1
|
Shekarian T, Ritz MF, Hogan S, Martins TA, Schmassmann P, Gerber A, Roux J, Kaymak D, Durano C, Burger B, Matter M, Hutter G. Multidimensional analysis of matched primary and recurrent glioblastoma identifies contributors to tumor recurrence influencing time to relapse. J Neuropathol Exp Neurol 2025; 84:45-58. [PMID: 39423857 DOI: 10.1093/jnen/nlae108] [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] [Indexed: 10/21/2024] Open
Abstract
Glioblastoma (GBM) is a lethal brain tumor without effective treatment options. This study aimed to characterize longitudinal tumor changes in order to find potentially actionable targets to prevent GBM relapse. We extracted RNA and proteins from fresh frozen tumor samples from patient-matched IDHwt WHO grade 4 primary (pGBM) and recurrent (rGBM) tumors for transcriptomics and proteomics analysis. A tissue microarray containing paired tumor samples was processed for spatial transcriptomics analysis. Differentially expressed genes and proteins between pGBM and rGBM were involved in synapse development and myelination. By categorizing patients into short (STTR) and long (LTTR) time-to-lapse, we identified genes/proteins whose expression levels positively or negatively correlated with TTR. In rGBM, expressions of Fcγ receptors (FCGRs) and complement system genes were negatively correlated with TTR, whereas expression of genes involved in DNA methylation was positively correlated with TTR. Spatial transcriptomics of the tumor cells showed enrichment of oligodendrocytes in rGBM. Besides, we observed changes in the myeloid compartment such as a switch from quiescent to activated microglia and an enrichment in B and T cells in rGBM with STTR. Our results uncover a role for activated microglia/macrophages in GBM recurrence and suggest that interfering with these cells may hinder GBM relapse.
Collapse
Affiliation(s)
- Tala Shekarian
- Brain Tumor Immunotherapy and Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Marie-Françoise Ritz
- Brain Tumor Immunotherapy and Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Sabrina Hogan
- Brain Tumor Immunotherapy and Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Tomás A Martins
- Brain Tumor Immunotherapy and Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Philip Schmassmann
- Brain Tumor Immunotherapy and Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Alexandra Gerber
- Brain Tumor Immunotherapy and Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Julien Roux
- Bioinformatics Core Facility, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Deniz Kaymak
- Brain Tumor Immunotherapy and Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Célia Durano
- Brain Tumor Immunotherapy and Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Bettina Burger
- Dermatology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Matthias Matter
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Gregor Hutter
- Brain Tumor Immunotherapy and Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
- Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
2
|
Pan Q, Jiang L, Xiong Y, Chao FL, Liu S, Zhang SS, Zhu L, Luo YM, Xiao Q, Tang J, Liang X, Tang Y, Zhou CN, Zhang L. Voluntary running exercise promotes maturation differentiation and myelination of oligodendrocytes around Aβ plaques in the medial prefrontal cortex of APP/PS1 mice. Brain Res Bull 2024; 220:111170. [PMID: 39675487 DOI: 10.1016/j.brainresbull.2024.111170] [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: 10/15/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Previous studies have reported that running exercise could improves myelinization in hippocampus. However, the effects of running exercise on the differentiation and maturation of oligodendrocytes, and myelination surrounding Aβ plaques in the medial prefrontal cortex (mPFC) of the Alzheimer's disease (AD) brain have not been reported. METHODS Forty 10-month-old male APP/PS1 AD mice were randomly divided into the AD group and the AD running (AD+RUN) group, while 20 age-matched wild-type littermate mice were included in the WT group. The running group received three-month voluntary running exercise in a running cage, while the AD and WT groups were untreated. After the exercise intervention, all mice were given behavioral tests. The total number of mature oligodendrocytes (CC1+) in the mPFC of mice was precisely quantified using unbiased stereology. Myelin basic protein (MBP) and Aβ plaque, as well as the fluorescence area of MBP surrounding Aβ plaques, and the density and morphology of PDGFα+ cells in the mPFC were analyzed using immunofluorescence. RESULTS The levels of working memory, cognitive memory, spatial learning and memory ability were decreased significantly in the AD group compared to the WT group, while these functions were significantly improved in the AD+RUN group compared to the AD group. The Aβ plaques in the mPFC were significantly reduced in the AD+RUN group compared to the AD group. The total number of CC1+ cells and the percentage of MBP fluorescence area surrounding Aβ plaques in the mPFC were significantly lower in the AD group compared to the WT group, but they were significantly higher in the AD+RUN group compared to the AD group. The density and branching complexity of PDGFα+ cells surrounding Aβ plaques in the mPFC were significantly higher in the AD group than in the WT group, while the AD+RUN group showed significantly lower density and branching complexity than the AD group. Changes in MBP expression around Aβ plaques, cell density and cell branching complexity of PDGFα+ cells around Aβ plaques were closely related to the number of Aβ plaques in mPFC, and they were also closely related to behavioral changes in mice. CONCLUSIONS Voluntary running exercise could reduce Aβ plaque deposition and promote the maturation and myelination capacity of oligodendrocytes surrounding Aβ plaques in the mPFC of AD mice, thereby improving the learning and memory abilities of APP/PS1 transgenic AD mice.
Collapse
Affiliation(s)
- Qing Pan
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Jiang
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Yao Xiong
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Feng-Lei Chao
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Shan Liu
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Shan-Shan Zhang
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Zhu
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Yan-Min Luo
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Physiology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Qian Xiao
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Radioactive Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Tang
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Xin Liang
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Pathology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Yong Tang
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Chun-Ni Zhou
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China.
| | - Lei Zhang
- Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
3
|
Pivoňková H, Sitnikov S, Kamen Y, Vanhaesebrouck A, Matthey M, Spitzer SO, Ng YT, Tao C, de Faria O, Varga BV, Káradóttir RT. Heterogeneity in oligodendrocyte precursor cell proliferation is dynamic and driven by passive bioelectrical properties. Cell Rep 2024; 43:114873. [PMID: 39423130 PMCID: PMC11602547 DOI: 10.1016/j.celrep.2024.114873] [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: 01/10/2023] [Revised: 07/12/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) generate myelinating oligodendrocytes and are the main proliferative cells in the adult central nervous system. OPCs are a heterogeneous population, with proliferation and differentiation capacity varying with brain region and age. We demonstrate that during early postnatal maturation, cortical, but not callosal, OPCs begin to show altered passive bioelectrical properties, particularly increased inward potassium (K+) conductance, which correlates with G1 cell cycle stage and affects their proliferation potential. Neuronal activity-evoked transient K+ currents in OPCs with high inward K+ conductance potentially release OPCs from cell cycle arrest. Eventually, OPCs in all regions acquire high inward K+ conductance, the magnitude of which may underlie differences in OPC proliferation between regions, with cells being pushed into a dormant state as they acquire high inward K+ conductance and released from dormancy by synchronous neuronal activity. Age-related accumulation of OPCs with high inward K+ conductance might contribute to differentiation failure.
Collapse
Affiliation(s)
- Helena Pivoňková
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Sergey Sitnikov
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Yasmine Kamen
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - An Vanhaesebrouck
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Moritz Matthey
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Sonia Olivia Spitzer
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Yan Ting Ng
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Chenyue Tao
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Omar de Faria
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Balazs Viktor Varga
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Ragnhildur Thóra Káradóttir
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK; Department of Physiology, BioMedical Center, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland.
| |
Collapse
|
4
|
He Y, Xu Z, He Y, Liu J, Li J, Wang S, Xiao L. Preventing production of new oligodendrocytes impairs remyelination and sustains behavioural deficits after demyelination. Biochem Biophys Res Commun 2024; 733:150592. [PMID: 39213705 DOI: 10.1016/j.bbrc.2024.150592] [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: 05/23/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Damage to oligodendrocytes (OLs) and myelin sheaths (demyelination) has been shown to be associated with numerous neurological and psychiatric disorders. Remyelination is a rare and reliable regenerative response that occurs in the central nervous system (CNS). It is generally believed that OL progenitor cells (OPCs) are the cell source to generate new OLs to remyelinate the demyelinated axons. However, several recent studies have argued that pre-existing mature OLs that survive within the demyelinated area are responsible for remyelination. Here, by conditional knock-out (KO) of a transcription factor gene that is essential for OPC differentiation, namely myelin regulatory factor (Myrf), to block the production of adult new OLs and examined its effect on remyelination after cuprizone (CPZ)-induced demyelination. We found that OPCs specific Myrf cKO mice show dramatic impairment in remyelination after 4 weeks of recovery from 5 weeks of CPZ diet and they leave over significant behavioral deficits such as anxiety-like behavior, decreased motor skills, and impaired memory compared to control mice that have recovered for the same time. Our data support the idea that OPCs are the major cell sources for myelin regeneration, suggesting that targeting the activation of OPCs and promoting their differentiation to boost new OLs production is critical for therapeutic intervention for demyelinating diseases such as multiple sclerosis (MS).
Collapse
Affiliation(s)
- Yuehua He
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Zhengtao Xu
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Yongxiang He
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Junhong Liu
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Jiong Li
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Shuming Wang
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Lin Xiao
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
5
|
Bromley-Coolidge S, Iruegas D, Appel B. Cspg4 sculpts oligodendrocyte precursor cell morphology. Differentiation 2024; 140:100819. [PMID: 39566199 PMCID: PMC11637897 DOI: 10.1016/j.diff.2024.100819] [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: 08/08/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024]
Abstract
The extracellular matrix (ECM) provides critical biochemical and structural cues that regulate neural development. Chondroitin sulfate proteoglycans (CSPGs), a major ECM component, have been implicated in modulating oligodendrocyte precursor cell (OPC) proliferation, migration, and maturation, but their specific roles in oligodendrocyte lineage cell (OLC) development and myelination in vivo remain poorly understood. Here, we use zebrafish as a model system to investigate the spatiotemporal dynamics of ECM deposition and CSPG localization during central nervous system (CNS) development, with a focus on their relationship to OLCs. We demonstrate that ECM components, including CSPGs, are dynamically expressed in distinct spatiotemporal patterns coinciding with OLC development and myelination. We found that zebrafish lacking cspg4 function produced normal numbers of OLCs, which appeared to undergo proper differentiation. However, OPC morphology in mutant larvae was aberrant. Nevertheless, the number and length of myelin sheaths produced by mature oligodendrocytes were unaffected. These data indicate that Cspg4 regulates OPC morphogenesis in vivo, supporting the role of the ECM in neural development.
Collapse
Affiliation(s)
- Samantha Bromley-Coolidge
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80445, USA
| | - Diego Iruegas
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80445, USA
| | - Bruce Appel
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80445, USA.
| |
Collapse
|
6
|
Heo D, Kim AA, Neumann B, Doze VN, Xu YKT, Mironova YA, Slosberg J, Goff LA, Franklin RJM, Bergles DE. Transcriptional profiles of murine oligodendrocyte precursor cells across the lifespan. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.27.620502. [PMID: 39554158 PMCID: PMC11565715 DOI: 10.1101/2024.10.27.620502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Oligodendrocyte progenitor cells (OPCs) are highly dynamic, widely distributed glial cells of the central nervous system (CNS) that are responsible for generating myelinating oligodendrocytes during development. By also generating new oligodendrocytes in the adult CNS, OPCs allow formation of new myelin sheaths in response to environmental and behavioral changes and play a crucial role in regenerating myelin following demyelination (remyelination). However, the rates of OPC proliferation and differentiation decline dramatically with aging, which may impair homeostasis, remyelination, and adaptive myelination during learning. To determine how aging influences OPCs, we generated a novel transgenic mouse line that expresses membrane-anchored EGFP under the endogenous promoter/enhancer of Matrilin-4 (Matn4-mEGFP) and performed high-throughput single-cell RNA sequencing, providing enhanced resolution of transcriptional changes during key transitions from quiescence to proliferation and differentiation across the lifespan. Comparative analysis of OPCs isolated from mice aged 30 to 720 days, revealed that aging induces distinct inflammatory transcriptomic changes in OPCs in different states, including enhanced activation of HIF-1α and Wnt pathways. Inhibition of these pathways in acutely isolated OPCs from aged animals restored their ability to differentiate, suggesting that this enhanced signaling may contribute to the decreased regenerative potential of OPCs with aging. This Matn4-mEGFP mouse line and single-cell mRNA datasets of cortical OPCs across ages help to define the molecular changes guiding their behavior in various physiological and pathological contexts.
Collapse
Affiliation(s)
- Dongeun Heo
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Anya A. Kim
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Björn Neumann
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Valerie N. Doze
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yu Kang T. Xu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yevgeniya A. Mironova
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jared Slosberg
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Loyal A. Goff
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Robin J. M. Franklin
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Dwight E. Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
McDonough A, Weinstein JR. Glial 'omics in ischemia: Acute stroke and chronic cerebral small vessel disease. Glia 2024. [PMID: 39463002 DOI: 10.1002/glia.24634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
Vascular injury and pathologies underlie common diseases including ischemic stroke and cerebral small vessel disease (CSVD). Prior work has identified a key role for glial cells, including microglia, in the multifaceted and temporally evolving neuroimmune response to both stroke and CSVD. Transcriptional profiling has led to important advances including identification of distinct gene expression signatures in ischemia-exposed, flow cytometrically sorted microglia and more recently single cell RNA sequencing-identified microglial subpopulations or clusters. There is a reassuring degree of overlap in the results from these two distinct methodologies with both identifying a proliferative and a separate type I interferon responsive microglial element. Similar patterns were later seen using multimodal and spatial transcriptomal profiling in ischemia-exposed microglia and astrocytes. Methodological advances including enrichment of specific neuroanatomic/functional regions (such as the neurovascular unit) prior to single cell RNA sequencing has led to identification of novel cellular subtypes and generation of new credible hypotheses as to cellular function based on the enhanced cell sub-type specific gene expression patterns. A ribosomal tagging strategy focusing on the cellular translatome analyses carried out in the acute phases post stroke has revealed distinct inflammation-regulating roles for microglia and astrocytes in this setting. Early spatial transcriptomics experiments using cerebral ischemia models have identified regionally distinct microglial cell clusters in ischemic core versus penumbra. There is great potential for combination of these methods for multi-omics approaches to further elucidate glial responses in the context of both acute ischemic stroke and chronic CSVD.
Collapse
Affiliation(s)
- Ashley McDonough
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Jonathan R Weinstein
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA
- Department of Neurological Surgery, School of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
8
|
Collins HY, Doan RA, Li J, Early JE, Madden ME, Simkins T, Lyons DA, Monk KR, Emery B. FBXW7 regulates MYRF levels to control myelin capacity and homeostasis in the adult CNS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618515. [PMID: 39464137 PMCID: PMC11507870 DOI: 10.1101/2024.10.15.618515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Myelin, along with the oligodendrocytes (OLs) that produce it, is essential for proper central nervous system (CNS) function in vertebrates. Although the accurate targeting of myelin to axons and its maintenance are critical for CNS performance, the molecular pathways that regulate these processes remain poorly understood. Through a combination of zebrafish genetics, mouse models, and primary OL cultures, we found FBXW7, a recognition subunit of an E3 ubiquitin ligase complex, is a regulator of adult myelination in the CNS. Loss of Fbxw7 in myelinating OLs resulted in increased myelin sheath lengths with no change in myelin thickness. As the animals aged, they developed progressive abnormalities including myelin outfolds, disrupted paranodal organization, and ectopic ensheathment of neuronal cell bodies with myelin. Through biochemical studies we found that FBXW7 directly binds and degrades the N-terminal of Myelin Regulatory Factor (N-MYRF), to control the balance between oligodendrocyte myelin growth and homeostasis.
Collapse
Affiliation(s)
- Hannah Y. Collins
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239, USA
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Ryan A. Doan
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Jiaxing Li
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Jason E. Early
- Centre for Discovery Brain Sciences, MS society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Megan E. Madden
- Centre for Discovery Brain Sciences, MS society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Tyrell Simkins
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239, USA
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - David A. Lyons
- Centre for Discovery Brain Sciences, MS society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Kelly R. Monk
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239, USA
| |
Collapse
|
9
|
Brown RI, Barber HM, Kucenas S. Satellite glial cell manipulation prior to axotomy enhances developing dorsal root ganglion central branch regrowth into the spinal cord. Glia 2024; 72:1766-1784. [PMID: 39141572 PMCID: PMC11325082 DOI: 10.1002/glia.24581] [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] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 08/16/2024]
Abstract
The central and peripheral nervous systems (CNS and PNS, respectively) exhibit remarkable diversity in the capacity to regenerate following neuronal injury with PNS injuries being much more likely to regenerate than those that occur in the CNS. Glial responses to damage greatly influence the likelihood of regeneration by either promoting or inhibiting axonal regrowth over time. However, despite our understanding of how some glial lineages participate in nerve degeneration and regeneration, less is known about the contributions of peripheral satellite glial cells (SGC) to regeneration failure following central axon branch injury of dorsal root ganglia (DRG) sensory neurons. Here, using in vivo, time-lapse imaging in larval zebrafish coupled with laser axotomy, we investigate the role of SGCs in axonal regeneration. In our studies we show that SGCs respond to injury by relocating their nuclei to the injury site during the same period that DRG neurons produce new central branch neurites. Laser ablation of SGCs prior to axon injury results in more neurite growth attempts and ultimately a higher rate of successful central axon regrowth, implicating SGCs as inhibitors of regeneration. We also demonstrate that this SGC response is mediated in part by ErbB signaling, as chemical inhibition of this receptor results in reduced SGC motility and enhanced central axon regrowth. These findings provide new insights into SGC-neuron interactions under injury conditions and how these interactions influence nervous system repair.
Collapse
Affiliation(s)
- Robin I Brown
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Heather M Barber
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
- Cell & Developmental Biology Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
10
|
Simons M, Gibson EM, Nave KA. Oligodendrocytes: Myelination, Plasticity, and Axonal Support. Cold Spring Harb Perspect Biol 2024; 16:a041359. [PMID: 38621824 PMCID: PMC11444305 DOI: 10.1101/cshperspect.a041359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The myelination of axons has evolved to enable fast and efficient transduction of electrical signals in the vertebrate nervous system. Acting as an electric insulator, the myelin sheath is a multilamellar membrane structure around axonal segments generated by the spiral wrapping and subsequent compaction of oligodendroglial plasma membranes. These oligodendrocytes are metabolically active and remain functionally connected to the subjacent axon via cytoplasmic-rich myelinic channels for movement of metabolites and macromolecules to and from the internodal periaxonal space under the myelin sheath. Increasing evidence indicates that oligodendrocyte numbers, specifically in the forebrain, and myelin as a dynamic cellular compartment can both respond to physiological demands, collectively referred to as adaptive myelination. This review summarizes our current understanding of how myelin is generated, how its function is dynamically regulated, and how oligodendrocytes support the long-term integrity of myelinated axons.
Collapse
Affiliation(s)
- Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich 80802, Germany
- German Center for Neurodegenerative Diseases, Munich Cluster of Systems Neurology (SyNergy), Institute for Stroke and Dementia Research, Munich 81377, Germany
| | - Erin M Gibson
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford 94305, California, USA
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37075, Germany
| |
Collapse
|
11
|
Ma Z, Zhang W, Wang C, Su Y, Yi C, Niu J. A New Acquaintance of Oligodendrocyte Precursor Cells in the Central Nervous System. Neurosci Bull 2024; 40:1573-1589. [PMID: 39042298 PMCID: PMC11422404 DOI: 10.1007/s12264-024-01261-8] [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: 11/06/2023] [Accepted: 03/21/2024] [Indexed: 07/24/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are a heterogeneous multipotent population in the central nervous system (CNS) that appear during embryogenesis and persist as resident cells in the adult brain parenchyma. OPCs could generate oligodendrocytes to participate in myelination. Recent advances have renewed our knowledge of OPC biology by discovering novel markers of oligodendroglial cells, the myelin-independent roles of OPCs, and the regulatory mechanism of OPC development. In this review, we will explore the updated knowledge on OPC identity, their multifaceted roles in the CNS in health and diseases, as well as the regulatory mechanisms that are involved in their developmental stages, which hopefully would contribute to a further understanding of OPCs and attract attention in the field of OPC biology.
Collapse
Affiliation(s)
- Zexuan Ma
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
| | - Wei Zhang
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
| | - Chenmeng Wang
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China.
- Shenzhen Key Laboratory of Chinese Medicine Active substance screening and Translational Research, Shenzhen, 518107, China.
| | - Jianqin Niu
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China.
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400038, China.
| |
Collapse
|
12
|
Das S, Shaw AK, Das Sarma S, Koval M, Das Sarma J, Maulik M. Neurotropic Murine β-Coronavirus Infection Causes Differential Expression of Connexin 47 in Oligodendrocyte Subpopulations Associated with Demyelination. Mol Neurobiol 2024:10.1007/s12035-024-04482-0. [PMID: 39292341 DOI: 10.1007/s12035-024-04482-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
Gap junctions (GJs) play a crucial role in the survival of oligodendrocytes and myelination of the central nervous system (CNS). In this study, we investigated the spatiotemporal changes in the expression of oligodendroglial GJ protein connexin 47 (Cx47), its primary astroglial coupling partner, Cx43, and their association with demyelination following intracerebral infection with mouse hepatitis virus (MHV). Neurotropic strains of MHV, a β-coronavirus, induce an acute encephalomyelitis followed by a chronic demyelinating disease that shares similarities with the human disease multiple sclerosis (MS). Our results reveal that Cx47 GJs are persistently lost in mature oligodendrocytes, not only in demyelinating lesions but also in surrounding normal appearing white and gray matter areas, following an initial loss of astroglial Cx43 GJs during acute infection. At later stages after viral clearance, astroglial Cx43 GJs re-emerge but mature oligodendrocytes fail to fully re-establish GJs with astrocytes due to lack of Cx47 GJ expression. In contrast, at this later demyelinating stage, the increased oligodendrocyte precursor cells appear to exhibit Cx47 GJs. Our findings further highlight varying degrees of demyelination in distinct spinal cord regions, with the thoracic cord showing the most pronounced demyelination. The regional difference in demyelination correlates well with dynamic changes in the proportion of different oligodendrocyte lineage cells exhibiting differential Cx47 GJ expression, suggesting an important mechanism of progressive demyelination even after viral clearance.
Collapse
Affiliation(s)
- Soubhik Das
- Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, 741251, West Bengal, India
| | - Archana Kumari Shaw
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, West Bengal, India
| | - Subhajit Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, West Bengal, India
| | - Michael Koval
- Departments of Medicine and Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, West Bengal, India
| | - Mahua Maulik
- Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, 741251, West Bengal, India.
| |
Collapse
|
13
|
Chen H, Yang G, Xu DE, Du YT, Zhu C, Hu H, Luo L, Feng L, Huang W, Sun YY, Ma QH. Autophagy in Oligodendrocyte Lineage Cells Controls Oligodendrocyte Numbers and Myelin Integrity in an Age-dependent Manner. Neurosci Bull 2024:10.1007/s12264-024-01292-1. [PMID: 39283565 DOI: 10.1007/s12264-024-01292-1] [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: 04/24/2024] [Accepted: 07/10/2024] [Indexed: 12/08/2024] Open
Abstract
Oligodendrocyte lineage cells, including oligodendrocyte precursor cells (OPCs) and oligodendrocytes (OLs), are essential in establishing and maintaining brain circuits. Autophagy is a conserved process that keeps the quality of organelles and proteostasis. The role of autophagy in oligodendrocyte lineage cells remains unclear. The present study shows that autophagy is required to maintain the number of OPCs/OLs and myelin integrity during brain aging. Inactivation of autophagy in oligodendrocyte lineage cells increases the number of OPCs/OLs in the developing brain while exaggerating the loss of OPCs/OLs with brain aging. Inactivation of autophagy in oligodendrocyte lineage cells impairs the turnover of myelin basic protein (MBP). It causes MBP to accumulate in the cytoplasm as multimeric aggregates and fails to be incorporated into integral myelin, which is associated with attenuated endocytic recycling. Inactivation of autophagy in oligodendrocyte lineage cells impairs myelin integrity and causes demyelination. Thus, this study shows autophagy is required to maintain myelin quality during aging by controlling the turnover of myelin components.
Collapse
Affiliation(s)
- Hong Chen
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Gang Yang
- Lab Center, Medical College of Soochow University, Suzhou, 215021, China
| | - De-En Xu
- The Wuxi No.2 People Hospital, Wuxi, 214002, China
| | - Yu-Tong Du
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Chao Zhu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Hua Hu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Li Luo
- School of Physical Education and Sports Science, Soochow University, Suzhou, 215021, China
| | - Lei Feng
- Monash Suzhou Research Institute, Suzhou, 215000, China
| | - Wenhui Huang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421, Homburg, Germany
| | - Yan-Yun Sun
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| | - Quan-Hong Ma
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
14
|
Giacomoni J, Bruzelius A, Habekost M, Kajtez J, Ottosson DR, Fiorenzano A, Storm P, Parmar M. 3D model for human glia conversion into subtype-specific neurons, including dopamine neurons. CELL REPORTS METHODS 2024; 4:100845. [PMID: 39236715 PMCID: PMC11440053 DOI: 10.1016/j.crmeth.2024.100845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/05/2024] [Accepted: 08/08/2024] [Indexed: 09/07/2024]
Abstract
Two-dimensional neuronal cultures have a limited ability to recapitulate the in vivo environment of the brain. Here, we introduce a three-dimensional in vitro model for human glia-to-neuron conversion, surpassing the spatial and temporal constrains of two-dimensional cultures. Focused on direct conversion to induced dopamine neurons (iDANs) relevant to Parkinson disease, the model generates functionally mature iDANs in 2 weeks and allows long-term survival. As proof of concept, we use single-nucleus RNA sequencing and molecular lineage tracing during iDAN generation and find that all glial subtypes generate neurons and that conversion relies on the coordinated expression of three neural conversion factors. We also show the formation of mature and functional iDANs over time. The model facilitates molecular investigations of the conversion process to enhance understanding of conversion outcomes and offers a system for in vitro reprogramming studies aimed at advancing alternative therapeutic strategies in the diseased brain.
Collapse
Affiliation(s)
- Jessica Giacomoni
- Developmental and Regenerative Neurobiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Andreas Bruzelius
- Regenerative Neurophysiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Mette Habekost
- Developmental and Regenerative Neurobiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Janko Kajtez
- Developmental and Regenerative Neurobiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Daniella Rylander Ottosson
- Regenerative Neurophysiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Alessandro Fiorenzano
- Developmental and Regenerative Neurobiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Petter Storm
- Developmental and Regenerative Neurobiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden.
| |
Collapse
|
15
|
Jeon S, Park J, Likhite S, Moon JH, Shin D, Li L, Meyer KC, Lee JW, Lee SK. The postnatal injection of AAV9-FOXG1 rescues corpus callosum agenesis and other brain deficits in the mouse model of FOXG1 syndrome. Mol Ther Methods Clin Dev 2024; 32:101275. [PMID: 39022742 PMCID: PMC11253142 DOI: 10.1016/j.omtm.2024.101275] [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: 01/11/2023] [Accepted: 05/31/2024] [Indexed: 07/20/2024]
Abstract
Heterozygous mutations in the FOXG1 gene manifest as FOXG1 syndrome, a severe neurodevelopmental disorder characterized by structural brain anomalies, including agenesis of the corpus callosum, hippocampal reduction, and myelination delays. Despite the well-defined genetic basis of FOXG1 syndrome, therapeutic interventions targeting the underlying cause of the disorder are nonexistent. In this study, we explore the therapeutic potential of adeno-associated virus 9 (AAV9)-mediated delivery of the FOXG1 gene. Remarkably, intracerebroventricular injection of AAV9-FOXG1 to Foxg1 heterozygous mouse model at the postnatal stage rescues a wide range of brain pathologies. This includes the amelioration of corpus callosum deficiencies, the restoration of dentate gyrus morphology in the hippocampus, the normalization of oligodendrocyte lineage cell numbers, and the rectification of myelination anomalies. Our findings highlight the efficacy of AAV9-based gene therapy as a viable treatment strategy for FOXG1 syndrome and potentially other neurodevelopmental disorders with similar brain malformations, asserting its therapeutic relevance in postnatal stages.
Collapse
Affiliation(s)
- Shin Jeon
- Department of Biological Sciences, College of Arts and Sciences, FOXG1 Research Center, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
- Department of Systems Pharmacology & Translational Therapeutics, Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jaein Park
- Department of Biological Sciences, College of Arts and Sciences, FOXG1 Research Center, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
| | - Shibi Likhite
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Ji Hwan Moon
- Department of Biological Sciences, College of Arts and Sciences, FOXG1 Research Center, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea
| | - Dongjun Shin
- Department of Biological Sciences, College of Arts and Sciences, FOXG1 Research Center, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
| | - Liwen Li
- Department of Biological Sciences, College of Arts and Sciences, FOXG1 Research Center, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
| | - Kathrin C. Meyer
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Jae W. Lee
- Department of Biological Sciences, College of Arts and Sciences, FOXG1 Research Center, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
| | - Soo-Kyung Lee
- Department of Biological Sciences, College of Arts and Sciences, FOXG1 Research Center, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
| |
Collapse
|
16
|
Beachum AN, Salazar G, Nachbar A, Krause K, Klose H, Meyer K, Maserejian A, Ross G, Boyd H, Weigel T, Ambaye L, Miller H, Coutinho-Budd J. Glia multitask to compensate for neighboring glial cell dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611719. [PMID: 39314422 PMCID: PMC11418964 DOI: 10.1101/2024.09.06.611719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
As glia mature, they undergo glial tiling to abut one another without invading each other's boundaries. Upon the loss of the secreted neurotrophin Spätzle3 (Spz3), Drosophila cortex glia transform morphologically and lose their intricate interactions with neurons and surrounding glial subtypes. Here, we reveal that all neighboring glial cell types (astrocytes, ensheathing glia, and subperineurial glia) react by extending processes into the previous cortex glial territory to compensate for lost cortex glial function and reduce the buildup of neuronal debris. However, the loss of Spz3 alone is not sufficient for glia to cross their natural borders, as blocking CNS growth via nutrient-restriction blocks the aberrant infiltration induced by the loss of Spz3. Surprisingly, even when these neighboring glia divert their cellular resources beyond their typical borders to take on new compensatory roles, they are able to multitask to continue to preserve their own normal functions to maintain CNS homeostasis.
Collapse
Affiliation(s)
- Allison N. Beachum
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Gabriela Salazar
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Amelia Nachbar
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Kevin Krause
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Hannah Klose
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Kate Meyer
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | | | - Grace Ross
- Department of Biology, University of Vermont, Burlington, VT 05405
| | - Hannah Boyd
- Department of Biology, University of Vermont, Burlington, VT 05405
| | - Thaddeus Weigel
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Lydia Ambaye
- Department of Biology, University of Vermont, Burlington, VT 05405
| | - Hayes Miller
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Jaeda Coutinho-Budd
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| |
Collapse
|
17
|
Xin W, Kaneko M, Roth RH, Zhang A, Nocera S, Ding JB, Stryker MP, Chan JR. Oligodendrocytes and myelin limit neuronal plasticity in visual cortex. Nature 2024; 633:856-863. [PMID: 39169185 PMCID: PMC11424474 DOI: 10.1038/s41586-024-07853-8] [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: 10/03/2023] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
Developmental myelination is a protracted process in the mammalian brain1. One theory for why oligodendrocytes mature so slowly posits that myelination may stabilize neuronal circuits and temper neuronal plasticity as animals age2-4. We tested this theory in the visual cortex, which has a well-defined critical period for experience-dependent neuronal plasticity5. During adolescence, visual experience modulated the rate of oligodendrocyte maturation in visual cortex. To determine whether oligodendrocyte maturation in turn regulates neuronal plasticity, we genetically blocked oligodendrocyte differentiation and myelination in adolescent mice. In adult mice lacking adolescent oligodendrogenesis, a brief period of monocular deprivation led to a significant decrease in visual cortex responses to the deprived eye, reminiscent of the plasticity normally restricted to adolescence. This enhanced functional plasticity was accompanied by a greater turnover of dendritic spines and coordinated reductions in spine size following deprivation. Furthermore, inhibitory synaptic transmission, which gates experience-dependent plasticity at the circuit level, was diminished in the absence of adolescent oligodendrogenesis. These results establish a critical role for oligodendrocytes in shaping the maturation and stabilization of cortical circuits and support the concept of developmental myelination acting as a functional brake on neuronal plasticity.
Collapse
Affiliation(s)
- Wendy Xin
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| | - Megumi Kaneko
- Department of Physiology, Kavli Institute for Fundamental Neuroscience and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Richard H Roth
- Departments of Neurosurgery and Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Albert Zhang
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Sonia Nocera
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Jun B Ding
- Departments of Neurosurgery and Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michael P Stryker
- Department of Physiology, Kavli Institute for Fundamental Neuroscience and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Jonah R Chan
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
18
|
Xu C, Nedergaard M, Fowell DJ, Friedl P, Ji N. Multiphoton fluorescence microscopy for in vivo imaging. Cell 2024; 187:4458-4487. [PMID: 39178829 PMCID: PMC11373887 DOI: 10.1016/j.cell.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/26/2024]
Abstract
Multiphoton fluorescence microscopy (MPFM) has been a game-changer for optical imaging, particularly for studying biological tissues deep within living organisms. MPFM overcomes the strong scattering of light in heterogeneous tissue by utilizing nonlinear excitation that confines fluorescence emission mostly to the microscope focal volume. This enables high-resolution imaging deep within intact tissue and has opened new avenues for structural and functional studies. MPFM has found widespread applications and has led to numerous scientific discoveries and insights into complex biological processes. Today, MPFM is an indispensable tool in many research communities. Its versatility and effectiveness make it a go-to technique for researchers investigating biological phenomena at the cellular and subcellular levels in their native environments. In this Review, the principles, implementations, capabilities, and limitations of MPFM are presented. Three application areas of MPFM, neuroscience, cancer biology, and immunology, are reviewed in detail and serve as examples for applying MPFM to biological research.
Collapse
Affiliation(s)
- Chris Xu
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14850, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Alle 3B, 2200 Copenhagen, Denmark; University of Rochester Medical School, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Deborah J Fowell
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Peter Friedl
- Department of Medical BioSciences, Radboud University Medical Centre, Geert Grooteplein 26-28, Nijmegen HB 6500, the Netherlands
| | - Na Ji
- Department of Neuroscience, Department of Physics, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
19
|
Bromley-Coolidge S, Iruegas D, Appel B. Cspg4 sculpts oligodendrocyte precursor cell morphology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607226. [PMID: 39149260 PMCID: PMC11326215 DOI: 10.1101/2024.08.08.607226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The extracellular matrix (ECM) provides critical biochemical and structural cues that regulate neural development. Chondroitin sulfate proteoglycans (CSPGs), a major ECM component, have been implicated in modulating oligodendrocyte precursor cell (OPC) proliferation, migration, and maturation, but their specific roles in oligodendrocyte lineage cell (OLC) development and myelination in vivo remain poorly understood. Here, we use zebrafish as a model system to investigate the spatiotemporal dynamics of ECM deposition and CSPG localization during central nervous system (CNS) development, with a focus on their relationship to OLCs. We demonstrate that ECM components, including CSPGs, are dynamically expressed in distinct spatiotemporal patterns coinciding with OLC development and myelination. We found that zebrafish lacking cspg4 function produced normal numbers of OLCs, which appeared to undergo proper differentiation. However, OPC morphology in mutant larvae was aberrant. Nevertheless, the number and length of myelin sheaths produced by mature oligodendrocytes were unaffected. These data indicate that Cspg4 regulates OPC morphogenesis in vivo, supporting the role of the ECM in neural development.
Collapse
Affiliation(s)
- Samantha Bromley-Coolidge
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA, 80445
| | - Diego Iruegas
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA, 80445
| | - Bruce Appel
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA, 80445
| |
Collapse
|
20
|
Glial plasticity dysregulation contributes to learning impairments in the neurogenetic disorder NF1. Nat Neurosci 2024; 27:1447-1448. [PMID: 38831040 DOI: 10.1038/s41593-024-01667-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
|
21
|
Pan Y, Hysinger JD, Yalçın B, Lennon JJ, Byun YG, Raghavan P, Schindler NF, Anastasaki C, Chatterjee J, Ni L, Xu H, Malacon K, Jahan SM, Ivec AE, Aghoghovwia BE, Mount CW, Nagaraja S, Scheaffer S, Attardi LD, Gutmann DH, Monje M. Nf1 mutation disrupts activity-dependent oligodendroglial plasticity and motor learning in mice. Nat Neurosci 2024; 27:1555-1564. [PMID: 38816530 PMCID: PMC11303248 DOI: 10.1038/s41593-024-01654-y] [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: 09/22/2022] [Accepted: 04/18/2024] [Indexed: 06/01/2024]
Abstract
Neurogenetic disorders, such as neurofibromatosis type 1 (NF1), can cause cognitive and motor impairments, traditionally attributed to intrinsic neuronal defects such as disruption of synaptic function. Activity-regulated oligodendroglial plasticity also contributes to cognitive and motor functions by tuning neural circuit dynamics. However, the relevance of oligodendroglial plasticity to neurological dysfunction in NF1 is unclear. Here we explore the contribution of oligodendrocyte progenitor cells (OPCs) to pathological features of the NF1 syndrome in mice. Both male and female littermates (4-24 weeks of age) were used equally in this study. We demonstrate that mice with global or OPC-specific Nf1 heterozygosity exhibit defects in activity-dependent oligodendrogenesis and harbor focal OPC hyperdensities with disrupted homeostatic OPC territorial boundaries. These OPC hyperdensities develop in a cell-intrinsic Nf1 mutation-specific manner due to differential PI3K/AKT activation. OPC-specific Nf1 loss impairs oligodendroglial differentiation and abrogates the normal oligodendroglial response to neuronal activity, leading to impaired motor learning performance. Collectively, these findings show that Nf1 mutation delays oligodendroglial development and disrupts activity-dependent OPC function essential for normal motor learning in mice.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jared D Hysinger
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Belgin Yalçın
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - James J Lennon
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Youkyeong Gloria Byun
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Preethi Raghavan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Nicole F Schindler
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lijun Ni
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Haojun Xu
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Karen Malacon
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Samin M Jahan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Alexis E Ivec
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Benjamin E Aghoghovwia
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher W Mount
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Surya Nagaraja
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Suzanne Scheaffer
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Laura D Attardi
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
22
|
Zveik O, Rechtman A, Ganz T, Vaknin-Dembinsky A. The interplay of inflammation and remyelination: rethinking MS treatment with a focus on oligodendrocyte progenitor cells. Mol Neurodegener 2024; 19:53. [PMID: 38997755 PMCID: PMC11245841 DOI: 10.1186/s13024-024-00742-8] [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: 05/29/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) therapeutic goals have traditionally been dichotomized into two distinct avenues: immune-modulatory-centric interventions and pro-regenerative strategies. Oligodendrocyte progenitor cells (OPCs) were regarded for many years solely in concern to their potential to generate oligodendrocytes and myelin in the central nervous system (CNS). However, accumulating data elucidate the multifaceted roles of OPCs, including their immunomodulatory functions, positioning them as cardinal constituents of the CNS's immune landscape. MAIN BODY In this review, we will discuss how the two therapeutic approaches converge. We present a model by which (1) an inflammation is required for the appropriate pro-myelinating immune function of OPCs in the chronically inflamed CNS, and (2) the immune function of OPCs is crucial for their ability to differentiate and promote remyelination. This model highlights the reciprocal interactions between OPCs' pro-myelinating and immune-modulating functions. Additionally, we review the specific effects of anti- and pro-inflammatory interventions on OPCs, suggesting that immunosuppression adversely affects OPCs' differentiation and immune functions. CONCLUSION We suggest a multi-systemic therapeutic approach, which necessitates not a unidimensional focus but a harmonious balance between OPCs' pro-myelinating and immune-modulatory functions.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel.
| |
Collapse
|
23
|
Czopka T. A new gatekeeper to control oligodendrogenesis. PLoS Biol 2024; 22:e3002691. [PMID: 38990827 PMCID: PMC11239056 DOI: 10.1371/journal.pbio.3002691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
The diversity of oligodendrocyte precursor cells (OPCs) is not well understood and is actively discussed in the field. A new study in PLOS Biology describes a novel marker for an OPC subpopulation that controls oligodendrogenesis and myelination.
Collapse
Affiliation(s)
- Tim Czopka
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
24
|
Moghimyfiroozabad S, Paul MA, Bellenger L, Selimi F. A molecularly defined subpopulation of oligodendrocyte precursor cells controls the generation of myelinating oligodendrocytes during postnatal development. PLoS Biol 2024; 22:e3002655. [PMID: 38985832 PMCID: PMC11236193 DOI: 10.1371/journal.pbio.3002655] [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: 08/08/2023] [Accepted: 05/02/2024] [Indexed: 07/12/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are a class of glial cells that uniformly tiles the entire central nervous system (CNS). They play several key functions across the brain including the generation of oligodendrocytes and the control of myelination. Whether the functional diversity of OPCs is the result of genetically defined subpopulations or of their regulation by external factors has not been definitely established. We discovered that a subpopulation of OPCs found across the brain is defined by the expression of C1ql1, a gene previously described for its synaptic function in neurons. This subpopulation starts to appear during the first postnatal week in the mouse cortex. Ablation of C1ql1-expressing OPCs in the mouse leads to a massive lack of oligodendrocytes and myelination in many brain regions. This deficit cannot be rescued, even though some OPCs escape Sox10-driven ablation and end up partially compensating the OPC loss in the adult. Therefore, C1ql1 is a molecular marker of a functionally non-redundant subpopulation of OPCs, which controls the generation of myelinating oligodendrocytes.
Collapse
Affiliation(s)
- Shayan Moghimyfiroozabad
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Maela A Paul
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Lea Bellenger
- ARTbio Bioinformatics Analysis Facility, Sorbonne Université, Inserm U1156, CNRS FR 3631, Institut Français de Bioinformatique (IFB), Paris, France
| | - Fekrije Selimi
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| |
Collapse
|
25
|
Marangon D, Castro e Silva JH, Cerrato V, Boda E, Lecca D. Oligodendrocyte Progenitors in Glial Scar: A Bet on Remyelination. Cells 2024; 13:1024. [PMID: 38920654 PMCID: PMC11202012 DOI: 10.3390/cells13121024] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) represent a subtype of glia, giving rise to oligodendrocytes, the myelin-forming cells in the central nervous system (CNS). While OPCs are highly proliferative during development, they become relatively quiescent during adulthood, when their fate is strictly influenced by the extracellular context. In traumatic injuries and chronic neurodegenerative conditions, including those of autoimmune origin, oligodendrocytes undergo apoptosis, and demyelination starts. Adult OPCs become immediately activated; they migrate at the lesion site and proliferate to replenish the damaged area, but their efficiency is hampered by the presence of a glial scar-a barrier mainly formed by reactive astrocytes, microglia and the deposition of inhibitory extracellular matrix components. If, on the one hand, a glial scar limits the lesion spreading, it also blocks tissue regeneration. Therapeutic strategies aimed at reducing astrocyte or microglia activation and shifting them toward a neuroprotective phenotype have been proposed, whereas the role of OPCs has been largely overlooked. In this review, we have considered the glial scar from the perspective of OPCs, analysing their behaviour when lesions originate and exploring the potential therapies aimed at sustaining OPCs to efficiently differentiate and promote remyelination.
Collapse
Affiliation(s)
- Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| | - Juliana Helena Castro e Silva
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| | - Valentina Cerrato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; (V.C.); (E.B.)
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; (V.C.); (E.B.)
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| |
Collapse
|
26
|
Emery B, Wood TL. Regulators of Oligodendrocyte Differentiation. Cold Spring Harb Perspect Biol 2024; 16:a041358. [PMID: 38503504 PMCID: PMC11146316 DOI: 10.1101/cshperspect.a041358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Myelination has evolved as a mechanism to ensure fast and efficient propagation of nerve impulses along axons. Within the central nervous system (CNS), myelination is carried out by highly specialized glial cells, oligodendrocytes. The formation of myelin is a prolonged aspect of CNS development that occurs well into adulthood in humans, continuing throughout life in response to injury or as a component of neuroplasticity. The timing of myelination is tightly tied to the generation of oligodendrocytes through the differentiation of their committed progenitors, oligodendrocyte precursor cells (OPCs), which reside throughout the developing and adult CNS. In this article, we summarize our current understanding of some of the signals and pathways that regulate the differentiation of OPCs, and thus the myelination of CNS axons.
Collapse
Affiliation(s)
- Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, USA
| |
Collapse
|
27
|
Piscopo VEC, Chapleau A, Blaszczyk GJ, Sirois J, You Z, Soubannier V, Chen CXQ, Bernard G, Antel JP, Durcan TM. The use of a SOX10 reporter toward ameliorating oligodendrocyte lineage differentiation from human induced pluripotent stem cells. Glia 2024; 72:1165-1182. [PMID: 38497409 DOI: 10.1002/glia.24524] [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: 12/06/2023] [Revised: 02/01/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
Oligodendrocytes (OLs) are key players in the central nervous system, critical for the formation and maintenance of the myelin sheaths insulating axons, ensuring efficient neuronal communication. In the last decade, the use of human induced pluripotent stem cells (iPSCs) has become essential for recapitulating and understanding the differentiation and role of OLs in vitro. Current methods include overexpression of transcription factors for rapid OL generation, neglecting the complexity of OL lineage development. Alternatively, growth factor-based protocols offer physiological relevance but struggle with efficiency and cell heterogeneity. To address these issues, we created a novel SOX10-P2A-mOrange iPSC reporter line to track and purify oligodendrocyte precursor cells. Using this reporter cell line, we analyzed an existing differentiation protocol and shed light on the origin of glial cell heterogeneity. Additionally, we have modified the differentiation protocol, toward enhancing reproducibility, efficiency, and terminal maturity. Our approach not only advances OL biology but also holds promise to accelerate research and translational work with iPSC-derived OLs.
Collapse
Affiliation(s)
- Valerio E C Piscopo
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Alexandra Chapleau
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Gabriela J Blaszczyk
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Julien Sirois
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Zhipeng You
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Vincent Soubannier
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Carol X-Q Chen
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Geneviève Bernard
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
- Department of Pediatrics and Human Genetics, McGill University, Montreal, Quebec, Canada
- Division of Medical Genetics, Department of Internal Medicine, McGill University Health Center, Montreal, Quebec, Canada
| | - Jack P Antel
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
28
|
Thornton MA, Futia GL, Stockton ME, Budoff SA, Ramirez AN, Ozbay B, Tzang O, Kilborn K, Poleg-Polsky A, Restrepo D, Gibson EA, Hughes EG. Long-term in vivo three-photon imaging reveals region-specific differences in healthy and regenerative oligodendrogenesis. Nat Neurosci 2024; 27:846-861. [PMID: 38539013 PMCID: PMC11104262 DOI: 10.1038/s41593-024-01613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/26/2024] [Indexed: 04/09/2024]
Abstract
The generation of new myelin-forming oligodendrocytes in the adult central nervous system is critical for cognitive function and regeneration following injury. Oligodendrogenesis varies between gray and white matter regions, suggesting that local cues drive regional differences in myelination and the capacity for regeneration. However, the layer- and region-specific regulation of oligodendrocyte populations is unclear due to the inability to monitor deep brain structures in vivo. Here we harnessed the superior imaging depth of three-photon microscopy to permit long-term, longitudinal in vivo three-photon imaging of the entire cortical column and subcortical white matter in adult mice. We find that cortical oligodendrocyte populations expand at a higher rate in the adult brain than those of the white matter. Following demyelination, oligodendrocyte replacement is enhanced in the white matter, while the deep cortical layers show deficits in regenerative oligodendrogenesis and the restoration of transcriptional heterogeneity. Together, our findings demonstrate that regional microenvironments regulate oligodendrocyte population dynamics and heterogeneity in the healthy and diseased brain.
Collapse
Affiliation(s)
- Michael A Thornton
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Gregory L Futia
- Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael E Stockton
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Samuel A Budoff
- Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexandra N Ramirez
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Baris Ozbay
- Intelligent Imaging Innovations, Denver, CO, USA
| | - Omer Tzang
- Intelligent Imaging Innovations, Denver, CO, USA
| | - Karl Kilborn
- Intelligent Imaging Innovations, Denver, CO, USA
| | - Alon Poleg-Polsky
- Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Diego Restrepo
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emily A Gibson
- Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ethan G Hughes
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
29
|
Ozkan A, Padmanabhan HK, Shipman SL, Azim E, Kumar P, Sadegh C, Basak AN, Macklis JD. Directed differentiation of functional corticospinal-like neurons from endogenous SOX6+/NG2+ cortical progenitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.21.590488. [PMID: 38712174 PMCID: PMC11071355 DOI: 10.1101/2024.04.21.590488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Corticospinal neurons (CSN) centrally degenerate in amyotrophic lateral sclerosis (ALS), along with spinal motor neurons, and loss of voluntary motor function in spinal cord injury (SCI) results from damage to CSN axons. For functional regeneration of specifically affected neuronal circuitry in vivo , or for optimally informative disease modeling and/or therapeutic screening in vitro , it is important to reproduce the type or subtype of neurons involved. No such appropriate in vitro models exist with which to investigate CSN selective vulnerability and degeneration in ALS, or to investigate routes to regeneration of CSN circuitry for ALS or SCI, critically limiting the relevance of much research. Here, we identify that the HMG-domain transcription factor Sox6 is expressed by a subset of NG2+ endogenous cortical progenitors in postnatal and adult cortex, and that Sox6 suppresses a latent neurogenic program by repressing inappropriate proneural Neurog2 expression by progenitors. We FACS-purify these genetically accessible progenitors from postnatal mouse cortex and establish a pure culture system to investigate their potential for directed differentiation into CSN. We then employ a multi-component construct with complementary and differentiation-sharpening transcriptional controls (activating Neurog2, Fezf2 , while antagonizing Olig2 with VP16:Olig2 ). We generate corticospinal-like neurons from SOX6+/NG2+ cortical progenitors, and find that these neurons differentiate with remarkable fidelity compared with corticospinal neurons in vivo . They possess appropriate morphological, molecular, transcriptomic, and electrophysiological characteristics, without characteristics of the alternate intracortical or other neuronal subtypes. We identify that these critical specifics of differentiation are not reproduced by commonly employed Neurog2 -driven differentiation. Neurons induced by Neurog2 instead exhibit aberrant multi-axon morphology and express molecular hallmarks of alternate cortical projection subtypes, often in mixed form. Together, this developmentally-based directed differentiation from genetically accessible cortical progenitors sets a precedent and foundation for in vitro mechanistic and therapeutic disease modeling, and toward regenerative neuronal repopulation and circuit repair.
Collapse
|
30
|
Hupp MT, Santos EN, Heo D, Call CL, Guttenplan KA. Vesicle Fusion in Oligodendrocyte Maturation and Myelination. J Neurosci 2024; 44:e2203232024. [PMID: 38631918 PMCID: PMC11026360 DOI: 10.1523/jneurosci.2203-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Affiliation(s)
- Madison T Hupp
- Vollum Institute, Oregon Health and Science University, Portland, Oregon
| | - Erin N Santos
- Vollum Institute, Oregon Health and Science University, Portland, Oregon
| | - Dongeun Heo
- Vollum Institute, Oregon Health and Science University, Portland, Oregon
| | - Cody L Call
- Vollum Institute, Oregon Health and Science University, Portland, Oregon
| | - Kevin A Guttenplan
- Vollum Institute, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
31
|
Cheng YJ, Wang F, Feng J, Yu B, Wang B, Gao Q, Wang TY, Hu B, Gao X, Chen JF, Chen YJ, Lv SQ, Feng H, Xiao L, Mei F. Prolonged myelin deficits contribute to neuron loss and functional impairments after ischaemic stroke. Brain 2024; 147:1294-1311. [PMID: 38289861 DOI: 10.1093/brain/awae029] [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: 08/06/2023] [Revised: 12/29/2023] [Accepted: 01/13/2024] [Indexed: 02/01/2024] Open
Abstract
Ischaemic stroke causes neuron loss and long-term functional deficits. Unfortunately, effective approaches to preserving neurons and promoting functional recovery remain unavailable. Oligodendrocytes, the myelinating cells in the CNS, are susceptible to oxygen and nutrition deprivation and undergo degeneration after ischaemic stroke. Technically, new oligodendrocytes and myelin can be generated by the differentiation of oligodendrocyte precursor cells (OPCs). However, myelin dynamics and their functional significance after ischaemic stroke remain poorly understood. Here, we report numerous denuded axons accompanied by decreased neuron density in sections from ischaemic stroke lesions in human brain, suggesting that neuron loss correlates with myelin deficits in these lesions. To investigate the longitudinal changes in myelin dynamics after stroke, we labelled and traced pre-existing and newly-formed myelin, respectively, using cell-specific genetic approaches. Our results indicated massive oligodendrocyte death and myelin loss 2 weeks after stroke in the transient middle cerebral artery occlusion (tMCAO) mouse model. In contrast, myelin regeneration remained insufficient 4 and 8 weeks post-stroke. Notably, neuronal loss and functional impairments worsened in aged brains, and new myelin generation was diminished. To analyse the causal relationship between remyelination and neuron survival, we manipulated myelinogenesis by conditional deletion of Olig2 (a positive regulator) or muscarinic receptor 1 (M1R, a negative regulator) in OPCs. Deleting Olig2 inhibited remyelination, reducing neuron survival and functional recovery after tMCAO. Conversely, enhancing remyelination by M1R conditional knockout or treatment with the pro-myelination drug clemastine after tMCAO preserved white matter integrity and neuronal survival, accelerating functional recovery. Together, our findings demonstrate that enhancing myelinogenesis is a promising strategy to preserve neurons and promote functional recovery after ischaemic stroke.
Collapse
Affiliation(s)
- Yong-Jie Cheng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, 1st affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fei Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jie Feng
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Yu
- Department of Neurosurgery, 2nd affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Wang
- Department of Physiology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Qing Gao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Mathematical Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Teng-Yue Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Mathematical Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Bo Hu
- Department of Physiology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Xing Gao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing-Fei Chen
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu-Jie Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, 1st affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, 2nd affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, 1st affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Xiao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Department of Neurosurgery, 2nd affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Feng Mei
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
32
|
Koupourtidou C, Schwarz V, Aliee H, Frerich S, Fischer-Sternjak J, Bocchi R, Simon-Ebert T, Bai X, Sirko S, Kirchhoff F, Dichgans M, Götz M, Theis FJ, Ninkovic J. Shared inflammatory glial cell signature after stab wound injury, revealed by spatial, temporal, and cell-type-specific profiling of the murine cerebral cortex. Nat Commun 2024; 15:2866. [PMID: 38570482 PMCID: PMC10991294 DOI: 10.1038/s41467-024-46625-w] [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: 03/03/2023] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Traumatic brain injury leads to a highly orchestrated immune- and glial cell response partially responsible for long-lasting disability and the development of secondary neurodegenerative diseases. A holistic understanding of the mechanisms controlling the responses of specific cell types and their crosstalk is required to develop an efficient strategy for better regeneration. Here, we combine spatial and single-cell transcriptomics to chart the transcriptomic signature of the injured male murine cerebral cortex, and identify specific states of different glial cells contributing to this signature. Interestingly, distinct glial cells share a large fraction of injury-regulated genes, including inflammatory programs downstream of the innate immune-associated pathways Cxcr3 and Tlr1/2. Systemic manipulation of these pathways decreases the reactivity state of glial cells associated with poor regeneration. The functional relevance of the discovered shared signature of glial cells highlights the importance of our resource enabling comprehensive analysis of early events after brain injury.
Collapse
Affiliation(s)
- Christina Koupourtidou
- Chair of Cell Biology and Anatomy, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Veronika Schwarz
- Chair of Cell Biology and Anatomy, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Hananeh Aliee
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Simon Frerich
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Judith Fischer-Sternjak
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Riccardo Bocchi
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Tatiana Simon-Ebert
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany
| | - Swetlana Sirko
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology SYNERGY, LMU Munich, Munich, Germany
- German Centre for Neurodegenerative Diseases, Munich, Germany
| | - Magdalena Götz
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Munich Cluster for Systems Neurology SYNERGY, LMU Munich, Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, Munich, Germany
| | - Jovica Ninkovic
- Chair of Cell Biology and Anatomy, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany.
- Munich Cluster for Systems Neurology SYNERGY, LMU Munich, Munich, Germany.
| |
Collapse
|
33
|
Huang HT, Wang CY, Ho CH, Tzeng SF. Interleukin-6 Inhibits Expression of miR-204-5p, a Regulator of Oligodendrocyte Differentiation: Involvement of miR-204-5p in the Prevention of Chemical-Induced Oligodendrocyte Impairment. Mol Neurobiol 2024; 61:1953-1968. [PMID: 37817030 DOI: 10.1007/s12035-023-03681-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023]
Abstract
Oligodendrocytes (OLs) form myelin sheaths around axons in the central nervous system (CNS) facilitate the propagation of action potentials. The studies have shown that the differentiation and maturation of OLs involve microRNA (miR) regulation. The recent findings have addressed that miR-204 regulates OL differentiation in culture. In this study, through in situ hybridization in combination with immunohistochemistry, we showed that microRNA-204-5p in the corpus callosum was mainly expressed in OLs immunoreactive with adenomatous polyposis coli (APC), an OL marker. We also found miR-204-5p expression in mature OLs was suppressed by the addition of interleukin-6 (IL-6). Moreover, IL-6-induced inhibition of miR-204-5p expression was blocked by the addition of the inhibitors specific for p38 mitogen-activated protein kinase (p38MAPK) or phosphatidylinositol 3-kinase (PI3K) pathway. We further utilized a rat model by feeding cuprizone (CPZ)-containing diet for 3 weeks to induce demyelination and gliosis in the corpus callosum, as well as the upregulation of IL-6 gene expression significantly. Despite that miR-204-5p expression in the corpus callosum was not altered after feeding by CPZ for 3 weeks, its expression was increased and IL-6 transcription was decreased in the corpus callosum of the recovery group that was fed by CPZ for the first 2 weeks and by the regular diet for one more week. Our data demonstrate that miR-204-5p expression in OLs declined under the influence of the inflamed microenvironment. The findings that an increase in miR-204-5p and declined IL-6 expression observed in the recovery group might be involved with OL repair in the corpus callosum, and also shed light on a potential role for miR-204-5p in OL homeostasis following the white matter injury.
Collapse
Affiliation(s)
- Hui-Ting Huang
- Department of Life Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Yen Wang
- Department of Life Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, Taiwan
- Department of Biotechnology and Bioindustry Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Hsin Ho
- Department of Life Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, College of Biosciences and Biotechnology, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
34
|
Zveik O, Rechtman A, Brill L, Vaknin-Dembinsky A. Anti- and pro-inflammatory milieu differentially regulate differentiation and immune functions of oligodendrocyte progenitor cells. Immunology 2024; 171:618-633. [PMID: 38243672 DOI: 10.1111/imm.13757] [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: 10/23/2023] [Accepted: 01/05/2024] [Indexed: 01/21/2024] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) were regarded for years solely for their regenerative role; however, their immune-modulatory roles have gained much attention recently, particularly in the context of multiple sclerosis (MS). Despite extensive studies on OPCs, there are limited data elucidating the interactions between their intrinsic regenerative and immune functions, as well as their relationship with the inflamed central nervous system (CNS) environment, a key factor in MS pathology. We examined the effects of pro-inflammatory cytokines, represented by interferon (IFN)-γ and tumour necrosis factor (TNF)-α, as well as anti-inflammatory cytokines, represented by interleukin (IL)-4 and IL-10, on OPC differentiation and immune characteristics. Using primary cultures, enzyme-linked immunosorbent assay and immunofluorescence stainings, we assessed differentiation capacity, phagocytic activity, major histocompatibility complex (MHC)-II expression, and cytokine secretion. We observed that the anti-inflammatory milieu (IL4 and IL10) reduced both OPC differentiation and immune functions. Conversely, exposure to TNF-α led to intact differentiation, increased phagocytic activity, high levels of MHC-II expression, and cytokines secretion. Those effects were attributed to signalling via TNF-receptor-2 and counteracted the detrimental effects of IFN-γ on OPC differentiation. Our findings suggest that a pro-regenerative, permissive inflammatory environment is needed for OPCs to execute both regenerative and immune-modulatory functions.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Livnat Brill
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
35
|
Rokach M, Portioli C, Brahmachari S, Estevão BM, Decuzzi P, Barak B. Tackling myelin deficits in neurodevelopmental disorders using drug delivery systems. Adv Drug Deliv Rev 2024; 207:115218. [PMID: 38403255 DOI: 10.1016/j.addr.2024.115218] [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: 11/14/2023] [Revised: 01/27/2024] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
Interest in myelin and its roles in almost all brain functions has been greatly increasing in recent years, leading to countless new studies on myelination, as a dominant process in the development of cognitive functions. Here, we explore the unique role myelin plays in the central nervous system and specifically discuss the results of altered myelination in neurodevelopmental disorders. We present parallel developmental trajectories involving myelination that correlate with the onset of cognitive impairment in neurodevelopmental disorders and discuss the key challenges in the treatment of these chronic disorders. Recent developments in drug repurposing and nano/micro particle-based therapies are reviewed as a possible pathway to circumvent some of the main hurdles associated with early intervention, including patient's adherence and compliance, side effects, relapse, and faster route to possible treatment of these disorders. The strategy of drug encapsulation overcomes drug solubility and metabolism, with the possibility of drug targeting to a specific compartment, reducing side effects upon systemic administration.
Collapse
Affiliation(s)
- May Rokach
- Sagol School of Neuroscience, Tel-Aviv University, Israel
| | - Corinne Portioli
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Sayanti Brahmachari
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Bianca Martins Estevão
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Boaz Barak
- Sagol School of Neuroscience, Tel-Aviv University, Israel; Faculty of Social Sciences, The School of Psychological Sciences, Tel-Aviv University, Israel.
| |
Collapse
|
36
|
Chapman TW, Kamen Y, Piedra ET, Hill RA. Oligodendrocyte Maturation Alters the Cell Death Mechanisms That Cause Demyelination. J Neurosci 2024; 44:e1794232024. [PMID: 38395617 PMCID: PMC10977033 DOI: 10.1523/jneurosci.1794-23.2024] [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: 09/21/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Myelinating oligodendrocytes die in human disease and early in aging. Despite this, the mechanisms that underly oligodendrocyte death are not resolved and it is also not clear whether these mechanisms change as oligodendrocyte lineage cells are undergoing differentiation and maturation. Here, we used a combination of intravital imaging, single-cell ablation, and cuprizone-mediated demyelination, in both female and male mice, to discover that oligodendrocyte maturation dictates the dynamics and mechanisms of cell death. After single-cell phototoxic damage, oligodendrocyte precursor cells underwent programmed cell death within hours, differentiating oligodendrocytes died over several days, while mature oligodendrocytes took weeks to die. Importantly cells at each maturation stage all eventually died but did so with drastically different temporal dynamics and morphological features. Consistent with this, cuprizone treatment initiated a caspase-3-dependent form of rapid cell death in differentiating oligodendrocytes, while mature oligodendrocytes never activated this executioner caspase. Instead, mature oligodendrocytes exhibited delayed cell death which was marked by DNA damage and disruption in poly-ADP-ribose subcellular localization. Thus, oligodendrocyte maturation plays a key role in determining the mechanism of death a cell undergoes in response to the same insult. This means that oligodendrocyte maturation is important to consider when designing strategies for preventing cell death and preserving myelin while also enhancing the survival of new oligodendrocytes in demyelinating conditions.
Collapse
Affiliation(s)
- Timothy W Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Yasmine Kamen
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Enrique T Piedra
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| |
Collapse
|
37
|
Rogujski P, Lukomska B, Janowski M, Stanaszek L. Glial-restricted progenitor cells: a cure for diseased brain? Biol Res 2024; 57:8. [PMID: 38475854 DOI: 10.1186/s40659-024-00486-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The central nervous system (CNS) is home to neuronal and glial cells. Traditionally, glia was disregarded as just the structural support across the brain and spinal cord, in striking contrast to neurons, always considered critical players in CNS functioning. In modern times this outdated dogma is continuously repelled by new evidence unravelling the importance of glia in neuronal maintenance and function. Therefore, glia replacement has been considered a potentially powerful therapeutic strategy. Glial progenitors are at the center of this hope, as they are the source of new glial cells. Indeed, sophisticated experimental therapies and exciting clinical trials shed light on the utility of exogenous glia in disease treatment. Therefore, this review article will elaborate on glial-restricted progenitor cells (GRPs), their origin and characteristics, available sources, and adaptation to current therapeutic approaches aimed at various CNS diseases, with particular attention paid to myelin-related disorders with a focus on recent progress and emerging concepts. The landscape of GRP clinical applications is also comprehensively presented, and future perspectives on promising, GRP-based therapeutic strategies for brain and spinal cord diseases are described in detail.
Collapse
Affiliation(s)
- Piotr Rogujski
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, USA
| | - Luiza Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| |
Collapse
|
38
|
Hill RA, Nishiyama A, Hughes EG. Features, Fates, and Functions of Oligodendrocyte Precursor Cells. Cold Spring Harb Perspect Biol 2024; 16:a041425. [PMID: 38052500 PMCID: PMC10910408 DOI: 10.1101/cshperspect.a041425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) are a central nervous system resident population of glia with a distinct molecular identity and an ever-increasing list of functions. OPCs generate oligodendrocytes throughout development and across the life span in most regions of the brain and spinal cord. This process involves a complex coordination of molecular checkpoints and biophysical cues from the environment that initiate the differentiation and integration of new oligodendrocytes that synthesize myelin sheaths on axons. Outside of their progenitor role, OPCs have been proposed to play other functions including the modulation of axonal and synaptic development and the participation in bidirectional signaling with neurons and other glia. Here, we review OPC identity and known functions and discuss recent findings implying other roles for these glial cells in brain physiology and pathology.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Ethan G Hughes
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
39
|
Perdaens O, Bottemanne P, van Pesch V. MicroRNAs dysregulated in multiple sclerosis affect the differentiation of CG-4 cells, an oligodendrocyte progenitor cell line. Front Cell Neurosci 2024; 18:1336439. [PMID: 38486710 PMCID: PMC10937391 DOI: 10.3389/fncel.2024.1336439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/24/2024] [Indexed: 03/17/2024] Open
Abstract
Introduction Demyelination is one of the hallmarks of multiple sclerosis (MS). While remyelination occurs during the disease, it is incomplete from the start and strongly decreases with its progression, mainly due to the harm to oligodendrocyte progenitor cells (OPCs), causing irreversible neurological deficits and contributing to neurodegeneration. Therapeutic strategies promoting remyelination are still very preliminary and lacking within the current treatment panel for MS. Methods In a previous study, we identified 21 microRNAs dysregulated mostly in the CSF of relapsing and/or remitting MS patients. In this study we transfected the mimics/inhibitors of several of these microRNAs separately in an OPC cell line, called CG-4. We aimed (1) to phenotypically characterize their effect on OPC differentiation and (2) to identify corroborating potential mRNA targets via immunocytochemistry, RT-qPCR analysis, RNA sequencing, and Gene Ontology enrichment analysis. Results We observed that the majority of 13 transfected microRNA mimics decreased the differentiation of CG-4 cells. We demonstrate, by RNA sequencing and independent RT-qPCR analyses, that miR-33-3p, miR-34c-5p, and miR-124-5p arrest OPC differentiation at a late progenitor stage and miR-145-5p at a premyelinating stage as evidenced by the downregulation of premyelinating oligodendrocyte (OL) [Tcf7l2, Cnp (except for miR-145-5p)] and mature OL (Plp1, Mbp, and Mobp) markers, whereas only miR-214-3p promotes OPC differentiation. We further propose a comprehensive exploration of their change in cell fate through Gene Ontology enrichment analysis. We finally confirm by RT-qPCR analyses the downregulation of several predicted mRNA targets for each microRNA that possibly support their effect on OPC differentiation by very distinctive mechanisms, of which some are still unexplored in OPC/OL physiology. Conclusion miR-33-3p, miR-34c-5p, and miR-124-5p arrest OPC differentiation at a late progenitor stage and miR-145-5p at a premyelinating stage, whereas miR-214-3p promotes the differentiation of CG-4 cells. We propose several potential mRNA targets and hypothetical mechanisms by which each microRNA exerts its effect. We hereby open new perspectives in the research on OPC differentiation and the pathophysiology of demyelination/remyelination, and possibly even in the search for new remyelinating therapeutic strategies in the scope of MS.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pauline Bottemanne
- Bioanalysis and Pharmacology of Bioactive Lipids, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Neurology, Cliniques universitaires Saint-Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
40
|
Aranda S, Muntané G, Vilella E. Coexpression network analysis of the adult brain sheds light on the pathogenic mechanism of DDR1 in schizophrenia and bipolar disorder. Transl Psychiatry 2024; 14:112. [PMID: 38395959 PMCID: PMC10891045 DOI: 10.1038/s41398-024-02823-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
DDR1 has been linked to schizophrenia (SCZ) and bipolar disorder (BD) in association studies. DDR1 encodes 58 distinct transcripts, which can be translated into five isoforms (DDR1a-e) and are expressed in the brain. However, the transcripts expressed in each brain cell type, their functions and their involvement in SCZ and BD remain unknown. Here, to infer the processes in which DDR1 transcripts are involved, we used transcriptomic data from the human brain dorsolateral prefrontal cortex of healthy controls (N = 936) and performed weighted gene coexpression network analysis followed by enrichment analyses. Then, to explore the involvement of DDR1 transcripts in SCZ (N = 563) and BD (N = 222), we studied the association of coexpression modules with disease and performed differential expression and transcript significance analyses. Some DDR1 transcripts were distributed across five coexpression modules identified in healthy controls (MHC). MHC1 and MHC2 were enriched in the cell cycle and proliferation of astrocytes and OPCs; MHC3 and MHC4 were enriched in oligodendrocyte differentiation and myelination; and MHC5 was enriched in neurons and synaptic transmission. Most of the DDR1 transcripts associated with SCZ and BD pertained to MHC1 and MHC2. Altogether, our results suggest that DDR1 expression might be altered in SCZ and BD via the proliferation of astrocytes and OPCs, suggesting that these processes are relevant in psychiatric disorders.
Collapse
Affiliation(s)
- Selena Aranda
- Institut d'Investigació Sanitària Pere Virgili-CERCA, Reus, Spain
- Hospital Universitari Institut Pere Mata, Reus, Spain
- Universitat Rovira i Virgili, Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Gerard Muntané
- Institut d'Investigació Sanitària Pere Virgili-CERCA, Reus, Spain
- Hospital Universitari Institut Pere Mata, Reus, Spain
- Universitat Rovira i Virgili, Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM)-Instituto de Salud Carlos III, Madrid, Spain
- Institut de Biologia Evolutiva (UPF-CSIC), Departament de Medicina i Ciències de la Vida (MELIS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Elisabet Vilella
- Institut d'Investigació Sanitària Pere Virgili-CERCA, Reus, Spain.
- Hospital Universitari Institut Pere Mata, Reus, Spain.
- Universitat Rovira i Virgili, Reus, Spain.
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM)-Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
41
|
Huang B, Chen Z, Huang F, Gao F, Chen J, Liu P, Lu Z, Chen W, Wu J. Demyelination in the medial prefrontal cortex by withdrawal from chronic nicotine causes impaired cognitive memory. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110901. [PMID: 38036034 DOI: 10.1016/j.pnpbp.2023.110901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Epidemiological studies revealed deficits in cognitive learning and memory in smokers who withdrawal from smoking, but the molecular mechanisms underlying it is unclear. Here, we employed the novel object recognition task (NORT) to evaluate cognitive memory and found impaired memory and motor skills after withdrawal from chronic nicotine. Myelin sheath hastens the conduction of signals along axons and thus plays a critical role in learning and memory. We found no effect of nicotine withdrawal on the myelination in both of the Ventral tegmental area (VTA) and Nucleus accumbens (NAc) regions, but unexpectedly, we observed a demyelination phenomenon in the medial prefrontal cortex (mPFC) after withdrawal from chronic nicotine. Moreover, we found a positive correlation between the impaired memory and demyelination, and pharmaceutical rescue of myelination by clemastine specifically improved the impaired recognition memory but not the decreased motor skills caused by withdrawal from chronic nicotine. We further found nicotine directly acts on oligodendrocytes with OPCs potential to decrease their myelination process. Taken together, these results demonstrate demyelination in the mPFC causes impaired recognition memory and reveal a potential of enhancing myelination as a therapeutic strategy to alleviate cognitive memory deficits caused by smoking withdrawal.
Collapse
Affiliation(s)
- Bing Huang
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Guangdong, China; Brain Function and Disease Laboratory, Shantou University Medical College, Guangdong, China; Department of Pharmacology, Shantou University Medical College, 22 Xinling Road, Shantou 515041, Guangdong Province, China.
| | - Zifei Chen
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Guangdong, China
| | - Fang Huang
- Joint Shantou International Eye Center, Shantou University and the Chinese University of Hong Kong, 515041 Shantou, Guangdong, China
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, 22 Xinling Road, Shantou 515041, Guangdong Province, China
| | - Jieling Chen
- Brain Function and Disease Laboratory, Shantou University Medical College, Guangdong, China
| | - Peipei Liu
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhijie Lu
- Brain Function and Disease Laboratory, Shantou University Medical College, Guangdong, China
| | - Weiyuan Chen
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Guangdong, China
| | - Jie Wu
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Guangdong, China; Brain Function and Disease Laboratory, Shantou University Medical College, Guangdong, China
| |
Collapse
|
42
|
Janeckova L, Knotek T, Kriska J, Hermanova Z, Kirdajova D, Kubovciak J, Berkova L, Tureckova J, Camacho Garcia S, Galuskova K, Kolar M, Anderova M, Korinek V. Astrocyte-like subpopulation of NG2 glia in the adult mouse cortex exhibits characteristics of neural progenitor cells. Glia 2024; 72:245-273. [PMID: 37772368 DOI: 10.1002/glia.24471] [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: 02/20/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/30/2023]
Abstract
Glial cells expressing neuron-glial antigen 2 (NG2), also known as oligodendrocyte progenitor cells (OPCs), play a critical role in maintaining brain health. However, their ability to differentiate after ischemic injury is poorly understood. The aim of this study was to investigate the properties and functions of NG2 glia in the ischemic brain. Using transgenic mice, we selectively labeled NG2-expressing cells and their progeny in both healthy brain and after focal cerebral ischemia (FCI). Using single-cell RNA sequencing, we classified the labeled glial cells into five distinct subpopulations based on their gene expression patterns. Additionally, we examined the membrane properties of these cells using the patch-clamp technique. Of the identified subpopulations, three were identified as OPCs, whereas the fourth subpopulation had characteristics indicative of cells likely to develop into oligodendrocytes. The fifth subpopulation of NG2 glia showed astrocytic markers and had similarities to neural progenitor cells. Interestingly, this subpopulation was present in both healthy and post-ischemic tissue; however, its gene expression profile changed after ischemia, with increased numbers of genes related to neurogenesis. Immunohistochemical analysis confirmed the temporal expression of neurogenic genes and showed an increased presence of NG2 cells positive for Purkinje cell protein-4 at the periphery of the ischemic lesion 12 days after FCI, as well as NeuN-positive NG2 cells 28 and 60 days after injury. These results suggest the potential development of neuron-like cells arising from NG2 glia in the ischemic tissue. Our study provides insights into the plasticity of NG2 glia and their capacity for neurogenesis after stroke.
Collapse
Affiliation(s)
- Lucie Janeckova
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomas Knotek
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Denisa Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Kubovciak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Linda Berkova
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Sara Camacho Garcia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Katerina Galuskova
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michal Kolar
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vladimir Korinek
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
43
|
Morrissey ZD, Gao J, Shetti A, Li W, Zhan L, Li W, Fortel I, Saido T, Saito T, Ajilore O, Cologna SM, Lazarov O, Leow AD. Temporal Alterations in White Matter in An App Knock-In Mouse Model of Alzheimer's Disease. eNeuro 2024; 11:ENEURO.0496-23.2024. [PMID: 38290851 PMCID: PMC10897532 DOI: 10.1523/eneuro.0496-23.2024] [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: 11/27/2023] [Revised: 01/05/2024] [Accepted: 01/17/2024] [Indexed: 02/01/2024] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and results in neurodegeneration and cognitive impairment. White matter (WM) is affected in AD and has implications for neural circuitry and cognitive function. The trajectory of these changes across age, however, is still not well understood, especially at earlier stages in life. To address this, we used the AppNL-G-F/NL-G-F knock-in (APPKI) mouse model that harbors a single copy knock-in of the human amyloid precursor protein (APP) gene with three familial AD mutations. We performed in vivo diffusion tensor imaging (DTI) to study how the structural properties of the brain change across age in the context of AD. In late age APPKI mice, we observed reduced fractional anisotropy (FA), a proxy of WM integrity, in multiple brain regions, including the hippocampus, anterior commissure (AC), neocortex, and hypothalamus. At the cellular level, we observed greater numbers of oligodendrocytes in middle age (prior to observations in DTI) in both the AC, a major interhemispheric WM tract, and the hippocampus, which is involved in memory and heavily affected in AD, prior to observations in DTI. Proteomics analysis of the hippocampus also revealed altered expression of oligodendrocyte-related proteins with age and in APPKI mice. Together, these results help to improve our understanding of the development of AD pathology with age, and imply that middle age may be an important temporal window for potential therapeutic intervention.
Collapse
Affiliation(s)
- Zachery D Morrissey
- Graduate Program in Neuroscience, University of Illinois Chicago, Chicago, Illinois 60612
- Department of Psychiatry, University of Illinois Chicago, Chicago, Illinois 60612
- Department of Anatomy & Cell Biology, University of Illinois Chicago, Chicago, Illinois 60612
| | - Jin Gao
- Department of Electrical & Computer Engineering, University of Illinois Chicago, Chicago, Illinois 60607
- Preclinical Imaging Core, University of Illinois Chicago, Chicago, Illinois 60612
| | - Aashutosh Shetti
- Department of Anatomy & Cell Biology, University of Illinois Chicago, Chicago, Illinois 60612
| | - Wenping Li
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607
| | - Liang Zhan
- Department of Electrical & Computer Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Weiguo Li
- Preclinical Imaging Core, University of Illinois Chicago, Chicago, Illinois 60612
- Department of Bioengineering, University of Illinois Chicago, Chicago, Illinois 60607
- Department of Radiology, Northwestern University, Chicago, Illinois 60611
| | - Igor Fortel
- Department of Bioengineering, University of Illinois Chicago, Chicago, Illinois 60607
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako 351-0198, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Nagoya 467-8601, Japan
| | - Olusola Ajilore
- Department of Psychiatry, University of Illinois Chicago, Chicago, Illinois 60612
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607
| | - Orly Lazarov
- Department of Anatomy & Cell Biology, University of Illinois Chicago, Chicago, Illinois 60612
| | - Alex D Leow
- Department of Psychiatry, University of Illinois Chicago, Chicago, Illinois 60612
- Department of Bioengineering, University of Illinois Chicago, Chicago, Illinois 60607
- Department of Computer Science, University of Illinois Chicago, Chicago, Illinois 60607
| |
Collapse
|
44
|
Haak A, Lesslich HM, Dietzel ID. Visualization of the membrane surface and cytoskeleton of oligodendrocyte progenitor cell growth cones using a combination of scanning ion conductance and four times expansion microscopy. Biol Chem 2024; 405:31-41. [PMID: 37950644 DOI: 10.1515/hsz-2023-0217] [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: 05/16/2023] [Accepted: 10/17/2023] [Indexed: 11/13/2023]
Abstract
Growth cones of oligodendrocyte progenitor cells (OPCs) are challenging to investigate with conventional light microscopy due to their small size. Especially substructures such as filopodia, lamellipodia and their underlying cytoskeleton are difficult to resolve with diffraction limited microscopy. Light microscopy techniques, which surpass the diffraction limit such as stimulated emission depletion microscopy, often require expensive setups and specially trained personnel rendering them inaccessible to smaller research groups. Lately, the invention of expansion microscopy (ExM) has enabled super-resolution imaging with any light microscope without the need for additional equipment. Apart from the necessary resolution, investigating OPC growth cones comes with another challenge: Imaging the topography of membranes, especially label- and contact-free, is only possible with very few microscopy techniques one of them being scanning ion conductance microscopy (SICM). We here present a new imaging workflow combining SICM and ExM, which enables the visualization of OPC growth cone nanostructures. We correlated SICM recordings and ExM images of OPC growth cones captured with a conventional widefield microscope. This enabled the visualization of the growth cones' membrane topography as well as their underlying actin and tubulin cytoskeleton.
Collapse
Affiliation(s)
- Annika Haak
- Nanoscopy, RUBION, Ruhr-Universität Bochum, Universitätsstraße 150, D-44801 Bochum, Germany
| | - Heiko M Lesslich
- Nanoscopy, RUBION, Ruhr-Universität Bochum, Universitätsstraße 150, D-44801 Bochum, Germany
| | - Irmgard D Dietzel
- Department of Biochemistry II, Electrobiochemistry of Neural Cells, Ruhr-Universität Bochum, D-44801 Bochum, Germany
| |
Collapse
|
45
|
Patil N, Korenfeld O, Scalf RN, Lavoie N, Huntemer-Silveira A, Han G, Swenson R, Parr AM. Electrical stimulation affects the differentiation of transplanted regionally specific human spinal neural progenitor cells (sNPCs) after chronic spinal cord injury. Stem Cell Res Ther 2023; 14:378. [PMID: 38124191 PMCID: PMC10734202 DOI: 10.1186/s13287-023-03597-w] [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: 03/27/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND There are currently no effective clinical therapies to ameliorate the loss of function that occurs after spinal cord injury. Electrical stimulation of the rat spinal cord through the rat tail has previously been described by our laboratory. We propose combinatorial treatment with human induced pluripotent stem cell-derived spinal neural progenitor cells (sNPCs) along with tail nerve electrical stimulation (TANES). The purpose of this study was to examine the influence of TANES on the differentiation of sNPCs with the hypothesis that the addition of TANES would affect incorporation of sNPCs into the injured spinal cord, which is our ultimate goal. METHODS Chronically injured athymic nude rats were allocated to one of three treatment groups: injury only, sNPC only, or sNPC + TANES. Rats were sacrificed at 16 weeks post-transplantation, and tissue was processed and analyzed utilizing standard histological and tissue clearing techniques. Functional testing was performed. All quantitative data were presented as mean ± standard error of the mean. Statistics were conducted using GraphPad Prism. RESULTS We found that sNPCs were multi-potent and retained the ability to differentiate into mainly neurons or oligodendrocytes after this transplantation paradigm. The addition of TANES resulted in more transplanted cells differentiating into oligodendrocytes compared with no TANES treatment, and more myelin was found. TANES not only promoted significantly higher numbers of sNPCs migrating away from the site of injection but also influenced long-distance axonal/dendritic projections especially in the rostral direction. Further, we observed localization of synaptophysin on SC121-positive cells, suggesting integration with host or surrounding neurons, and this finding was enhanced when TANES was applied. Also, rats that were transplanted with sNPCs in combination with TANES resulted in an increase in serotonergic fibers in the lumbar region. This suggests that TANES contributes to integration of sNPCs, as well as activity-dependent oligodendrocyte and myelin remodeling of the chronically injured spinal cord. CONCLUSIONS Together, the data suggest that the added electrical stimulation promoted cellular integration and influenced the fate of human induced pluripotent stem cell-derived sNPCs transplanted into the injured spinal cord.
Collapse
Affiliation(s)
- Nandadevi Patil
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, 2-214 MTRF, 2001 6th St. SE, Minneapolis, MN, 55455, USA
| | - Olivia Korenfeld
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, 2-214 MTRF, 2001 6th St. SE, Minneapolis, MN, 55455, USA
| | - Rachel N Scalf
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, 2-214 MTRF, 2001 6th St. SE, Minneapolis, MN, 55455, USA
| | - Nicolas Lavoie
- Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Anne Huntemer-Silveira
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, 2-214 MTRF, 2001 6th St. SE, Minneapolis, MN, 55455, USA
| | - Guebum Han
- Department of Mechanical Engineering, College of Science and Engineering, University of Minnesota, 1100 Mechanical Engineering Building, 111 Church St. SE, Minneapolis, MN, 55455, USA
| | - Riley Swenson
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, 2-214 MTRF, 2001 6th St. SE, Minneapolis, MN, 55455, USA
| | - Ann M Parr
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, MMC 96, 420 Delaware St. SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
46
|
Maden SK, Kwon SH, Huuki-Myers LA, Collado-Torres L, Hicks SC, Maynard KR. Challenges and opportunities to computationally deconvolve heterogeneous tissue with varying cell sizes using single-cell RNA-sequencing datasets. Genome Biol 2023; 24:288. [PMID: 38098055 PMCID: PMC10722720 DOI: 10.1186/s13059-023-03123-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Deconvolution of cell mixtures in "bulk" transcriptomic samples from homogenate human tissue is important for understanding disease pathologies. However, several experimental and computational challenges impede transcriptomics-based deconvolution approaches using single-cell/nucleus RNA-seq reference atlases. Cells from the brain and blood have substantially different sizes, total mRNA, and transcriptional activities, and existing approaches may quantify total mRNA instead of cell type proportions. Further, standards are lacking for the use of cell reference atlases and integrative analyses of single-cell and spatial transcriptomics data. We discuss how to approach these key challenges with orthogonal "gold standard" datasets for evaluating deconvolution methods.
Collapse
Affiliation(s)
- Sean K Maden
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Louise A Huuki-Myers
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Leonardo Collado-Torres
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA.
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA.
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
47
|
Brousse B, Mercier O, Magalon K, Gubellini P, Malapert P, Cayre M, Durbec P. Characterization of a new mouse line triggering transient oligodendrocyte progenitor depletion. Sci Rep 2023; 13:21959. [PMID: 38081969 PMCID: PMC10713661 DOI: 10.1038/s41598-023-48926-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Oligodendrocyte progenitor cells (OPC) are the main proliferative cells in the healthy adult brain. They produce new myelinating oligodendrocytes to ensure physiological myelin remodeling and regeneration after various pathological insults. Growing evidence suggests that OPC have other functions. Here, we aimed to develop an experimental model that allows the specific ablation of OPC at the adult stage to unravel possible new functions. We generated a transgenic mouse expressing a floxed human diphtheria toxin receptor under the control of the PDGFRa promoter, crossed with an Olig2Cre mouse to limit the recombination to the oligodendrocyte lineage in the central nervous system. We determined a diphtheria toxin dose to substantially decrease OPC density in the cortex and the corpus callosum without triggering side toxicity after a few daily injections. OPC density was normalized 7 days post-treatment, showing high repopulation capacity from few surviving OPC. We took advantage of this strong but transient depletion to show that OPC loss was associated with behavioral impairment, which was restored by OPC recovery, as well as disruption of the excitation/inhibition balance in the sensorimotor cortex, reinforcing the hypothesis of a neuromodulatory role of OPC in the adult brain.
Collapse
Affiliation(s)
- B Brousse
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
| | - O Mercier
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
| | - K Magalon
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
| | - P Gubellini
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
- Aix Marseille Univ, CNRS, LNC UMR7291, 3 Place Victor Hugo, 13331, Marseille Cedex 3, France
| | - P Malapert
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
| | - M Cayre
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
- Aix Marseille Univ, CNRS, LNC UMR7291, 3 Place Victor Hugo, 13331, Marseille Cedex 3, France
| | - P Durbec
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France.
| |
Collapse
|
48
|
Fiore F, Alhalaseh K, Dereddi RR, Bodaleo Torres F, Çoban I, Harb A, Agarwal A. Norepinephrine regulates calcium signals and fate of oligodendrocyte precursor cells in the mouse cerebral cortex. Nat Commun 2023; 14:8122. [PMID: 38065932 PMCID: PMC10709653 DOI: 10.1038/s41467-023-43920-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Oligodendrocyte precursor cells (OPCs) generate oligodendrocytes, contributing to myelination and myelin repair. OPCs contact axons and respond to neuronal activity, but how the information relayed by the neuronal activity translates into OPC Ca2+ signals, which in turn influence their fate, remains unknown. We generated transgenic mice for concomitant monitoring of OPCs Ca2+ signals and cell fate using 2-photon microscopy in the somatosensory cortex of awake-behaving mice. Ca2+ signals in OPCs mainly occur within processes and confine to Ca2+ microdomains. A subpopulation of OPCs enhances Ca2+ transients while mice engaged in exploratory locomotion. We found that OPCs responsive to locomotion preferentially differentiate into oligodendrocytes, and locomotion-non-responsive OPCs divide. Norepinephrine mediates locomotion-evoked Ca2+ increases in OPCs by activating α1 adrenergic receptors, and chemogenetic activation of OPCs or noradrenergic neurons promotes OPC differentiation. Hence, we uncovered that for fate decisions OPCs integrate Ca2+ signals, and norepinephrine is a potent regulator of OPC fate.
Collapse
Affiliation(s)
- Frederic Fiore
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Khaleel Alhalaseh
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Ram R Dereddi
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Felipe Bodaleo Torres
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Ilknur Çoban
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Ali Harb
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Amit Agarwal
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
49
|
Gudi V, Grieb P, Linker RA, Skripuletz T. CDP-choline to promote remyelination in multiple sclerosis: the need for a clinical trial. Neural Regen Res 2023; 18:2599-2605. [PMID: 37449595 DOI: 10.4103/1673-5374.373671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis is a multifactorial chronic inflammatory disease of the central nervous system that leads to demyelination and neuronal cell death, resulting in functional disability. Remyelination is the natural repair process of demyelination, but it is often incomplete or fails in multiple sclerosis. Available therapies reduce the inflammatory state and prevent clinical relapses. However, therapeutic approaches to increase myelin repair in humans are not yet available. The substance cytidine-5'-diphosphocholine, CDP-choline, is ubiquitously present in eukaryotic cells and plays a crucial role in the synthesis of cellular phospholipids. Regenerative properties have been shown in various animal models of diseases of the central nervous system. We have already shown that the compound CDP-choline improves myelin regeneration in two animal models of multiple sclerosis. However, the results from the animal models have not yet been studied in patients with multiple sclerosis. In this review, we summarise the beneficial effects of CDP-choline on biolipid metabolism and turnover with regard to inflammatory and regenerative processes. We also explain changes in phospholipid and sphingolipid homeostasis in multiple sclerosis and suggest a possible therapeutic link to CDP-choline.
Collapse
Affiliation(s)
- Viktoria Gudi
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Paweł Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Ralf A Linker
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | | |
Collapse
|
50
|
Ganz T, Zveik O, Fainstein N, Lachish M, Rechtman A, Sofer L, Brill L, Ben-Hur T, Vaknin-Dembinsky A. Oligodendrocyte progenitor cells differentiation induction with MAPK/ERK inhibitor fails to support repair processes in the chronically demyelinated CNS. Glia 2023; 71:2815-2831. [PMID: 37610097 DOI: 10.1002/glia.24453] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023]
Abstract
Remyelination failure is considered a major obstacle in treating chronic-progressive multiple sclerosis (MS). Studies have shown blockage in the differentiation of resident oligodendrocyte progenitor cells (OPC) into myelin-forming cells, suggesting that pushing OPC into a differentiation program might be sufficient to overcome remyelination failure. Others stressed the need for a permissive environment to allow proper activation, migration, and differentiation of OPC. PD0325901, a MAPK/ERK inhibitor, was previously shown to induce OPC differentiation, non-specific immunosuppression, and a significant therapeutic effect in acute demyelinating MS models. We examined PD0325901 effects in the chronically inflamed central nervous system. Treatment with PD0325901 induced OPC differentiation into mature oligodendrocytes with high morphological complexity. However, treatment of Biozzi mice with chronic-progressive experimental autoimmune encephalomyelitis with PD0325901 showed no clinical improvement in comparison to the control group, no reduction in demyelination, nor induction of OPC migration into foci of demyelination. PD0325901 induced a direct general immunosuppressive effect on various cell populations, leading to a diminished phagocytic capability of microglia and less activation of lymph-node cells. It also significantly impaired the immune-modulatory functions of OPC. Our findings suggest OPC regenerative function depends on a permissive environment, which may include pro-regenerative inflammatory elements. Furthermore, they indicate that maintaining a delicate balance between the pro-myelinating and immune functions of OPC is of importance. Thus, the highly complex mission of creating a pro-regenerative environment depends upon an appropriate immune response controlled in time, place, and intensity. We suggest the need to employ a multi-systematic therapeutic approach, which cannot be achieved through a single molecule-based therapy.
Collapse
Affiliation(s)
- Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lihi Sofer
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Livnat Brill
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|