Pub Date : 2025-12-04DOI: 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.
{"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}
Pub Date : 2025-11-27DOI: 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.
{"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}
Pub Date : 2025-11-20DOI: 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}
Pub Date : 2025-11-05DOI: 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.
{"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}
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}
Pub Date : 2025-10-29DOI: 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.
{"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}
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.
{"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}
Pub Date : 2025-10-11DOI: 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.
{"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}
Pub Date : 2025-10-10DOI: 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}
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.
{"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}