1
|
Foutch K, Tilton I, Cooney A, Bender C, Licharz C, Baldemor M, Rock C, Asal Sahagun A, Brock R, Franzia C, Garcia MF, Gupta R, Arellano Reyes C, Lokhandwala M, Moura D, Noguchi H, Cocas L. Adolescent seizure impacts oligodendrocyte maturation, neuronal-glial circuit Formation, and myelination in the mammalian forebrain. Neuroscience 2025; 564:144-159. [PMID: 39571961 DOI: 10.1016/j.neuroscience.2024.11.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Oligodendrocyte progenitor cells differentiate into oligodendrocytes, which myelinate axons during development and following demyelinating injury. However, the mechanisms that drive the timing and specificity of developmental myelination are not well understood. We hypothesized that oligodendrocyte progenitor cell proliferation and differentiation would be affected by pathological neuronal activity during adolescent development when developmental myelination is occurring and that this would also impact neuron-to-oligodendrocyte progenitor cell connectivity and myelination. We used kainic acid to induce a seizure in mice, treating equal numbers of males and females, in sample sizes of at least five animals. We found that the seizures led to increased cell death overall, specifically in the oligodendrocyte-lineage cells. We found that both oligodendrocyte progenitor cell proliferation and overall numbers increased, and the number of mature oligodendrocytes decreased. We found a decrease in myelin in the cerebral cortex, corpus callosum, and hippocampus after a seizure. We observed an increase in demyelinating lesions, but no change in neuronal process length, in brains after seizure, suggesting that the demyelination was due primarily to the loss of both oligodendrocyte-lineage cells. We found that Kir4.1 potassium channel expression on oligodendrocyte progenitor cells decreased after seizure, but not mature oligodendrocytes. Finally, we found a decrease in neuron-to-oligodendrocyte progenitor cell connections in seizure mice compared to controls. These findings provide insight into the response of the adolescent brain to seizure activity, as well as how seizures affect oligodendrocyte development, neuronal-glial connections, and myelin formation.
Collapse
Affiliation(s)
- Kylie Foutch
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Iris Tilton
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States; University of California, San Francisco, Neurology Department, San Francisco, CA, United States
| | - Aundrea Cooney
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Cole Bender
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Collin Licharz
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Megan Baldemor
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Caitlyn Rock
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Atehsa Asal Sahagun
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Robert Brock
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Chloe Franzia
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Mary Francis Garcia
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Raghav Gupta
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | | | - Mariyam Lokhandwala
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States
| | - Daniela Moura
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States; University of California, San Francisco, Neurology Department, San Francisco, CA, United States
| | - Hirofumi Noguchi
- University of California, San Francisco, Neurology Department, San Francisco, CA, United States
| | - Laura Cocas
- 500 El Camino Real, Santa Clara University, Biology Department, Santa Clara, CA, United States; University of California, San Francisco, Neurology Department, San Francisco, CA, United States.
| |
Collapse
|
2
|
Margolis ET, Gabard-Durnam LJ. Prenatal influences on postnatal neuroplasticity: Integrating DOHaD and sensitive/critical period frameworks to understand biological embedding in early development. INFANCY 2025; 30:e12588. [PMID: 38449347 DOI: 10.1111/infa.12588] [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: 09/02/2023] [Revised: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
Early environments can have significant and lasting effects on brain, body, and behavior across the lifecourse. Here, we address current research efforts to understand how experiences impact neurodevelopment with a new perspective integrating two well-known conceptual frameworks - the Developmental Origins of Health and Disease (DOHaD) and sensitive/critical period frameworks. Specifically, we consider how prenatal experiences characterized in the DOHaD model impact two key neurobiological mechanisms of sensitive/critical periods for adapting to and learning from the postnatal environment. We draw from both animal and human research to summarize the current state of knowledge on how particular prenatal substance exposures (psychoactive substances and heavy metals) and nutritional profiles (protein-energy malnutrition and iron deficiency) each differentially impact brain circuits' excitation/GABAergic inhibition balance and myelination. Finally, we highlight new research directions that emerge from this integrated framework, including testing how prenatal environments alter sensitive/critical period timing and learning and identifying potential promotional/buffering prenatal exposures to impact postnatal sensitive/critical periods. We hope this integrative framework considering prenatal influences on postnatal neuroplasticity will stimulate new research to understand how early environments have lasting consequences on our brains, behavior, and health.
Collapse
Affiliation(s)
- Emma T Margolis
- Department of Psychology, Northeastern University, Boston, Massachusetts, USA
| | | |
Collapse
|
3
|
Xu X, Fang M, Chen L, Huang H, Dai ZM, Yang J, Qiu M. Nzf2 promotes oligodendrocyte differentiation and regeneration via repressing HDAC1-mediated histone deacetylation. SCIENCE ADVANCES 2024; 10:eadf8405. [PMID: 39671488 PMCID: PMC11641009 DOI: 10.1126/sciadv.adf8405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/07/2024] [Indexed: 12/15/2024]
Abstract
Proper axonal myelination and function of the vertebrate central nervous system rely largely on the timely differentiation of oligodendrocytes (OLs), yet key regulatory factors remain enigmatic. Our study reveals neural zinc finger (Nzf2) as a crucial orchestrator that controls the timing of OL differentiation both during development and myelin repair, contrasting with its previously suggested role in direct myelin gene regulation. Nzf2 ablation delays the onset of OL differentiation, while hyperactivation stimulates OL differentiation both during development and remyelination. Using RNA-seq and ChIP-seq, we pinpoint Nkx2.2 as a critical downstream target of Nzf2. Specific binding of Nzf2 in the Nkx2.2 gene locus inhibits histone deacetylation by disrupting the HDAC1 repressor complex and reducing deacetylase activity. Furthermore, Nzf2 overrides the inhibitory Notch signaling to initiate OL differentiation. Thus, we propose that the Notch-Nzf2-Nkx2.2 axis is a vital component of OL differentiation timing mechanism, suggesting Nzf2 as a potential therapeutic target for stimulating OL differentiation and boosting myelin repair in demyelinating diseases.
Collapse
Affiliation(s)
- Xiaofeng Xu
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Minxi Fang
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lixia Chen
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Hao Huang
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Zhong-Min Dai
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Junlin Yang
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
4
|
Palazzo C, Nutarelli S, Mastrantonio R, Tamagnone L, Viscomi MT. Glia-glia crosstalk via semaphorins: Emerging implications in neurodegeneration. Ageing Res Rev 2024; 104:102618. [PMID: 39638095 DOI: 10.1016/j.arr.2024.102618] [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: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
The central nervous system (CNS) is wired by a complex network of integrated glial and neuronal signals, which is critical for its development and homeostasis. In this context, glia-glia communication is a complex and dynamic process that is essential for ensuring optimal CNS function. Semaphorins, which include secreted and transmembrane molecules, and their receptors, mainly found in the plexin and neuropilin families, are expressed in a wide range of cell types, including glia. In the CNS, semaphorin signalling is involved in a spectrum of processes, including neurogenesis, neuronal migration and wiring, and glial cell recruitment. Recently, semaphorins and plexins have attracted intense research aimed at elucidating their roles in instructing glial cell behavior during development or in response to inflammatory stimuli. In this review, we provide an overview of the multifaceted role of semaphorins in glia-glia communication, highlighting recent discoveries about semaphoring-dependent regulation of glia functions in healthy conditions. We also discuss the mechanisms of gliaglia crosstalk mediated by semaphorins under pathological conditions, and how these interactions may provide potential avenues for therapeutic intervention in neuroinflammation-mediated neurodegeneration.
Collapse
Affiliation(s)
- Claudia Palazzo
- Department of Life Sciences and Public Health, Section of Histology and Embryology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sofia Nutarelli
- Department of Life Sciences and Public Health, Section of Histology and Embryology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberta Mastrantonio
- Department of Life Sciences and Public Health, Section of Histology and Embryology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Tamagnone
- Department of Life Sciences and Public Health, Section of Histology and Embryology, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, Rome, Italy.
| | - Maria Teresa Viscomi
- Department of Life Sciences and Public Health, Section of Histology and Embryology, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, Rome, Italy.
| |
Collapse
|
5
|
Back AM, Connor B, McCaughey-Chapman A. Oligodendrocytes in Huntington's Disease: A Review of Oligodendrocyte Pathology and Current Cell Reprogramming Approaches for Oligodendrocyte Modelling of Huntington's Disease. J Neurosci Res 2024; 102:e70010. [PMID: 39714111 DOI: 10.1002/jnr.70010] [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: 07/03/2024] [Revised: 11/24/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder traditionally characterized by the selective loss of medium spiny neurons in the basal ganglia. However, it has become apparent that white matter injury and oligodendrocyte dysfunction precede the degeneration of medium spiny neurons, garnering interest as a key pathogenic mechanism of HD. Oligodendrocytes are glial cells found within the central nervous system involved in the production of myelin and the myelination of axons. Myelin is a lipid-rich sheath that wraps around axons, facilitating signal conduction and neuronal viability. The degeneration of myelin hinders effective communication and leaves neurons vulnerable to external damage and subsequent degeneration. Abnormalities in oligodendrocyte maturation have been established in the HD human brain, however, investigations into the underlying dysfunction of human oligodendrocytes in HD are limited. This review will detail the involvement of oligodendrocytes and white matter damage in HD. Recent developments in modeling human-specific oligodendrocyte pathology in HD will be discussed, with a particular focus on emerging somatic cell reprogramming approaches.
Collapse
Affiliation(s)
- Amelie Marie Back
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, School of Medical Science, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, School of Medical Science, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Amy McCaughey-Chapman
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, School of Medical Science, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
6
|
Yang Y, Suo N, Cui SH, Wu X, Ren XY, Liu Y, Guo R, Xie X. Trametinib, an anti-tumor drug, promotes oligodendrocytes generation and myelin formation. Acta Pharmacol Sin 2024; 45:2527-2539. [PMID: 38871922 PMCID: PMC11579360 DOI: 10.1038/s41401-024-01313-9] [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: 12/24/2023] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
Oligodendrocytes (OLs) are differentiated from oligodendrocyte precursor cells (OPCs) in the central nervous system (CNS). Demyelination is a common feature of many neurological diseases such as multiple sclerosis (MS) and leukodystrophies. Although spontaneous remyelination can happen after myelin injury, nevertheless, it is often insufficient and may lead to aggravated neurodegeneration and neurological disabilities. Our previous study has discovered that MEK/ERK pathway negatively regulates OPC-to-OL differentiation and remyelination in mouse models. To facilitate possible clinical evaluation, here we investigate several MEK inhibitors which have been approved by FDA for cancer therapies in both mouse and human OPC-to-OL differentiation systems. Trametinib, the first FDA approved MEK inhibitor, displays the best effect in stimulating OL generation in vitro among the four MEK inhibitors examined. Trametinib also significantly enhances remyelination in both MOG-induced EAE model and LPC-induced focal demyelination model. More exciting, trametinib facilitates the generation of MBP+ OLs from human embryonic stem cells (ESCs)-derived OPCs. Mechanism study indicates that trametinib promotes OL generation by reducing E2F1 nuclear translocation and subsequent transcriptional activity. In summary, our studies indicate a similar inhibitory role of MEK/ERK in human and mouse OL generation. Targeting the MEK/ERK pathway might help to develop new therapies or repurpose existing drugs for demyelinating diseases.
Collapse
Affiliation(s)
- Ying Yang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Na Suo
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Shi-Hao Cui
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuan Wu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xin-Yue Ren
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yin Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ren Guo
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Xin Xie
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
7
|
Lee VY, Nils AVM, Arruda BP, Xavier GF, Nogueira MI, Motta-Teixeira LC, Takada SH. Spontaneous running wheel exercise during pregnancy prevents later neonatal-anoxia-induced somatic and neurodevelopmental alterations. IBRO Neurosci Rep 2024; 17:263-279. [PMID: 39310269 PMCID: PMC11414703 DOI: 10.1016/j.ibneur.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction About 15-20 % of babies that suffer perinatal asphyxia die and around 25 % of the survivors exhibit permanent neural outcomes. Minimization of this global health problem has been warranted. This study investigated if the offspring of pregnant female rats allowed to spontaneously exercise on running wheels along a 11-day pregnancy period were protected for somatic and neurodevelopmental disturbs that usually follow neonatal anoxia. Methods spontaneous exercise was applied to female rats which were housed in cages allowing free access to running wheels along a 11-day pregnancy period. Their offspring were submitted to anoxia 24-36 h after birth. Somatic and sensory-motor development of the pups were recorded until postnatal day 21 (P21). Myelin basic protein (MBP)-stained areas of sensory and motor cortices were measured at P21. Neuronal nuclei (NeuN)-immunopositive cells and synapsin-I levels in hippocampal formation were estimated at P21 and P75. Results gestational exercise and / or neonatal anoxia increased the weight and the size of the pups. In addition, gestational exercise accelerated somatic and sensory-motor development of the pups and protected them against neonatal-anoxia-induced delay in development. Further, neonatal anoxia reduced MBP stained area in the secondary motor cortex and decreased hippocampal neuronal estimates and synapsin-I levels at P21; gestational exercise prevented these effects. Therefore, spontaneous exercise along pregnancy is a valuable strategy to prevent neonatal-anoxia-induced disturbs in the offspring. Conclusion spontaneous gestational running wheel exercise protects against neonatal anoxia-induced disturbs in the offspring, including (1) physical and neurobehavioral developmental impairments, and (2) hippocampal and cortical changes. Thus, spontaneous exercise during pregnancy may represent a valuable strategy to prevent disturbs which usually follow neonatal anoxia.
Collapse
Affiliation(s)
- Vitor Yonamine Lee
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
| | - Aline Vilar Machado Nils
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, R. do Matão, Travessa 14, 101, Sao Paulo 05508-900, Brazil
| | - Bruna Petrucelli Arruda
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Alameda da Universidade, s/n, Bloco Delta, São Bernardo do Campo, SP 09606-070, Brazil
| | - Gilberto Fernando Xavier
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, R. do Matão, Travessa 14, 101, Sao Paulo 05508-900, Brazil
| | - Maria Inês Nogueira
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
| | - Lívia Clemente Motta-Teixeira
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, R. do Matão, Travessa 14, 101, Sao Paulo 05508-900, Brazil
- Departamento de Ciências Fisiológicas, Faculdade de Ciências Médicas da Santa Casa de São Paulo, R. Jaguaribe, 155 - Vila Buarque, Sao Paulo, SP 01224-001, Brazil
| | - Silvia Honda Takada
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Alameda da Universidade, s/n, Bloco Delta, São Bernardo do Campo, SP 09606-070, Brazil
| |
Collapse
|
8
|
Ozgür-Gunes Y, Le Stunff C, Bougnères P. Oligodendrocytes, the Forgotten Target of Gene Therapy. Cells 2024; 13:1973. [PMID: 39682723 DOI: 10.3390/cells13231973] [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: 10/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
If the billions of oligodendrocytes (OLs) populating the central nervous system (CNS) of patients could express their feelings, they would undoubtedly tell gene therapists about their frustration with the other neural cell populations, neurons, microglia, or astrocytes, which have been the favorite targets of gene transfer experiments. This review questions why OLs have been left out of most gene therapy attempts. The first explanation is that the pathogenic role of OLs is still discussed in most CNS diseases. Another reason is that the so-called ubiquitous CAG, CBA, CBh, or CMV promoters-widely used in gene therapy studies-are unable or poorly able to activate the transcription of episomal transgene copies brought by adeno-associated virus (AAV) vectors in OLs. Accordingly, transgene expression in OLs has either not been found or not been evaluated in most gene therapy studies in rodents or non-human primates. The aims of the current review are to give OLs their rightful place among the neural cells that future gene therapy could target and to encourage researchers to test the effect of OL transduction in various CNS diseases.
Collapse
Affiliation(s)
- Yasemin Ozgür-Gunes
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Catherine Le Stunff
- MIRCen Institute, Laboratoire des Maladies Neurodégénératives, Commissariat à l'Energie Atomique, 92260 Fontenay-aux-Roses, France
- NEURATRIS at MIRCen, 92260 Fontenay-aux-Roses, France
- UMR1195 Inserm and University Paris Saclay, 94270 Le Kremlin-Bicêtre, France
| | - Pierre Bougnères
- MIRCen Institute, Laboratoire des Maladies Neurodégénératives, Commissariat à l'Energie Atomique, 92260 Fontenay-aux-Roses, France
- NEURATRIS at MIRCen, 92260 Fontenay-aux-Roses, France
- Therapy Design Consulting, 94300 Vincennes, France
| |
Collapse
|
9
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 DOI: 10.3390/ijms252312637] [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: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
10
|
Kamen Y, Chapman TW, Piedra ET, Ciolkowski ME, Hill RA. Transient upregulation of procaspase-3 during oligodendrocyte fate decisions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623446. [PMID: 39605489 PMCID: PMC11601457 DOI: 10.1101/2024.11.13.623446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Oligodendrocytes are generated throughout life and in neurodegenerative conditions from brain resident oligodendrocyte precursor cells (OPCs). The transition from OPC to oligodendrocyte involves a complex cascade of molecular and morphological states that position the cell to make a fate decision to integrate as a myelinating oligodendrocyte or die through apoptosis. Oligodendrocyte maturation impacts the cell death mechanisms that occur in degenerative conditions, but it is unclear if and how the cell death machinery changes as OPCs transition into oligodendrocytes. Here, we discovered that differentiating oligodendrocytes transiently upregulate the zymogen procaspase-3, equipping these cells to make a survival decision during differentiation. Pharmacological inhibition of caspase-3 decreases oligodendrocyte density, indicating that procaspase-3 upregulation promotes differentiation. Moreover, using procaspase-3 as a marker, we show that oligodendrocyte differentiation continues in the aging cortex and white matter. Taken together, our data establish procaspase-3 as a differentiating oligodendrocyte marker and provide insight into the underlying mechanisms occurring during the decision to integrate or die.
Collapse
Affiliation(s)
- Yasmine Kamen
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Timothy W. Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Enrique T. Piedra
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | | | - Robert A. Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
11
|
Jaftha M, Robertson F, van Rensburg SJ, Kidd M, van Toorn R, Kemp MC, Johannes C, Moremi KE, Whati L, Kotze MJ, Engel-Hills P. White Matter Lesion Volumes on 3-T MRI in People With MS Who Had Followed a Diet and Lifestyle Program for More Than 10 Years. Mult Scler Int 2024; 2024:8818934. [PMID: 39524062 PMCID: PMC11548950 DOI: 10.1155/2024/8818934] [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: 05/30/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Cerebral white matter lesion (WML) formation in people with multiple sclerosis (pwMS) is linked to the death of myelin-producing oligodendrocytes. Current MS treatment strategies focus on limiting WML accumulation and disability. Using a pathology-supported genetic testing (PSGT) program, we identified specific risk factors for MS, categorized as deficiencies and aggravators. We developed a novel clinical methodology to mitigate these risk factors, including personalized lifestyle interventions and optimization of cerebral nutrients to prevent oligodendrocyte demise and promote remyelination. Objective: To conduct a pilot case-control study over a 10-year period to ascertain whether the PSGT Program can reduce or prevent WML formation in pwMS. Methods: MRI was performed at baseline as well as after an interval period of at least 10 years or longer in 22 pwMS. WML volumes were determined using Sequence Adaptive Multimodal SEGmentation (SAMSEG) software, part of FreeSurfer 7.2. Other variables included age at MRI, disease duration, disability status, and medication. Results: PwMS (n = 13) who had followed the PSGT program for more than 10 years, had significantly smaller lesion volumes (mm3) compared to pwMS who did not adhere to the program (n = 9) (4950 ± 5303 vs. 17934 ± 11139; p = 0.002). WML volumes were significantly associated (p = 0.02) with disability (EDSS) but not with age (p = 0.350), disease duration (p = 0.709), or Interferon-β treatment (p = 0.70). Conclusion: Dietary and lifestyle changes may lower the risk of developing cerebral WMLs in pwMS and potentially slow disease progression. Larger studies are required to confirm the effectiveness of such interventions in pwMS.
Collapse
Affiliation(s)
- Mariaan Jaftha
- Department of Medical Imaging and Therapeutic Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, Cape Town, South Africa
- Cape University Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frances Robertson
- Cape University Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Susan J. van Rensburg
- Division of Chemical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Martin Kidd
- Centre for Statistical Consultation, Department of Statistics and Actuarial Sciences, Stellenbosch University, Private Bag X1, Matieland 7602, Cape Town, South Africa
| | - Ronald van Toorn
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Merlisa C. Kemp
- Department of Medical Imaging and Therapeutic Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, Cape Town, South Africa
- Medical Imaging, Department of Health and Care Professions, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Clint Johannes
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7500, Cape Town, South Africa
| | - Kelebogile E. Moremi
- Division of Chemical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, and National Health Laboratory Service (NHLS), Cape Town, South Africa
| | | | - Maritha J. Kotze
- Division of Chemical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, and National Health Laboratory Service (NHLS), Cape Town, South Africa
| | - Penelope Engel-Hills
- Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| |
Collapse
|
12
|
Huang G, Li Z, Liu X, Guan M, Zhou S, Zhong X, Zheng T, Xin D, Gu X, Mu D, Guo Y, Zhang L, Zhang L, Lu QR, He X. DOR activation in mature oligodendrocytes regulates α-ketoglutarate metabolism leading to enhanced remyelination in aged mice. Nat Neurosci 2024; 27:2073-2085. [PMID: 39266660 DOI: 10.1038/s41593-024-01754-9] [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: 07/20/2023] [Accepted: 08/07/2024] [Indexed: 09/14/2024]
Abstract
The decreased ability of mature oligodendrocytes to produce myelin negatively affects remyelination in demyelinating diseases and aging, but the underlying mechanisms are incompletely understood. In the present study, we identify a mature oligodendrocyte-enriched transcriptional coregulator diabetes- and obesity-related gene (DOR)/tumor protein p53-inducible nuclear protein 2 (TP53INP2), downregulated in demyelinated lesions of donors with multiple sclerosis and in aged oligodendrocyte-lineage cells. Dor ablation in mice of both sexes results in defective myelinogenesis and remyelination. Genomic occupancy in oligodendrocytes and transcriptome profiling of the optic nerves of wild-type and Dor conditional knockout mice reveal that DOR and SOX10 co-occupy enhancers of critical myelinogenesis-associated genes including Prr18, encoding an oligodendrocyte-enriched, proline-rich factor. We show that DOR targets regulatory elements of genes responsible for α-ketoglutarate biosynthesis in mature oligodendrocytes and is essential for α-ketoglutarate production and lipid biosynthesis. Supplementation with α-ketoglutarate restores oligodendrocyte-maturation defects in Dor-deficient adult mice and improves remyelination after lysolecithin-induced demyelination and cognitive function in 17-month-old wild-type mice. Our data suggest that activation of α-ketoglutarate metabolism in mature oligodendrocytes can promote myelin production during demyelination and aging.
Collapse
Affiliation(s)
- Guojiao Huang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhidan Li
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xuezhao Liu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Menglong Guan
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaowen Zhong
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tao Zheng
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dazhuan Xin
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Dezhi Mu
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yingkun Guo
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lin Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liguo Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xuelian He
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Liu LL, Emir U, Gu H, Sang LT, Sawiak SJ, Cannon JR, Du Y, Zheng W. Demyelination and impaired oligodendrogenesis in the corpus callosum following lead exposure. Toxicol Sci 2024; 202:123-141. [PMID: 39150886 PMCID: PMC11514834 DOI: 10.1093/toxsci/kfae100] [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: 08/18/2024] Open
Abstract
The corpus callosum is an oligodendrocyte-enriched brain region, replenished by newborn oligodendrocytes from oligodendrocyte progenitor cells (OPCs) in subventricular zone (SVZ). Lead (Pb) exposure has been associated with multiple sclerosis, a disease characterized by the loss of oligodendrocytes. This study aimed to investigate the effects of Pb exposure on oligodendrogenesis in SVZ and myelination in the corpus callosum. Adult female mice were used for a disproportionately higher prevalence of multiple sclerosis in females. Acute Pb exposure (one ip-injection of 27 mg Pb/kg as PbAc2 24 hr before sampling) caused mild Pb accumulation in the corpus callosum. Ex vivo assay using isolated SVZ tissues collected from acute Pb-exposed brains showed a diminished oligodendrogenesis in SVZ-derived neurospheres compared with controls. In vivo subchronic Pb exposure (13.5 mg Pb/kg by daily oral gavage 4 wk) revealed significantly decreased newborn BrdU+/MBP+ oligodendrocytes in the corpus callosum, suggesting demyelination. Mechanistic investigations indicated decreased Rictor in SVZ OPCs, defective self-defense pathways, and reactive gliosis in the corpus callosum. Given the interwined pathologies between multiple sclerosis and Alzheimer's disease, the effect of Pb on myelination was evaluated in AD-modeled APP/PS1 mice. Myelin MRI on mice following chronic exposure (1,000 ppm Pb in drinking water as PbAc2 for 20 wk) revealed a profound demyelination in the corpus callosum compared with controls. Immunostaining of the choroid plexus showed diminished signaling molecule (Klotho, OTX2) expressions in Pb-treated animals. These observations suggest that Pb caused demyelination in the corpus callosum, likely by disrupting oligodendrogenesis from SVZ OPCs. Pb-induced demyelination represents a crucial pathogenic pathway in Pb neurotoxicity, including multiple sclerosis.
Collapse
Affiliation(s)
- Luke L Liu
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - Uzay Emir
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - Huiying Gu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Lara T Sang
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - Stephen J Sawiak
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge CB2 3EB, United Kingdom
| | - Jason R Cannon
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, United States
| |
Collapse
|
14
|
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
|
15
|
Harvey BM, Baxter M, Garcia AM, Granato M. Glial cell derived pathway directs regenerating optic nerve axons toward the CNS midline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618346. [PMID: 39464127 PMCID: PMC11507804 DOI: 10.1101/2024.10.15.618346] [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: 10/29/2024]
Abstract
Several RGC intrinsic signaling pathways have been shown to enhance RGC survival and RGC axonal growth after optic nerve injury. Yet an unresolved challenge for regenerating RGC axons is to properly navigate the optic chiasm located at the Central Nervous System midline. Here, we use live-cell imaging in larval zebrafish to show that regrowing RGC axons initiate growth toward the midline and extend along a trajectory similar to their original projection. From a candidate genetic screen, we identify the glycosyltransferase Lh3 to be required during the process of regeneration to direct regrowing RGC axons toward the midline. Moreover, we find that mutants in collagen 18a1 (col18a1), a putative Lh3 substrate, display RGC axonal misguidance phenotypes similar to those we observe in lh3 mutants, suggesting that lh3 may act through col18a1 during regeneration. Finally, we show that transgenic lh3 expression in sox10+ presumptive olig2+ oligodendrocytes located near the optic chiasm restores directed axonal growth. Combined these data identify lh3 and col18a1 as part of a glial derived molecular pathway critical for guiding in vivo regenerating RGC axons towards and across the optic chiasm.
Collapse
Affiliation(s)
- Beth M. Harvey
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Melissa Baxter
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Alexis M. Garcia
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
16
|
Raj S, Sarangi P, Goyal D, Kumar H. The Hidden Hand in White Matter: Pericytes and the Puzzle of Demyelination. ACS Pharmacol Transl Sci 2024; 7:2912-2923. [PMID: 39421660 PMCID: PMC11480894 DOI: 10.1021/acsptsci.4c00192] [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: 04/03/2024] [Revised: 08/01/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Disruption of myelin, the fatty sheath-insulating nerve fibers in the white matter, blocks or slows the rapid transmission of electrical signals along nerve cells and contributes to several neurodegenerative diseases such as multiple sclerosis. Traditionally, research has focused on neuronal dysfunction as the primary factor, including autoimmunity, infections, inflammation, and genetic disorders causing demyelination. However, recent insights emphasize the critical role of pericytes, non-neuronal cells that regulate blood flow and maintain the health of blood vessels within white matter. This Perspective explores the principal mechanisms through which pericyte dysfunction contributes to damage and demyelination, including impaired communication with neurons (neurovascular uncoupling), excessive formation of scar tissue (fibrosis), and the infiltration of detrimental substances from the bloodstream. Understanding these mechanisms of pericyte-driven demyelination may lead to the creation of new therapeutic strategies for tackling a range of neurodegenerative conditions.
Collapse
Affiliation(s)
- Siddharth Raj
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Priyabrata Sarangi
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Divya Goyal
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Hemant Kumar
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| |
Collapse
|
17
|
Liu P, Zhang K, Tong C, Liu T, Zheng J. Progesterone alleviates esketamine-induced hypomyelination via PI3K/Akt signaling pathway in the developing rat brain. Biotechnol Genet Eng Rev 2024; 40:1202-1217. [PMID: 36946765 DOI: 10.1080/02648725.2023.2193058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/14/2023] [Indexed: 03/23/2023]
Abstract
The neurodevelopmental toxicity of anesthetics has been confirmed repeatedly, and esketamine is now widely used in pediatric surgeries. Oligodendrocyte precursor cells (OPCs) evolved into mature oligodendrocytes (OLs) and formed myeline sheath during the early brain development. In this study, we investigated whether esketamine exposure interrupted development of OPCs and induced hypomyelination in rats. Further we explored the roles of PI3K/Akt phosphorylation in OPCs development and myelination. Sprague Dawley rats with different ages (postnatal day (P) 1, 3, 7 and 12) were exposed to 40mg/kg esketamine. Progesterone treatment was given (16 mg/kg per day for 3 days) 24 h after esketamine exposure via the intraperitoneal route. Corpus callosum tissues were collected at P8 or P14 for western blot and immunofluorescence analyses. Esketamine exposure at P7 and P12 significantly reduced myelin basic protein (MBP) expression and CC1+ OLs number in corpus callosum. Esketamine exposure at P7 not only aggravated the mature OLs apoptosis, also decreased the OPCs proliferation and differentiation, which was related with dephosphorylation of PI3K/Akt. Progesterone was able to promote OPCs differentiation and ameliorate esketamine-induced hypomyelination by enhancing PI3K/Akt phosphorylation. Stage-dependent abnormality of OPCs/OLs after esketamine leads to the esketamine-induced hypomyelination. Esketamine interrupted OPCs evolution via PI3K/Akt signaling pathway, which can be ameliorated by progesterone.
Collapse
Affiliation(s)
- Peiwen Liu
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kan Zhang
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chaoyang Tong
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Liu
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jijian Zheng
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Brandt M, Kosmeijer C, Achterberg E, de Theije C, Nijboer C. Timed fetal inflammation and postnatal hypoxia cause cortical white matter injury, interneuron imbalances, and behavioral deficits in a double-hit rat model of encephalopathy of prematurity. Brain Behav Immun Health 2024; 40:100817. [PMID: 39188404 PMCID: PMC11345510 DOI: 10.1016/j.bbih.2024.100817] [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: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 08/28/2024] Open
Abstract
Extreme preterm birth-associated adversities are a major risk factor for aberrant brain development, known as encephalopathy of prematurity (EoP), which can lead to long-term neurodevelopmental impairments. Although progress in clinical care for preterm infants has markedly improved perinatal outcomes, there are currently no curative treatment options available to combat EoP. EoP has a multifactorial etiology, including but not limited to pre- or postnatal immune activation and oxygen fluctuations. Elucidating the underlying mechanisms of EoP and determining the efficacy of potential therapies relies on valid, clinically translatable experimental models that reflect the neurodevelopmental and pathophysiological hallmarks of EoP. Here, we expand on our double-hit rat model that can be used to study EoP disease mechanisms and therapeutic options in a preclinical setting. Pregnant Wistar dams were intraperitoneally injected with 10 μg/kg LPS on embryonic day (E)20 and offspring was subjected to hypoxia (140 min, 8% O2) at postnatal day 4. Rats exposed to fetal inflammation and postnatal hypoxia (FIPH) showed neurodevelopmental impairments, such as reduced nest-seeking ability, ultrasonic vocalizations, social engagement, and working memory, and increased anxiety and sensitivity. Impairments in myelination, oligodendrocyte maturation and interneuron development were examined as hallmarks for EoP, in different layers and coordinates of the cortex using histological and molecular techniques. Myelin density and complexity was decreased in the cortex, which partially coincided with a decrease in mature oligodendrocytes. Furthermore, interneuron populations (GAD67+ and PVALB+) were affected. To determine if the timing of inducing fetal inflammation affected the severity of EoP hallmarks in the cortex, multiple timepoints of fetal inflammation were compared. Inflammation at E20 combined with postnatal hypoxia gave the most severe EoP phenotype in the cortex. In conclusion, we present a double-hit rat model which displays various behavioral, anatomical and molecular hallmarks of EoP, including diffuse white matter injury. This double-hit model can be used to investigate pathophysiological mechanisms and potential therapies for EoP.
Collapse
Affiliation(s)
- M.J.V. Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands
| | - C.M. Kosmeijer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands
| | - E.J.M. Achterberg
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, the Netherlands
| | - C.G.M. de Theije
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands
| | - C.H. Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands
| |
Collapse
|
19
|
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
|
20
|
Kaller MS, Lazari A, Feng Y, van der Toorn A, Rühling S, Thomas CW, Shimizu T, Bannerman D, Vyazovskiy V, Richardson WD, Sampaio-Baptista C, Johansen-Berg H. Ablation of oligodendrogenesis in adult mice alters brain microstructure and activity independently of behavioral deficits. Glia 2024; 72:1728-1745. [PMID: 38982743 DOI: 10.1002/glia.24576] [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: 10/13/2023] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 07/11/2024]
Abstract
Oligodendrocytes continue to differentiate from their precursor cells even in adulthood, a process that can be modulated by neuronal activity and experience. Previous work has indicated that conditional ablation of oligodendrogenesis in adult mice leads to learning and memory deficits in a range of behavioral tasks. The current study replicated and re-evaluated evidence for a role of oligodendrogenesis in motor learning, using a complex running wheel task. Further, we found that ablating oligodendrogenesis alters brain microstructure (ex vivo MRI) and brain activity (in vivo EEG) independent of experience with the task. This suggests a role for adult oligodendrocyte formation in the maintenance of brain function and indicates that task-independent changes due to oligodendrogenesis ablation need to be considered when interpreting learning and memory deficits in this model.
Collapse
Affiliation(s)
- Malte S Kaller
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Alberto Lazari
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Yingshi Feng
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Annette van der Toorn
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht & Utrecht University, Utrecht, The Netherlands
| | - Sebastian Rühling
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christopher W Thomas
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Takahiro Shimizu
- The Wolfson Institute for Biomedical Research, University College London, London, UK
| | - David Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Vladyslav Vyazovskiy
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, UK
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - William D Richardson
- The Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Cassandra Sampaio-Baptista
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, UK
| | - Heidi Johansen-Berg
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
21
|
Bagheri H, Friedman H, Hadwen A, Jarweh C, Cooper E, Oprea L, Guerrier C, Khadra A, Collin A, Cohen‐Adad J, Young A, Victoriano GM, Swire M, Jarjour A, Bechler ME, Pryce RS, Chaurand P, Cougnaud L, Vuckovic D, Wilion E, Greene O, Nishiyama A, Benmamar‐Badel A, Owens T, Grouza V, Tuznik M, Liu H, Rudko DA, Zhang J, Siminovitch KA, Peterson AC. Myelin basic protein mRNA levels affect myelin sheath dimensions, architecture, plasticity, and density of resident glial cells. Glia 2024; 72:1893-1914. [PMID: 39023138 PMCID: PMC11426340 DOI: 10.1002/glia.24589] [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: 08/07/2024] [Revised: 05/29/2024] [Accepted: 06/23/2024] [Indexed: 07/20/2024]
Abstract
Myelin Basic Protein (MBP) is essential for both elaboration and maintenance of CNS myelin, and its reduced accumulation results in hypomyelination. How different Mbp mRNA levels affect myelin dimensions across the lifespan and how resident glial cells may respond to such changes are unknown. Here, to investigate these questions, we used enhancer-edited mouse lines that accumulate Mbp mRNA levels ranging from 8% to 160% of wild type. In young mice, reduced Mbp mRNA levels resulted in corresponding decreases in Mbp protein accumulation and myelin sheath thickness, confirming the previously demonstrated rate-limiting role of Mbp transcription in the control of initial myelin synthesis. However, despite maintaining lower line specific Mbp mRNA levels into old age, both MBP protein levels and myelin thickness improved or fully normalized at rates defined by the relative Mbp mRNA level. Sheath length, in contrast, was affected only when mRNA levels were very low, demonstrating that sheath thickness and length are not equally coupled to Mbp mRNA level. Striking abnormalities in sheath structure also emerged with reduced mRNA levels. Unexpectedly, an increase in the density of all glial cell types arose in response to reduced Mbp mRNA levels. This investigation extends understanding of the role MBP plays in myelin sheath elaboration, architecture, and plasticity across the mouse lifespan and illuminates a novel axis of glial cell crosstalk.
Collapse
Affiliation(s)
- Hooman Bagheri
- Department of Human GeneticsMcGill UniversityMontrealQuebecCanada
| | - Hana Friedman
- Department of Human GeneticsMcGill UniversityMontrealQuebecCanada
| | - Amanda Hadwen
- Department of PhysiologyMcGill UniversityMontrealQuebecCanada
| | - Celia Jarweh
- Department of Pharmacology & TherapeuticsMcGill UniversityMontrealQuebecCanada
| | - Ellis Cooper
- Department of PhysiologyMcGill UniversityMontrealQuebecCanada
| | - Lawrence Oprea
- Integrated Program in NeuroscienceMcGill UniversityMontréalQuebecCanada
| | | | - Anmar Khadra
- Integrated Program in NeuroscienceMcGill UniversityMontréalQuebecCanada
| | - Armand Collin
- Institute of Biomedical Engineering, Ecole Polytechnique de MontrealMontrealQuebecCanada
| | - Julien Cohen‐Adad
- Institute of Biomedical Engineering, Ecole Polytechnique de MontrealMontrealQuebecCanada
| | - Amanda Young
- Department of Cell and Developmental BiologyState University of New York Upstate Medical UniversitySyracuseNew YorkUSA
- Department of Neuroscience and PhysiologyState University of New York Upstate Medical UniversitySyracuseNew YorkUSA
| | - Gerardo Mendez Victoriano
- Department of Cell and Developmental BiologyState University of New York Upstate Medical UniversitySyracuseNew YorkUSA
- Department of Neuroscience and PhysiologyState University of New York Upstate Medical UniversitySyracuseNew YorkUSA
| | - Matthew Swire
- Department of Cell and Developmental BiologyState University of New York Upstate Medical UniversitySyracuseNew YorkUSA
- Department of Neuroscience and PhysiologyState University of New York Upstate Medical UniversitySyracuseNew YorkUSA
| | - Andrew Jarjour
- Department of Cell and Developmental BiologyState University of New York Upstate Medical UniversitySyracuseNew YorkUSA
- Department of Neuroscience and PhysiologyState University of New York Upstate Medical UniversitySyracuseNew YorkUSA
| | - Marie E. Bechler
- Department of Cell and Developmental BiologyState University of New York Upstate Medical UniversitySyracuseNew YorkUSA
- Department of Neuroscience and PhysiologyState University of New York Upstate Medical UniversitySyracuseNew YorkUSA
| | - Rachel S. Pryce
- Department of ChemistryUniversité de MontréalMontrealQuebecCanada
| | - Pierre Chaurand
- Department of ChemistryUniversité de MontréalMontrealQuebecCanada
| | - Lise Cougnaud
- Department of Chemistry and BiochemistryConcordia UniversityMontrealQuebecCanada
| | - Dajana Vuckovic
- Department of Chemistry and BiochemistryConcordia UniversityMontrealQuebecCanada
| | - Elliott Wilion
- Department of Physiology and NeurobiologyUniversity of ConnecticutStorrsConnecticutUSA
| | - Owen Greene
- Department of Physiology and NeurobiologyUniversity of ConnecticutStorrsConnecticutUSA
| | - Akiko Nishiyama
- Department of Physiology and NeurobiologyUniversity of ConnecticutStorrsConnecticutUSA
- Institute for Systems Genomics, University of ConnecticutStorrsConnecticutUSA
- The Connecticut Institute for Brain and Cognitive Sciences, University of ConnecticutStorrsConnecticutUSA
| | - Anouk Benmamar‐Badel
- Department of Neurobiology ResearchInstitute for Molecular Medicine, University of Southern DenmarkOdenseDenmark
| | - Trevor Owens
- Department of Neurobiology ResearchInstitute for Molecular Medicine, University of Southern DenmarkOdenseDenmark
| | - Vladimir Grouza
- McConnell Brain Imaging Centre, Montreal Neurological Institute and HospitalMontrealQuebecCanada
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada
| | - Marius Tuznik
- McConnell Brain Imaging Centre, Montreal Neurological Institute and HospitalMontrealQuebecCanada
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada
| | - Hanwen Liu
- McConnell Brain Imaging Centre, Montreal Neurological Institute and HospitalMontrealQuebecCanada
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada
| | - David A. Rudko
- McConnell Brain Imaging Centre, Montreal Neurological Institute and HospitalMontrealQuebecCanada
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada
- Department of Biomedical EngineeringMcGill UniversityMontrealQuebecCanada
| | - Jinyi Zhang
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
- Mount Sinai Hospital, Lunenfeld‐Tanenbaum and Toronto General Hospital Research InstitutesTorontoOntarioCanada
| | - Katherine A. Siminovitch
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
- Mount Sinai Hospital, Lunenfeld‐Tanenbaum and Toronto General Hospital Research InstitutesTorontoOntarioCanada
| | - Alan C. Peterson
- Department of Human GeneticsMcGill UniversityMontrealQuebecCanada
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada
- Gerald Bronfman Department of OncologyMcGill UniversityQuebecCanada
| |
Collapse
|
22
|
Marshall-Phelps KL, Almeida R. Axonal neurotransmitter release in the regulation of myelination. Biosci Rep 2024; 44:BSR20231616. [PMID: 39230890 PMCID: PMC11427734 DOI: 10.1042/bsr20231616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/05/2024] Open
Abstract
Myelination of axons is a key determinant of fast action potential propagation, axonal health and circuit function. Previously considered a static structure, it is now clear that myelin is dynamically regulated in response to neuronal activity in the central nervous system (CNS). However, how activity-dependent signals are conveyed to oligodendrocytes remains unclear. Here, we review the potential mechanisms by which neurons could communicate changing activity levels to myelin, with a focus on the accumulating body of evidence to support activity-dependent vesicular signalling directly onto myelin sheaths. We discuss recent in vivo findings of activity-dependent fusion of neurotransmitter vesicles from non-synaptic axonal sites, and how modulation of this vesicular fusion regulates the stability and growth of myelin sheaths. We also consider the potential mechanisms by which myelin could sense and respond to axon-derived signals to initiate remodelling, and the relevance of these adaptations for circuit function. We propose that axonal vesicular signalling represents an important and underappreciated mode of communication by which neurons can transmit activity-regulated signals to myelinating oligodendrocytes and, potentially, more broadly to other cell types in the CNS.
Collapse
Affiliation(s)
- Katy L.H. Marshall-Phelps
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, U.K
- MS Society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, U.K
| | - Rafael G. Almeida
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, U.K
- MS Society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
23
|
Li J, Yang F, Tian Y, Wang Z, Qi D, Yang Z, Song J, Ding J, Wang X, Zhang Z. Lateral/caudal ganglionic eminence makes limited contribution to cortical oligodendrocytes. eLife 2024; 13:RP94317. [PMID: 39259197 PMCID: PMC11390106 DOI: 10.7554/elife.94317] [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: 09/12/2024] Open
Abstract
The emergence of myelinating oligodendrocytes represents a pivotal developmental milestone in vertebrates, given their capacity to ensheath axons and facilitate the swift conduction of action potentials. It is widely accepted that cortical oligodendrocyte progenitor cells (OPCs) arise from medial ganglionic eminence (MGE), lateral/caudal ganglionic eminence (LGE/CGE), and cortical radial glial cells (RGCs). Here, we used two different fate mapping strategies to challenge the established notion that the LGE generates cortical OPCs. Furthermore, we used a Cre/loxP-dependent exclusion strategy to reveal that the LGE/CGE does not give rise to cortical OPCs. Additionally, we showed that specifically eliminating MGE-derived OPCs leads to a significant reduction of cortical OPCs. Together, our findings indicate that the LGE does not generate cortical OPCs, contrary to previous beliefs. These findings provide a new view of the developmental origins of cortical OPCs and a valuable foundation for future research on both normal development and oligodendrocyte-related disease.
Collapse
Affiliation(s)
- Jialin Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Feihong Yang
- Department of Anesthesiology, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Tian
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ziwu Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dashi Qi
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiangang Song
- Department of Anesthesiology, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Zota I, Chanoumidou K, Gravanis A, Charalampopoulos I. Stimulating myelin restoration with BDNF: a promising therapeutic approach for Alzheimer's disease. Front Cell Neurosci 2024; 18:1422130. [PMID: 39285941 PMCID: PMC11402763 DOI: 10.3389/fncel.2024.1422130] [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: 04/23/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Alzheimer's Disease (AD) is a chronic neurodegenerative disorder constituting the most common form of dementia (60%-70% of cases). Although AD presents majorly a neurodegenerative pathology, recent clinical evidence highlights myelin impairment as a key factor in disease pathogenesis. The lack of preventive or restorative treatment is emphasizing the need to develop novel therapeutic approaches targeting to the causes of the disease. Recent studies in animals and patients have highlighted the loss of myelination of the neuronal axons as an extremely aggravating factor in AD, in addition to the formation of amyloid plaques and neurofibrillary tangles that are to date the main pathological hallmarks of the disease. Myelin breakdown represents an early stage event in AD. However, it is still unclear whether myelin loss is attributed only to exogenous factors like inflammatory processes of the tissue or to impaired oligodendrogenesis as well. Neurotrophic factors are well established protective molecules under many pathological conditions of the neural tissue, contributing also to proper myelination. Due to their inability to be used as drugs, many research efforts are focused on substituting neurotrophic activity with small molecules. Our research team has recently developed novel micromolecular synthetic neurotrophin mimetics (MNTs), selectively acting on neurotrophin receptors, and thus offering a unique opportunity for innovative therapies against neurodegenerative diseases. These small sized, lipophilic molecules address the underlying biological effect of these diseases (neuroprotective action), but also they exert significant neurogenic actions inducing neuronal replacement of the disease areas. One of the significant neurotrophin molecules in the Central Nervous System is Brain-Derived-Neurotrophin-Factor (BDNF). BDNF is a neurotrophin that not only supports neuroprotection and adult neurogenesis, but also mediates pro-myelinating effects in the CNS. BDNF binds with high-affinity on the TrkB neurotrophin receptor and enhances myelination by increasing the density of oligodendrocyte progenitor cells (OPCs) and playing an important role in CNS myelination. Conclusively, in the present review, we discuss the myelin pathophysiology in Alzheimer's Diseases, as well as the role of neurotrophins, and specifically BDNF, in myelin maintenance and restoration, revealing its valuable therapeutic potential against AD.
Collapse
Affiliation(s)
- Ioanna Zota
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| | - Konstantina Chanoumidou
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| |
Collapse
|
25
|
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
|
26
|
Yu Q, Zhang L, Xu T, Shao J, Yuan F, Yang Z, Wu Y, Lyu H. Oligodendroglia-to-pericyte conversion after lipopolysaccharide exposure is gender-dependent. PLoS One 2024; 19:e0308132. [PMID: 39106252 DOI: 10.1371/journal.pone.0308132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/17/2024] [Indexed: 08/09/2024] Open
Abstract
To investigate the sex-dependent differentiation of Sox10 cells and their response to pathological conditions such as lipopolysaccharide (LPS) exposure or ischemia, we utilized Sox10 Cre-ERT2, tdTomato mice. Tamoxifen administration induced the expression of red fluorescent protein (RFP) in these cells, facilitating their subsequent tracking and analysis after LPS injection and ischemia via immunofluorescence staining. Propidium iodide (PI) was injected to label necrotic cells following LPS administration. We found that the conversion of Sox10 cells to pericytes in female mice was significantly higher than in male mice, especially in those exposed to LPS. After LPS injection, the number of PI+ necrotic cells were significantly greater in females than in males. Moreover, RFP+ cells did not co-localize with glial fibrillary acidic protein (GFAP) or cluster of differentiation 11b (CD11b). Similarly, after brain ischemia, RFP+ cells did not express cluster of differentiation 13 (CD13), neuronal nuclei (NeuN), GFAP, or ionised calcium binding adaptor molecule 1 (Iba-1). These findings indicate that the conversion of Sox10 cells to pericytes following LPS exposure is sex-dependent, with neither male nor female groups showing differentiation into other cell types after LPS exposure or under ischemic conditions. The differences in LPS-induced necrosis of pericytes between sexes may explain the variations in the conversion of Sox10 cells to pericytes in both sexes.
Collapse
Affiliation(s)
- Qingting Yu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Linyuan Zhang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Xu
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Jiapeng Shao
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Falei Yuan
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Zuisu Yang
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Yuncheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyan Lyu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Jacquens A, Csaba Z, Soleimanzad H, Bokobza C, Delmotte PR, Userovici C, Boussemart P, Chhor V, Bouvier D, van de Looij Y, Faivre V, Diao S, Lemoine S, Blugeon C, Schwendimann L, Young-Ten P, Naffaa V, Laprevote O, Tanter M, Dournaud P, Van Steenwinckel J, Degos V, Gressens P. Deleterious effect of sustained neuroinflammation in pediatric traumatic brain injury. Brain Behav Immun 2024; 120:99-116. [PMID: 38705494 DOI: 10.1016/j.bbi.2024.04.029] [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: 11/28/2023] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024] Open
Abstract
INTRODUCTION Despite improved management of traumatic brain injury (TBI), it still leads to lifelong sequelae and disability, particularly in children. Chronic neuroinflammation (the so-called tertiary phase), in particular, microglia/macrophage and astrocyte reactivity, is among the main mechanisms suspected of playing a role in the generation of lesions associated with TBI. The role of acute neuroinflammation is now well understood, but its persistent effect and impact on the brain, particularly during development, are not. Here, we investigated the long-term effects of pediatric TBI on the brain in a mouse model. METHODS Pediatric TBI was induced in mice on postnatal day (P) 7 by weight-drop trauma. The time course of neuroinflammation and myelination was examined in the TBI mice. They were also assessed by magnetic resonance, functional ultrasound, and behavioral tests at P45. RESULTS TBI induced robust neuroinflammation, characterized by acute microglia/macrophage and astrocyte reactivity. The long-term consequences of pediatric TBI studied on P45 involved localized scarring astrogliosis, persistent microgliosis associated with a specific transcriptomic signature, and a long-lasting myelination defect consisting of the loss of myelinated axons, a decreased level of myelin binding protein, and severe thinning of the corpus callosum. These results were confirmed by reduced fractional anisotropy, measured by diffusion tensor imaging, and altered inter- and intra-hemispheric connectivity, measured by functional ultrasound imaging. In addition, adolescent mice with pediatric TBI showed persistent social interaction deficits and signs of anxiety and depressive behaviors. CONCLUSIONS We show that pediatric TBI induces tertiary neuroinflammatory processes associated with white matter lesions and altered behavior. These results support our model as a model for preclinical studies for tertiary lesions following TBI.
Collapse
Affiliation(s)
- Alice Jacquens
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France; Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anaesthesiology and Critical Care Medicine, Pitié-Salpêtrière Hospital, 47-83, boulevard de l'Hôpital, 75013 Paris, France.
| | - Zsolt Csaba
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France
| | - Haleh Soleimanzad
- Physics for Medicine Paris, Inserm, ESPCI Paris, PSL Research University, CNRS, 75005 Paris, France
| | - Cindy Bokobza
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France
| | | | | | | | - Vibol Chhor
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France
| | - Damien Bouvier
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France
| | - Yohan van de Looij
- Université de Genève, Service Développement et Croissance, Département de Pédiatrie, Faculté de Médecine, 1211 Genève, Suisse; Centre d'Imagerie Biomédicale, Section Technologie d'Imagerie Animale, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Suisse
| | - Valérie Faivre
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France
| | - Siaho Diao
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France
| | - Sophie Lemoine
- Genomics Core Facility, Département de Biologie, École Normale Supérieure, Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Université PSL, Paris, France
| | - Corinne Blugeon
- Genomics Core Facility, Département de Biologie, École Normale Supérieure, Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Université PSL, Paris, France
| | | | | | - Vanessa Naffaa
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France
| | - Olivier Laprevote
- Université de Paris, CNRS, CiTCoM, 75006 Paris, France; Hôpital Européen Georges Pompidou, AP-HP, Service de Biochimie, 75015 Paris, France
| | - Mickael Tanter
- Physics for Medicine Paris, Inserm, ESPCI Paris, PSL Research University, CNRS, 75005 Paris, France
| | - Pascal Dournaud
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France
| | | | - Vincent Degos
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France; Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anaesthesiology and Critical Care Medicine, Pitié-Salpêtrière Hospital, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France
| |
Collapse
|
28
|
He Y, Xie H, Xu Z, Zhang L, Feng Y, Long Y, Wang S, He Y, Li J, Zou Y, Zheng W, Xiao L. Rapid and prolonged response of oligodendrocyte lineage cells in standard acute cuprizone demyelination model revealed by in situ hybridization. Neurosci Lett 2024; 836:137869. [PMID: 38852766 DOI: 10.1016/j.neulet.2024.137869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Dietary administration of a copper chelator, cuprizone (CPZ), has long been reported to induce intense and reproducible demyelination of several brain structures such as the corpus callosum. Despite the widespread use of CPZ as an animal model for demyelinating diseases such as multiple sclerosis (MS), the mechanism by which it induces demyelination and then allows robust remyelination is still unclear. An intensive mapping of the cell dynamics of oligodendrocyte (OL) lineage during the de- and remyelination course would be particularly important for a deeper understanding of this model. Here, using a panel of OL lineage cell markers as in situ hybridization (ISH) probes, including Pdgfra, Plp, Mbp, Mog, Enpp6, combined with immunofluorescence staining of CC1, SOX10, we provide a detailed dynamic profile of OL lineage cells during the entire course of the model from 1, 2, 3.5 days, 1, 2, 3, 4,5 weeks of CPZ treatment, as well as after 1, 2, 3, 4 weeks of recovery from CPZ treatment. The result showed an unexpected early death of mature OLs and response of OL progenitor cells (OPCs) in vivo upon CPZ challenge, and a prolonged upregulation of myelin-forming OLs compared to the intact control even 4 weeks after CPZ withdrawal. These data may serve as a basic reference system for future studies of the effects of any intervention on de- and remyelination using the CPZ model, and imply the need to optimize the timing windows for the introduction of pro-remyelination therapies in demyelinating diseases such as 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
| | - Hua Xie
- 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
| | - Liuning Zhang
- 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
| | - Yuanyu Feng
- 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
| | - Yu Long
- 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
| | - 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
| | - 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
| | - Yanping Zou
- 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
| | - Wei Zheng
- 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
|
29
|
Guardiola-Diaz HM, DiBenedictis BT, Prendaj E, Bansal R. Diverse Responses of Oligodendrocytes to Different FGF-Family Members: Uncoupling Structure-Function Relationship Within FGF Subfamilies. ASN Neuro 2024; 16:2371163. [PMID: 39024549 PMCID: PMC11262039 DOI: 10.1080/17590914.2024.2371163] [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: 11/16/2023] [Accepted: 03/01/2024] [Indexed: 07/20/2024] Open
Abstract
The fifteen canonical paracrine fibroblast growth factors (FGFs) are organized in five subfamilies that interact with four FGF-receptors (FGFRs) and heparan sulfate proteoglycan (HSPG) co-receptors. Many of these FGFs are expressed in CNS regions where oligodendrocyte (OL) progenitors originate, migrate or differentiate. FGF2 (basic FGF) is considered a prototype FGF and the information about the effects of FGF signaling on OL-lineage cells has evolved largely from the study of FGF2. However, other FGFs from four subfamilies ((FGF1 (FGF1,-2), FGF4 (FGF4,-5,-6), FGF8 (FGF8,-17,-18) and FGF9 (FGF9,-16,-20)) that can interact with the isoforms of FGFRs expressed in OL-lineage cells may also play important roles. We previously reported OL-responses to FGF8 family members. Here, we investigate the effects of members of the FGF1,-4, and -9 subfamilies on proliferation and differentiation of OL progenitors (OPCs), and on cell cycle re-entry and down-regulation of myelin proteins by mature OLs. We found that while FGF2 induced all these responses strongly, FGF4,-6,-9 could do so only transiently and in the presence of exogenous HSPGs, and that FGF5,-16,-20 could not do so even in the presence of heparin or at higher concentrations. Furthermore, we noted that structurally similar FGFs within subfamilies did not always show similarities in their biological effects on OL-lineage cells. Taken together, these studies reveal that FGFs differ in the way they regulate the OL-lineage cells, emphasizes the selectivity and importance of HSPGs as FGF co-receptors in OL-lineage cells and suggests that structural similarity among FGF-subfamily members may not always predict their overlapping biological functions.
Collapse
Affiliation(s)
- Hebe M Guardiola-Diaz
- Department of Biology and Neuroscience Program, Trinity College, Hartford, Connecticut, USA
| | - Brett T DiBenedictis
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut, USA
| | - Erealda Prendaj
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut, USA
| | - Rashmi Bansal
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut, USA
| |
Collapse
|
30
|
Głowacka P, Oszajca K, Pudlarz A, Szemraj J, Witusik-Perkowska M. Postbiotics as Molecules Targeting Cellular Events of Aging Brain-The Role in Pathogenesis, Prophylaxis and Treatment of Neurodegenerative Diseases. Nutrients 2024; 16:2244. [PMID: 39064687 PMCID: PMC11279795 DOI: 10.3390/nu16142244] [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] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Aging is the most prominent risk factor for neurodegeneration occurrence. The most common neurodegenerative diseases (NDs), Alzheimer's (AD) and Parkinson's (PD) diseases, are characterized by the incidence of proteinopathy, abnormal activation of glial cells, oxidative stress, neuroinflammation, impaired autophagy and cellular senescence excessive for the patient's age. Moreover, mitochondrial disfunction, epigenetic alterations and neurogenesis inhibition, together with increased blood-brain barrier permeability and gut dysbiosis, have been linked to ND pathogenesis. Since NDs still lack curative treatment, recent research has sought therapeutic options in restoring gut microbiota and supplementing probiotic bacteria-derived metabolites with beneficial action to the host-so called postbiotics. The current review focuses on literature explaining cellular mechanisms involved in ND pathogenesis and research addressing the impact that postbiotics as a whole mixture and particular metabolites, such as short-chain fatty acids (SCFAs), lactate, polyamines, polyphenols, tryptophan metabolites, exopolysaccharides and bacterial extracellular vesicles, have on the ageing-associated processes underlying ND occurrence. The review also discusses the issue of implementing postbiotics into ND prophylaxis and therapy, depicting them as compounds addressing senescence-triggered dysfunctions that are worth translating from bench to pharmaceutical market in response to "silver consumers" demands.
Collapse
Affiliation(s)
- Pola Głowacka
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland; (P.G.); (K.O.); (A.P.); (J.S.)
- International Doctoral School, Medical University of Lodz, 90-419 Lodz, Poland
| | - Katarzyna Oszajca
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland; (P.G.); (K.O.); (A.P.); (J.S.)
| | - Agnieszka Pudlarz
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland; (P.G.); (K.O.); (A.P.); (J.S.)
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland; (P.G.); (K.O.); (A.P.); (J.S.)
| | - Monika Witusik-Perkowska
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland; (P.G.); (K.O.); (A.P.); (J.S.)
| |
Collapse
|
31
|
Wang J, Yang L, Du Y, Wang J, Weng Q, Liu X, Nicholson E, Xin M, Lu QR. BRG1 programs PRC2-complex repression and controls oligodendrocyte differentiation and remyelination. J Cell Biol 2024; 223:e202310143. [PMID: 38652118 PMCID: PMC11040499 DOI: 10.1083/jcb.202310143] [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: 10/30/2023] [Revised: 03/01/2024] [Accepted: 03/28/2024] [Indexed: 04/25/2024] Open
Abstract
Chromatin-remodeling protein BRG1/SMARCA4 is pivotal for establishing oligodendrocyte (OL) lineage identity. However, its functions for oligodendrocyte-precursor cell (OPC) differentiation within the postnatal brain and during remyelination remain elusive. Here, we demonstrate that Brg1 loss profoundly impairs OPC differentiation in the brain with a comparatively lesser effect in the spinal cord. Moreover, BRG1 is critical for OPC remyelination after injury. Integrative transcriptomic/genomic profiling reveals that BRG1 exhibits a dual role by promoting OPC differentiation networks while repressing OL-inhibitory cues and proneuronal programs. Furthermore, we find that BRG1 interacts with EED/PRC2 polycomb-repressive-complexes to enhance H3K27me3-mediated repression at gene loci associated with OL-differentiation inhibition and neurogenesis. Notably, BRG1 depletion decreases H3K27me3 deposition, leading to the upregulation of BMP/WNT signaling and proneurogenic genes, which suppresses OL programs. Thus, our findings reveal a hitherto unexplored spatiotemporal-specific role of BRG1 for OPC differentiation in the developing CNS and underscore a new insight into BRG1/PRC2-mediated epigenetic regulation that promotes and safeguards OL lineage commitment and differentiation.
Collapse
Affiliation(s)
- Jiajia Wang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Lijun Yang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Yiwen Du
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jincheng Wang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xuezhao Liu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Eva Nicholson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Mei Xin
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Qing Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| |
Collapse
|
32
|
Zhou F, Guo YX, Gao R, Ji XY, Tang YX, Wang LB, Zhang Y, Li X. Quercetin regulates dendritic cell activation by targeting STAT4 in the treatment of experimental autoimmune encephalomyelitis. Toxicol Appl Pharmacol 2024; 488:116980. [PMID: 38823456 DOI: 10.1016/j.taap.2024.116980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/13/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Multiple sclerosis (MS) is a class of autoimmune diseases mainly caused by the immune system attacking the myelin sheath of the axons in the nervous system. Although the pathogenesis of MS is complex, studies have shown that dendritic cells (DCs) play a vital role in the pathogenesis of MS. Quercetin (QU) has a unique advantage in clinical application, especially for treating autoimmune diseases. However, the mechanism of QU in the treatment of experimental autoimmune encephalomyelitis (EAE) remains unclear. In this study, we explore the potential role of QU in EAE. Finally, we find that QU has anti-inflammatory activities and neural protective effects in EAE. The experimental results suggest that the cellular basis for QU's function is to inhibit the activation of DCs while modulating the Th17 cell differentiation in the co-culture system. Further, QU may target STAT4 to inhibit its activation in DCs. This work will be of great significance for the future development and utilization of QU.
Collapse
Affiliation(s)
- Fang Zhou
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yu-Xin Guo
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Rui Gao
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xiao-Yu Ji
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yu-Xuan Tang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Li-Bin Wang
- Huazhong University of Science and Technology Union Shenzhen Hospital/Shenzhen Nanshan Hospital, Shenzhen, China
| | - Yuan Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xing Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| |
Collapse
|
33
|
D'Antoni S, Spatuzza M, Bonaccorso CM, Catania MV. Role of fragile X messenger ribonucleoprotein 1 in the pathophysiology of brain disorders: a glia perspective. Neurosci Biobehav Rev 2024; 162:105731. [PMID: 38763180 DOI: 10.1016/j.neubiorev.2024.105731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Fragile X messenger ribonucleoprotein 1 (FMRP) is a widely expressed RNA binding protein involved in several steps of mRNA metabolism. Mutations in the FMR1 gene encoding FMRP are responsible for fragile X syndrome (FXS), a leading genetic cause of intellectual disability and autism spectrum disorder, and fragile X-associated tremor-ataxia syndrome (FXTAS), a neurodegenerative disorder in aging men. Although FMRP is mainly expressed in neurons, it is also present in glial cells and its deficiency or altered expression can affect functions of glial cells with implications for the pathophysiology of brain disorders. The present review focuses on recent advances on the role of glial subtypes, astrocytes, oligodendrocytes and microglia, in the pathophysiology of FXS and FXTAS, and describes how the absence or reduced expression of FMRP in these cells can impact on glial and neuronal functions. We will also briefly address the role of FMRP in radial glial cells and its effects on neural development, and gliomas and will speculate on the role of glial FMRP in other brain disorders.
Collapse
Affiliation(s)
- S D'Antoni
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy
| | - M Spatuzza
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy
| | - C M Bonaccorso
- Oasi Research Institute - IRCCS, via Conte Ruggero 73, Troina 94018, Italy
| | - M V Catania
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy.
| |
Collapse
|
34
|
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
|
35
|
Sheloukhova L, Watanabe H. Evolution of glial cells: a non-bilaterian perspective. Neural Dev 2024; 19:10. [PMID: 38907299 PMCID: PMC11193209 DOI: 10.1186/s13064-024-00184-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 06/06/2024] [Indexed: 06/23/2024] Open
Abstract
Nervous systems of bilaterian animals generally consist of two cell types: neurons and glial cells. Despite accumulating data about the many important functions glial cells serve in bilaterian nervous systems, the evolutionary origin of this abundant cell type remains unclear. Current hypotheses regarding glial evolution are mostly based on data from model bilaterians. Non-bilaterian animals have been largely overlooked in glial studies and have been subjected only to morphological analysis. Here, we provide a comprehensive overview of conservation of the bilateral gliogenic genetic repertoire of non-bilaterian phyla (Cnidaria, Placozoa, Ctenophora, and Porifera). We overview molecular and functional features of bilaterian glial cell types and discuss their possible evolutionary history. We then examine which glial features are present in non-bilaterians. Of these, cnidarians show the highest degree of gliogenic program conservation and may therefore be crucial to answer questions about glial evolution.
Collapse
Affiliation(s)
- Larisa Sheloukhova
- Evolutionary Neurobiology Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0412, Japan
| | - Hiroshi Watanabe
- Evolutionary Neurobiology Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0412, Japan.
| |
Collapse
|
36
|
Graham JH, Schlachetzki JCM, Yang X, Breuss MW. Genomic Mosaicism of the Brain: Origin, Impact, and Utility. Neurosci Bull 2024; 40:759-776. [PMID: 37898991 PMCID: PMC11178748 DOI: 10.1007/s12264-023-01124-8] [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: 05/04/2023] [Accepted: 07/16/2023] [Indexed: 10/31/2023] Open
Abstract
Genomic mosaicism describes the phenomenon where some but not all cells within a tissue harbor unique genetic mutations. Traditionally, research focused on the impact of genomic mosaicism on clinical phenotype-motivated by its involvement in cancers and overgrowth syndromes. More recently, we increasingly shifted towards the plethora of neutral mosaic variants that can act as recorders of cellular lineage and environmental exposures. Here, we summarize the current state of the field of genomic mosaicism research with a special emphasis on our current understanding of this phenomenon in brain development and homeostasis. Although the field of genomic mosaicism has a rich history, technological advances in the last decade have changed our approaches and greatly improved our knowledge. We will provide current definitions and an overview of contemporary detection approaches for genomic mosaicism. Finally, we will discuss the impact and utility of genomic mosaicism.
Collapse
Affiliation(s)
- Jared H Graham
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, 80045-2581, CO, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, 92093-0021, San Diego, CA, USA
| | - Xiaoxu Yang
- Department of Neurosciences, University of California San Diego, La Jolla, 92093-0021, San Diego, CA, USA
- Rady Children's Institute for Genomic Medicine, San Diego, 92123, CA, USA
| | - Martin W Breuss
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, 80045-2581, CO, USA.
| |
Collapse
|
37
|
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
|
38
|
Huang D, Mela A, Bhanu NV, Garcia BA, Canoll P, Casaccia P. PDGF-BB overexpression in p53 null oligodendrocyte progenitors increases H3K27me3 and induces transcriptional changes which favor proliferation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594214. [PMID: 38798631 PMCID: PMC11118351 DOI: 10.1101/2024.05.14.594214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Proneural gliomas are brain tumors characterized by enrichment of oligodendrocyte progenitor cell (OPC) transcripts and genetic alterations. In this study we sought to identify transcriptional and epigenetic differences between OPCs with Trp53 deletion and PDGF-BB overexpression (BB-p53n), which form tumors when transplanted in mouse brains, and those carrying only p53 deletion (p53n), which do not. We used unbiased histone proteomics and RNA-seq analysis on these two genetically modified OPC populations and detected higher levels of H3K27me3 in BB-p53n compared to p53n OPCs. The BB-p53n OPC were characterized by higher levels of transcripts related to proliferation and lower levels of those related to differentiation. Pharmacological inhibition of histone H3K27 trimethylation in BB-p53n OPC reduced cell cycle transcripts and increased the expression of differentiation markers. These data suggest that PDGF-BB overexpression in p53 null OPC results in histone post-translational modifications and consequent transcriptional changes favoring proliferation while halting differentiation, thereby promoting the early stages of transformation.
Collapse
|
39
|
Starikov L, Ghinia-Tegla M, Kottmann AH. Lateral Motor Column specific expression of Sonic Hedgehog contributes to maintenance and scaling of pMN progenitor cell populations during oligodendrogenesis. RESEARCH SQUARE 2024:rs.3.rs-4249282. [PMID: 38798533 PMCID: PMC11118686 DOI: 10.21203/rs.3.rs-4249282/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Motor neurons (MNs) and oligodendrocyte precursor cells (OPCs) emerge sequentially from the pMN precursor domain during spinal cord development. MNs diversify into muscle specific subtypes and settle in stereotypic locations in the ventral horns. In contrast, OPCs are mobile and appear to evenly populate the parenchyma. Whether earlier born MNs influence OPC production is controversial. We found that Sonic Hedgehog signaling emanating from nascent MNs of the lateral motor column is critical for maintaining a larger and more yielding pMN domain at limb levels compared to trunk levels during OPC production. Reduced Shh signaling resulted in unrecoverable diminishment of pMN domain based OPC production leaving the spinal cord impoverished of OPC. Our results suggest that production of OPC at limb levels is contingent on completion of MN production.
Collapse
Affiliation(s)
- Lev Starikov
- City University of New York School of Medicine (CSOM), Dept. of Molecular, Cellular and Biomedical Sciences, New York City, NY 10031, USA
- City University of New York Graduate Center, Molecular, Cellular, and Developmental Biology Subprogram, New York City, NY 10016, USA
| | | | - Andreas H. Kottmann
- City University of New York School of Medicine (CSOM), Dept. of Molecular, Cellular and Biomedical Sciences, New York City, NY 10031, USA
- City University of New York Graduate Center, Molecular, Cellular, and Developmental Biology Subprogram, New York City, NY 10016, USA
- City University of New York Graduate Center, Neuroscience Subprogram, New York City, NY 10016, USA3
| |
Collapse
|
40
|
Marziali LN, Hwang Y, Palmisano M, Cuenda A, Sim FJ, Gonzalez A, Volsko C, Dutta R, Trapp BD, Wrabetz L, Feltri ML. p38γ MAPK delays myelination and remyelination and is abundant in multiple sclerosis lesions. Brain 2024; 147:1871-1886. [PMID: 38128553 PMCID: PMC11068213 DOI: 10.1093/brain/awad421] [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: 10/21/2022] [Revised: 10/05/2023] [Accepted: 11/12/2023] [Indexed: 12/23/2023] Open
Abstract
Multiple sclerosis is a chronic inflammatory disease in which disability results from the disruption of myelin and axons. During the initial stages of the disease, injured myelin is replaced by mature myelinating oligodendrocytes that differentiate from oligodendrocyte precursor cells. However, myelin repair fails in secondary and chronic progressive stages of the disease and with ageing, as the environment becomes progressively more hostile. This may be attributable to inhibitory molecules in the multiple sclerosis environment including activation of the p38MAPK family of kinases. We explored oligodendrocyte precursor cell differentiation and myelin repair using animals with conditional ablation of p38MAPKγ from oligodendrocyte precursors. We found that p38γMAPK ablation accelerated oligodendrocyte precursor cell differentiation and myelination. This resulted in an increase in both the total number of oligodendrocytes and the migration of progenitors ex vivo and faster remyelination in the cuprizone model of demyelination/remyelination. Consistent with its role as an inhibitor of myelination, p38γMAPK was significantly downregulated as oligodendrocyte precursor cells matured into oligodendrocytes. Notably, p38γMAPK was enriched in multiple sclerosis lesions from patients. Oligodendrocyte progenitors expressed high levels of p38γMAPK in areas of failed remyelination but did not express detectable levels of p38γMAPK in areas where remyelination was apparent. Our data suggest that p38γ could be targeted to improve myelin repair in multiple sclerosis.
Collapse
Affiliation(s)
- Leandro N Marziali
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Yoonchan Hwang
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Marilena Palmisano
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Ana Cuenda
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid 28049, Spain
| | - Fraser J Sim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Alberto Gonzalez
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Christina Volsko
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lawrence Wrabetz
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Maria L Feltri
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
- Università degli studi di Milano, Biometra department and IRCcs Carlo Besta, Milano 20133, Italy
| |
Collapse
|
41
|
Poggi G, Klaus F, Pryce CR. Pathophysiology in cortico-amygdala circuits and excessive aversion processing: the role of oligodendrocytes and myelination. Brain Commun 2024; 6:fcae140. [PMID: 38712320 PMCID: PMC11073757 DOI: 10.1093/braincomms/fcae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/27/2023] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Stress-related psychiatric illnesses, such as major depressive disorder, anxiety and post-traumatic stress disorder, present with alterations in emotional processing, including excessive processing of negative/aversive stimuli and events. The bidirectional human/primate brain circuit comprising anterior cingulate cortex and amygdala is of fundamental importance in processing emotional stimuli, and in rodents the medial prefrontal cortex-amygdala circuit is to some extent analogous in structure and function. Here, we assess the comparative evidence for: (i) Anterior cingulate/medial prefrontal cortex<->amygdala bidirectional neural circuits as major contributors to aversive stimulus processing; (ii) Structural and functional changes in anterior cingulate cortex<->amygdala circuit associated with excessive aversion processing in stress-related neuropsychiatric disorders, and in medial prefrontal cortex<->amygdala circuit in rodent models of chronic stress-induced increased aversion reactivity; and (iii) Altered status of oligodendrocytes and their oligodendrocyte lineage cells and myelination in anterior cingulate/medial prefrontal cortex<->amygdala circuits in stress-related neuropsychiatric disorders and stress models. The comparative evidence from humans and rodents is that their respective anterior cingulate/medial prefrontal cortex<->amygdala circuits are integral to adaptive aversion processing. However, at the sub-regional level, the anterior cingulate/medial prefrontal cortex structure-function analogy is incomplete, and differences as well as similarities need to be taken into account. Structure-function imaging studies demonstrate that these neural circuits are altered in both human stress-related neuropsychiatric disorders and rodent models of stress-induced increased aversion processing. In both cases, the changes include altered white matter integrity, albeit the current evidence indicates that this is decreased in humans and increased in rodent models. At the cellular-molecular level, in both humans and rodents, the current evidence is that stress disorders do present with changes in oligodendrocyte lineage, oligodendrocytes and/or myelin in these neural circuits, but these changes are often discordant between and even within species. Nonetheless, by integrating the current comparative evidence, this review provides a timely insight into this field and should function to inform future studies-human, monkey and rodent-to ascertain whether or not the oligodendrocyte lineage and myelination are causally involved in the pathophysiology of stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Giulia Poggi
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, CH-8008 Zurich, Switzerland
| | - Federica Klaus
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA
- Desert-Pacific Mental Illness Research Education and Clinical Center, VA San Diego Healthcare System, San Diego, CA 92093, USA
| | - Christopher R Pryce
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, CH-8008 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
- URPP Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
42
|
Lim YA, Tan LS, Lee WT, Sim WL, Lv Y, Takakuni M, Saito S, Ihara M, Arumugam TV, Chen C, Wong FWS, Dawe GS. Hope for vascular cognitive impairment: Ac-YVAD-cmk as a novel treatment against white matter rarefaction. PLoS One 2024; 19:e0299703. [PMID: 38630707 PMCID: PMC11023579 DOI: 10.1371/journal.pone.0299703] [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: 09/27/2023] [Accepted: 02/14/2024] [Indexed: 04/19/2024] Open
Abstract
Vascular cognitive impairment (VCI) is the second leading cause of dementia with limited treatment options, characterised by cerebral hypoperfusion-induced white matter rarefaction (WMR). Subcortical VCI is the most common form of VCI, but the underlying reasons for region susceptibility remain elusive. Recent studies employing the bilateral cortical artery stenosis (BCAS) method demonstrate that various inflammasomes regulate white matter injury and blood-brain barrier dysfunction but whether caspase-1 inhibition will be beneficial remains unclear. To address this, we performed BCAS on C57/BL6 mice to study the effects of Ac-YVAD-cmk, a caspase-1 inhibitor, on the subcortical and cortical regions. Cerebral blood flow (CBF), WMR, neuroinflammation and the expression of tight junction-related proteins associated with blood-brain barrier integrity were assessed 15 days post BCAS. We observed that Ac-YVAD-cmk restored CBF, attenuated BCAS-induced WMR and restored subcortical myelin expression. Within the subcortical region, BCAS activated the NLRP3/caspase-1/interleukin-1beta axis only within the subcortical region, which was attenuated by Ac-YVAD-cmk. Although we observed that BCAS induced significant increases in VCAM-1 expression in both brain regions that were attenuated with Ac-YVAD-cmk, only ZO-1 and occludin were observed to be significantly altered in the subcortical region. Here we show that caspase-1 may contribute to subcortical regional susceptibility in a mouse model of VCI. In addition, our results support further investigations into the potential of Ac-YVAD-cmk as a novel treatment strategy against subcortical VCI and other conditions exhibiting cerebral hypoperfusion-induced WMR.
Collapse
Affiliation(s)
- Yun-An Lim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Li Si Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Thye Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Liang Sim
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Yang Lv
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Maki Takakuni
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
- Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | | | - Christopher Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Fred Wai-Shiu Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gavin Stewart Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
43
|
Khelfaoui H, Ibaceta-Gonzalez C, Angulo MC. Functional myelin in cognition and neurodevelopmental disorders. Cell Mol Life Sci 2024; 81:181. [PMID: 38615095 PMCID: PMC11016012 DOI: 10.1007/s00018-024-05222-2] [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: 12/08/2023] [Revised: 03/18/2024] [Accepted: 03/30/2024] [Indexed: 04/15/2024]
Abstract
In vertebrates, oligodendrocytes (OLs) are glial cells of the central nervous system (CNS) responsible for the formation of the myelin sheath that surrounds the axons of neurons. The myelin sheath plays a crucial role in the transmission of neuronal information by promoting the rapid saltatory conduction of action potentials and providing neurons with structural and metabolic support. Saltatory conduction, first described in the peripheral nervous system (PNS), is now generally recognized as a universal evolutionary innovation to respond quickly to the environment: myelin helps us think and act fast. Nevertheless, the role of myelin in the central nervous system, especially in the brain, may not be primarily focused on accelerating conduction speed but rather on ensuring precision. Its principal function could be to coordinate various neuronal networks, promoting their synchronization through oscillations (or rhythms) relevant for specific information processing tasks. Interestingly, myelin has been directly involved in different types of cognitive processes relying on brain oscillations, and myelin plasticity is currently considered to be part of the fundamental mechanisms for memory formation and maintenance. However, despite ample evidence showing the involvement of myelin in cognition and neurodevelopmental disorders characterized by cognitive impairments, the link between myelin, brain oscillations, cognition and disease is not yet fully understood. In this review, we aim to highlight what is known and what remains to be explored to understand the role of myelin in high order brain processes.
Collapse
Affiliation(s)
- Hasni Khelfaoui
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France
| | - Cristobal Ibaceta-Gonzalez
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France
| | - Maria Cecilia Angulo
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France.
- GHU-PARIS Psychiatrie Et Neurosciences, Hôpital Sainte Anne, 75014, Paris, France.
| |
Collapse
|
44
|
Qi YB, Xu Z, Shen S, Wang Z, Wang Z. MYRF: A unique transmembrane transcription factor- from proteolytic self-processing to its multifaceted roles in animal development. Bioessays 2024; 46:e2300209. [PMID: 38488284 DOI: 10.1002/bies.202300209] [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: 10/27/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 03/28/2024]
Abstract
The Myelin Regulator Factor (MYRF) is a master regulator governing myelin formation and maintenance in the central nervous system. The conservation of MYRF across metazoans and its broad tissue expression suggest it has functions extending beyond the well-established role in myelination. Loss of MYRF results in developmental lethality in both invertebrates and vertebrates, and MYRF haploinsufficiency in humans causes MYRF-related Cardiac Urogenital Syndrome, underscoring its importance in animal development; however, these mechanisms are largely unexplored. MYRF, an unconventional transcription factor, begins embedded in the membrane and undergoes intramolecular chaperone mediated trimerization, which triggers self-cleavage, allowing its N-terminal segment with an Ig-fold DNA-binding domain to enter the nucleus for transcriptional regulation. Recent research suggests developmental regulation of cleavage, yet the mechanisms remain enigmatic. While some parts of MYRF's structure have been elucidated, others remain obscure, leaving questions about how these motifs are linked to its intricate processing and function.
Collapse
Affiliation(s)
- Yingchuan B Qi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhimin Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shiqian Shen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhao Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhizhi Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
45
|
Liu Y, Yuan J, Dong Y, Jiang S, Zhang M, Zhao X. Interaction between Oligodendrocytes and Interneurons in Brain Development and Related Neuropsychiatric Disorders. Int J Mol Sci 2024; 25:3620. [PMID: 38612430 PMCID: PMC11011273 DOI: 10.3390/ijms25073620] [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: 02/14/2024] [Revised: 03/17/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
A variety of neurological and psychiatric disorders have recently been shown to be highly associated with the abnormal development and function of oligodendrocytes (OLs) and interneurons. OLs are the myelin-forming cells in the central nervous system (CNS), while interneurons are important neural types gating the function of excitatory neurons. These two types of cells are of great significance for the establishment and function of neural circuits, and they share similar developmental origins and transcriptional architectures, and interact with each other in multiple ways during development. In this review, we compare the similarities and differences in these two cell types, providing an important reference and further revealing the pathogenesis of related brain disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Xianghui Zhao
- Department of Neuroscience, Air Force Medical University, Xi’an 710032, China
| |
Collapse
|
46
|
Stancu M, Wohlfrom H, Heß M, Grothe B, Leibold C, Kopp-Scheinpflug C. Ambient sound stimulation tunes axonal conduction velocity by regulating radial growth of myelin on an individual, axon-by-axon basis. Proc Natl Acad Sci U S A 2024; 121:e2316439121. [PMID: 38442165 PMCID: PMC10945791 DOI: 10.1073/pnas.2316439121] [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: 09/22/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Adaptive myelination is the emerging concept of tuning axonal conduction velocity to the activity within specific neural circuits over time. Sound processing circuits exhibit structural and functional specifications to process signals with microsecond precision: a time scale that is amenable to adjustment in length and thickness of myelin. Increasing activity of auditory axons by introducing sound-evoked responses during postnatal development enhances myelin thickness, while sensory deprivation prevents such radial growth during development. When deprivation occurs during adulthood, myelin thickness was reduced. However, it is unclear whether sensory stimulation adjusts myelination in a global fashion (whole fiber bundles) or whether such adaptation occurs at the level of individual fibers. Using temporary monaural deprivation in mice provided an internal control for a) differentially tracing structural changes in active and deprived fibers and b) for monitoring neural activity in response to acoustic stimulation of the control and the deprived ear within the same animal. The data show that sound-evoked activity increased the number of myelin layers around individual active axons, even when located in mixed bundles of active and deprived fibers. Thicker myelination correlated with faster axonal conduction velocity and caused shorter auditory brainstem response wave VI-I delays, providing a physiologically relevant readout. The lack of global compensation emphasizes the importance of balanced sensory experience in both ears throughout the lifespan of an individual.
Collapse
Affiliation(s)
- Mihai Stancu
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, Planegg-Martinsried82152, Germany
- Munich Cluster for Systems Neurology, Munich81377, Germany
- Graduate School of Systemic Neurosciences, Planegg-Martinsried82152, Germany
| | - Hilde Wohlfrom
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, Planegg-Martinsried82152, Germany
| | - Martin Heß
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, Planegg-Martinsried82152, Germany
| | - Benedikt Grothe
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, Planegg-Martinsried82152, Germany
- Munich Cluster for Systems Neurology, Munich81377, Germany
| | - Christian Leibold
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, Planegg-Martinsried82152, Germany
- Faculty of Biology, Bernstein Center Freiburg, BrainLinks-BrainTools, University of Freiburg, Freiburg im Breisgau79110, Germany
| | - Conny Kopp-Scheinpflug
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, Planegg-Martinsried82152, Germany
| |
Collapse
|
47
|
Cui SH, Suo N, Yang Y, Wu X, Guo SM, Xie X. The aminosteroid U73122 promotes oligodendrocytes generation and myelin formation. Acta Pharmacol Sin 2024; 45:490-501. [PMID: 37935896 PMCID: PMC10834981 DOI: 10.1038/s41401-023-01183-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/13/2023] [Indexed: 11/09/2023] Open
Abstract
Oligodendrocytes (OLs) are glial cells that ensheath neuronal axons and form myelin in the central nervous system (CNS). OLs are differentiated from oligodendrocyte precursor cells (OPCs) during development and myelin repair, which is often insufficient in the latter case in demyelinating diseases such as multiple sclerosis (MS). Many factors have been reported to regulate OPC-to-OL differentiation, including a number of G protein-coupled receptors (GPCRs). In an effort to search pathways downstream of GPCRs that might be involved in OPC differentiation, we discover that U73122, a phosphoinositide specific phospholipase C (PI-PLC) inhibitor, dramatically promotes OPC-to-OL differentiation and myelin regeneration in experimental autoimmune encephalomyelitis model. Unexpectedly, U73343, a close analog of U73122 which lacks PI-PLC inhibitory activity also promotes OL differentiation, while another reported PI-PLC inhibitor edelfosine does not have such effect, suggesting that U73122 and U73343 enhance OPC differentiation independent of PLC. Although the structures of U73122 and U73343 closely resemble 17β-estradiol, and both compounds do activate estrogen receptors Erα and Erβ with low efficacy and potency, further study indicates that these compounds do not act through Erα and/or Erβ to promote OPC differentiation. RNA-Seq and bioinformatic analysis indicate that U73122 and U73343 may regulate cholesterol biosynthesis. Further study shows both compounds increase 14-dehydrozymostenol, a steroid reported to promote OPC differentiation, in OPC culture. In conclusion, the aminosteroids U73122 and U73343 promote OPC-to-OL generation and myelin formation by regulating cholesterol biosynthesis pathway.
Collapse
Affiliation(s)
- Shi-Hao Cui
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Na Suo
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Ying Yang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xuan Wu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shi-Meng Guo
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xin Xie
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
48
|
Clark KJ, Lubin EE, Gonzalez EM, Sangree AK, Layo-Carris DE, Durham EL, Ahrens-Nicklas RC, Nomakuchi TT, Bhoj EJ. NeuroTri2-VISDOT: An open-access tool to harness the power of second trimester human single cell data to inform models of Mendelian neurodevelopmental disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578438. [PMID: 38352329 PMCID: PMC10862881 DOI: 10.1101/2024.02.01.578438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Whole exome and genome sequencing, coupled with refined bioinformatic pipelines, have enabled improved diagnostic yields for individuals with Mendelian conditions and have led to the rapid identification of novel syndromes. For many Mendelian neurodevelopmental disorders (NDDs), there is a lack of pre-existing model systems for mechanistic work. Thus, it is critical for translational researchers to have an accessible phenotype- and genotype-informed approach for model system selection. Single-cell RNA sequencing data can be informative in such an approach, as it can indicate which cell types express a gene of interest at the highest levels across time. For Mendelian NDDs, such data for the developing human brain is especially useful. A valuable single-cell RNA sequencing dataset of the second trimester developing human brain was produced by Bhaduri et al in 2021, but access to these data can be limited by computing power and the learning curve of single-cell data analysis. To reduce these barriers for translational research on Mendelian NDDs, we have built the web-based tool, Neurodevelopment in Trimester 2 - VIsualization of Single cell Data Online Tool (NeuroTri2-VISDOT), for exploring this single-cell dataset, and we have employed it in several different settings to demonstrate its utility for the translational research community.
Collapse
Affiliation(s)
- Kelly J. Clark
- Biomedical Graduate School, University of Pennsylvania, Perelman School of Medicine
- Children’s Hospital of Philadelphia
| | - Emily E. Lubin
- Biomedical Graduate School, University of Pennsylvania, Perelman School of Medicine
- Children’s Hospital of Philadelphia
| | - Elizabeth M. Gonzalez
- Biomedical Graduate School, University of Pennsylvania, Perelman School of Medicine
- Children’s Hospital of Philadelphia
| | - Annabel K. Sangree
- Biomedical Graduate School, University of Pennsylvania, Perelman School of Medicine
- Children’s Hospital of Philadelphia
| | | | | | - Rebecca C. Ahrens-Nicklas
- Children’s Hospital of Philadelphia
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine
| | | | - Elizabeth J. Bhoj
- Children’s Hospital of Philadelphia
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine
| |
Collapse
|
49
|
Mattera V, Occhiuzzi F, Correale J, Pasquini JM. Remyelinating effect driven by transferrin-loaded extracellular vesicles. Glia 2024; 72:338-361. [PMID: 37860913 DOI: 10.1002/glia.24478] [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: 02/11/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/21/2023]
Abstract
Extracellular vesicles (EVs) are involved in diverse cellular functions, playing a significant role in cell-to-cell communication in both physiological conditions and pathological scenarios. Therefore, EVs represent a promising therapeutic strategy. Oligodendrocytes (OLs) are myelinating glial cells developed from oligodendrocyte progenitor cells (OPCs) and damaged in chronic demyelinating diseases such as multiple sclerosis (MS). Glycoprotein transferrin (Tf) plays a critical role in iron homeostasis and has pro-differentiating effects on OLs in vivo and in vitro. In the current work, we evaluated the use of EVs as transporters of Tf to the central nervous system (CNS) through the intranasal (IN) route. For the in vitro mechanistic studies, we used rat plasma EVs. Our results show that EVTf enter OPCs through clathrin-caveolae and cholesterol-rich lipid raft endocytic pathways, releasing the cargo and exerting a pro-maturation effect on OPCs. These effects were also observed in vivo using the animal model of demyelination induced by cuprizone (CPZ). In this model, IN administered Tf-loaded EVs isolated from mouse plasma reached the brain parenchyma, internalizing into OPCs, promoting their differentiation, and accelerating remyelination. Furthermore, in vivo experiments demonstrated that EVs protected the Tf cargo and significantly reduced the amount of Tf required to induce remyelination as compared to soluble Tf. Collectively, these findings unveil EVs as functional nanocarriers of Tf to induce remyelination.
Collapse
Affiliation(s)
- Vanesa Mattera
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológica (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Federico Occhiuzzi
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológica (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Jorge Correale
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológica (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
- Departamento de Neurología, Fleni, Buenos Aires, Argentina
| | - Juana M Pasquini
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológica (IQUIFIB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| |
Collapse
|
50
|
Li J, Miramontes TG, Czopka T, Monk KR. Synaptic input and Ca 2+ activity in zebrafish oligodendrocyte precursor cells contribute to myelin sheath formation. Nat Neurosci 2024; 27:219-231. [PMID: 38216650 DOI: 10.1038/s41593-023-01553-8] [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: 03/25/2022] [Accepted: 12/13/2023] [Indexed: 01/14/2024]
Abstract
In the nervous system, only one type of neuron-glial synapse is known to exist: that between neurons and oligodendrocyte precursor cells (OPCs), yet their composition, assembly, downstream signaling and in vivo functions remain largely unclear. Here, we address these questions using in vivo microscopy in zebrafish spinal cord and identify postsynaptic molecules PSD-95 and gephyrin in OPCs. The puncta containing these molecules in OPCs increase during early development and decrease upon OPC differentiation. These puncta are highly dynamic and frequently assemble at 'hotspots'. Gephyrin hotspots and synapse-associated Ca2+ activity in OPCs predict where a subset of myelin sheaths forms in differentiated oligodendrocytes. Further analyses reveal that spontaneous synaptic release is integral to OPC Ca2+ activity, while evoked synaptic release contributes only in early development. Finally, disruption of the synaptic genes dlg4a/dlg4b, gphnb and nlgn3b impairs OPC differentiation and myelination. Together, we propose that neuron-OPC synapses are dynamically assembled and can predetermine myelination patterns through Ca2+ signaling.
Collapse
Affiliation(s)
- Jiaxing Li
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| | | | - Tim Czopka
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Kelly R Monk
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|