1
|
Shapira G, Karmon G, Hacohen-Kleiman G, Ganaiem M, Shazman S, Theotokis P, Grigoriadis N, Shomron N, Gozes I. ADNP is essential for sex-dependent hippocampal neurogenesis, through male unfolded protein response and female mitochondrial gene regulation. Mol Psychiatry 2025; 30:2696-2706. [PMID: 39715923 DOI: 10.1038/s41380-024-02879-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/20/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Essential for brain formation and protective against tauopathy, activity-dependent neuroprotective protein (ADNP) is critical for neurogenesis and cognitive functions, while regulating steroid hormone biogenesis. As such, de novo mutations in ADNP lead to syndromic autism and somatic ADNP mutations parallel Alzheimer's disease progression. Furthermore, clinical trials with the ADNP fragment NAP (the investigational drug davunetide) showed efficacy in women suffering from the tauopathy progressive supranuclear palsy and differentially boosted memory in men (spatial) and women (verbal), exhibiting prodromal Alzheimer's disease. While autism is more prevalent in boys and Alzheimer's disease in women, both involve impaired neurogenesis. Here, we asked whether ADNP sex-dependently regulates neurogenesis. Using bromodeoxyuridine (BrdU) as a marker of neurogenesis, we identified two-fold higher labeling in the hippocampal sub-ventricular zone of ADNP-intact male versus female mice. Adnp haplo-insufficient (Adnp+/-) mice or mice CRSIPR/Cas9-edited to present the most prevalent neurodevelopmental ADNP syndrome mutation, p.Tyr718* (Tyr) showed dramatic reductions in male BrdU incorporation, resulting in mutated females presenting higher labeling than males. Treatment with NAP compensated for the male reduction of BrdU labeling. Mechanistically, hippocampal RNAseq revealed male-specific Tyr down-regulation of endoplasmic reticulum unfolded protein response genes critical for sex-dependent organogenesis. Newly discovered mitochondrial accessibility of ADNP was inhibited by the Tyr718* mutation further revealing female-specific Tyr downregulation of mitochondrial ATP6. NAP moderated much of the differential expression caused by p.Tyr718*, accompanied by the down-regulation of neurotoxic, pro-inflammatory and pro-apoptotic genes. Thus, ADNP is a key regulator of sex-dependent neurogenesis that acts by controlling canonical pathways, with NAP compensating for fundamental ADNP deficiencies, striding toward clinical development targeting the ADNP syndrome and related neurodevelopmental/neurodegenerative diseases.
Collapse
Affiliation(s)
- Guy Shapira
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Sagol School of Neuroscience, Edmond J Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Gidon Karmon
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Gal Hacohen-Kleiman
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Maram Ganaiem
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Shula Shazman
- Department of Mathematics and Computer Science, The Open University of Israel, Ra'anana, 4353701, Israel
| | - Paschalis Theotokis
- Department of Neurology, Laboratory of Experimental Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Department of Neurology, Laboratory of Experimental Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Noam Shomron
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Sagol School of Neuroscience, Edmond J Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Illana Gozes
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
2
|
Zhang K, Du Y, Yang S, Sun G. Irisin suppressed the progression of TBI via modulating AMPK/MerTK/autophagy and SYK/ROS/inflammatory signaling. Sci Rep 2025; 15:15583. [PMID: 40320408 PMCID: PMC12050266 DOI: 10.1038/s41598-025-00066-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Irisin is a hormone-like peptide secreted by muscle tissues and generated by hydrolysis of type III fibronectin domain-containing protein 5 by proteolytic hydrolases. Whether Irisin has a potential protective role in traumatic brain injury (TBI). In this study, we will investigate the relevant research progress of Irisin's protective role in traumatic brain injury (TBI) in recent years in terms of attenuating oxidative stress, inhibiting pyroptosis, suppressing inflammatory response, and improving autophagy, with the aim of providing valuable references for the diagnosis and treatment of traumatic brain injury (TBI). Utilize bioinformatics analysis to study the interactions between genes in TBI (Traumatic Brain Injury). Construct a TBI mouse model to observe the effects of Irisin on TBI. The Morris water maze test is used to assess the learning and spatial memory abilities of mice, TUNEL fluorescence is used to detect cell apoptosis, Nissl staining is employed to observe the survival of hippocampal neurons in mice, and HE staining is used to observe the extent of brain injury in mice. Western blot is used to detect protein expression in both in vivo and in vitro experiments. Q-PCR is employed to detect the levels of proteins related to autophagy/pyroptosis/inflammation. Irisin promotes MerTK overexpression by enhancing AMPK activation. Irisin can increase the expression of LC3I and Beclin-1 proteins, indicating the promotion of autophagic response. Additionally, Irisin reduces ROS levels and decreases SYK expression, thereby inhibiting the inflammatory response. Irisin improves the learning and spatial memory abilities of TBI mice and reduces cell apoptosis, as well as decreases hippocampal neuron death. HE staining shows that the brain injury in mice treated with Irisin is significantly alleviated. Irisin can enhance the expression of phosphorylated AMPK and phosphorylated MerTK proteins, promote autophagic response, and inhibit pyroptosis/inflammatory response. Correction experiments confirmed that after stimulation with an AMPK agonist, the expression of phosphorylated MerTK protein is significantly increased, autophagic response is enhanced, and pyroptosis/inflammatory response is weakened. When treated with a MerTK inhibitor during AMPK agonist stimulation, the autophagic response is weakened while pyroptosis/inflammatory response is enhanced. Irisin can inhibit the progression of traumatic brain injury by regulating AMPK/MerTK/autophagy and SYK/ROS/inflammatory signaling.
Collapse
Affiliation(s)
- Kuo Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang City, 050000, Hebei Province, China
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang City, 050000, Hebei Province, China
| | - Yihui Du
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang City, 050000, Hebei Province, China
| | - Sihui Yang
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang City, 050000, Hebei Province, China
| | - Guozhu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang City, 050000, Hebei Province, China.
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang City, 050000, Hebei Province, China.
| |
Collapse
|
3
|
Jiao X, Lu Y, Huang Y, Chen J, Gu Z, Gao X, Yuan L, Du B, Bi X. Plasma proteomic profiling reveals Parkinson's disease-associated proteins: A UK Biobank study. Parkinsonism Relat Disord 2025; 135:107851. [PMID: 40300504 DOI: 10.1016/j.parkreldis.2025.107851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
INTRODUCTION The rapid advancement of proteomics has provided new insights into early detection and prediction of Parkinson's disease (PD), particularly in identifying risk factors for PD. This study aims to develop a proteomics-based model to predict the risk of PD in patients. METHODS We analyzed data from the UK Biobank cohort, including 52,851 PD-free participants at baseline, with a median follow-up of 15.3 years and 811 newly diagnosed PD cases. A prospective proteomic analysis was conducted to assess the predictive value of 2,923 plasma proteins, and LightGBM models were used to calculate protein importance, followed by an evaluation of the proteins' predictive performance. RESULTS The study found that higher levels of NEFL and MERTK were significantly associated with future PD events, while lower levels of ITGAV, BAG3, CLEC10A, ITGAM, HNMT, and TPK1 were identified as potential risk factors for PD. Notably, the axonal injury marker NEFL and the thiamine metabolism-related protein TPK1 ranked higher than other proteins in terms of importance. The combination of NEFL and TPK1 significantly enhanced the predictive accuracy of conventional clinical models, increasing the Area Under the Curve (AUC) of the full-cohort prediction model from 0.784 to 0.842 and the 5-year prediction model from 0.780 to 0.908. CONCLUSIONS This study provides a novel insight for screening high-risk PD populations and underscores the significant role of nutritional metabolism in PD development, offering valuable insights for precision prevention strategies.
Collapse
Affiliation(s)
- Xuehao Jiao
- Department of Neurology, Changhai Hospital, Naval Medical University, Shanghai, PR China
| | - Yue Lu
- Department of Neurology, Changhai Hospital, Naval Medical University, Shanghai, PR China
| | - Yuxin Huang
- Department of Neurology, Changhai Hospital, Naval Medical University, Shanghai, PR China
| | - Jingjing Chen
- Department of Neurology, Changhai Hospital, Naval Medical University, Shanghai, PR China
| | - Zhengsheng Gu
- Department of Neurology, Changhai Hospital, Naval Medical University, Shanghai, PR China
| | - Xin Gao
- Department of Neurology, Changhai Hospital, Naval Medical University, Shanghai, PR China
| | - Lei Yuan
- Department of Health Management, Faculty of Military Health Service, Naval Medical University, Shanghai, PR China.
| | - Bingying Du
- Institute for Translational Brain Research, Fudan University, Shanghai, PR China.
| | - Xiaoying Bi
- Department of Neurology, Changhai Hospital, Naval Medical University, Shanghai, PR China.
| |
Collapse
|
4
|
Wang X, Li K, Guo L, Liu X, Guo Y, Zhang W. The Influence of Changes in Microglia Development on the Plasticity of the Developing Visual Cortex Circuit in Juvenile Mice. Invest Ophthalmol Vis Sci 2025; 66:45. [PMID: 40244609 PMCID: PMC12013681 DOI: 10.1167/iovs.66.4.45] [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: 09/15/2024] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose To investigate the role of microglial subtypes in mouse visual cortex development, focusing on ocular dominance plasticity and interactions with GABAergic neurons and the extracellular matrix. Methods Immunofluorescence and single-nucleus RNA-sequencing (snRNA-seq) were used to study microglia in the binocular primary visual cortex (V1) from postnatal day (P) 11 to P42. Gene ontology (GO) analysis assessed synapse organization, and the impact of microglial disruption on ocular dominance plasticity was examined. Visual evoked potentials and miniature postsynaptic current recordings are used to monitor functional changes in V1. Results Microglia underwent a marked expansion between P11 and P21 and stabilized after P35, coinciding with notable changes in gene expression that aligned with synaptic remodeling. GO analysis at P14 and P28 revealed significant enrichment in synaptic organization linked to microglia. Single-nucleus RNA sequencing identified six distinct microglial clusters, among which two functionally relevant subpopulations were closely linked to cortical synaptic plasticity. One cluster, enriched in inflammatory responses and endocytosis, peaked at P21, whereas another cluster, associated with synapse organization and signaling, exhibited dynamic changes after eye opening and during the critical period, significantly influencing cortical synaptic plasticity. In parallel, perineuronal nets (PNNs) and PV(+) interneuron populations increased and reached steady levels by P42, suggesting that microglia help coordinate the timing of inhibitory circuit maturation. Disrupting microglial function during the critical period impaired ocular dominance plasticity, but this effect was reversed after treatment cessation. Mechanistically, microglial depletion enhanced PV(+) interneuron numbers, elevated PNN expression, and altered synapse development. Conclusions Our findings highlight specific microglial subtypes as key regulators of cortical synapse development and plasticity through their interactions with PV(+) interneurons and PNNs. These insights advance our understanding of microglial contributions to visual cortex development and provide potential avenues for targeting microglial function to modulate cortical plasticity.
Collapse
Affiliation(s)
- Xuechun Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Kuan Li
- Department of Respiratory Medicine, Haihe Hospital, Tianjin University, Tianjin, China
| | - Lingzhi Guo
- Institute of Ophthalmology, Nankai University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xinlong Liu
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Yatu Guo
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
| | - Wei Zhang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
| |
Collapse
|
5
|
Spargo TP, Sands CF, Juan IR, Mitchell J, Ravanmehr V, Butts JC, De-Paula RB, Kim Y, Hu F, Wang Q, Vitsios D, Garg M, Middleton L, Tyrlik M, Messa M, Del Angel G, Calame DG, Saade H, Robak L, Hollis B, Cuddapah VA, Zoghbi HY, Shulman JM, Petrovski S, Al-Ramahi I, Tachmazidou I, Dhindsa RS. Haploinsufficiency of ITSN1 is associated with a substantial increased risk of Parkinson's disease. Cell Rep 2025; 44:115355. [PMID: 40056900 PMCID: PMC12124131 DOI: 10.1016/j.celrep.2025.115355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/18/2024] [Accepted: 02/06/2025] [Indexed: 03/10/2025] Open
Abstract
Despite its significant heritability, the genetic basis of Parkinson's disease (PD) remains incompletely understood. Here, in analyzing whole-genome sequence data from 3,809 PD cases and 247,101 controls in the UK Biobank, we discover that protein-truncating variants in ITSN1 confer a substantially increased risk of PD (p = 6.1 × 10-7; odds ratio [95% confidence interval] = 10.5 [5.2, 21.3]). We replicate this association in three independent datasets totaling 8,407 cases and 413,432 controls (combined p = 4.5 × 10-12). Notably, ITSN1 haploinsufficiency has also been associated with autism spectrum disorder, suggesting variable penetrance/expressivity. In Drosophila, we find that loss of the ITSN1 ortholog Dap160 exacerbates α-synuclein-induced neuronal toxicity and motor deficits, and in vitro assays further suggest a physical interaction between ITSN1 and α-synuclein. These results firmly establish ITSN1 as a PD risk gene with an effect size exceeding previously established loci, implicate vesicular trafficking dysfunction in PD pathogenesis, and potentially open new avenues for therapeutic development.
Collapse
Affiliation(s)
- Thomas P Spargo
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Chloe F Sands
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA; Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA
| | - Isabella R Juan
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan Mitchell
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Vida Ravanmehr
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA
| | - Jessica C Butts
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX, USA
| | - Ruth B De-Paula
- Quantitative and Computational Biology Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Youngdoo Kim
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Fengyuan Hu
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Quanli Wang
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Waltham, MA, USA
| | - Dimitrios Vitsios
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Manik Garg
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Lawrence Middleton
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Michal Tyrlik
- Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Mirko Messa
- Translational Genomics, Centre for Genomics Research, Discovery Sciences BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Guillermo Del Angel
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Waltham, MA, USA
| | - Daniel G Calame
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Hiba Saade
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Laurie Robak
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ben Hollis
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Vishnu A Cuddapah
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Huda Y Zoghbi
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Joshua M Shulman
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Department of Neurology, Baylor College of Medicine, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK; Department of Medicine, Austin Health, University of Melbourne, Melbourne, VIC, Australia.
| | - Ismael Al-Ramahi
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Ioanna Tachmazidou
- Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Ryan S Dhindsa
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St., Suite N.1150, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
6
|
Parhizkar S, Holtzman DM. The night's watch: Exploring how sleep protects against neurodegeneration. Neuron 2025; 113:817-837. [PMID: 40054454 PMCID: PMC11925672 DOI: 10.1016/j.neuron.2025.02.004] [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: 06/12/2024] [Revised: 10/15/2024] [Accepted: 02/04/2025] [Indexed: 03/21/2025]
Abstract
Sleep loss is often regarded as an early manifestation of neurodegenerative diseases given its common occurrence and link to cognitive dysfunction. However, the precise mechanisms by which sleep disturbances contribute to neurodegeneration are not fully understood, nor is it clear why some individuals are more susceptible to these effects than others. This review addresses critical unanswered questions in the field, including whether sleep disturbances precede or result from neurodegenerative diseases, the functional significance of sleep changes during the preclinical disease phase, and the potential role of sleep homeostasis as an adaptive mechanism enhancing resilience against cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Samira Parhizkar
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
7
|
Wu KM, Xu QH, Liu YQ, Feng YW, Han SD, Zhang YR, Chen SD, Guo Y, Wu BS, Ma LZ, Zhang Y, Chen YL, Yang L, Yang ZF, Xiao YJ, Wang TT, Zhao J, Chen SF, Cui M, Lu BX, Le WD, Shu YS, Ye K, Li JY, Li WS, Wang J, Liu C, Yuan P, Yu JT. Neuronal FAM171A2 mediates α-synuclein fibril uptake and drives Parkinson's disease. Science 2025; 387:892-900. [PMID: 39977508 DOI: 10.1126/science.adp3645] [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: 03/22/2024] [Revised: 10/21/2024] [Accepted: 01/10/2025] [Indexed: 02/22/2025]
Abstract
Neuronal accumulation and spread of pathological α-synuclein (α-syn) fibrils are key events in Parkinson's disease (PD) pathophysiology. However, the neuronal mechanisms underlying the uptake of α-syn fibrils remain unclear. In this work, we identified FAM171A2 as a PD risk gene that affects α-syn aggregation. Overexpressing FAM171A2 promotes α-syn fibril endocytosis and exacerbates the spread and neurotoxicity of α-syn pathology. Neuronal-specific knockdown of FAM171A2 expression shows protective effects. Mechanistically, the FAM171A2 extracellular domain 1 interacts with the α-syn C terminus through electrostatic forces, with >1000 times more selective for fibrils. Furthermore, we identified bemcentinib as an effective blocker of FAM171A2-α-syn fibril interaction with an in vitro binding assay, in cellular models, and in mice. Our findings identified FAM171A2 as a potential receptor for the neuronal uptake of α-syn fibrils and, thus, as a therapeutic target against PD.
Collapse
Affiliation(s)
- Kai-Min Wu
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qian-Hui Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yi-Qi Liu
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Wei Feng
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Si-Da Han
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling-Zhi Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Lin Chen
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhao-Fei Yang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu-Jie Xiao
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ting-Ting Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Jue Zhao
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shu-Fen Chen
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo-Xun Lu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei-Dong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Neurology, Sichuan Provincial People's Hospital, Medical School of University of Electronic Science and Technology of China, Chengdu, China
| | - You-Sheng Shu
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
- Institute of Health Sciences, China Medical University, Liaoning, Shenyang, China
| | - Wen-Sheng Li
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jian Wang
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Small Molecule Modulation of Biological Processes, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Shanghai, China
- Shanghai Academy of Natural Sciences (SANS), Fudan University, Shanghai, China
| | - Peng Yuan
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Rehabilitation Medicine, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Wang J, Li M, Zhao B, Chang R, Wu W, Zhang H, Usman M, Loor JJ, Xu C. A Disintegrin and Metalloproteinase 17 Disrupts Bovine Macrophage MER Proto-Oncogene Tyrosine Kinase Integrity to Impede Apoptotic Cell Clearance and Promote Inflammation in Clinical Mastitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:549-561. [PMID: 39731564 DOI: 10.1021/acs.jafc.4c09164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2024]
Abstract
In clinical mastitis of dairy cows, the abnormal accumulation of apoptotic cells (ACs) and subsequent secondary necrosis and inflammation pose significant concerns, with macrophage-mediated efferocytosis, crucial for ACs clearance, remaining unexplored in this context. In nonruminants, MER proto-oncogene tyrosine kinase (MERTK) receptors are essential for efferocytosis and A Disintegrin and Metalloproteinase 17 (ADAM17) is thought to play a role in regulating MERTK integrity. This study aimed to delineate the in situ role of efferocytosis in clinical mastitis, with a particular focus on the interaction between MERTK and ADAM17 in bovine macrophages. In mastitic mammary tissue, a significant accumulation of ACs was observed, along with active macrophage efferocytosis. Western blotting analysis revealed elevated expressions of MERTK and ADAM17, and immunofluorescence confirmed that MERTK is predominantly localized within CD163+ macrophages. Additionally, elevated levels of soluble MERTK (sol-MER) in serum indicated impaired integrity and functionality of MERTK. In vitro experiments with the bovine macrophage cell line Bo-mac cells selectively phagocytosed apoptotic MAC-T cells, a process associated with increased MERTK phosphorylation and an anti-inflammatory phenotype. The activation of ADAM17 by Phorbol 12-myristate 13-acetate (PMA) induced sol-Mer release and impaired efferocytosis, with these effects reversed by the ADAM17 inhibitor TAPI-1. Bo-mac efferocytosis was influenced by the presence and activation of MERTK. Silencing MERTK interrupted efferocytosis, a phenomenon also observed with the inhibition of MERTK phosphorylation by UNC2025, which concurrently suppressed anti-inflammatory cytokine production. These findings suggest that targeting the MERTK-ADAM17 axis could enhance inflammatory resolution, providing a novel therapeutic strategy for dairy cattle health management.
Collapse
Affiliation(s)
- Jingyi Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Ming Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Bichen Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Renxu Chang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Wenda Wu
- School of Food and Biological Engineering, Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230009, China
| | - Huijing Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Muhammad Usman
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Juan J Loor
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Chuang Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| |
Collapse
|
9
|
Sagredo GT, Tanglay O, Shahdadpuri S, Fu Y, Halliday GM. ⍺-Synuclein levels in Parkinson's disease - Cell types and forms that contribute to pathogenesis. Exp Neurol 2024; 379:114887. [PMID: 39009177 DOI: 10.1016/j.expneurol.2024.114887] [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: 04/02/2024] [Revised: 06/28/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Parkinson's disease (PD) has two main pathological hallmarks, the loss of nigral dopamine neurons and the proteinaceous aggregations of ⍺-synuclein (⍺Syn) in neuronal Lewy pathology. These two co-existing features suggest a causative association between ⍺Syn aggregation and the underpinning mechanism of neuronal degeneration in PD. Both increased levels and post-translational modifications of ⍺Syn can contribute to the formation of pathological aggregations of ⍺Syn in neurons. Recent studies have shown that the protein is also expressed by multiple types of non-neuronal cells in the brain and peripheral tissues, suggesting additional roles of the protein and potential diversity in non-neuronal pathogenic triggers. It is important to determine (1) the threshold levels triggering ⍺Syn to convert from a biological to a pathologic form in different brain cells in PD; (2) the dominant form of pathologic ⍺Syn and the associated post-translational modification of the protein in each cell type involved in PD; and (3) the cell type associated biological processes impacted by pathologic ⍺Syn in PD. This review integrates these aspects and speculates on potential pathological mechanisms and their impact on neuronal and non-neuronal ⍺Syn in the brains of patients with PD.
Collapse
Affiliation(s)
- Giselle Tatiana Sagredo
- The University of Sydney, Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, Sydney, NSW, Australia; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America
| | - Onur Tanglay
- The University of Sydney, Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, Sydney, NSW, Australia
| | - Shrey Shahdadpuri
- The University of Sydney, Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, Sydney, NSW, Australia
| | - YuHong Fu
- The University of Sydney, Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, Sydney, NSW, Australia; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America
| | - Glenda M Halliday
- The University of Sydney, Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, Sydney, NSW, Australia; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America.
| |
Collapse
|
10
|
Dorion MF, Casas D, Shlaifer I, Yaqubi M, Fleming P, Karpilovsky N, Chen CXQ, Nicouleau M, Piscopo VEC, MacDougall EJ, Alluli A, Goldsmith TM, Schneider A, Dorion S, Aprahamian N, MacDonald A, Thomas RA, Dudley RWR, Hall JA, Fon EA, Antel JP, Stratton JA, Durcan TM, La Piana R, Healy LM. An adapted protocol to derive microglia from stem cells and its application in the study of CSF1R-related disorders. Mol Neurodegener 2024; 19:31. [PMID: 38576039 PMCID: PMC10996091 DOI: 10.1186/s13024-024-00723-x] [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] [Accepted: 03/17/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Induced pluripotent stem cell-derived microglia (iMGL) represent an excellent tool in studying microglial function in health and disease. Yet, since differentiation and survival of iMGL are highly reliant on colony-stimulating factor 1 receptor (CSF1R) signaling, it is difficult to use iMGL to study microglial dysfunction associated with pathogenic defects in CSF1R. METHODS Serial modifications to an existing iMGL protocol were made, including but not limited to changes in growth factor combination to drive microglial differentiation, until successful derivation of microglia-like cells from an adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) patient carrying a c.2350G > A (p.V784M) CSF1R variant. Using healthy control lines, the quality of the new iMGL protocol was validated through cell yield assessment, measurement of microglia marker expression, transcriptomic comparison to primary microglia, and evaluation of inflammatory and phagocytic activities. Similarly, molecular and functional characterization of the ALSP patient-derived iMGL was carried out in comparison to healthy control iMGL. RESULTS The newly devised protocol allowed the generation of iMGL with enhanced transcriptomic similarity to cultured primary human microglia and with higher scavenging and inflammatory competence at ~ threefold greater yield compared to the original protocol. Using this protocol, decreased CSF1R autophosphorylation and cell surface expression was observed in iMGL derived from the ALSP patient compared to those derived from healthy controls. Additionally, ALSP patient-derived iMGL presented a migratory defect accompanying a temporal reduction in purinergic receptor P2Y12 (P2RY12) expression, a heightened capacity to internalize myelin, as well as heightened inflammatory response to Pam3CSK4. Poor P2RY12 expression was confirmed to be a consequence of CSF1R haploinsufficiency, as this feature was also observed following CSF1R knockdown or inhibition in mature control iMGL, and in CSF1RWT/KO and CSF1RWT/E633K iMGL compared to their respective isogenic controls. CONCLUSIONS We optimized a pre-existing iMGL protocol, generating a powerful tool to study microglial involvement in human neurological diseases. Using the optimized protocol, we have generated for the first time iMGL from an ALSP patient carrying a pathogenic CSF1R variant, with preliminary characterization pointing toward functional alterations in migratory, phagocytic and inflammatory activities.
Collapse
Affiliation(s)
- Marie-France Dorion
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Diana Casas
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Irina Shlaifer
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Moein Yaqubi
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Peter Fleming
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Nathan Karpilovsky
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- McGill Parkinson Program and Neurodegenerative Disorders Research Group, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Carol X-Q Chen
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Michael Nicouleau
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Valerio E C Piscopo
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Emma J MacDougall
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- McGill Parkinson Program and Neurodegenerative Disorders Research Group, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Aeshah Alluli
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Taylor M Goldsmith
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Alexandria Schneider
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Samuel Dorion
- Faculty of Arts and Sciences, Université de Montréal, Montreal, H3T 1NB, Canada
| | - Nathalia Aprahamian
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Adam MacDonald
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Rhalena A Thomas
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- McGill Parkinson Program and Neurodegenerative Disorders Research Group, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Roy W R Dudley
- Department of Pediatric Surgery, Division of Neurosurgery, Montreal Children's Hospital, McGill University Health Centers, Montreal, H4A 3J1, Canada
| | - Jeffrey A Hall
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- McGill Parkinson Program and Neurodegenerative Disorders Research Group, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Jo Anne Stratton
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Roberta La Piana
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada.
| | - Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada.
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada.
| |
Collapse
|