首页 > 最新文献

Experimental Neurology最新文献

英文 中文
Daily intranasal resveratrol-conjugated gold nanoparticles administration promotes neuroprotection and improves neurological outcome in the R6/2 mouse model of Huntington's disease 每日鼻内白藜芦醇结合金纳米颗粒可促进亨廷顿病R6/2小鼠模型的神经保护并改善神经预后
IF 4.2 2区 医学 Q1 NEUROSCIENCES Pub Date : 2026-01-07 DOI: 10.1016/j.expneurol.2026.115639
Emanuela Paldino , Emiliano Montalesi , Marco Fiocchetti , Flavia Dioguardi , Iole Venditti , Elena Olivieri , Maria Marino , Francesca R. Fusco
Pan-apoptosis and involvement of the inflammatory process are the hallmarks of Huntington's disease (HD). Inflammation currently represents one of the potential therapeutic targets for slowing and fighting the pathological phenotype of HD. The immunomodulatory properties of natural compounds, such as resveratrol, have been demonstrated in various disease models and human clinical trials. In the present study, we evaluated the neuroprotective and anti-inflammatory effects of the daily intranasal administration of resveratrol-conjugated gold nanoparticles in awake R6/2 mice, the genetic animal model of HD. Transgenic mice were treated daily with resveratrol-conjugated gold nanoparticles (0.1 mg/kg/day) starting from 5 weeks of age corresponding to the prodromal stage of the disease. After sacrifice, histological and immunofluorescence studies were performed. We found that resveratrol treated R6/2 mice survived longer and displayed a significant partial recovery of motor performance compared with R6/2 mice that received the nanoparticles with vehicle. Primary outcome measures such as striatal atrophy, neuronal intranuclear inclusions, and modulation of microglial reaction revealed a neuroprotective effect of resveratrol conjugated gold nanoparticles. Resveratrol provided a significant increase of neuroglobin, a neuroprotective globin, along with activated CREB and BDNF in the mice medium spiny neurons, accompanied by a down modulation of neuroinflammation, which, combined, might explain the beneficial effects observed in this model. Our findings showed that nanoparticles loaded with a specific compound which acts on the mutated protein intranuclear inclusions and inflammatory components may represent a valid therapeutic strategy in slowing down the symptoms of HD neurodegeneration.
泛细胞凋亡和炎症过程的参与是亨廷顿舞蹈病(HD)的标志。炎症目前是减缓和对抗HD病理表型的潜在治疗靶点之一。天然化合物(如白藜芦醇)的免疫调节特性已在各种疾病模型和人体临床试验中得到证实。在本研究中,我们在清醒的HD遗传动物模型R6/2小鼠中评估了每日鼻内给药白藜芦醇结合金纳米颗粒的神经保护和抗炎作用。从与疾病前驱期相对应的5周龄开始,每天用白藜芦醇偶联金纳米颗粒(0.1 mg/kg/天)治疗转基因小鼠。牺牲后,进行组织学和免疫荧光研究。我们发现白藜芦醇处理的R6/2小鼠存活时间更长,运动能力明显部分恢复。纹状体萎缩、神经元核内包涵体和小胶质细胞反应的调节等主要结局指标显示白藜芦醇共轭金纳米颗粒具有神经保护作用。白藜芦醇显著增加了小鼠中棘神经元中的神经球蛋白(一种神经保护性球蛋白),同时激活了CREB和BDNF,并伴有神经炎症的下调,这可能解释了该模型中观察到的有益效果。我们的研究结果表明,携带特定化合物的纳米颗粒可作用于突变蛋白核内包涵体和炎症成分,可能是减缓HD神经退行性疾病症状的有效治疗策略。
{"title":"Daily intranasal resveratrol-conjugated gold nanoparticles administration promotes neuroprotection and improves neurological outcome in the R6/2 mouse model of Huntington's disease","authors":"Emanuela Paldino ,&nbsp;Emiliano Montalesi ,&nbsp;Marco Fiocchetti ,&nbsp;Flavia Dioguardi ,&nbsp;Iole Venditti ,&nbsp;Elena Olivieri ,&nbsp;Maria Marino ,&nbsp;Francesca R. Fusco","doi":"10.1016/j.expneurol.2026.115639","DOIUrl":"10.1016/j.expneurol.2026.115639","url":null,"abstract":"<div><div>Pan-apoptosis and involvement of the inflammatory process are the hallmarks of Huntington's disease (HD). Inflammation currently represents one of the potential therapeutic targets for slowing and fighting the pathological phenotype of HD. The immunomodulatory properties of natural compounds, such as resveratrol, have been demonstrated in various disease models and human clinical trials. In the present study, we evaluated the neuroprotective and anti-inflammatory effects of the daily intranasal administration of resveratrol-conjugated gold nanoparticles in awake R6/2 mice, the genetic animal model of HD. Transgenic mice were treated daily with resveratrol-conjugated gold nanoparticles (0.1 mg/kg/day) starting from 5 weeks of age corresponding to the prodromal stage of the disease. After sacrifice, histological and immunofluorescence studies were performed. We found that resveratrol treated R6/2 mice survived longer and displayed a significant partial recovery of motor performance compared with R6/2 mice that received the nanoparticles with vehicle. Primary outcome measures such as striatal atrophy, neuronal intranuclear inclusions, and modulation of microglial reaction revealed a neuroprotective effect of resveratrol conjugated gold nanoparticles. Resveratrol provided a significant increase of neuroglobin, a neuroprotective globin, along with activated CREB and BDNF in the mice medium spiny neurons, accompanied by a down modulation of neuroinflammation, which, combined, might explain the beneficial effects observed in this model. Our findings showed that nanoparticles loaded with a specific compound which acts on the mutated protein intranuclear inclusions and inflammatory components may represent a valid therapeutic strategy in slowing down the symptoms of HD neurodegeneration.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":"398 ","pages":"Article 115639"},"PeriodicalIF":4.2,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145922537","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
NFKBIZ mediates neuroprotection and maintains blood-brain barrier integrity in cerebral ischemia/reperfusion via STAT3-regulated Nrf2/ARE signaling NFKBIZ通过stat3调控的Nrf2/ARE信号介导脑缺血/再灌注时的神经保护和维持血脑屏障完整性。
IF 4.2 2区 医学 Q1 NEUROSCIENCES Pub Date : 2026-01-07 DOI: 10.1016/j.expneurol.2026.115638
Cheng Huang , Xiaocong Mo , Ying Liu , Di Hu , Cun Li , Yuan Zhao , Shuxin Wang , Jinjun Xia , Xiaoyan Chen , Wei Sun , Rui Xu
Ischemic stroke (IS) is a leading cause of adult disability and mortality worldwide. It is characterized by a complex series of cellular and molecular events that lead to neuronal injury and disruption of the blood-brain barrier (BBB). The BBB plays a crucial role in maintaining brain homeostasis, and its disruption during IS exacerbates secondary brain injury. However, the molecular mechanisms that preserve BBB integrity and provide neuroprotection remain poorly understood. In this study, we used mRNA sequencing to identify genes differentially expressed in IS models. To investigate the role of NFKBIZ in BBB maintenance, we conducted additional in vitro and in vivo experiments. Mechanistic studies focused on the STAT3-mediated Nrf2/ARE signaling pathway, and metabolomic profiling was used to identify metabolites associated with NFKBIZ activity. Our findings indicate that NFKBIZ is essential for maintaining BBB integrity. Overexpression of NFKBIZ reduced ischemic injury and preserved BBB function, while its downregulation significantly worsened neurological deficits and BBB damage. The STAT3-Nrf2/ARE axis, a critical pathway for antioxidant defense, was activated by NFKBIZ, contributing to its protective effects. Furthermore, metabolomic analysis identified a set of metabolites linked to NFKBIZ function, providing insight into the underlying biological mechanisms. This study underscores the potential of NFKBIZ as a therapeutic target for neuroprotection and BBB preservation in IS, suggesting new avenues for developing treatments to improve outcomes in stroke patients.
缺血性脑卒中(IS)是世界范围内导致成人残疾和死亡的主要原因。它的特点是一系列复杂的细胞和分子事件,导致神经元损伤和血脑屏障(BBB)的破坏。血脑屏障在维持大脑稳态中起着至关重要的作用,在IS期间,血脑屏障的破坏加剧了继发性脑损伤。然而,保持血脑屏障完整性和提供神经保护的分子机制仍然知之甚少。在这项研究中,我们使用mRNA测序来鉴定IS模型中差异表达的基因。为了研究NFKBIZ在血脑屏障维持中的作用,我们进行了额外的体外和体内实验。机制研究侧重于stat3介导的Nrf2/ARE信号通路,代谢组学分析用于鉴定与NFKBIZ活性相关的代谢物。我们的研究结果表明NFKBIZ对于维持血脑屏障的完整性至关重要。NFKBIZ过表达可减轻缺血性损伤,保留血脑屏障功能,而其下调可显著加重神经功能缺损和血脑屏障损伤。STAT3-Nrf2/ARE轴是抗氧化防御的关键途径,NFKBIZ激活了STAT3-Nrf2/ARE轴,有助于其保护作用。此外,代谢组学分析确定了一组与NFKBIZ功能相关的代谢物,为潜在的生物学机制提供了见解。这项研究强调了NFKBIZ作为IS神经保护和血脑屏障保存的治疗靶点的潜力,为开发改善卒中患者预后的治疗方法提供了新的途径。
{"title":"NFKBIZ mediates neuroprotection and maintains blood-brain barrier integrity in cerebral ischemia/reperfusion via STAT3-regulated Nrf2/ARE signaling","authors":"Cheng Huang ,&nbsp;Xiaocong Mo ,&nbsp;Ying Liu ,&nbsp;Di Hu ,&nbsp;Cun Li ,&nbsp;Yuan Zhao ,&nbsp;Shuxin Wang ,&nbsp;Jinjun Xia ,&nbsp;Xiaoyan Chen ,&nbsp;Wei Sun ,&nbsp;Rui Xu","doi":"10.1016/j.expneurol.2026.115638","DOIUrl":"10.1016/j.expneurol.2026.115638","url":null,"abstract":"<div><div>Ischemic stroke (IS) is a leading cause of adult disability and mortality worldwide. It is characterized by a complex series of cellular and molecular events that lead to neuronal injury and disruption of the blood-brain barrier (BBB). The BBB plays a crucial role in maintaining brain homeostasis, and its disruption during IS exacerbates secondary brain injury. However, the molecular mechanisms that preserve BBB integrity and provide neuroprotection remain poorly understood. In this study, we used mRNA sequencing to identify genes differentially expressed in IS models. To investigate the role of NFKBIZ in BBB maintenance, we conducted additional in vitro and in vivo experiments. Mechanistic studies focused on the STAT3-mediated Nrf2/ARE signaling pathway, and metabolomic profiling was used to identify metabolites associated with NFKBIZ activity. Our findings indicate that NFKBIZ is essential for maintaining BBB integrity. Overexpression of NFKBIZ reduced ischemic injury and preserved BBB function, while its downregulation significantly worsened neurological deficits and BBB damage. The STAT3-Nrf2/ARE axis, a critical pathway for antioxidant defense, was activated by NFKBIZ, contributing to its protective effects. Furthermore, metabolomic analysis identified a set of metabolites linked to NFKBIZ function, providing insight into the underlying biological mechanisms. This study underscores the potential of NFKBIZ as a therapeutic target for neuroprotection and BBB preservation in IS, suggesting new avenues for developing treatments to improve outcomes in stroke patients.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":"398 ","pages":"Article 115638"},"PeriodicalIF":4.2,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145943025","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Astrocytic TPK1 mitigates amyloid pathology via TFEB-mediated endocytosis 星形细胞TPK1通过tfeb介导的内吞作用减轻淀粉样蛋白病理。
IF 4.2 2区 医学 Q1 NEUROSCIENCES Pub Date : 2026-01-06 DOI: 10.1016/j.expneurol.2026.115640
Shu-Zhen Zhang , Yuan Ma , Yu Ding , Yan-Qing Yin , Bing-Wei Wang , Gang Hu , Jia-Wei Zhou
Alzheimer's disease (AD), the leading cause of dementia, is characterized by amyloid-beta (Aβ) plaques, neurofibrillary tangles, and progressive neurodegeneration. Deregulation of glial cell activity plays an important role in the amyloid pathology. However, it is still unclear how changes in astrocytes contribute to Aβ deposition and clearance in AD. Here, we showed that deficiency of astrocytic thiamine pyrophosphokinase 1 (Tpk1), exacerbated Aβ burden leading to exacerbated spatial memory deficits in a mouse model of AD. While selective overexpression of Tpk1 in astrocytes ameliorated cognitive decline and significantly reduced hippocampal and cortical Aβ plaque burden. Enhanced Tpk1 expression augmented astrocyte endocytic capacity. Mechanistically, Tpk1-promoted endocytic activity depended on the activation of transcription factor EB (TFEB)-mediated pathways. Collectively, our findings demonstrate that astrocytic TPK1 mitigates cognitive impairment in 5xFAD mice by upregulating TFEB expression, thereby enhancing astrocyte-mediated engulfment and degradation of neurotoxic aggregates, including Aβ. This study suggests that astrocytic TPK1/TFEB pathway is a promising target for developing disease-modifying AD therapies.
阿尔茨海默病(AD)是痴呆症的主要原因,其特征是β淀粉样蛋白(Aβ)斑块、神经原纤维缠结和进行性神经变性。神经胶质细胞活性的失调在淀粉样蛋白病理中起着重要作用。然而,目前尚不清楚星形胶质细胞的变化如何促进AD中Aβ的沉积和清除。在本研究中,我们发现星形细胞硫胺素焦磷酸激酶1 (Tpk1)的缺乏加重了阿尔茨海默病小鼠模型中的a β负荷,从而加重了空间记忆缺陷。而在星形胶质细胞中选择性过表达Tpk1可改善认知能力下降,并显著减少海马和皮质β斑块负担。Tpk1表达的增强增强了星形胶质细胞的内吞能力。在机制上,tpk1促进的内吞活性依赖于转录因子EB (TFEB)介导途径的激活。总的来说,我们的研究结果表明,星形胶质细胞TPK1通过上调TFEB表达来减轻5xFAD小鼠的认知障碍,从而增强星形胶质细胞介导的神经毒性聚集体(包括Aβ)的吞噬和降解。这项研究表明星形细胞TPK1/TFEB通路是开发疾病修饰性AD治疗的一个有希望的靶点。
{"title":"Astrocytic TPK1 mitigates amyloid pathology via TFEB-mediated endocytosis","authors":"Shu-Zhen Zhang ,&nbsp;Yuan Ma ,&nbsp;Yu Ding ,&nbsp;Yan-Qing Yin ,&nbsp;Bing-Wei Wang ,&nbsp;Gang Hu ,&nbsp;Jia-Wei Zhou","doi":"10.1016/j.expneurol.2026.115640","DOIUrl":"10.1016/j.expneurol.2026.115640","url":null,"abstract":"<div><div>Alzheimer's disease (AD), the leading cause of dementia, is characterized by amyloid-beta (Aβ) plaques, neurofibrillary tangles, and progressive neurodegeneration. Deregulation of glial cell activity plays an important role in the amyloid pathology. However, it is still unclear how changes in astrocytes contribute to Aβ deposition and clearance in AD. Here, we showed that deficiency of astrocytic thiamine pyrophosphokinase 1 (<em>Tpk1</em>), exacerbated Aβ burden leading to exacerbated spatial memory deficits in a mouse model of AD. While selective overexpression of <em>Tpk1</em> in astrocytes ameliorated cognitive decline and significantly reduced hippocampal and cortical Aβ plaque burden. Enhanced <em>Tpk1</em> expression augmented astrocyte endocytic capacity. Mechanistically, <em>Tpk1</em>-promoted endocytic activity depended on the activation of transcription factor EB (TFEB)-mediated pathways. Collectively, our findings demonstrate that astrocytic TPK1 mitigates cognitive impairment in 5xFAD mice by upregulating TFEB expression, thereby enhancing astrocyte-mediated engulfment and degradation of neurotoxic aggregates, including Aβ. This study suggests that astrocytic TPK1/TFEB pathway is a promising target for developing disease-modifying AD therapies.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":"398 ","pages":"Article 115640"},"PeriodicalIF":4.2,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145931587","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Sleep deprivation attenuates acute ischemic stroke-induced hippocampal neuronal injury via the IL-38/NF-κB axis 睡眠剥夺通过IL-38/NF-κB轴减弱急性缺血性卒中诱导的海马神经元损伤。
IF 4.2 2区 医学 Q1 NEUROSCIENCES Pub Date : 2026-01-02 DOI: 10.1016/j.expneurol.2025.115635
Anlin Yue , Yongfei Liu , Zhongting Wang , Mian Zhang , Yuan Qin , Zhaoyan Zhao , Jingxiang Wang , Shuxuan He , Jiangjing Li , Xude Sun , Li Sun

Background

Although epidemiological studies have identified sleep disorders as a risk factor for stroke, animal studies have yielded conflicting results regarding the impact of sleep deprivation (SD). This study aimed to investigate whether SD preconditioning can alleviate early neurological impairment following acute ischemic stroke and to explore the potential role of the anti-inflammatory cytokine interleukin-38 (IL-38) in this process.

Methods

Male Sprague-Dawley rats underwent 10-day SD preconditioning using a modified multi-platform method before being subjected to transient middle cerebral artery occlusion (tMCAO). Neurological function was evaluated using the Longa scoring system, while cerebral infarct volume was quantified via 2,3,5-triphenyltetrazolium chloride (TTC) staining. Histopathological changes in brain tissues were assessed through hematoxylin-eosin (H&E) staining, Nissl staining, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining. The expression level of IL-38 and the status of inflammatory responses were analyzed using Western blotting, immunohistochemistry, and immunofluorescence. IL-38 knockout (IL-38 KO) rats were employed to verify the essential role of IL-38 in SD-induced neuroprotection.

Results

SD preconditioning significantly improved neurological function, reduced cerebral infarct volume, and decreased neuronal apoptosis in the CA1 region of the hippocampus. Notably, SD preconditioning markedly upregulated IL-38 expression, which was predominantly localized in neurons and exhibited a time-dependent increase pattern. In IL-38 KO rats, the neuroprotective effects of SD preconditioning were completely abolished, with no improvements observed in neurological scores, infarct size, or neuronal survival. Mechanistically, SD preconditioning reduced the levels of pro-inflammatory cytokines (tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6], and interleukin-1β [IL-1β]) and inhibited the phosphorylation of p65 in the nuclear factor-κB (NF-κB) signaling pathway, and these regulatory effects were largely dependent on IL-38.

Conclusions

SD preconditioning confers neuroprotection against acute ischemic stroke by upregulating IL-38 expression. IL-38 mediates this protection, at least in part, by attenuating neuroinflammation through suppression of the NF-κB signaling pathway. These findings identify IL-38 as a critical mediator of SD-induced ischemic tolerance, warranting further clinical investigation.
背景:虽然流行病学研究已经确定睡眠障碍是中风的一个危险因素,但动物研究却得出了关于睡眠剥夺(SD)影响的相互矛盾的结果。本研究旨在探讨SD预处理是否能减轻急性缺血性脑卒中后早期神经功能损害,并探讨抗炎细胞因子白细胞介素-38 (IL-38)在这一过程中的潜在作用。方法:雄性Sprague-Dawley大鼠在进行短暂性大脑中动脉闭塞(tMCAO)前,采用改良的多平台方法进行10 d SD预处理。使用Longa评分系统评估神经功能,通过2,3,5-三苯四唑氯(TTC)染色定量脑梗死体积。通过苏木精-伊红(H&E)染色、尼氏染色和末端脱氧核苷酸转移酶介导的dUTP镍端标记(TUNEL)染色评估脑组织的组织病理学变化。采用Western blotting、免疫组织化学和免疫荧光分析IL-38表达水平和炎症反应状态。采用IL-38敲除(IL-38 KO)大鼠验证IL-38在sd诱导的神经保护中的重要作用。结果:SD预处理能显著改善大鼠神经功能,减少脑梗死体积,减少海马CA1区神经元凋亡。值得注意的是,SD预处理显著上调了IL-38的表达,IL-38的表达主要集中在神经元中,并呈现出时间依赖性的增加模式。在IL-38 KO大鼠中,SD预处理的神经保护作用完全消失,未观察到神经评分、梗死面积或神经元存活的改善。机制上,SD预处理降低促炎因子(肿瘤坏死因子-α [TNF-α]、白细胞介素-6 [IL-6]、白细胞介素-1β [IL-1β])水平,抑制核因子-κB (NF-κB)信号通路p65磷酸化,这些调节作用主要依赖于IL-38。结论:SD预处理通过上调IL-38表达对急性缺血性脑卒中具有神经保护作用。IL-38至少在一定程度上通过抑制NF-κB信号通路减轻神经炎症来介导这种保护作用。这些发现表明IL-38是sd诱导的缺血耐受的关键介质,值得进一步的临床研究。
{"title":"Sleep deprivation attenuates acute ischemic stroke-induced hippocampal neuronal injury via the IL-38/NF-κB axis","authors":"Anlin Yue ,&nbsp;Yongfei Liu ,&nbsp;Zhongting Wang ,&nbsp;Mian Zhang ,&nbsp;Yuan Qin ,&nbsp;Zhaoyan Zhao ,&nbsp;Jingxiang Wang ,&nbsp;Shuxuan He ,&nbsp;Jiangjing Li ,&nbsp;Xude Sun ,&nbsp;Li Sun","doi":"10.1016/j.expneurol.2025.115635","DOIUrl":"10.1016/j.expneurol.2025.115635","url":null,"abstract":"<div><h3>Background</h3><div>Although epidemiological studies have identified sleep disorders as a risk factor for stroke, animal studies have yielded conflicting results regarding the impact of sleep deprivation (SD). This study aimed to investigate whether SD preconditioning can alleviate early neurological impairment following acute ischemic stroke and to explore the potential role of the anti-inflammatory cytokine interleukin-38 (IL-38) in this process.</div></div><div><h3>Methods</h3><div>Male Sprague-Dawley rats underwent 10-day SD preconditioning using a modified multi-platform method before being subjected to transient middle cerebral artery occlusion (tMCAO). Neurological function was evaluated using the Longa scoring system, while cerebral infarct volume was quantified via 2,3,5-triphenyltetrazolium chloride (TTC) staining. Histopathological changes in brain tissues were assessed through hematoxylin-eosin (H&amp;E) staining, Nissl staining, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining. The expression level of IL-38 and the status of inflammatory responses were analyzed using Western blotting, immunohistochemistry, and immunofluorescence. IL-38 knockout (IL-38 KO) rats were employed to verify the essential role of IL-38 in SD-induced neuroprotection.</div></div><div><h3>Results</h3><div>SD preconditioning significantly improved neurological function, reduced cerebral infarct volume, and decreased neuronal apoptosis in the CA1 region of the hippocampus. Notably, SD preconditioning markedly upregulated IL-38 expression, which was predominantly localized in neurons and exhibited a time-dependent increase pattern. In IL-38 KO rats, the neuroprotective effects of SD preconditioning were completely abolished, with no improvements observed in neurological scores, infarct size, or neuronal survival. Mechanistically, SD preconditioning reduced the levels of pro-inflammatory cytokines (tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6], and interleukin-1β [IL-1β]) and inhibited the phosphorylation of p65 in the nuclear factor-κB (NF-κB) signaling pathway, and these regulatory effects were largely dependent on IL-38.</div></div><div><h3>Conclusions</h3><div>SD preconditioning confers neuroprotection against acute ischemic stroke by upregulating IL-38 expression. IL-38 mediates this protection, at least in part, by attenuating neuroinflammation through suppression of the NF-κB signaling pathway. These findings identify IL-38 as a critical mediator of SD-induced ischemic tolerance, warranting further clinical investigation.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":"398 ","pages":"Article 115635"},"PeriodicalIF":4.2,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145899637","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Biochanin A exerts neuroprotective effects in Parkinson's disease both in vivo and in vitro by improving mitochondrial dysfunction through the Sirt1 signaling pathway 生物茶素a通过Sirt1信号通路改善线粒体功能障碍,在体内和体外均对帕金森病发挥神经保护作用。
IF 4.2 2区 医学 Q1 NEUROSCIENCES Pub Date : 2026-01-01 DOI: 10.1016/j.expneurol.2025.115636
Shuxiang Tian , Mingguang Niu , Qian Qian, Kexian Zhang, Han Yang, Yang Xiao, Xinyue Jin, Yanyan Yin
The accumulation of reactive oxygen species (ROS) leading to mitochondrial dysfunction is the pathological characteristics underlying the damage to dopaminergic neurons in the substantia nigra (SN) of Parkinson's disease (PD). Therefore, through improving mitochondrial dysfunction may be a potential strategy for PD treatment. Biochanin A (Bioch A), as a natural isoflavone phytoestrogen, has been implicated in studies for its therapeutic potential in neurodegenerative diseases. However, the precise molecular mechanisms by which it modulates PD-related neuronal damage remain unclear, limiting its clinical translational application. This study focuses on the neuroprotective mechanism of Bioch A, systematically revealing the key pathways and regulatory mechanisms through which it exerts its neuroprotective effects in PD by targeting mitochondrial dysfunction. By establishing lipopolysaccharide (LPS)-induced PD model mice and tumor necrosis factor-α (TNF-α)-induced SH-SY5Y cell models, combined with in vivo and in vitro experiments confirmed that Bioch A significantly alleviates dopaminergic neuronal damage by downregulating ROS levels, modulating adenosine triphosphate (ATP) production, promoting mitochondrial biogenesis, and improving abnormal mitochondrial dynamics. More importantly, this study has for the first time revealed that the Sirt1 pathway is a core target for Bioch A in regulating mitochondrial dysfunction. Furthermore, Bioch A promotes mitochondrial biogenesis by activating the Sirt1 pathway and reduces apoptosis levels by promoting mitochondrial fusion. In conclusion, this study provides novel experimental evidence for Bioch A's regulation of mitochondrial dysfunction, establishing the Sirt1 pathway as a key neuroprotective target. This discovery paves the way for Bioch A's clinical translation and targeted therapeutic research in PD.
活性氧(ROS)的积累导致线粒体功能障碍是帕金森病(PD)黑质(SN)多巴胺能神经元损伤的病理特征。因此,通过改善线粒体功能障碍可能是帕金森病治疗的一种潜在策略。生物茶素A (Biochanin A, Bioch A)作为一种天然的异黄酮植物雌激素,因其在神经退行性疾病中的治疗潜力而被研究。然而,其调节pd相关神经元损伤的精确分子机制尚不清楚,限制了其临床转化应用。本研究重点关注Bioch A的神经保护机制,系统揭示其针对线粒体功能障碍在PD中发挥神经保护作用的关键通路和调控机制。通过建立脂多糖(LPS)诱导的PD模型小鼠和肿瘤坏死因子-α (TNF-α)诱导的SH-SY5Y细胞模型,结合体内和体外实验证实,Bioch A通过下调ROS水平、调节三磷酸腺苷(ATP)生成、促进线粒体生物发生、改善线粒体异常动力学等方式,显著缓解多巴胺能神经元损伤。更重要的是,本研究首次揭示了Sirt1通路是Bioch a调节线粒体功能障碍的核心靶点。此外,Bioch A通过激活Sirt1途径促进线粒体生物发生,并通过促进线粒体融合降低细胞凋亡水平。总之,本研究为Bioch A调控线粒体功能障碍提供了新的实验证据,确立了Sirt1通路作为关键的神经保护靶点。这一发现为Bioch A在帕金森病的临床转化和靶向治疗研究铺平了道路。
{"title":"Biochanin A exerts neuroprotective effects in Parkinson's disease both in vivo and in vitro by improving mitochondrial dysfunction through the Sirt1 signaling pathway","authors":"Shuxiang Tian ,&nbsp;Mingguang Niu ,&nbsp;Qian Qian,&nbsp;Kexian Zhang,&nbsp;Han Yang,&nbsp;Yang Xiao,&nbsp;Xinyue Jin,&nbsp;Yanyan Yin","doi":"10.1016/j.expneurol.2025.115636","DOIUrl":"10.1016/j.expneurol.2025.115636","url":null,"abstract":"<div><div>The accumulation of reactive oxygen species (ROS) leading to mitochondrial dysfunction is the pathological characteristics underlying the damage to dopaminergic neurons in the substantia nigra (SN) of Parkinson's disease (PD). Therefore, through improving mitochondrial dysfunction may be a potential strategy for PD treatment. Biochanin A (Bioch A), as a natural isoflavone phytoestrogen, has been implicated in studies for its therapeutic potential in neurodegenerative diseases. However, the precise molecular mechanisms by which it modulates PD-related neuronal damage remain unclear, limiting its clinical translational application. This study focuses on the neuroprotective mechanism of Bioch A, systematically revealing the key pathways and regulatory mechanisms through which it exerts its neuroprotective effects in PD by targeting mitochondrial dysfunction. By establishing lipopolysaccharide (LPS)-induced PD model mice and tumor necrosis factor-α (TNF-α)-induced SH-SY5Y cell models, combined with in vivo and in vitro experiments confirmed that Bioch A significantly alleviates dopaminergic neuronal damage by downregulating ROS levels, modulating adenosine triphosphate (ATP) production, promoting mitochondrial biogenesis, and improving abnormal mitochondrial dynamics. More importantly, this study has for the first time revealed that the Sirt1 pathway is a core target for Bioch A in regulating mitochondrial dysfunction. Furthermore, Bioch A promotes mitochondrial biogenesis by activating the Sirt1 pathway and reduces apoptosis levels by promoting mitochondrial fusion. In conclusion, this study provides novel experimental evidence for Bioch A's regulation of mitochondrial dysfunction, establishing the Sirt1 pathway as a key neuroprotective target. This discovery paves the way for Bioch A's clinical translation and targeted therapeutic research in PD.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":"398 ","pages":"Article 115636"},"PeriodicalIF":4.2,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896397","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Towards zebrafish models of migraine 偏头痛的斑马鱼模型。
IF 4.2 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-31 DOI: 10.1016/j.expneurol.2025.115627
Yubo Xiao , Zichen Li , Haojun Yao , Yimeng Wang , Ruyue Bai , Zhili Yu , Gaomuyi Wang , Zhixuan He , Jingbo Yang , Jiale Chu , Yixuan Pang , Qiuyin Li , Jiahao Cui , Longen Yang , Adam Michael Stewart , Valentina N. Perfilova , Clement Napo , Murilo S. de Abreu , Allan V. Kalueff
Second among global disability causes, migraine is a severely debilitating neurological disorder that affects over 1 billion people. Recognized clinically for millennia, migraine exhibits complex multifactorial pathogenesis, whose mechanisms, risk factors, and therapy remain poorly understood. This also necessitates robust animal models that recapitulate this disorder. Complementing rodent models, the zebrafish (Danio rerio) is a commonly used organism in neuroscience research. Can these fish be used to study pathophysiology of migraine? Here, we discuss the developing utility of these fish for modeling migraine-like conditions and the potential for high-throughput pharmacological testing of its therapies. Critically evaluating the existing challenges of modeling migraine in zebrafish, we also outline potential future lines of research in this field.
在全球致残原因中排名第二的是,偏头痛是一种严重使人衰弱的神经系统疾病,影响超过10亿人。偏头痛的发病机制、危险因素和治疗方法在临床上已有几千年的历史。这也需要强大的动物模型来概括这种疾病。作为啮齿动物模型的补充,斑马鱼(Danio rerio)通常用于神经科学研究。这些鱼可以用来研究偏头痛的病理生理吗?在这里,我们讨论了这些鱼在模拟偏头痛样疾病方面的发展效用,以及其疗法的高通量药理学测试的潜力。批判性地评估斑马鱼偏头痛建模的现有挑战,我们还概述了该领域潜在的未来研究方向。
{"title":"Towards zebrafish models of migraine","authors":"Yubo Xiao ,&nbsp;Zichen Li ,&nbsp;Haojun Yao ,&nbsp;Yimeng Wang ,&nbsp;Ruyue Bai ,&nbsp;Zhili Yu ,&nbsp;Gaomuyi Wang ,&nbsp;Zhixuan He ,&nbsp;Jingbo Yang ,&nbsp;Jiale Chu ,&nbsp;Yixuan Pang ,&nbsp;Qiuyin Li ,&nbsp;Jiahao Cui ,&nbsp;Longen Yang ,&nbsp;Adam Michael Stewart ,&nbsp;Valentina N. Perfilova ,&nbsp;Clement Napo ,&nbsp;Murilo S. de Abreu ,&nbsp;Allan V. Kalueff","doi":"10.1016/j.expneurol.2025.115627","DOIUrl":"10.1016/j.expneurol.2025.115627","url":null,"abstract":"<div><div>Second among global disability causes, migraine is a severely debilitating neurological disorder that affects over 1 billion people. Recognized clinically for millennia, migraine exhibits complex multifactorial pathogenesis, whose mechanisms, risk factors, and therapy remain poorly understood. This also necessitates robust animal models that recapitulate this disorder. Complementing rodent models, the zebrafish (<em>Danio rerio</em>) is a commonly used organism in neuroscience research. Can these fish be used to study pathophysiology of migraine? Here, we discuss the developing utility of these fish for modeling migraine-like conditions and the potential for high-throughput pharmacological testing of its therapies. Critically evaluating the existing challenges of modeling migraine in zebrafish, we also outline potential future lines of research in this field.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":"398 ","pages":"Article 115627"},"PeriodicalIF":4.2,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145891723","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Demethyleneberberine attenuates combined cognitive and metabolic dysfunctions in an insulin-resistance-induced Alzheimer's disease rat model: Synthesis, in-silico and in-vivo insights 去亚甲基小檗碱减轻胰岛素抵抗诱导的阿尔茨海默病大鼠模型中的认知和代谢功能障碍:合成、计算机和体内观察
IF 4.2 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-31 DOI: 10.1016/j.expneurol.2025.115634
Amritpal Kaur , Shareen Singh , Manjinder Singh , Pragati Silakari , Ashi Mannan , Sukriti Vishwas , Puneet Kumar , Vetriselvan Subramaniyan , Thakur Gurjeet Singh
In this study, we evaluated the therapeutic potential of DMB, a berberine derivative known for its enhanced bioavailability and reduced toxicity. DMB was synthesized and administered orally at doses of 5 and 10 mg/kg in an in vivo rat model of insulin resistance-induced Alzheimer's disease (AD). This model was established using a combination of a high-fat diet (HFD), streptozotocin (35 mg/kg; intraperitoneally), and amyloid-β2535. In-silico docking studies revealed that DMB exhibits a high binding affinity for key proteins implicated in both AD and diabetes, including insulin receptors, leptin receptors, protein tyrosine phosphatase 1B (PTP1B), HMG-CoA reductase, acetylcholinesterase (AChE), and butyrylcholinesterase (BChE). Molecular dynamics simulations confirmed the stable binding and inhibitory potential of Demethyleneberberine (DMB) against Insulin Receptor Tyrosine Kinase and AChE. Pharmacological network analysis indicated that DMB modulates multiple pathways involved in metabolic and cognitive decline, suggesting its promise as a therapeutic candidate for insulin resistance-induced AD. Neurobehavioral assessments demonstrated that DMB significantly (p < 0.001) improved cognitive function, ameliorated metabolic disruptions (elevated blood glucose and insulin levels), and normalized pro-inflammatory markers (Tumor Necrosis Factor-alpha (TNF-α), Interleukin 1-beta (IL-1β)) and oxidative stress parameters (Thiobarbituric Acid Reactive Substances (TBARS), glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT). Additionally, DMB reduced levels of AD-related biomarkers, including BACE-1 (β-secretase 1), amyloid-β, and acetylcholinesterase, indicating its capacity to mitigate oxidative stress and amyloidogenesis. This multidisciplinary approach, integrating in vivo and in-silico methodologies, provides a comprehensive understanding of DMB's neuroprotective effects and underscores its potential as a therapeutic agent for both AD and diabetes.
在这项研究中,我们评估了DMB的治疗潜力,DMB是一种以提高生物利用度和降低毒性而闻名的小檗碱衍生物。合成DMB并在胰岛素抵抗性阿尔茨海默病(AD)大鼠模型中以5和10 mg/kg的剂量口服。采用高脂肪饲粮(HFD)、链脲佐菌素(35 mg/kg;腹腔注射)和淀粉样蛋白-β25-35联合建立模型。硅对接研究显示,DMB对AD和糖尿病相关的关键蛋白具有高结合亲和力,包括胰岛素受体、瘦素受体、蛋白酪氨酸磷酸酶1B (PTP1B)、HMG-CoA还原酶、乙酰胆碱酯酶(AChE)和丁基胆碱酯酶(BChE)。分子动力学模拟证实了去亚甲基小檗碱(DMB)对胰岛素受体酪氨酸激酶和AChE的稳定结合和抑制潜力。药理网络分析表明,DMB可调节代谢和认知能力下降的多种途径,提示其有望成为胰岛素抵抗性AD的治疗候选药物。神经行为评估显示DMB显著(p
{"title":"Demethyleneberberine attenuates combined cognitive and metabolic dysfunctions in an insulin-resistance-induced Alzheimer's disease rat model: Synthesis, in-silico and in-vivo insights","authors":"Amritpal Kaur ,&nbsp;Shareen Singh ,&nbsp;Manjinder Singh ,&nbsp;Pragati Silakari ,&nbsp;Ashi Mannan ,&nbsp;Sukriti Vishwas ,&nbsp;Puneet Kumar ,&nbsp;Vetriselvan Subramaniyan ,&nbsp;Thakur Gurjeet Singh","doi":"10.1016/j.expneurol.2025.115634","DOIUrl":"10.1016/j.expneurol.2025.115634","url":null,"abstract":"<div><div>In this study, we evaluated the therapeutic potential of DMB, a berberine derivative known for its enhanced bioavailability and reduced toxicity. DMB was synthesized and administered orally at doses of 5 and 10 mg/kg in an in vivo rat model of insulin resistance-induced Alzheimer's disease (AD). This model was established using a combination of a high-fat diet (HFD), streptozotocin (35 mg/kg; intraperitoneally), and amyloid-β<sub>25</sub><sub>–</sub><sub>35</sub>. In-silico docking studies revealed that DMB exhibits a high binding affinity for key proteins implicated in both AD and diabetes, including insulin receptors, leptin receptors, protein tyrosine phosphatase 1B (PTP1B), HMG-CoA reductase, acetylcholinesterase (AChE), and butyrylcholinesterase (BChE). Molecular dynamics simulations confirmed the stable binding and inhibitory potential of Demethyleneberberine (DMB) against Insulin Receptor Tyrosine Kinase and AChE. Pharmacological network analysis indicated that DMB modulates multiple pathways involved in metabolic and cognitive decline, suggesting its promise as a therapeutic candidate for insulin resistance-induced AD. Neurobehavioral assessments demonstrated that DMB significantly (<em>p</em> &lt; 0.001) improved cognitive function, ameliorated metabolic disruptions (elevated blood glucose and insulin levels), and normalized pro-inflammatory markers (Tumor Necrosis Factor-alpha (TNF-α), Interleukin 1-beta (IL-1β)) and oxidative stress parameters (Thiobarbituric Acid Reactive Substances (TBARS), glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT). Additionally, DMB reduced levels of AD-related biomarkers, including BACE-1 (β-secretase 1), amyloid-β, and acetylcholinesterase, indicating its capacity to mitigate oxidative stress and amyloidogenesis. This multidisciplinary approach, integrating in vivo and in-silico methodologies, provides a comprehensive understanding of DMB's neuroprotective effects and underscores its potential as a therapeutic agent for both AD and diabetes.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":"398 ","pages":"Article 115634"},"PeriodicalIF":4.2,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892330","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Electroacupuncture ameliorates tau-driven cognitive decline by modulating NF-κB/NLRP3 inflammasome signaling in P301S mice 电针通过调节P301S小鼠NF-κB/NLRP3炎性体信号传导改善tau驱动的认知衰退。
IF 4.2 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-31 DOI: 10.1016/j.expneurol.2025.115637
Ruixue Zheng , Xueyun Liu , Zhenge Liao , Runjie Wan , Gengbin Qiu , Min Li , Chunzhi Tang , Runjin Zhou , Juxian Song
Alzheimer's disease (AD) progression is driven by a vicious cycle wherein pathological Tau hyperphosphorylation promotes microglial activation and NF-κB/NLRP3 inflammasome signaling, leading to excessive secretion of proinflammatory cytokines that reciprocally exacerbate Tau pathology. While pharmacological NLRP3 inhibitors hold therapeutic potential for AD, critical barriers—including poor blood-brain barrier penetration, suboptimal target selectivity, and safety concerns—persist. This study investigated whether electroacupuncture (EA), a non-pharmacological neuromodulatory approach, could disrupt this Tau-inflammasome cycle. Using P301S Tau transgenic mice, two EA regimens were tested at the GV20 (Baihui) acupoint: 6-month-old mice receiving a 1-month EA intervention, and 6-month-old mice undergoing a prolonged 3-month EA intervention. Cognitive function was evaluated via Y-maze, novel object recognition (NOR), and Morris water maze (MWM) tests, while corticospinal function was assessed using tail-suspension limb-clasping scoring. Hippocampal Tau pathology and inflammatory signaling were analyzed by Western blot and immunohistochemistry, targeting total Tau, phosphorylated Tau, NF-κB, NLRP3, caspase-1, IL-1β, IL-18, TNF-α, and microglial morphology. Short-term (1-month) EA treatment significantly improved spatial working memory and recognition memory. Mechanistically, EA reduced p-Tau levels, suppressed NF-κB activation (decreased p-P65/P65 ratio), downregulated NLRP3 inflammasome components (NLRP3, cleaved caspase-1) and proinflammatory cytokines (IL-1β, IL-18 and TNF-α), and mitigated microglial hyperactivation. Importantly, long-term (3-month) EA treatment persistently suppressed p-Tau accumulation and neuroinflammation, thereby consolidating cognitive benefits even in P301S mice with severe corticospinal dysfunction. These findings establish EA as a multi-targeted immunomodulatory strategy that attenuates Tau-driven neuroinflammation through the TNF-α/NF-κB/NLRP3 signaling axis, highlighting its potential as a safe, non-pharmacological adjunct or alternative therapy for AD and related tauopathies.
阿尔茨海默病(AD)的进展是由一个恶性循环驱动的,其中病理性Tau过度磷酸化促进小胶质细胞活化和NF-κB/NLRP3炎性小体信号传导,导致促炎细胞因子的过度分泌,从而相互加剧Tau病理。虽然药物NLRP3抑制剂具有治疗AD的潜力,但关键障碍-包括血脑屏障穿透性差,次优靶标选择性和安全性问题-仍然存在。这项研究调查了电针(EA),一种非药物神经调节方法,是否可以破坏这种tau -炎性体周期。采用P301S Tau转基因小鼠,在GV20(百会)穴测试两种EA方案:6月龄小鼠接受1个月的EA干预,6月龄小鼠接受3个月的延长EA干预。通过y形迷宫、新物体识别(NOR)和Morris水迷宫(MWM)测试评估认知功能,采用悬尾扣肢评分评估皮质脊髓功能。以总Tau蛋白、磷酸化Tau蛋白、NF-κB、NLRP3、caspase-1、IL-1β、IL-18、TNF-α和小胶质细胞形态为指标,采用Western blot和免疫组化方法分析海马Tau蛋白病理和炎症信号传导。短期(1个月)EA治疗显著改善了空间工作记忆和识别记忆。从机制上讲,EA降低了p-Tau水平,抑制了NF-κB的活化(降低了p-P65/P65比值),下调了NLRP3炎性小体成分(NLRP3、cleaved caspase-1)和促炎细胞因子(IL-1β、IL-18和TNF-α),减轻了小胶质细胞的过度活化。重要的是,长期(3个月)EA治疗持续抑制p-Tau积聚和神经炎症,从而巩固认知益处,甚至在严重皮质脊髓功能障碍的P301S小鼠中也是如此。这些发现表明EA是一种多靶点的免疫调节策略,通过TNF-α/NF-κB/NLRP3信号轴减弱tau驱动的神经炎症,突出了其作为AD和相关tau病变的安全,非药物辅助或替代疗法的潜力。
{"title":"Electroacupuncture ameliorates tau-driven cognitive decline by modulating NF-κB/NLRP3 inflammasome signaling in P301S mice","authors":"Ruixue Zheng ,&nbsp;Xueyun Liu ,&nbsp;Zhenge Liao ,&nbsp;Runjie Wan ,&nbsp;Gengbin Qiu ,&nbsp;Min Li ,&nbsp;Chunzhi Tang ,&nbsp;Runjin Zhou ,&nbsp;Juxian Song","doi":"10.1016/j.expneurol.2025.115637","DOIUrl":"10.1016/j.expneurol.2025.115637","url":null,"abstract":"<div><div>Alzheimer's disease (AD) progression is driven by a vicious cycle wherein pathological Tau hyperphosphorylation promotes microglial activation and NF-κB/NLRP3 inflammasome signaling, leading to excessive secretion of proinflammatory cytokines that reciprocally exacerbate Tau pathology. While pharmacological NLRP3 inhibitors hold therapeutic potential for AD, critical barriers—including poor blood-brain barrier penetration, suboptimal target selectivity, and safety concerns—persist. This study investigated whether electroacupuncture (EA), a non-pharmacological neuromodulatory approach, could disrupt this Tau-inflammasome cycle. Using P301S Tau transgenic mice, two EA regimens were tested at the GV20 (Baihui) acupoint: 6-month-old mice receiving a 1-month EA intervention, and 6-month-old mice undergoing a prolonged 3-month EA intervention. Cognitive function was evaluated via Y-maze, novel object recognition (NOR), and Morris water maze (MWM) tests, while corticospinal function was assessed using tail-suspension limb-clasping scoring. Hippocampal Tau pathology and inflammatory signaling were analyzed by Western blot and immunohistochemistry, targeting total Tau, phosphorylated Tau, NF-κB, NLRP3, caspase-1, IL-1β, IL-18, TNF-α, and microglial morphology. Short-term (1-month) EA treatment significantly improved spatial working memory and recognition memory. Mechanistically, EA reduced p-Tau levels, suppressed NF-κB activation (decreased p-P65/P65 ratio), downregulated NLRP3 inflammasome components (NLRP3, cleaved caspase-1) and proinflammatory cytokines (IL-1β, IL-18 and TNF-α), and mitigated microglial hyperactivation. Importantly, long-term (3-month) EA treatment persistently suppressed p-Tau accumulation and neuroinflammation, thereby consolidating cognitive benefits even in P301S mice with severe corticospinal dysfunction. These findings establish EA as a multi-targeted immunomodulatory strategy that attenuates Tau-driven neuroinflammation through the TNF-α/NF-κB/NLRP3 signaling axis, highlighting its potential as a safe, non-pharmacological adjunct or alternative therapy for AD and related tauopathies.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":"398 ","pages":"Article 115637"},"PeriodicalIF":4.2,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892349","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Apelin-13 attenuates microglia-mediated neuroinflammation following intracerebral hemorrhage via targeting JAK2/STAT3 signaling pathway Apelin-13通过靶向JAK2/STAT3信号通路减轻脑出血后小胶质细胞介导的神经炎症
IF 4.2 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-31 DOI: 10.1016/j.expneurol.2025.115633
Pingping Guo , Jingjing Li , Xiangyu Zhang , Qingli Wang , Yang Liu , Huizhen Zhou , Yun Chen , V. Wee Yong , Mengzhou Xue

Background

Neuroinflammation is a critical contributor to secondary brain injury and subsequent neurological decline after intracerebral hemorrhage (ICH). Apelin-13, the most bioactive isoform of the endogenous G protein-coupled receptor (GPCR) ligand, exhibits protective roles in multiple neurological disorders. Nevertheless, its therapeutic effects and underlying mechanisms in neuroinflammation following ICH remain elusive.

Methods

In vivo, ICH was induced in mice with collagenase type VII, followed by intracerebroventricular injection of Apelin-13. In vitro, BV2 microglia were pretreated with Apelin-13 overnight, followed by lipopolysaccharide (LPS) stimulation. To investigate the mechanistic role of Apelin-13, we employed specific shRNA for APJ knockdown and the selective JAK2/STAT3 inhibitor WP1066 for pathway blockage. Western blotting and immunofluorescence assays were applied to assess JAK2/STAT3 signaling activation and pro-inflammatory mediator expression.

Results

Apelin-13 significantly decreased hematoma volume and mitigated neurological impairments in ICH mice. Correspondingly, both in vivo and in vitro studies confirmed its efficacy in attenuating microglia-mediated neuroinflammation. Mechanistically, Apelin-13 significantly suppressed JAK2/STAT3 signaling pathway in LPS-stimulated BV2 microglia. This suppression was reversed by APJ knockdown, verifying the necessity of the Apelin-13/APJ interaction. Furthermore, combining WP1066 with Apelin-13 significantly enhanced its anti-inflammatory effects, as evidenced by a more pronounced reduction in p-JAK2/p-STAT3 levels and pro-inflammatory cytokine secretion. Finally, the inhibition of the microglial JAK2/STAT3 pathway by Apelin-13 was also confirmed in the perihematomal brain tissues of ICH mice.

Conclusions

Apelin-13 attenuated brain injury after ICH by suppressing microglia-mediated neuroinflammation through APJ receptor-dependent inhibition of the JAK2/STAT3 pathway.
背景神经炎症是脑出血(ICH)后继发性脑损伤和随后的神经功能衰退的重要因素。Apelin-13是内源性G蛋白偶联受体(GPCR)配体中最具生物活性的异构体,在多种神经系统疾病中具有保护作用。然而,其治疗脑出血后神经炎症的效果和潜在机制仍不清楚。方法采用7型胶原酶诱导小鼠脑出血,然后脑室内注射Apelin-13。体外,用Apelin-13预处理BV2小胶质细胞过夜,然后进行脂多糖(LPS)刺激。为了研究Apelin-13的机制作用,我们使用了特异性shRNA来下调APJ,并使用了选择性JAK2/STAT3抑制剂WP1066来阻断通路。Western blotting和免疫荧光法检测JAK2/STAT3信号的激活和促炎介质的表达。结果apelin -13能显著减少脑出血小鼠血肿体积,减轻神经损伤。相应地,体内和体外研究都证实了其减轻小胶质细胞介导的神经炎症的功效。机制上,Apelin-13在lps刺激的BV2小胶质细胞中显著抑制JAK2/STAT3信号通路。这种抑制被APJ敲除逆转,验证了Apelin-13/APJ相互作用的必要性。此外,WP1066与Apelin-13联合使用可显著增强其抗炎作用,p-JAK2/p-STAT3水平和促炎细胞因子分泌均显著降低。最后,在脑出血小鼠血肿周围脑组织中也证实了Apelin-13对小胶质细胞JAK2/STAT3通路的抑制作用。结论sapelin -13通过APJ受体依赖性抑制JAK2/STAT3通路,抑制小胶质细胞介导的神经炎症,减轻脑出血后脑损伤。
{"title":"Apelin-13 attenuates microglia-mediated neuroinflammation following intracerebral hemorrhage via targeting JAK2/STAT3 signaling pathway","authors":"Pingping Guo ,&nbsp;Jingjing Li ,&nbsp;Xiangyu Zhang ,&nbsp;Qingli Wang ,&nbsp;Yang Liu ,&nbsp;Huizhen Zhou ,&nbsp;Yun Chen ,&nbsp;V. Wee Yong ,&nbsp;Mengzhou Xue","doi":"10.1016/j.expneurol.2025.115633","DOIUrl":"10.1016/j.expneurol.2025.115633","url":null,"abstract":"<div><h3>Background</h3><div>Neuroinflammation is a critical contributor to secondary brain injury and subsequent neurological decline after intracerebral hemorrhage (ICH). Apelin-13, the most bioactive isoform of the endogenous G protein-coupled receptor (GPCR) ligand, exhibits protective roles in multiple neurological disorders. Nevertheless, its therapeutic effects and underlying mechanisms in neuroinflammation following ICH remain elusive.</div></div><div><h3>Methods</h3><div>In vivo, ICH was induced in mice with collagenase type VII, followed by intracerebroventricular injection of Apelin-13. In vitro, BV2 microglia were pretreated with Apelin-13 overnight, followed by lipopolysaccharide (LPS) stimulation. To investigate the mechanistic role of Apelin-13, we employed specific shRNA for APJ knockdown and the selective JAK2/STAT3 inhibitor WP1066 for pathway blockage. Western blotting and immunofluorescence assays were applied to assess JAK2/STAT3 signaling activation and pro-inflammatory mediator expression.</div></div><div><h3>Results</h3><div>Apelin-13 significantly decreased hematoma volume and mitigated neurological impairments in ICH mice. Correspondingly, both in vivo and in vitro studies confirmed its efficacy in attenuating microglia-mediated neuroinflammation. Mechanistically, Apelin-13 significantly suppressed JAK2/STAT3 signaling pathway in LPS-stimulated BV2 microglia. This suppression was reversed by APJ knockdown, verifying the necessity of the Apelin-13/APJ interaction. Furthermore, combining WP1066 with Apelin-13 significantly enhanced its anti-inflammatory effects, as evidenced by a more pronounced reduction in p-JAK2/p-STAT3 levels and pro-inflammatory cytokine secretion. Finally, the inhibition of the microglial JAK2/STAT3 pathway by Apelin-13 was also confirmed in the perihematomal brain tissues of ICH mice.</div></div><div><h3>Conclusions</h3><div>Apelin-13 attenuated brain injury after ICH by suppressing microglia-mediated neuroinflammation through APJ receptor-dependent inhibition of the JAK2/STAT3 pathway.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":"398 ","pages":"Article 115633"},"PeriodicalIF":4.2,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145882228","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Role of A-delta low threshold mechanoreceptors and tropomyosin receptor kinase B plasticity in at-level aversive pain after spinal cord injury a - δ低阈机械受体和原肌球蛋白受体激酶B可塑性在脊髓损伤后水平厌恶性疼痛中的作用
IF 4.2 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-31 DOI: 10.1016/j.expneurol.2025.115628
Shangrila Parvin, Kyeongran Jang, Sandra M. Garraway
The mechanisms of neuropathic pain after spinal cord injury (SCI) are not fully understood, although spinal and peripheral processes are involved. Maladaptive tropomyosin receptor kinase-B (TrkB) signaling has been implicated in pain hypersensitivity after SCI. A-delta-low threshold mechanoreceptors (Aδ-LTMRs) innervate the hairy skin and normally signal directional touch and are identified by their preferential TrkB expression. This study investigated whether Aδ-LTMRs play a role in at-level pain after thoracic contusion SCI. Using a modified light-dark chamber conditioned place aversion (CPA) paradigm, we assessed chamber preferences and transitions between chambers in response to mechanical stimulation, and optogenetic stimulation of Aδ-LTMRs in the trunk skin of adult TrkBCre mice of both sexes. Respiratory rates (RRs) were monitored at baseline and during truncal stimulation. The expression of brain-derived neurotrophic factor (BDNF), TrkB and pERK1/2 in the lesioned spinal cord and skin, and histological changes in Aδ-LTMRs in trunk skin were assessed. In addition, electrophysiological studies examined changes in Aδ-LTMRs membrane and firing properties, and response to bath-applied 7, 8-dihydroxyflavone (7,8-DHF), a TrkB agonist. The results showed that whereas brush stimulation evoked an aversive response at 4 weeks post-SCI that was accompanied by increased RRs, targeted stimulation of Aδ-LTMRs produced an aversive response 7 weeks post-SCI. SCI increased BDNF, TrkB and pERK1/2 expression in the skin, and augmented 7,8-DHF-induced inward current in Aδ-LTMRs. Together, these results suggest that plasticity of Aδ-LTMR, including an increase in TrkB signaling in the periphery, contribute to at-level affective pain following chronic SCI in adult mice.
脊髓损伤(SCI)后神经性疼痛的机制尚不完全清楚,尽管涉及脊髓和外周过程。原肌球蛋白受体激酶- b (TrkB)信号的不适应与脊髓损伤后的疼痛超敏反应有关。a- δ-低阈值机械感受器(a- δ- ltmrs)支配毛茸茸的皮肤,通常指示定向触摸,并通过其优先表达TrkB来识别。本研究探讨了a - δ- ltmr是否在胸挫伤脊髓损伤后的水平疼痛中起作用。利用改进的光-暗室条件下的场所厌恶(CPA)范式,我们评估了成年TrkBCre小鼠躯干皮肤a δ- ltmrs在机械刺激和光遗传刺激下的室偏好和室间转换。在基线和躯干刺激期间监测呼吸频率(rr)。观察脑源性神经营养因子(BDNF)、TrkB和pERK1/2在损伤脊髓和皮肤中的表达及躯干皮肤Aδ-LTMRs的组织学变化。此外,电生理研究还检测了a - δ- ltmrs膜和放电特性的变化,以及对TrkB激动剂7,8-二羟黄酮(7,8- dhf)的反应。结果表明,刷刺激在脊髓损伤后4周引起厌恶反应,并伴有rr升高,而a δ- ltmr靶向刺激在脊髓损伤后7周产生厌恶反应。SCI增加了皮肤中BDNF、TrkB和pERK1/2的表达,增强了Aδ-LTMRs中7,8- dhf诱导的内向电流。总之,这些结果表明,a - δ- ltmr的可塑性,包括外周TrkB信号的增加,有助于成年小鼠慢性脊髓损伤后的情感性疼痛。
{"title":"Role of A-delta low threshold mechanoreceptors and tropomyosin receptor kinase B plasticity in at-level aversive pain after spinal cord injury","authors":"Shangrila Parvin,&nbsp;Kyeongran Jang,&nbsp;Sandra M. Garraway","doi":"10.1016/j.expneurol.2025.115628","DOIUrl":"10.1016/j.expneurol.2025.115628","url":null,"abstract":"<div><div>The mechanisms of neuropathic pain after spinal cord injury (SCI) are not fully understood, although spinal and peripheral processes are involved. Maladaptive tropomyosin receptor kinase-B (TrkB) signaling has been implicated in pain hypersensitivity after SCI. A-delta-low threshold mechanoreceptors (Aδ-LTMRs) innervate the hairy skin and normally signal directional touch and are identified by their preferential TrkB expression. This study investigated whether Aδ-LTMRs play a role in at-level pain after thoracic contusion SCI. Using a modified light-dark chamber conditioned place aversion (CPA) paradigm, we assessed chamber preferences and transitions between chambers in response to mechanical stimulation, and optogenetic stimulation of Aδ-LTMRs in the trunk skin of adult TrkB<sup>Cre</sup> mice of both sexes. Respiratory rates (RRs) were monitored at baseline and during truncal stimulation. The expression of brain-derived neurotrophic factor (BDNF), TrkB and pERK1/2 in the lesioned spinal cord and skin, and histological changes in Aδ-LTMRs in trunk skin were assessed. In addition, electrophysiological studies examined changes in Aδ-LTMRs membrane and firing properties, and response to bath-applied 7, 8-dihydroxyflavone (7,8-DHF), a TrkB agonist. The results showed that whereas brush stimulation evoked an aversive response at 4 weeks post-SCI that was accompanied by increased RRs, targeted stimulation of Aδ-LTMRs produced an aversive response 7 weeks post-SCI. SCI increased BDNF, TrkB and pERK1/2 expression in the skin, and augmented 7,8-DHF-induced inward current in Aδ-LTMRs. Together, these results suggest that plasticity of Aδ-LTMR, including an increase in TrkB signaling in the periphery, contribute to at-level affective pain following chronic SCI in adult mice.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":"398 ","pages":"Article 115628"},"PeriodicalIF":4.2,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145882229","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Experimental Neurology
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