Pub Date : 2025-08-14DOI: 10.1007/s10571-025-01599-1
Tong Shang, Binglin Kuang, Lei Zheng, Baiwen Zhang, Xueting Liu, Yaxin Shang, Jia Zheng, Baochun Luo, Wei Zou
Mitochondrial dysfunction has been identified as a key factor in the pathophysiological changes associated with intracerebral hemorrhage (ICH). As the core of intracellular energy metabolism, mitochondrial homeostasis is highly dependent on the precise regulation of its mitochondrial quality control (MtQC) system. After ICH, dysfunctional mitochondria lead to impaired oxidative phosphorylation and cellular bioenergetic stress, inducing oxidative stress, inflammatory responses, and programmed cell death, further exacerbating cellular damage. To counteract this injury, cells activate a series of MtQC mechanisms for compensatory repair, including mitochondrial dynamics, mitochondrial biogenesis, mitophagy, and intercellular mitochondrial transfer. These stringent mechanisms help maintain the mitochondrial network, restore the integrity of mitochondrial structural and functional integrity, improve neural function, and mitigate brain injury. In this review, we discuss key evidence regarding the role of mitochondrial dysfunction in ICH, focusing on the MtQC mechanisms involved in ICH. We also summarize potential therapeutic strategies targeting MtQC to intervene in ICH, providing valuable insights for clinical applications.
{"title":"Mitochondrial Quality Control: Insights into Intracerebral Hemorrhage.","authors":"Tong Shang, Binglin Kuang, Lei Zheng, Baiwen Zhang, Xueting Liu, Yaxin Shang, Jia Zheng, Baochun Luo, Wei Zou","doi":"10.1007/s10571-025-01599-1","DOIUrl":"10.1007/s10571-025-01599-1","url":null,"abstract":"<p><p>Mitochondrial dysfunction has been identified as a key factor in the pathophysiological changes associated with intracerebral hemorrhage (ICH). As the core of intracellular energy metabolism, mitochondrial homeostasis is highly dependent on the precise regulation of its mitochondrial quality control (MtQC) system. After ICH, dysfunctional mitochondria lead to impaired oxidative phosphorylation and cellular bioenergetic stress, inducing oxidative stress, inflammatory responses, and programmed cell death, further exacerbating cellular damage. To counteract this injury, cells activate a series of MtQC mechanisms for compensatory repair, including mitochondrial dynamics, mitochondrial biogenesis, mitophagy, and intercellular mitochondrial transfer. These stringent mechanisms help maintain the mitochondrial network, restore the integrity of mitochondrial structural and functional integrity, improve neural function, and mitigate brain injury. In this review, we discuss key evidence regarding the role of mitochondrial dysfunction in ICH, focusing on the MtQC mechanisms involved in ICH. We also summarize potential therapeutic strategies targeting MtQC to intervene in ICH, providing valuable insights for clinical applications.</p>","PeriodicalId":9742,"journal":{"name":"Cellular and Molecular Neurobiology","volume":"45 1","pages":"79"},"PeriodicalIF":4.8,"publicationDate":"2025-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12354944/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144854719","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 : 2025-08-11DOI: 10.1007/s10571-025-01597-3
Arpan Mukhoti, P K Annapoorna, Ashutosh Kumar, Pratishtha Wadnerkar, Ayesha Atqa Khan, Salil Saurav Pathak, Sumana Chakravarty, Arvind Kumar
Major depressive disorder (MDD) results from repeated and constant exposure to stress over prolonged periods. The highly variable response to stress and the low heritability suggests that MDD has a strong epigenetic basis. Studies show global dysregulation of histone modifications in both susceptible and resilient animals after chronic stress suggesting involvement of epigenetics in stress response in the brain. Given that the hippocampus and dentate gyrus (DG) show epigenetic changes in neurogenesis in Rodent models of stress that is known to be highly affected in MDD, we hypothesized that epigenetic changes might be involved in the advent of depressive phenotype during the progressive stress paradigm. To study the stress progression into the depression-like phenotype at the molecular level, we designed a novel progressive social defeat stress (PSDS) paradigm based on the popular chronic social defeat stress (CSDS) paradigm but involving only 5 days of defeat stress. Our molecular studies revealed consistent downregulation of H3K9me2 marks in the hippocampus and DG after the 4th day of stress while H3K27me2 showed an early upregulation in the hippocampus and a late downregulation after the 5th day of stress in the DG. In parallel, an early increase in phf8 and phf2 in hippocampus and DG, respectively, was observed. These findings of variable changes like repressive histone methylation marks and expression of corresponding demethylase genes after different durations of defeat stress, led to better understanding of the important role epigenetics play in stress progression into depression at molecular level in establishing resilient and susceptible phenotypes.
{"title":"Role of Repressive Histone Lysine Demethylases and Methylases in Susceptibility to Depression Using a Novel Progressive Social Defeat Stress Mouse Model.","authors":"Arpan Mukhoti, P K Annapoorna, Ashutosh Kumar, Pratishtha Wadnerkar, Ayesha Atqa Khan, Salil Saurav Pathak, Sumana Chakravarty, Arvind Kumar","doi":"10.1007/s10571-025-01597-3","DOIUrl":"10.1007/s10571-025-01597-3","url":null,"abstract":"<p><p>Major depressive disorder (MDD) results from repeated and constant exposure to stress over prolonged periods. The highly variable response to stress and the low heritability suggests that MDD has a strong epigenetic basis. Studies show global dysregulation of histone modifications in both susceptible and resilient animals after chronic stress suggesting involvement of epigenetics in stress response in the brain. Given that the hippocampus and dentate gyrus (DG) show epigenetic changes in neurogenesis in Rodent models of stress that is known to be highly affected in MDD, we hypothesized that epigenetic changes might be involved in the advent of depressive phenotype during the progressive stress paradigm. To study the stress progression into the depression-like phenotype at the molecular level, we designed a novel progressive social defeat stress (PSDS) paradigm based on the popular chronic social defeat stress (CSDS) paradigm but involving only 5 days of defeat stress. Our molecular studies revealed consistent downregulation of H3K9me2 marks in the hippocampus and DG after the 4th day of stress while H3K27me2 showed an early upregulation in the hippocampus and a late downregulation after the 5th day of stress in the DG. In parallel, an early increase in phf8 and phf2 in hippocampus and DG, respectively, was observed. These findings of variable changes like repressive histone methylation marks and expression of corresponding demethylase genes after different durations of defeat stress, led to better understanding of the important role epigenetics play in stress progression into depression at molecular level in establishing resilient and susceptible phenotypes.</p>","PeriodicalId":9742,"journal":{"name":"Cellular and Molecular Neurobiology","volume":"45 1","pages":"78"},"PeriodicalIF":4.8,"publicationDate":"2025-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12339800/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144815888","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 : 2025-08-04DOI: 10.1007/s10571-025-01568-8
Shafiul Haque, Darin M Mathkor, Mohd Wahid, Harshika Suri, Faraz Ahmad
Alzheimer's disease (AD) is one of the most common causes of dementia in elderly populations. A multifactorial and complex etiology has hindered the establishment of successful disease-modifying and retarding treatments. An important molecular target that has a close link with the disease's pathophysiology is glycogen synthase kinase 3β (GSK-3β). GSK-3β is thought to be an important bridge between amyloid and tau pathologies, the two principle pathogenic hallmarks of the disease. In particular, its kinase activity is thought to be a contributing factor for initiating aberrant tau hyperphosphorylation toward neurodegenerative progression. To identify potential inhibitors for GSK-3β, a pharmacophore-based virtual screening was used on the VITAS-M Lab database, a large database of small molecules. A co-crystal ligand employed as the template allowed the screening of roughly 200,000 compounds. Compounds successfully screened were selected on the basis of the Phase screen score combining vector alignments, volume scores, and site matching parameters. Using a cutoff score of 1.7, 174 compounds were docked using the Glide tool for molecular docking to further identify potential high-affinity binding ligands. Finally, four chemicals with the best binding scores (cutoff Glide GScore values of - 8 kcal/mol) were identified. Among these, 3-(2-oxo-2H-chromen-3-yl)-N-(4-sulfamoylphenyl) benzamide (VL-1) and trimethylsilyl trifluoromethanesulfonate (VL-2) showed strong and stable binding interactions, as evidenced by molecular dynamics simulation (MDS). The results suggest that VL-1 and VL-2 may serve as promising lead compounds for GSK-3β-based anti-AD therapeutics. However, further in vivo mechanistic validation is warrantied to confirm their therapeutic applicability.
{"title":"Identification of Novel Scaffolds Against GSK-3β for Targeting Alzheimer's Disease Through Molecular Modeling Techniques.","authors":"Shafiul Haque, Darin M Mathkor, Mohd Wahid, Harshika Suri, Faraz Ahmad","doi":"10.1007/s10571-025-01568-8","DOIUrl":"10.1007/s10571-025-01568-8","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is one of the most common causes of dementia in elderly populations. A multifactorial and complex etiology has hindered the establishment of successful disease-modifying and retarding treatments. An important molecular target that has a close link with the disease's pathophysiology is glycogen synthase kinase 3β (GSK-3β). GSK-3β is thought to be an important bridge between amyloid and tau pathologies, the two principle pathogenic hallmarks of the disease. In particular, its kinase activity is thought to be a contributing factor for initiating aberrant tau hyperphosphorylation toward neurodegenerative progression. To identify potential inhibitors for GSK-3β, a pharmacophore-based virtual screening was used on the VITAS-M Lab database, a large database of small molecules. A co-crystal ligand employed as the template allowed the screening of roughly 200,000 compounds. Compounds successfully screened were selected on the basis of the Phase screen score combining vector alignments, volume scores, and site matching parameters. Using a cutoff score of 1.7, 174 compounds were docked using the Glide tool for molecular docking to further identify potential high-affinity binding ligands. Finally, four chemicals with the best binding scores (cutoff Glide GScore values of - 8 kcal/mol) were identified. Among these, 3-(2-oxo-2H-chromen-3-yl)-N-(4-sulfamoylphenyl) benzamide (VL-1) and trimethylsilyl trifluoromethanesulfonate (VL-2) showed strong and stable binding interactions, as evidenced by molecular dynamics simulation (MDS). The results suggest that VL-1 and VL-2 may serve as promising lead compounds for GSK-3β-based anti-AD therapeutics. However, further in vivo mechanistic validation is warrantied to confirm their therapeutic applicability.</p>","PeriodicalId":9742,"journal":{"name":"Cellular and Molecular Neurobiology","volume":"45 1","pages":"77"},"PeriodicalIF":4.8,"publicationDate":"2025-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12321713/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144783615","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 : 2025-07-31DOI: 10.1007/s10571-025-01587-5
Alireza Sharafshah, Majid Motovali-Bashi, Kenneth Blum, Kai-Uwe Lewandrowski, Mark S Gold, Parvaneh Keshavarz, Panayotis K Thanos
This study aimed to integrate genome-wide association studies (GWAS) with pharmacogenomics data to develop personalized pain and inflammatory therapeutics. Despite recent developments in the clinical utilities of pharmacogenomics, it needs more investigations for uncovering the complicated mechanisms of drugs from a genetic standpoint. The research addresses the increasing misuse of opioids during recovery, emphasizing personalized interventions for opioid use disorder (OUD). Key pain-related pathways were analyzed to uncover their interactions. Five GWAS traits, including pain, inflammatory biomarkers, immune system abnormalities, and opioid-related traits, were examined. Candidate genes extracted from GWAS datasets were refined through in silico analyses, including protein-protein interactions (PPIs), TF-miRNA coregulatory interactions, enrichment analysis (EA), and clustering enrichment analysis (CEA). A network of 50 highly connected genes was identified, with APOE emerging as a top candidate due to its role in cholesterol metabolism and opioid-induced lipid effects. Pharmacogenomics analysis highlighted significant gene annotations, including OPRM1, DRD2, APOE, GRIN2B, and GPR98, linking them to opioid dependence, neurological disorders, and lipid traits. Protein interaction analyses further validated these connections, with implications for epigenetic repair. Our findings reveal a strong association between APOE, opioid use, and Alzheimer's disease, suggesting potential for novel recovery strategies. Combining HDL-boosting drugs with pro-dopaminergic regulators like KB220 may help prevent relapse. This study underscores the importance of integrating genetic and pharmacogenomic data to advance personalized therapies.
{"title":"A GWAS Meta-meta-analysis and In-depth Silico Pharmacogenomic Investigations in Identification of APOE and Other Genes Associated with Pain, Anti-inflammatory, and Immunomodulating Agents in Opioid Use Disorder (OUD) Derived from 14.91 M Subjects.","authors":"Alireza Sharafshah, Majid Motovali-Bashi, Kenneth Blum, Kai-Uwe Lewandrowski, Mark S Gold, Parvaneh Keshavarz, Panayotis K Thanos","doi":"10.1007/s10571-025-01587-5","DOIUrl":"10.1007/s10571-025-01587-5","url":null,"abstract":"<p><p>This study aimed to integrate genome-wide association studies (GWAS) with pharmacogenomics data to develop personalized pain and inflammatory therapeutics. Despite recent developments in the clinical utilities of pharmacogenomics, it needs more investigations for uncovering the complicated mechanisms of drugs from a genetic standpoint. The research addresses the increasing misuse of opioids during recovery, emphasizing personalized interventions for opioid use disorder (OUD). Key pain-related pathways were analyzed to uncover their interactions. Five GWAS traits, including pain, inflammatory biomarkers, immune system abnormalities, and opioid-related traits, were examined. Candidate genes extracted from GWAS datasets were refined through in silico analyses, including protein-protein interactions (PPIs), TF-miRNA coregulatory interactions, enrichment analysis (EA), and clustering enrichment analysis (CEA). A network of 50 highly connected genes was identified, with APOE emerging as a top candidate due to its role in cholesterol metabolism and opioid-induced lipid effects. Pharmacogenomics analysis highlighted significant gene annotations, including OPRM1, DRD2, APOE, GRIN2B, and GPR98, linking them to opioid dependence, neurological disorders, and lipid traits. Protein interaction analyses further validated these connections, with implications for epigenetic repair. Our findings reveal a strong association between APOE, opioid use, and Alzheimer's disease, suggesting potential for novel recovery strategies. Combining HDL-boosting drugs with pro-dopaminergic regulators like KB220 may help prevent relapse. This study underscores the importance of integrating genetic and pharmacogenomic data to advance personalized therapies.</p>","PeriodicalId":9742,"journal":{"name":"Cellular and Molecular Neurobiology","volume":"45 1","pages":"76"},"PeriodicalIF":4.8,"publicationDate":"2025-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12314298/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144752528","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 : 2025-07-26DOI: 10.1007/s10571-025-01595-5
Hany E Marei
Recent developments in neural circuit mapping and neurotherapy are changing our understanding of the dynamic network structure of the brain and offering new treatment options. In many neurological and psychiatric diseases, targeted control of specific brain circuits has proven to be a successful strategy to reduce cognitive, behavioral, and motor abnormalities. Sophisticated retrograde tracing techniques, transcranial magnetic stimulation (TMS), chemogenetics, optogenetics, and other technologies have greatly improved our ability to outline, observe, and control neuronal circuits with remarkable accuracy. These sophisticated techniques have revealed crucial information on neuroplasticity, circuit remodeling following injury, and the therapeutic potential of neuromodulatory interventions. Disorders include depression, anxiety, stroke, and neurodegenerative diseases are treated using techniques such as optogenetic stimulation, chemogenetic activation, and non-invasive brain stimulation to restore circuit function. Emerging multifunctional probes like Tetracysteine Display of Optogenetic Elements (Tetro-DOpE) provide real-time monitoring and modification of neuronal populations, improving circuit-level interventions' precision. At the same time, especially following severe brain injury and neurodegeneration, stem cell treatments combined with neurogenesis-promoting strategies show great promise in increasing circuit repair and functional recovery. The development of drug delivery methods like tailored nanoparticle systems and multifunctional probes is helping to improve the accuracy and safety of treatments by reducing off-target effects. These developments taken together draw attention to a notable shift toward precision neuromedicine. These techniques are meant to offer more efficient, focused, and specialized treatments for various neurological and psychiatric diseases by combining sophisticated circuit mapping with tailored therapeutic interventions.
神经回路制图和神经疗法的最新发展正在改变我们对大脑动态网络结构的理解,并提供新的治疗选择。在许多神经和精神疾病中,有针对性地控制特定的脑回路已被证明是减少认知、行为和运动异常的成功策略。复杂的逆行追踪技术、经颅磁刺激(TMS)、化学遗传学、光遗传学和其他技术极大地提高了我们以惊人的精度勾勒、观察和控制神经元回路的能力。这些复杂的技术揭示了神经可塑性、损伤后的电路重塑以及神经调节干预的治疗潜力的重要信息。包括抑郁、焦虑、中风和神经退行性疾病在内的疾病,使用光遗传刺激、化学发生激活和非侵入性脑刺激等技术来恢复电路功能。新兴的多功能探针如Tetracysteine Display of Optogenetic Elements (Tetro-DOpE)提供了对神经元群的实时监测和修改,提高了电路级干预的精度。同时,特别是在严重的脑损伤和神经变性后,干细胞治疗结合神经发生促进策略在增加电路修复和功能恢复方面显示出很大的希望。量身定制的纳米颗粒系统和多功能探针等药物输送方法的发展,通过减少脱靶效应,有助于提高治疗的准确性和安全性。这些进展加在一起,引起了人们对精确神经医学的显著转变的关注。这些技术旨在通过将复杂的电路测绘与量身定制的治疗干预相结合,为各种神经和精神疾病提供更有效、更集中和更专业的治疗。
{"title":"Neural Circuit Mapping and Neurotherapy-Based Strategies.","authors":"Hany E Marei","doi":"10.1007/s10571-025-01595-5","DOIUrl":"10.1007/s10571-025-01595-5","url":null,"abstract":"<p><p>Recent developments in neural circuit mapping and neurotherapy are changing our understanding of the dynamic network structure of the brain and offering new treatment options. In many neurological and psychiatric diseases, targeted control of specific brain circuits has proven to be a successful strategy to reduce cognitive, behavioral, and motor abnormalities. Sophisticated retrograde tracing techniques, transcranial magnetic stimulation (TMS), chemogenetics, optogenetics, and other technologies have greatly improved our ability to outline, observe, and control neuronal circuits with remarkable accuracy. These sophisticated techniques have revealed crucial information on neuroplasticity, circuit remodeling following injury, and the therapeutic potential of neuromodulatory interventions. Disorders include depression, anxiety, stroke, and neurodegenerative diseases are treated using techniques such as optogenetic stimulation, chemogenetic activation, and non-invasive brain stimulation to restore circuit function. Emerging multifunctional probes like Tetracysteine Display of Optogenetic Elements (Tetro-DOpE) provide real-time monitoring and modification of neuronal populations, improving circuit-level interventions' precision. At the same time, especially following severe brain injury and neurodegeneration, stem cell treatments combined with neurogenesis-promoting strategies show great promise in increasing circuit repair and functional recovery. The development of drug delivery methods like tailored nanoparticle systems and multifunctional probes is helping to improve the accuracy and safety of treatments by reducing off-target effects. These developments taken together draw attention to a notable shift toward precision neuromedicine. These techniques are meant to offer more efficient, focused, and specialized treatments for various neurological and psychiatric diseases by combining sophisticated circuit mapping with tailored therapeutic interventions.</p>","PeriodicalId":9742,"journal":{"name":"Cellular and Molecular Neurobiology","volume":"45 1","pages":"75"},"PeriodicalIF":4.8,"publicationDate":"2025-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12297084/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144717633","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 : 2025-07-22DOI: 10.1007/s10571-025-01596-4
Rastegar Hoseini, Elnaz Ghafari
Multiple Sclerosis (MS) is a chronic, inflammatory, and neurodegenerative disease of the Central Nervous System (CNS) that is characterized by immune dysregulation and neuroinflammation. Owing to the generation of neuroactive metabolites, the kynurenine pathway (KP), one of the key pathways of tryptophan metabolism, influences the pathogenesis of MS by regulating immune responses and neuronal homeostasis. KP dysregulation results in the overproduction of neurotoxic metabolites such as quinolinic acid (QUIN), characterized by the loss of homeostasis between the neuroprotective (e.g., kynurenic acid, KYNA) and neurotoxic (e.g., QUIN) metabolites, contributing to neuroinflammation, excitotoxicity, and neurodegeneration. Recent evidence suggests that exercise may serve as a non-pharmacological intervention to modulate KP and limit MS progression. Both acute and chronic exercise, especially high-intensity interval training (HIIT), have been demonstrated to decrease the systemic levels of these neurotoxic KP metabolites and increase the neuroprotective KYNA production. Through the modulation of cytokine profiles toward an anti-inflammatory response and Aryl Hydrocarbon Receptor (AhR) activation that promotes immune tolerance, exercise is also an important regulator of the immune response. These findings imply that exercise normalizes KP homeostasis, decreases neuro-axonal damage and improves neuroprotection in MS, but the mechanisms of exercise-induced KP regulation as well as its long-term therapeutic role in MS treatment need further investigation. This review highlights the therapeutic potential of exercise as a complementary approach to existing drugs to ameliorate neuroinflammation and neurodegeneration in MS.
{"title":"Kynurenine Pathway Modulation by Exercise in Multiple Sclerosis: Implications for Neuroprotection and Inflammation.","authors":"Rastegar Hoseini, Elnaz Ghafari","doi":"10.1007/s10571-025-01596-4","DOIUrl":"10.1007/s10571-025-01596-4","url":null,"abstract":"<p><p>Multiple Sclerosis (MS) is a chronic, inflammatory, and neurodegenerative disease of the Central Nervous System (CNS) that is characterized by immune dysregulation and neuroinflammation. Owing to the generation of neuroactive metabolites, the kynurenine pathway (KP), one of the key pathways of tryptophan metabolism, influences the pathogenesis of MS by regulating immune responses and neuronal homeostasis. KP dysregulation results in the overproduction of neurotoxic metabolites such as quinolinic acid (QUIN), characterized by the loss of homeostasis between the neuroprotective (e.g., kynurenic acid, KYNA) and neurotoxic (e.g., QUIN) metabolites, contributing to neuroinflammation, excitotoxicity, and neurodegeneration. Recent evidence suggests that exercise may serve as a non-pharmacological intervention to modulate KP and limit MS progression. Both acute and chronic exercise, especially high-intensity interval training (HIIT), have been demonstrated to decrease the systemic levels of these neurotoxic KP metabolites and increase the neuroprotective KYNA production. Through the modulation of cytokine profiles toward an anti-inflammatory response and Aryl Hydrocarbon Receptor (AhR) activation that promotes immune tolerance, exercise is also an important regulator of the immune response. These findings imply that exercise normalizes KP homeostasis, decreases neuro-axonal damage and improves neuroprotection in MS, but the mechanisms of exercise-induced KP regulation as well as its long-term therapeutic role in MS treatment need further investigation. This review highlights the therapeutic potential of exercise as a complementary approach to existing drugs to ameliorate neuroinflammation and neurodegeneration in MS.</p>","PeriodicalId":9742,"journal":{"name":"Cellular and Molecular Neurobiology","volume":"45 1","pages":"74"},"PeriodicalIF":4.8,"publicationDate":"2025-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12283535/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144689035","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 : 2025-07-20DOI: 10.1007/s10571-025-01593-7
Zhanhua Shi, Kelong Chen, Yin Wang, Haixia Du
Ischemic stroke is a common cerebrovascular disease accompanied by a large number of neuronal death and severe functional impairment. In recent years, the role of ferroptosis and ferritinophagy in neuronal death after cerebral infarction has attracted great interest in the field of ischemic stroke. Ferroptosis is a newly discovered programmed cell death pattern characterized by iron overload, dysregulation of the xCT/GSH/GPX4 system, and lipid peroxidation system, which is closely associated with neurological damage after ischemic stroke. Ferritinophagy is a selective autophagy mediated by NCOA4 that regulates intracellular iron metabolism, and can be regulated by factors such as intracellular iron content and HERC2-FBXL5-IPR2 axis. Under normal physiological conditions, ferritinophagy maintains the balance of intracellular iron elements, and excessive activation can cause ferroptosis. Here, we mainly review the general mechanisms of ferroptosis and ferritinophagy, and focus on the relationship between ischemic stroke and ferroptosis/ferritinophagy. Specifically, we explored the crosstalk of ferroptosis and ferritinophagy in ischemic stroke and outlined current treatment strategies and key challenges. These observations may help to further understand the pathological events of ischemic stroke and bridge the gap between basic and translational research to provide novel insights for its treatment.
{"title":"The Crosstalk Between Ferritinophagy and Ferroptosis in Ischemic Stroke: Regulatory Mechanisms and Therapeutic Implications.","authors":"Zhanhua Shi, Kelong Chen, Yin Wang, Haixia Du","doi":"10.1007/s10571-025-01593-7","DOIUrl":"10.1007/s10571-025-01593-7","url":null,"abstract":"<p><p>Ischemic stroke is a common cerebrovascular disease accompanied by a large number of neuronal death and severe functional impairment. In recent years, the role of ferroptosis and ferritinophagy in neuronal death after cerebral infarction has attracted great interest in the field of ischemic stroke. Ferroptosis is a newly discovered programmed cell death pattern characterized by iron overload, dysregulation of the xCT/GSH/GPX4 system, and lipid peroxidation system, which is closely associated with neurological damage after ischemic stroke. Ferritinophagy is a selective autophagy mediated by NCOA4 that regulates intracellular iron metabolism, and can be regulated by factors such as intracellular iron content and HERC2-FBXL5-IPR2 axis. Under normal physiological conditions, ferritinophagy maintains the balance of intracellular iron elements, and excessive activation can cause ferroptosis. Here, we mainly review the general mechanisms of ferroptosis and ferritinophagy, and focus on the relationship between ischemic stroke and ferroptosis/ferritinophagy. Specifically, we explored the crosstalk of ferroptosis and ferritinophagy in ischemic stroke and outlined current treatment strategies and key challenges. These observations may help to further understand the pathological events of ischemic stroke and bridge the gap between basic and translational research to provide novel insights for its treatment.</p>","PeriodicalId":9742,"journal":{"name":"Cellular and Molecular Neurobiology","volume":"45 1","pages":"73"},"PeriodicalIF":4.8,"publicationDate":"2025-07-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12277238/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144667279","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 : 2025-07-19DOI: 10.1007/s10571-025-01582-w
Wei-Sheng Lin, Pei-Yu Wang, Sheng-Rong Yeh, Zoe Lai, Andrew Chengyu Lee, Shou-Zen Fan
Depression during pregnancy is often overlooked and undertreated. Ketamine has been shown to exert prompt and sustained antidepressant effects in patients with depression, although concerns of potential neurotoxicity prohibit its use in pregnant women. Here, we aim to investigate the neurobehavioral effects of subanesthetic ketamine on pregnant mice and their offspring. We found that pregnant C57BL/6 mice receiving ketamine (10 mg/kg/day intraperitoneal) from gestation day 15 to 17 exhibited less depression-like behaviors. Prenatal ketamine treatment induced male-specific reduction in depression- and anxiety-like behaviors in adult offspring, without alterations in social and memory performance. These behavioral outcomes were associated with a male-specific increase in dendritic spine density of dentate gyrus granule cells, while neither dendritic architecture nor hippocampal neurogenesis was affected. N-methyl-D-aspartate receptor subunits GluN2A and GluN3A were expressed at significantly higher levels in the hippocampus of male as compared to female mouse embryos, suggesting sex-dependent actions of ketamine on developing brain. Overall, our study showed that prenatal exposure to subanesthetic ketamine could exert long-lasting neurobehavioral effects in a sex-dependent manner, with male offspring being more resilient to stress. These findings may have implications concerning ketamine use during pregnancy, and also provide clues about the developmental origins of emotional problems.
{"title":"Sex-Specific Neuropsychiatric Effects of Subanesthetic Ketamine Exposure in Pregnant Mice and Their Offspring.","authors":"Wei-Sheng Lin, Pei-Yu Wang, Sheng-Rong Yeh, Zoe Lai, Andrew Chengyu Lee, Shou-Zen Fan","doi":"10.1007/s10571-025-01582-w","DOIUrl":"10.1007/s10571-025-01582-w","url":null,"abstract":"<p><p>Depression during pregnancy is often overlooked and undertreated. Ketamine has been shown to exert prompt and sustained antidepressant effects in patients with depression, although concerns of potential neurotoxicity prohibit its use in pregnant women. Here, we aim to investigate the neurobehavioral effects of subanesthetic ketamine on pregnant mice and their offspring. We found that pregnant C57BL/6 mice receiving ketamine (10 mg/kg/day intraperitoneal) from gestation day 15 to 17 exhibited less depression-like behaviors. Prenatal ketamine treatment induced male-specific reduction in depression- and anxiety-like behaviors in adult offspring, without alterations in social and memory performance. These behavioral outcomes were associated with a male-specific increase in dendritic spine density of dentate gyrus granule cells, while neither dendritic architecture nor hippocampal neurogenesis was affected. N-methyl-D-aspartate receptor subunits GluN2A and GluN3A were expressed at significantly higher levels in the hippocampus of male as compared to female mouse embryos, suggesting sex-dependent actions of ketamine on developing brain. Overall, our study showed that prenatal exposure to subanesthetic ketamine could exert long-lasting neurobehavioral effects in a sex-dependent manner, with male offspring being more resilient to stress. These findings may have implications concerning ketamine use during pregnancy, and also provide clues about the developmental origins of emotional problems.</p>","PeriodicalId":9742,"journal":{"name":"Cellular and Molecular Neurobiology","volume":"45 1","pages":"72"},"PeriodicalIF":4.8,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12276194/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144667278","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 : 2025-07-18DOI: 10.1007/s10571-025-01585-7
Prayash Paudel, Asutosh Sah, Poonam Paudel
Myasthenia gravis (MG) is an autoimmune neuromuscular disorder characterized by fluctuating muscle weakness. MicroRNAs (miRNAs) have emerged as potential biomarkers for MG diagnosis, offering noninvasive and reliable detection. This systematic review and meta-analysis evaluated the diagnostic accuracy of miRNAs in MG. A comprehensive search of PubMed, Embase, and Google Scholar was conducted up to March 9, 2025. Eligible studies assessing miRNAs as MG biomarkers were selected on the basis of predefined criteria. Pooled sensitivity, specificity, and diagnostic odds ratios (DORs) were calculated via random effects model. Heterogeneity was assessed via I2, and publication bias was evaluated via Deeks' funnel plot. Nine studies including 1,797 participants were analysed. The pooled sensitivity and specificity were 0.80 (95% CI: 0.75-0.84) and 0.71 (95% CI: 0.65-0.77), respectively, with an area under the curve (AUC) of 0.83. Bivariate heterogeneity analysis indicated moderate variability, the cause of which were identified using subgroup analysis with region, clinical subtypes and seropositivity as subgroups. miRNAs demonstrate strong diagnostic potential for MG, with good sensitivity and specificity. However, standardized methodologies and further validation in large, multicentre studies is warranted.
{"title":"MicroRNAs as Diagnostic Biomarkers of Myasthenia Gravis: A Systematic Review and Meta-Analysis.","authors":"Prayash Paudel, Asutosh Sah, Poonam Paudel","doi":"10.1007/s10571-025-01585-7","DOIUrl":"10.1007/s10571-025-01585-7","url":null,"abstract":"<p><p>Myasthenia gravis (MG) is an autoimmune neuromuscular disorder characterized by fluctuating muscle weakness. MicroRNAs (miRNAs) have emerged as potential biomarkers for MG diagnosis, offering noninvasive and reliable detection. This systematic review and meta-analysis evaluated the diagnostic accuracy of miRNAs in MG. A comprehensive search of PubMed, Embase, and Google Scholar was conducted up to March 9, 2025. Eligible studies assessing miRNAs as MG biomarkers were selected on the basis of predefined criteria. Pooled sensitivity, specificity, and diagnostic odds ratios (DORs) were calculated via random effects model. Heterogeneity was assessed via I<sup>2</sup>, and publication bias was evaluated via Deeks' funnel plot. Nine studies including 1,797 participants were analysed. The pooled sensitivity and specificity were 0.80 (95% CI: 0.75-0.84) and 0.71 (95% CI: 0.65-0.77), respectively, with an area under the curve (AUC) of 0.83. Bivariate heterogeneity analysis indicated moderate variability, the cause of which were identified using subgroup analysis with region, clinical subtypes and seropositivity as subgroups. miRNAs demonstrate strong diagnostic potential for MG, with good sensitivity and specificity. However, standardized methodologies and further validation in large, multicentre studies is warranted.</p>","PeriodicalId":9742,"journal":{"name":"Cellular and Molecular Neurobiology","volume":"45 1","pages":"71"},"PeriodicalIF":4.8,"publicationDate":"2025-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12274162/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144658541","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 : 2025-07-16DOI: 10.1007/s10571-025-01586-6
Sharon Mariam Abraham, Sneha Suresh, Pragya Komal
The multifunctional roles of alpha7 nicotinic acetylcholine receptors (α7nAChRs), ranging from cognitive enhancement, neuroprotection, and anti-inflammatory action, credit tagging this receptor as "unique" among the cholinergic receptor family. The uniqueness of α7nAChRs in neuronal function and communication lies in their high calcium permeability among the cholinergic receptor family. The ionotropic function of α7nAChRs is governed by protein kinases' post-translational modification (PTMs), which alter their expression and function, affecting neuronal communication. A decrease in the ionotropic function of α7nAChRs and its downstream signaling pathways is observed across many neurologic disorders. The loss of α7nAChRs, decreased cholinergic function, and increased acetylcholinesterase levels are commonly associated with neuronal degeneration, cognitive impairment, and decreased memory function. An extensive body of evidence suggests the cognitive benefits of simple nutraceutical supplementation, Vitamin D3 (VD), in many neurologic disorders (Skv et al. in Mol Neurobiol 61:7211-7238, 2024). The present review will, however, focus on recent and past evidence deciphering the unique properties of α7nAChRs crucial for brain function. We have also emphasized on the therapeutic benefits of VD supplementation in restoring cholinergic neurotransmission and α7nAChRs expression in various neuropsychiatric and neurologic disorders.
α7烟碱乙酰胆碱受体(α7nAChRs)的多功能作用,包括认知增强、神经保护和抗炎作用,使其在胆碱能受体家族中具有“独特”的地位。α7nAChRs在神经元功能和通讯中的独特之处在于其在胆碱能受体家族中的高钙通透性。α7nAChRs的亲离子功能受蛋白激酶的翻译后修饰(PTMs)调控,PTMs改变了α7nAChRs的表达和功能,影响了神经元的通讯。α 7nachr及其下游信号通路的离子化功能降低在许多神经系统疾病中都有发现。α 7nachr的缺失、胆碱能功能的下降和乙酰胆碱酯酶水平的升高通常与神经元变性、认知障碍和记忆功能下降有关。大量证据表明,简单的营养补充剂维生素D3 (VD)对许多神经系统疾病有认知益处(Skv et al. in Mol Neurobiol 61:7211-7238, 2024)。然而,目前的综述将集中在最近和过去的证据上,以破译α 7nachr对脑功能至关重要的独特特性。我们还强调了VD补充剂在恢复各种神经精神和神经疾病的胆碱能神经传递和α7nAChRs表达方面的治疗益处。
{"title":"Targeting Neuronal Alpha7 Nicotinic Acetylcholine Receptor Upregulation in Age-Related Neurological Disorders.","authors":"Sharon Mariam Abraham, Sneha Suresh, Pragya Komal","doi":"10.1007/s10571-025-01586-6","DOIUrl":"10.1007/s10571-025-01586-6","url":null,"abstract":"<p><p>The multifunctional roles of alpha7 nicotinic acetylcholine receptors (α7nAChRs), ranging from cognitive enhancement, neuroprotection, and anti-inflammatory action, credit tagging this receptor as \"unique\" among the cholinergic receptor family. The uniqueness of α7nAChRs in neuronal function and communication lies in their high calcium permeability among the cholinergic receptor family. The ionotropic function of α7nAChRs is governed by protein kinases' post-translational modification (PTMs), which alter their expression and function, affecting neuronal communication. A decrease in the ionotropic function of α7nAChRs and its downstream signaling pathways is observed across many neurologic disorders. The loss of α7nAChRs, decreased cholinergic function, and increased acetylcholinesterase levels are commonly associated with neuronal degeneration, cognitive impairment, and decreased memory function. An extensive body of evidence suggests the cognitive benefits of simple nutraceutical supplementation, Vitamin D3 (VD), in many neurologic disorders (Skv et al. in Mol Neurobiol 61:7211-7238, 2024). The present review will, however, focus on recent and past evidence deciphering the unique properties of α7nAChRs crucial for brain function. We have also emphasized on the therapeutic benefits of VD supplementation in restoring cholinergic neurotransmission and α7nAChRs expression in various neuropsychiatric and neurologic disorders.</p>","PeriodicalId":9742,"journal":{"name":"Cellular and Molecular Neurobiology","volume":"45 1","pages":"70"},"PeriodicalIF":4.8,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12267820/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144641934","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}