Pub Date : 2025-12-18DOI: 10.1016/j.neurot.2025.e00821
Roberto Santalucia, Carlotta Carosio, Antonio Giulio Gennari, Amir G Baroumand, Pascal Vrielynck, Alexane Fierain, Antoine Guilmot, Vincent Joris, Susana Ferrao Santos, Enrique Germany Morrison, Pieter van Mierlo, Georgia Ramantani, Riëm El Tahry
The aim of this study was to evaluate the feasibility of low-density (LD) interictal (IIC) and ictal (IC) electrical source imaging (ESI), and to assess their individual and combined diagnostic accuracy and predictive value in a cohort of children with drug-resistant epilepsy (DRE) who underwent resective surgery before the age of 7. Retrospective analysis was conducted on de-identified EEG and MRI data, which were (semi)-automatically processed, blinded to clinical information, to compute both IIC and IC-ESI. The concordance of ESI localizations with the resection cavity at sublobar level, and the association with surgical outcome were assessed. Thirty-two children were included. IIC- and IC-ESI showed an accuracy of 66 % (CI 95 % 47-81 %) and 72 % (CI 95 % 53-86 %) and a diagnostic odds ratio (DOR) of 3.0 (CI 95 % 0,66-13,69; p = 0,15) and 5.0 (CI 95 % 0,91-27,47; p = 0,06), respectively. The combined approach increased diagnostic performance, achieving an overall accuracy of 75 % and a DOR of 11.4 (CI 95 % 1.08-120,35; p = 0,042). In multivariate logistic regression analysis, the combined IIC/IC ESI result emerged as the strongest predictor of postsurgical seizure freedom (OR: 222,28; p = 0,0262; AUC: 0.87). These findings demonstrate that combined (semi)-automated LD-IIC and IC-ESI is feasible and can accurately localize the epileptogenic zone and predict postsurgical seizure freedom in children under 7 years of age. ESI may support earlier surgical referral, reduce the time from epilepsy onset to surgery, and ultimately improve long-term outcomes.
本研究的目的是评估低密度(LD)间期(IIC)和间期(IC)电源成像(ESI)的可行性,并评估其在7岁前接受切除手术的耐药癫痫(DRE)儿童队列中的单独和联合诊断准确性和预测价值。对去识别的EEG和MRI数据进行回顾性分析,经(半)自动处理,不考虑临床信息,计算IIC和IC-ESI。评估ESI定位与叶下水平切除腔的一致性,以及与手术结果的关系。其中包括32名儿童。IIC-和IC-ESI的准确率分别为66% (CI 95% 47 ~ 81%)和72% (CI 95% 53 ~ 86%),诊断优势比(DOR)分别为3.0 (CI 95% 0,66 ~ 13,69; p = 0,15)和5.0 (CI 95% 0,91 ~ 27,47; p = 0,06)。联合方法提高了诊断性能,达到75%的总体准确率和11.4的DOR (CI 95% 1.08-120,35; p = 0,042)。在多变量logistic回归分析中,IIC/IC ESI联合结果是术后癫痫发作自由的最强预测因子(OR: 222,28; p = 0,0262; AUC: 0.87)。以上结果表明,结合(半)自动化的LD-IIC和IC-ESI是可行的,可以准确定位7岁以下儿童的癫痫区,预测术后癫痫发作自由度。ESI可以支持早期手术转诊,减少癫痫发作到手术的时间,并最终改善长期预后。
{"title":"Combining interictal and ictal low-density EEG source imaging to delineate the epileptogenic zone in young children.","authors":"Roberto Santalucia, Carlotta Carosio, Antonio Giulio Gennari, Amir G Baroumand, Pascal Vrielynck, Alexane Fierain, Antoine Guilmot, Vincent Joris, Susana Ferrao Santos, Enrique Germany Morrison, Pieter van Mierlo, Georgia Ramantani, Riëm El Tahry","doi":"10.1016/j.neurot.2025.e00821","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00821","url":null,"abstract":"<p><p>The aim of this study was to evaluate the feasibility of low-density (LD) interictal (IIC) and ictal (IC) electrical source imaging (ESI), and to assess their individual and combined diagnostic accuracy and predictive value in a cohort of children with drug-resistant epilepsy (DRE) who underwent resective surgery before the age of 7. Retrospective analysis was conducted on de-identified EEG and MRI data, which were (semi)-automatically processed, blinded to clinical information, to compute both IIC and IC-ESI. The concordance of ESI localizations with the resection cavity at sublobar level, and the association with surgical outcome were assessed. Thirty-two children were included. IIC- and IC-ESI showed an accuracy of 66 % (CI 95 % 47-81 %) and 72 % (CI 95 % 53-86 %) and a diagnostic odds ratio (DOR) of 3.0 (CI 95 % 0,66-13,69; p = 0,15) and 5.0 (CI 95 % 0,91-27,47; p = 0,06), respectively. The combined approach increased diagnostic performance, achieving an overall accuracy of 75 % and a DOR of 11.4 (CI 95 % 1.08-120,35; p = 0,042). In multivariate logistic regression analysis, the combined IIC/IC ESI result emerged as the strongest predictor of postsurgical seizure freedom (OR: 222,28; p = 0,0262; AUC: 0.87). These findings demonstrate that combined (semi)-automated LD-IIC and IC-ESI is feasible and can accurately localize the epileptogenic zone and predict postsurgical seizure freedom in children under 7 years of age. ESI may support earlier surgical referral, reduce the time from epilepsy onset to surgery, and ultimately improve long-term outcomes.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00821"},"PeriodicalIF":6.9,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145794401","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-18DOI: 10.1016/j.neurot.2025.e00824
Suhayl Dhib-Jalbut
{"title":"Switching high efficacy therapies in Multiple Sclerosis: Does real world experience support such a strategy?","authors":"Suhayl Dhib-Jalbut","doi":"10.1016/j.neurot.2025.e00824","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00824","url":null,"abstract":"","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00824"},"PeriodicalIF":6.9,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145794333","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-18DOI: 10.1016/j.neurot.2025.e00822
Carl E Stafstrom
Infantile epileptic spasms syndrome (IESS) is a developmental and. epileptic encephalopathy with unique clinical and electrographic features, including seizure semiology (spasms), numerous and diverse etiologies spanning structural, genetic and metabolic causes, characteristic interictal (hypsarrhythmia) and ictal (electrodecrement) electroencephalogram (EEG) patterns, and responsiveness to "standard" pharmacological therapies (adrenocorticotrophic hormone, high-dose corticosteroids, vigabatrin) that are not commonly used in other epilepsy syndromes. Despite these long-recognized clinical features and laboratory investigations using a multiplicity of animal models with different epileptogenic mechanisms, the neurobiological underpinnings of IESS remain poorly understood, hampering the development of alternative treatments. This commentary discusses three aspects of IESS intended to raise fundamental clinical and mechanistic issues to afford greater understanding of the syndrome - nomenclature, EEG findings, and selected emerging animal models that might shed light on IESS pathophysiology and guide therapy development.
{"title":"Infantile epileptic spasms syndrome: Mechanisms and therapeutic approaches.","authors":"Carl E Stafstrom","doi":"10.1016/j.neurot.2025.e00822","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00822","url":null,"abstract":"<p><p>Infantile epileptic spasms syndrome (IESS) is a developmental and. epileptic encephalopathy with unique clinical and electrographic features, including seizure semiology (spasms), numerous and diverse etiologies spanning structural, genetic and metabolic causes, characteristic interictal (hypsarrhythmia) and ictal (electrodecrement) electroencephalogram (EEG) patterns, and responsiveness to \"standard\" pharmacological therapies (adrenocorticotrophic hormone, high-dose corticosteroids, vigabatrin) that are not commonly used in other epilepsy syndromes. Despite these long-recognized clinical features and laboratory investigations using a multiplicity of animal models with different epileptogenic mechanisms, the neurobiological underpinnings of IESS remain poorly understood, hampering the development of alternative treatments. This commentary discusses three aspects of IESS intended to raise fundamental clinical and mechanistic issues to afford greater understanding of the syndrome - nomenclature, EEG findings, and selected emerging animal models that might shed light on IESS pathophysiology and guide therapy development.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00822"},"PeriodicalIF":6.9,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145794309","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-16DOI: 10.1016/j.neurot.2025.e00813
Ran Yan, Wenhua Zhang, Wenjing Wang, Jiaqi Wu, Jun Zhang, Yingjie Xu, Wei Xu, Wen Yang
Alzheimer's disease (AD), closely associated with mitochondrial dysfunction, currently lacks convenient and non-invasive biomarkers for mitochondrial assessment. In this study, we developed an artificial intelligence framework leveraging live urine-derived stem cell (USC) mitochondrial fluorescence imaging to investigate differences between cognitively impaired individuals (AD and mild cognitive impairment (MCI)) and cognitively normal (CN) subjects. Mitochondrial fluorescence images from living HeLa cells were first segmented, and two binary classification models based on the ResNet-18 convolutional neural network were trained to identify mitochondrial hyperfission and hyperfusion relative to normal morphology. The models demonstrated robust performance in detecting intermediate mitochondrial states during validation. When applied to USCs, the system effectively distinguished mitochondrial patterns associated with cognitive impairment, highlighting its potential for the early detection of Alzheimer's disease and merits further validation in larger, independent cohorts.
{"title":"Deep learning analysis of urine-derived stem cell mitochondrial morphology as a non-invasive Alzheimer's disease biomarker.","authors":"Ran Yan, Wenhua Zhang, Wenjing Wang, Jiaqi Wu, Jun Zhang, Yingjie Xu, Wei Xu, Wen Yang","doi":"10.1016/j.neurot.2025.e00813","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00813","url":null,"abstract":"<p><p>Alzheimer's disease (AD), closely associated with mitochondrial dysfunction, currently lacks convenient and non-invasive biomarkers for mitochondrial assessment. In this study, we developed an artificial intelligence framework leveraging live urine-derived stem cell (USC) mitochondrial fluorescence imaging to investigate differences between cognitively impaired individuals (AD and mild cognitive impairment (MCI)) and cognitively normal (CN) subjects. Mitochondrial fluorescence images from living HeLa cells were first segmented, and two binary classification models based on the ResNet-18 convolutional neural network were trained to identify mitochondrial hyperfission and hyperfusion relative to normal morphology. The models demonstrated robust performance in detecting intermediate mitochondrial states during validation. When applied to USCs, the system effectively distinguished mitochondrial patterns associated with cognitive impairment, highlighting its potential for the early detection of Alzheimer's disease and merits further validation in larger, independent cohorts.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00813"},"PeriodicalIF":6.9,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145775109","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
More than half of subarachnoid hemorrhage (SAH) survivors develop delayed cognitive dysfunction, but the underlying mechanisms remain elusive. This study investigated the role of meningeal lymphatic vessels (mLVs) in this complication by examining their structural integrity, drainage capacity, and association with cognitive deficits post-SAH. In adult male C57BL/6J mice in which SAH was induced by intracisternal injection of autologous blood, spatial learning and memory, and hippocampal CA1 neuronal activity were impaired as early as 1 month post-surgery, with a marked exacerbation of these deficits at 2 months. SAH induced mLV fragmentation and atrophy, subsequent cerebrospinal and interstitial fluid drainage impairment, metabolite accumulation, and ultimately delayed cognitive dysfunction. Notably, lymphatic vessel ablation exacerbated these pathologies. In vitro experiments confirmed that vascular endothelial growth factor C (VEGF-C) reduced oxyhemoglobin-induced lymphatic endothelial cell apoptosis. Furthermore, in vivo studies demonstrated that VEGF-C therapy inhibited amyloid-β (Aβ) deposition in the hippocampal CA1 region and ameliorated cognitive dysfunction. Additional studies revealed that VEGF-C's protective effect on mLVs may be mediated via PI3K-AKT pathway activation. Collectively, these findings indicate that disrupted mLV integrity and drainage contribute to post-SAH cognitive impairment. Activation of VEGF-C-mediated PI3K-AKT signaling may preserve mLV function and represent a potential therapeutic strategy for preventing delayed cognitive impairment after SAH.
{"title":"Meningeal lymphatic dysfunction drives cognitive impairment after experimental subarachnoid hemorrhage.","authors":"Yichen Cai, Yanxin Shao, Hui Yuan, Lina Feng, Jing Wang, Mingfeng Yang, Cong Li, Baoliang Sun, Leilei Mao","doi":"10.1016/j.neurot.2025.e00819","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00819","url":null,"abstract":"<p><p>More than half of subarachnoid hemorrhage (SAH) survivors develop delayed cognitive dysfunction, but the underlying mechanisms remain elusive. This study investigated the role of meningeal lymphatic vessels (mLVs) in this complication by examining their structural integrity, drainage capacity, and association with cognitive deficits post-SAH. In adult male C57BL/6J mice in which SAH was induced by intracisternal injection of autologous blood, spatial learning and memory, and hippocampal CA1 neuronal activity were impaired as early as 1 month post-surgery, with a marked exacerbation of these deficits at 2 months. SAH induced mLV fragmentation and atrophy, subsequent cerebrospinal and interstitial fluid drainage impairment, metabolite accumulation, and ultimately delayed cognitive dysfunction. Notably, lymphatic vessel ablation exacerbated these pathologies. In vitro experiments confirmed that vascular endothelial growth factor C (VEGF-C) reduced oxyhemoglobin-induced lymphatic endothelial cell apoptosis. Furthermore, in vivo studies demonstrated that VEGF-C therapy inhibited amyloid-β (Aβ) deposition in the hippocampal CA1 region and ameliorated cognitive dysfunction. Additional studies revealed that VEGF-C's protective effect on mLVs may be mediated via PI3K-AKT pathway activation. Collectively, these findings indicate that disrupted mLV integrity and drainage contribute to post-SAH cognitive impairment. Activation of VEGF-C-mediated PI3K-AKT signaling may preserve mLV function and represent a potential therapeutic strategy for preventing delayed cognitive impairment after SAH.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00819"},"PeriodicalIF":6.9,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145752032","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-12DOI: 10.1016/j.neurot.2025.e00816
Fabiana Filogamo, Fabrizio Maria Liguori, Giovanna La Rana, Roberto Russo, Claudia Cristiano
Several studies show that neurosteroids currently play a significant role in autism spectrum disorders (ASD). However, the pathway of neurosteroid synthesis involved in ASD remains unclear. This study aimed to investigate the crosstalk between autism and neurosteroids, focusing on the mechanism of allopregnanolone production. We used the BTBR T+ tf/J (BTBR) mouse, a well-established animal model of ASD that exhibits typical autism-like behaviors along with neuroinflammation. In the hippocampus of BTBR mice, we observed a marked overexpression of pregnenolone and a related reduction in allopregnanolone levels. This neurosteroid imbalance also appears to be associated with an inflammatory pattern and the manifestation of repetitive and asocial behaviors. The combination of low doses of ultramicronized palmitoylethanolamide (PEA-um) and docosahexaenoic acid (DHA) restores allopregnanolone production modulating neurosteroidogenesis. In association with neurosteroid modulation, this restoration reduces repetitive behaviors and improves social interactions in BTBR mice, also modulating the inflammatory profile with a significant reduction in proinflammatory cytokines and brain-derived neurotrophic factor (BDNF) levels in the hippocampus. These effects demonstrate an important role of the peroxisome proliferator-activated receptor alpha (PPAR-α), whose expression is particularly reduced in BTBR mice. In addition, the pivotal involvement of PPAR-α was further supported by administering a specific antagonist that abolished the advantageous effects of PEA-um + DHA. Overall, our findings demonstrate the potential synergistic effect of the low-dose combination of PEA-um and DHA, confirming their therapeutic effect in ASD and the involvement of neurosteroids in their mechanism of action.
{"title":"Low-dose combination of ultramicronized palmitoylethanolamide and docosahexaenoic acid on neurosteroid and neuroinflammatory dysregulation in autism spectrum disorders.","authors":"Fabiana Filogamo, Fabrizio Maria Liguori, Giovanna La Rana, Roberto Russo, Claudia Cristiano","doi":"10.1016/j.neurot.2025.e00816","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00816","url":null,"abstract":"<p><p>Several studies show that neurosteroids currently play a significant role in autism spectrum disorders (ASD). However, the pathway of neurosteroid synthesis involved in ASD remains unclear. This study aimed to investigate the crosstalk between autism and neurosteroids, focusing on the mechanism of allopregnanolone production. We used the BTBR T+ tf/J (BTBR) mouse, a well-established animal model of ASD that exhibits typical autism-like behaviors along with neuroinflammation. In the hippocampus of BTBR mice, we observed a marked overexpression of pregnenolone and a related reduction in allopregnanolone levels. This neurosteroid imbalance also appears to be associated with an inflammatory pattern and the manifestation of repetitive and asocial behaviors. The combination of low doses of ultramicronized palmitoylethanolamide (PEA-um) and docosahexaenoic acid (DHA) restores allopregnanolone production modulating neurosteroidogenesis. In association with neurosteroid modulation, this restoration reduces repetitive behaviors and improves social interactions in BTBR mice, also modulating the inflammatory profile with a significant reduction in proinflammatory cytokines and brain-derived neurotrophic factor (BDNF) levels in the hippocampus. These effects demonstrate an important role of the peroxisome proliferator-activated receptor alpha (PPAR-α), whose expression is particularly reduced in BTBR mice. In addition, the pivotal involvement of PPAR-α was further supported by administering a specific antagonist that abolished the advantageous effects of PEA-um + DHA. Overall, our findings demonstrate the potential synergistic effect of the low-dose combination of PEA-um and DHA, confirming their therapeutic effect in ASD and the involvement of neurosteroids in their mechanism of action.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00816"},"PeriodicalIF":6.9,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145752094","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1016/j.neurot.2025.e00805
Leonardo E Ariello, David P W Rastall, Liana S Rosenthal
Primary adult-onset neurodegenerative cerebellar ataxias (PANCA) are a clinically and genetically diverse group of disorders for which disease-modifying therapies remain limited. In this review, we provide a comprehensive analysis of therapeutic strategies for PANCA, with a primary focus on clinical trials-randomized controlled and open-label-that have evaluated pharmacological agents, rehabilitation programs, and neuromodulatory interventions. Where clinical trial data are lacking, we incorporate relevant observational studies, expert consensus, and mechanistic rationale to contextualize current practices. Rehabilitation and multidisciplinary care remain foundational across all subtypes and are supported by growing clinical trial evidence. Pharmacological approaches, including omaveloxolone for Friedreich's ataxia and off-label agents such as riluzole, 4-aminopyridine, and varenicline, demonstrate subtype-specific benefits. Neuromodulation techniques, such as transcranial direct current stimulation and repetitive transcranial magnetic stimulation, have shown early promise in improving motor outcomes. In parallel, molecular and gene-based therapies-including antisense oligonucleotides, viral vector delivery systems, and CRISPR-based strategies-are advancing into preclinical and early-phase clinical studies. This evolving therapeutic landscape underscores a shift toward personalized, multimodal care for cerebellar ataxias and highlights the need for continued translational efforts to bridge mechanistic insights with clinical impact.
{"title":"Treatment of primary adult-onset neurodegenerative cerebellar ataxias.","authors":"Leonardo E Ariello, David P W Rastall, Liana S Rosenthal","doi":"10.1016/j.neurot.2025.e00805","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00805","url":null,"abstract":"<p><p>Primary adult-onset neurodegenerative cerebellar ataxias (PANCA) are a clinically and genetically diverse group of disorders for which disease-modifying therapies remain limited. In this review, we provide a comprehensive analysis of therapeutic strategies for PANCA, with a primary focus on clinical trials-randomized controlled and open-label-that have evaluated pharmacological agents, rehabilitation programs, and neuromodulatory interventions. Where clinical trial data are lacking, we incorporate relevant observational studies, expert consensus, and mechanistic rationale to contextualize current practices. Rehabilitation and multidisciplinary care remain foundational across all subtypes and are supported by growing clinical trial evidence. Pharmacological approaches, including omaveloxolone for Friedreich's ataxia and off-label agents such as riluzole, 4-aminopyridine, and varenicline, demonstrate subtype-specific benefits. Neuromodulation techniques, such as transcranial direct current stimulation and repetitive transcranial magnetic stimulation, have shown early promise in improving motor outcomes. In parallel, molecular and gene-based therapies-including antisense oligonucleotides, viral vector delivery systems, and CRISPR-based strategies-are advancing into preclinical and early-phase clinical studies. This evolving therapeutic landscape underscores a shift toward personalized, multimodal care for cerebellar ataxias and highlights the need for continued translational efforts to bridge mechanistic insights with clinical impact.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00805"},"PeriodicalIF":6.9,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145743519","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1016/j.neurot.2025.e00818
Yiyong Zeng, Jinhan Cai, Meilin Zheng, Yujie Jiang, Jingyang Le, Shengjun Zhou, Xiang Gao, Chenhui Zhou, Wei Cui
Intracerebral hemorrhage (ICH) is a highly fatal stroke subtype with limited treatment options, where pathological activation of peri-hematomal microglia drives acute secondary injury. Colony-stimulating factor 1 receptor (CSF-1R), highly expressed in microglia, is a potential therapeutic target. This study evaluated the effects of short-term administration of sunitinib, a clinically used CSF-1R inhibitor, in a collagenase-induced mouse ICH model and an in vitro hemoglobin (Hb)-treated BV2 microglial model. Sunitinib significantly improved motor functions, reduced myelin damage, and attenuated microglial activation and neuroinflammation in peri-hematomal tissue. RNA sequencing revealed that sunitinib might modulate lipid metabolism, phagocytosis, and immune response. In BV2 cells, sunitinib inhibited Hb-induced lipid droplet accumulation, phagocytic reduction, and pro-inflammatory cytokine production, effects mirrored by CSF-1R knockdown. These findings suggest that sunitinib alleviates acute ICH injury by modulating microglial functions, likely through inhibition of the CSF-1R axis, supporting its potential repurposing for central nervous system disorders like ICH.
{"title":"Sunitinib attenuates secondary injury via the regulation of peri-hematomal microglia at the acute phase of intracerebral hemorrhage.","authors":"Yiyong Zeng, Jinhan Cai, Meilin Zheng, Yujie Jiang, Jingyang Le, Shengjun Zhou, Xiang Gao, Chenhui Zhou, Wei Cui","doi":"10.1016/j.neurot.2025.e00818","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00818","url":null,"abstract":"<p><p>Intracerebral hemorrhage (ICH) is a highly fatal stroke subtype with limited treatment options, where pathological activation of peri-hematomal microglia drives acute secondary injury. Colony-stimulating factor 1 receptor (CSF-1R), highly expressed in microglia, is a potential therapeutic target. This study evaluated the effects of short-term administration of sunitinib, a clinically used CSF-1R inhibitor, in a collagenase-induced mouse ICH model and an in vitro hemoglobin (Hb)-treated BV2 microglial model. Sunitinib significantly improved motor functions, reduced myelin damage, and attenuated microglial activation and neuroinflammation in peri-hematomal tissue. RNA sequencing revealed that sunitinib might modulate lipid metabolism, phagocytosis, and immune response. In BV2 cells, sunitinib inhibited Hb-induced lipid droplet accumulation, phagocytic reduction, and pro-inflammatory cytokine production, effects mirrored by CSF-1R knockdown. These findings suggest that sunitinib alleviates acute ICH injury by modulating microglial functions, likely through inhibition of the CSF-1R axis, supporting its potential repurposing for central nervous system disorders like ICH.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00818"},"PeriodicalIF":6.9,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145743481","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-04DOI: 10.1016/j.neurot.2025.e00814
Charlie J Maddox, Francis S Lee, Anjali M Rajadhyaksha, Arlene Martínez-Rivera
Substance use disorder (SUD) remains a critical public health issue characterized by high rates of relapse and limited effective pharmacotherapies, particularly for non-opioid substances. A key challenge in addressing SUD lies in the persistent neuroadaptations within the brain's reward circuitry. The endocannabinoid (eCB) system plays a crucial role in modulating reward and reinforcement processes and is disrupted by chronic drug exposure. Recent work highlights the therapeutic potential of indirectly modulating cannabinoid 1 (CB1) receptor signaling by targeting eCB-metabolizing enzymes, fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), to restore homeostatic eCB tone. We review and synthesize findings from both genetic and pharmacological studies, highlighting the contributions of FAAH and MAGL across major classes of abused substances and considering their potential as therapeutic targets for SUD treatment.
{"title":"FAAH and MAGL inhibition: Evolving approaches to treating substance use disorders.","authors":"Charlie J Maddox, Francis S Lee, Anjali M Rajadhyaksha, Arlene Martínez-Rivera","doi":"10.1016/j.neurot.2025.e00814","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00814","url":null,"abstract":"<p><p>Substance use disorder (SUD) remains a critical public health issue characterized by high rates of relapse and limited effective pharmacotherapies, particularly for non-opioid substances. A key challenge in addressing SUD lies in the persistent neuroadaptations within the brain's reward circuitry. The endocannabinoid (eCB) system plays a crucial role in modulating reward and reinforcement processes and is disrupted by chronic drug exposure. Recent work highlights the therapeutic potential of indirectly modulating cannabinoid 1 (CB1) receptor signaling by targeting eCB-metabolizing enzymes, fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), to restore homeostatic eCB tone. We review and synthesize findings from both genetic and pharmacological studies, highlighting the contributions of FAAH and MAGL across major classes of abused substances and considering their potential as therapeutic targets for SUD treatment.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00814"},"PeriodicalIF":6.9,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145687746","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03DOI: 10.1016/j.neurot.2025.e00812
Franz Felix Konen, Steffen Pfeuffer, Konstantin Fritz Jendretzky, Klaus Gehring, Birte Elias-Hamp, Kurt-Wolfram Sühs, Stephan Halle, Korbinian Brand, Ralf Lichtinghagen, Eline Willemse, Marc Pawlitzki, Jens Kuhle, Sven G Meuth, Christoph Kleinschnitz, Refik Pul, Thomas Skripuletz
Anti-CD20 antibodies and cladribine are established therapies for active relapsing multiple sclerosis (RMS). Increasing evidence suggests that switching between these therapies may be beneficial in patients with ongoing disease activity under current treatment. In this multicenter retrospective study across six German MS centres, a total of 90 patients with active RMS were considered for inclusion, of whom 71 patients were switched either from anti-CD20 antibodies to cladribine (n = 31) or from cladribine to anti-CD20 antibodies (n = 40), with a minimum follow-up of 12 months. At treatment initiation, patients switching from anti-CD20 antibodies were older, had a longer disease duration, and a higher disability score compared to those switching from cladribine (p = 0.0040, p = 0.0447, p = 0.0028, respectively). The primary reason for switching was disease activity. Following the switch, the proportion of patients with relapsing disease activity was markedly reduced (from 55 % to 16 % for anti-CD20 to cladribine, and from 83 % to 25 % for cladribine to anti-CD20). Clinical outcomes improved, while serum biomarkers such as neurofilament light chain and glial fibrillary acidic protein remained stable over six months. Notably, the prevalence of hypogammaglobulinemia decreased after switching from anti-CD20 therapies to cladribine. These results indicate that patients with active RMS can achieve clinical stabilization after switching therapies in either direction, underscoring the complementary mechanisms of action and the safety of such an approach in real-world practice.
抗cd20抗体和克拉德滨是治疗活动性复发性多发性硬化症(RMS)的既定疗法。越来越多的证据表明,在目前的治疗下,在这些疗法之间切换可能对正在进行疾病活动的患者有益。在这项横跨6个德国MS中心的多中心回顾性研究中,共纳入了90例活动性RMS患者,其中71例患者从抗cd20抗体切换到cladriine (n = 31)或从cladriine切换到抗cd20抗体(n = 40),随访时间至少为12个月。在治疗开始时,与改用克拉德滨的患者相比,改用抗cd20抗体的患者年龄更大,病程更长,残疾评分更高(p = 0.0040, p = 0.0447, p = 0.0028)。转换的主要原因是疾病活动。转换后,复发疾病活动的患者比例显著降低(抗cd20到克拉德里宾的比例从55%降至16%,克拉德里宾到抗cd20的比例从83%降至25%)。临床结果得到改善,而血清生物标志物如神经丝轻链和胶质纤维酸性蛋白在6个月内保持稳定。值得注意的是,从抗cd20治疗转为克拉宾治疗后,低γ -球蛋白血症的患病率下降。这些结果表明,活动性RMS患者在转换治疗方向后可以达到临床稳定,强调了这种方法在现实实践中的互补机制和安全性。
{"title":"Switching from anti-CD20 therapies to cladribine and vice versa - Analysis of a German relapsing multiple sclerosis cohort.","authors":"Franz Felix Konen, Steffen Pfeuffer, Konstantin Fritz Jendretzky, Klaus Gehring, Birte Elias-Hamp, Kurt-Wolfram Sühs, Stephan Halle, Korbinian Brand, Ralf Lichtinghagen, Eline Willemse, Marc Pawlitzki, Jens Kuhle, Sven G Meuth, Christoph Kleinschnitz, Refik Pul, Thomas Skripuletz","doi":"10.1016/j.neurot.2025.e00812","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00812","url":null,"abstract":"<p><p>Anti-CD20 antibodies and cladribine are established therapies for active relapsing multiple sclerosis (RMS). Increasing evidence suggests that switching between these therapies may be beneficial in patients with ongoing disease activity under current treatment. In this multicenter retrospective study across six German MS centres, a total of 90 patients with active RMS were considered for inclusion, of whom 71 patients were switched either from anti-CD20 antibodies to cladribine (n = 31) or from cladribine to anti-CD20 antibodies (n = 40), with a minimum follow-up of 12 months. At treatment initiation, patients switching from anti-CD20 antibodies were older, had a longer disease duration, and a higher disability score compared to those switching from cladribine (p = 0.0040, p = 0.0447, p = 0.0028, respectively). The primary reason for switching was disease activity. Following the switch, the proportion of patients with relapsing disease activity was markedly reduced (from 55 % to 16 % for anti-CD20 to cladribine, and from 83 % to 25 % for cladribine to anti-CD20). Clinical outcomes improved, while serum biomarkers such as neurofilament light chain and glial fibrillary acidic protein remained stable over six months. Notably, the prevalence of hypogammaglobulinemia decreased after switching from anti-CD20 therapies to cladribine. These results indicate that patients with active RMS can achieve clinical stabilization after switching therapies in either direction, underscoring the complementary mechanisms of action and the safety of such an approach in real-world practice.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00812"},"PeriodicalIF":6.9,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145678208","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}