首页 > 最新文献

NeuroMolecular Medicine最新文献

英文 中文
Targeting Microglial Activation to Modulate Neuroinflammation in Alzheimer's Disease. 靶向小胶质细胞激活调节阿尔茨海默病的神经炎症。
IF 3.9 4区 医学 Q2 NEUROSCIENCES Pub Date : 2025-12-04 DOI: 10.1007/s12017-025-08898-2
Vinay Patil, Amit Sharma, Bhavin Parekh, Husni Farah, S Renuka Jyothi, Swati Mishra, Anima Nanda, Shaker Al-Hasnaawei, Manoj Kumar Mishra

Alzheimer's disease is a multifaceted neurodegenerative condition marked by the build-up of amyloid plaques and neurofibrillary tangles that lead to progressive cognitive impairment. Neuroinflammation, especially the activation of microglia, plays a pivotal part in driving this pathology. Microglia are the brain's resident immune cells and can adopt a spectrum of activation states that support either neuroprotection or neurodegeneration. Evidence shows that their phenotypes are highly dynamic and shaped by environmental influences and pathological signals. During the early phases of the disease, microglia tend to assume anti-inflammatory roles that facilitate plaque clearance and promote tissue recovery. Prolonged or dysregulated activation, however, shifts them toward a pro-inflammatory state that amplifies neuronal damage. Several molecular pathways including JAK STAT, PI3K AKT, and MAPK are central to regulating these processes and have emerged as promising therapeutic targets. This review summarizes current insights into microglial phenotypic transitions, the signaling mechanisms governing their activation, and the therapeutic potential of modulating neuroinflammation. Enhancing the neuroprotective capacity of microglia, suppressing chronic inflammatory responses, and targeting key receptors such as TREM2 and P2 × 7 represent potential strategies. A deeper understanding of microglial interactions with other glial cells and the molecular drivers of their activation may provide new avenues for slowing or halting the progression of Alzheimer's disease and related neurodegenerative disorders.

阿尔茨海默病是一种多方面的神经退行性疾病,其特征是淀粉样斑块和神经原纤维缠结的积累,导致进行性认知障碍。神经炎症,尤其是小胶质细胞的激活,在驱动这种病理中起着关键作用。小胶质细胞是大脑的常驻免疫细胞,可以采取一系列激活状态,支持神经保护或神经退化。有证据表明,它们的表型是高度动态的,受环境影响和病理信号的影响。在疾病的早期阶段,小胶质细胞倾向于承担抗炎作用,促进斑块清除和促进组织恢复。然而,长时间或失调的激活会使它们转向促炎状态,从而放大神经元损伤。包括JAK STAT、PI3K AKT和MAPK在内的一些分子通路是调节这些过程的核心,并已成为有希望的治疗靶点。这篇综述总结了目前对小胶质细胞表型转变、控制其激活的信号机制以及调节神经炎症的治疗潜力的见解。增强小胶质细胞的神经保护能力,抑制慢性炎症反应,靶向tre2和P2 × 7等关键受体是潜在的策略。更深入地了解小胶质细胞与其他胶质细胞的相互作用及其激活的分子驱动因素,可能为减缓或阻止阿尔茨海默病和相关神经退行性疾病的进展提供新的途径。
{"title":"Targeting Microglial Activation to Modulate Neuroinflammation in Alzheimer's Disease.","authors":"Vinay Patil, Amit Sharma, Bhavin Parekh, Husni Farah, S Renuka Jyothi, Swati Mishra, Anima Nanda, Shaker Al-Hasnaawei, Manoj Kumar Mishra","doi":"10.1007/s12017-025-08898-2","DOIUrl":"https://doi.org/10.1007/s12017-025-08898-2","url":null,"abstract":"<p><p>Alzheimer's disease is a multifaceted neurodegenerative condition marked by the build-up of amyloid plaques and neurofibrillary tangles that lead to progressive cognitive impairment. Neuroinflammation, especially the activation of microglia, plays a pivotal part in driving this pathology. Microglia are the brain's resident immune cells and can adopt a spectrum of activation states that support either neuroprotection or neurodegeneration. Evidence shows that their phenotypes are highly dynamic and shaped by environmental influences and pathological signals. During the early phases of the disease, microglia tend to assume anti-inflammatory roles that facilitate plaque clearance and promote tissue recovery. Prolonged or dysregulated activation, however, shifts them toward a pro-inflammatory state that amplifies neuronal damage. Several molecular pathways including JAK STAT, PI3K AKT, and MAPK are central to regulating these processes and have emerged as promising therapeutic targets. This review summarizes current insights into microglial phenotypic transitions, the signaling mechanisms governing their activation, and the therapeutic potential of modulating neuroinflammation. Enhancing the neuroprotective capacity of microglia, suppressing chronic inflammatory responses, and targeting key receptors such as TREM2 and P2 × 7 represent potential strategies. A deeper understanding of microglial interactions with other glial cells and the molecular drivers of their activation may provide new avenues for slowing or halting the progression of Alzheimer's disease and related neurodegenerative disorders.</p>","PeriodicalId":19304,"journal":{"name":"NeuroMolecular Medicine","volume":"27 1","pages":"76"},"PeriodicalIF":3.9,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668591","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Comparative Hippocampal Transcriptomics Reveals Model-Specific Pathways and Convergent Inflammation in Sepsis-Associated Encephalopathy. 比较海马转录组学揭示脓毒症相关脑病的模型特异性途径和会聚性炎症。
IF 3.9 4区 医学 Q2 NEUROSCIENCES Pub Date : 2025-11-27 DOI: 10.1007/s12017-025-08896-4
Xueling Zhang, Mengshi Yang, Yaxuan Zhang, Limin Tao, Xiyu Chen, Fei Liu, Bin Zhang, Guangzhi Shi

Sepsis-associated encephalopathy (SAE) is a serious sepsis complication with high mortality. Animal models, including cecal ligation and puncture (CLP), lipopolysaccharide (LPS) injection, and peritoneal contamination and infection (PCI), are known to trigger distinct inflammatory responses with differential hippocampal impact. This study aimed to comprehensively compare the hippocampal transcriptomic profiles and validate key findings through independent experimentation. Transcriptomic datasets GSE253309 (CLP), GSE226120 (LPS), and GSE167610 (PCI) were retrieved from the GEO database. Bioinformatics analyses were employed to identify DEGs and enriched pathways. WGCNA pinpointed characteristic modules, and PPI networks were constructed and analyzed. Critically, an independent CLP-induced SAE mouse model was established, and hippocampal RNA sequencing was performed for confirmation. DEG analysis revealed 381, 533, and 85 significant DEGs in the CLP, LPS, and PCI datasets, respectively. CLP and LPS models shared a robust signature of neuroinflammation, significantly enriching GO terms related to immune response and inflammatory response, and KEGG pathways such as TNF, NF-κB, IL-17. In stark contrast, the PCI model was predominantly associated with cell migration, aldarate metabolism, and enriched in metabolic pathways, including bile secretion, ascorbate and aldarate metabolism. Cross-dataset analysis identified 29 common DEGs, from which a PPI network of 16 hub genes was constructed. Importantly, independent validation confirmed a strong concordance (r = 0.576) between the CLP-seq discovery cohort and the experimental CLP-seq data. Lcn2, S100a8, S100a9, Lrg1 and the TNF/IL-17 signaling pathways were robustly verified. CLP and LPS models demonstrate convergent hippocampal transcriptomic profiles distinct from PCI. Lcn2, S100a8, S100a9, Lrg1 and the TNF and IL-17 signaling pathways are highly reliable core features in SAE.

脓毒症相关脑病(SAE)是一种严重的脓毒症并发症,死亡率高。动物模型,包括盲肠结扎和穿刺(CLP)、脂多糖(LPS)注射和腹膜污染和感染(PCI),已知会引发不同的炎症反应,并对海马产生不同的影响。本研究旨在全面比较海马转录组谱,并通过独立实验验证关键发现。转录组学数据集GSE253309 (CLP)、GSE226120 (LPS)和GSE167610 (PCI)从GEO数据库中检索。生物信息学分析用于鉴定deg和富集途径。WGCNA确定了特征模块,构建并分析了PPI网络。重要的是,建立了独立的clp诱导的SAE小鼠模型,并进行了海马RNA测序以进行确认。DEG分析显示CLP、LPS和PCI数据集中分别有381、533和85个显著DEG。CLP和LPS模型都具有强大的神经炎症特征,显著丰富了与免疫反应和炎症反应相关的GO术语,以及TNF、NF-κB、IL-17等KEGG通路。与之形成鲜明对比的是,PCI模型主要与细胞迁移、醛酸盐代谢相关,并且代谢途径丰富,包括胆汁分泌、抗坏血酸盐和醛酸盐代谢。交叉数据集分析确定了29个共同的deg,从中构建了16个中心基因的PPI网络。重要的是,独立验证证实了CLP-seq发现队列与实验CLP-seq数据之间的强一致性(r = 0.576)。Lcn2、S100a8、S100a9、Lrg1和TNF/IL-17信号通路得到了强有力的验证。CLP和LPS模型显示与PCI不同的海马转录组特征趋同。Lcn2、S100a8、S100a9、Lrg1以及TNF和IL-17信号通路是SAE高度可靠的核心特征。
{"title":"Comparative Hippocampal Transcriptomics Reveals Model-Specific Pathways and Convergent Inflammation in Sepsis-Associated Encephalopathy.","authors":"Xueling Zhang, Mengshi Yang, Yaxuan Zhang, Limin Tao, Xiyu Chen, Fei Liu, Bin Zhang, Guangzhi Shi","doi":"10.1007/s12017-025-08896-4","DOIUrl":"https://doi.org/10.1007/s12017-025-08896-4","url":null,"abstract":"<p><p>Sepsis-associated encephalopathy (SAE) is a serious sepsis complication with high mortality. Animal models, including cecal ligation and puncture (CLP), lipopolysaccharide (LPS) injection, and peritoneal contamination and infection (PCI), are known to trigger distinct inflammatory responses with differential hippocampal impact. This study aimed to comprehensively compare the hippocampal transcriptomic profiles and validate key findings through independent experimentation. Transcriptomic datasets GSE253309 (CLP), GSE226120 (LPS), and GSE167610 (PCI) were retrieved from the GEO database. Bioinformatics analyses were employed to identify DEGs and enriched pathways. WGCNA pinpointed characteristic modules, and PPI networks were constructed and analyzed. Critically, an independent CLP-induced SAE mouse model was established, and hippocampal RNA sequencing was performed for confirmation. DEG analysis revealed 381, 533, and 85 significant DEGs in the CLP, LPS, and PCI datasets, respectively. CLP and LPS models shared a robust signature of neuroinflammation, significantly enriching GO terms related to immune response and inflammatory response, and KEGG pathways such as TNF, NF-κB, IL-17. In stark contrast, the PCI model was predominantly associated with cell migration, aldarate metabolism, and enriched in metabolic pathways, including bile secretion, ascorbate and aldarate metabolism. Cross-dataset analysis identified 29 common DEGs, from which a PPI network of 16 hub genes was constructed. Importantly, independent validation confirmed a strong concordance (r = 0.576) between the CLP-seq discovery cohort and the experimental CLP-seq data. Lcn2, S100a8, S100a9, Lrg1 and the TNF/IL-17 signaling pathways were robustly verified. CLP and LPS models demonstrate convergent hippocampal transcriptomic profiles distinct from PCI. Lcn2, S100a8, S100a9, Lrg1 and the TNF and IL-17 signaling pathways are highly reliable core features in SAE.</p>","PeriodicalId":19304,"journal":{"name":"NeuroMolecular Medicine","volume":"27 1","pages":"75"},"PeriodicalIF":3.9,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145636613","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Longikaurin A - Mediated Regulation of ROS/JNK Signaling Counteracts Epithelial-Mesenchymal Transition in Glioblastoma. Longikaurin A介导的ROS/JNK信号调控抑制胶质母细胞瘤上皮-间质转化。
IF 3.9 4区 医学 Q2 NEUROSCIENCES Pub Date : 2025-11-20 DOI: 10.1007/s12017-025-08897-3
Hua Chen, Yangming Mao, Xiang Chen, Xia Zhao

Glioblastoma (GBM) is a highly aggressive brain tumor characterized by rapid proliferation, therapy resistance, and extensive invasion, largely driven by epithelial-mesenchymal transition (EMT). Longikaurin A (LK-A), a natural kaurane diterpenoid, has demonstrated promising anti-cancer properties, yet its role in EMT regulation within GBM remains unclear. This study aimed to systematically investigate the inhibitory effects of LK-A on TGF-β1-induced EMT and to elucidate the underlying molecular mechanisms contributing to its anti-invasive potential in GBM. LK-A inhibited EMT-associated phenotypic changes, including reduced expression of mesenchymal markers (N-cadherin, Vimentin) and increased expression of epithelial markers (ZO-1, Occludin), alongside suppression of key EMT transcription factors (Snail, Twist1). Functionally, LK-A impaired EMT-induced cell migration, invasion, and glioma stem cell-like traits, evidenced by decreased gliosphere formation and downregulation of stemness markers such as Sox2 and Oct4. Mechanistic analyses revealed that LK-A triggered reactive oxygen species (ROS) accumulation, leading to the activation of the JNK/p38 MAPK signaling cascade. Pharmacological inhibition of JNK or ROS scavenging reversed the anti-EMT effects of LK-A, confirming that EMT suppression is mediated through ROS-dependent JNK activation. In vivo, LK-A significantly suppressed tumor growth, EMT marker expression, and stemness in xenograft models. Collectively, these findings identify LK-A as a potent regulator of EMT and glioma stemness via ROS/JNK signaling. This work provides new mechanistic insight into the anti-tumor effects of LK-A and highlights its potential as a promising therapeutic strategy for combating GBM aggressiveness.

胶质母细胞瘤(GBM)是一种高度侵袭性的脑肿瘤,主要由上皮-间质转化(EMT)驱动,具有快速增殖、耐药和广泛侵袭的特点。Longikaurin A (k -A)是一种天然的kaurane二萜,已被证明具有良好的抗癌特性,但其在GBM中EMT调控中的作用尚不清楚。本研究旨在系统探讨LK-A对TGF-β1诱导的EMT的抑制作用,并阐明其抗GBM侵袭潜力的潜在分子机制。LK-A抑制EMT相关的表型变化,包括间质标记物(N-cadherin, Vimentin)的表达减少,上皮标记物(ZO-1, Occludin)的表达增加,以及关键EMT转录因子(Snail, Twist1)的抑制。在功能上,LK-A损害了emt诱导的细胞迁移、侵袭和胶质瘤干细胞样性状,胶质层形成减少,Sox2和Oct4等干性标志物下调。机制分析显示LK-A触发活性氧(ROS)积累,导致JNK/p38 MAPK信号级联激活。药理抑制JNK或清除ROS可逆转LK-A的抗EMT作用,证实EMT抑制是通过ROS依赖性JNK激活介导的。在体内,LK-A显著抑制异种移植物模型的肿瘤生长、EMT标志物表达和干细胞性。总的来说,这些发现表明LK-A是通过ROS/JNK信号传导EMT和胶质瘤干细胞的有效调节剂。这项工作为LK-A的抗肿瘤作用提供了新的机制见解,并突出了其作为对抗GBM侵袭性的有前途的治疗策略的潜力。
{"title":"Longikaurin A - Mediated Regulation of ROS/JNK Signaling Counteracts Epithelial-Mesenchymal Transition in Glioblastoma.","authors":"Hua Chen, Yangming Mao, Xiang Chen, Xia Zhao","doi":"10.1007/s12017-025-08897-3","DOIUrl":"https://doi.org/10.1007/s12017-025-08897-3","url":null,"abstract":"<p><p>Glioblastoma (GBM) is a highly aggressive brain tumor characterized by rapid proliferation, therapy resistance, and extensive invasion, largely driven by epithelial-mesenchymal transition (EMT). Longikaurin A (LK-A), a natural kaurane diterpenoid, has demonstrated promising anti-cancer properties, yet its role in EMT regulation within GBM remains unclear. This study aimed to systematically investigate the inhibitory effects of LK-A on TGF-β1-induced EMT and to elucidate the underlying molecular mechanisms contributing to its anti-invasive potential in GBM. LK-A inhibited EMT-associated phenotypic changes, including reduced expression of mesenchymal markers (N-cadherin, Vimentin) and increased expression of epithelial markers (ZO-1, Occludin), alongside suppression of key EMT transcription factors (Snail, Twist1). Functionally, LK-A impaired EMT-induced cell migration, invasion, and glioma stem cell-like traits, evidenced by decreased gliosphere formation and downregulation of stemness markers such as Sox2 and Oct4. Mechanistic analyses revealed that LK-A triggered reactive oxygen species (ROS) accumulation, leading to the activation of the JNK/p38 MAPK signaling cascade. Pharmacological inhibition of JNK or ROS scavenging reversed the anti-EMT effects of LK-A, confirming that EMT suppression is mediated through ROS-dependent JNK activation. In vivo, LK-A significantly suppressed tumor growth, EMT marker expression, and stemness in xenograft models. Collectively, these findings identify LK-A as a potent regulator of EMT and glioma stemness via ROS/JNK signaling. This work provides new mechanistic insight into the anti-tumor effects of LK-A and highlights its potential as a promising therapeutic strategy for combating GBM aggressiveness.</p>","PeriodicalId":19304,"journal":{"name":"NeuroMolecular Medicine","volume":"27 1","pages":"74"},"PeriodicalIF":3.9,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145564536","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Glutamic Acid at Position 168 Is a Constitutive Activator of Tank Binding Kinase 1 Catalytic Function. 位置168的谷氨酸是罐结合激酶1催化功能的组成激活剂。
IF 3.9 4区 医学 Q2 NEUROSCIENCES Pub Date : 2025-11-05 DOI: 10.1007/s12017-025-08894-6
Noopur Bhore, Anubhuti Sarkar, Zhi Yao, Susanne Herbst, Patrick A Lewis

TANK binding kinase 1 (TBK1) is serine/threonine protein kinase member of the inhibitor of nuclear factor-kB kinase family, with links to the etiology of familial as well as idiopathic Amyotrophic Lateral Sclerosis. It contributes to several regulatory cellular processes such as autophagy, inflammation and apoptosis. Reduction or loss of TBK1 kinase activity is associated with increased risk of ALS, and so understanding the molecular basis of this activity is an important research priority. In this current study, the role of the E168 residue, located adjacent to the active site of TBK1, has been assessed using a combination of artificial and naturally occurring variants found at this codon - evaluated using multiple readouts for TBK1 kinase activity. The results suggest that the negative charge resulting from the presence of a glutamic acid at this codon is a constitutive activator of TBK1 activity.

TANK结合激酶1 (TBK1)是核因子- kb激酶家族抑制剂的丝氨酸/苏氨酸蛋白激酶成员,与家族性和特发性肌萎缩侧索硬化症的病因有关。它参与了一些调节细胞过程,如自噬、炎症和凋亡。TBK1激酶活性的降低或丧失与ALS的风险增加有关,因此了解这种活性的分子基础是一个重要的研究重点。在目前的研究中,位于TBK1活性位点附近的E168残基的作用已经通过在该密码子上发现的人工和自然变异的组合进行了评估,并使用TBK1激酶活性的多次读数进行了评估。结果表明,由于该密码子上存在谷氨酸而产生的负电荷是TBK1活性的组成激活因子。
{"title":"Glutamic Acid at Position 168 Is a Constitutive Activator of Tank Binding Kinase 1 Catalytic Function.","authors":"Noopur Bhore, Anubhuti Sarkar, Zhi Yao, Susanne Herbst, Patrick A Lewis","doi":"10.1007/s12017-025-08894-6","DOIUrl":"10.1007/s12017-025-08894-6","url":null,"abstract":"<p><p>TANK binding kinase 1 (TBK1) is serine/threonine protein kinase member of the inhibitor of nuclear factor-kB kinase family, with links to the etiology of familial as well as idiopathic Amyotrophic Lateral Sclerosis. It contributes to several regulatory cellular processes such as autophagy, inflammation and apoptosis. Reduction or loss of TBK1 kinase activity is associated with increased risk of ALS, and so understanding the molecular basis of this activity is an important research priority. In this current study, the role of the E168 residue, located adjacent to the active site of TBK1, has been assessed using a combination of artificial and naturally occurring variants found at this codon - evaluated using multiple readouts for TBK1 kinase activity. The results suggest that the negative charge resulting from the presence of a glutamic acid at this codon is a constitutive activator of TBK1 activity.</p>","PeriodicalId":19304,"journal":{"name":"NeuroMolecular Medicine","volume":"27 1","pages":"73"},"PeriodicalIF":3.9,"publicationDate":"2025-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12589330/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145445619","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
HDAC3 Epigenetic Suppression by Electroacupuncture Restores AMPA Receptor Function and Synaptic Plasticity in Alzheimer's Disease Models. 电针抑制HDAC3表观遗传可恢复阿尔茨海默病模型AMPA受体功能和突触可塑性
IF 3.9 4区 医学 Q2 NEUROSCIENCES Pub Date : 2025-11-04 DOI: 10.1007/s12017-025-08892-8
Hongzhu Li, Bing Deng, Mucan Lin, Lanfeng Lai, Jiaying Zhao, Yuemei Li

Alzheimer's disease (AD) is a progressive neurodegenerative condition marked by continuous cognitive deterioration, primarily resulting from the accumulation of amyloid-β (Aβ) plaques and tau-induced neurofibrillary tangles (NFTs). Recent studies have also highlighted histone deacetylase 3 (HDAC3) as a critical suppressor of synaptic plasticity. Although pharmacological inhibition of HDAC3 has been shown to facilitate long-term potentiation (LTP), the precise relationship between HDAC3 activity and AMPA receptor signaling, key components in LTP induction and maintenance, remains insufficiently understood. Electroacupuncture (EA), known to modulate epigenetic markers like H3K9/H3K27 acetylation and HDAC3/4 activity, may offer therapeutic potential by targeting these pathways. Here, we investigated EA's effects on AD-related pathology in APP/PS1 transgenic mice, focusing on HDAC3-AMPA receptor interactions in synaptic plasticity. Behavioral assays (Morris water maze) and electrophysiological recordings revealed that EA improved spatial learning ability and reinstated LTP in APP/PS1 transgenic mice. Mechanistically, EA reduced hippocampal HDAC3 expression while upregulating GluR1/GluR2 subunits and increasing acetylated H3K9K14/H3 levels, suggesting HDAC3-mediated transcriptional regulation of AMPA receptor genes. Co-immunoprecipitation assays further supported HDAC3's physical interaction with AMPA receptor components. Crucially, conditional knockout of HDAC3 in neurons rescued both LTP impairments and memory deficits, reinforcing its pivotal role in synaptic dysfunction. Our findings unveil a novel epigenetic mechanism whereby EA mitigates AD-associated synaptic damage by suppressing HDAC3 and enhancing AMPA receptor-dependent plasticity, highlighting HDAC3 as a promising therapeutic target for AD intervention.

阿尔茨海默病(AD)是一种进行性神经退行性疾病,其特征是持续的认知能力下降,主要是由于淀粉样蛋白-β (a β)斑块和tau诱导的神经原纤维缠结(nft)的积累。最近的研究也强调了组蛋白去乙酰化酶3 (HDAC3)是突触可塑性的关键抑制因子。尽管HDAC3的药理抑制已被证明可促进长期增强(LTP),但HDAC3活性与AMPA受体信号传导(LTP诱导和维持的关键成分)之间的确切关系仍未得到充分了解。已知电针(EA)可以调节表观遗传标记,如H3K9/H3K27乙酰化和HDAC3/4活性,可能通过靶向这些途径提供治疗潜力。在此,我们研究了EA对APP/PS1转基因小鼠ad相关病理的影响,重点关注HDAC3-AMPA受体在突触可塑性中的相互作用。行为学分析(Morris水迷宫)和电生理记录显示,EA提高了APP/PS1转基因小鼠的空间学习能力,恢复了LTP。在机制上,EA降低了海马HDAC3的表达,上调了GluR1/GluR2亚基,增加了乙酰化的H3K9K14/H3水平,提示HDAC3介导了AMPA受体基因的转录调节。共免疫沉淀实验进一步支持了HDAC3与AMPA受体组分的物理相互作用。至关重要的是,有条件地敲除神经元中的HDAC3可以挽救LTP损伤和记忆缺陷,从而加强其在突触功能障碍中的关键作用。我们的发现揭示了一种新的表观遗传机制,即EA通过抑制HDAC3和增强AMPA受体依赖的可塑性来减轻AD相关的突触损伤,强调HDAC3是AD干预的一个有希望的治疗靶点。
{"title":"HDAC3 Epigenetic Suppression by Electroacupuncture Restores AMPA Receptor Function and Synaptic Plasticity in Alzheimer's Disease Models.","authors":"Hongzhu Li, Bing Deng, Mucan Lin, Lanfeng Lai, Jiaying Zhao, Yuemei Li","doi":"10.1007/s12017-025-08892-8","DOIUrl":"https://doi.org/10.1007/s12017-025-08892-8","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is a progressive neurodegenerative condition marked by continuous cognitive deterioration, primarily resulting from the accumulation of amyloid-β (Aβ) plaques and tau-induced neurofibrillary tangles (NFTs). Recent studies have also highlighted histone deacetylase 3 (HDAC3) as a critical suppressor of synaptic plasticity. Although pharmacological inhibition of HDAC3 has been shown to facilitate long-term potentiation (LTP), the precise relationship between HDAC3 activity and AMPA receptor signaling, key components in LTP induction and maintenance, remains insufficiently understood. Electroacupuncture (EA), known to modulate epigenetic markers like H3K9/H3K27 acetylation and HDAC3/4 activity, may offer therapeutic potential by targeting these pathways. Here, we investigated EA's effects on AD-related pathology in APP/PS1 transgenic mice, focusing on HDAC3-AMPA receptor interactions in synaptic plasticity. Behavioral assays (Morris water maze) and electrophysiological recordings revealed that EA improved spatial learning ability and reinstated LTP in APP/PS1 transgenic mice. Mechanistically, EA reduced hippocampal HDAC3 expression while upregulating GluR1/GluR2 subunits and increasing acetylated H3K9K14/H3 levels, suggesting HDAC3-mediated transcriptional regulation of AMPA receptor genes. Co-immunoprecipitation assays further supported HDAC3's physical interaction with AMPA receptor components. Crucially, conditional knockout of HDAC3 in neurons rescued both LTP impairments and memory deficits, reinforcing its pivotal role in synaptic dysfunction. Our findings unveil a novel epigenetic mechanism whereby EA mitigates AD-associated synaptic damage by suppressing HDAC3 and enhancing AMPA receptor-dependent plasticity, highlighting HDAC3 as a promising therapeutic target for AD intervention.</p>","PeriodicalId":19304,"journal":{"name":"NeuroMolecular Medicine","volume":"27 1","pages":"72"},"PeriodicalIF":3.9,"publicationDate":"2025-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145438699","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Aurintricarboxylic Acid Attenuates Tramadol Withdrawal Syndrome Via TWEAK/FN14 Inhibition and CREB Modulation in Mice Model. 金羧酸通过TWEAK/FN14抑制和CREB调节减轻小鼠模型曲马多戒断综合征
IF 3.9 4区 医学 Q2 NEUROSCIENCES Pub Date : 2025-10-29 DOI: 10.1007/s12017-025-08893-7
Rajalaxmi, Shahid Nazir Wani, Amarjot Kaur Grewal, Varinder Singh, Amit Kumar, Heena Khan, Thakur Gurjeet Singh

Tramadol withdrawal presents a significant clinical challenge, characterized by neurobehavioral impairments linked to neuroinflammation, oxidative stress and neurotransmitter dysregulation. The TNF-like weak inducer of apoptosis (TWEAK)/fibroblast growth factor-inducer 14 (Fn14) pathway and downstream effectors like cAMP response element binding protein (CREB) are implicated, but effective targeted therapies are lacking. Aurintricarboxylic acid (ATA), a TWEAK inhibitor, exhibits neuroprotective potential. This study aims to evaluate the therapeutic efficacy of ATA in mitigating the tramadol withdrawal-induced neurobehavioral alterations in mice model, focusing on the role of TWEAK/Fn14 pathway and CREB phosphorylation. Swiss albino mice were subjected to chronic tramadol administration (50 mg/kg, s.c.) for 57 days, with withdrawal precipitated with naloxone (5 mg/kg, i.p.) on day 57. Behavioural assessments included withdrawal severity score (WSS), jumping frequency, and hyperalgesia. Biochemical analyses measured the level of oxidative stress markers (TBARS, SOD, GSH and catalase), inflammatory biomarkers (TNF-α, IL-6, IL-1β), and neurotransmitters (glutamate, dopamine and serotonin). ATA (5 mg/kg and 10 mg/kg i.p.) dose-dependently reduced the WSS, jumping frequency and hyperalgesia. It also mitigated the oxidative stress, neuroinflammation, and glutamate level, while restoring the neurotransmitter level. Notably, pretreatment with CREB inhibitor (666 - 15) (10 mg/kg i.p) significantly attenuated the protective effect of ATA, underscoring the pivotal role of CREB phosphorylation in its mechanism. Our findings demonstrate that ATA offers significant neuroprotection against tramadol withdrawal, primarily by inhibiting the TWEAK/Fn14 pathway and subsequently promoting the CREB phosphorylation. This study highlights ATA as a promising therapeutic candidate for managing tramadol withdrawal syndrome by targeting oxidative stress, neuroinflammation, and its downstream effectors.

曲马多戒断表现出显著的临床挑战,其特征是与神经炎症、氧化应激和神经递质失调相关的神经行为损伤。涉及到tnf样细胞凋亡弱诱导剂(TWEAK)/成纤维细胞生长因子诱导剂14 (Fn14)途径和下游效应物,如cAMP反应元件结合蛋白(CREB),但缺乏有效的靶向治疗。Aurintricarboxylic acid (ATA)是一种TWEAK抑制剂,具有神经保护作用。本研究旨在评价ATA在小鼠模型中减轻曲马多戒断诱导的神经行为改变的治疗效果,重点关注TWEAK/Fn14通路和CREB磷酸化的作用。给瑞士白化小鼠慢性曲马多(50 mg/kg, s.c) 57天,第57天纳洛酮(5 mg/kg, i.p)沉淀停药。行为评估包括戒断严重程度评分(WSS)、跳跃频率和痛觉过敏。生化分析测量氧化应激标志物(TBARS、SOD、GSH和过氧化氢酶)、炎症生物标志物(TNF-α、IL-6、IL-1β)和神经递质(谷氨酸、多巴胺和血清素)的水平。ATA (5mg /kg和10mg /kg i.p.)剂量依赖性地降低了WSS、跳频和痛觉过敏。它还能减轻氧化应激、神经炎症和谷氨酸水平,同时恢复神经递质水平。值得注意的是,CREB抑制剂(666 - 15)预处理(10 mg/kg i.p)显著减弱了ATA的保护作用,强调了CREB磷酸化在其机制中的关键作用。我们的研究结果表明,ATA对曲马多戒断具有显著的神经保护作用,主要是通过抑制TWEAK/Fn14途径,随后促进CREB磷酸化。这项研究强调了ATA作为一种有希望的治疗候选药物,通过靶向氧化应激、神经炎症及其下游效应来治疗曲马多戒断综合征。
{"title":"Aurintricarboxylic Acid Attenuates Tramadol Withdrawal Syndrome Via TWEAK/FN14 Inhibition and CREB Modulation in Mice Model.","authors":"Rajalaxmi, Shahid Nazir Wani, Amarjot Kaur Grewal, Varinder Singh, Amit Kumar, Heena Khan, Thakur Gurjeet Singh","doi":"10.1007/s12017-025-08893-7","DOIUrl":"https://doi.org/10.1007/s12017-025-08893-7","url":null,"abstract":"<p><p>Tramadol withdrawal presents a significant clinical challenge, characterized by neurobehavioral impairments linked to neuroinflammation, oxidative stress and neurotransmitter dysregulation. The TNF-like weak inducer of apoptosis (TWEAK)/fibroblast growth factor-inducer 14 (Fn14) pathway and downstream effectors like cAMP response element binding protein (CREB) are implicated, but effective targeted therapies are lacking. Aurintricarboxylic acid (ATA), a TWEAK inhibitor, exhibits neuroprotective potential. This study aims to evaluate the therapeutic efficacy of ATA in mitigating the tramadol withdrawal-induced neurobehavioral alterations in mice model, focusing on the role of TWEAK/Fn14 pathway and CREB phosphorylation. Swiss albino mice were subjected to chronic tramadol administration (50 mg/kg, s.c.) for 57 days, with withdrawal precipitated with naloxone (5 mg/kg, i.p.) on day 57. Behavioural assessments included withdrawal severity score (WSS), jumping frequency, and hyperalgesia. Biochemical analyses measured the level of oxidative stress markers (TBARS, SOD, GSH and catalase), inflammatory biomarkers (TNF-α, IL-6, IL-1β), and neurotransmitters (glutamate, dopamine and serotonin). ATA (5 mg/kg and 10 mg/kg i.p.) dose-dependently reduced the WSS, jumping frequency and hyperalgesia. It also mitigated the oxidative stress, neuroinflammation, and glutamate level, while restoring the neurotransmitter level. Notably, pretreatment with CREB inhibitor (666 - 15) (10 mg/kg i.p) significantly attenuated the protective effect of ATA, underscoring the pivotal role of CREB phosphorylation in its mechanism. Our findings demonstrate that ATA offers significant neuroprotection against tramadol withdrawal, primarily by inhibiting the TWEAK/Fn14 pathway and subsequently promoting the CREB phosphorylation. This study highlights ATA as a promising therapeutic candidate for managing tramadol withdrawal syndrome by targeting oxidative stress, neuroinflammation, and its downstream effectors.</p>","PeriodicalId":19304,"journal":{"name":"NeuroMolecular Medicine","volume":"27 1","pages":"71"},"PeriodicalIF":3.9,"publicationDate":"2025-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145401414","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Suppression of Glial Activation in Tau Transgenic Mice Through Inhibition of CRMP2 Phosphorylation: a Morphometric Analysis. 通过抑制CRMP2磷酸化抑制Tau转基因小鼠的胶质细胞活化:形态计量学分析。
IF 3.9 4区 医学 Q2 NEUROSCIENCES Pub Date : 2025-10-14 DOI: 10.1007/s12017-025-08891-9
Wanying Li, Toshiki Kubota, Valeria Ayala Guevara, Yoshio Goshima, Takaomi C Saido, Toshio Ohshima

Tauopathies, including Alzheimer's disease (AD), are neurodegenerative diseases characterized by abnormal tau aggregation in neurons and glial cells. Various tauopathy mouse models have been developed, including PS19, a tau-overexpressing transgenic mouse model with the P301S mutation, in which glial activation has been reported prior to the accumulation of tau. In this mouse model, tau pathology was improved by the administration of the immunosuppressant FKB506, suggesting that inflammatory responses promote the progression of tau pathology. Our previous studies have shown that the inhibition of Collapsin response mediator protein 2 (CRMP2) phosphorylation suppresses inflammation and ameliorates pathological progression in spinal cord injury and MPTP-induced Parkinson's disease models using CRMP2KI/KI mice, in which the phosphorylation site Ser522 was replaced with Ala. Therefore, we compared glial cell activation in male PS19 and PS19; CRMP2KI/KI mice using morphometric analysis at 5 months of age, before the onset of tau pathology. We found that the morphological changes caused by the activation of microglia and astrocytes were normalized by suppressing CRMP2 phosphorylation compared with those in PS19 mice. Cox-2 expression in hippocampal neurons was increased in PS19 mice, but this increase was suppressed in PS19; CRMP2KI/KI mice, suggesting that the suppression of CRMP2 phosphorylation in neurons is also involved in this process. These results suggest that the inhibition of CRMP2 phosphorylation may improve neuroinflammation in tauopathy.

tau病,包括阿尔茨海默病(AD),是一种神经退行性疾病,其特征是神经元和神经胶质细胞中的tau异常聚集。各种tau病小鼠模型已经被开发出来,包括PS19,一种带有P301S突变的tau过表达转基因小鼠模型,其中胶质细胞激活已被报道在tau积累之前。在该小鼠模型中,免疫抑制剂FKB506改善了tau病理,表明炎症反应促进了tau病理的进展。我们之前的研究表明,抑制坍缩反应介质蛋白2 (CRMP2)磷酸化可以抑制脊髓损伤和mptp诱导的帕金森病模型的炎症并改善病理进展,使用CRMP2KI/KI小鼠,其中磷酸化位点Ser522被Ala取代。因此,我们比较了雄性PS19和PS19的胶质细胞活化;使用形态计量学分析的CRMP2KI/KI小鼠在5个月大时,在tau病理发作之前。我们发现,与PS19小鼠相比,小胶质细胞和星形胶质细胞激活引起的形态学变化通过抑制CRMP2磷酸化而正常化。Cox-2在PS19小鼠海马神经元中的表达增加,但在PS19小鼠中这种增加受到抑制;CRMP2KI/KI小鼠,提示神经元中CRMP2磷酸化的抑制也参与了这一过程。这些结果表明,抑制CRMP2磷酸化可能改善牛头病的神经炎症。
{"title":"Suppression of Glial Activation in Tau Transgenic Mice Through Inhibition of CRMP2 Phosphorylation: a Morphometric Analysis.","authors":"Wanying Li, Toshiki Kubota, Valeria Ayala Guevara, Yoshio Goshima, Takaomi C Saido, Toshio Ohshima","doi":"10.1007/s12017-025-08891-9","DOIUrl":"10.1007/s12017-025-08891-9","url":null,"abstract":"<p><p>Tauopathies, including Alzheimer's disease (AD), are neurodegenerative diseases characterized by abnormal tau aggregation in neurons and glial cells. Various tauopathy mouse models have been developed, including PS19, a tau-overexpressing transgenic mouse model with the P301S mutation, in which glial activation has been reported prior to the accumulation of tau. In this mouse model, tau pathology was improved by the administration of the immunosuppressant FKB506, suggesting that inflammatory responses promote the progression of tau pathology. Our previous studies have shown that the inhibition of Collapsin response mediator protein 2 (CRMP2) phosphorylation suppresses inflammation and ameliorates pathological progression in spinal cord injury and MPTP-induced Parkinson's disease models using CRMP2KI/KI mice, in which the phosphorylation site Ser522 was replaced with Ala. Therefore, we compared glial cell activation in male PS19 and PS19; CRMP2KI/KI mice using morphometric analysis at 5 months of age, before the onset of tau pathology. We found that the morphological changes caused by the activation of microglia and astrocytes were normalized by suppressing CRMP2 phosphorylation compared with those in PS19 mice. Cox-2 expression in hippocampal neurons was increased in PS19 mice, but this increase was suppressed in PS19; CRMP2KI/KI mice, suggesting that the suppression of CRMP2 phosphorylation in neurons is also involved in this process. These results suggest that the inhibition of CRMP2 phosphorylation may improve neuroinflammation in tauopathy.</p>","PeriodicalId":19304,"journal":{"name":"NeuroMolecular Medicine","volume":"27 1","pages":"70"},"PeriodicalIF":3.9,"publicationDate":"2025-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12521301/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145286699","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Morpholino Knockdown in Zebrafish: A Tool to Investigate the Functional Impact of Variants of Unknown Significance in Mitochondrial Diseases. 斑马鱼的Morpholino敲低:研究线粒体疾病中未知意义变异的功能影响的工具。
IF 3.9 4区 医学 Q2 NEUROSCIENCES Pub Date : 2025-10-11 DOI: 10.1007/s12017-025-08890-w
Mateus Laranjeira, Jorge M A Oliveira, Filippo M Santorelli, Maria Marchese, Célia Nogueira

Mitochondrial diseases (MDs) are heterogeneous multisystemic disorders often caused by genetic defects in either nuclear or mitochondrial DNA. Although next-generation sequencing technologies have dramatically expanded the number of variants associated with these diseases, many remain variants of unknown significance (VUS). This review explores the utility of zebrafish (Danio rerio) as a vertebrate model system for studying mitochondrial dysfunction, with a focused analysis on the application of morpholino oligonucleotides (MOs) to functionally characterize and interpret VUS. MO-induced knockdown produces a transient suppression of target genes during zebrafish early development, recapitulating key MD phenotypes. Furthermore, rescue experiments involving co-injection of MO and either wild-type or mutant mRNA have proven useful to functionally assess the pathogenicity of specific variants. Specifically, while wild-type mRNA rescues the morphant phenotype, failure of mutant mRNA to do so confirms the variant's pathogenic effect. This approach has successfully linked previously uncharacterized genes to MD and helped reclassify ambiguous variants. The use of MO-based strategies in zebrafish remains a valuable tool for variant interpretation and functional validation, bridging the gap between genomic data and clinical action, and ultimately reducing the diagnostic odyssey. Overall, this review places MO knockdown and rescue assays in zebrafish as a robust and versatile platform to address functional genomics in MD research.

线粒体疾病(MDs)是一种异质性多系统疾病,通常由核或线粒体DNA的遗传缺陷引起。尽管新一代测序技术极大地增加了与这些疾病相关的变异数量,但仍有许多未知意义的变异(VUS)。本文探讨了斑马鱼(Danio rerio)作为研究线粒体功能障碍的脊椎动物模型系统的用途,重点分析了morpholino寡核苷酸(MOs)在VUS功能表征和解释中的应用。mo诱导的敲低在斑马鱼早期发育过程中产生靶基因的短暂抑制,重现关键的MD表型。此外,包括MO和野生型或突变型mRNA共同注射的救援实验已被证明有助于功能性评估特定变异的致病性。具体来说,虽然野生型mRNA挽救了变异表型,但突变mRNA未能做到这一点,证实了变异的致病作用。这种方法已经成功地将以前未表征的基因与MD联系起来,并帮助重新分类模棱两可的变异。在斑马鱼中使用基于mo的策略仍然是变异解释和功能验证的有价值的工具,弥合了基因组数据和临床行动之间的差距,并最终减少了诊断过程。总的来说,这篇综述将斑马鱼的MO敲除和拯救试验作为一个强大的和通用的平台,以解决医学研究中的功能基因组学问题。
{"title":"Morpholino Knockdown in Zebrafish: A Tool to Investigate the Functional Impact of Variants of Unknown Significance in Mitochondrial Diseases.","authors":"Mateus Laranjeira, Jorge M A Oliveira, Filippo M Santorelli, Maria Marchese, Célia Nogueira","doi":"10.1007/s12017-025-08890-w","DOIUrl":"https://doi.org/10.1007/s12017-025-08890-w","url":null,"abstract":"<p><p>Mitochondrial diseases (MDs) are heterogeneous multisystemic disorders often caused by genetic defects in either nuclear or mitochondrial DNA. Although next-generation sequencing technologies have dramatically expanded the number of variants associated with these diseases, many remain variants of unknown significance (VUS). This review explores the utility of zebrafish (Danio rerio) as a vertebrate model system for studying mitochondrial dysfunction, with a focused analysis on the application of morpholino oligonucleotides (MOs) to functionally characterize and interpret VUS. MO-induced knockdown produces a transient suppression of target genes during zebrafish early development, recapitulating key MD phenotypes. Furthermore, rescue experiments involving co-injection of MO and either wild-type or mutant mRNA have proven useful to functionally assess the pathogenicity of specific variants. Specifically, while wild-type mRNA rescues the morphant phenotype, failure of mutant mRNA to do so confirms the variant's pathogenic effect. This approach has successfully linked previously uncharacterized genes to MD and helped reclassify ambiguous variants. The use of MO-based strategies in zebrafish remains a valuable tool for variant interpretation and functional validation, bridging the gap between genomic data and clinical action, and ultimately reducing the diagnostic odyssey. Overall, this review places MO knockdown and rescue assays in zebrafish as a robust and versatile platform to address functional genomics in MD research.</p>","PeriodicalId":19304,"journal":{"name":"NeuroMolecular Medicine","volume":"27 1","pages":"69"},"PeriodicalIF":3.9,"publicationDate":"2025-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145275219","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Picroside II Alleviates the Progression of Alzheimer's Disease via the NLRP3/Caspase-1/GSDMD Pathway. Picroside II通过NLRP3/Caspase-1/GSDMD通路缓解阿尔茨海默病的进展
IF 3.9 4区 医学 Q2 NEUROSCIENCES Pub Date : 2025-10-10 DOI: 10.1007/s12017-025-08889-3
Shuyue Li, Ya Gao, Yidan Zhang, Jian Zhang, Yuan Zhao, Cui Chang, Xuan Gao, Jungang Zhang, Guofeng Yang

Alzheimer's disease (AD), an irreversible, degenerative disorder, affects the central nervous system. However, its accurate pathology remains unclear, and studies on treatment modalities are ongoing. Picroside II (PII) is an active compound in the medicinal herb Rhizoma coptis. It has strong effects, including antioxidation, anti-inflammatory, antiapoptotic, and neuroprotective effects. In this study, we analyzed how PII affects cognitive impairment in mice with AD and its underlying mechanism. PII at doses of 20 or 40 mg/kg was given to APP/PS1 mice through intraperitoneal injection for 2 months. Moreover, we carried out the Morris water maze test to evaluate cognitive function. Immunofluorescence analysis was performed to observe cortical Aβ plaque deposition, neuronal loss, and inflammatory cell expression. An enzyme-linked immunosorbent assay (ELISA) was performed to measure the levels of the cortical inflammatory factors tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β. Western blotting and quantitative polymerase chain reaction (qPCR) were performed to measure NLRP3, ASC, GSDMD, and caspase-1 expression. PII improved cognitive function, reduced Aβ plaque deposition and glial activation, and alleviated cortical neuronal loss in APP/PS1 mice. Furthermore, PII decreased the levels of cortical inflammatory factors (TNF-α, IL-6, and IL-1β). In addition, it suppressed NLRP3, ASC, GSDMD, and caspase-1 expression at the mRNA and protein levels. PII enhances the cognitive function of APP/PS1 mice by reducing inflammation and pyroptosis via the suppression of the NLRP3/caspase-1/GSDMD pathway. Therefore, PII is a candidate anti-AD therapeutic agent.

阿尔茨海默病(AD)是一种不可逆的退行性疾病,影响中枢神经系统。然而,其准确的病理尚不清楚,治疗方式的研究仍在进行中。Picroside II (PII)是中药黄连中的一种活性化合物。它具有很强的作用,包括抗氧化、抗炎、抗细胞凋亡和神经保护作用。在这项研究中,我们分析了PII如何影响AD小鼠的认知障碍及其潜在机制。APP/PS1小鼠腹腔注射PII,剂量分别为20或40 mg/kg,持续2个月。此外,我们还进行了Morris水迷宫测试来评估认知功能。免疫荧光分析观察皮层Aβ斑块沉积、神经元丢失和炎症细胞表达。采用酶联免疫吸附试验(ELISA)测定皮质炎性因子肿瘤坏死因子(TNF)-α、白细胞介素(IL)-6和IL-1β的水平。采用Western blotting和定量聚合酶链反应(qPCR)检测NLRP3、ASC、GSDMD和caspase-1的表达。PII可改善APP/PS1小鼠的认知功能,减少Aβ斑块沉积和神经胶质活化,减轻皮质神经元损失。此外,PII降低了皮质炎症因子(TNF-α, IL-6和IL-1β)的水平。此外,它在mRNA和蛋白水平上抑制NLRP3、ASC、GSDMD和caspase-1的表达。PII通过抑制NLRP3/caspase-1/GSDMD通路减少炎症和焦亡,从而增强APP/PS1小鼠的认知功能。因此,PII是一种候选的抗ad治疗剂。
{"title":"Picroside II Alleviates the Progression of Alzheimer's Disease via the NLRP3/Caspase-1/GSDMD Pathway.","authors":"Shuyue Li, Ya Gao, Yidan Zhang, Jian Zhang, Yuan Zhao, Cui Chang, Xuan Gao, Jungang Zhang, Guofeng Yang","doi":"10.1007/s12017-025-08889-3","DOIUrl":"10.1007/s12017-025-08889-3","url":null,"abstract":"<p><p>Alzheimer's disease (AD), an irreversible, degenerative disorder, affects the central nervous system. However, its accurate pathology remains unclear, and studies on treatment modalities are ongoing. Picroside II (PII) is an active compound in the medicinal herb Rhizoma coptis. It has strong effects, including antioxidation, anti-inflammatory, antiapoptotic, and neuroprotective effects. In this study, we analyzed how PII affects cognitive impairment in mice with AD and its underlying mechanism. PII at doses of 20 or 40 mg/kg was given to APP/PS1 mice through intraperitoneal injection for 2 months. Moreover, we carried out the Morris water maze test to evaluate cognitive function. Immunofluorescence analysis was performed to observe cortical Aβ plaque deposition, neuronal loss, and inflammatory cell expression. An enzyme-linked immunosorbent assay (ELISA) was performed to measure the levels of the cortical inflammatory factors tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β. Western blotting and quantitative polymerase chain reaction (qPCR) were performed to measure NLRP3, ASC, GSDMD, and caspase-1 expression. PII improved cognitive function, reduced Aβ plaque deposition and glial activation, and alleviated cortical neuronal loss in APP/PS1 mice. Furthermore, PII decreased the levels of cortical inflammatory factors (TNF-α, IL-6, and IL-1β). In addition, it suppressed NLRP3, ASC, GSDMD, and caspase-1 expression at the mRNA and protein levels. PII enhances the cognitive function of APP/PS1 mice by reducing inflammation and pyroptosis via the suppression of the NLRP3/caspase-1/GSDMD pathway. Therefore, PII is a candidate anti-AD therapeutic agent.</p>","PeriodicalId":19304,"journal":{"name":"NeuroMolecular Medicine","volume":"27 1","pages":"68"},"PeriodicalIF":3.9,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12513885/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145275362","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sex-Dependent Regional Vulnerability and Recovery Mechanisms in a Mouse Model of Transient Global Cerebral Ischemia. 小鼠短暂性全脑缺血模型中性别依赖的区域易损性和恢复机制。
IF 3.9 4区 医学 Q2 NEUROSCIENCES Pub Date : 2025-10-08 DOI: 10.1007/s12017-025-08887-5
Debiprasad Sinha, Roli Kushwaha, Shashikant Patel, Sainath Sunil Dhaygude, Mydhili Radhakrishnan, Papia Basuthakur, Arvind Kumar, Sumana Chakravarty

This study investigates the influence of sex on region-specific neural vulnerability following global cerebral ischemia using a Bilateral Common Carotid Artery Occlusion (BCCAo) mouse model that mimics severe ischemic brain stroke condition in humans. Comprehensive behavioral assessments, neuropathological analyses, and molecular profiling were conducted across multiple time points post-ischemia in male and female CD1 mice. Both sexes exhibited early motor deficits, cortical-striatal mitochondrial dysfunction, inflammation, and cell death at day 1, with gradual behavioral recovery. However, the hippocampus demonstrated a clear sex-specific divergence: males exhibited delayed yet prolonged inflammation, apoptotic cell death, and increased autophagy/mitophagy activity, while females were largely protected despite hypoxic and inflammatory gene expression. Molecular assays revealed prolonged upregulation of hypoxia-inducible factor 1α (HIF-1α), IL-1β, IL-6, TNF-α, and apoptotic markers in males, especially in the hippocampus, alongside increased expression of autophagy (Beclin-1, LC3-II, ATG7) and mitophagy (PINK1, BNIP3L) regulators and a shift in mitochondrial dynamics favoring fission.

本研究利用双侧颈总动脉闭塞(BCCAo)小鼠模型模拟人类严重缺血性脑卒中情况,研究性别对脑缺血后区域特异性神经易感性的影响。在雄性和雌性CD1小鼠缺血后的多个时间点进行综合行为评估、神经病理分析和分子谱分析。在第1天,两性都表现出早期运动障碍、皮质纹状体线粒体功能障碍、炎症和细胞死亡,行为逐渐恢复。然而,海马表现出明显的性别特异性差异:雄性表现出延迟但延长的炎症、凋亡细胞死亡和增加的自噬/有丝分裂活性,而雌性尽管缺氧和炎症基因表达,但在很大程度上受到保护。分子分析显示,在雄性中,特别是在海马中,低氧诱导因子1α (HIF-1α)、IL-1β、IL-6、TNF-α和凋亡标志物的持续上调,同时自噬(Beclin-1、LC3-II、ATG7)和线粒体自噬(PINK1、BNIP3L)调节因子的表达增加,线粒体动力学向有利于裂变的方向转变。
{"title":"Sex-Dependent Regional Vulnerability and Recovery Mechanisms in a Mouse Model of Transient Global Cerebral Ischemia.","authors":"Debiprasad Sinha, Roli Kushwaha, Shashikant Patel, Sainath Sunil Dhaygude, Mydhili Radhakrishnan, Papia Basuthakur, Arvind Kumar, Sumana Chakravarty","doi":"10.1007/s12017-025-08887-5","DOIUrl":"https://doi.org/10.1007/s12017-025-08887-5","url":null,"abstract":"<p><p>This study investigates the influence of sex on region-specific neural vulnerability following global cerebral ischemia using a Bilateral Common Carotid Artery Occlusion (BCCAo) mouse model that mimics severe ischemic brain stroke condition in humans. Comprehensive behavioral assessments, neuropathological analyses, and molecular profiling were conducted across multiple time points post-ischemia in male and female CD1 mice. Both sexes exhibited early motor deficits, cortical-striatal mitochondrial dysfunction, inflammation, and cell death at day 1, with gradual behavioral recovery. However, the hippocampus demonstrated a clear sex-specific divergence: males exhibited delayed yet prolonged inflammation, apoptotic cell death, and increased autophagy/mitophagy activity, while females were largely protected despite hypoxic and inflammatory gene expression. Molecular assays revealed prolonged upregulation of hypoxia-inducible factor 1α (HIF-1α), IL-1β, IL-6, TNF-α, and apoptotic markers in males, especially in the hippocampus, alongside increased expression of autophagy (Beclin-1, LC3-II, ATG7) and mitophagy (PINK1, BNIP3L) regulators and a shift in mitochondrial dynamics favoring fission.</p>","PeriodicalId":19304,"journal":{"name":"NeuroMolecular Medicine","volume":"27 1","pages":"67"},"PeriodicalIF":3.9,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145252180","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
NeuroMolecular Medicine
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