首页 > 最新文献

Neurochemistry international最新文献

英文 中文
Investigating the role of GABAergic interneurons in the antidepressant-like mechanism of agomelatine gaba能中间神经元在阿戈美拉汀抗抑郁机制中的作用
IF 4 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-09 DOI: 10.1016/j.neuint.2025.106101
Suhong Ye , Qiaolu Xu , Nashwa Amin , Ling Bao , Yang Yang , Irum Naz Abbasi , Marong Fang
Depression is a prevalent and debilitating mental disorder with substantial impacts on global health and socioeconomic costs. Despite various antidepressants targeting monoaminergic neurotransmission, a significant proportion of patients fail to achieve remission with existing treatments. Agomelatine (AGO), as a novel antidepressant, has shown promise in treating depression. However, the neural circuits and molecular mechanisms underlying its therapeutic effects remain largely unknown. This study aimed to investigate the role of GABAergic neural circuits on the antidepressant effects of AGO and elucidate the underlying cellular and molecular mechanisms. A chronic unpredictable mild stress (CUMS) mouse model was used to induce depressive-like behaviors. Genetic manipulation was employed to selectively ablate GABAergic neurons, and the effects of AGO treatment on behavioral performance and neuronal morphology were assessed. Additionally, the expression of synaptic and clock genes was analyzed to explore underlying molecular mechanisms. We found that AGO treatment significantly improved the behavioral performance of CUMS mice and rescued the structural integrity and quantity of central neurons. It regulated the protein expressions of VGAT, VGLUT1, and Gad65 in the brain tissues of CUMS mice. Notably, AGO altered the protein and gene expressions in GABAergic neural circuits across different brain regions. Morphological analysis revealed that AGO improved dendritic spine density and length in neurons in the selective ablation of GABAergic interneurons. The antidepressant effects of AGO involve the modulation of GABAergic neural circuits as a critical but non-exclusive target, alongside the restoration of GABAergic-glutamatergic balance, synaptic function, and clock gene expressions. These findings highlight AGO's potential in normalizing disrupted neuronal function in depression and offer insights into novel multi-target therapeutic strategies.
抑郁症是一种普遍存在的使人衰弱的精神障碍,对全球健康和社会经济成本产生重大影响。尽管有各种针对单胺能神经传递的抗抑郁药物,但很大一部分患者无法通过现有的治疗获得缓解。阿戈美拉汀(AGO)作为一种新型抗抑郁药,在治疗抑郁症方面显示出良好的前景。然而,其治疗效果背后的神经回路和分子机制在很大程度上仍然未知。本研究旨在探讨gaba能神经回路在AGO抗抑郁作用中的作用,并阐明其潜在的细胞和分子机制。采用慢性不可预测轻度应激(CUMS)小鼠模型诱导抑郁样行为。采用基因操作选择性消融gaba能神经元,评估AGO处理对行为表现和神经元形态的影响。此外,我们还分析了突触和时钟基因的表达,以探索潜在的分子机制。我们发现AGO治疗显著改善了CUMS小鼠的行为表现,恢复了中枢神经元的结构完整性和数量。调节CUMS小鼠脑组织中VGAT、VGLUT1、Gad65蛋白的表达。值得注意的是,AGO改变了gaba能神经回路中不同脑区的蛋白质和基因表达。形态学分析显示,在选择性消融gaba能中间神经元时,AGO可改善神经元的树突棘密度和长度。AGO的抗抑郁作用包括gaba能神经回路的调节,这是一个关键但非排他的目标,同时还包括gaba能-谷氨酸能平衡、突触功能和时钟基因表达的恢复。这些发现突出了AGO在使抑郁症中受损的神经元功能正常化方面的潜力,并为新的多靶点治疗策略提供了见解。
{"title":"Investigating the role of GABAergic interneurons in the antidepressant-like mechanism of agomelatine","authors":"Suhong Ye ,&nbsp;Qiaolu Xu ,&nbsp;Nashwa Amin ,&nbsp;Ling Bao ,&nbsp;Yang Yang ,&nbsp;Irum Naz Abbasi ,&nbsp;Marong Fang","doi":"10.1016/j.neuint.2025.106101","DOIUrl":"10.1016/j.neuint.2025.106101","url":null,"abstract":"<div><div>Depression is a prevalent and debilitating mental disorder with substantial impacts on global health and socioeconomic costs. Despite various antidepressants targeting monoaminergic neurotransmission, a significant proportion of patients fail to achieve remission with existing treatments. Agomelatine (AGO), as a novel antidepressant, has shown promise in treating depression. However, the neural circuits and molecular mechanisms underlying its therapeutic effects remain largely unknown. This study aimed to investigate the role of GABAergic neural circuits on the antidepressant effects of AGO and elucidate the underlying cellular and molecular mechanisms. A chronic unpredictable mild stress (CUMS) mouse model was used to induce depressive-like behaviors. Genetic manipulation was employed to selectively ablate GABAergic neurons, and the effects of AGO treatment on behavioral performance and neuronal morphology were assessed. Additionally, the expression of synaptic and clock genes was analyzed to explore underlying molecular mechanisms. We found that AGO treatment significantly improved the behavioral performance of CUMS mice and rescued the structural integrity and quantity of central neurons. It regulated the protein expressions of VGAT, VGLUT1, and Gad65 in the brain tissues of CUMS mice. Notably, AGO altered the protein and gene expressions in GABAergic neural circuits across different brain regions. Morphological analysis revealed that AGO improved dendritic spine density and length in neurons in the selective ablation of GABAergic interneurons. The antidepressant effects of AGO involve the modulation of GABAergic neural circuits as a critical but non-exclusive target, alongside the restoration of GABAergic-glutamatergic balance, synaptic function, and clock gene expressions. These findings highlight AGO's potential in normalizing disrupted neuronal function in depression and offer insights into novel multi-target therapeutic strategies.</div></div>","PeriodicalId":398,"journal":{"name":"Neurochemistry international","volume":"192 ","pages":"Article 106101"},"PeriodicalIF":4.0,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145734559","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
SPI1 suppresses YAP phosphorylation in vascular endothelial cells to prevent intracranial aneurysm progression SPI1抑制血管内皮细胞中YAP磷酸化以防止颅内动脉瘤进展
IF 4 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-09 DOI: 10.1016/j.neuint.2025.106100
Yikui Liu , Danyi Zheng , Lanlan Zhao , Bing Leng , Qingfang Sun , Liuguan Bian , Yongtao Zheng
This study aims to elucidate the mechanism by which YAP mediates the activity of vascular endothelial cells (ECs) in the biological process of intracranial aneurysms (IAs) and to provide a novel target for noninvasive IAs treatment. Single-nuclei RNA profiling of aneurysmal cells revealed that ECs within aneurysms exhibit an intermediate identity between arterial and venous/capillary cells, rather than clustering within the normal arterial population. These specific human ECs showed downregulated YAP expression under turbulent flow. Immunostaining of human IA tissues demonstrated reduced YAP and increased phosphorylated YAP (p-YAP) compared with superficial temporal artery walls. Using YAP-knockdown human brain microvascular endothelial cells (HBMECs), we observed elevated expression of senescence markers p21 and p16, accompanied by diminished proliferation and migration capacities. Furthermore, SPI1 (also known as PU.1) overexpression alleviated EC degeneration induced by turbulent flow through suppression of YAP phosphorylation. Collectively, our findings indicate that turbulent flow markedly reduces YAP expression while promoting its phosphorylation, thereby accelerating endothelial senescence. Importantly, SPI1 overexpression effectively mitigated turbulent-flow-induced endothelial senescence, suggesting that SPI1 may serve as a potential therapeutic target for preventing aneurysmal progression.
本研究旨在阐明YAP在颅内动脉瘤(IAs)生物学过程中介导血管内皮细胞(ECs)活性的机制,为IAs的无创治疗提供新的靶点。动脉瘤细胞的单核RNA谱显示,动脉瘤内的内皮细胞表现出动脉和静脉/毛细血管细胞之间的中间身份,而不是聚集在正常动脉群中。这些特异性的人类ECs在湍流中表现出下调的YAP表达。与颞浅动脉壁相比,人IA组织的免疫染色显示YAP减少,磷酸化YAP (p-YAP)增加。使用yap敲除的人脑微血管内皮细胞(HBMECs),我们观察到衰老标志物p21和p16的表达升高,同时增殖和迁移能力减弱。此外,SPI1(又称PU.1)过表达通过抑制YAP磷酸化减轻湍流诱导的EC变性。总的来说,我们的研究结果表明,湍流显著降低YAP的表达,同时促进其磷酸化,从而加速内皮细胞衰老。重要的是,SPI1过表达有效地减轻了湍流诱导的内皮衰老,这表明SPI1可能作为预防动脉瘤进展的潜在治疗靶点。
{"title":"SPI1 suppresses YAP phosphorylation in vascular endothelial cells to prevent intracranial aneurysm progression","authors":"Yikui Liu ,&nbsp;Danyi Zheng ,&nbsp;Lanlan Zhao ,&nbsp;Bing Leng ,&nbsp;Qingfang Sun ,&nbsp;Liuguan Bian ,&nbsp;Yongtao Zheng","doi":"10.1016/j.neuint.2025.106100","DOIUrl":"10.1016/j.neuint.2025.106100","url":null,"abstract":"<div><div>This study aims to elucidate the mechanism by which YAP mediates the activity of vascular endothelial cells (ECs) in the biological process of intracranial aneurysms (IAs) and to provide a novel target for noninvasive IAs treatment. Single-nuclei RNA profiling of aneurysmal cells revealed that ECs within aneurysms exhibit an intermediate identity between arterial and venous/capillary cells, rather than clustering within the normal arterial population. These specific human ECs showed downregulated YAP expression under turbulent flow. Immunostaining of human IA tissues demonstrated reduced YAP and increased phosphorylated YAP (p-YAP) compared with superficial temporal artery walls. Using YAP-knockdown human brain microvascular endothelial cells (HBMECs), we observed elevated expression of senescence markers p21 and p16, accompanied by diminished proliferation and migration capacities. Furthermore, SPI1 (also known as PU.1) overexpression alleviated EC degeneration induced by turbulent flow through suppression of YAP phosphorylation. Collectively, our findings indicate that turbulent flow markedly reduces YAP expression while promoting its phosphorylation, thereby accelerating endothelial senescence. Importantly, SPI1 overexpression effectively mitigated turbulent-flow-induced endothelial senescence, suggesting that SPI1 may serve as a potential therapeutic target for preventing aneurysmal progression.</div></div>","PeriodicalId":398,"journal":{"name":"Neurochemistry international","volume":"192 ","pages":"Article 106100"},"PeriodicalIF":4.0,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145734557","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
Transcription factor EB inhibition in response to genotoxic stress promotes apoptosis of glioblastoma cells 基因毒性应激对转录因子EB的抑制促进胶质母细胞瘤细胞凋亡。
IF 4 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-01 DOI: 10.1016/j.neuint.2025.106087
Seung-Ho Park , Minseok Jeong , Min-Jeong Kong, Kyung-Chul Choi
Glioblastoma multiforme (GBM), one of the most malignant brain cancers, responds poorly to chemotherapy and surgery. Transcription factor EB (TFEB) is markedly overexpressed in GBM cells. We investigated whether TFEB contributes to resistance to genotoxic stress and whether its inhibition promotes apoptosis of GBM cells and glioma stem cells (GSCs). Specifically, we examined whether combined treatment with etoposide and SAHA overcomes TFEB-mediated resistance and enhances apoptotic cell death. We examined the effects of etoposide, a topoisomerase II inhibitor, and SAHA, a histone deacetylase inhibitor, on TFEB expression and apoptotic signaling in human GBM cells and GSCs. To assess TFEB-mediated drug resistance, we measured cell viability, proliferation, and tumorsphere formation following single or combined treatments. Apoptotic signaling was analyzed by western blotting, MTT assays, and tumorsphere formation assays. Functional roles of TFEB were further investigated using overexpression and shRNA knockdown approaches. Treatment with etoposide induced apoptosis and reduced TFEB expression in GBM cells. Co-treatment with etoposide and SAHA synergistically increased cleaved PARP and phosphorylated H2AX levels, indicating enhanced apoptotic activity. In TFEB-overexpressing and knockdown GBM cells, apoptosis sensitivity varied according to TFEB expression levels. In GSCs, combination treatment significantly suppressed cell proliferation and tumorsphere formation, accompanied by reduced TFEB expression and oligomerization, and increased apoptosis. Our findings suggest that TFEB promotes the chemoresistance of GBM tumors and GSCs by suppressing apoptosis. Co-treatment with etoposide and SAHA inhibits TFEB activity and enhances apoptotic cell death, representing a promising therapeutic strategy for treating malignant brain tumors.
多形性胶质母细胞瘤(GBM)是最恶性的脑癌之一,对化疗和手术的反应很差。转录因子EB (TFEB)在GBM细胞中明显过表达。我们研究了TFEB是否有助于抵抗基因毒性应激,以及它的抑制是否促进GBM细胞和胶质瘤干细胞(GSCs)的凋亡。具体来说,我们研究了依托泊苷和SAHA联合治疗是否能克服tfeb介导的耐药并增强凋亡细胞死亡。我们研究了依托泊苷(一种拓扑异构酶II抑制剂)和SAHA(一种组蛋白去乙酰化酶抑制剂)对人GBM细胞和GSCs中TFEB表达和凋亡信号的影响。为了评估tfeb介导的耐药性,我们测量了单一或联合治疗后的细胞活力、增殖和肿瘤球形成。凋亡信号通过western blotting、MTT和肿瘤球形成实验进行分析。通过过表达和shRNA敲低的方法进一步研究了TFEB的功能作用。依托泊苷可诱导GBM细胞凋亡并降低TFEB表达。依托opo苷和SAHA共处理可协同增加裂解的PARP和磷酸化的H2AX水平,表明细胞凋亡活性增强。在TFEB过表达和低表达的GBM细胞中,凋亡敏感性随TFEB表达水平的变化而变化。在GSCs中,联合治疗显著抑制细胞增殖和肿瘤球形成,同时降低TFEB表达和寡聚化,增加凋亡。我们的研究结果表明,TFEB通过抑制细胞凋亡来促进GBM肿瘤和GSCs的化疗耐药。依托泊苷和SAHA联合治疗可抑制TFEB活性,增强凋亡细胞死亡,是治疗恶性脑肿瘤的一种有前景的治疗策略。
{"title":"Transcription factor EB inhibition in response to genotoxic stress promotes apoptosis of glioblastoma cells","authors":"Seung-Ho Park ,&nbsp;Minseok Jeong ,&nbsp;Min-Jeong Kong,&nbsp;Kyung-Chul Choi","doi":"10.1016/j.neuint.2025.106087","DOIUrl":"10.1016/j.neuint.2025.106087","url":null,"abstract":"<div><div>Glioblastoma multiforme (GBM), one of the most malignant brain cancers, responds poorly to chemotherapy and surgery. Transcription factor EB (TFEB) is markedly overexpressed in GBM cells. We investigated whether TFEB contributes to resistance to genotoxic stress and whether its inhibition promotes apoptosis of GBM cells and glioma stem cells (GSCs). Specifically, we examined whether combined treatment with etoposide and SAHA overcomes TFEB-mediated resistance and enhances apoptotic cell death. We examined the effects of etoposide, a topoisomerase II inhibitor, and SAHA, a histone deacetylase inhibitor, on TFEB expression and apoptotic signaling in human GBM cells and GSCs. To assess TFEB-mediated drug resistance, we measured cell viability, proliferation, and tumorsphere formation following single or combined treatments. Apoptotic signaling was analyzed by western blotting, MTT assays, and tumorsphere formation assays. Functional roles of TFEB were further investigated using overexpression and shRNA knockdown approaches. Treatment with etoposide induced apoptosis and reduced TFEB expression in GBM cells. Co-treatment with etoposide and SAHA synergistically increased cleaved PARP and phosphorylated H2AX levels, indicating enhanced apoptotic activity. In TFEB-overexpressing and knockdown GBM cells, apoptosis sensitivity varied according to TFEB expression levels. In GSCs, combination treatment significantly suppressed cell proliferation and tumorsphere formation, accompanied by reduced TFEB expression and oligomerization, and increased apoptosis. Our findings suggest that TFEB promotes the chemoresistance of GBM tumors and GSCs by suppressing apoptosis. Co-treatment with etoposide and SAHA inhibits TFEB activity and enhances apoptotic cell death, representing a promising therapeutic strategy for treating malignant brain tumors.</div></div>","PeriodicalId":398,"journal":{"name":"Neurochemistry international","volume":"191 ","pages":"Article 106087"},"PeriodicalIF":4.0,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145538119","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
A novel GSK3β inhibitor ameliorates tau aggregation and neuroinflammation in Alzheimer's disease 一种新的GSK3β抑制剂改善阿尔茨海默病的tau聚集和神经炎症。
IF 4 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-27 DOI: 10.1016/j.neuint.2025.106090
Xin-Yue Ning , Wen-Jie Liu , Li-Jun Zhou , Nan Wang , Xin-Zhu Li , Li-Meng Wu , Zhen-Shu Li , Ai-Zhu Yang , Si-Yuan Liu , Zong-He Xu , Fang-Hua Xun , Zi-Hua Xu , Qing-Chun Zhao
In Alzheimer's disease, increased GSK3β activity drives tau phosphorylation and directly or indirectly triggers neuroinflammation, neuronal damage and cognitive decline. We previously developed a novel GSK3β inhibitor, ZLWH-60, which demonstrated inhibitory activity with an IC50 of 11.5 nM. Here, we comprehensively evaluated the therapeutic potential of ZLWH-60 in suppressing tau pathology and neuroinflammation using multiple chemically-induced AD models. Our results demonstrate that ZLWH-60 could reduce the phosphorylation of multiple tau epitopes by inhibiting the activity of GSK3β, thereby ameliorating cognitive impairments in OKA-induced mouse model. In the LPS-induced mouse model, ZLWH-60 also reduced the secretion of inflammatory factors in the brain, exerting a neuroprotective effect. Our data highlight that ZLWH-60, as a GSK3β inhibitor, has a powerful ability to reduce the phosphorylation of tau protein and shift the balance of the inflammatory response from pro-inflammatory to anti-inflammatory, demonstrating the potential for therapeutic use of this drug to control AD.
在阿尔茨海默病中,GSK3β活性增加驱动tau磷酸化,直接或间接引发神经炎症、神经元损伤和认知能力下降。我们之前开发了一种新的GSK3β抑制剂ZLWH-60,显示出抑制活性,IC50为11.5 nM。在这里,我们通过多种化学诱导的AD模型,综合评估了ZLWH-60在抑制tau病理和神经炎症方面的治疗潜力。我们的研究结果表明,ZLWH-60可以通过抑制GSK3β的活性来降低多个tau表位的磷酸化,从而改善oka诱导的小鼠模型的认知障碍。在lps诱导小鼠模型中,ZLWH-60还能减少脑内炎症因子的分泌,发挥神经保护作用。我们的数据强调,ZLWH-60作为一种GSK3β抑制剂,具有降低tau蛋白磷酸化的强大能力,并将炎症反应的平衡从促炎转变为抗炎,这表明该药物具有控制AD的治疗潜力。
{"title":"A novel GSK3β inhibitor ameliorates tau aggregation and neuroinflammation in Alzheimer's disease","authors":"Xin-Yue Ning ,&nbsp;Wen-Jie Liu ,&nbsp;Li-Jun Zhou ,&nbsp;Nan Wang ,&nbsp;Xin-Zhu Li ,&nbsp;Li-Meng Wu ,&nbsp;Zhen-Shu Li ,&nbsp;Ai-Zhu Yang ,&nbsp;Si-Yuan Liu ,&nbsp;Zong-He Xu ,&nbsp;Fang-Hua Xun ,&nbsp;Zi-Hua Xu ,&nbsp;Qing-Chun Zhao","doi":"10.1016/j.neuint.2025.106090","DOIUrl":"10.1016/j.neuint.2025.106090","url":null,"abstract":"<div><div>In Alzheimer's disease, increased GSK3β activity drives tau phosphorylation and directly or indirectly triggers neuroinflammation, neuronal damage and cognitive decline. We previously developed a novel GSK3β inhibitor, ZLWH-60, which demonstrated inhibitory activity with an IC50 of 11.5 nM. Here, we comprehensively evaluated the therapeutic potential of ZLWH-60 in suppressing tau pathology and neuroinflammation using multiple chemically-induced AD models. Our results demonstrate that ZLWH-60 could reduce the phosphorylation of multiple tau epitopes by inhibiting the activity of GSK3β, thereby ameliorating cognitive impairments in OKA-induced mouse model. In the LPS-induced mouse model, ZLWH-60 also reduced the secretion of inflammatory factors in the brain, exerting a neuroprotective effect. Our data highlight that ZLWH-60, as a GSK3β inhibitor, has a powerful ability to reduce the phosphorylation of tau protein and shift the balance of the inflammatory response from pro-inflammatory to anti-inflammatory, demonstrating the potential for therapeutic use of this drug to control AD.</div></div>","PeriodicalId":398,"journal":{"name":"Neurochemistry international","volume":"192 ","pages":"Article 106090"},"PeriodicalIF":4.0,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145627325","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
Interplay and intervention of epigenetic dysregulation in traumatic brain injury pathology 外伤性脑损伤病理中表观遗传失调的相互作用与干预。
IF 4 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-27 DOI: 10.1016/j.neuint.2025.106094
Samuel G. Faasen, Vijay Arruri, Reid S. Alisch
Traumatic brain injury (TBI) occurs when an external mechanical force damages brain tissue, leading to temporary or lasting disturbances in brain structure and function. The heterogeneous molecular and phenotypic nature of TBI poses a major challenge to translating basic research discoveries into clinically effective interventions. Emerging evidence indicates that epigenetic and epitranscriptomic mechanisms, including histone modifications, DNA methylation, and RNA modifications, play pivotal roles in the molecular response to TBI. In this review, we discuss post-TBI epigenomic alterations with a focus on histone modifications, DNA methylation, and RNA modifications, and we highlight preclinical interventions that modulate these alterations and improve related post-TBI behavioral outcomes.
外伤性脑损伤(TBI)发生时,外部机械力损害脑组织,导致暂时或持久的大脑结构和功能紊乱。TBI的异质性分子和表型性质对将基础研究发现转化为临床有效的干预措施提出了重大挑战。新出现的证据表明,表观遗传和表转录组学机制,包括组蛋白修饰、DNA甲基化和RNA修饰,在TBI的分子反应中起着关键作用。在这篇综述中,我们讨论了脑外伤后的表观基因组改变,重点是组蛋白修饰、DNA甲基化和RNA修饰,并强调了调节这些改变和改善相关脑外伤后行为结果的临床前干预。
{"title":"Interplay and intervention of epigenetic dysregulation in traumatic brain injury pathology","authors":"Samuel G. Faasen,&nbsp;Vijay Arruri,&nbsp;Reid S. Alisch","doi":"10.1016/j.neuint.2025.106094","DOIUrl":"10.1016/j.neuint.2025.106094","url":null,"abstract":"<div><div>Traumatic brain injury (TBI) occurs when an external mechanical force damages brain tissue, leading to temporary or lasting disturbances in brain structure and function. The heterogeneous molecular and phenotypic nature of TBI poses a major challenge to translating basic research discoveries into clinically effective interventions. Emerging evidence indicates that epigenetic and epitranscriptomic mechanisms, including histone modifications, DNA methylation, and RNA modifications, play pivotal roles in the molecular response to TBI. In this review, we discuss post-TBI epigenomic alterations with a focus on histone modifications, DNA methylation, and RNA modifications, and we highlight preclinical interventions that modulate these alterations and improve related post-TBI behavioral outcomes.</div></div>","PeriodicalId":398,"journal":{"name":"Neurochemistry international","volume":"192 ","pages":"Article 106094"},"PeriodicalIF":4.0,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145627306","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
Essential oils by name and by nature: a review of their antioxidant and neuroprotective potential in Parkinson's disease 精油的名称和性质:综述其在帕金森病中的抗氧化和神经保护潜力
IF 4 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-24 DOI: 10.1016/j.neuint.2025.106092
Claudia Cannas , Stefano Zoroddu , Alessandra Tiziana Peana , Gaia Rocchitta , Luigi Bagella , Rossana Migheli
Oxidative stress (OS), resulting from an imbalance between reactive oxygen species (ROS) and endogenous antioxidants, plays a central role in the pathogenesis of neurodegenerative diseases, including Parkinson's disease (PD). The brain's high oxygen demand and abundance of polyunsaturated fatty acids make it particularly vulnerable to ROS-induced damage. Despite major advances in research, no disease-modifying treatments for PD are currently available. Consequently, increasing attention has been directed toward natural bioactive compounds with antioxidant and neuroprotective properties. Among these, essential oils (EOs), volatile plant-derived mixtures with documented antioxidant, anti-inflammatory, and neuroactive effects, are emerging as promising adjuvants for PD management. This review critically examines the antioxidant and neuroprotective effects of well-characterized EOs evaluated in both in vitro and in vivo models of neurodegeneration. Literature searches were conducted in PubMed and Scopus up to March 2025, identifying studies investigating EOs or their major components in PD-related experimental settings. Evidence indicates that essential oils derived from the Citrus and Rosa genus, and the Lamiaceae family, can reduce intracellular ROS accumulation, inhibit lipid peroxidation, enhance endogenous antioxidant enzyme activity, and modulate both apoptotic and inflammatory pathways. These multitarget actions are often attributed to synergistic interactions among EO constituents, such as limonene, linalool, thymol, and carvacrol. Owing to their low toxicity and ability to cross the blood–brain barrier, EOs represent promising natural candidates for the development of complementary therapeutic strategies in PD. Further mechanistic and translational studies are warranted to substantiate their clinical potential.
氧化应激(OS)是由活性氧(ROS)和内源性抗氧化剂之间的失衡引起的,在包括帕金森病(PD)在内的神经退行性疾病的发病机制中起着核心作用。大脑的高需氧量和丰富的多不饱和脂肪酸使它特别容易受到ros引起的损伤。尽管研究取得了重大进展,但目前尚无PD的疾病改善治疗方法。因此,人们越来越关注具有抗氧化和神经保护特性的天然生物活性化合物。其中,精油(EOs),挥发性植物来源的混合物,具有抗氧化,抗炎和神经活性的作用,正在成为PD治疗的有前途的佐剂。本文在体外和体内神经退行性变模型中对特征明确的EOs的抗氧化和神经保护作用进行了严格的研究。截至2025年3月,在PubMed和Scopus中进行了文献检索,确定了在pd相关实验环境中调查EOs或其主要成分的研究。有证据表明,柑橘属和蔷薇属以及Lamiaceae家族的精油可以减少细胞内ROS积累,抑制脂质过氧化,增强内源性抗氧化酶活性,调节细胞凋亡和炎症途径。这些多靶点作用通常归因于EO成分之间的协同相互作用,如柠檬烯、芳樟醇、百里香酚和香芹酚。由于其低毒性和穿越血脑屏障的能力,EOs代表了PD补充治疗策略发展的有希望的天然候选者。进一步的机制和转化研究是必要的,以证实其临床潜力。
{"title":"Essential oils by name and by nature: a review of their antioxidant and neuroprotective potential in Parkinson's disease","authors":"Claudia Cannas ,&nbsp;Stefano Zoroddu ,&nbsp;Alessandra Tiziana Peana ,&nbsp;Gaia Rocchitta ,&nbsp;Luigi Bagella ,&nbsp;Rossana Migheli","doi":"10.1016/j.neuint.2025.106092","DOIUrl":"10.1016/j.neuint.2025.106092","url":null,"abstract":"<div><div>Oxidative stress (OS), resulting from an imbalance between reactive oxygen species (ROS) and endogenous antioxidants, plays a central role in the pathogenesis of neurodegenerative diseases, including Parkinson's disease (PD). The brain's high oxygen demand and abundance of polyunsaturated fatty acids make it particularly vulnerable to ROS-induced damage. Despite major advances in research, no disease-modifying treatments for PD are currently available. Consequently, increasing attention has been directed toward natural bioactive compounds with antioxidant and neuroprotective properties. Among these, essential oils (EOs), volatile plant-derived mixtures with documented antioxidant, anti-inflammatory, and neuroactive effects, are emerging as promising adjuvants for PD management. This review critically examines the antioxidant and neuroprotective effects of well-characterized EOs evaluated in both <em>in vitro</em> and <em>in vivo</em> models of neurodegeneration. Literature searches were conducted in PubMed and Scopus up to March 2025, identifying studies investigating EOs or their major components in PD-related experimental settings. Evidence indicates that essential oils derived from the <em>Citrus</em> and <em>Rosa</em> genus, and the <em>Lamiaceae</em> family, can reduce intracellular ROS accumulation, inhibit lipid peroxidation, enhance endogenous antioxidant enzyme activity, and modulate both apoptotic and inflammatory pathways. These multitarget actions are often attributed to synergistic interactions among EO constituents, such as limonene, linalool, thymol, and carvacrol. Owing to their low toxicity and ability to cross the blood–brain barrier, EOs represent promising natural candidates for the development of complementary therapeutic strategies in PD. Further mechanistic and translational studies are warranted to substantiate their clinical potential.</div></div>","PeriodicalId":398,"journal":{"name":"Neurochemistry international","volume":"192 ","pages":"Article 106092"},"PeriodicalIF":4.0,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145617187","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
ERBB4 colocalizes with phosphorylated tau aggregates in multiple tauopathies 在多种tau病变中,ERBB4与磷酸化的tau聚集物共定位
IF 4 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-24 DOI: 10.1016/j.neuint.2025.106093
Chihiro Matsumoto , Tomohiro Kabuta , Terunori Sano , Shigeo Murayama , Yuko Saito , Yuji Takahashi
The neuregulin-ERBB4 pathway is essential for maintaining cellular function. Upon stimulation by its ligand, neuregulin, ERBB4—a receptor tyrosine kinase—triggers multiple cellular responses, including proliferation, apoptosis, differentiation, and neuromuscular junction formation. Previous research has implicated dysregulated ERBB4 signaling in the pathophysiology of several neurodegenerative disorders, such as Alzheimer's disease, progressive supranuclear palsy, amyotrophic lateral sclerosis, and Parkinson's disease. In this study, we examined ERBB4 expression in diseases characterized by phosphorylated tau (MAPT) pathology. We found that ERBB4 colocalized with neuronal and glial phosphorylated tau-positive inclusions in multiple tauopathies, including Pick's disease, Alzheimer's disease, corticobasal degeneration, progressive supranuclear palsy, argyrophilic grain disease, and frontotemporal lobar degeneration with MAPT mutation. Conversely, ERBB4 did not colocalize with α-synuclein aggregates in α-synucleinopathies (Parkinson's disease and multiple system atrophy) or with neuronal intranuclear inclusions in triplet repeat disorders (Huntington's disease and dentatorubral-pallidoluysian atrophy). A co-immunoprecipitation assay indicated that ERBB4 can interact with tau intracellularly. Notably, in corticobasal degeneration, we observed ectopic ERBB4 expression in astrocytes lacking apparent phosphorylated tau aggregates. These findings suggest a potential role for ERBB4 in the pathophysiology of tau-related neurodegenerative diseases.
神经调节- erbb4通路对维持细胞功能至关重要。erbb4受体酪氨酸激酶在其配体神经调节蛋白的刺激下,引发多种细胞反应,包括增殖、凋亡、分化和神经肌肉连接形成。先前的研究表明ERBB4信号失调在一些神经退行性疾病的病理生理中,如阿尔茨海默病、进行性核上性麻痹、肌萎缩侧索硬化症和帕金森病。在这项研究中,我们检测了ERBB4在以磷酸化tau (MAPT)病理为特征的疾病中的表达。我们发现ERBB4在多种tau病变中与神经元和胶质磷酸化的tau阳性包涵体共定位,包括匹克病、阿尔茨海默病、皮质基底变性、进行性核上性麻痹、嗜阿糖颗粒病和伴有MAPT突变的额颞叶变性。相反,在α-突触核蛋白病(帕金森病和多系统萎缩)中,ERBB4不与α-突触核蛋白聚集体共定位,也不与三重重复疾病(亨廷顿病和齿状小网膜-白球萎缩)中的神经元核内包涵体共定位。免疫共沉淀法表明ERBB4可与细胞内tau蛋白相互作用。值得注意的是,在皮质基底变性中,我们在缺乏明显磷酸化tau聚集物的星形胶质细胞中观察到异位ERBB4表达。这些发现提示ERBB4在tau相关神经退行性疾病的病理生理中具有潜在的作用。
{"title":"ERBB4 colocalizes with phosphorylated tau aggregates in multiple tauopathies","authors":"Chihiro Matsumoto ,&nbsp;Tomohiro Kabuta ,&nbsp;Terunori Sano ,&nbsp;Shigeo Murayama ,&nbsp;Yuko Saito ,&nbsp;Yuji Takahashi","doi":"10.1016/j.neuint.2025.106093","DOIUrl":"10.1016/j.neuint.2025.106093","url":null,"abstract":"<div><div>The neuregulin-ERBB4 pathway is essential for maintaining cellular function. Upon stimulation by its ligand, neuregulin, ERBB4—a receptor tyrosine kinase—triggers multiple cellular responses, including proliferation, apoptosis, differentiation, and neuromuscular junction formation. Previous research has implicated dysregulated ERBB4 signaling in the pathophysiology of several neurodegenerative disorders, such as Alzheimer's disease, progressive supranuclear palsy, amyotrophic lateral sclerosis, and Parkinson's disease. In this study, we examined ERBB4 expression in diseases characterized by phosphorylated tau (MAPT) pathology. We found that ERBB4 colocalized with neuronal and glial phosphorylated tau-positive inclusions in multiple tauopathies, including Pick's disease, Alzheimer's disease, corticobasal degeneration, progressive supranuclear palsy, argyrophilic grain disease, and frontotemporal lobar degeneration with <em>MAPT</em> mutation. Conversely, ERBB4 did not colocalize with α-synuclein aggregates in α-synucleinopathies (Parkinson's disease and multiple system atrophy) or with neuronal intranuclear inclusions in triplet repeat disorders (Huntington's disease and dentatorubral-pallidoluysian atrophy). A co-immunoprecipitation assay indicated that ERBB4 can interact with tau intracellularly. Notably, in corticobasal degeneration, we observed ectopic ERBB4 expression in astrocytes lacking apparent phosphorylated tau aggregates. These findings suggest a potential role for ERBB4 in the pathophysiology of tau-related neurodegenerative diseases.</div></div>","PeriodicalId":398,"journal":{"name":"Neurochemistry international","volume":"192 ","pages":"Article 106093"},"PeriodicalIF":4.0,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145617186","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
GDNF-AS1 mediated LHX2/METTL3/NCOA4 axis inhibits glioma progression via induction of ferroptosis GDNF-AS1介导的LHX2/METTL3/NCOA4轴通过诱导铁下垂抑制胶质瘤进展。
IF 4 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-21 DOI: 10.1016/j.neuint.2025.106085
Yangbo zhou, Zhongyue Liu, Wenjia Ma
Glioma, particularly glioblastoma (GBM), represents the most aggressive primary brain tumor with limited treatment options and poor prognosis. Emerging evidence highlights ferroptosis induction as a promising therapeutic strategy, while long non-coding RNAs (lncRNAs) have gained attention as potential biomarkers and regulators in glioma pathogenesis. This study aimed to investigate the molecular mechanism of lncRNA Glial Cell Line-Derived Neurotrophic Factor Antisense RNA 1 (GDNF-AS1) in glioma cell ferroptosis through the LIM Homeobox 2 (LHX2)/Methyltransferase-Like 3 (METTL3)/Nuclear Receptor Coactivator 4 (NCOA4) pathway using Normal Human Astrocytes (NHA) and glioma cell lines (U87MG, T98G, U251, and A172), along with intracranial and subcutaneous xenotransplantation models established in BALB/c nude mice. Functional experiments demonstrated that GDNF-AS1, LHX2, and NCOA4 were downregulated while METTL3 was upregulated in glioma cells. GDNF-AS1 overexpression promoted mitochondrial damage and oxidative stress by enhancing ferroptosis, ultimately impairing glioma cell biological functions. METTL3 silencing augmented GDNF-AS1's effects, further exacerbating ferroptosis and oxidative stress while inhibiting glioma progression. Mechanistically, GDNF-AS1 recruited transcription factor LHX2 to upregulate its enrichment at the METTL3 promoter, thereby suppressing METTL3 transcription, reducing N6-Methyladenosine (m6A) levels, promoting NCOA4 expression, and inducing ferroautophagy and ferroptosis in glioma cells. These findings demonstrate that GDNF-AS1 inhibits glioma development by activating ferroptosis through the LHX2/METTL3/NCOA4 axis.
胶质瘤,尤其是胶质母细胞瘤(GBM),是最具侵袭性的原发性脑肿瘤,治疗方案有限,预后差。新出现的证据表明,诱导铁下垂是一种很有前景的治疗策略,而长链非编码rna (lncRNAs)作为神经胶质瘤发病机制中的潜在生物标志物和调节因子已受到关注。本研究旨在利用正常人星状细胞(NHA)和胶质瘤细胞系(U87MG、T98G、U251和A172),以及建立BALB/c裸鼠颅内和皮下异种移植模型,通过LIM Homeobox 2 (LHX2)/甲基转移酶样3 (METTL3)/核受体共激活因子4 (NCOA4)通路,探讨lncRNA胶质细胞系衍生的神经营养因子反义RNA 1 (GDNF-AS1)在胶质瘤细胞铁凋亡中的分子机制。功能实验表明,胶质瘤细胞中GDNF-AS1、LHX2和NCOA4下调,而METTL3上调。GDNF-AS1过表达通过增强铁凋亡促进线粒体损伤和氧化应激,最终损害胶质瘤细胞的生物学功能。METTL3沉默增强GDNF-AS1的作用,进一步加剧铁下垂和氧化应激,同时抑制胶质瘤的进展。机制上,GDNF-AS1募集转录因子LHX2上调其在METTL3启动子的富集,从而抑制METTL3转录,降低n6 -甲基腺苷(m6A)水平,促进NCOA4表达,诱导胶质瘤细胞铁自噬和铁凋亡。这些发现表明GDNF-AS1通过LHX2/METTL3/NCOA4轴激活铁下垂来抑制胶质瘤的发展。
{"title":"GDNF-AS1 mediated LHX2/METTL3/NCOA4 axis inhibits glioma progression via induction of ferroptosis","authors":"Yangbo zhou,&nbsp;Zhongyue Liu,&nbsp;Wenjia Ma","doi":"10.1016/j.neuint.2025.106085","DOIUrl":"10.1016/j.neuint.2025.106085","url":null,"abstract":"<div><div>Glioma, particularly glioblastoma (GBM), represents the most aggressive primary brain tumor with limited treatment options and poor prognosis. Emerging evidence highlights ferroptosis induction as a promising therapeutic strategy, while long non-coding RNAs (lncRNAs) have gained attention as potential biomarkers and regulators in glioma pathogenesis. This study aimed to investigate the molecular mechanism of lncRNA Glial Cell Line-Derived Neurotrophic Factor Antisense RNA 1 (GDNF-AS1) in glioma cell ferroptosis through the LIM Homeobox 2 (LHX2)/Methyltransferase-Like 3 (METTL3)/Nuclear Receptor Coactivator 4 (NCOA4) pathway using Normal Human Astrocytes (NHA) and glioma cell lines (U87MG, T98G, U251, and A172), along with intracranial and subcutaneous xenotransplantation models established in BALB/c nude mice. Functional experiments demonstrated that GDNF-AS1, LHX2, and NCOA4 were downregulated while METTL3 was upregulated in glioma cells. GDNF-AS1 overexpression promoted mitochondrial damage and oxidative stress by enhancing ferroptosis, ultimately impairing glioma cell biological functions. METTL3 silencing augmented GDNF-AS1's effects, further exacerbating ferroptosis and oxidative stress while inhibiting glioma progression. Mechanistically, GDNF-AS1 recruited transcription factor LHX2 to upregulate its enrichment at the METTL3 promoter, thereby suppressing METTL3 transcription, reducing N6-Methyladenosine (m6A) levels, promoting NCOA4 expression, and inducing ferroautophagy and ferroptosis in glioma cells. These findings demonstrate that GDNF-AS1 inhibits glioma development by activating ferroptosis through the LHX2/METTL3/NCOA4 axis.</div></div>","PeriodicalId":398,"journal":{"name":"Neurochemistry international","volume":"192 ","pages":"Article 106085"},"PeriodicalIF":4.0,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145585640","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
Therapeutic potential of KATP channels in the attenuation of Parkinson's disease pathogenesis and progression – A review KATP通道在减缓帕金森病发病和进展中的治疗潜力综述
IF 4 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-21 DOI: 10.1016/j.neuint.2025.106091
Barbara Gundi , Hio Lam Ho , Xinyang Zhang , Amanda He , Danielle Xin , Ana Flávia F. Ferreira , Luiz Roberto Britto , Zhong-Ping Feng , Hong-Shuo Sun
Parkinson's disease (PD) is one of the most prevalent progressive neurodegenerative diseases today. However, existing treatments primarily focus on symptom management rather than attenuating disease progression and pathogenesis. ATP-sensitive potassium (KATP) ion channels play a significant role in motor control and coordination within the basal ganglia and have been implicated in the dopaminergic depletion mechanisms underlying PD. Recent studies have explored the potential of KATP channel inhibitors to slow PD pathogenesis and progression. Both pharmacological inhibition and genetic inactivation of these channels have been shown to reduce oxidative stress, dopamine (DA) depletion, and subsequent motor deficits. Contrastingly, alternative evidence suggests that KATP channel openers (KCOs) may elicit similar effects, highlighting the need for further exploration of KATP-mediated DA depletion mechanisms in PD. Future studies expanding our understanding of the mechanistic action of KATP in PD are essential to effectively leverage the channel's potential as a therapeutic target for combating PD pathology.
帕金森病(PD)是当今最常见的进行性神经退行性疾病之一。然而,现有的治疗主要侧重于症状管理,而不是减轻疾病的进展和发病机制。atp敏感钾离子通道在基底神经节内的运动控制和协调中起着重要作用,并与PD的多巴胺能耗竭机制有关。最近的研究已经探索了KATP通道抑制剂减缓PD发病和进展的潜力。这些通道的药理抑制和基因失活均可减少氧化应激、多巴胺(DA)消耗和随后的运动缺陷。相反,其他证据表明,KATP通道打开剂(KCOs)可能会产生类似的效果,这突出了进一步探索KATP介导的PD中DA消耗机制的必要性。未来的研究扩大了我们对KATP在PD中的机制作用的理解,对于有效利用该通道作为对抗PD病理的治疗靶点的潜力至关重要。
{"title":"Therapeutic potential of KATP channels in the attenuation of Parkinson's disease pathogenesis and progression – A review","authors":"Barbara Gundi ,&nbsp;Hio Lam Ho ,&nbsp;Xinyang Zhang ,&nbsp;Amanda He ,&nbsp;Danielle Xin ,&nbsp;Ana Flávia F. Ferreira ,&nbsp;Luiz Roberto Britto ,&nbsp;Zhong-Ping Feng ,&nbsp;Hong-Shuo Sun","doi":"10.1016/j.neuint.2025.106091","DOIUrl":"10.1016/j.neuint.2025.106091","url":null,"abstract":"<div><div>Parkinson's disease (PD) is one of the most prevalent progressive neurodegenerative diseases today. However, existing treatments primarily focus on symptom management rather than attenuating disease progression and pathogenesis. ATP-sensitive potassium (K<sub>ATP</sub>) ion channels play a significant role in motor control and coordination within the basal ganglia and have been implicated in the dopaminergic depletion mechanisms underlying PD. Recent studies have explored the potential of K<sub>ATP</sub> channel inhibitors to slow PD pathogenesis and progression. Both pharmacological inhibition and genetic inactivation of these channels have been shown to reduce oxidative stress, dopamine (DA) depletion, and subsequent motor deficits. Contrastingly, alternative evidence suggests that K<sub>ATP</sub> channel openers (KCOs) may elicit similar effects, highlighting the need for further exploration of K<sub>ATP</sub>-mediated DA depletion mechanisms in PD. Future studies expanding our understanding of the mechanistic action of K<sub>ATP</sub> in PD are essential to effectively leverage the channel's potential as a therapeutic target for combating PD pathology.</div></div>","PeriodicalId":398,"journal":{"name":"Neurochemistry international","volume":"192 ","pages":"Article 106091"},"PeriodicalIF":4.0,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145585674","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
Esketamine attenuates post-traumatic stress disorder via suppressing neuroinflammation and abnormal myelination 艾氯胺酮通过抑制神经炎症和异常髓鞘形成来减轻创伤后应激障碍。
IF 4 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-19 DOI: 10.1016/j.neuint.2025.106089
Zhaoliang Gu , Ruixue Song , Guoqiang Liu , Hao Yu , Liangyu Ju , Yu Su , Jianming Bi , Jianhao Qiu , Yuan Dong , Aijie Liu

Background

Post-traumatic stress disorder (PTSD) is a chronic psychological disorder that is induced by traumatic events. The pathophysiological mechanism of PTSD involves complex neurobiological processes. However, the underlying mechanism is not clear, leading to lack of effective therapeutic interventions.

Methods

Mice were exposed to the electric foot shocks using the contextual fear memory paradigm. A subanesthetic dose (30 mg/kg) of esketamine or saline was administered via intraperitoneal (i.p.) injection 1 h after the electric foot shocks. Fear retrieval was tested on day 1 and day 7 after fear conditioning. Anxiety-like and depressive-like behaviors were evaluated using the open field test and elevated plus maze on day 1 and day 2, respectively, after the foot-shocks. The medial prefrontal cortex (mPFC) was freshly collected 1 h after esketamine administration following the foot-shocks for RNA sequencing. Additionally, the mPFC were collected 4 days after fear conditioning and subjected to quantitative real-time PCR (qPCR) analysis and immunofluorescence staining.

Results

A single subanesthetic dose of esketamine significantly alleviated PTSD-like symptoms in mice induced by electric foot-shocks. RNA sequencing revealed the involvement of neuroinflammation and aberrant myelination in the pathogenesis of PTSD. Subsequently, we observed a significant increase in the number of ionized calcium binding adaptor molecule 1 (Iba1)-positive microglial cells and transcriptional upregulation of pro-inflammatory cytokines, such as interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α), in the mPFC of mice subjected electric foot shocks, indicating elevated neuroinflammation. Subanesthetic esketamine administration significantly attenuated this neuroinflammatory response. Furthermore, electric foot shocks caused significantly increased the expression of myelin basic protein (MBP), myelin-associated glycoprotein (MAG), oligodendrocyte transcription factor 2 (Olig2) and platelet-derived growth factor receptor-α (PDGFRα), suggesting increased myelination associated with PTSD. Esketamine treatment also rescued this abnormal myelination.

Conclusion

Our study demonstrates the contribution of neuroinflammation and abnormal myelination are closely related to the development of PTSD. Moreover, a subanesthetic dose of esketamine alleviated the PTSD-like symptoms in mice by suppressing foot-shock-induced increases in neuroinflammation and myelination. These results highlight the therapeutic potential of subanesthetic esketamine in mitigating PTSD.
背景:创伤后应激障碍(PTSD)是一种由创伤性事件诱发的慢性心理障碍。创伤后应激障碍的病理生理机制涉及复杂的神经生物学过程。然而,其潜在机制尚不清楚,导致缺乏有效的治疗干预措施。方法:采用情境恐惧记忆模式对小鼠进行足电刺激。足部电刺激1小时后腹腔注射亚麻醉剂量(30mg /kg)艾氯胺酮或生理盐水。恐惧条件反射后第1天和第7天分别进行恐惧恢复测试。在足震后的第1天和第2天,分别采用开放场测试和升高加迷宫的方法评估焦虑样行为和抑郁样行为。足部电击后给予艾氯胺酮1小时后新鲜采集内侧前额叶皮层(mPFC)进行RNA测序。另外,在恐惧条件反射4天后收集mPFC,进行实时荧光定量PCR (qPCR)分析和免疫荧光染色。结果:单次亚麻醉剂量艾氯胺酮可显著减轻足电小鼠ptsd样症状。RNA测序显示神经炎症和异常髓鞘形成参与PTSD的发病机制。随后,我们观察到电化钙结合接头分子1 (Iba1)阳性的小胶质细胞数量显著增加,并且在电击足的小鼠mPFC中,促炎细胞因子,如白细胞介素-6 (IL-6)和肿瘤坏死因子α (TNF-α)的转录上调,表明神经炎症升高。亚麻醉艾氯胺酮显著减轻了这种神经炎症反应。此外,电足刺激引起髓鞘碱性蛋白(MBP)、髓鞘相关糖蛋白(MAG)、少突胶质细胞转录因子2 (Olig2)和血小板衍生生长因子受体-α (PDGFRα)的表达显著升高,提示髓鞘形成增加与PTSD相关。艾氯胺酮治疗也挽救了这种异常髓鞘形成。结论:神经炎症和髓鞘形成异常与PTSD的发生发展密切相关。此外,亚麻醉剂量的艾氯胺酮通过抑制足冲击引起的神经炎症和髓鞘形成的增加,减轻了小鼠ptsd样症状。这些结果突出了亚麻醉艾氯胺酮在缓解创伤后应激障碍方面的治疗潜力。
{"title":"Esketamine attenuates post-traumatic stress disorder via suppressing neuroinflammation and abnormal myelination","authors":"Zhaoliang Gu ,&nbsp;Ruixue Song ,&nbsp;Guoqiang Liu ,&nbsp;Hao Yu ,&nbsp;Liangyu Ju ,&nbsp;Yu Su ,&nbsp;Jianming Bi ,&nbsp;Jianhao Qiu ,&nbsp;Yuan Dong ,&nbsp;Aijie Liu","doi":"10.1016/j.neuint.2025.106089","DOIUrl":"10.1016/j.neuint.2025.106089","url":null,"abstract":"<div><h3>Background</h3><div>Post-traumatic stress disorder (PTSD) is a chronic psychological disorder that is induced by traumatic events. The pathophysiological mechanism of PTSD involves complex neurobiological processes. However, the underlying mechanism is not clear, leading to lack of effective therapeutic interventions.</div></div><div><h3>Methods</h3><div>Mice were exposed to the electric foot shocks using the contextual fear memory paradigm. A subanesthetic dose (30 mg/kg) of esketamine or saline was administered via intraperitoneal (i.p.) injection 1 h after the electric foot shocks. Fear retrieval was tested on day 1 and day 7 after fear conditioning. Anxiety-like and depressive-like behaviors were evaluated using the open field test and elevated plus maze on day 1 and day 2, respectively, after the foot-shocks. The medial prefrontal cortex (mPFC) was freshly collected 1 h after esketamine administration following the foot-shocks for RNA sequencing. Additionally, the mPFC were collected 4 days after fear conditioning and subjected to quantitative real-time PCR (qPCR) analysis and immunofluorescence staining.</div></div><div><h3>Results</h3><div>A single subanesthetic dose of esketamine significantly alleviated PTSD-like symptoms in mice induced by electric foot-shocks. RNA sequencing revealed the involvement of neuroinflammation and aberrant myelination in the pathogenesis of PTSD. Subsequently, we observed a significant increase in the number of ionized calcium binding adaptor molecule 1 (Iba1)-positive microglial cells and transcriptional upregulation of pro-inflammatory cytokines, such as interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α), in the mPFC of mice subjected electric foot shocks, indicating elevated neuroinflammation. Subanesthetic esketamine administration significantly attenuated this neuroinflammatory response. Furthermore, electric foot shocks caused significantly increased the expression of myelin basic protein (MBP), myelin-associated glycoprotein (MAG), oligodendrocyte transcription factor 2 (Olig2) and platelet-derived growth factor receptor-α (PDGFRα), suggesting increased myelination associated with PTSD. Esketamine treatment also rescued this abnormal myelination.</div></div><div><h3>Conclusion</h3><div>Our study demonstrates the contribution of neuroinflammation and abnormal myelination are closely related to the development of PTSD. Moreover, a subanesthetic dose of esketamine alleviated the PTSD-like symptoms in mice by suppressing foot-shock-induced increases in neuroinflammation and myelination. These results highlight the therapeutic potential of subanesthetic esketamine in mitigating PTSD.</div></div>","PeriodicalId":398,"journal":{"name":"Neurochemistry international","volume":"191 ","pages":"Article 106089"},"PeriodicalIF":4.0,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145562291","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
Neurochemistry international
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:604180095
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