Pub Date : 2025-11-18DOI: 10.1016/j.neurot.2025.e00783
Shabnam Babakry, Jana V P Devos, Catharine A Hellingman, Linda Ackermans, Jasper V Smit, Michelle Moerel, Carsten Leue, Annelien A Duits, Yasin Temel, Marcus L F Janssen
Tinnitus disorder can have a significant negative impact on quality of life, especially when refractory to standard care. Deep brain stimulation (DBS) of the medial geniculate body (MGB) attenuates pathological neuronal activity in the central auditory pathway and is a potential treatment for severe tinnitus. The aim of this pilot study was to assess the safety and feasibility of bilateral MGB DBS in patients with refractory tinnitus disorder. This randomised double-blind 2 × 2 cross-over study was conducted at Maastricht University Medical Centre, Maastricht, the Netherlands. The included patients had treatment refractory, severe, and chronic tinnitus without an anatomical substrate. Patients with bilateral MGB DBS were randomised to an ON-OFF or OFF-ON stimulation order for two cross-over phases. Primary outcomes consisted of safety and feasibility. Secondary outcomes on tinnitus severity, psychiatric and cognitive functioning and quality of life were assessed at screening, after both cross-over phases and at one-year follow-up. Four patients were included. No irreversible stimulation-induced side effects were observed. Surgical-related side effects were transient and resolved within two weeks. All patients experienced DBS as an acceptable treatment. Three of four patients showed improvement of tinnitus complaints based on the Tinnitus Functional Index. In the non-responder, electrodes had the largest distance from the centre of the MGB. To conclude, this study shows that bilateral MGB DBS is safe and feasible for patients with refractory tinnitus. Findings suggest the potential for clinically meaningful reduction in tinnitus burden through DBS. Effectiveness needs to be further evaluated in a follow-up study.
{"title":"Deep brain stimulation of the medial geniculate body for refractory tinnitus: A feasibility study.","authors":"Shabnam Babakry, Jana V P Devos, Catharine A Hellingman, Linda Ackermans, Jasper V Smit, Michelle Moerel, Carsten Leue, Annelien A Duits, Yasin Temel, Marcus L F Janssen","doi":"10.1016/j.neurot.2025.e00783","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00783","url":null,"abstract":"<p><p>Tinnitus disorder can have a significant negative impact on quality of life, especially when refractory to standard care. Deep brain stimulation (DBS) of the medial geniculate body (MGB) attenuates pathological neuronal activity in the central auditory pathway and is a potential treatment for severe tinnitus. The aim of this pilot study was to assess the safety and feasibility of bilateral MGB DBS in patients with refractory tinnitus disorder. This randomised double-blind 2 × 2 cross-over study was conducted at Maastricht University Medical Centre, Maastricht, the Netherlands. The included patients had treatment refractory, severe, and chronic tinnitus without an anatomical substrate. Patients with bilateral MGB DBS were randomised to an ON-OFF or OFF-ON stimulation order for two cross-over phases. Primary outcomes consisted of safety and feasibility. Secondary outcomes on tinnitus severity, psychiatric and cognitive functioning and quality of life were assessed at screening, after both cross-over phases and at one-year follow-up. Four patients were included. No irreversible stimulation-induced side effects were observed. Surgical-related side effects were transient and resolved within two weeks. All patients experienced DBS as an acceptable treatment. Three of four patients showed improvement of tinnitus complaints based on the Tinnitus Functional Index. In the non-responder, electrodes had the largest distance from the centre of the MGB. To conclude, this study shows that bilateral MGB DBS is safe and feasible for patients with refractory tinnitus. Findings suggest the potential for clinically meaningful reduction in tinnitus burden through DBS. Effectiveness needs to be further evaluated in a follow-up study.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00783"},"PeriodicalIF":6.9,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145557618","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 : 2025-11-18DOI: 10.1016/j.neurot.2025.e00792
Jessica P Hayes, Velda X Han, Brooke A Keating, Hiroya Nishida, Erica Tsang, Xianzhong Lau, Ruwani Dissanayake, Nader Aryamanesh, Wendy Gold, Melanie Wong, Carolyn Ellaway, Brian S Gloss, Shekeeb S Mohammad, Markus J Hofer, Peter Valtchev, Shrujna Patel, Russell C Dale
Pathogenic DNA variants in chromatin-related genes constitute an important minority of neurodevelopmental disorders (NDDs). Epigenetic mechanisms, including chromatin regulation driven by genetic or environmental factors, are increasingly recognised as key contributors to pathogenesis of diverse NDDs. We hypothesise that therapeutic strategies targeting chromatin dysregulation, such as histone deacetylase inhibition with butyrate, may be a potential disease modifying therapy for NDDs. We first performed peripheral blood bulk RNA sequencing (RNA-seq) to explore baseline gene regulation in children with chromatin-related NDDs (Kabuki syndrome (KMT2D, n = 4), CHARGE syndrome (CHD7, n = 2), and Rett syndrome (MECP2, n = 5), and children with NDDs but without a monogenic diagnosis (non-monogenic, n = 8), compared with sex-matched healthy controls (total n = 21). Next, to explore the effects of butyrate, single-cell RNA sequencing (scRNA-seq) was performed on 101,539 peripheral immune cells from four selected patients (one per condition) and two controls, before and after butyrate treatment. At baseline, dysregulation of ribosomal and immune pathways was seen in all four NDD cohorts (KMT2D, CHD7, MECP2, non-monogenic) compared to controls. Butyrate largely reversed these pathways, normalising ribosomal and immune pathways in patient and control cells. Butyrate induced up-regulation of ribosome, GTPase, cytoskeletal, mitochondrial pathways, and down-regulation of epigenetic and immune pathways. In conclusion, we identified a common ribosomal-immune RNA signature in chromatin-related NDDs, and a similar signature in non-monogenic NDDs. We showed that butyrate modulates epigenetic and immune gene networks in monogenic and non-monogenic NDDs, positioning butyrate as a promising therapeutic modulator across diverse NDDs.
染色质相关基因的致病性DNA变异构成了神经发育障碍(ndd)的重要少数。表观遗传机制,包括由遗传或环境因素驱动的染色质调控,越来越被认为是各种ndd发病机制的关键因素。我们假设针对染色质失调的治疗策略,如用丁酸抑制组蛋白去乙酰化酶,可能是ndd的潜在疾病修饰疗法。我们首先进行了外周血大容量RNA测序(RNA-seq),以探索与染色质相关的ndd儿童(Kabuki综合征(KMT2D, n = 4)、CHARGE综合征(CHD7, n = 2)和Rett综合征(MECP2, n = 5)以及非单基因诊断的ndd儿童(非单基因,n = 8)的基线基因调节,并与性别匹配的健康对照组(总n = 21)进行比较。接下来,为了探索丁酸盐的作用,在丁酸盐治疗前后,对来自4名患者(每种情况一个)和2名对照组的101539个外周免疫细胞进行了单细胞RNA测序(scRNA-seq)。基线时,与对照组相比,所有四个NDD队列(KMT2D、CHD7、MECP2、非单基因)的核糖体和免疫途径均出现失调。丁酸盐在很大程度上逆转了这些途径,使患者和对照细胞中的核糖体和免疫途径正常化。丁酸盐诱导核糖体、GTPase、细胞骨架、线粒体途径上调,表观遗传和免疫途径下调。总之,我们在染色质相关ndd中发现了共同的核糖体免疫RNA特征,在非单基因ndd中也发现了类似的特征。我们发现丁酸盐调节单基因和非单基因ndd的表观遗传和免疫基因网络,将丁酸盐定位为多种ndd的有前途的治疗调节剂。
{"title":"Butyrate modifies epigenetic and immune pathways in peripheral mononuclear cells from children with neurodevelopmental disorders associated with chromatin dysregulation.","authors":"Jessica P Hayes, Velda X Han, Brooke A Keating, Hiroya Nishida, Erica Tsang, Xianzhong Lau, Ruwani Dissanayake, Nader Aryamanesh, Wendy Gold, Melanie Wong, Carolyn Ellaway, Brian S Gloss, Shekeeb S Mohammad, Markus J Hofer, Peter Valtchev, Shrujna Patel, Russell C Dale","doi":"10.1016/j.neurot.2025.e00792","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00792","url":null,"abstract":"<p><p>Pathogenic DNA variants in chromatin-related genes constitute an important minority of neurodevelopmental disorders (NDDs). Epigenetic mechanisms, including chromatin regulation driven by genetic or environmental factors, are increasingly recognised as key contributors to pathogenesis of diverse NDDs. We hypothesise that therapeutic strategies targeting chromatin dysregulation, such as histone deacetylase inhibition with butyrate, may be a potential disease modifying therapy for NDDs. We first performed peripheral blood bulk RNA sequencing (RNA-seq) to explore baseline gene regulation in children with chromatin-related NDDs (Kabuki syndrome (KMT2D, n = 4), CHARGE syndrome (CHD7, n = 2), and Rett syndrome (MECP2, n = 5), and children with NDDs but without a monogenic diagnosis (non-monogenic, n = 8), compared with sex-matched healthy controls (total n = 21). Next, to explore the effects of butyrate, single-cell RNA sequencing (scRNA-seq) was performed on 101,539 peripheral immune cells from four selected patients (one per condition) and two controls, before and after butyrate treatment. At baseline, dysregulation of ribosomal and immune pathways was seen in all four NDD cohorts (KMT2D, CHD7, MECP2, non-monogenic) compared to controls. Butyrate largely reversed these pathways, normalising ribosomal and immune pathways in patient and control cells. Butyrate induced up-regulation of ribosome, GTPase, cytoskeletal, mitochondrial pathways, and down-regulation of epigenetic and immune pathways. In conclusion, we identified a common ribosomal-immune RNA signature in chromatin-related NDDs, and a similar signature in non-monogenic NDDs. We showed that butyrate modulates epigenetic and immune gene networks in monogenic and non-monogenic NDDs, positioning butyrate as a promising therapeutic modulator across diverse NDDs.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00792"},"PeriodicalIF":6.9,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145557517","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 : 2025-11-18DOI: 10.1016/j.neurot.2025.e00804
Yu-Lin Li, Jia-Jia Wu, Xi Zhang, Yi Wu, Jian-Guang Xu
Functional connectivity (FC) is crucial for the execution of functional activities and the progression of diseases. From a functional perspective, changes in subthalamic nucleus (STN)-based FC and network properties across lifespan and their effectiveness in predicting age require further investigation. The STN-based atlas was constructed using neuroimaging data from 1060 healthy participants. Based on this atlas, the FC and brain network properties were quantified subsequently. Both linear and non-linear changes in FC and network properties with aging, and relationships across brain regions were analyzed with generalized additive model (GAM). Connectome-based predictive modeling (CPM) was used to evaluate the performance of FC in predicting age, while computational lesion analysis was utilized to identify the FC with significant contributions to model performance. The STN-based atlas mainly involved bilateral sensory, motor, and sensory-motor integration cortices, subcortical nuclei, and cerebellar crus I and II. Global network properties were declined with aging (all P < 0.05). Significant non-linear age-related changes were identified in regional network properties of bilateral M1 and right STN (all P < 0.05). There were non-linear negative relationships between bilateral M1, and between right M1 and right STN (all P < 0.05). The FC within motor cortex, and between limbic areas and subcortical nuclei, significantly contributed to the performance of CPM. Therefore, from a functional perspective, dynamic changes in FC and network properties across the lifespan were highlighted, particularly within motor-related circuits. This provides valuable insights into neural mechanisms underlying motor dysfunction during aging and informs circuit-based neuromodulation strategies.
{"title":"Lifespan dynamics of STN-based functional connectivity and network properties: Implications for motor dysfunction and circuit-based neuromodulation.","authors":"Yu-Lin Li, Jia-Jia Wu, Xi Zhang, Yi Wu, Jian-Guang Xu","doi":"10.1016/j.neurot.2025.e00804","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00804","url":null,"abstract":"<p><p>Functional connectivity (FC) is crucial for the execution of functional activities and the progression of diseases. From a functional perspective, changes in subthalamic nucleus (STN)-based FC and network properties across lifespan and their effectiveness in predicting age require further investigation. The STN-based atlas was constructed using neuroimaging data from 1060 healthy participants. Based on this atlas, the FC and brain network properties were quantified subsequently. Both linear and non-linear changes in FC and network properties with aging, and relationships across brain regions were analyzed with generalized additive model (GAM). Connectome-based predictive modeling (CPM) was used to evaluate the performance of FC in predicting age, while computational lesion analysis was utilized to identify the FC with significant contributions to model performance. The STN-based atlas mainly involved bilateral sensory, motor, and sensory-motor integration cortices, subcortical nuclei, and cerebellar crus I and II. Global network properties were declined with aging (all P < 0.05). Significant non-linear age-related changes were identified in regional network properties of bilateral M1 and right STN (all P < 0.05). There were non-linear negative relationships between bilateral M1, and between right M1 and right STN (all P < 0.05). The FC within motor cortex, and between limbic areas and subcortical nuclei, significantly contributed to the performance of CPM. Therefore, from a functional perspective, dynamic changes in FC and network properties across the lifespan were highlighted, particularly within motor-related circuits. This provides valuable insights into neural mechanisms underlying motor dysfunction during aging and informs circuit-based neuromodulation strategies.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00804"},"PeriodicalIF":6.9,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145557648","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}
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by progressive motor neuron degeneration and muscle weakness, generally leading to death due to respiratory failure within 2-5 years of symptom onset. Current Food and Drug Administration-approved drugs -riluzole, edaravone, and tofersen - offer limited clinical benefit due to ALS multifactorial etiology and high heterogeneity. To bypass this therapeutic letdown, we previously exploited network medicine and drug repurposing strategies. Leveraging the SAveRUNNER algorithm, we identified several potentially repurposable candidates, including clomipramine (Anafranil®), mianserin (Lantanon®/Tolvon®), and modafinil (Provigil®). Here, we evaluated the in vivo efficacy of these compounds in Drosophila models of ALS, precisely those expressing pan-neuronal human SOD1A4V or SOD1G85R mutations. Our results demonstrate that clomipramine is the most promising candidate, ameliorating lifespan reduction, improving climbing abilities, and mitigating both genomic instability and inflammation, key pathological hallmarks of these SOD1-ALS models. Despite needing further validation in higher organisms, our Drosophila findings represent preliminary yet significant support for clomipramine's action as an add-on treatment for SOD1-ALS.
{"title":"Validation in Drosophila of the in silico predicted clomipramine as repurposable for SOD1-ALS.","authors":"Francesco Liguori, Susanna Amadio, Chiara Angioli, Angelo Ferriero, Iolanda Passaro, Francesca Alberti, Fiammetta Vernì, Cinzia Volonté","doi":"10.1016/j.neurot.2025.e00793","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00793","url":null,"abstract":"<p><p>Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by progressive motor neuron degeneration and muscle weakness, generally leading to death due to respiratory failure within 2-5 years of symptom onset. Current Food and Drug Administration-approved drugs -riluzole, edaravone, and tofersen - offer limited clinical benefit due to ALS multifactorial etiology and high heterogeneity. To bypass this therapeutic letdown, we previously exploited network medicine and drug repurposing strategies. Leveraging the SAveRUNNER algorithm, we identified several potentially repurposable candidates, including clomipramine (Anafranil®), mianserin (Lantanon®/Tolvon®), and modafinil (Provigil®). Here, we evaluated the in vivo efficacy of these compounds in Drosophila models of ALS, precisely those expressing pan-neuronal human SOD1<sup>A4V</sup> or SOD1<sup>G85R</sup> mutations. Our results demonstrate that clomipramine is the most promising candidate, ameliorating lifespan reduction, improving climbing abilities, and mitigating both genomic instability and inflammation, key pathological hallmarks of these SOD1-ALS models. Despite needing further validation in higher organisms, our Drosophila findings represent preliminary yet significant support for clomipramine's action as an add-on treatment for SOD1-ALS.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00793"},"PeriodicalIF":6.9,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145523960","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}
Traumatic brain injury (TBI) constitutes a major global public health concern associated with substantial mortality and long-term disability, while its diagnostic approaches and underlying pathophysiological mechanisms remain to be fully elucidated. In this study, we conducted longitudinal metabolomic profiling of cerebrospinal fluid (CSF) from 23 TBI patients (9 mild, 14 severe) and 5 uninjured controls using high-resolution mass spectrometry. Comprehensive quantification of metabolites was performed at three critical post-injury time points (days 1, 3, and 7), revealing distinct temporal metabolic patterns. Our results demonstrated significant alterations in the CSF metabolome following TBI. Early-phase changes (day 1) predominantly involved energy-related metabolites, including sphingosine, glucose, and dl-lactate. More pronounced metabolic shifts were observed by day 3, characterized by marked variations in amino acids (l-glutamine, l-histidine) and medium-chain fatty acids (caprylic acid, octanoic acid), suggesting the transition from primary to secondary injury mechanisms. The day 7 profile revealed accumulation of repair-associated metabolites such as 2'-deoxyuridine 5'-monophosphate and 1,2-dihexadecanoyl-sn-glycerol, potentially indicative of processes occurring in the chronic phase, which may include both reparative mechanisms and ongoing pathology. Notably, we identified significant alterations in established biomarkers (trimethylamine N-oxide) and novel small peptides (e.g., Gly-His-Lys), with distinct metabolic signatures differentiating mild versus severe TBI cases. These findings delineate temporally dynamic and severity-dependent metabolic reprogramming in TBI, providing mechanistic insights into the progression from acute injury through secondary pathogenesis to chronic recovery phases. The identified metabolic signatures may serve as potential biomarkers for injury staging and therapeutic monitoring.
{"title":"Metabolomics of cerebrospinal fluid following traumatic brain injury: Exploration of biomarkers for secondary injuries and severity.","authors":"Shuai Zhang, Zhuohang Wang, Jiangxin Mao, Aikang Zhang, Sufang Zhang, Juxiang Chen","doi":"10.1016/j.neurot.2025.e00789","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00789","url":null,"abstract":"<p><p>Traumatic brain injury (TBI) constitutes a major global public health concern associated with substantial mortality and long-term disability, while its diagnostic approaches and underlying pathophysiological mechanisms remain to be fully elucidated. In this study, we conducted longitudinal metabolomic profiling of cerebrospinal fluid (CSF) from 23 TBI patients (9 mild, 14 severe) and 5 uninjured controls using high-resolution mass spectrometry. Comprehensive quantification of metabolites was performed at three critical post-injury time points (days 1, 3, and 7), revealing distinct temporal metabolic patterns. Our results demonstrated significant alterations in the CSF metabolome following TBI. Early-phase changes (day 1) predominantly involved energy-related metabolites, including sphingosine, glucose, and dl-lactate. More pronounced metabolic shifts were observed by day 3, characterized by marked variations in amino acids (l-glutamine, l-histidine) and medium-chain fatty acids (caprylic acid, octanoic acid), suggesting the transition from primary to secondary injury mechanisms. The day 7 profile revealed accumulation of repair-associated metabolites such as 2'-deoxyuridine 5'-monophosphate and 1,2-dihexadecanoyl-sn-glycerol, potentially indicative of processes occurring in the chronic phase, which may include both reparative mechanisms and ongoing pathology. Notably, we identified significant alterations in established biomarkers (trimethylamine N-oxide) and novel small peptides (e.g., Gly-His-Lys), with distinct metabolic signatures differentiating mild versus severe TBI cases. These findings delineate temporally dynamic and severity-dependent metabolic reprogramming in TBI, providing mechanistic insights into the progression from acute injury through secondary pathogenesis to chronic recovery phases. The identified metabolic signatures may serve as potential biomarkers for injury staging and therapeutic monitoring.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00789"},"PeriodicalIF":6.9,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145523993","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 : 2025-11-11DOI: 10.1016/j.neurot.2025.e00786
Zhaojuan Ke, Ying Zhang, Xin Jiang, Jie Luo, Hengsheng Chen, Yao Ma, Qibin Chen, Li Zhao, Binyang Cai
Ketamine exhibits superior efficacy compared to conventional antidepressants, yet its clinical application remains limited by dose-dependent side effects. Ascorbic acid (AA) augments ketamine's antidepressant-like effects, suggesting that AA co-administration with subtherapeutic ketamine doses may achieve optimal efficacy while improving safety. Since AA itself lacks independently clinical antidepressant efficacy, its enhancement on the efficacy of ketamine raises a mechanistic question. This study investigated the antidepressant-like effects of AA combined with esketamine (ketamine's S-enantiomer) and elucidated the role of hippocampal TARP-γ8-containing AMPA receptors (AMPARs) in this enhancement. We employed a chronic restraint stress-induced mouse model of depression to evaluate depressive-like behaviors, hippocampal synaptic markers, and neural plasticity. The effects of AA, esketamine, and their combination were examined, along with pharmacological modulation of hippocampal TARP-γ8-containing AMPARs. Our findings demonstrated that AA enhanced the action of a subeffective dose of esketamine, fully reversing both behavioral and synaptic deficits in depressed mice to levels comparable with healthy controls. This combinatorial effect was equivalent to that achieved by an effective dose of esketamine alone. Selective pharmacological blockade of hippocampal TARP-γ8-containing AMPARs completely abolished the antidepressant-like efficacy of the subeffective-dosed AA-esketamine combination. However, the same blockade did not affect baseline depressive-like phenotypes in depressed mice or the inactivity of either agent at subeffective dose alone. These results indicate that AA's enhancement of esketamine's antidepressant-like effects requires the dependent mediation of hippocampal TARP-γ8-containing AMPARs for synaptic modulation, providing both mechanistic insight and potential clinical implications for optimizing ketamine-based strategies of therapeutics for depression.
{"title":"Ascorbic acid enhances antidepressant-like efficacy of esketamine: Hippocampal TARP-γ8-containing AMPA receptors mediate synaptic modulation.","authors":"Zhaojuan Ke, Ying Zhang, Xin Jiang, Jie Luo, Hengsheng Chen, Yao Ma, Qibin Chen, Li Zhao, Binyang Cai","doi":"10.1016/j.neurot.2025.e00786","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00786","url":null,"abstract":"<p><p>Ketamine exhibits superior efficacy compared to conventional antidepressants, yet its clinical application remains limited by dose-dependent side effects. Ascorbic acid (AA) augments ketamine's antidepressant-like effects, suggesting that AA co-administration with subtherapeutic ketamine doses may achieve optimal efficacy while improving safety. Since AA itself lacks independently clinical antidepressant efficacy, its enhancement on the efficacy of ketamine raises a mechanistic question. This study investigated the antidepressant-like effects of AA combined with esketamine (ketamine's S-enantiomer) and elucidated the role of hippocampal TARP-γ8-containing AMPA receptors (AMPARs) in this enhancement. We employed a chronic restraint stress-induced mouse model of depression to evaluate depressive-like behaviors, hippocampal synaptic markers, and neural plasticity. The effects of AA, esketamine, and their combination were examined, along with pharmacological modulation of hippocampal TARP-γ8-containing AMPARs. Our findings demonstrated that AA enhanced the action of a subeffective dose of esketamine, fully reversing both behavioral and synaptic deficits in depressed mice to levels comparable with healthy controls. This combinatorial effect was equivalent to that achieved by an effective dose of esketamine alone. Selective pharmacological blockade of hippocampal TARP-γ8-containing AMPARs completely abolished the antidepressant-like efficacy of the subeffective-dosed AA-esketamine combination. However, the same blockade did not affect baseline depressive-like phenotypes in depressed mice or the inactivity of either agent at subeffective dose alone. These results indicate that AA's enhancement of esketamine's antidepressant-like effects requires the dependent mediation of hippocampal TARP-γ8-containing AMPARs for synaptic modulation, providing both mechanistic insight and potential clinical implications for optimizing ketamine-based strategies of therapeutics for depression.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00786"},"PeriodicalIF":6.9,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145506076","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 : 2025-11-11DOI: 10.1016/j.neurot.2025.e00785
Tracy W Brown, Francesca M Filbey
Pain and sleep disturbances are primary reasons for medicinal cannabis use. Cannabis influences both pain and sleep through its modulation of the endocannabinoid system, which regulates pain and sleep signaling. Despite their interconnected roles, the effects of cannabis and chronic pain on sleep architecture have been studied mainly in isolation. An integrated understanding is needed to guide use and minimize risks in this population. Our primary aim was to examine the potential interactive effect of regular cannabis use on chronic pain and sleep. A total of 339 nights (2273.43 h) of in-home sleep electroencephalogram (EEG) recordings were collected from 60 adults (50 % male; 32 % chronic pain; 47 % cannabis use; Mage = 25.25; SE = 1.05) over seven consecutive nights per participant. A mixed-model repeated-measures ANCOVA tested the main effects and interactions of chronic pain and regular cannabis use on total sleep time (TST), total slow-wave sleep (SWS%), total rapid-eye-movement (REM%), sleep onset latency (SOL), and number of awakenings. There was a significant main effect of cannabis use on SWS, TST, SOL, and REM. There was a significant main effect of chronic pain on TST. Significant interactions emerged between cannabis use and chronic pain on SWS and REM. These findings may reflect a dysregulated sleep response in individuals using cannabis to manage chronic pain, highlighting the need to consider both beneficial and detrimental effects of cannabis on specific sleep stages.
疼痛和睡眠障碍是使用医用大麻的主要原因。大麻通过调节内源性大麻素系统来影响疼痛和睡眠,内源性大麻素系统调节疼痛和睡眠信号。尽管大麻和慢性疼痛对睡眠结构的影响相互关联,但它们主要是单独研究的。需要一个综合的认识来指导这一人群的使用并将风险降到最低。我们的主要目的是研究经常使用大麻对慢性疼痛和睡眠的潜在相互作用。从60名成年人(50%为男性,32%为慢性疼痛,47%为大麻使用者,Mage = 25.25, SE = 1.05)连续7个晚上收集了339个晚上(2273.43小时)的在家睡眠脑电图(EEG)记录。混合模型重复测量ANCOVA测试了慢性疼痛和经常使用大麻对总睡眠时间(TST)、总慢波睡眠(SWS%)、总快速眼动(REM%)、睡眠发作潜伏期(SOL)和觉醒次数的主要影响和相互作用。大麻使用对SWS、TST、SOL和REM有显著的主影响,慢性疼痛对TST有显著的主影响。大麻使用与SWS和REM的慢性疼痛之间出现了显著的相互作用。这些发现可能反映了使用大麻来控制慢性疼痛的个体的睡眠反应失调,强调需要考虑大麻对特定睡眠阶段的有益和有害影响。
{"title":"Interactions between cannabis use and chronic pain on sleep architecture: Findings from in-home EEG recordings.","authors":"Tracy W Brown, Francesca M Filbey","doi":"10.1016/j.neurot.2025.e00785","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00785","url":null,"abstract":"<p><p>Pain and sleep disturbances are primary reasons for medicinal cannabis use. Cannabis influences both pain and sleep through its modulation of the endocannabinoid system, which regulates pain and sleep signaling. Despite their interconnected roles, the effects of cannabis and chronic pain on sleep architecture have been studied mainly in isolation. An integrated understanding is needed to guide use and minimize risks in this population. Our primary aim was to examine the potential interactive effect of regular cannabis use on chronic pain and sleep. A total of 339 nights (2273.43 h) of in-home sleep electroencephalogram (EEG) recordings were collected from 60 adults (50 % male; 32 % chronic pain; 47 % cannabis use; M<sub>age</sub> = 25.25; SE = 1.05) over seven consecutive nights per participant. A mixed-model repeated-measures ANCOVA tested the main effects and interactions of chronic pain and regular cannabis use on total sleep time (TST), total slow-wave sleep (SWS%), total rapid-eye-movement (REM%), sleep onset latency (SOL), and number of awakenings. There was a significant main effect of cannabis use on SWS, TST, SOL, and REM. There was a significant main effect of chronic pain on TST. Significant interactions emerged between cannabis use and chronic pain on SWS and REM. These findings may reflect a dysregulated sleep response in individuals using cannabis to manage chronic pain, highlighting the need to consider both beneficial and detrimental effects of cannabis on specific sleep stages.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00785"},"PeriodicalIF":6.9,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145506063","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 : 2025-11-11DOI: 10.1016/j.neurot.2025.e00790
Zhenxiang Zang, An'ning Li, Zhifang Zhang, Tingfang Wu, Xiongying Chen, Kaini Qiao, Alimire Paerhati, Zhi Yang, Gang Wang
Racemic (R,S)-ketamine exerts rapid antidepressant effects, and growing evidence suggests its R-isomer may offer sustained efficacy with fewer side effects. However, the neurobiological mechanisms underlying (R)-ketamine's action in the human brain are largely unknown. To address this, we acquired resting-state fMRI data from 32 healthy volunteers 24 h before and after intranasal administration of (R)-ketamine (n = 24) or placebo (n = 8). We primarily assessed changes in long-range functional synchrony using degree centrality (DC) and elucidated the sources of these changes with functional connectivity (FC) analysis. (R)-ketamine significantly decreased DC in a key cognitive-motor integration hub: the supplementary motor area/middle cingulate cortex (SMA/MCC, cluster-corrected P < 0.05). Critically, the reduction of DC was absent under the placebo condition, yielding a significant group-by-time interaction (P = 0.01). The reduction in long-range synchrony of the SMA/MCC was primarily driven by attenuated FC with both the dorsal medial prefrontal cortex/dorsal anterior cingulate cortex (dMPFC/dACC) and the cerebellum, and was spatially correlated with serotonin, norepinephrine, and acetylcholine neurotransmitter profiles. More importantly, the clinical relevance of the neuroimaging phenotypes was established in an independent Major Depressive Disorder (MDD) cohort, where FC between the SMA/MCC and dMPFC/dACC significantly correlated with depressive symptom severity (HAMD score, P = 0.019). This study provides novel, system-level evidence that intranasal (R)-ketamine modulates specific human brain networks by attenuating long-range synchrony in the SMA/MCC. The link between the neuroimaging phenotype, depression-relevant neurotransmitter profiles, and clinical symptom severity may offer a plausible therapeutic mechanism of (R)-ketamine.
{"title":"Intranasal (R)-ketamine modulates depression symptom and neurotransmitters-associated human brain connectivity.","authors":"Zhenxiang Zang, An'ning Li, Zhifang Zhang, Tingfang Wu, Xiongying Chen, Kaini Qiao, Alimire Paerhati, Zhi Yang, Gang Wang","doi":"10.1016/j.neurot.2025.e00790","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00790","url":null,"abstract":"<p><p>Racemic (R,S)-ketamine exerts rapid antidepressant effects, and growing evidence suggests its R-isomer may offer sustained efficacy with fewer side effects. However, the neurobiological mechanisms underlying (R)-ketamine's action in the human brain are largely unknown. To address this, we acquired resting-state fMRI data from 32 healthy volunteers 24 h before and after intranasal administration of (R)-ketamine (n = 24) or placebo (n = 8). We primarily assessed changes in long-range functional synchrony using degree centrality (DC) and elucidated the sources of these changes with functional connectivity (FC) analysis. (R)-ketamine significantly decreased DC in a key cognitive-motor integration hub: the supplementary motor area/middle cingulate cortex (SMA/MCC, cluster-corrected P < 0.05). Critically, the reduction of DC was absent under the placebo condition, yielding a significant group-by-time interaction (P = 0.01). The reduction in long-range synchrony of the SMA/MCC was primarily driven by attenuated FC with both the dorsal medial prefrontal cortex/dorsal anterior cingulate cortex (dMPFC/dACC) and the cerebellum, and was spatially correlated with serotonin, norepinephrine, and acetylcholine neurotransmitter profiles. More importantly, the clinical relevance of the neuroimaging phenotypes was established in an independent Major Depressive Disorder (MDD) cohort, where FC between the SMA/MCC and dMPFC/dACC significantly correlated with depressive symptom severity (HAMD score, P = 0.019). This study provides novel, system-level evidence that intranasal (R)-ketamine modulates specific human brain networks by attenuating long-range synchrony in the SMA/MCC. The link between the neuroimaging phenotype, depression-relevant neurotransmitter profiles, and clinical symptom severity may offer a plausible therapeutic mechanism of (R)-ketamine.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00790"},"PeriodicalIF":6.9,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145506013","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 : 2025-11-10DOI: 10.1016/j.neurot.2025.e00788
Deborah K Rose, Constantine G Lyketsos, Paul B Rosenberg, Milap A Nowrangi
Neuropsychiatric symptoms (NPS) are among the most distressing and functionally disruptive features of Alzheimer's disease (AD), affecting the vast majority of individuals across the disease continuum. These symptoms, ranging from apathy and depression to agitation and psychosis, not only worsen quality of life but also predict faster decline, earlier institutionalization, and heightened caregiver burden. Yet, despite their clinical significance, NPS remain under-recognized and undertreated. This review synthesizes current understanding of the biological underpinnings of NPS in AD, highlighting network-level dysfunction, neurotransmitter imbalances, neuroinflammation, and emerging roles for tau pathology and circadian disruption. We critically examine current treatment paradigms, noting that pharmacologic interventions offer benefit but often carry significant risks. In contrast, non-pharmacological approaches, particularly those that integrate caregiver training, environmental design, and sensory engagement, hold promise but are inconsistently applied in routine care. Emerging innovations, including neuromodulation, repurposed agents (e.g., beta-blockers, cannabinoids), and digital therapeutics such as virtual reality and AI-enabled monitoring tools, offer new therapeutic avenues. We call for a paradigm shift toward person-centered, mechanistically-informed care that aligns intervention strategies with biological drivers of NPS. Future progress hinges on inclusive clinical trials, implementation of first-line behavioral strategies, and development of biomarker-guided, precision approaches to symptom management. Effective care for NPS in AD demands integration, not substitution, of pharmacologic and non-pharmacologic strategies, grounded in a deeper understanding of both disease biology and lived patient experience.
{"title":"Neuropsychiatric symptoms in Alzheimer's disease: Bridging mechanisms, management, and emerging innovations.","authors":"Deborah K Rose, Constantine G Lyketsos, Paul B Rosenberg, Milap A Nowrangi","doi":"10.1016/j.neurot.2025.e00788","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00788","url":null,"abstract":"<p><p>Neuropsychiatric symptoms (NPS) are among the most distressing and functionally disruptive features of Alzheimer's disease (AD), affecting the vast majority of individuals across the disease continuum. These symptoms, ranging from apathy and depression to agitation and psychosis, not only worsen quality of life but also predict faster decline, earlier institutionalization, and heightened caregiver burden. Yet, despite their clinical significance, NPS remain under-recognized and undertreated. This review synthesizes current understanding of the biological underpinnings of NPS in AD, highlighting network-level dysfunction, neurotransmitter imbalances, neuroinflammation, and emerging roles for tau pathology and circadian disruption. We critically examine current treatment paradigms, noting that pharmacologic interventions offer benefit but often carry significant risks. In contrast, non-pharmacological approaches, particularly those that integrate caregiver training, environmental design, and sensory engagement, hold promise but are inconsistently applied in routine care. Emerging innovations, including neuromodulation, repurposed agents (e.g., beta-blockers, cannabinoids), and digital therapeutics such as virtual reality and AI-enabled monitoring tools, offer new therapeutic avenues. We call for a paradigm shift toward person-centered, mechanistically-informed care that aligns intervention strategies with biological drivers of NPS. Future progress hinges on inclusive clinical trials, implementation of first-line behavioral strategies, and development of biomarker-guided, precision approaches to symptom management. Effective care for NPS in AD demands integration, not substitution, of pharmacologic and non-pharmacologic strategies, grounded in a deeper understanding of both disease biology and lived patient experience.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00788"},"PeriodicalIF":6.9,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145496329","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 : 2025-11-07DOI: 10.1016/j.neurot.2025.e00781
Qianxiong He, Yannan Chen, Peiwen Chen, Yi Wang, Rong Zou, Feng Zhao, Guangqun Zeng, Lin Zhang, Haiping Liu, Yuanjiang Shi, Liuyi Xiao, Xiaorong Xin
Ischemic retinal damage is the most common cause of severe vision impairment and blindness. Anti-vascular endothelial growth factor (anti-VEGF) agents have transformed the treatment of retinal ischemic disorders and have become the cornerstone therapy for these conditions. Nonetheless, the risk for systemic and ocular adverse effects necessitates careful consideration. Meanwhile, the therapeutic potential of natural compounds for ischemic retinal injury is increasingly attracting attention. In this study, piperine (PIP), a natural compound derived from pepper, was found to reduce apoptosis by reducing the severity of retinal and optic nerve ischemic damage. However, the precise pharmacological mechanisms of PIP are yet to be fully elucidated. Molecular docking (MD) studies, MD simulations, and surface plasmon resonance experiments were conducted to determine the molecular targets of PIP. Our data revealed that PIP can bind to apurinic/apyrimidinic endonuclease 1 (APE1), thereby inhibiting apoptosis by decreasing the expression of caspase-9 and caspase-3 and regulating the mitochondrial pathway. In summary, PIP may directly targets the APE1 protein and further regulates the caspase-9/caspase-3 axis to provide neuroprotection against ischemic retinal injury.
{"title":"Piperine alleviates retinal ischemic injury by mediating the APE1 signaling.","authors":"Qianxiong He, Yannan Chen, Peiwen Chen, Yi Wang, Rong Zou, Feng Zhao, Guangqun Zeng, Lin Zhang, Haiping Liu, Yuanjiang Shi, Liuyi Xiao, Xiaorong Xin","doi":"10.1016/j.neurot.2025.e00781","DOIUrl":"https://doi.org/10.1016/j.neurot.2025.e00781","url":null,"abstract":"<p><p>Ischemic retinal damage is the most common cause of severe vision impairment and blindness. Anti-vascular endothelial growth factor (anti-VEGF) agents have transformed the treatment of retinal ischemic disorders and have become the cornerstone therapy for these conditions. Nonetheless, the risk for systemic and ocular adverse effects necessitates careful consideration. Meanwhile, the therapeutic potential of natural compounds for ischemic retinal injury is increasingly attracting attention. In this study, piperine (PIP), a natural compound derived from pepper, was found to reduce apoptosis by reducing the severity of retinal and optic nerve ischemic damage. However, the precise pharmacological mechanisms of PIP are yet to be fully elucidated. Molecular docking (MD) studies, MD simulations, and surface plasmon resonance experiments were conducted to determine the molecular targets of PIP. Our data revealed that PIP can bind to apurinic/apyrimidinic endonuclease 1 (APE1), thereby inhibiting apoptosis by decreasing the expression of caspase-9 and caspase-3 and regulating the mitochondrial pathway. In summary, PIP may directly targets the APE1 protein and further regulates the caspase-9/caspase-3 axis to provide neuroprotection against ischemic retinal injury.</p>","PeriodicalId":19159,"journal":{"name":"Neurotherapeutics","volume":" ","pages":"e00781"},"PeriodicalIF":6.9,"publicationDate":"2025-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145477008","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}