Methamphetamine (METH) abuse increases worldwide. In addition to its acute life-threatening effects, METH is toxic for dopaminergic neurons, increasing the risk of developing Parkinson's disease. The impact of repeated METH binge consumption on dopaminergic and neurotoxicity markers remains unclear. We exposed mice to a repeated "binge-like" METH regime, consisting of three doses over a 6 h interval, repeated three times with 14-day intervals. After the first binge, spontaneous motor activity decreased markedly but remained normal after subsequent binges. Following the first binge, we observed a 25 % loss of nigral dopaminergic cell bodies and significant axon terminal damage as assessed through striatal silver staining, with minimal further degeneration after additional binges. Dopaminergic markers were substantially depleted after the first and second binges, despite partial recovery between binges, dropping to below 20 % of control levels. By one day after the third binge, tyrosine hydroxylase (TH) and vesicular monoamine transporter 2 (VMAT2) stabilized at 50-60 % of control levels, but the dopamine transporter (DAT) remained at only 25 %, showing less recovery. These changes were accompanied by an evolving neuroinflammation pattern, with a transient microglial surge after the first binge and persistent astroglial and temperature responses. Overall, our findings indicate partial recovery of dopaminergic markers and the development of tolerance to METH neurotoxicity. The robust reduction of DAT after the first binge may contribute to this tolerance to subsequence binges by limiting METH entry into neurons thereby mitigating its neurotoxic effects.
{"title":"Attenuated neurotoxicity after repeated methamphetamine binges linked to dopamine transporter (DAT) decline.","authors":"Noelia Granado, Liliana Mendieta, Yousef Tizabi, Mario Gustavo Murer, Rosario Moratalla","doi":"10.1016/j.nbd.2025.106839","DOIUrl":"https://doi.org/10.1016/j.nbd.2025.106839","url":null,"abstract":"<p><p>Methamphetamine (METH) abuse increases worldwide. In addition to its acute life-threatening effects, METH is toxic for dopaminergic neurons, increasing the risk of developing Parkinson's disease. The impact of repeated METH binge consumption on dopaminergic and neurotoxicity markers remains unclear. We exposed mice to a repeated \"binge-like\" METH regime, consisting of three doses over a 6 h interval, repeated three times with 14-day intervals. After the first binge, spontaneous motor activity decreased markedly but remained normal after subsequent binges. Following the first binge, we observed a 25 % loss of nigral dopaminergic cell bodies and significant axon terminal damage as assessed through striatal silver staining, with minimal further degeneration after additional binges. Dopaminergic markers were substantially depleted after the first and second binges, despite partial recovery between binges, dropping to below 20 % of control levels. By one day after the third binge, tyrosine hydroxylase (TH) and vesicular monoamine transporter 2 (VMAT2) stabilized at 50-60 % of control levels, but the dopamine transporter (DAT) remained at only 25 %, showing less recovery. These changes were accompanied by an evolving neuroinflammation pattern, with a transient microglial surge after the first binge and persistent astroglial and temperature responses. Overall, our findings indicate partial recovery of dopaminergic markers and the development of tolerance to METH neurotoxicity. The robust reduction of DAT after the first binge may contribute to this tolerance to subsequence binges by limiting METH entry into neurons thereby mitigating its neurotoxic effects.</p>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":" ","pages":"106839"},"PeriodicalIF":5.1,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143414222","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 : 2025-02-10DOI: 10.1016/j.nbd.2025.106838
Minjia Xie , Xin Wu , Xi Liu , Longyuan Li, Feng Gu, Xinyu Tao, Bingyi Song, Lei Bai, Di Li, Haitao Shen, Zongqi Wang, Wei Gao
Background
Despite the availability of various antiepileptic treatments, approximately 30 % of epilepsy patients remain refractory to conventional therapies, underscoring the need for neuroprotective strategies. This study investigates the role of GrpEL1 in modulating the mitochondrial unfolded protein response (UPRmt) and its potential protective effects on hippocampal neurons following experimental status epilepticus (SE).
Methods
The effects of GrpEL1 were assessed in vivo using a Lithium-pilocarpine rat model of SE and in vitro with glutamate-treated HT22 hippocampal cells. Protein expression and interactions were analyzed by Western blot, immunofluorescence, and co-immunoprecipitation. Neuronal survival was evaluated through Nissl staining. Mitochondrial function was evaluated aggresome formation, mitochondrial membrane potential (MMP) assays, mitochondrial oxygen consumption rate (OCR) measurements, and behavioral assessments using the Morris water maze.
Results
In the SE rat model, mtHSP70 levels were significantly upregulated in mitochondria, while GrpEL1 expression remained relatively stable. Overexpression of GrpEL1 led to a reduction in neuronal damage and improved functional recovery post-SE. In vitro, GrpEL1 overexpression enhanced the GrpEL1-mtHSP70 interaction, reduced the accumulation of misfolded proteins, and decreased neuronal apoptosis. Furthermore, GrpEL1 overexpression mitigated mitochondrial dysfunction by preserving MMP and improving mitochondrial bioenergetics, as evidenced by enhanced mitochondrial OCR.
Conclusion
GrpEL1 plays a crucial role in maintaining mitochondrial proteostasis and mitigating hippocampal neuronal injury following SE by regulating UPRmt. These findings suggest that GrpEL1 may represent a promising target for therapeutic intervention to protect against seizure-induced neurodegeneration.
{"title":"GrpEL1 overexpression mitigates hippocampal neuron damage via mitochondrial unfolded protein response after experimental status epilepticus","authors":"Minjia Xie , Xin Wu , Xi Liu , Longyuan Li, Feng Gu, Xinyu Tao, Bingyi Song, Lei Bai, Di Li, Haitao Shen, Zongqi Wang, Wei Gao","doi":"10.1016/j.nbd.2025.106838","DOIUrl":"10.1016/j.nbd.2025.106838","url":null,"abstract":"<div><h3>Background</h3><div>Despite the availability of various antiepileptic treatments, approximately 30 % of epilepsy patients remain refractory to conventional therapies, underscoring the need for neuroprotective strategies. This study investigates the role of GrpEL1 in modulating the mitochondrial unfolded protein response (UPRmt) and its potential protective effects on hippocampal neurons following experimental status epilepticus (SE).</div></div><div><h3>Methods</h3><div>The effects of GrpEL1 were assessed <em>in vivo</em> using a Lithium-pilocarpine rat model of SE and <em>in vitro</em> with glutamate-treated HT22 hippocampal cells. Protein expression and interactions were analyzed by Western blot, immunofluorescence, and co-immunoprecipitation. Neuronal survival was evaluated through Nissl staining. Mitochondrial function was evaluated aggresome formation, mitochondrial membrane potential (MMP) assays, mitochondrial oxygen consumption rate (OCR) measurements, and behavioral assessments using the Morris water maze.</div></div><div><h3>Results</h3><div>In the SE rat model, mtHSP70 levels were significantly upregulated in mitochondria, while GrpEL1 expression remained relatively stable. Overexpression of GrpEL1 led to a reduction in neuronal damage and improved functional recovery post-SE. <em>In vitro</em>, GrpEL1 overexpression enhanced the GrpEL1-mtHSP70 interaction, reduced the accumulation of misfolded proteins, and decreased neuronal apoptosis. Furthermore, GrpEL1 overexpression mitigated mitochondrial dysfunction by preserving MMP and improving mitochondrial bioenergetics, as evidenced by enhanced mitochondrial OCR.</div></div><div><h3>Conclusion</h3><div>GrpEL1 plays a crucial role in maintaining mitochondrial proteostasis and mitigating hippocampal neuronal injury following SE by regulating UPRmt. These findings suggest that GrpEL1 may represent a promising target for therapeutic intervention to protect against seizure-induced neurodegeneration.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"206 ","pages":"Article 106838"},"PeriodicalIF":5.1,"publicationDate":"2025-02-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143388313","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-10DOI: 10.1016/j.nbd.2025.106837
Alishah Lakhani, Washington Huang, Chris C Rodgers, Peter Wenner
Mouse models of Fragile X Syndrome (FXS) have demonstrated impairments in sensory-evoked neuronal firing of excitatory and inhibitory neurons. Homeostatic plasticity does not compensate for these changes in activity. Previous work has shown that impairments in homeostatic plasticity mechanisms are observed in FXS, including deficits in synaptic scaling and intrinsic excitability. Here, we aimed to examine how sensory integration changes in vivo following a homeostatic perturbation, unilateral whisker deprivation (WD), in an Fmr1 knock out (KO) mouse model. We used multi-electrode array recordings of neurons in the lightly anesthetized juvenile mouse somatosensory cortex, and found that whisker-evoked responses in layer 5/6 (L5/6) excitatory neurons were weaker in the KO compared to the wild-type (WT). We show that WD in the WT leads to a compensatory increase in the proportion of L5/6 somatosensory neurons that were recruited following whisker stimulation, but this did not occur in the KO. On the other hand, certain compensatory responses were observed in the KO following WD; the firing rate of the whisker-responsive neurons was increased following both a 2- and 7-day WD. Similar to excitatory neurons, we observed increased recruitment of fast spiking (presumed inhibitory) neurons following WD in the WT, but not KO. Our results suggest that certain homeostatic mechanisms are impaired in the KO, while others appear to remain intact. Compromised homeostatic plasticity in development could influence adult sensory processing and long-term cortical organization.
{"title":"Impairment in the homeostatic recruitment of layer 5/6 neurons following whisker stimulation in Fmr1 KO mice.","authors":"Alishah Lakhani, Washington Huang, Chris C Rodgers, Peter Wenner","doi":"10.1016/j.nbd.2025.106837","DOIUrl":"https://doi.org/10.1016/j.nbd.2025.106837","url":null,"abstract":"<p><p>Mouse models of Fragile X Syndrome (FXS) have demonstrated impairments in sensory-evoked neuronal firing of excitatory and inhibitory neurons. Homeostatic plasticity does not compensate for these changes in activity. Previous work has shown that impairments in homeostatic plasticity mechanisms are observed in FXS, including deficits in synaptic scaling and intrinsic excitability. Here, we aimed to examine how sensory integration changes in vivo following a homeostatic perturbation, unilateral whisker deprivation (WD), in an Fmr1 knock out (KO) mouse model. We used multi-electrode array recordings of neurons in the lightly anesthetized juvenile mouse somatosensory cortex, and found that whisker-evoked responses in layer 5/6 (L5/6) excitatory neurons were weaker in the KO compared to the wild-type (WT). We show that WD in the WT leads to a compensatory increase in the proportion of L5/6 somatosensory neurons that were recruited following whisker stimulation, but this did not occur in the KO. On the other hand, certain compensatory responses were observed in the KO following WD; the firing rate of the whisker-responsive neurons was increased following both a 2- and 7-day WD. Similar to excitatory neurons, we observed increased recruitment of fast spiking (presumed inhibitory) neurons following WD in the WT, but not KO. Our results suggest that certain homeostatic mechanisms are impaired in the KO, while others appear to remain intact. Compromised homeostatic plasticity in development could influence adult sensory processing and long-term cortical organization.</p>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":" ","pages":"106837"},"PeriodicalIF":5.1,"publicationDate":"2025-02-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143409336","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}
Background: Glioblastoma (GB) is incurable with a dismal prognosis. Single-cell RNA sequencing (scRNA-seq) is a pivotal tool for studying tumor heterogeneity. The dysregulation of the urea cycle (UC) frequently occurs in tumors, but its characteristics in GB have not been illuminated. This study integrated scRNA-seq UC scores and bulk RNA-seq data to build a GB prognostic model.
Methods: Samples from 3 pairs of GB patients were collected for scRNA-seq analysis. GB-mRNA expression data, clinical data, and SNV mutation data were sourced from the Cancer Genome Atlas (TCGA). GB-mRNA expression data and clinical data were downloaded from the Chinese Glioma Genome Atlas (CGGA). GB RNA-seq data and clinical data were obtained from Gene Expression Omnibus (GEO) database. The R package Seurat was applied for scRNA-seq data processing. UMAP and TSNE were used for dimensionality reduction. UCell enrichment method was employed to score each astrocyte. Monocle algorithm was applied for pseudotime trajectory analysis. CellChat R package was applied for cell communication analysis. Cell labeling was performed on the results of the nine subclusters of astrocytes. The GSE138794 dataset was used to validate the results of single-cell classification. For bulk RNA-seq, univariate Cox and LASSO analyses were undertaken to screen prognostic genes, while multivariate Cox regression analysis was applied to set up a prognostic model. The differences between high-risk (HR) and low-risk (LR) groups were studied in terms of immune infiltration, sensitivity to anti-tumor drugs, etc. We verified the effect of the marker gene on the function of GB cells at the cellular level.
Results: The analysis of scRNA-seq data yielded 7 core cell types. Further clustering of the largest proportion of astrocytes resulted in 9 subclusters. UC score and pseudotime analysis revealed the heterogeneity and differentiation process among subclusters. Subcluster 8 was annotated as an immature astrocyte (marker: CXCL8), and this cell cluster had a higher UC score. The results were validated in the GSE138794 dataset. Combining UC scores, we performed univariate Cox and LASSO to select prognostic genes on bulk RNA-seq data. A prognostic model based on 5 feature genes (RGS4, CTSB, SERPINE2, ID1, and CALD1) was established through multivariate Cox analysis. In addition, patients in the HR group had higher immune infiltration and immune function. Final cell experiments demonstrated that 5 feature genes were highly expressed in GB cells. CALD1 promoted the malignant phenotype of GB cells.
Conclusion: We set up a novel prognostic model for predicting the survival of GB patients by integrating bulk RNA-seq and scRNA-seq data. The risk score was closely correlated with immune infiltration and drug sensitivity, pinpointing it as a promising independent prognostic factor.
{"title":"Joint analysis of single-cell RNA sequencing and bulk transcriptome reveals the heterogeneity of the urea cycle of astrocytes in glioblastoma.","authors":"Minfeng Tong, Qi Tu, Lude Wang, Huahui Chen, Xing Wan, Zhijian Xu","doi":"10.1016/j.nbd.2025.106835","DOIUrl":"https://doi.org/10.1016/j.nbd.2025.106835","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma (GB) is incurable with a dismal prognosis. Single-cell RNA sequencing (scRNA-seq) is a pivotal tool for studying tumor heterogeneity. The dysregulation of the urea cycle (UC) frequently occurs in tumors, but its characteristics in GB have not been illuminated. This study integrated scRNA-seq UC scores and bulk RNA-seq data to build a GB prognostic model.</p><p><strong>Methods: </strong>Samples from 3 pairs of GB patients were collected for scRNA-seq analysis. GB-mRNA expression data, clinical data, and SNV mutation data were sourced from the Cancer Genome Atlas (TCGA). GB-mRNA expression data and clinical data were downloaded from the Chinese Glioma Genome Atlas (CGGA). GB RNA-seq data and clinical data were obtained from Gene Expression Omnibus (GEO) database. The R package Seurat was applied for scRNA-seq data processing. UMAP and TSNE were used for dimensionality reduction. UCell enrichment method was employed to score each astrocyte. Monocle algorithm was applied for pseudotime trajectory analysis. CellChat R package was applied for cell communication analysis. Cell labeling was performed on the results of the nine subclusters of astrocytes. The GSE138794 dataset was used to validate the results of single-cell classification. For bulk RNA-seq, univariate Cox and LASSO analyses were undertaken to screen prognostic genes, while multivariate Cox regression analysis was applied to set up a prognostic model. The differences between high-risk (HR) and low-risk (LR) groups were studied in terms of immune infiltration, sensitivity to anti-tumor drugs, etc. We verified the effect of the marker gene on the function of GB cells at the cellular level.</p><p><strong>Results: </strong>The analysis of scRNA-seq data yielded 7 core cell types. Further clustering of the largest proportion of astrocytes resulted in 9 subclusters. UC score and pseudotime analysis revealed the heterogeneity and differentiation process among subclusters. Subcluster 8 was annotated as an immature astrocyte (marker: CXCL8), and this cell cluster had a higher UC score. The results were validated in the GSE138794 dataset. Combining UC scores, we performed univariate Cox and LASSO to select prognostic genes on bulk RNA-seq data. A prognostic model based on 5 feature genes (RGS4, CTSB, SERPINE2, ID1, and CALD1) was established through multivariate Cox analysis. In addition, patients in the HR group had higher immune infiltration and immune function. Final cell experiments demonstrated that 5 feature genes were highly expressed in GB cells. CALD1 promoted the malignant phenotype of GB cells.</p><p><strong>Conclusion: </strong>We set up a novel prognostic model for predicting the survival of GB patients by integrating bulk RNA-seq and scRNA-seq data. The risk score was closely correlated with immune infiltration and drug sensitivity, pinpointing it as a promising independent prognostic factor.</p>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":" ","pages":"106835"},"PeriodicalIF":5.1,"publicationDate":"2025-02-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143409351","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 : 2025-02-06DOI: 10.1016/j.nbd.2025.106833
Huinan Zhang , Xinzhe Du , Tian Gao , Xing Wang , Huifeng Zhang , Manyang Yu , Jing Huang
Microglial canonical transient receptor potential channel 1 (TRPC1) has been proposed to influence neuroinflammation after cerebral ischemia and reperfusion injury (CIRI), however, the underlying mechanism remains poorly understood. This study demonstrates that TRPC1 is modified by small ubiquitin-related modifier (SUMO)ylation. Our findings suggest a notable increase in microglial TRPC1 SUMOylation within both the middle cerebral artery occlusion reperfusion (MCAO/R) model and the in vitro oxygen-glucose deprivation/regeneration model. Mice with a loss of TRPC1 SUMOylation in microglia exhibited improved stroke outcomes including reduced behavior deficits, infarct volume, blood brain barrier damage as well as neuronal apoptosis. Mechanistically, SUMOylation of microglial TRPC1 exacerbated neutrophil infiltration into the peri-infarct area. Additionally, SUMOylated TRPC1 activates the Nod-like receptor protein (NLRP) 3 signaling pathway in microglia and stimulates multiple CC-chemokine ligands and C-X-C motif ligand chemokines after MCAO/R. SUMOylated TRPC1 facilitates the interaction between TRPC1 and β-arrestin2 (ARRB2), a negative regulator of NLRP3 inflammasome, which disrupts the NLPR3/ARRB2 complex and stimulates the activation of the NLPR3 signaling pathway. Furthermore, ARRB2 directly binds to the residues 46 to 61 of TRPC1 N terminus, which is enhanced by TRPC1 SUMOylation. Collectively, our findings demonstrate a previously unidentified mechanism by which SUMOylated TRPC1 in microglia regulates leukocyte infiltration after stroke, suggesting that the inhibition of microglial TRPC1 SUMOylation may provide therapeutic benefits for CIRI.
{"title":"Microglia TRPC1 SUMOylation drives neuroinflammation after stroke by modulating NLRP3 activity via increasing TRPC1 interaction with ARRB2","authors":"Huinan Zhang , Xinzhe Du , Tian Gao , Xing Wang , Huifeng Zhang , Manyang Yu , Jing Huang","doi":"10.1016/j.nbd.2025.106833","DOIUrl":"10.1016/j.nbd.2025.106833","url":null,"abstract":"<div><div>Microglial canonical transient receptor potential channel 1 (TRPC1) has been proposed to influence neuroinflammation after cerebral ischemia and reperfusion injury (CIRI), however, the underlying mechanism remains poorly understood. This study demonstrates that TRPC1 is modified by small ubiquitin-related modifier (SUMO)ylation. Our findings suggest a notable increase in microglial TRPC1 SUMOylation within both the middle cerebral artery occlusion reperfusion (MCAO/R) model and the in vitro oxygen-glucose deprivation/regeneration model. Mice with a loss of TRPC1 SUMOylation in microglia exhibited improved stroke outcomes including reduced behavior deficits, infarct volume, blood brain barrier damage as well as neuronal apoptosis. Mechanistically, SUMOylation of microglial TRPC1 exacerbated neutrophil infiltration into the peri-infarct area. Additionally, SUMOylated TRPC1 activates the Nod-like receptor protein (NLRP) 3 signaling pathway in microglia and stimulates multiple CC-chemokine ligands and C-X-C motif ligand chemokines after MCAO/R. SUMOylated TRPC1 facilitates the interaction between TRPC1 and β-arrestin2 (ARRB2), a negative regulator of NLRP3 inflammasome, which disrupts the NLPR3/ARRB2 complex and stimulates the activation of the NLPR3 signaling pathway. Furthermore, ARRB2 directly binds to the residues 46 to 61 of TRPC1 N terminus, which is enhanced by TRPC1 SUMOylation. Collectively, our findings demonstrate a previously unidentified mechanism by which SUMOylated TRPC1 in microglia regulates leukocyte infiltration after stroke, suggesting that the inhibition of microglial TRPC1 SUMOylation may provide therapeutic benefits for CIRI.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"206 ","pages":"Article 106833"},"PeriodicalIF":5.1,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143372691","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-05DOI: 10.1016/j.nbd.2025.106834
Matthew V Prifti, Oluwademilade Nuga, Ryan O Dulay, Nikhil C Patel, Truman Kula, Kozeta Libohova, Autumn Jackson-Butler, Wei-Ling Tsou, Kristin Richardson, Sokol V Todi
Dentatorubral-pallidoluysian atrophy (DRPLA) is a neurodegenerative disorder that presents with ataxia, dementia and epilepsy. As a member of the polyglutamine family of diseases, DRPLA is caused by abnormal CAG triplet expansion beyond 48 repeats in the protein-coding region of ATROPHIN 1 (ATN1), a transcriptional co-repressor. To better understand DRPLA, we generated new Drosophila lines that can be induced to express full-length, human ATN1 with a normal (Q7) or pathogenic (Q88) repeat in a variety of cells, including neuronal, glial or any other type of tissue. Expression of ATN1 is toxic, with the polyglutamine-expanded version being consistently more problematic than wild-type ATN1. Fly motility, longevity and internal structures are negatively impacted by pathogenic ATN1. RNA-seq identified altered protein quality control and immune pathways in the presence of pathogenic ATN1. Based on these data, we conducted genetic experiments that confirmed the role of protein quality control components that ameliorate or exacerbate ATN1 toxicity. Hsc70-3, a chaperone, arose as a likely suppressor of toxicity. VCP (a proteasome-related AAA ATPase), Rpn11 (a proteasome-related deubiquitinase) and select DnaJ proteins (co-chaperones) were inconsistently protective, depending on the tissues where they were expressed. Lastly, informed by RNA-seq data that exercise-related genes may also be involved in this model of DRPLA, we conducted short-term exercise, which improved overall fly motility. This new model of DRPLA will prove important to understanding this understudied disease and will help to identify therapeutic targets for it.
{"title":"Insights into dentatorubral-pallidoluysian atrophy from a new Drosophila model of disease.","authors":"Matthew V Prifti, Oluwademilade Nuga, Ryan O Dulay, Nikhil C Patel, Truman Kula, Kozeta Libohova, Autumn Jackson-Butler, Wei-Ling Tsou, Kristin Richardson, Sokol V Todi","doi":"10.1016/j.nbd.2025.106834","DOIUrl":"10.1016/j.nbd.2025.106834","url":null,"abstract":"<p><p>Dentatorubral-pallidoluysian atrophy (DRPLA) is a neurodegenerative disorder that presents with ataxia, dementia and epilepsy. As a member of the polyglutamine family of diseases, DRPLA is caused by abnormal CAG triplet expansion beyond 48 repeats in the protein-coding region of ATROPHIN 1 (ATN1), a transcriptional co-repressor. To better understand DRPLA, we generated new Drosophila lines that can be induced to express full-length, human ATN1 with a normal (Q7) or pathogenic (Q88) repeat in a variety of cells, including neuronal, glial or any other type of tissue. Expression of ATN1 is toxic, with the polyglutamine-expanded version being consistently more problematic than wild-type ATN1. Fly motility, longevity and internal structures are negatively impacted by pathogenic ATN1. RNA-seq identified altered protein quality control and immune pathways in the presence of pathogenic ATN1. Based on these data, we conducted genetic experiments that confirmed the role of protein quality control components that ameliorate or exacerbate ATN1 toxicity. Hsc70-3, a chaperone, arose as a likely suppressor of toxicity. VCP (a proteasome-related AAA ATPase), Rpn11 (a proteasome-related deubiquitinase) and select DnaJ proteins (co-chaperones) were inconsistently protective, depending on the tissues where they were expressed. Lastly, informed by RNA-seq data that exercise-related genes may also be involved in this model of DRPLA, we conducted short-term exercise, which improved overall fly motility. This new model of DRPLA will prove important to understanding this understudied disease and will help to identify therapeutic targets for it.</p>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":" ","pages":"106834"},"PeriodicalIF":5.1,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143374478","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 : 2025-02-05DOI: 10.1016/j.nbd.2025.106816
LiHua Deng , Liu Feng , JingWen Li , YongHua Huang , PeiLing Ou , LinFeng Shi , Hui Chen , YuHan Zhang , LiMeng Dai , Yuan He , Chen Wei , HuaFu Chen , Jian Wang , Leinian Li , Chen Liu
Spinocerebellar ataxia type 3 (SCA3), a neurodegenerative disorder caused by excess CAG repeats in the ATXN3 gene, leads to progressive cerebellar ataxia and other symptoms. The results of previous studies suggest that trace element dysregulation contributes to neurodegenerative disorder onset. Here, we investigated the relationships of trace element dysregulation with CAG repeat length, clinical severity, and brain structural and functional connectivity in 45 patients with SCA3 and 44 healthy controls (HCs). Blood levels of lithium (Li), selenium (Se), and copper (Cu) were significantly lower in patients with SCA3 than in HCs; Li and Se levels were negatively correlated with CAG repeat length, especially in the manifest subgroup. Diffusion tensor imaging combined with resting-state functional magnetic resonance imaging revealed that Li levels were negatively correlated with fractional anisotropy in the white matter (WM) of bilateral frontal and parietal regions; tractography mapping showed disorder structural connectivity of Li-associated region nerve fiber pathways in patients with SCA3. Dynamic causal modeling analyses showed bidirectional causal connectivity from the inferior parietal lobule(IPL) to the cerebellum was significantly correlated with the blood level of Li in patients with SCA3. Time series correlation-based functional connectivity analysis revealed that the intrinsic connectivities of the bilateral dorsal premotor cortex(PMd) and IPL with local cerebellar regions were significantly weaker in patients with SCA3 than in HCs. Our results suggest that trace element dysregulation, especially Li deficiency, induces brain alterations and clinical manifestations in patients with SCA3; Li supplementation may be beneficial for WM or astrocytes in this patient population.
{"title":"Effects of trace element dysregulation on brain structure and function in spinocerebellar Ataxia type 3","authors":"LiHua Deng , Liu Feng , JingWen Li , YongHua Huang , PeiLing Ou , LinFeng Shi , Hui Chen , YuHan Zhang , LiMeng Dai , Yuan He , Chen Wei , HuaFu Chen , Jian Wang , Leinian Li , Chen Liu","doi":"10.1016/j.nbd.2025.106816","DOIUrl":"10.1016/j.nbd.2025.106816","url":null,"abstract":"<div><div>Spinocerebellar ataxia type 3 (SCA3), a neurodegenerative disorder caused by excess CAG repeats in the <em>ATXN3</em> gene, leads to progressive cerebellar ataxia and other symptoms. The results of previous studies suggest that trace element dysregulation contributes to neurodegenerative disorder onset. Here, we investigated the relationships of trace element dysregulation with CAG repeat length, clinical severity, and brain structural and functional connectivity in 45 patients with SCA3 and 44 healthy controls (HCs). Blood levels of lithium (Li), selenium (Se), and copper (Cu) were significantly lower in patients with SCA3 than in HCs; Li and Se levels were negatively correlated with CAG repeat length, especially in the manifest subgroup. Diffusion tensor imaging combined with resting-state functional magnetic resonance imaging revealed that Li levels were negatively correlated with fractional anisotropy in the white matter (WM) of bilateral frontal and parietal regions; tractography mapping showed disorder structural connectivity of Li-associated region nerve fiber pathways in patients with SCA3. Dynamic causal modeling analyses showed bidirectional causal connectivity from the inferior parietal lobule(IPL) to the cerebellum was significantly correlated with the blood level of Li in patients with SCA3. Time series correlation-based functional connectivity analysis revealed that the intrinsic connectivities of the bilateral dorsal premotor cortex(PMd) and IPL with local cerebellar regions were significantly weaker in patients with SCA3 than in HCs. Our results suggest that trace element dysregulation, especially Li deficiency, induces brain alterations and clinical manifestations in patients with SCA3; Li supplementation may be beneficial for WM or astrocytes in this patient population.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"207 ","pages":"Article 106816"},"PeriodicalIF":5.1,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143374463","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-04DOI: 10.1016/j.nbd.2025.106832
Akram Zamani , Adam K. Walker , David K. Wright
Dysfunctional Tar DNA binding protein-43 (TDP-43) is found in approximately 95 % of all people with amyotrophic lateral sclerosis (ALS). Recent evidence suggests that the glymphatic system, which clears the brain of waste proteins, is impaired in ALS and may contribute to the accumulation of TDP-43. This study extends this work to investigate how glymphatic function changes over time in the rNLS8 doxycycline (Dox)-dependent TDP-43 mouse model of ALS. Motor function, advanced MRI biomarkers of neurodegeneration, and cortical glymphatic pathway gene expression were assessed together with dynamic contrast-enhanced MRI (DCE-MRI) assessment of glymphatic function at 0-, 3-, 7-, and 21-days after removing mice from Dox feed to initiate cytoplasmic human TDP-43 expression. A trend toward increased glymphatic influx was observed at 3-days post-Dox, together with MRI evidence of brain changes that occurred in the absence of hind-limb clasping and motor impairment. Glymphatic flow is facilitated by aquaporin-4 (AQP4) water channels polarized to astrocytic end feet. We found that while glymphatic function normalized to control levels at 7-days post-Dox, AQP4 expression in the cortex was significantly decreased. After 3-weeks of human TDP-43 expression, glymphatic dysfunction, weight loss, neurodegeneration, motor impairments and astrogliosis were observed. Our findings highlight early glymphatic dysfunction in ALS, suggesting its potential as a therapeutic target.
{"title":"Glymphatic dysfunction and neurodegeneration in ALS: Longitudinal insights from rNLS8 TDP-43 mice","authors":"Akram Zamani , Adam K. Walker , David K. Wright","doi":"10.1016/j.nbd.2025.106832","DOIUrl":"10.1016/j.nbd.2025.106832","url":null,"abstract":"<div><div>Dysfunctional Tar DNA binding protein-43 (TDP-43) is found in approximately 95 % of all people with amyotrophic lateral sclerosis (ALS). Recent evidence suggests that the glymphatic system, which clears the brain of waste proteins, is impaired in ALS and may contribute to the accumulation of TDP-43. This study extends this work to investigate how glymphatic function changes over time in the rNLS8 doxycycline (Dox)-dependent TDP-43 mouse model of ALS. Motor function, advanced MRI biomarkers of neurodegeneration, and cortical glymphatic pathway gene expression were assessed together with dynamic contrast-enhanced MRI (DCE-MRI) assessment of glymphatic function at 0-, 3-, 7-, and 21-days after removing mice from Dox feed to initiate cytoplasmic human TDP-43 expression. A trend toward increased glymphatic influx was observed at 3-days post-Dox, together with MRI evidence of brain changes that occurred in the absence of hind-limb clasping and motor impairment. Glymphatic flow is facilitated by aquaporin-4 (AQP4) water channels polarized to astrocytic end feet. We found that while glymphatic function normalized to control levels at 7-days post-Dox, AQP4 expression in the cortex was significantly decreased. After 3-weeks of human TDP-43 expression, glymphatic dysfunction, weight loss, neurodegeneration, motor impairments and astrogliosis were observed. Our findings highlight early glymphatic dysfunction in ALS, suggesting its potential as a therapeutic target.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"206 ","pages":"Article 106832"},"PeriodicalIF":5.1,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143236057","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-04DOI: 10.1016/j.nbd.2025.106831
Enrique Pérez-Garci , Kateryna Pysanenko , Giorgio Rizzi , Florian Studer , Daniel Ulrich , Thorsten Fritzius , Simon Früh , Alessandra Porcu , Valérie Besseyrias , Adolf Melichar , Martin Gassmann , Tania Rinaldi Barkat , Rostislav Tureček , Kelly R. Tan , Bernhard Bettler
During GABAergic synaptic transmission, G protein-coupled GABAB receptors (GBRs) activate K+ channels that prolong the duration of inhibitory postsynaptic potentials (IPSPs). We now show that KCTD16, an auxiliary GBR subunit, anchors hyperpolarization-activated cyclic nucleotide-gated (HCN) channels containing HCN2/HCN3 subunits to GBRs. In dopamine neurons of the VTA (DAVTA neurons), this interaction facilitates activation of HCN channels via hyperpolarization during IPSPs, counteracting the GBR-mediated late phase of these IPSPs. Consequently, disruption of the GBR/HCN complex in KCTD16−/− mice leads to prolonged optogenetic inhibition of DAVTA neuron firing. KCTD16−/− mice exhibit increased anxiety-like behavior in response to stress - a behavior replicated by CRISPR/Cas9-mediated KCTD16 ablation in DAVTA neurons or by intra-VTA infusion of an HCN antagonist in wild-type mice. Our findings support that the retention of HCN channels at GABAergic synapses by GBRs in DAVTA neurons provides a negative feedback mechanism that restricts IPSP duration and mitigates the development of anxiety.
{"title":"Binding of HCN channels to GABAB receptors in dopamine neurons of the VTA limits synaptic inhibition and prevents the development of anxiety","authors":"Enrique Pérez-Garci , Kateryna Pysanenko , Giorgio Rizzi , Florian Studer , Daniel Ulrich , Thorsten Fritzius , Simon Früh , Alessandra Porcu , Valérie Besseyrias , Adolf Melichar , Martin Gassmann , Tania Rinaldi Barkat , Rostislav Tureček , Kelly R. Tan , Bernhard Bettler","doi":"10.1016/j.nbd.2025.106831","DOIUrl":"10.1016/j.nbd.2025.106831","url":null,"abstract":"<div><div>During GABAergic synaptic transmission, G protein-coupled GABA<sub>B</sub> receptors (GBRs) activate K<sup>+</sup> channels that prolong the duration of inhibitory postsynaptic potentials (IPSPs). We now show that KCTD16, an auxiliary GBR subunit, anchors hyperpolarization-activated cyclic nucleotide-gated (HCN) channels containing HCN2/HCN3 subunits to GBRs. In dopamine neurons of the VTA (DA<sup>VTA</sup> neurons), this interaction facilitates activation of HCN channels via hyperpolarization during IPSPs, counteracting the GBR-mediated late phase of these IPSPs. Consequently, disruption of the GBR/HCN complex in <em>KCTD16</em><sup>−/−</sup> mice leads to prolonged optogenetic inhibition of DA<sup>VTA</sup> neuron firing. <em>KCTD16</em><sup><em>−/−</em></sup> mice exhibit increased anxiety-like behavior in response to stress - a behavior replicated by CRISPR/Cas9-mediated <em>KCTD16</em> ablation in DA<sup>VTA</sup> neurons or by intra-VTA infusion of an HCN antagonist in wild-type mice. Our findings support that the retention of HCN channels at GABAergic synapses by GBRs in DA<sup>VTA</sup> neurons provides a negative feedback mechanism that restricts IPSP duration and mitigates the development of anxiety.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"206 ","pages":"Article 106831"},"PeriodicalIF":5.1,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143350522","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: This study investigated the subthalamic nucleus (STN) function and deep brain stimulation (DBS) effects on single-unit activity (SUA) in Parkinson's disease (PD) patients with dysarthria.
Methods: After presurgical speech analysis, we recorded STN neuronal activities while PD patients (n = 16) articulated Chinese Pinyin consonants. The Pinyin consonants were categorized by the manner and place of articulation for SUA cluster analysis. The cohort was then divided into normal articulation and dysarthria groups based on diadochokinetic (DDK) assessments. The STN SUA patterns, represented by the mean firing rate (FR), peak time, and response intensity during articulation, were analyzed and compared between the two groups. Finally, a stimulation cohort of 7 PD patients was included to test articulation and SUA pattern changes following intraoperative DBS.
Results: Clustering analysis of STN neuronal firing patterns demonstrated that neurons encode articulation by grouping consonants with the same manner of articulation into distinct clusters. Using k-means clustering, we further classified SUAs into two waveform types: negative spikes (type 1) and positive spikes (type 2). Dysarthria patients exhibited an increased mean FR of type 1 spikes and a reduced response intensity of type 2 spikes. During intraoperative stimulation, PD patients showed accelerated DDK, accompanied by a decrease in type 1 mean FR and an increase in type 2 mean FR.
Conclusion: Our findings indicate the crucial role of the STN in consonant encoding and dysarthria at the single-unit level. Both SUA firing patterns in the STN and DDK performance can be modulated by DBS.
{"title":"Abnormal neuronal activity in the subthalamic nucleus contributes to dysarthria in patients with Parkinson's disease.","authors":"Yu Diao, Zixiao Yin, Baotian Zhao, Yichen Xu, Yin Jiang, Yanling Yin, Anchao Yang, Yanming Zhu, Jan Hlavnicka, Jianguo Zhang","doi":"10.1016/j.nbd.2025.106830","DOIUrl":"https://doi.org/10.1016/j.nbd.2025.106830","url":null,"abstract":"<p><strong>Background: </strong>This study investigated the subthalamic nucleus (STN) function and deep brain stimulation (DBS) effects on single-unit activity (SUA) in Parkinson's disease (PD) patients with dysarthria.</p><p><strong>Methods: </strong>After presurgical speech analysis, we recorded STN neuronal activities while PD patients (n = 16) articulated Chinese Pinyin consonants. The Pinyin consonants were categorized by the manner and place of articulation for SUA cluster analysis. The cohort was then divided into normal articulation and dysarthria groups based on diadochokinetic (DDK) assessments. The STN SUA patterns, represented by the mean firing rate (FR), peak time, and response intensity during articulation, were analyzed and compared between the two groups. Finally, a stimulation cohort of 7 PD patients was included to test articulation and SUA pattern changes following intraoperative DBS.</p><p><strong>Results: </strong>Clustering analysis of STN neuronal firing patterns demonstrated that neurons encode articulation by grouping consonants with the same manner of articulation into distinct clusters. Using k-means clustering, we further classified SUAs into two waveform types: negative spikes (type 1) and positive spikes (type 2). Dysarthria patients exhibited an increased mean FR of type 1 spikes and a reduced response intensity of type 2 spikes. During intraoperative stimulation, PD patients showed accelerated DDK, accompanied by a decrease in type 1 mean FR and an increase in type 2 mean FR.</p><p><strong>Conclusion: </strong>Our findings indicate the crucial role of the STN in consonant encoding and dysarthria at the single-unit level. Both SUA firing patterns in the STN and DDK performance can be modulated by DBS.</p>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":" ","pages":"106830"},"PeriodicalIF":5.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143256017","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}