on Non-Coding RNAs. 铝诱导神经毒性和阿尔茨海默病的表观遗传学机制:聚焦非编码 RNA。
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-27 DOI: 10.1007/s11064-024-04214-9
Michael Aschner, Anatoly V Skalny, Abel Santamaria, Joao B T Rocha, Borhan Mansouri, Yousef Tizabi, Roberto Madeddu, Rongzu Lu, Eunsook Lee, Alexey A Tinkov

Aluminum (Al) is known to induce neurotoxic effects, potentially contributing to Alzheimer's disease (AD) pathogenesis. Recent studies suggest that epigenetic modification may contribute to Al neurotoxicity, although the mechanisms are still debatable. Therefore, the objective of the present study was to summarize existing data on the involvement of epigenetic mechanisms in Al-induced neurotoxicity, especially AD-type pathology. Existing data demonstrate that Al exposure induces disruption in DNA methylation, histone modifications, and non-coding RNA expression in brains. Alterations in DNA methylation following Al exposure were shown to be mediated by changes in expression and activity of DNA methyltransferases (DNMTs) and ten-eleven translocation proteins (TETs). Al exposure was shown to reduce histone acetylation by up-regulating expression of histone deacetylases (HDACs) and impair histone methylation, ultimately contributing to down-regulation of brain-derived neurotrophic factor (BDNF) expression and activation of nuclear factor κB (NF-κB) signaling. Neurotoxic effects of Al exposure were also associated with aberrant expression of non-coding RNAs, especially microRNAs (miR). Al-induced patterns of miR expression were involved in development of AD-type pathology by increasing amyloid β (Aβ) production through up-regulation of Aβ precursor protein (APP) and β secretase (BACE1) expression (down-regulation of miR-29a/b, miR-101, miR-124, and Let-7c expression), increasing in neuroinflammation through NF-κB signaling (up-regulation of miR-9, miR-125b, miR-128, and 146a), as well as modulating other signaling pathways. Furthermore, reduced global DNA methylation, altered histone modification, and aberrant miRNA expression were associated with cognitive decline in Al-exposed subjects. However, further studies are required to evaluate the contribution of epigenetic mechanisms to Al-induced neurotoxicity and/or AD development.

众所周知,铝(Al)具有神经毒性作用,可能导致阿尔茨海默病(AD)的发病。最近的研究表明,表观遗传修饰可能有助于铝的神经毒性,但其机制仍有待商榷。因此,本研究旨在总结表观遗传机制参与铝诱导的神经毒性,尤其是 AD 型病理学的现有数据。现有数据表明,铝暴露会诱导大脑中DNA甲基化、组蛋白修饰和非编码RNA表达的破坏。研究表明,暴露于铝后DNA甲基化的改变是由DNA甲基转移酶(DNMTs)和十-十一转位蛋白(TETs)的表达和活性变化介导的。研究表明,接触铝会通过上调组蛋白去乙酰化酶(HDACs)的表达来减少组蛋白乙酰化,并损害组蛋白甲基化,最终导致脑源性神经营养因子(BDNF)表达下调和核因子κB(NF-κB)信号的激活。铝暴露的神经毒性效应还与非编码 RNA,特别是微 RNA(miR)的异常表达有关。铝诱导的 miR 表达模式通过上调 Aβ 前体蛋白(APP)和 β 分泌酶(BACE1)的表达(下调 miR-29a/b、miR-101、miR-124 和 Let-7c 的表达),通过 NF-κB 信号转导增加神经炎症(上调 miR-9、miR-125b、miR-128 和 146a),以及调节其他信号通路。此外,全局 DNA 甲基化的减少、组蛋白修饰的改变和 miRNA 表达的异常与暴露于铝的受试者认知能力的下降有关。然而,要评估表观遗传机制对铝诱导的神经毒性和/或注意力缺失症发展的贡献,还需要进一步的研究。
{"title":"Epigenetic Mechanisms of Aluminum-Induced Neurotoxicity and Alzheimer's Disease: A Focus on Non-Coding RNAs.","authors":"Michael Aschner, Anatoly V Skalny, Abel Santamaria, Joao B T Rocha, Borhan Mansouri, Yousef Tizabi, Roberto Madeddu, Rongzu Lu, Eunsook Lee, Alexey A Tinkov","doi":"10.1007/s11064-024-04214-9","DOIUrl":"https://doi.org/10.1007/s11064-024-04214-9","url":null,"abstract":"<p><p>Aluminum (Al) is known to induce neurotoxic effects, potentially contributing to Alzheimer's disease (AD) pathogenesis. Recent studies suggest that epigenetic modification may contribute to Al neurotoxicity, although the mechanisms are still debatable. Therefore, the objective of the present study was to summarize existing data on the involvement of epigenetic mechanisms in Al-induced neurotoxicity, especially AD-type pathology. Existing data demonstrate that Al exposure induces disruption in DNA methylation, histone modifications, and non-coding RNA expression in brains. Alterations in DNA methylation following Al exposure were shown to be mediated by changes in expression and activity of DNA methyltransferases (DNMTs) and ten-eleven translocation proteins (TETs). Al exposure was shown to reduce histone acetylation by up-regulating expression of histone deacetylases (HDACs) and impair histone methylation, ultimately contributing to down-regulation of brain-derived neurotrophic factor (BDNF) expression and activation of nuclear factor κB (NF-κB) signaling. Neurotoxic effects of Al exposure were also associated with aberrant expression of non-coding RNAs, especially microRNAs (miR). Al-induced patterns of miR expression were involved in development of AD-type pathology by increasing amyloid β (Aβ) production through up-regulation of Aβ precursor protein (APP) and β secretase (BACE1) expression (down-regulation of miR-29a/b, miR-101, miR-124, and Let-7c expression), increasing in neuroinflammation through NF-κB signaling (up-regulation of miR-9, miR-125b, miR-128, and 146a), as well as modulating other signaling pathways. Furthermore, reduced global DNA methylation, altered histone modification, and aberrant miRNA expression were associated with cognitive decline in Al-exposed subjects. However, further studies are required to evaluate the contribution of epigenetic mechanisms to Al-induced neurotoxicity and/or AD development.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141764722","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
Targeting Adipokines: A Promising Therapeutic Strategy for Epilepsy. 靶向脂肪因子:治疗癫痫病的前景广阔的策略
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-26 DOI: 10.1007/s11064-024-04219-4
Iqraa Shaikh, Lokesh Kumar Bhatt

Epilepsy affects 65 million people globally and causes neurobehavioral, cognitive, and psychological defects. Although research on the disease is progressing and a wide range of treatments are available, approximately 30% of people have refractory epilepsy that cannot be managed with conventional medications. This underlines the importance of further understanding the condition and exploring cutting-edge targets for treatment. Adipokines are peptides secreted by adipocyte's white adipose tissue, involved in controlling food intake and metabolism. Their regulatory functions in the central nervous system (CNS) are multifaceted and identified in several physiology and pathologies. Adipokines play a role in oxidative stress and neuroinflammation which are associated with brain degeneration and connected neurological diseases. This review aims to highlight the potential impacts of leptin, adiponectin, apelin, vaspin, visfatin, and chimerin in the pathogenesis of epilepsy.

癫痫影响着全球 6500 万人,并导致神经行为、认知和心理缺陷。尽管对这种疾病的研究正在取得进展,并有多种治疗方法可供选择,但仍有约 30% 的人患有难治性癫痫,无法用常规药物治疗。这凸显了进一步了解这种疾病并探索前沿治疗目标的重要性。脂肪因子是脂肪细胞白色脂肪组织分泌的多肽,参与控制食物摄入和新陈代谢。它们在中枢神经系统(CNS)中的调控功能是多方面的,并在多种生理和病理现象中被发现。脂肪因子在氧化应激和神经炎症中发挥作用,而氧化应激和神经炎症与大脑退化和相关神经疾病有关。本综述旨在强调瘦素、脂肪连素、apelin、vaspin、visfatin 和 chimerin 在癫痫发病机制中的潜在影响。
{"title":"Targeting Adipokines: A Promising Therapeutic Strategy for Epilepsy.","authors":"Iqraa Shaikh, Lokesh Kumar Bhatt","doi":"10.1007/s11064-024-04219-4","DOIUrl":"10.1007/s11064-024-04219-4","url":null,"abstract":"<p><p>Epilepsy affects 65 million people globally and causes neurobehavioral, cognitive, and psychological defects. Although research on the disease is progressing and a wide range of treatments are available, approximately 30% of people have refractory epilepsy that cannot be managed with conventional medications. This underlines the importance of further understanding the condition and exploring cutting-edge targets for treatment. Adipokines are peptides secreted by adipocyte's white adipose tissue, involved in controlling food intake and metabolism. Their regulatory functions in the central nervous system (CNS) are multifaceted and identified in several physiology and pathologies. Adipokines play a role in oxidative stress and neuroinflammation which are associated with brain degeneration and connected neurological diseases. This review aims to highlight the potential impacts of leptin, adiponectin, apelin, vaspin, visfatin, and chimerin in the pathogenesis of epilepsy.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141764724","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
Physical Exercise-Induced Activation of NRF2 and BDNF as a Promising Strategy for Ferroptosis Regulation in Parkinson's Disease. 体育锻炼诱导的 NRF2 和 BDNF 激活有望成为调节帕金森病铁蛋白沉积的策略。
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-01 Epub Date: 2024-05-24 DOI: 10.1007/s11064-024-04152-6
Anand Thirupathi, Luis Felipe Marqueze, Tiago F Outeiro, Zsolt Radak, Ricardo A Pinho

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra. Ferroptosis, an iron-dependent form of regulated cell death, may contribute to the progression of PD owing to an unbalanced brain redox status. Physical exercise is a complementary therapy that can modulate ferroptosis in PD by regulating the redox system through the activation of nuclear factor (erythroid-derived 2)-like 2 (NRF2) and brain-derived neurotrophic factor (BDNF) signaling. However, the precise effects of physical exercise on ferroptosis in PD remain unclear. In this review, we explored how physical exercise influences NRF2 and BDNF signaling and affects ferroptosis in PD. We further investigated relevant publications over the past two decades by searching the PubMed, Web of Science, and Google Scholar databases using keywords related to physical exercise, PD, ferroptosis, and neurotrophic factor antioxidant signaling. This review provides insights into current research gaps and demonstrates the necessity for future research to elucidate the specific mechanisms by which exercise regulates ferroptosis in PD, including the assessment of different exercise protocols and their long-term effects. Ultimately, exploring these aspects may lead to the development of improved exercise interventions for the better management of patients with PD.

帕金森病(PD)是一种进行性神经退行性疾病,其特征是黑质中多巴胺能神经元的丧失。由于大脑氧化还原状态失衡,铁中毒(一种依赖铁的调节性细胞死亡形式)可能会导致帕金森病的进展。体育锻炼是一种辅助疗法,可通过激活核因子(红细胞衍生 2)-类 2(NRF2)和脑源性神经营养因子(BDNF)信号来调节氧化还原系统,从而调节帕金森病的铁氧化。然而,体育锻炼对帕金森病铁氧化的确切影响仍不清楚。在这篇综述中,我们探讨了体育锻炼如何影响 NRF2 和 BDNF 信号传导,以及如何影响帕金森病的铁蛋白沉积。我们使用与体育锻炼、帕金森病、铁突变和神经营养因子抗氧化信号转导相关的关键词搜索了 PubMed、Web of Science 和 Google Scholar 数据库,进一步研究了过去二十年中的相关文献。本综述深入探讨了目前的研究空白,并表明未来研究有必要阐明运动调节帕金森病铁蛋白沉积的具体机制,包括评估不同的运动方案及其长期影响。最终,对这些方面的探索可能会开发出更好的运动干预措施,从而更好地管理帕金森病患者。
{"title":"Physical Exercise-Induced Activation of NRF2 and BDNF as a Promising Strategy for Ferroptosis Regulation in Parkinson's Disease.","authors":"Anand Thirupathi, Luis Felipe Marqueze, Tiago F Outeiro, Zsolt Radak, Ricardo A Pinho","doi":"10.1007/s11064-024-04152-6","DOIUrl":"10.1007/s11064-024-04152-6","url":null,"abstract":"<p><p>Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra. Ferroptosis, an iron-dependent form of regulated cell death, may contribute to the progression of PD owing to an unbalanced brain redox status. Physical exercise is a complementary therapy that can modulate ferroptosis in PD by regulating the redox system through the activation of nuclear factor (erythroid-derived 2)-like 2 (NRF2) and brain-derived neurotrophic factor (BDNF) signaling. However, the precise effects of physical exercise on ferroptosis in PD remain unclear. In this review, we explored how physical exercise influences NRF2 and BDNF signaling and affects ferroptosis in PD. We further investigated relevant publications over the past two decades by searching the PubMed, Web of Science, and Google Scholar databases using keywords related to physical exercise, PD, ferroptosis, and neurotrophic factor antioxidant signaling. This review provides insights into current research gaps and demonstrates the necessity for future research to elucidate the specific mechanisms by which exercise regulates ferroptosis in PD, including the assessment of different exercise protocols and their long-term effects. Ultimately, exploring these aspects may lead to the development of improved exercise interventions for the better management of patients with PD.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141086405","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
Prolactin is an Endogenous Antioxidant Factor in Astrocytes That Limits Oxidative Stress-Induced Astrocytic Cell Death via the STAT3/NRF2 Signaling Pathway. 催乳素是星形胶质细胞中的内源性抗氧化因子,可通过 STAT3/NRF2 信号通路限制氧化应激诱导的星形胶质细胞死亡
IF 4.4 3区 医学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-07-01 Epub Date: 2024-05-17 DOI: 10.1007/s11064-024-04147-3
Miriam Ulloa, Fernando Macías, Carmen Clapp, Gonzalo Martínez de la Escalera, Edith Arnold

Oxidative stress-induced death of neurons and astrocytes contributes to the pathogenesis of numerous neurodegenerative diseases. While significant progress has been made in identifying neuroprotective molecules against neuronal oxidative damage, little is known about their counterparts for astrocytes. Prolactin (PRL), a hormone known to stimulate astroglial proliferation, viability, and cytokine expression, exhibits antioxidant effects in neurons. However, its role in protecting astrocytes from oxidative stress remains unexplored. Here, we investigated the effect of PRL against hydrogen peroxide (H2O2)-induced oxidative insult in primary cortical astrocyte cultures. Incubation of astrocytes with PRL led to increased enzymatic activity of superoxide dismutase (SOD) and glutathione peroxidase (GPX), resulting in higher total antioxidant capacity. Concomitantly, PRL prevented H2O2-induced cell death, reactive oxygen species accumulation, and protein and lipid oxidation. The protective effect of PRL upon H2O2-induced cell death can be explained by the activation of both signal transducer and activator of transcription 3 (STAT3) and NFE2 like bZIP transcription factor 2 (NRF2) transduction cascades. We demonstrated that PRL induced nuclear translocation and transcriptional upregulation of Nrf2, concurrently with the transcriptional upregulation of the NRF2-dependent genes heme oxygenase 1, Sod1, Sod2, and Gpx1. Pharmacological blockade of STAT3 suppressed PRL-induced transcriptional upregulation of Nrf2, Sod1 and Gpx1 mRNA, and SOD and GPX activities. Furthermore, genetic ablation of the PRL receptor increased astroglial susceptibility to H2O2-induced cell death and superoxide accumulation, while diminishing their intrinsic antioxidant capacity. Overall, these findings unveil PRL as a potent antioxidant hormone that protects astrocytes from oxidative insult, which may contribute to brain neuroprotection.

氧化应激引起的神经元和星形胶质细胞死亡是多种神经退行性疾病的发病机制之一。尽管在确定神经元氧化损伤的神经保护分子方面取得了重大进展,但人们对星形胶质细胞的相应分子却知之甚少。催乳素(PRL)是一种已知能刺激星形胶质细胞增殖、活力和细胞因子表达的激素,在神经元中具有抗氧化作用。然而,它在保护星形胶质细胞免受氧化应激方面的作用仍有待探索。在这里,我们研究了 PRL 对原代皮质星形胶质细胞培养物中过氧化氢(H2O2)诱导的氧化损伤的影响。用 PRL 培养星形胶质细胞可提高超氧化物歧化酶(SOD)和谷胱甘肽过氧化物酶(GPX)的酶活性,从而提高总抗氧化能力。同时,PRL 还能防止 H2O2 诱导的细胞死亡、活性氧积累以及蛋白质和脂质氧化。PRL对H2O2诱导的细胞死亡的保护作用可通过激活信号转导子和转录激活子3(STAT3)以及类似于bZIP转录因子2(NRF2)的NFE2转导级联来解释。我们证实,PRL诱导Nrf2的核转位和转录上调,同时NRF2依赖基因血红素加氧酶1、Sod1、Sod2和Gpx1也转录上调。药物阻断 STAT3 可抑制 PRL 诱导的 Nrf2、Sod1 和 Gpx1 mRNA 转录上调以及 SOD 和 GPX 活性。此外,PRL 受体的遗传消减增加了星形胶质细胞对 H2O2 诱导的细胞死亡和超氧化物积累的敏感性,同时降低了其内在的抗氧化能力。总之,这些研究结果揭示了 PRL 是一种有效的抗氧化激素,它能保护星形胶质细胞免受氧化损伤,这可能有助于脑神经保护。
{"title":"Prolactin is an Endogenous Antioxidant Factor in Astrocytes That Limits Oxidative Stress-Induced Astrocytic Cell Death via the STAT3/NRF2 Signaling Pathway.","authors":"Miriam Ulloa, Fernando Macías, Carmen Clapp, Gonzalo Martínez de la Escalera, Edith Arnold","doi":"10.1007/s11064-024-04147-3","DOIUrl":"10.1007/s11064-024-04147-3","url":null,"abstract":"<p><p>Oxidative stress-induced death of neurons and astrocytes contributes to the pathogenesis of numerous neurodegenerative diseases. While significant progress has been made in identifying neuroprotective molecules against neuronal oxidative damage, little is known about their counterparts for astrocytes. Prolactin (PRL), a hormone known to stimulate astroglial proliferation, viability, and cytokine expression, exhibits antioxidant effects in neurons. However, its role in protecting astrocytes from oxidative stress remains unexplored. Here, we investigated the effect of PRL against hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>)-induced oxidative insult in primary cortical astrocyte cultures. Incubation of astrocytes with PRL led to increased enzymatic activity of superoxide dismutase (SOD) and glutathione peroxidase (GPX), resulting in higher total antioxidant capacity. Concomitantly, PRL prevented H<sub>2</sub>O<sub>2</sub>-induced cell death, reactive oxygen species accumulation, and protein and lipid oxidation. The protective effect of PRL upon H<sub>2</sub>O<sub>2</sub>-induced cell death can be explained by the activation of both signal transducer and activator of transcription 3 (STAT3) and NFE2 like bZIP transcription factor 2 (NRF2) transduction cascades. We demonstrated that PRL induced nuclear translocation and transcriptional upregulation of Nrf2, concurrently with the transcriptional upregulation of the NRF2-dependent genes heme oxygenase 1, Sod1, Sod2, and Gpx1. Pharmacological blockade of STAT3 suppressed PRL-induced transcriptional upregulation of Nrf2, Sod1 and Gpx1 mRNA, and SOD and GPX activities. Furthermore, genetic ablation of the PRL receptor increased astroglial susceptibility to H<sub>2</sub>O<sub>2</sub>-induced cell death and superoxide accumulation, while diminishing their intrinsic antioxidant capacity. Overall, these findings unveil PRL as a potent antioxidant hormone that protects astrocytes from oxidative insult, which may contribute to brain neuroprotection.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11144156/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140955674","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
Is Methylglyoxal a Potential Biomarker for the Warburg Effect Induced by the Lipopolysaccharide Neuroinflammation Model? 甲基乙二酸是脂多糖神经炎症模型诱发沃伯格效应的潜在生物标志物吗?
IF 4.4 3区 医学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-07-01 Epub Date: 2024-05-10 DOI: 10.1007/s11064-024-04142-8
Adriana Fernanda Kuckartz Vizuete, Carlos-Alberto Gonçalves

Methylglyoxal (MG) is considered a classical biomarker of diabetes mellitus and its comorbidities. However, a role for this compound in exacerbated immune responses, such as septicemia, is being increasingly observed and requires clarification, particularly in the context of neuroinflammatory responses. Herein, we used two different approaches (in vivo and acute hippocampal slice models) to investigate MG as a biomarker of neuroinflammation and the neuroimmunometabolic shift to glycolysis in lipopolysaccharide (LPS) inflammation models. Our data reinforce the hypothesis that LPS-induced neuroinflammation stimulates the cerebral innate immune response by increasing IL-1β, a classical pro-inflammatory cytokine, and the astrocyte reactive response, via elevating S100B secretion and GFAP levels. Acute neuroinflammation promotes an early neuroimmunometabolic shift to glycolysis by elevating glucose uptake, lactate release, PFK1, and PK activities. We observed high serum and cerebral MG levels, in association with a reduction in glyoxalase 1 detoxification activity, and a close correlation between serum and hippocampus MG levels with the systemic and neuroinflammatory responses to LPS. Findings strongly suggest a role for MG in immune responses.

甲基乙二酸(MG)被认为是糖尿病及其并发症的经典生物标志物。然而,越来越多的人观察到这种化合物在败血症等免疫反应恶化中的作用,需要加以澄清,尤其是在神经炎症反应的背景下。在此,我们采用两种不同的方法(体内和急性海马片模型)研究了作为神经炎症生物标志物的 MG 以及脂多糖(LPS)炎症模型中神经免疫代谢向糖酵解的转变。我们的数据加强了这样的假设:LPS诱导的神经炎症通过增加经典的促炎细胞因子IL-1β和星形胶质细胞反应性反应,刺激大脑先天性免疫反应,从而提高S100B分泌和GFAP水平。急性神经炎症通过提高葡萄糖摄取、乳酸释放、PFK1 和 PK 活性,促进早期神经免疫代谢向糖酵解转变。我们观察到血清和大脑中的 MG 含量较高,同时乙二醛酶 1 的解毒活性降低,而且血清和海马 MG 含量与 LPS 的全身反应和神经炎症反应密切相关。研究结果有力地说明了 MG 在免疫反应中的作用。
{"title":"Is Methylglyoxal a Potential Biomarker for the Warburg Effect Induced by the Lipopolysaccharide Neuroinflammation Model?","authors":"Adriana Fernanda Kuckartz Vizuete, Carlos-Alberto Gonçalves","doi":"10.1007/s11064-024-04142-8","DOIUrl":"10.1007/s11064-024-04142-8","url":null,"abstract":"<p><p>Methylglyoxal (MG) is considered a classical biomarker of diabetes mellitus and its comorbidities. However, a role for this compound in exacerbated immune responses, such as septicemia, is being increasingly observed and requires clarification, particularly in the context of neuroinflammatory responses. Herein, we used two different approaches (in vivo and acute hippocampal slice models) to investigate MG as a biomarker of neuroinflammation and the neuroimmunometabolic shift to glycolysis in lipopolysaccharide (LPS) inflammation models. Our data reinforce the hypothesis that LPS-induced neuroinflammation stimulates the cerebral innate immune response by increasing IL-1β, a classical pro-inflammatory cytokine, and the astrocyte reactive response, via elevating S100B secretion and GFAP levels. Acute neuroinflammation promotes an early neuroimmunometabolic shift to glycolysis by elevating glucose uptake, lactate release, PFK1, and PK activities. We observed high serum and cerebral MG levels, in association with a reduction in glyoxalase 1 detoxification activity, and a close correlation between serum and hippocampus MG levels with the systemic and neuroinflammatory responses to LPS. Findings strongly suggest a role for MG in immune responses.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140896843","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
Mir155hg Accelerates Hippocampal Neuron Injury in Convulsive Status Epilepticus by Inhibiting Microglial Phagocytosis. Mir155hg 通过抑制小胶质细胞的吞噬作用加速惊厥性癫痫状态中海马神经元的损伤
IF 4.4 3区 医学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-07-01 Epub Date: 2024-03-30 DOI: 10.1007/s11064-024-04131-x
Ming Wang, Binyuan Xu, Yangmei Xie, Ge Yao, Yinghui Chen

Convulsive status epilepticus (CSE) is a common critical neurological condition that can lead to irreversible hippocampal neuron damage and cognitive dysfunction. Multiple studies have demonstrated the critical roles that long non-coding RNA Mir155hg plays in a variety of diseases. However, less is known about the function and mechanism of Mir155hg in CSE. Here we investigate and elucidate the mechanism underlying the contribution of Mir155hg to CSE-induced hippocampal neuron injury. By applying high-throughput sequencing, we examined the expression of differentially expressed genes in normal and CSE rats. Subsequent RT-qPCR enabled us to measure the level of Mir155hg in rat hippocampal tissue. Targeted knockdown of Mir155hg was achieved by the AAV9 virus. Additionally, we utilized HE and Tunel staining to evaluate neuronal injury. Immunofluorescence (IF), Golgi staining, and brain path clamping were also used to detect the synaptic plasticity of hippocampal neurons. Finally, through IF staining and Sholl analysis, we assessed the degree of microglial phagocytic function. It was found that the expression of Mir155hg was elevated in CSE rats. HE and Tunel staining results showed that Mir155hg knockdown suppressed the hippocampal neuron loss and apoptosis followed CSE. IF, Golgi staining and brain path clamp data found that Mir155hg knockdown enhanced neuronal synaptic plasticity. The results from IF staining and Sholl analysis showed that Mir155hg knockdown enhanced microglial phagocytosis. Our findings suggest that Mir155hg promotes CSE-induced hippocampal neuron injury by inhibiting microglial phagocytosis.

惊厥性癫痫(CSE)是一种常见的危重神经系统疾病,可导致不可逆的海马神经元损伤和认知功能障碍。多项研究表明,长非编码 RNA Mir155hg 在多种疾病中发挥着关键作用。然而,人们对 Mir155hg 在 CSE 中的功能和机制知之甚少。在这里,我们研究并阐明了 Mir155hg 在 CSE 诱导的海马神经元损伤中的作用机制。通过应用高通量测序技术,我们检测了正常大鼠和 CSE 大鼠中差异表达基因的表达情况。随后的 RT-qPCR 使我们能够测量 Mir155hg 在大鼠海马组织中的水平。通过 AAV9 病毒实现了 Mir155hg 的靶向敲除。此外,我们还利用 HE 和 Tunel 染色来评估神经元损伤。免疫荧光(IF)、高尔基体染色和脑路径钳夹也被用来检测海马神经元的突触可塑性。最后,通过 IF 染色和 Sholl 分析,我们评估了小胶质细胞吞噬功能的程度。结果发现,Mir155hg在CSE大鼠中表达升高。HE和Tunel染色结果显示,敲除Mir155hg抑制了CSE后海马神经元的丢失和凋亡。IF、高尔基体染色和脑路径钳夹数据发现,Mir155hg敲除增强了神经元突触的可塑性。IF染色和Sholl分析结果显示,Mir155hg敲除增强了小胶质细胞的吞噬能力。我们的研究结果表明,Mir155hg通过抑制小胶质细胞的吞噬作用促进了CSE诱导的海马神经元损伤。
{"title":"Mir155hg Accelerates Hippocampal Neuron Injury in Convulsive Status Epilepticus by Inhibiting Microglial Phagocytosis.","authors":"Ming Wang, Binyuan Xu, Yangmei Xie, Ge Yao, Yinghui Chen","doi":"10.1007/s11064-024-04131-x","DOIUrl":"10.1007/s11064-024-04131-x","url":null,"abstract":"<p><p>Convulsive status epilepticus (CSE) is a common critical neurological condition that can lead to irreversible hippocampal neuron damage and cognitive dysfunction. Multiple studies have demonstrated the critical roles that long non-coding RNA Mir155hg plays in a variety of diseases. However, less is known about the function and mechanism of Mir155hg in CSE. Here we investigate and elucidate the mechanism underlying the contribution of Mir155hg to CSE-induced hippocampal neuron injury. By applying high-throughput sequencing, we examined the expression of differentially expressed genes in normal and CSE rats. Subsequent RT-qPCR enabled us to measure the level of Mir155hg in rat hippocampal tissue. Targeted knockdown of Mir155hg was achieved by the AAV9 virus. Additionally, we utilized HE and Tunel staining to evaluate neuronal injury. Immunofluorescence (IF), Golgi staining, and brain path clamping were also used to detect the synaptic plasticity of hippocampal neurons. Finally, through IF staining and Sholl analysis, we assessed the degree of microglial phagocytic function. It was found that the expression of Mir155hg was elevated in CSE rats. HE and Tunel staining results showed that Mir155hg knockdown suppressed the hippocampal neuron loss and apoptosis followed CSE. IF, Golgi staining and brain path clamp data found that Mir155hg knockdown enhanced neuronal synaptic plasticity. The results from IF staining and Sholl analysis showed that Mir155hg knockdown enhanced microglial phagocytosis. Our findings suggest that Mir155hg promotes CSE-induced hippocampal neuron injury by inhibiting microglial phagocytosis.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140329467","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-Mediated Hippocampus Ferroptosis is Involved in Cognitive Impairment in Immature Rats Induced by Hypoxia Combined with Propofol. 缺氧联合异丙酚诱导的未成熟大鼠认知功能受损与铁蛋白吞噬介导的海马铁突变有关
IF 4.4 3区 医学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-07-01 Epub Date: 2024-03-21 DOI: 10.1007/s11064-024-04128-6
Ling Liu, Wen Gao, Shun Yang, Fei Yang, Shangyingying Li, Yaqiong Tian, Li Yang, Qianyu Deng, Zhengwei Gan, Shengfen Tu

Propofol is a clinically common intravenous general anesthetic and is widely used for anesthesia induction, maintenance and intensive care unit (ICU) sedation in children. Hypoxemia is a common perioperative complication. In clinical work, we found that children with hypoxemia who received propofol anesthesia experienced significant postoperative cognitive changes. To explore the causes of this phenomenon, we conducted the study. In this study, our in vivo experiments found that immature rats exposed to hypoxia combined with propofol (HCWP) could develop cognitive impairment. We performed the RNA-seq analysis of its hippocampal tissues and found that autophagy and ferroptosis may play a role in our model. Next, we verified the participation of the two modes of death by detecting the expression of autophagy-related indexes Sequestosome 1 (SQSTM1) and Beclin1, and ferroptosis-related indicators Fe2+, reactive oxygen species (ROS) and glutathione peroxidase 4 (GPX4). Meanwhile, we found that ferrostatin-1 (Fer-1), an inhibitor of ferroptosis, could improve cognitive impairment in immature rats caused by HCWP. In addition, we found that nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy, which acted as a key junction between autophagy and ferroptosis, was also involved. Finally, our in vitro experiments concluded that autophagy activation was an upstream factor in HCWP-induced hippocampus ferroptosis through the intervention of autophagy inhibitor 3-methyladenine (3-MA). Our study was expected to provide an attractive therapeutic target for cognitive impairment that occurred after HCWP exposures.

丙泊酚是一种临床常用的静脉注射全身麻醉药,广泛用于儿童麻醉诱导、维持和重症监护室(ICU)镇静。低氧血症是围术期常见的并发症。在临床工作中,我们发现接受异丙酚麻醉的低氧血症患儿术后会出现明显的认知改变。为了探索这一现象的原因,我们开展了这项研究。在这项研究中,我们的体内实验发现,未成熟大鼠暴露于缺氧联合丙泊酚(HCWP)会出现认知障碍。我们对其海马组织进行了RNA-seq分析,发现自噬和铁突变可能在我们的模型中发挥作用。接下来,我们通过检测自噬相关指标Sequestosome 1(SQSTM1)和Beclin1的表达,以及铁突变相关指标Fe2+、活性氧(ROS)和谷胱甘肽过氧化物酶4(GPX4)的表达,验证了这两种死亡模式的参与。同时,我们还发现,铁色素沉着抑制剂铁前列素-1(Fer-1)可改善高氯性水肿引起的未成熟大鼠认知障碍。此外,我们还发现核受体辅激活子 4(NCOA4)介导的铁蛋白吞噬也参与其中,而铁蛋白吞噬是自噬和铁变态反应之间的关键连接点。最后,我们的体外实验得出结论,通过自噬抑制剂 3-甲基腺嘌呤(3-MA)的干预,自噬激活是 HCWP 诱导的海马铁突变的上游因素。我们的研究有望为暴露于六氯环己烷后出现的认知障碍提供一个有吸引力的治疗靶点。
{"title":"Ferritinophagy-Mediated Hippocampus Ferroptosis is Involved in Cognitive Impairment in Immature Rats Induced by Hypoxia Combined with Propofol.","authors":"Ling Liu, Wen Gao, Shun Yang, Fei Yang, Shangyingying Li, Yaqiong Tian, Li Yang, Qianyu Deng, Zhengwei Gan, Shengfen Tu","doi":"10.1007/s11064-024-04128-6","DOIUrl":"10.1007/s11064-024-04128-6","url":null,"abstract":"<p><p>Propofol is a clinically common intravenous general anesthetic and is widely used for anesthesia induction, maintenance and intensive care unit (ICU) sedation in children. Hypoxemia is a common perioperative complication. In clinical work, we found that children with hypoxemia who received propofol anesthesia experienced significant postoperative cognitive changes. To explore the causes of this phenomenon, we conducted the study. In this study, our in vivo experiments found that immature rats exposed to hypoxia combined with propofol (HCWP) could develop cognitive impairment. We performed the RNA-seq analysis of its hippocampal tissues and found that autophagy and ferroptosis may play a role in our model. Next, we verified the participation of the two modes of death by detecting the expression of autophagy-related indexes Sequestosome 1 (SQSTM1) and Beclin1, and ferroptosis-related indicators Fe<sup>2+</sup>, reactive oxygen species (ROS) and glutathione peroxidase 4 (GPX4). Meanwhile, we found that ferrostatin-1 (Fer-1), an inhibitor of ferroptosis, could improve cognitive impairment in immature rats caused by HCWP. In addition, we found that nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy, which acted as a key junction between autophagy and ferroptosis, was also involved. Finally, our in vitro experiments concluded that autophagy activation was an upstream factor in HCWP-induced hippocampus ferroptosis through the intervention of autophagy inhibitor 3-methyladenine (3-MA). Our study was expected to provide an attractive therapeutic target for cognitive impairment that occurred after HCWP exposures.</p>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140183445","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
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