1
|
Bayam E, Tilly P, Collins SC, Rivera Alvarez J, Kannan M, Tonneau L, Brivio E, Rinaldi B, Lecat R, Schwaller N, Cotellessa L, Maddirevula S, Monteiro F, Guardia CM, Kitajima JP, Kok F, Kato M, Hamed AAA, Salih MA, Al Tala S, Hashem MO, Tada H, Saitsu H, Stabile M, Giacobini P, Friant S, Yüksel Z, Nakashima M, Alkuraya FS, Yalcin B, Godin JD. Bi-allelic variants in WDR47 cause a complex neurodevelopmental syndrome. EMBO Mol Med 2025; 17:129-168. [PMID: 39609633 DOI: 10.1038/s44321-024-00178-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/30/2024] Open
Abstract
Brain development requires the coordinated growth of structures and cues that are essential for forming neural circuits and cognitive functions. The corpus callosum, the largest interhemispheric connection, is formed by the axons of callosal projection neurons through a series of tightly regulated cellular events, including neuronal specification, migration, axon extension and branching. Defects in any of those steps can lead to a range of disorders known as syndromic corpus callosum dysgenesis (CCD). We report five unrelated families carrying bi-allelic variants in WDR47 presenting with CCD together with other neuroanatomical phenotypes such as microcephaly and enlarged ventricles. Using in vitro and in vivo mouse models and complementation assays, we show that WDR47 is required for survival of callosal neurons by contributing to the maintenance of mitochondrial and microtubule homeostasis. We further propose that severity of the CCD phenotype is determined by the degree of the loss of function caused by the human variants. Taken together, we identify WDR47 as a causative gene of a new neurodevelopmental syndrome characterized by corpus callosum abnormalities and other neuroanatomical malformations.
Collapse
Affiliation(s)
- Efil Bayam
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, F-67404, France.
- Centre National de la Recherche Scientifique, CNRS, UMR7104, Illkirch, F-67404, France.
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, F-67404, France.
- Université de Strasbourg, Strasbourg, F-67000, France.
| | - Peggy Tilly
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, F-67404, France
- Centre National de la Recherche Scientifique, CNRS, UMR7104, Illkirch, F-67404, France
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, F-67404, France
- Université de Strasbourg, Strasbourg, F-67000, France
| | - Stephan C Collins
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, F-67404, France
- Centre National de la Recherche Scientifique, CNRS, UMR7104, Illkirch, F-67404, France
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, F-67404, France
- Université de Strasbourg, Strasbourg, F-67000, France
- Université de Bourgogne, INSERM UMR1231, 21000, Dijon, France
| | - José Rivera Alvarez
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, F-67404, France
- Centre National de la Recherche Scientifique, CNRS, UMR7104, Illkirch, F-67404, France
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, F-67404, France
- Université de Strasbourg, Strasbourg, F-67000, France
| | - Meghna Kannan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, F-67404, France
- Centre National de la Recherche Scientifique, CNRS, UMR7104, Illkirch, F-67404, France
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, F-67404, France
- Université de Strasbourg, Strasbourg, F-67000, France
| | - Lucile Tonneau
- Université de Bourgogne, INSERM UMR1231, 21000, Dijon, France
| | - Elena Brivio
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, F-67404, France
- Centre National de la Recherche Scientifique, CNRS, UMR7104, Illkirch, F-67404, France
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, F-67404, France
- Université de Strasbourg, Strasbourg, F-67000, France
| | - Bruno Rinaldi
- Université de Strasbourg, CNRS, GMGM UMR7156, F-67000, Strasbourg, France
- INSERM, U1112, CRBS (Centre de recherche en biomédecine de Strasbourg), Université de Strasbourg, Strasbourg, F-67000, France
| | - Romain Lecat
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, F-67404, France
- Centre National de la Recherche Scientifique, CNRS, UMR7104, Illkirch, F-67404, France
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, F-67404, France
- Université de Strasbourg, Strasbourg, F-67000, France
| | - Noémie Schwaller
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, F-67404, France
- Centre National de la Recherche Scientifique, CNRS, UMR7104, Illkirch, F-67404, France
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, F-67404, France
- Université de Strasbourg, Strasbourg, F-67000, France
| | - Ludovica Cotellessa
- Université de Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition UMR-S 1172, Lille, France
| | - Sateesh Maddirevula
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Carlos M Guardia
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | | | - Fernando Kok
- Mendelics Análise Genomica SA, CEP 02511-000, Sao Paulo, Brazil
- Department of Neurology, University of Sao Paulo School of Medicine, 01246-903, Sao Paulo, Brazil
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Ahlam A A Hamed
- Department of Pediatric and Child Health, Faculty of Medicine University of Khartoum, Khartoum, Sudan
| | - Mustafa A Salih
- Health Sector, King Abdulaziz City for Science and Technology, Riyadh, 11442, Saudi Arabia
| | - Saeed Al Tala
- Department of Pediatrics, Genetic Unit, Armed Forces Hospital, Khamis Mushayt, Saudi Arabia
| | - Mais O Hashem
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Hiroko Tada
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-0057, Japan
- Division of Pediatrics, Chibaken Saiseikai Narashino Hospital, Chiba, 275-8580, Japan
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuuo-ku, Hamamatsu, 431-3192, Japan
| | - Mariano Stabile
- Center of Genetics and Prenatal Diagnosis "Zygote", 84131, Salerno, Italy
| | - Paolo Giacobini
- Université de Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition UMR-S 1172, Lille, France
| | - Sylvie Friant
- Université de Strasbourg, CNRS, GMGM UMR7156, F-67000, Strasbourg, France
- PCBIS-IMPReSs, Plateforme de Chimie Biologique Intégrative de Strasbourg, UAR 3286 CNRS/Université de Strasbourg, 67400, Illkirch, France
| | - Zafer Yüksel
- Human Genetics, Bioscientia GmbH, Ingelheim, Germany
| | - Mitsuko Nakashima
- Department of Biochemistry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuuo-ku, Hamamatsu, 431-3192, Japan
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Binnaz Yalcin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, F-67404, France.
- Centre National de la Recherche Scientifique, CNRS, UMR7104, Illkirch, F-67404, France.
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, F-67404, France.
- Université de Strasbourg, Strasbourg, F-67000, France.
- INSERM UMR1231, Université de Bourgogne, 21000, Dijon, France.
| | - Juliette D Godin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, F-67404, France.
- Centre National de la Recherche Scientifique, CNRS, UMR7104, Illkirch, F-67404, France.
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, F-67404, France.
- Université de Strasbourg, Strasbourg, F-67000, France.
| |
Collapse
|
2
|
Gao L, Yang XN, Dong YX, Han YJ, Zhang XY, Zhou XL, Liu Y, Liu F, Fang JS, Ji JL, Gao ZR, Qin XM. The potential therapeutic strategy in combating neurodegenerative diseases: Focusing on natural products. Pharmacol Ther 2024; 264:108751. [PMID: 39522697 DOI: 10.1016/j.pharmthera.2024.108751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/25/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), Huntington disease (HD), and Multiple sclerosis (MS), pose a significant global health challenge due to their intricate pathology and limited therapeutic interventions. Natural products represent invaluable reservoirs for combating these neurodegenerative diseases by targeting key pathological hallmarks such as protein aggregation, synaptic dysfunction, aberrant proteostasis, cytoskeletal abnormalities, altered energy homeostasis, inflammation, and neuronal cell death. This review provides an in-depth analysis of the mechanisms and therapeutic targets of natural products for their neuroprotective effects. Furthermore, it elucidates the current progress of clinical trials investigating the potential of natural products in delaying neurodegeneration. The objective of this review is to enhance the comprehension of natural products in the prevention and treatment of neurodegenerative diseases, offering new insights and potential avenues for future pharmaceutical research.
Collapse
Affiliation(s)
- Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Xi-Na Yang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Yi-Xiao Dong
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Yi-Jia Han
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Xin-Yue Zhang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Xin-Le Zhou
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Ying Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Fang Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Jian-Song Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jian-Long Ji
- College of Integrated Circuits, Taiyuan University of Technology, Taiyuan, China.
| | - Zheng-Run Gao
- Songjiang Research Institute, Songjiang Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China.
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China.
| |
Collapse
|
3
|
Chu L, Zeng D, He Y, Dong X, Li Q, Liao X, Zhao T, Chen X, Lei T, Men W, Wang Y, Wang D, Hu M, Pan Z, Tan S, Gao JH, Qin S, Tao S, Dong Q, He Y, Li S. Segregation of the regional radiomics similarity network exhibited an increase from late childhood to early adolescence: A developmental investigation. Neuroimage 2024; 302:120893. [PMID: 39426642 DOI: 10.1016/j.neuroimage.2024.120893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 09/15/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024] Open
Abstract
Brain development is characterized by an increase in structural and functional segregation, which supports the specialization of cognitive processes within the context of network neuroscience. In this study, we investigated age-related changes in morphological segregation using individual Regional Radiomics Similarity Networks (R2SNs) constructed with a longitudinal dataset of 494 T1-weighted MR scans from 309 typically developing children aged 6.2 to 13 years at baseline. Segertation indices were defined as the relative difference in connectivity strengths within and between modules and cacluated at the global, system and local levels. Linear mixed-effect models revealed longitudinal increases in both global and system segregation indices, particularly within the limbic and dorsal attention network, and decreases within the ventral attention network. Superior performance in working memory and inhibitory control was associated with higher system-level segregation indices in default, frontoparietal, ventral attention, somatomotor and subcortical systems, and lower local segregation indices in visual network regions, regardless of age. Furthermore, gene enrichment analysis revealed correlations between age-related changes in local segregation indices and regional expression levels of genes related to developmental processes. These findings provide novel insights into typical brain developmental changes using R2SN-derived segregation indices, offering a valuable tool for understanding human brain structural and cognitive maturation.
Collapse
Affiliation(s)
- Lei Chu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science & Medical Engineering, Beihang University, Beijing 100083, China
| | - Debin Zeng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science & Medical Engineering, Beihang University, Beijing 100083, China
| | - Yirong He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Xiaoxi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Qiongling Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Xuhong Liao
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Tengda Zhao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Xiaodan Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Tianyuan Lei
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Weiwei Men
- Beijing City Key Laboratory for Medical Physics and Engineering, Institute of Heavy Ion Physics, School of Physics, Peking University, Beijing 100871, China; Zhejiang Philosophy and Social Science Laboratory for Research in Early Development and Childcare, Hangzhou Normal University, Hangzhou 311121, China
| | - Yanpei Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Daoyang Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Zhejiang Philosophy and Social Science Laboratory for Research in Early Development and Childcare, Hangzhou Normal University, Hangzhou 311121, China
| | - Mingming Hu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Zhiying Pan
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Shuping Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Jia-Hong Gao
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing City Key Laboratory for Medical Physics and Engineering, Institute of Heavy Ion Physics, School of Physics, Peking University, Beijing 100871, China; IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Shaozheng Qin
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Sha Tao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Qi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Yong He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; Chinese Institute for Brain Research, Beijing 102206, China.
| | - Shuyu Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
4
|
Bartelt LC, Switonski PM, Adamek G, Longo F, Carvalho J, Duvick LA, Jarrah SI, McLoughlin HS, Scoles DR, Pulst SM, Orr HT, Hull C, Lowe CB, La Spada AR. Dysregulation of zebrin-II cell subtypes in the cerebellum is a shared feature across polyglutamine ataxia mouse models and patients. Sci Transl Med 2024; 16:eadn5449. [PMID: 39504355 DOI: 10.1126/scitranslmed.adn5449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/12/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024]
Abstract
Spinocerebellar ataxia type 7 (SCA7) is a genetic neurodegenerative disorder caused by a CAG-polyglutamine repeat expansion. Purkinje cells (PCs) are central to the pathology of ataxias, but their low abundance in the cerebellum underrepresents their transcriptomes in sequencing assays. To address this issue, we developed a PC enrichment protocol and sequenced individual nuclei from mice and patients with SCA7. Single-nucleus RNA sequencing in SCA7-266Q mice revealed dysregulation of cell identity genes affecting glia and PCs. Specifically, genes marking zebrin-II PC subtypes accounted for the highest proportion of DEGs in symptomatic SCA7-266Q mice. These transcriptomic changes in SCA7-266Q mice were associated with increased numbers of inhibitory synapses as quantified by immunohistochemistry and reduced spiking of PCs in acute brain slices. Dysregulation of zebrin-II cell subtypes was the predominant signal in PCs of SCA7-266Q mice and was associated with the loss of zebrin-II striping in the cerebellum at motor symptom onset. We furthermore demonstrated zebrin-II stripe degradation in additional mouse models of polyglutamine ataxia and observed decreased zebrin-II expression in the cerebella of patients with SCA7. Our results suggest that a breakdown of zebrin subtype regulation is a shared pathological feature of polyglutamine ataxias.
Collapse
Affiliation(s)
- Luke C Bartelt
- University Program in Genetics & Genomics, Duke University Medical Center, Durham, NC 27710, USA
- Departments of Pathology & Laboratory Medicine, Neurology, Biological Chemistry, and Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92697, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Pawel M Switonski
- Department of Neuronal Cell Biology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland
| | - Grażyna Adamek
- Department of Neuronal Cell Biology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland
| | - Fabiana Longo
- Departments of Pathology & Laboratory Medicine, Neurology, Biological Chemistry, and Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Juliana Carvalho
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lisa A Duvick
- Institute for Translational Neuroscience, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sabrina I Jarrah
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Daniel R Scoles
- Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Harry T Orr
- Institute for Translational Neuroscience, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Court Hull
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Craig B Lowe
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Albert R La Spada
- Departments of Pathology & Laboratory Medicine, Neurology, Biological Chemistry, and Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92697, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
- UCI Center for Neurotherapeutics, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
5
|
Prange SE, Bhakta IN, Sysoeva D, Jean GE, Madisetti A, Le HHN, Duong LU, Hwu PT, Melton JG, Thompson-Peer KL. Dendrite injury triggers neuroprotection in Drosophila models of neurodegenerative disease. Sci Rep 2024; 14:24766. [PMID: 39433621 PMCID: PMC11494097 DOI: 10.1038/s41598-024-74670-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/26/2024] [Indexed: 10/23/2024] Open
Abstract
Dendrite defects and loss are early cellular alterations observed across neurodegenerative diseases that play a role in early disease pathogenesis. Dendrite degeneration can be modeled by expressing pathogenic polyglutamine disease transgenes in Drosophila neurons in vivo. Here, we show that we can protect against dendrite loss in neurons modeling neurodegenerative polyglutamine diseases through injury to a single primary dendrite branch. We find that this neuroprotection is specific to injury-induced activation of dendrite regeneration: neither injury to the axon nor injury just to surrounding tissues induces this response. We show that the mechanism of this regenerative response is stabilization of the actin (but not microtubule) cytoskeleton. We also demonstrate that this regenerative response may extend to other neurodegenerative diseases. Together, we provide evidence that activating dendrite regeneration pathways has the potential to slow-or even reverse-dendrite loss in neurodegenerative disease.
Collapse
Affiliation(s)
- Sydney E Prange
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, Irvine, CA, USA
| | - Isha N Bhakta
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Daria Sysoeva
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Grace E Jean
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Anjali Madisetti
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Hieu H N Le
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Ly U Duong
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Patrick T Hwu
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Jaela G Melton
- Center for the Neurobiology of Learning and Memory, Irvine, CA, USA
| | - Katherine L Thompson-Peer
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA.
- Center for the Neurobiology of Learning and Memory, Irvine, CA, USA.
- Sue and Bill Gross Stem Cell Research Center, Irvine, CA, USA.
- Reeve-Irvine Research Center, Irvine, CA, USA.
| |
Collapse
|
6
|
Cepeda C, Holley SM, Barry J, Oikonomou KD, Yazon VW, Peng A, Argueta D, Levine MS. Corticostriatal Maldevelopment in the R6/2 Mouse Model of Juvenile Huntington's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618500. [PMID: 39464124 PMCID: PMC11507867 DOI: 10.1101/2024.10.15.618500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
There is a growing consensus that brain development in Huntington's disease (HD) is abnormal, leading to the idea that HD is not only a neurodegenerative but also a neurodevelopmental disorder. Indeed, structural and functional abnormalities have been observed during brain development in both humans and animal models of HD. However, a concurrent study of cortical and striatal development in a genetic model of HD is still lacking. Here we report significant alterations of corticostriatal development in the R6/2 mouse model of juvenile HD. We examined wildtype (WT) and R6/2 mice at postnatal (P) days 7, 14, and 21. Morphological examination demonstrated early structural and cellular alterations reminiscent of malformations of cortical development, and ex vivo electrophysiological recordings of cortical pyramidal neurons (CPNs) demonstrated significant age- and genotype-dependent changes of intrinsic membrane and synaptic properties. In general, R6/2 CPNs had reduced cell membrane capacitance and increased input resistance (P7 and P14), along with reduced frequency of spontaneous excitatory and inhibitory synaptic events during early development (P7), suggesting delayed cortical maturation. This was confirmed by increased occurrence of GABA A receptor-mediated giant depolarizing potentials at P7. At P14, the rheobase of CPNs was significantly reduced, along with increased excitability. Altered membrane and synaptic properties of R6/2 CPNs recovered progressively, and by P21 they were similar to WT CPNs. In striatal medium-sized spiny neurons (MSNs), a different picture emerged. Intrinsic membrane properties were relatively normal throughout development, except for a transient increase in membrane capacitance at P14. The first alterations in MSNs synaptic activity were observed at P14 and consisted of significant deficits in GABAergic inputs, however, these also were normalized by P21. In contrast, excitatory inputs began to decrease at this age. We conclude that the developing HD brain is capable of compensating for early developmental abnormalities and that cortical alterations precede and are a main contributor of striatal changes. Addressing cortical maldevelopment could help prevent or delay disease manifestations.
Collapse
|
7
|
Louçã M, El Akrouti D, Lemesle A, Louessard M, Dufour N, Baroin C, de la Fouchardière A, Cotter L, Jean-Jacques H, Redeker V, Perrier AL. Huntingtin lowering impairs the maturation and synchronized synaptic activity of human cortical neuronal networks derived from induced pluripotent stem cells. Neurobiol Dis 2024; 200:106630. [PMID: 39106928 DOI: 10.1016/j.nbd.2024.106630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024] Open
Abstract
Despite growing descriptions of wild-type Huntingtin (wt-HTT) roles in both adult brain function and, more recently, development, several clinical trials are exploring HTT-lowering approaches that target both wt-HTT and the mutant isoform (mut-HTT) responsible for Huntington's disease (HD). This non-selective targeting is based on the autosomal dominant inheritance of HD, supporting the idea that mut-HTT exerts its harmful effects through a toxic gain-of-function or a dominant-negative mechanism. However, the precise amount of wt-HTT needed for healthy neurons in adults and during development remains unclear. In this study, we address this question by examining how wt-HTT loss affects human neuronal network formation, synaptic maturation, and homeostasis in vitro. Our findings establish a role of wt-HTT in the maturation of dendritic arborization and the acquisition of network-wide synchronized activity by human cortical neuronal networks modeled in vitro. Interestingly, the network synchronization defects only became apparent when more than two-thirds of the wt-HTT protein was depleted. Our study underscores the critical need to precisely understand wt-HTT role in neuronal health. It also emphasizes the potential risks of excessive wt-HTT loss associated with non-selective therapeutic approaches targeting both wt- and mut-HTT isoforms in HD patients.
Collapse
Affiliation(s)
- Mathilde Louçã
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Donya El Akrouti
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Aude Lemesle
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Morgane Louessard
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Noëlle Dufour
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Chloé Baroin
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Aurore de la Fouchardière
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Laurent Cotter
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Hélène Jean-Jacques
- Université Paris-Saclay, CNRS, CEA, Institute for Integrative Biology of the Cell, 91198 Gif-sur-Yvette, France
| | - Virginie Redeker
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CNRS, CEA, Institute for Integrative Biology of the Cell, 91198 Gif-sur-Yvette, France
| | - Anselme L Perrier
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives : Mécanismes, Thérapies, Imagerie, 92265 Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France.
| |
Collapse
|
8
|
Park J, Wu Y, Suk Kim J, Byun J, Lee J, Oh YK. Cytoskeleton-modulating nanomaterials and their therapeutic potentials. Adv Drug Deliv Rev 2024; 211:115362. [PMID: 38906478 DOI: 10.1016/j.addr.2024.115362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/25/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
The cytoskeleton, an intricate network of protein fibers within cells, plays a pivotal role in maintaining cell shape, enabling movement, and facilitating intracellular transport. Its involvement in various pathological states, ranging from cancer proliferation and metastasis to the progression of neurodegenerative disorders, underscores its potential as a target for therapeutic intervention. The exploration of nanotechnology in this realm, particularly the use of nanomaterials for cytoskeletal modulation, represents a cutting-edge approach with the promise of novel treatments. Inorganic nanomaterials, including those derived from gold, metal oxides, carbon, and black phosphorus, alongside organic variants such as peptides and proteins, are at the forefront of this research. These materials offer diverse mechanisms of action, either by directly interacting with cytoskeletal components or by influencing cellular signaling pathways that, in turn, modulate the cytoskeleton. Recent advancements have introduced magnetic field-responsive and light-responsive nanomaterials, which allow for targeted and controlled manipulation of the cytoskeleton. Such precision is crucial in minimizing off-target effects and enhancing therapeutic efficacy. This review explores the importance of research into cytoskeleton-targeting nanomaterials for developing therapeutic interventions for a range of diseases. It also addresses the progress made in this field, the challenges encountered, and future directions for using nanomaterials to modulate the cytoskeleton. The continued exploration of nanomaterials for cytoskeleton modulation holds great promise for advancing therapeutic strategies against a broad spectrum of diseases, marking a significant step forward in the intersection of nanotechnology and medicine.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jung Suk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junho Byun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
9
|
Karliner J, Liu Y, Merry DE. Mutant androgen receptor induces neurite loss and senescence independently of ARE binding in a neuronal model of SBMA. Proc Natl Acad Sci U S A 2024; 121:e2321408121. [PMID: 38976730 PMCID: PMC11260106 DOI: 10.1073/pnas.2321408121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 06/11/2024] [Indexed: 07/10/2024] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a slowly progressing neuromuscular disease caused by a polyglutamine (polyQ)-encoding CAG trinucleotide repeat expansion in the androgen receptor (AR) gene, leading to AR aggregation, lower motor neuron death, and muscle atrophy. AR is a ligand-activated transcription factor that regulates neuronal architecture and promotes axon regeneration; however, whether AR transcriptional functions contribute to disease pathogenesis is not fully understood. Using a differentiated PC12 cell model of SBMA, we identified dysfunction of polyQ-expanded AR in its regulation of neurite growth and maintenance. Specifically, we found that in the presence of androgens, polyQ-expanded AR inhibited neurite outgrowth, induced neurite retraction, and inhibited neurite regrowth. This dysfunction was independent of polyQ-expanded AR transcriptional activity at androgen response elements (ARE). We further showed that the formation of polyQ-expanded AR intranuclear inclusions promoted neurite retraction, which coincided with reduced expression of the neuronal differentiation marker β-III-Tubulin. Finally, we revealed that cell death is not the primary outcome for cells undergoing neurite retraction; rather, these cells become senescent. Our findings reveal that mechanisms independent of AR canonical transcriptional activity underly neurite defects in a cell model of SBMA and identify senescence as a pathway implicated in this pathology. These findings suggest that in the absence of a role for AR canonical transcriptional activity in the SBMA pathologies described here, the development of SBMA therapeutics that preserve this activity may be desirable. This approach may be broadly applicable to other polyglutamine diseases such as Huntington's disease and spinocerebellar ataxias.
Collapse
Affiliation(s)
- Jordyn Karliner
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107
| | - Yuhong Liu
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107
| | - Diane E. Merry
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107
| |
Collapse
|
10
|
Catlin JP, Tooley CES. Exploring potential developmental origins of common neurodegenerative disorders. Biochem Soc Trans 2024; 52:1035-1044. [PMID: 38661189 PMCID: PMC11440815 DOI: 10.1042/bst20230422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/12/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024]
Abstract
In the United States, it is now estimated that 6.7 million people over the age of 65 are afflicted by Alzheimer's disease (AD), over 1 million people are living with Parkinson's disease (PD), and over 200 000 have or are at risk for developing Huntington's disease (HD). All three of these neurodegenerative diseases result in the ultimate death of distinct neuronal subtypes, and it is widely thought that age-related damage is the single biggest contributing factor to this neuronal death. However, recent studies are now suggesting that developmental defects during early neurogenesis could also play a role in the pathology of neurodegenerative diseases. Loss or overexpression of proteins associated with HD, PD, and AD also result in embryonic phenotypes but whether these developmental defects slowly unmask over time and contribute to age-related neurodegeneration remains highly debated. Here, we discuss known links between embryonic neurogenesis and neurodegenerative disorders (including common signaling pathways), potential compensatory mechanisms that could delay presentation of neurodegenerative disorders, and the types of model systems that could be used to study these links in vivo.
Collapse
Affiliation(s)
- James P. Catlin
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Christine E. Schaner Tooley
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
11
|
Hu H, Wang C, Tao R, Liu B, Peng D, Chen Y, Zhang W. Evidences of neurological injury caused by COVID-19 from glioma tissues and glioma organoids. CNS Neurosci Ther 2024; 30:e14822. [PMID: 38923860 PMCID: PMC11199819 DOI: 10.1111/cns.14822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION Despite the extensive neurological symptoms induced by COVID-19 and the identification of SARS-CoV-2 in post-mortem brain samples from COVID-19 patients months after death, the precise mechanisms of SARS-CoV-2 invasion into the central nervous system remain unclear due to the lack of research models. METHODS We collected glioma tissue samples from glioma patients who had a recent history of COVID-19 and examined the presence of the SARS-CoV-2 spike protein. Subsequently, spatial transcriptomic analyses were conducted on normal brain tissues, glioma tissues, and glioma tissues from glioma patients with recent COVID-19 history. Additionally, single-cell sequencing data from both glioma tissues and glioma organoids were collected and analyzed. Glioma organoids were utilized to evaluate the efficacy of potential COVID-19 blocking agents. RESULTS Glioma tissues from glioma patients with recent COVID-19 history exhibited the presence of the SARS-CoV-2 spike protein. Differences between glioma tissues from glioma patients who had a recent history of COVID-19 and healthy brain tissues primarily manifested in neuronal cells. Notably, neuronal cells within glioma tissues of COVID-19 history demonstrated heightened susceptibility to Alzheimer's disease, depression, and synaptic dysfunction, indicative of neuronal aberrations. Expressions of SARS-CoV-2 entry factors were confirmed in both glioma tissues and glioma organoids. Moreover, glioma organoids were susceptible to pseudo-SARS-CoV-2 infection and the infections could be partly blocked by the potential COVID-19 drugs. CONCLUSIONS Gliomas had inherent traits that render them susceptible to SARS-CoV-2 infection, leading to their representability of COVID-19 neurological symptoms. This established a biological foundation for the rationality and feasibility of utilization of glioma organoids as research and blocking drug testing model in SARS-CoV-2 infection within the central nervous system.
Collapse
Affiliation(s)
- Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Center of Brain Tumor, Beijing Institute for Brain DisordersBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Chinese Glioma Genome Atlas Network (CGGA)BeijingChina
| | - Chen Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Center of Brain Tumor, Beijing Institute for Brain DisordersBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Chinese Glioma Genome Atlas Network (CGGA)BeijingChina
| | - Rui Tao
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Center of Brain Tumor, Beijing Institute for Brain DisordersBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Chinese Glioma Genome Atlas Network (CGGA)BeijingChina
| | - Bohan Liu
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Center of Brain Tumor, Beijing Institute for Brain DisordersBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Chinese Glioma Genome Atlas Network (CGGA)BeijingChina
| | - Dazhao Peng
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Center of Brain Tumor, Beijing Institute for Brain DisordersBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Chinese Glioma Genome Atlas Network (CGGA)BeijingChina
| | - Yankun Chen
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Center of Brain Tumor, Beijing Institute for Brain DisordersBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Chinese Glioma Genome Atlas Network (CGGA)BeijingChina
| | - Wei Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Center of Brain Tumor, Beijing Institute for Brain DisordersBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Chinese Glioma Genome Atlas Network (CGGA)BeijingChina
| |
Collapse
|
12
|
Zaninello M, Schlegel T, Nolte H, Pirzada M, Savino E, Barth E, Klein I, Wüstenberg H, Uddin T, Wolff L, Wirth B, Lehmann HC, Cioni JM, Langer T, Rugarli EI. CLUH maintains functional mitochondria and translation in motoneuronal axons and prevents peripheral neuropathy. SCIENCE ADVANCES 2024; 10:eadn2050. [PMID: 38809982 PMCID: PMC11135423 DOI: 10.1126/sciadv.adn2050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/24/2024] [Indexed: 05/31/2024]
Abstract
Transporting and translating mRNAs in axons is crucial for neuronal viability. Local synthesis of nuclear-encoded mitochondrial proteins protects long-lived axonal mitochondria from damage; however, the regulatory factors involved are largely unknown. We show that CLUH, which binds mRNAs encoding mitochondrial proteins, prevents peripheral neuropathy and motor deficits in the mouse. CLUH is enriched in the growth cone of developing spinal motoneurons and is required for their growth. The lack of CLUH affects the abundance of target mRNAs and the corresponding mitochondrial proteins more prominently in axons, leading to ATP deficits in the growth cone. CLUH interacts with ribosomal subunits, translation initiation, and ribosome recycling components and preserves axonal translation. Overexpression of the ribosome recycling factor ABCE1 rescues the mRNA and translation defects, as well as the growth cone size, in CLUH-deficient motoneurons. Thus, we demonstrate a role for CLUH in mitochondrial quality control and translational regulation in axons, which is essential for their development and long-term integrity and function.
Collapse
Affiliation(s)
- Marta Zaninello
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Tim Schlegel
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Hendrik Nolte
- Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Mujeeb Pirzada
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Elisa Savino
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Esther Barth
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Ines Klein
- Department of Neurology, University of Cologne, Cologne 50931, Germany
| | - Hauke Wüstenberg
- Department of Neurology, University of Cologne, Cologne 50931, Germany
| | - Tesmin Uddin
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Lisa Wolff
- Institute of Human Genetics, University of Cologne, Cologne 50931, Germany
| | - Brunhilde Wirth
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Institute of Human Genetics, University of Cologne, Cologne 50931, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
- Center for Rare Diseases Cologne (CESEK), University Hospital of Cologne, Cologne 50937, Germany
| | - Helmar C. Lehmann
- Department of Neurology, University of Cologne, Cologne 50931, Germany
| | - Jean-Michel Cioni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Thomas Langer
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Elena I. Rugarli
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| |
Collapse
|
13
|
Ratié L, Humbert S. A developmental component to Huntington's disease. Rev Neurol (Paris) 2024; 180:357-362. [PMID: 38614929 DOI: 10.1016/j.neurol.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 04/15/2024]
Abstract
Huntington's disease is a dominantly inherited disorder characterized by the dysfunction and death of cortical and striatal neurons. Striatal degeneration in Huntington's disease is due, at least in part, to defective cortical signalling to the striatum. Although Huntington's disease generally manifests at the adult stage, mouse and neuroimaging studies of presymptomatic mutation carriers suggest that it may affect neurodevelopment. In support of this notion, the development of the cortex is altered in mice with Huntington's disease and the foetuses of human Huntington's disease gene carriers. We will discuss these studies and the contribution of abnormal brain development to the later appearance of the disease.
Collapse
Affiliation(s)
- L Ratié
- U1216, CEA, Grenoble Institute Neurosciences, Inserm, université Grenoble Alpes, 38000 Grenoble, France
| | - S Humbert
- Institut du Cerveau-Paris Brain Institute, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France.
| |
Collapse
|
14
|
Dubey SK, Lloyd TE, Tapadia MG. Disrupted nuclear import of cell cycle proteins in Huntington's/PolyQ disease causes neurodevelopment defects in cellular and Drosophila model. Heliyon 2024; 10:e26393. [PMID: 38434042 PMCID: PMC10906312 DOI: 10.1016/j.heliyon.2024.e26393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/15/2024] [Accepted: 02/12/2024] [Indexed: 03/05/2024] Open
Abstract
Huntington's disease is caused by an expansion of CAG repeats in exon 1 of the huntingtin gene encoding an extended PolyQ tract within the Huntingtin protein (mHtt). This expansion results in selective degeneration of striatal medium spiny projection neurons in the basal ganglia. The mutation causes abnormalities during neurodevelopment in human and mouse models. Here, we report that mHtt/PolyQ aggregates inhibit the cell cycle in the Drosophila brain during development. PolyQ aggregates disrupt the nuclear pore complexes of the cells preventing the translocation of cell cycle proteins such as Cyclin E, E2F and PCNA from cytoplasm to the nucleus, thus affecting cell cycle progression. PolyQ aggregates also disrupt the nuclear pore complex and nuclear import in mHtt expressing mammalian CAD neurons. PolyQ toxicity and cell cycle defects can be restored by enhancing RanGAP-mediated nuclear import, suggesting a potential therapeutic approach for this disease.
Collapse
Affiliation(s)
- Sandeep Kumar Dubey
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Thomas E. Lloyd
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Madhu G. Tapadia
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
15
|
Khoshnan A. Gut Microbiota as a Modifier of Huntington's Disease Pathogenesis. J Huntingtons Dis 2024; 13:133-147. [PMID: 38728199 PMCID: PMC11307070 DOI: 10.3233/jhd-240012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2024] [Indexed: 05/12/2024]
Abstract
Huntingtin (HTT) protein is expressed in most cell lineages, and the toxicity of mutant HTT in multiple organs may contribute to the neurological and psychiatric symptoms observed in Huntington's disease (HD). The proteostasis and neurotoxicity of mutant HTT are influenced by the intracellular milieu and responses to environmental signals. Recent research has highlighted a prominent role of gut microbiota in brain and immune system development, aging, and the progression of neurological disorders. Several studies suggest that mutant HTT might disrupt the homeostasis of gut microbiota (known as dysbiosis) and impact the pathogenesis of HD. Dysbiosis has been observed in HD patients, and in animal models of the disease it coincides with mutant HTT aggregation, abnormal behaviors, and reduced lifespan. This review article aims to highlight the potential toxicity of mutant HTT in organs and pathways within the microbiota-gut-immune-central nervous system (CNS) axis. Understanding the functions of Wild-Type (WT) HTT and the toxicity of mutant HTT in these organs and the associated networks may elucidate novel pathogenic pathways, identify biomarkers and peripheral therapeutic targets for HD.
Collapse
Affiliation(s)
- Ali Khoshnan
- Keck School of Medicine, Physiology and Neuroscience, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
16
|
Louessard M, Cailleret M, Jarrige M, Bigarreau J, Lenoir S, Dufour N, Rey M, Saudou F, Deglon N, Perrier AL. Mono- and Biallelic Inactivation of Huntingtin Gene in Patient-Specific Induced Pluripotent Stem Cells Reveal HTT Roles in Striatal Development and Neuronal Functions. J Huntingtons Dis 2024; 13:41-53. [PMID: 38427495 DOI: 10.3233/jhd-231509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Background Mutations in the Huntingtin (HTT) gene cause Huntington's disease (HD), a neurodegenerative disorder. As a scaffold protein, HTT is involved in numerous cellular functions, but its normal and pathogenic functions during human forebrain development are poorly understood. Objective To investigate the developmental component of HD, with a specific emphasis on understanding the functions of wild-type and mutant HTT alleles during forebrain neuron development in individuals carrying HD mutations. Methods We used CRISPR/Cas9 gene-editing technology to disrupt the ATG region of the HTT gene via non-homologous end joining to produce mono- or biallelic HTT knock-out human induced pluripotent stem cell (iPSC) clones. Results We showed that the loss of wild-type, mutant, or both HTT isoforms does not affect the pluripotency of iPSCs or their transition into neural cells. However, we observed that HTT loss causes division impairments in forebrain neuro-epithelial cells and alters maturation of striatal projection neurons (SPNs) particularly in the acquisition of DARPP32 expression, a key functional marker of SPNs. Finally, young post-mitotic neurons derived from HTT-/- human iPSCs display cellular dysfunctions observed in adult HD neurons. Conclusions We described a novel collection of isogenic clones with mono- and biallelic HTT inactivation that complement existing HD-hiPSC isogenic series to explore HTT functions and test therapeutic strategies in particular HTT-lowering drugs. Characterizing neural and neuronal derivatives from human iPSCs of this collection, we show evidence that HTT loss or mutation has impacts on neuro-epithelial and striatal neurons maturation, and on basal DNA damage and BDNF axonal transport in post-mitotic neurons.
Collapse
Affiliation(s)
- Morgane Louessard
- Université Paris-Saclay, CEA, Molecular Imaging Research Center, Fontenay-aux-Roses, France
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives: Mécanismes, Thérapies, Imagerie, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, Univ Evry, Institut des Cellules Souches pour le Traitement et l'étude des Maladies Monogéniques, Corbeil-Essonne, France
| | - Michel Cailleret
- Université Paris-Saclay, Inserm, Univ Evry, Institut des Cellules Souches pour le Traitement et l'étude des Maladies Monogéniques, Corbeil-Essonne, France
| | - Margot Jarrige
- CECS/AFM, Institut des Cellules Souches pour le Traitement et l'étude des Maladies Monogéniques, Corbeil-Essonne, France
| | - Julie Bigarreau
- Université Paris-Saclay, Inserm, Univ Evry, Institut des Cellules Souches pour le Traitement et l'étude des Maladies Monogéniques, Corbeil-Essonne, France
| | - Sophie Lenoir
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Noëlle Dufour
- Université Paris-Saclay, CEA, Molecular Imaging Research Center, Fontenay-aux-Roses, France
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives: Mécanismes, Thérapies, Imagerie, Fontenay-aux-Roses, France
| | - Maria Rey
- Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Department of Clinical Neurosciences (DNC), and Neuroscience Research Center (CRN), Laboratory of Cellular and Molecular Neurotherapies, Lausanne, Switzerland
| | - Frédéric Saudou
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Nicole Deglon
- Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Department of Clinical Neurosciences (DNC), and Neuroscience Research Center (CRN), Laboratory of Cellular and Molecular Neurotherapies, Lausanne, Switzerland
| | - Anselme L Perrier
- Université Paris-Saclay, CEA, Molecular Imaging Research Center, Fontenay-aux-Roses, France
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives: Mécanismes, Thérapies, Imagerie, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, Univ Evry, Institut des Cellules Souches pour le Traitement et l'étude des Maladies Monogéniques, Corbeil-Essonne, France
| |
Collapse
|
17
|
Estevez-Fraga C, Altmann A, Parker CS, Scahill RI, Costa B, Chen Z, Manzoni C, Zarkali A, Durr A, Roos RAC, Landwehrmeyer B, Leavitt BR, Rees G, Tabrizi SJ, McColgan P. Genetic topography and cortical cell loss in Huntington's disease link development and neurodegeneration. Brain 2023; 146:4532-4546. [PMID: 37587097 PMCID: PMC10629790 DOI: 10.1093/brain/awad275] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/12/2023] [Accepted: 07/28/2023] [Indexed: 08/18/2023] Open
Abstract
Cortical cell loss is a core feature of Huntington's disease (HD), beginning many years before clinical motor diagnosis, during the premanifest stage. However, it is unclear how genetic topography relates to cortical cell loss. Here, we explore the biological processes and cell types underlying this relationship and validate these using cell-specific post-mortem data. Eighty premanifest participants on average 15 years from disease onset and 71 controls were included. Using volumetric and diffusion MRI we extracted HD-specific whole brain maps where lower grey matter volume and higher grey matter mean diffusivity, relative to controls, were used as proxies of cortical cell loss. These maps were combined with gene expression data from the Allen Human Brain Atlas (AHBA) to investigate the biological processes relating genetic topography and cortical cell loss. Cortical cell loss was positively correlated with the expression of developmental genes (i.e. higher expression correlated with greater atrophy and increased diffusivity) and negatively correlated with the expression of synaptic and metabolic genes that have been implicated in neurodegeneration. These findings were consistent for diffusion MRI and volumetric HD-specific brain maps. As wild-type huntingtin is known to play a role in neurodevelopment, we explored the association between wild-type huntingtin (HTT) expression and developmental gene expression across the AHBA. Co-expression network analyses in 134 human brains free of neurodegenerative disorders were also performed. HTT expression was correlated with the expression of genes involved in neurodevelopment while co-expression network analyses also revealed that HTT expression was associated with developmental biological processes. Expression weighted cell-type enrichment (EWCE) analyses were used to explore which specific cell types were associated with HD cortical cell loss and these associations were validated using cell specific single nucleus RNAseq (snRNAseq) data from post-mortem HD brains. The developmental transcriptomic profile of cortical cell loss in preHD was enriched in astrocytes and endothelial cells, while the neurodegenerative transcriptomic profile was enriched for neuronal and microglial cells. Astrocyte-specific genes differentially expressed in HD post-mortem brains relative to controls using snRNAseq were enriched in the developmental transcriptomic profile, while neuronal and microglial-specific genes were enriched in the neurodegenerative transcriptomic profile. Our findings suggest that cortical cell loss in preHD may arise from dual pathological processes, emerging as a consequence of neurodevelopmental changes, at the beginning of life, followed by neurodegeneration in adulthood, targeting areas with reduced expression of synaptic and metabolic genes. These events result in age-related cell death across multiple brain cell types.
Collapse
Affiliation(s)
- Carlos Estevez-Fraga
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
| | - Andre Altmann
- Centre for Medical Image Computing, University College London, London WC1V 6LJ, UK
| | - Christopher S Parker
- Centre for Medical Image Computing, University College London, London WC1V 6LJ, UK
| | - Rachael I Scahill
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
| | - Beatrice Costa
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Zhongbo Chen
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
| | - Claudia Manzoni
- School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Angeliki Zarkali
- Dementia Research Centre, University College London, London WC1N 3AR, UK
| | - Alexandra Durr
- Sorbonne Université, Paris Brain Institute (ICM), AP-HP, Inserm, CNRS, Paris 75013, France
| | - Raymund A C Roos
- Department of Neurology, Leiden University Medical Centre, Leiden 2333, The Netherlands
| | | | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver BC V5Z 4H4Canada
- Division of Neurology, Department of Medicine, University of British Columbia Hospital, Vancouver BC V6T 2B5, Canada
| | - Geraint Rees
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
| | - Peter McColgan
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
| |
Collapse
|
18
|
Deleuze V, Garcia L, Rouaisnel B, Salma M, Kinoo A, Andrieu-Soler C, Soler E. Efficient genome editing in erythroid cells unveils novel MYB target genes and regulatory functions. iScience 2023; 26:107641. [PMID: 37670779 PMCID: PMC10475484 DOI: 10.1016/j.isci.2023.107641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/09/2023] [Accepted: 08/11/2023] [Indexed: 09/07/2023] Open
Abstract
Targeted genome editing holds great promise in biology. However, efficient genome modification, including gene knock-in (KI), remains an unattained goal in multiple cell types and loci due to poor transfection efficiencies and low target genes expression, impeding the positive selection of recombined cells. Here, we describe a genome editing approach to achieve efficient gene targeting using hard to transfect erythroid cell lines. We demonstrate robust fluorescent protein KI efficiency in low expressed transcription factor (TF) genes (e.g., Myb or Zeb1). We further show the ability to target two independent loci in individual cells, exemplified by MYB-GFP and NuMA-Cherry double KI, allowing multicolor labeling of regulatory factors at physiological endogenous levels. Our KI tagging approach allowed us to perform genome-wide TF analysis at increased signal-to-noise ratios, and highlighted previously unidentified MYB target genes and pathways. Overall, we establish a versatile CRISPR-Cas9-based platform, offering attractive opportunities for the dissection of the erythroid differentiation process.
Collapse
Affiliation(s)
| | - Leonor Garcia
- IGMM, University Montpellier, CNRS, Montpellier, France
| | | | - Mohammad Salma
- IGMM, University Montpellier, CNRS, Montpellier, France
- Laboratory of Excellence GR-Ex, Université de Paris, Paris, France
| | - Alexia Kinoo
- IGMM, University Montpellier, CNRS, Montpellier, France
| | - Charlotte Andrieu-Soler
- IGMM, University Montpellier, CNRS, Montpellier, France
- Laboratory of Excellence GR-Ex, Université de Paris, Paris, France
| | - Eric Soler
- IGMM, University Montpellier, CNRS, Montpellier, France
- Laboratory of Excellence GR-Ex, Université de Paris, Paris, France
| |
Collapse
|
19
|
Burger T. Controlling for false discoveries subsequently to large scale one-way ANOVA testing in proteomics: Practical considerations. Proteomics 2023; 23:e2200406. [PMID: 37357151 DOI: 10.1002/pmic.202200406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/27/2023]
Abstract
In discovery proteomics, as well as many other "omic" approaches, the possibility to test for the differential abundance of hundreds (or of thousands) of features simultaneously is appealing, despite requiring specific statistical safeguards, among which controlling for the false discovery rate (FDR) has become standard. Moreover, when more than two biological conditions or group treatments are considered, it has become customary to rely on the one-way analysis of variance (ANOVA) framework, where a first global differential abundance landscape provided by an omnibus test can be subsequently refined using various post-hoc tests (PHTs). However, the interactions between the FDR control procedures and the PHTs are complex, because both correspond to different types of multiple test corrections (MTCs). This article surveys various ways to orchestrate them in a data processing workflow and discusses their pros and cons.
Collapse
Affiliation(s)
- Thomas Burger
- Univ. Grenoble Alpes, CNRS, CEA, INSERM, ProFI, EDyP, Grenoble, France
| |
Collapse
|
20
|
Yang S, Ma J, Zhang H, Chen L, Li Y, Pan M, Zhu H, Liang J, He D, Li S, Li XJ, Guo X. Mutant HTT does not affect glial development but impairs myelination in the early disease stage. Front Neurosci 2023; 17:1238306. [PMID: 37539389 PMCID: PMC10394243 DOI: 10.3389/fnins.2023.1238306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 07/03/2023] [Indexed: 08/05/2023] Open
Abstract
Introduction Huntington's disease (HD) is caused by expanded CAG repeats in the huntingtin gene (HTT) and is characterized by late-onset neurodegeneration that primarily affects the striatum. Several studies have shown that mutant HTT can also affect neuronal development, contributing to the late-onset neurodegeneration. However, it is currently unclear whether mutant HTT impairs the development of glial cells, which is important for understanding whether mutant HTT affects glial cells during early brain development. Methods Using HD knock-in mice that express full-length mutant HTT with a 140 glutamine repeat at the endogenous level, we analyzed the numbers of astrocytes and oligodendrocytes from postnatal day 1 to 3 months of age via Western blotting and immunocytochemistry. We also performed electron microscopy, RNAseq analysis, and quantitative RT-PCR. Results The numbers of astrocytes and oligodendrocytes were not significantly altered in postnatal HD KI mice compared to wild type (WT) mice. Consistently, glial protein expression levels were not significantly different between HD KI and WT mice. However, at 3 months of age, myelin protein expression was reduced in HD KI mice, as evidenced by Western blotting and immunocytochemical results. Electron microscopy revealed a slight but significant reduction in myelin thickness of axons in the HD KI mouse brain at 3 months of age. RNAseq analysis did not show significant reductions in myelin-related genes in postnatal HD KI mice. Conclusion These data suggest that cytoplasmic mutant HTT, rather than nuclear mutant HTT, mediates myelination defects in the early stages of the disease without impacting the differentiation and maturation of glial cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xiangyu Guo
- *Correspondence: Xiao-Jiang Li, ; Xiangyu Guo
| |
Collapse
|
21
|
Bartelt LC, Switonski PM, Adamek G, Carvalho J, Duvick LA, Jarrah SI, McLoughlin HS, Scoles DR, Pulst SM, Orr HT, Hull C, Lowe CB, La Spada AR. Purkinje-Enriched snRNA-seq in SCA7 Cerebellum Reveals Zebrin Identity Loss as a Central Feature of Polyglutamine Ataxias. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.19.533345. [PMID: 37214832 PMCID: PMC10197555 DOI: 10.1101/2023.03.19.533345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Spinocerebellar ataxia type 7 (SCA7) is an inherited neurodegenerative disorder caused by a CAG-polyglutamine repeat expansion. SCA7 patients display a striking loss of Purkinje cell (PC) neurons with disease progression; however, PCs are rare, making them difficult to characterize. We developed a PC nuclei enrichment protocol and applied it to single-nucleus RNA-seq of a SCA7 knock-in mouse model. Our results unify prior observations into a central mechanism of cell identity loss, impacting both glia and PCs, driving accumulation of inhibitory synapses and altered PC spiking. Zebrin-II subtype dysregulation is the predominant signal in PCs, leading to complete loss of zebrin-II striping at motor symptom onset in SCA7 mice. We show this zebrin-II subtype degradation is shared across Polyglutamine Ataxia mouse models and SCA7 patients. It has been speculated that PC subtype organization is critical for cerebellar function, and our results suggest that a breakdown of zebrin-II parasagittal striping is pathological.
Collapse
Affiliation(s)
- Luke C. Bartelt
- University Program in Genetics & Genomics, Duke University Medical Center, Durham, NC 27710, USA
- Departments of Pathology & Laboratory Medicine, Neurology, Biological Chemistry, and Neurobiology & Behavior, University of California, Irvine; Irvine, CA 92697, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Pawel M. Switonski
- Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland
| | - Grażyna Adamek
- Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland
| | - Juliana Carvalho
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lisa A. Duvick
- Institute for Translational Neuroscience, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sabrina I. Jarrah
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Daniel R. Scoles
- Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Stefan M. Pulst
- Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Harry T. Orr
- Institute for Translational Neuroscience, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Court Hull
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Craig B. Lowe
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Albert R. La Spada
- Departments of Pathology & Laboratory Medicine, Neurology, Biological Chemistry, and Neurobiology & Behavior, University of California, Irvine; Irvine, CA 92697, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
- UCI Center for Neurotherapeutics, University of California, Irvine; Irvine, CA 92697, USA
| |
Collapse
|
22
|
Wennagel D, Braz BY, Humbert S. [Treating early transient neuronal defects in a mouse model of Huntington's disease delays the signs of the disease in adulthood]. Med Sci (Paris) 2023; 39:313-316. [PMID: 37094259 DOI: 10.1051/medsci/2023036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Affiliation(s)
- Doris Wennagel
- Univ. Grenoble Alpes, Inserm U1216, Grenoble institut des neurosciences, Grenoble, France
| | - Barbara Yael Braz
- Univ. Grenoble Alpes, Inserm U1216, Grenoble institut des neurosciences, Grenoble, France
| | - Sandrine Humbert
- Univ. Grenoble Alpes, Inserm U1216, Grenoble institut des neurosciences, Grenoble, France - Sorbonne université, institut du cerveau, AP-HP, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
23
|
Li M, Peng L, Wang Z, Liu L, Cao M, Cui J, Wu F, Yang J. Roles of the cytoskeleton in human diseases. Mol Biol Rep 2023; 50:2847-2856. [PMID: 36609753 DOI: 10.1007/s11033-022-08025-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 10/12/2022] [Indexed: 01/08/2023]
Abstract
Recently, researches have revealed the key roles of the cytoskeleton in the occurrence and development of multiple diseases, suggesting that targeting the cytoskeleton is a viable approach for treating numerous refractory diseases. The cytoskeleton is a highly structured and complex network composed of actin filaments, microtubules, and intermediate filaments. In normal cells, these three cytoskeleton components are highly integrated and coordinated. However, the cytoskeleton undergoes drastic remodeling in cytoskeleton-related diseases, causing changes in cell polarity, affecting the cell cycle, leading to senescent diseases, and influencing cell migration to accelerate cancer metastasis. Additionally, mutations or abnormalities in cytoskeletal proteins and their related proteins are closely associated with several congenital diseases. Therefore, this review summarizes the roles of the cytoskeleton in cytoskeleton-related diseases as well as its potential roles in disease treatment to provide insights regarding the physiological functions and pathological roles of the cytoskeleton.
Collapse
Affiliation(s)
- Mengxin Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Li Peng
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, 610065, Chengdu, China
| | - Zhenming Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Lijia Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Mengjiao Cao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Jingyao Cui
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Fanzi Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China.
| |
Collapse
|
24
|
Humbert S, Barnat M. Huntington's disease and brain development. C R Biol 2022; 345:77-90. [PMID: 36847466 DOI: 10.5802/crbiol.93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Huntington's disease is a rare inherited neurological disorder that generally manifests in mild-adulthood. The disease is characterized by the dysfunction and the degeneration of specific brain structures leading progressively to psychiatric, cognitive and motor disorders. The disease is caused by a mutation in the gene coding for huntingtin and, although it appears in adulthood, embryos carry the mutated gene from their development in utero. Studies based on mouse models and human stem cells have reported altered developmental mechanisms in disease conditions. However, does the mutation affect development in humans? Focusing on the early stages of brain development in human fetuses carrying the HD mutation, we have identified abnormalities in the development of the neocortex, the structure that ensure higher cerebral functions. Altogether, these studies suggests that developmental defects could contribute to the onset symptoms in adults, changing the perspective on disease and thus the health care of patients.
Collapse
|
25
|
Braz BY, Wennagel D, Ratié L, de Souza DAR, Deloulme JC, Barbier EL, Buisson A, Lanté F, Humbert S. Treating early postnatal circuit defect delays Huntington's disease onset and pathology in mice. Science 2022; 377:eabq5011. [PMID: 36137051 DOI: 10.1126/science.abq5011] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Recent evidence has shown that even mild mutations in the Huntingtin gene that are associated with late-onset Huntington's disease (HD) disrupt various aspects of human neurodevelopment. To determine whether these seemingly subtle early defects affect adult neural function, we investigated neural circuit physiology in newborn HD mice. During the first postnatal week, HD mice have less cortical layer 2/3 excitatory synaptic activity than wild-type mice, express fewer glutamatergic receptors, and show sensorimotor deficits. The circuit self-normalizes in the second postnatal week but the mice nonetheless develop HD. Pharmacologically enhancing glutamatergic transmission during the neonatal period, however, rescues these deficits and preserves sensorimotor function, cognition, and spine and synapse density as well as brain region volume in HD adult mice.
Collapse
Affiliation(s)
- Barbara Yael Braz
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Doris Wennagel
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Leslie Ratié
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | | | | | - Emmanuel L Barbier
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Alain Buisson
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Fabien Lanté
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Sandrine Humbert
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France.,Institut du Cerveau-Paris Brain Institute, Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
26
|
Blumenstock S, Dudanova I. Balancing neuronal circuits. Science 2022; 377:1383-1384. [PMID: 36137024 DOI: 10.1126/science.ade3116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Correcting synaptic defects in development delays Huntington's disease symptoms in older mice.
Collapse
Affiliation(s)
- Sonja Blumenstock
- Molecular Neurodegeneration Group, Max Planck Institute for Biological Intelligence, i.f., Martinsried, Germany.,Department of Molecules-Signaling-Development, Max Planck Institute for Biological Intelligence, i.f., Martinsried, Germany.,Department of Neurobiology, Center for Neural Circuits and Behavior, Department of Neurosciences, University of California, San Diego, CA, USA
| | - Irina Dudanova
- Molecular Neurodegeneration Group, Max Planck Institute for Biological Intelligence, i.f., Martinsried, Germany.,Department of Molecules-Signaling-Development, Max Planck Institute for Biological Intelligence, i.f., Martinsried, Germany.,Center for Anatomy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
27
|
Eli S, Castagna R, Mapelli M, Parisini E. Recent Approaches to the Identification of Novel Microtubule-Targeting Agents. Front Mol Biosci 2022; 9:841777. [PMID: 35425809 PMCID: PMC9002125 DOI: 10.3389/fmolb.2022.841777] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/21/2022] [Indexed: 12/05/2022] Open
Abstract
Microtubules are key components of the eukaryotic cytoskeleton with essential roles in cell division, intercellular transport, cell morphology, motility, and signal transduction. They are composed of protofilaments of heterodimers of α-tubulin and β-tubulin organized as rigid hollow cylinders that can assemble into large and dynamic intracellular structures. Consistent with their involvement in core cellular processes, affecting microtubule assembly results in cytotoxicity and cell death. For these reasons, microtubules are among the most important targets for the therapeutic treatment of several diseases, including cancer. The vast literature related to microtubule stabilizers and destabilizers has been reviewed extensively in recent years. Here we summarize recent experimental and computational approaches for the identification of novel tubulin modulators and delivery strategies. These include orphan small molecules, PROTACs as well as light-sensitive compounds that can be activated with high spatio-temporal accuracy and that represent promising tools for precision-targeted chemotherapy.
Collapse
Affiliation(s)
- Susanna Eli
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Rossella Castagna
- Latvian Institute of Organic Synthesis, Aizkraukles Iela 21, Riga, Latvia
| | - Marina Mapelli
- IEO, European Institute of Oncology IRCCS, Milan, Italy
- *Correspondence: Marina Mapelli, ; Emilio Parisini,
| | - Emilio Parisini
- Latvian Institute of Organic Synthesis, Aizkraukles Iela 21, Riga, Latvia
- *Correspondence: Marina Mapelli, ; Emilio Parisini,
| |
Collapse
|
28
|
Stouffer MA, Khalaf-Nazzal R, Cifuentes-Diaz C, Albertini G, Bandet E, Grannec G, Lavilla V, Deleuze JF, Olaso R, Nosten-Bertrand M, Francis F. Doublecortin mutation leads to persistent defects in the Golgi apparatus and mitochondria in adult hippocampal pyramidal cells. Neurobiol Dis 2022; 168:105702. [PMID: 35339680 DOI: 10.1016/j.nbd.2022.105702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/08/2022] [Accepted: 03/17/2022] [Indexed: 11/08/2022] Open
Abstract
Human doublecortin (DCX) mutations are associated with severe brain malformations leading to aberrant neuron positioning (heterotopia), intellectual disability and epilepsy. DCX is a microtubule-associated protein which plays a key role during neurodevelopment in neuronal migration and differentiation. Dcx knockout (KO) mice show disorganized hippocampal pyramidal neurons. The CA2/CA3 pyramidal cell layer is present as two abnormal layers and disorganized CA3 KO pyramidal neurons are also more excitable than wild-type (WT) cells. To further identify abnormalities, we characterized Dcx KO hippocampal neurons at subcellular, molecular and ultrastructural levels. Severe defects were observed in mitochondria, affecting number and distribution. Also, the Golgi apparatus was visibly abnormal, increased in volume and abnormally organized. Transcriptome analyses from laser microdissected hippocampal tissue at postnatal day 60 (P60) highlighted organelle abnormalities. Ultrastructural studies of CA3 cells performed in P60 (young adult) and > 9 months (mature) tissue showed that organelle defects are persistent throughout life. Locomotor activity and fear memory of young and mature adults were also abnormal: Dcx KO mice consistently performed less well than WT littermates, with defects becoming more severe with age. Thus, we show that disruption of a neurodevelopmentally-regulated gene can lead to permanent organelle anomalies contributing to abnormal adult behavior.
Collapse
Affiliation(s)
- M A Stouffer
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - R Khalaf-Nazzal
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - C Cifuentes-Diaz
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - G Albertini
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - E Bandet
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - G Grannec
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - V Lavilla
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057 Evry, France
| | - J-F Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057 Evry, France
| | - R Olaso
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057 Evry, France
| | - M Nosten-Bertrand
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - F Francis
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France.
| |
Collapse
|
29
|
Serranilla M, Woodin MA. Striatal Chloride Dysregulation and Impaired GABAergic Signaling Due to Cation-Chloride Cotransporter Dysfunction in Huntington’s Disease. Front Cell Neurosci 2022; 15:817013. [PMID: 35095429 PMCID: PMC8795088 DOI: 10.3389/fncel.2021.817013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Intracellular chloride (Cl–) levels in mature neurons must be tightly regulated for the maintenance of fast synaptic inhibition. In the mature central nervous system (CNS), synaptic inhibition is primarily mediated by gamma-amino butyric acid (GABA), which binds to Cl– permeable GABAA receptors (GABAARs). The intracellular Cl– concentration is primarily maintained by the antagonistic actions of two cation-chloride cotransporters (CCCs): Cl–-importing Na+-K+-Cl– co-transporter-1 (NKCC1) and Cl– -exporting K+-Cl– co-transporter-2 (KCC2). In mature neurons in the healthy brain, KCC2 expression is higher than NKCC1, leading to lower levels of intracellular Cl–, and Cl– influx upon GABAAR activation. However, in neurons of the immature brain or in neurological disorders such as epilepsy and traumatic brain injury, impaired KCC2 function and/or enhanced NKCC1 expression lead to intracellular Cl– accumulation and GABA-mediated excitation. In Huntington’s disease (HD), KCC2- and NKCC1-mediated Cl–-regulation are also altered, which leads to GABA-mediated excitation and contributes to the development of cognitive and motor impairments. This review summarizes the role of Cl– (dys)regulation in the healthy and HD brain, with a focus on the basal ganglia (BG) circuitry and CCCs as potential therapeutic targets in the treatment of HD.
Collapse
|
30
|
Abstract
Neurodegenerative disorders can alter neural circuitry long before symptoms appear, but the path from early changes to later pathologies is obscure. In this issue of Neuron, Capizzi et al. (2021) show how early axonal growth defects in Huntington's disease create vulnerability to later degeneration.
Collapse
Affiliation(s)
- Morgan C Stephens
- Department of Human and Molecular Genetics, Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Vicky Brandt
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Juan Botas
- Department of Human and Molecular Genetics, Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|