1
|
Sade AN, Wiener G, Barak B. Intersection of mitochondrial dysfunction and myelination: An overlooked aspect in neurodevelopmental disorders. Neural Regen Res 2026; 21:659-660. [PMID: 39995084 DOI: 10.4103/nrr.nrr-d-24-01025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/27/2024] [Indexed: 02/26/2025] Open
Affiliation(s)
- Ariel Nir Sade
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel (Sade AN, Wiener G, Barak B)
| | - Gal Wiener
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel (Sade AN, Wiener G, Barak B)
| | - Boaz Barak
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel (Sade AN, Wiener G, Barak B)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel (Barak B)
| |
Collapse
|
2
|
Wu S, Chen J. Is age-related myelinodegenerative change an initial risk factor of neurodegenerative diseases? Neural Regen Res 2026; 21:648-658. [PMID: 40326982 DOI: 10.4103/nrr.nrr-d-24-00848] [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: 07/29/2024] [Accepted: 11/25/2024] [Indexed: 05/07/2025] Open
Abstract
Myelination, the continuous ensheathment of neuronal axons, is a lifelong process in the nervous system that is essential for the precise, temporospatial conduction of action potentials between neurons. Myelin also provides intercellular metabolic support to axons. Even minor disruptions in the integrity of myelin can impair neural performance and increase susceptibility to neurological diseases. In fact, myelin degeneration is a well-known neuropathological condition that is associated with normal aging and several neurodegenerative diseases, including multiple sclerosis and Alzheimer's disease. In the central nervous system, compact myelin sheaths are formed by fully mature oligodendrocytes. However, the entire oligodendrocyte lineage is susceptible to changes in the biological microenvironment and other risk factors that arise as the brain ages. In addition to their well-known role in action potential propagation, oligodendrocytes also provide intercellular metabolic support to axons by transferring energy metabolites and delivering exosomes. Therefore, myelin degeneration in the aging central nervous system is a significant contributor to the development of neurodegenerative diseases. Interventions that mitigate age-related myelin degeneration can improve neurological function in aging individuals. In this review, we investigate the changes in myelin that are associated with aging and their underlying mechanisms. We also discuss recent advances in understanding how myelin degeneration in the aging brain contributes to neurodegenerative diseases and explore the factors that can prevent, slow down, or even reverse age-related myelin degeneration. Future research will enhance our understanding of how reducing age-related myelin degeneration can be used as a therapeutic target for delaying or preventing neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuangchan Wu
- Sanhang Institute for Brain Science and Technology (SiBST), School of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi Province, China
- Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, Guangdong Province, China
| | - Jun Chen
- Sanhang Institute for Brain Science and Technology (SiBST), School of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi Province, China
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
3
|
Lawrence AB, Brown SM, Bradford BM, Mabbott NA, Bombail V, Rutherford KMD. Non-neuronal brain biology and its relevance to animal welfare. Neurosci Biobehav Rev 2025; 173:106136. [PMID: 40185375 DOI: 10.1016/j.neubiorev.2025.106136] [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: 07/10/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Non-neuronal cells constitute a significant portion of brain tissue and are seen as having key roles in brain homeostasis and responses to challenges. This review illustrates how non-neuronal biology can bring new perspectives to animal welfare through understanding mechanisms that determine welfare outcomes and highlighting interventions to improve welfare. Most obvious in this respect is the largely unrecognised relevance of neuroinflammation to animal welfare which is increasingly found to have roles in determining how animals respond to challenges. We start by introducing non-neuronal cells and review their involvement in affective states and cognition often seen as core psychological elements of animal welfare. We find that the evidence for a causal involvement of glia in cognition is currently more advanced than the corresponding evidence for affective states. We propose that translational research on affective disorders could usefully apply welfare science derived approaches for assessing affective states. Using evidence from translational research, we illustrate the involvement of non-neuronal cells and neuroinflammatory processes as mechanisms modulating resilience to welfare challenges including disease, pain, and social stress. We review research on impoverished environments and environmental enrichment which suggests that environmental conditions which improve animal welfare also improve resilience to challenges through balancing pro- and anti-inflammatory non-neuronal processes. We speculate that non-neuronal biology has relevance to animal welfare beyond neuro-inflammation including facilitating positive affective states. We acknowledge the relevance of neuronal biology to animal welfare whilst proposing that non-neuronal biology provides additional and relevant insights to improve animals' lives.
Collapse
Affiliation(s)
- Alistair B Lawrence
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK; Scotland's Rural College (SRUC), Edinburgh EH9 3JG, UK.
| | - Sarah M Brown
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Barry M Bradford
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Neil A Mabbott
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | | | | |
Collapse
|
4
|
Murayama R, Cai Y, Nakamura H, Hashimoto K. Demyelination in psychiatric and neurological disorders: Mechanisms, clinical impact, and novel therapeutic strategies. Neurosci Biobehav Rev 2025; 174:106209. [PMID: 40368261 DOI: 10.1016/j.neubiorev.2025.106209] [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: 01/07/2025] [Revised: 04/14/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Demyelination, defined as the loss of myelin sheaths around neuronal axons, is increasingly recognized as a key factor in a broad range of psychiatric and neurological disorders, including schizophrenia, major depressive disorder, bipolar disorder, post-traumatic stress disorder, autism spectrum disorder, substance use disorders, Alzheimer's disease, Parkinson's disease, and multiple sclerosis. This review investigates the core mechanisms driving demyelination, its clinical impact, and emerging therapeutic strategies aimed at maintaining or restoring myelin integrity. Disruption of myelin impairs crucial neural communication pathways, resulting in cognitive, motor, and behavioral deficits that substantially reduce quality of life and create significant economic and social challenges. Key contributors to demyelination include genetic predisposition, environmental triggers, immune dysregulation, neuroinflammation, and alterations in the gut-brain axis mediated by the vagus nerve. Promising therapies include sphingosine 1-phosphate receptor modulators and muscarinic acetylcholine receptor antagonists, both of which diminish immune-related myelin damage and may enhance neuroprotection. In addition, the novel antidepressant arketamine appears to boost myelination through transforming growth factor-β1 signaling pathways. Approaches targeting the gut-brain axis, such as noninvasive transcutaneous auricular vagus nerve stimulation and fecal microbiota transplantation, may also help reduce inflammation and support myelin repair. Future research should center on clarifying the precise molecular mechanisms of demyelination, developing targeted therapies, and leveraging advanced neuroimaging for earlier detection and personalized treatment. By combining immunomodulatory and neuroprotective strategies, there is potential to significantly improve outcomes for individuals affected by demyelinating psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Rumi Murayama
- Laboratory of Chemical Pharmacology, Chiba University Graduate School of Pharmaceutical Sciences, Chiba 260-8675, Japan; Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Yi Cai
- Laboratory of Chemical Pharmacology, Chiba University Graduate School of Pharmaceutical Sciences, Chiba 260-8675, Japan; Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Hiroyuki Nakamura
- Laboratory of Chemical Pharmacology, Chiba University Graduate School of Pharmaceutical Sciences, Chiba 260-8675, Japan
| | - Kenji Hashimoto
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| |
Collapse
|
5
|
Atashgar F, Shafieian M, Abolfathi N. From structure to mechanics: exploring the role of axons and interconnections in anisotropic behavior of brain white matter. Biomech Model Mechanobiol 2025:10.1007/s10237-025-01957-4. [PMID: 40295358 DOI: 10.1007/s10237-025-01957-4] [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: 12/03/2024] [Accepted: 03/28/2025] [Indexed: 04/30/2025]
Abstract
According to various experimental studies, the role of axons in the brain's white matter (WM) is still a subject of debate: Is the role of axons in brain white matter (WM) limited to their functional significance, or do they also play a pivotal mechanical role in defining its anisotropic behavior? Micromechanics and computational models provide valuable tools for scientists to comprehend the underlying mechanisms of tissue behavior, taking into account the contribution of microstructures. In this review, we delve into the consideration of strain level, strain rates, and injury threshold to determine when WM should be regarded as anisotropic, as well as when the assumption of isotropy can be deemed acceptable. Additionally, we emphasize the potential mechanical significance of interconnections between glial cells-axons and glial cells-vessels. Moreover, we elucidate the directionality of WM stiffness under various loading conditions and define the possible roles of microstructural components in each scenario. Ultimately, this review aims to shed light on the significant mechanical contributions of axons in conjunction with glial cells, paving the way for the development of future multiscale models capable of predicting injuries and facilitating the discovery of applicable treatments.
Collapse
Affiliation(s)
- Fatemeh Atashgar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Mehdi Shafieian
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Nabiollah Abolfathi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
6
|
Kujawa MJ, Grzywińska M, Sawicka AK, Marcinkowska AB, Chroboczek M, Jost Z, Szurowska E, Winklewski PJ, Szarmach A, Kujach S. Association between cardiorespiratory fitness and total brain myelin volume among older adults. Eur Rev Aging Phys Act 2025; 22:5. [PMID: 40200178 PMCID: PMC11980140 DOI: 10.1186/s11556-025-00371-0] [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: 05/13/2024] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Myelin, which insulates neurons, speeds up information transfer and provides the necessary conditions for cognitive and motor functioning. The direct link between physical performance and the total brain myelin volume remains unclear. METHODS This study involved 87 healthy participants (71 women, 16 men) with a mean age of 69.3 ± 3.14 years and a mean body mass index of 27.83 ± 3.93 kg/m2. Several measures of physical fitness (isometric muscle strength, handgrip strength, and cardiopulmonary exercise testing) were examined for their correlations with the total brain myelin volume using Synthetic MRI, an FDA-approved myelin assessment software. RESULTS A high maximal respiratory exchange ratio and low maximal heart rate achieved during cardiopulmonary exercise testing were associated with higher estimated brain myelin content. In addition, the handgrip strength test performance as well as the peak and average peak torque were associated with higher brain parenchymal myelin volumes. CONCLUSIONS We demonstrated that higher brain myelin content was positively associated with better cardiorespiratory fitness and higher upper and lower limb muscle strength in older individuals. These findings provide new insights into the development of improved rehabilitation and exercise schemes to preserve cognitive health in the older adult population.
Collapse
Affiliation(s)
- Mariusz J Kujawa
- 2nd Department of Radiology, Medical University of Gdansk, Gdansk, 80-214, Poland.
| | - Małgorzata Grzywińska
- Neuroinformatics and Artificial Intelligence Lab, Department of Neurophysiology, Neuropsychology and Neuroinformatics, Medical University of Gdansk, Gdansk, 80-210, Poland
| | - Angelika K Sawicka
- Applied Cognitive Neuroscience Lab, Department of Neurophysiology, Neuropsychology and Neuroinformatics, Medical University of Gdansk, Gdansk, 80-210, Poland
| | - Anna B Marcinkowska
- 2nd Department of Radiology, Medical University of Gdansk, Gdansk, 80-214, Poland
- Applied Cognitive Neuroscience Lab, Department of Neurophysiology, Neuropsychology and Neuroinformatics, Medical University of Gdansk, Gdansk, 80-210, Poland
| | - Maciej Chroboczek
- Department of Physiology, Gdansk University of Physical Education and Sport, Gdansk, 80-336, Poland
| | - Zbigniew Jost
- Department of Biochemistry, Gdansk University of Physical Education and Sport, Gdansk, 80-336, Poland
| | - Edyta Szurowska
- 2nd Department of Radiology, Medical University of Gdansk, Gdansk, 80-214, Poland
| | - Paweł J Winklewski
- 2nd Department of Radiology, Medical University of Gdansk, Gdansk, 80-214, Poland
- Department of Neurophysiology, Neuropsychology and Neuroinformatics, Medical University of Gdansk, Gdansk, 80-210, Poland
- Institute of Health Sciences, Pomeranian University in Slupsk, Slupsk, 76-200, Poland
| | - Arkadiusz Szarmach
- 2nd Department of Radiology, Medical University of Gdansk, Gdansk, 80-214, Poland.
| | - Sylwester Kujach
- Department of Physiology, Gdansk University of Physical Education and Sport, Gdansk, 80-336, Poland.
- Department of Physiology, Medical University of Gdansk, Gdansk, 80-210, Poland.
| |
Collapse
|
7
|
Freus NK, Wank I, Häfele M, Kalinichenko LS, Müller CP, Strobelt S, Ludwig A, Hess A, Kreitz S. Pharmacological and resting state fMRI reveal Osteocalcin's effects on mouse brain regions with high Gpr37 and Gpr158 expression. Sci Rep 2025; 15:10116. [PMID: 40128223 PMCID: PMC11933355 DOI: 10.1038/s41598-025-95000-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/18/2025] [Indexed: 03/26/2025] Open
Abstract
Osteocalcin (OCN) is an endocrine hormone that signals in the periphery, regulating male fertility, energy expenditure and glucose homeostasis. It can also cross the blood-brain-barrier and act on the brain via receptors GPR37 and GPR158. In the brain, OCN influences neurotransmitter synthesis of serotonin, norepinephrine, and dopamine. OCN's function is related to cognitive and memory performance and lack of OCN is associated with anxiety and depression-like behavior in mice. We used multiparametric magnetic resonance imaging (MRI) including pharmacological MRI and resting state functional MRI, along with gene expression data for Gpr37 and Gpr158 to investigate the physiological effects of intravenously administered OCN on the wild type mouse brain. We found four core brain regions (brainstem, limbic output, association cortex, and basal ganglia) that are highly relevant in all three analytical modalities (i.e. pharmacological, resting state MRI and gene expression) and play therefore a major role in mediating OCN's effect in the brain. This study provides the first imaging data of the physiological impact of OCN on the mouse brain, suggesting its potential role in modulating brain function and its relevance as a candidate for further investigation in anxiety, depression, and cognitive impairments.
Collapse
Affiliation(s)
- Natalia K Freus
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Isabel Wank
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Maximilian Häfele
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Liubov S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
- Central Institute for Mental Health, J5, 68159, Mannheim, Germany
| | - Sandra Strobelt
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Andreas Ludwig
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
- FAU NeW - Research Center for New Bioactive Compounds, Friedrich-Alexander- University Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
- FAU NeW - Research Center for New Bioactive Compounds, Friedrich-Alexander- University Erlangen-Nuremberg, Erlangen, Germany
| | - Silke Kreitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany.
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| |
Collapse
|
8
|
Kobayashi S, Yokoi T, Omata T, Yako H, Miyamoto Y, Yamauchi J. Claudin-11, a hypomyelinating leukodystrophy 22 (HLD22)-responsible protein, uniquely interacts with shroom-2 to change cell phenotypes. BBA ADVANCES 2025; 7:100159. [PMID: 40230506 PMCID: PMC11995805 DOI: 10.1016/j.bbadva.2025.100159] [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: 12/24/2024] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
Oligodendroglial cells are a type of glial cell in the central nervous system (CNS) that wrap neuronal axons with differentiated plasma membranes known as myelin sheaths. While the physiological functions of oligodendrocytes, such as generating saltatory conduction and protecting neuronal axons, are well understood, the physiological and/or pathophysiological molecular mechanisms governing their differentiation before myelination remain unclear. In this study, we describe for the first time that claudin-11, a protein associated with hypomyelinating leukodystrophy 22 (HLD22), interacts with shroom-2, a presumable adaptor protein containing the PSD95, DLG1, and ZO-1 (PDZ) domain. Knockdown of claudin-11 using specific siRNA resulted in a decrease in morphological changes and marker proteins in the FBD-102b oligodendroglial model undergoing differentiation. Transfection of the C-terminal PDZ ligand sequence of claudin-11, which was found to interact with the PDZ domain of shroom-2, also reduced these phenotypic changes. The HLD22-associated mutated sequence in claudin-11 failed to interact with the PDZ domain of shroom-2. Furthermore, knockdown of shroom-2 or transfection of the PDZ domain of shroom-2, which is involved in the interaction with claudin-11, resulted in decreased morphological changes and marker protein expression. These changes were linked to the phosphorylation states of Akt kinase, a key signaling molecule in oligodendroglial cell differentiation and myelination. These results suggest that the interaction between claudin-11 and shroom-2 plays a key role in shaping cell morphology, providing insights into the molecular mechanisms underlying oligodendroglial differentiation before myelination, as well as potential pathological mechanisms associated with HLD22 at the molecular and cellular levels.
Collapse
Affiliation(s)
- Sakurako Kobayashi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Takanori Yokoi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Takeru Omata
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Hideji Yako
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
9
|
Pascual-Guerra J, Torres-Rico M, Paíno CL, Rodríguez-Navarro JA, García AG. Fractal analysis to assess the differentiation state of oligodendroglia in culture. J Neurosci Methods 2025; 415:110336. [PMID: 39638053 DOI: 10.1016/j.jneumeth.2024.110336] [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: 04/01/2024] [Revised: 09/23/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Oligodendroglial development is accompanied by increased cell complexity. A simple and cost-effective evaluation of the pro-myelinating activity of different drugs and/or treatments would be of great interest. In cultured oligodendroglia, an evaluation of the pro-myelinating activity of different drugs and/or treatments can be achieved through fractal analysis, which allows measuring cell complexity. NEW METHOD Fractal dimension was assessed in two O4+ cell types (neural stem cell-derived and lineage-converted adipose tissue mesenchymal cells) under proliferating or differentiating conditions. COMPARISON WITH EXISTING METHODS This analysis, which was originally developed to analyze microglia, assigns a quantitative value (fractal dimension) to cellular profiles, obtaining higher coefficients as cells increase in size and arborizations instead of mRNA or protein quantification of mature oligodendroglial markers, such as MBP, MAG, O1 or PLP1/DM20. RESULTS This article describes a methodology to perform fractal analysis in immunofluorescent images of O4-positive (O4+) oligodendroglia using the FracLac plugin of ImageJ software. Pro-myelinating drug Benztropine-treated O4+ cells exhibit higher fractal dimension than control group. CONCLUSIONS The results demonstrated the effectiveness and sensitivity of the fractal dimension coefficient provided by FracLac software to assess the effects of treatments on oligodendroglial differentiation.
Collapse
Affiliation(s)
- J Pascual-Guerra
- Servicio de Neurobiología-Investigación, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain; Fundación Teófilo Hernando, Madrid, Spain.
| | | | - C L Paíno
- Servicio de Neurobiología-Investigación, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - J A Rodríguez-Navarro
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid, Spain
| | - A G García
- Fundación Teófilo Hernando, Madrid, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Spain
| |
Collapse
|
10
|
Chen J, He Y, Zhong J, Fu Y, Yuan S, Hou L, Zhang X, Meng F, Lin WJ, Ji F, Wang Z. Transcranial near-infrared light promotes remyelination through AKT1/mTOR pathway to ameliorate postoperative neurocognitive disorder in aged mice. Neuroscience 2025; 565:358-368. [PMID: 39653248 DOI: 10.1016/j.neuroscience.2024.12.007] [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: 07/04/2024] [Revised: 11/10/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Postoperative neurocognitive disorder (PND) is a prevalent complication following surgery and anesthesia, characterized by progressive cognitive decline. The precise etiology of PND remains unknown, and effective targeted therapeutic strategies are lacking. Transcranial near-infrared light (tNIRL) has shown potential benefits for cognitive dysfunction diseases, but its effect on PND remains unclear. Our previous research indicated a close association between demyelination and PND. In other central nervous system (CNS) disorders, tNIRL has been demonstrated to facilitate remyelination in response to demyelination. In this study, we established the PND model in 18-month-old male C57BL/6 mice using isoflurane anesthesia combined with left common carotid artery exposure. Following surgery, PND-aged mice were subjected to daily 2.5-minute tNIRL treatment at 810 nm for three consecutive days. Subsequently, we observed that tNIRL significantly improved cognitive performance and reduced inflammatory cytokine levels in the hippocampus of PND mice. Furthermore, tNIRL increased the expression of oligodendrocyte transcription factor 2 (OLIG2) and myelin basic protein (MBP), promoting remyelination while enhancing synaptic function-associated proteins such as synaptophysin (SYP) and postsynaptic density protein 95 (PSD95). Further investigation revealed that tNIRL may activate the AKT1/mTOR pathway to facilitate remyelination in PND mice. These findings indicate that tNIRL is a novel non-invasive therapeutic approach for treating PND.
Collapse
Affiliation(s)
- Jiawei Chen
- Department of Anesthesiology, Meishan City People's Hospital, Meishan, Sichuan, China
| | - Yuqing He
- Department of Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Junying Zhong
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanni Fu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shangyan Yuan
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Longjie Hou
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaojun Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fanqing Meng
- Department of Anesthesiology, Jinan Maternity and Child Care Hospital, Jinan, Shandong, China
| | - Wei-Jye Lin
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fengtao Ji
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Zhi Wang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Wang T, Huang X, Sun S, Wang Y, Han L, Zhang T, Zhang T, Chen X. Recent Advances in the Mechanisms of Postoperative Neurocognitive Dysfunction: A Narrative Review. Biomedicines 2025; 13:115. [PMID: 39857699 PMCID: PMC11762480 DOI: 10.3390/biomedicines13010115] [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: 12/03/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Postoperative neurocognitive dysfunction (PND) is a prevalent and debilitating complication in elderly surgical patients, characterized by persistent cognitive decline that negatively affects recovery and quality of life. As the aging population grows, the rising number of elderly surgical patients has made PND an urgent clinical challenge. Despite increasing research efforts, the pathophysiological mechanisms underlying PND remain inadequately characterized, underscoring the need for a more integrated framework to guide targeted interventions. To better understand the molecular mechanisms and therapeutic targets of PND, this narrative review synthesized evidence from peer-reviewed studies, identified through comprehensive searches of PubMed, Embase, Cochrane Library, and Web of Science. Key findings highlight neuroinflammation, oxidative stress, mitochondrial dysfunction, neurotransmitter imbalances, microvascular changes, and white matter lesions as central to PND pathophysiology, with particular parallels to encephalocele- and sepsis-associated cognitive impairments. Among these, neuroinflammation, mediated by pathways such as the NLRP3 inflammasome and blood-brain barrier disruption, emerges as a pivotal driver, triggering cascades that exacerbate neuronal injury. Oxidative stress and mitochondrial dysfunction synergistically amplify these effects, while neurotransmitter imbalances and microvascular alterations, including white matter lesions, contribute to synaptic dysfunction and cognitive decline. Anesthetic agents modulate these interconnected pathways, exhibiting both protective and detrimental effects. Propofol and dexmedetomidine demonstrate neuroprotective properties by suppressing neuroinflammation and microglial activation, whereas inhalational anesthetics like sevoflurane intensify oxidative stress and inflammatory responses. Ketamine, with its anti-inflammatory potential, offers promise but requires further evaluation to determine its long-term safety and efficacy. By bridging molecular insights with clinical practice, this review highlights the critical role of personalized anesthetic strategies in mitigating PND and improving cognitive recovery in elderly surgical patients. It aims to inform future research and clinical decision-making to address this multifaceted challenge.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Xin Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Tao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| |
Collapse
|
12
|
Liu X, Lv Z, Huang Q, Lei Y, Liu H, Xu P. The Role of Oligodendrocyte Lineage Cells in the Pathogenesis of Alzheimer's Disease. Neurochem Res 2025; 50:72. [PMID: 39751972 DOI: 10.1007/s11064-024-04325-3] [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: 07/29/2024] [Revised: 10/06/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Alzheimer's disease (AD) is a central nervous system degenerative disease with a stealthy onset and a progressive course characterized by memory loss, cognitive dysfunction, and abnormal psychological and behavioral symptoms. However, the pathogenesis of AD remains elusive. An increasing number of studies have shown that oligodendrocyte progenitor cells (OPCs) and oligodendroglial lineage cells (OLGs), especially OPCs and mature oligodendrocytes (OLGs), which are derived from OPCs, play important roles in the pathogenesis of AD. OLGs function mainly by myelinating axons, transmitting electrical signals, and regulating neural development. In addition to myelin, OPCs and OLGs can also participate in AD pathogenesis in other ways. This review summarizes the mechanisms by which OPCs and OLGs affect myelin formation, oxidative stress, neuroinflammation, the blood-brain barrier, synaptic function, and amyloid-beta protein and further elucidates the mechanisms by which oligodendrocyte lineage cells participate in AD pathogenesis and treatment, which is highly important for clarifying the pathogenesis of AD, clinical treatment, and prevention.
Collapse
Affiliation(s)
- Xiaodong Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Neurology, China Guihang Group 302 Hospital, Anshun, China
| | - Zhengxiang Lv
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Neurology, China Guihang Group 302 Hospital, Anshun, China
| | - Qin Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yihui Lei
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Haijun Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ping Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
13
|
Fitzgerald JC, Sun Y, Reinecke F, Bauer E, Garaschuk O, Kahle PJ, Pfeiffer F. Interactions of Oligodendrocyte Precursor Cells and Dopaminergic Neurons in the Mouse Substantia Nigra. J Neurochem 2025; 169:e16298. [PMID: 39871627 PMCID: PMC11773302 DOI: 10.1111/jnc.16298] [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: 08/15/2024] [Revised: 11/25/2024] [Accepted: 12/16/2024] [Indexed: 01/29/2025]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disease caused by the death of dopaminergic neurons within the substantia nigra pars compacta (SNpc) region of the midbrain. Recent genomic and single cell sequencing data identified oligodendrocytes and oligodendrocyte precursor cells (OPCs) to confer genetic risk in PD, but their biological role is unknown. Although SNpc dopaminergic neurons are scarcely or thinly myelinated, there is a gap in the knowledge concerning the physiological interactions between dopaminergic neurons and oligodendroglia. We sought to investigate the distribution of OPCs with regard to the myelination state in the mouse substantia nigra (SN) by high-resolution imaging to provide a morphological assessment of OPC-dopaminergic neuron interactions and quantification of cell numbers across different age groups. OPCs are evenly distributed in the midbrain throughout the lifespan and they physically interact with both the soma and axons of dopaminergic neurons. The presence of OPCs and their interaction with dopaminergic neurons does not correlate with the distribution of myelin. Myelination is sparse in the SNpc, including dopaminergic fibers originating from the SNpc and projecting through the substantia nigra pars reticulata (SNpr). We report that OPCs and dopaminergic neurons exist in a 1:1 ratio in the SNpc, with OPCs accounting for 15%-16% of all cells in the region across all age groups. This description of OPC-dopaminergic neuron interaction in the midbrain provides a first look at their longitudinal distribution in mice, suggesting additional functions of OPCs beyond their differentiation into myelinating oligodendrocytes.
Collapse
Affiliation(s)
- Julia C. Fitzgerald
- Hertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Ying Sun
- Institute for PhysiologyUniversity of TübingenTübingenGermany
| | | | - Elisabeth Bauer
- Hertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Olga Garaschuk
- Institute for PhysiologyUniversity of TübingenTübingenGermany
| | - Philipp J. Kahle
- Hertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- The German Centre for Neurodegenerative DiseasesTübingenGermany
- Institute of BiochemistryUniversity of TübingenTübingenGermany
| | | |
Collapse
|
14
|
Kim W, Angulo MC. Unraveling the role of oligodendrocytes and myelin in pain. J Neurochem 2025; 169:e16206. [PMID: 39162089 PMCID: PMC11657919 DOI: 10.1111/jnc.16206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/30/2024] [Accepted: 08/04/2024] [Indexed: 08/21/2024]
Abstract
Oligodendrocytes, the myelin-producing cells in the central nervous system (CNS), are crucial for rapid action potential conduction and neuronal communication. While extensively studied for their roles in neuronal support and axonal insulation, their involvement in pain modulation is an emerging research area. This review explores the interplay between oligodendrocytes, myelination, and pain, focusing on neuropathic pain following peripheral nerve injury, spinal cord injury (SCI), chemotherapy, and HIV infection. Studies indicate that a decrease in oligodendrocytes and increased cytokine production by oligodendroglia in response to injury can induce or exacerbate pain. An increase in endogenous oligodendrocyte precursor cells (OPCs) may be a compensatory response to repair damaged oligodendrocytes. Exogenous OPC transplantation shows promise in alleviating SCI-induced neuropathic pain and enhancing remyelination. Additionally, oligodendrocyte apoptosis in brain regions such as the medial prefrontal cortex is linked to opioid-induced hyperalgesia, highlighting their role in central pain mechanisms. Chemotherapeutic agents disrupt oligodendrocyte differentiation, leading to persistent pain, while HIV-associated neuropathy involves up-regulation of oligodendrocyte lineage cell markers. These findings underscore the multifaceted roles of oligodendrocytes in pain pathways, suggesting that targeting myelination processes could offer new therapeutic strategies for chronic pain management. Further research should elucidate the underlying molecular mechanisms to develop effective pain treatments.
Collapse
Affiliation(s)
- Woojin Kim
- Department of Physiology, College of Korean MedicineKyung Hee UniversitySeoulRepublic of Korea
- Korean Medicine‐Based Drug Repositioning Cancer Research Center, College of Korean MedicineKyung Hee UniversitySeoulRepublic of Korea
| | - María Cecilia Angulo
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, “Team: Interactions between Neurons and Oligodendroglia in Myelination and Myelin Repair”ParisFrance
- GHU PARIS Psychiatrie & NeurosciencesParisFrance
| |
Collapse
|
15
|
Rossini L, Maderna E, De Santis D, Rizzi M, Tassi L, Pastori C, Garbelli R, de Curtis M. Altered Gray Matter Myelin in Type IIb Focal Cortical Dysplasia. Neurology 2024; 103:e210057. [PMID: 39586045 DOI: 10.1212/wnl.0000000000210057] [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: 03/28/2024] [Accepted: 09/10/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Myelin is altered in several neurologic disorders. Published data demonstrate reduced white matter myelin content and lower oligodendrocyte cell number in postsurgical brain specimens obtained from patients with focal cortical dysplasia (FCD) and temporal lobe epilepsy; a pathogenic role of dysfunctional myelin in focal epilepsies has been proposed. Based on this evidence, our study aims to investigate the myelination status in the gray matter in postsurgical brain specimens from patients with FCDIIb. METHODS We collected specimens from patients with a histopathologic diagnosis of FCDIIb who underwent surgery between 1995 and 2022 in 2 epilepsy surgery centers in Milano; we used nonlesional samples and perilesional tissue within the same FCDIIb specimen as controls. Immunohistochemistry for myelin basic protein (MBP) and electron microscopy were used to quantify myelin alterations in the lesional core of FCDIIb specimens compared with nonlesional and perilesional control areas. Olig2 and breast carcinoma amplified sequence 1 immunohistochemistry, markers of oligodendrocytes, were also evaluated. RESULTS Sixteen patients with FCDIIb (24 ± 14 mean years at surgery, 44% female) and 4 controls (3 histopathology-negative epileptic patients and 1 patient with nonepileptic tumor; 32 ± 11 mean years at surgery, 50% female) were included. The cortical myeloarchitecture was disorganized in the FCD core lesion. MBP immunostained fiber density from 11 paired samples that included both the FCD lesional core and adjacent perilesional cortex in the same tissue section did not reveal a significant difference. Ultrastructural examination performed in the gray matter of 6 specimens from FCDIIb patients (both in the core and in the adjacent perilesional areas) and 2 controls revealed that exclusively in the FCDIIb core, myelinated fiber density was reduced and axons featured thin or no myelin coating and pathologic vacuoles. These changes were associated with a reduction of Olig2-immunostained cells in the FCDIIb cortex core. DISCUSSION Our findings demonstrate that the gray matter at the core of postsurgical FCDIIb specimens contains a high number of poorly myelinated axons and less oligodendrocytes; these findings suggest a potential contribution of altered myelination in the pathogenesis of FCDIIb.
Collapse
Affiliation(s)
- Laura Rossini
- From the Epilepsy Unit (L.R., D.D.S., C.P., R.G., M.d.C.), Division of Neurology V and Neuropathology (E.M.), and Neurosurgery Unit (M.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta; and Claudio Munari Epilepsy Surgery Centre (L.T.), Niguarda Hospital, Milano, Italy
| | - Emanuela Maderna
- From the Epilepsy Unit (L.R., D.D.S., C.P., R.G., M.d.C.), Division of Neurology V and Neuropathology (E.M.), and Neurosurgery Unit (M.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta; and Claudio Munari Epilepsy Surgery Centre (L.T.), Niguarda Hospital, Milano, Italy
| | - Dalia De Santis
- From the Epilepsy Unit (L.R., D.D.S., C.P., R.G., M.d.C.), Division of Neurology V and Neuropathology (E.M.), and Neurosurgery Unit (M.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta; and Claudio Munari Epilepsy Surgery Centre (L.T.), Niguarda Hospital, Milano, Italy
| | - Michele Rizzi
- From the Epilepsy Unit (L.R., D.D.S., C.P., R.G., M.d.C.), Division of Neurology V and Neuropathology (E.M.), and Neurosurgery Unit (M.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta; and Claudio Munari Epilepsy Surgery Centre (L.T.), Niguarda Hospital, Milano, Italy
| | - Laura Tassi
- From the Epilepsy Unit (L.R., D.D.S., C.P., R.G., M.d.C.), Division of Neurology V and Neuropathology (E.M.), and Neurosurgery Unit (M.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta; and Claudio Munari Epilepsy Surgery Centre (L.T.), Niguarda Hospital, Milano, Italy
| | - Chiara Pastori
- From the Epilepsy Unit (L.R., D.D.S., C.P., R.G., M.d.C.), Division of Neurology V and Neuropathology (E.M.), and Neurosurgery Unit (M.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta; and Claudio Munari Epilepsy Surgery Centre (L.T.), Niguarda Hospital, Milano, Italy
| | - Rita Garbelli
- From the Epilepsy Unit (L.R., D.D.S., C.P., R.G., M.d.C.), Division of Neurology V and Neuropathology (E.M.), and Neurosurgery Unit (M.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta; and Claudio Munari Epilepsy Surgery Centre (L.T.), Niguarda Hospital, Milano, Italy
| | - Marco de Curtis
- From the Epilepsy Unit (L.R., D.D.S., C.P., R.G., M.d.C.), Division of Neurology V and Neuropathology (E.M.), and Neurosurgery Unit (M.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta; and Claudio Munari Epilepsy Surgery Centre (L.T.), Niguarda Hospital, Milano, Italy
| |
Collapse
|
16
|
Al Mamun A, Quan Z, Geng P, Wang S, Shao C, Xiao J. Targeting Remyelination in Spinal Cord Injury: Insights and Emerging Therapeutic Strategies. CNS Neurosci Ther 2024; 30:1-15. [PMID: 39723448 DOI: 10.1111/cns.70193] [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: 08/05/2024] [Revised: 11/27/2024] [Accepted: 12/07/2024] [Indexed: 12/28/2024] Open
Abstract
INTRODUCTION Spinal cord injury (SCI) is a severe neurological disease characterized by significant motor, sensory, and autonomic dysfunctions. SCI is a major global disability cause, often resulting in long-term neurological impairments due to the impeded regeneration and remyelination of axons. A SCI interferes with communication between the brain and the spinal cord networks that control neurological functions. Recent advancements in understanding the molecular and cellular mechanisms of remyelination have opened novel therapeutic interventions. METHOD This review systematically sourced articles related to spinal chord injury, remyelination, regeneration and pathophysiology from major medical databases, including Scopus, PubMed, and Web of Science. RESULTS This review discusses the efficacy of targeted therapy in enhancing myelin repair after SCI by identifying key molecules and signaling pathways. This explores the effectiveness of specific pharmacological agents and biological factors in promoting oligodendrocyte precursor cell proliferation, differentiation, and myelin sheath formation using in vitro and in vivo models. Targeted therapies have shown promising results in improving remyelination, providing hope for functional recovery in SCI patients. CONCLUSIONS This review demonstrates challenges and future perspectives in translating findings into clinical practice, emphasizing safety profiles, delivery method optimization, and combinatory therapy potential. This review also supports the possibility of targeted remyelination therapies as a promising strategy for SCI treatment, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhou Quan
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
17
|
Tusnim J, Kutuzov P, Grasman JM. In Vitro Models for Peripheral Nerve Regeneration. Adv Healthc Mater 2024; 13:e2401605. [PMID: 39324286 DOI: 10.1002/adhm.202401605] [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: 04/30/2024] [Revised: 08/14/2024] [Indexed: 09/27/2024]
Abstract
Peripheral nerve injury (PNI) resulting in lesions is highly prevalent clinically, but current therapeutic approaches fail to provide satisfactory outcomes in many patients. While peripheral nerves have intrinsic regenerative capacity, the regenerative capabilities of peripheral nerves are often insufficient to restore full functionality. This highlights an unmet need for developing more effective strategies to repair damaged peripheral nerves and improve regenerative success. Consequently, researchers are actively exploring a variety of therapeutic strategies, encompassing the local delivery of trophic factors or bioactive molecules, the design of advanced biomaterials that interact with regenerating axons, and augmentation with nerve guidance conduits or complex prostheses. However, clinical translation of these technologies remains limited, emphasizing the need for continued research on peripheral nerve regeneration modalities that can enhance functional restoration. Experimental models that accurately recapitulate key aspects of peripheral nerve injury and repair biology can accelerate therapeutic development by enabling systematic testing of new techniques. Advancing regenerative therapies for PNI requires bridging the gap between basic science discoveries and clinical application. This review discusses different in vitro models of peripheral nerve injury and repair, including their advantages, limitations, and potential applications.
Collapse
Affiliation(s)
- Jarin Tusnim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Peter Kutuzov
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jonathan M Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| |
Collapse
|
18
|
Rugieł M, Setkowicz Z, Czyzycki M, Simon R, Baumbach T, Chwiej J. Element Changes Occurring in Brain Point at the White Matter Abnormalities in Rats Exposed to the Ketogenic Diet During Prenatal Life. ACS Chem Neurosci 2024; 15:3932-3944. [PMID: 39443296 PMCID: PMC11587514 DOI: 10.1021/acschemneuro.4c00283] [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: 05/06/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
A large number of clinical studies demonstrate that the ketogenic diet (KD) may be an effective approach to the reduction of epileptic seizures in children and adults. Such dietary therapy could also help pregnant women with epilepsy, especially since most antiseizure drugs have teratogenic action. However, there is a lack of medical data, considering the safety of using KD during gestation for the progeny. Therefore, we examined the influence of KD used prenatally in rats on the elemental composition of the selected brain regions in their offspring. For this purpose, synchrotron radiation-induced X-ray fluorescence (SR-XRF) microscopy was utilized, and elements such as P, S, K, Ca, Fe, and Zn were determined. Moreover, to verify whether the possible effects of KD are temporary or long-term, different stages of animal postnatal development were taken into account in our experiment. The obtained results confirmed the great applicability of SR-XRF microscopy to track the element changes occurring in the brain during postnatal development as well as those induced by prenatal exposure to the high-fat diet. The topographic analysis of the brains taken from offspring of mothers fed with KD during pregnancy and appropriate control individuals showed a potential influence of such dietary treatment on the brain levels of elements such as P and S. In the oldest progeny, a significant reduction of the surface of brain areas characterized by an increased P and S content, which histologically/morphologically correspond to white matter structures, was noticed. In turn, quantitative elemental analysis showed significantly decreased levels of Fe in the striatum and white matter of 30-day-old rats exposed prenatally to KD. This effect was temporary and was not noticed in adult animals. The observed abnormalities may be related to the changes in the accumulation of sphingomyelin and sulfatides and may testify about disturbances in the structure and integrity of the myelin, present in the white matter.
Collapse
Affiliation(s)
- Marzena Rugieł
- Faculty of
Physics and Applied Computer Science, AGH
University of Krakow, Al. Mickiewicza 30, Krakow 30-059, Poland
| | - Zuzanna Setkowicz
- Institute
of Zoology and Biomedical Research, Jagiellonian
University, Gronostajowa
9, Krakow 30-387, Poland
| | - Mateusz Czyzycki
- Institute
for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen D-76344, Germany
| | - Rolf Simon
- Institute
for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen D-76344, Germany
| | - Tilo Baumbach
- Institute
for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen D-76344, Germany
- Laboratory
for Applications of Synchrotron Radiation, Karlsruhe Institute of Technology, Kaiserstr. 12, Karlsruhe D-76131, Germany
| | - Joanna Chwiej
- Faculty of
Physics and Applied Computer Science, AGH
University of Krakow, Al. Mickiewicza 30, Krakow 30-059, Poland
| |
Collapse
|
19
|
Barbosa-Azevedo M, Dias-Carvalho A, Carvalho F, Costa VM. Chemotherapy-induced cognitive impairment and glia: A new take on chemobrain? Toxicol Appl Pharmacol 2024; 492:117085. [PMID: 39236990 DOI: 10.1016/j.taap.2024.117085] [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: 07/03/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
The significant rise in cancer survivorship stands out as one of the most notable achievements of modern science. However, this comes with a significant burden, as cancer treatment is not without adverse effects. Lately, there has been a growing focus on cognitive dysfunction associated with cancer treatment, often referred to as 'chemobrain'. It significantly impacts the quality of life for cancer survivors. The underlying mechanisms studied so far usually focus on neurons, while other cells of the central nervous system are often overlooked. This review seeks to place the hypothesis that glial cells may play a role in the development of chemotherapy-induced cognitive dysfunction. It summarizes the primary mechanisms proposed to date while underscoring the existing gaps in this research field. Inflammation and release of pro-inflammatory mediators by M1 microglia and A1 astrocytes are the most prevalent findings after chemotherapy. However, activation of A1 astrocytes by some chemotherapeutic agents may contribute to neuronal degeneration, alterations in synaptic branches, as well as glutamate excitotoxicity, which can contribute to cognitive impairment. Furthermore, the reduction in the number of oligodendrocytes after chemotherapy may also impact the myelin sheath, contributing to 'chemobrain'. Furthermore, some chemotherapeutic drugs activate M1 microglia, which is associated with decreased neuroplasticity and, possibly, cognitive impairment. In conclusion, data regarding the effects of chemotherapy on glial cells are scarce, and it is essential to understand how these cells are affected after cancer treatment to enable reliable therapeutic or preventive actions on cancer-treated patients.
Collapse
Affiliation(s)
- Maria Barbosa-Azevedo
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Ana Dias-Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
20
|
Fukushima N, Miyamoto Y, Yamauchi J. CRISPR/CasRx-Mediated Knockdown of Rab7B Restores Incomplete Cell Shape Induced by Pelizaeus-Merzbacher Disease-Associated PLP1 p.Ala243Val. Neurosci Insights 2024; 19:26331055241276873. [PMID: 39280331 PMCID: PMC11402064 DOI: 10.1177/26331055241276873] [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: 05/28/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024] Open
Abstract
Pelizaeus-Merzbacher disease (PMD, currently known as hypomyelinating leukodystrophy type 1 [HLD1]) is a hereditary hypomyelinating and/or demyelinating disease associated with the proteolipid protein 1 (plp1) gene in the central nervous system (CNS). One of the major causes of this condition is incomplete or defective oligodendroglial cell myelin sheath formation triggered by endoplasmic reticulum (ER) stress and subsequent unfolded protein response (UPR). The HLD1-associated Ala-243-to-Val mutation (p.Ala243Val) of PLP1 is widely recognized to trigger defective oligodendroglial cell morphological differentiation, primarily due to ER stress. We have previously reported that knockdown of Rab7B (also known as Rab42), a small GTP/GDP-binding protein involved in intracellular vesicle trafficking around the lysosome, can recover chemical ER stress-induced incomplete cell shapes in the FBD-102b cell line, a model of oligodendroglial cell morphological differentiation. Here, we present findings indicating that incomplete cell shapes induced by PLP1 p.Ala243Val can be restored by knockdown of Rab7B using the clustered regularly interspaced short palindromic repeats (CRISPR) and CasRx (also known as Cas13d) system. Also, the knockdown promoted the trafficking of PLP1 p.Ala243Val to lysosome-associated membrane protein 1 (LAMP1)-positive organelles. These results highlight the unique role of Rab7B knockdown in modulating oligodendroglial cell morphological changes and potentially facilitating the transport of mutated PLP1 to LAMP1-positive organelles, suggesting its potential as a therapeutic target for alleviating HLD1 phenotypes, at least in part, at the molecular and cellular levels.
Collapse
Affiliation(s)
- Nana Fukushima
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
21
|
Ames S, Brooks J, Jones E, Morehouse J, Cortez-Thomas F, Desta D, Stirling DP. NKCC1 inhibition reduces periaxonal swelling, increases white matter sparing, and improves neurological recovery after contusive SCI. Neurobiol Dis 2024; 199:106611. [PMID: 39032797 DOI: 10.1016/j.nbd.2024.106611] [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: 06/04/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024] Open
Abstract
Ultrastructural studies of contusive spinal cord injury (SCI) in mammals have shown that the most prominent acute changes in white matter are periaxonal swelling and separation of myelin away from their axon, axonal swelling, and axonal spheroid formation. However, the underlying cellular and molecular mechanisms that cause periaxonal swelling and the functional consequences are poorly understood. We hypothesized that periaxonal swelling and loss of connectivity between the axo-myelinic interface impedes neurological recovery by disrupting conduction velocity, and glial to axonal trophic support resulting in axonal swelling and spheroid formation. Utilizing in vivo longitudinal imaging of Thy1YFP+ axons and myelin labeled with Nile red, we reveal that periaxonal swelling significantly increases acutely following a contusive SCI (T13, 30 kdyn, IH Impactor) versus baseline recordings (laminectomy only) and often precedes axonal spheroid formation. In addition, using longitudinal imaging to determine the fate of myelinated fibers acutely after SCI, we show that ∼73% of myelinated fibers present with periaxonal swelling at 1 h post SCI and ∼ 51% of those fibers transition to axonal spheroids by 4 h post SCI. Next, we assessed whether cation-chloride cotransporters present within the internode contributed to periaxonal swelling and whether their modulation would increase white matter sparing and improve neurological recovery following a moderate contusive SCI (T9, 50 kdyn). Mechanistically, activation of the cation-chloride cotransporter KCC2 did not improve neurological recovery and acute axonal survival, but did improve chronic tissue sparing. In distinction, the NKKC1 antagonist bumetanide improved neurological recovery, tissue sparing, and axonal survival, in part through preventing periaxonal swelling and disruption of the axo-myelinic interface. Collectively, these data reveal a novel neuroprotective target to prevent periaxonal swelling and improve neurological recovery after SCI.
Collapse
Affiliation(s)
- Spencer Ames
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - Jesse Brooks
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - Emma Jones
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - Johnny Morehouse
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - Francisco Cortez-Thomas
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202; Bioengineering, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - Dereje Desta
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202
| | - David P Stirling
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY, USA 40202; Departments of Neurological Surgery, University of Louisville, School of Medicine, Louisville, KY, USA 40202; Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA 40202.
| |
Collapse
|
22
|
Liu H, Yi J, Zhang C, Li Y, Wang Q, Wang S, Dai S, Zheng Z, Jiang T, Gao P, Xue A, Huang Z, Kong F, Wang Y, He B, Guo X, Li Q, Chen J, Yin G, Zhao S. Macrophage GIT1 promotes oligodendrocyte precursor cell differentiation and remyelination after spinal cord injury. Glia 2024; 72:1674-1692. [PMID: 38899731 DOI: 10.1002/glia.24577] [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: 08/29/2023] [Revised: 05/02/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
Spinal cord injury (SCI) can result in severe motor and sensory deficits, for which currently no effective cure exists. The pathological process underlying this injury is extremely complex and involves many cell types in the central nervous system. In this study, we have uncovered a novel function for macrophage G protein-coupled receptor kinase-interactor 1 (GIT1) in promoting remyelination and functional repair after SCI. Using GIT1flox/flox Lyz2-Cre (GIT1 CKO) mice, we identified that GIT1 deficiency in macrophages led to an increased generation of tumor necrosis factor-alpha (TNFα), reduced proportion of mature oligodendrocytes (mOLs), impaired remyelination, and compromised functional recovery in vivo. These effects in GIT1 CKO mice were reversed with the administration of soluble TNF inhibitor. Moreover, bone marrow transplantation from GIT1 CWT mice reversed adverse outcomes in GIT1 CKO mice, further indicating the role of macrophage GIT1 in modulating spinal cord injury repair. Our in vitro experiments showed that macrophage GIT1 plays a critical role in secreting TNFα and influences the differentiation of oligodendrocyte precursor cells (OPCs) after stimulation with myelin debris. Collectively, our data uncovered a new role of macrophage GIT1 in regulating the transformation of OPCs into mOLs, essential for functional remyelination after SCI, suggesting that macrophage GIT1 could be a promising treatment target of SCI.
Collapse
Affiliation(s)
- Hao Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiang Yi
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Orthopedics, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Chenxi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yin Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shenyu Wang
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Siming Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ziyang Zheng
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peng Gao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ao Xue
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhenfei Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fanqi Kong
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yongxiang Wang
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Yangzhou, China
- Northern Jiangsu People's Hospital, Yangzhou, China
| | - Baorong He
- Department of Spine Surgery, Honghui-hospital, Xi'an Jiaotong Uinversity, School of Medicine, Xi'an, China
| | - Xiaodong Guo
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingqing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shujie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
23
|
Ahuja K, Vandenabeele M, Nami F, Lefevere E, Van Hoecke J, Bergmans S, Claes M, Vervliet T, Neyrinck K, Burg T, De Herdt D, Bhaskar P, Zhu Y, Looser ZJ, Loncke J, Gsell W, Plaas M, Agostinis P, Swinnen JV, Van Den Bosch L, Bultynck G, Saab AS, Wolfs E, Chai YC, Himmelreich U, Verfaillie C, Moons L, De Groef L. A deep phenotyping study in mouse and iPSC models to understand the role of oligodendroglia in optic neuropathy in Wolfram syndrome. Acta Neuropathol Commun 2024; 12:140. [PMID: 39198924 PMCID: PMC11351506 DOI: 10.1186/s40478-024-01851-7] [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: 04/29/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Wolfram syndrome (WS) is a rare childhood disease characterized by diabetes mellitus, diabetes insipidus, blindness, deafness, neurodegeneration and eventually early death, due to autosomal recessive mutations in the WFS1 (and WFS2) gene. While it is categorized as a neurodegenerative disease, it is increasingly becoming clear that other cell types besides neurons may be affected and contribute to the pathogenesis. MRI studies in patients and phenotyping studies in WS rodent models indicate white matter/myelin loss, implicating a role for oligodendroglia in WS-associated neurodegeneration. In this study, we sought to determine if oligodendroglia are affected in WS and whether their dysfunction may be the primary cause of the observed optic neuropathy and brain neurodegeneration. We demonstrate that 7.5-month-old Wfs1∆exon8 mice display signs of abnormal myelination and a reduced number of oligodendrocyte precursor cells (OPCs) as well as abnormal axonal conduction in the optic nerve. An MRI study of the brain furthermore revealed grey and white matter loss in the cerebellum, brainstem, and superior colliculus, as is seen in WS patients. To further dissect the role of oligodendroglia in WS, we performed a transcriptomics study of WS patient iPSC-derived OPCs and pre-myelinating oligodendrocytes. Transcriptional changes compared to isogenic control cells were found for genes with a role in ER function. However, a deep phenotyping study of these WS patient iPSC-derived oligodendroglia unveiled normal differentiation, mitochondria-associated endoplasmic reticulum (ER) membrane interactions and mitochondrial function, and no overt signs of ER stress. Overall, the current study indicates that oligodendroglia functions are largely preserved in the WS mouse and patient iPSC-derived models used in this study. These findings do not support a major defect in oligodendroglia function as the primary cause of WS, and warrant further investigation of neurons and neuron-oligodendroglia interactions as a target for future neuroprotective or -restorative treatments for WS.
Collapse
Affiliation(s)
- K Ahuja
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - M Vandenabeele
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - F Nami
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - E Lefevere
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - J Van Hoecke
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - S Bergmans
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - M Claes
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - T Vervliet
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - K Neyrinck
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - T Burg
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- VIB Center for Brain & Disease Research, Laboratory of Neurobiology, VIB-KU Leuven, Leuven, Belgium
| | - D De Herdt
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - P Bhaskar
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Y Zhu
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Z J Looser
- Institute of Pharmacology and Toxicology, Neuroscience Center Zurich, University of Zurich, University and ETH Zurich, Zurich, Switzerland
| | - J Loncke
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - W Gsell
- Biomedical MRI Group/MoSAIC, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - M Plaas
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - P Agostinis
- Laboratory for Cell Death Research & Therapy, Department of Cellular and Molecular Medicine, Leuven Center for Cancer Biology, VIB-KU, Leuven Cancer Institute, VIB-KU Leuven, Leuven, Belgium
| | - J V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - L Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- VIB Center for Brain & Disease Research, Laboratory of Neurobiology, VIB-KU Leuven, Leuven, Belgium
| | - G Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - A S Saab
- Institute of Pharmacology and Toxicology, Neuroscience Center Zurich, University of Zurich, University and ETH Zurich, Zurich, Switzerland
| | - E Wolfs
- Laboratory for Functional Imaging and Research on Stem Cells, BIOMED, UHasselt - Hasselt University, Diepenbeek, Belgium
| | - Y C Chai
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - U Himmelreich
- Biomedical MRI Group/MoSAIC, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - C Verfaillie
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - L Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - L De Groef
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
24
|
Fukushima N, Miyamoto Y, Yamauchi J. Knockdown of Rab9 Recovers Defective Morphological Differentiation Induced by Chemical ER Stress Inducer or PMD-Associated PLP1 Mutant Protein in FBD-102b Cells. PATHOPHYSIOLOGY 2024; 31:420-435. [PMID: 39311306 PMCID: PMC11417737 DOI: 10.3390/pathophysiology31030032] [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: 05/29/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/26/2024] Open
Abstract
Small GTP-binding proteins of the Rab family regulate intracellular vesicle trafficking across many aspects of the transport system. Among these, Rab9 is recognized for its role in controlling the transport system not only around the trans-Golgi network but also around the late endosome. However, the specific functions across different cell types and tissues remain unclear. Here, for the first time, we report that Rab9 negatively regulates morphological changes in the FBD-102b cell line, an oligodendroglial precursor cell line undergoing morphological differentiation. The knockdown of Rab9 led to an increase in cell shape alterations characterized by widespread membrane extensions. These changes were accompanied by increased expression levels of oligodendroglial cell differentiation and myelination marker proteins. Notably, the knockdown of Rab9 was capable of recovering defective cell morphological changes induced by tunicamycin, an inducer of endoplasmic reticulum (ER) stress, which is one of the major causes of oligodendroglial cell diseases such as Pelizaeus-Merzbacher disease (PMD, currently known as hypomyelinating leukodystrophy type 1 [HLD1]). In addition, Rab9 knockdown recovered levels of ER stress marker proteins and differentiation markers. Similar results were obtained in the cases of dithiothreitol (DTT), another chemical ER stress inducer, as well as HLD1-associated proteolipid protein 1 (PLP1) mutant protein. These results indicate a unique role for Rab9 in oligodendroglial cell morphological changes, suggesting its potential as a therapeutic target for mitigating diseases such as HLD1 at the molecular and cellular levels.
Collapse
Affiliation(s)
- Nana Fukushima
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (N.F.); (Y.M.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (N.F.); (Y.M.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (N.F.); (Y.M.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
25
|
Späte E, Zhou B, Sun T, Kusch K, Asadollahi E, Siems SB, Depp C, Werner HB, Saher G, Hirrlinger J, Möbius W, Nave KA, Goebbels S. Downregulated expression of lactate dehydrogenase in adult oligodendrocytes and its implication for the transfer of glycolysis products to axons. Glia 2024; 72:1374-1391. [PMID: 38587131 DOI: 10.1002/glia.24533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
Oligodendrocytes and astrocytes are metabolically coupled to neuronal compartments. Pyruvate and lactate can shuttle between glial cells and axons via monocarboxylate transporters. However, lactate can only be synthesized or used in metabolic reactions with the help of lactate dehydrogenase (LDH), a tetramer of LDHA and LDHB subunits in varying compositions. Here we show that mice with a cell type-specific disruption of both Ldha and Ldhb genes in oligodendrocytes lack a pathological phenotype that would be indicative of oligodendroglial dysfunctions or lack of axonal metabolic support. Indeed, when combining immunohistochemical, electron microscopical, and in situ hybridization analyses in adult mice, we found that the vast majority of mature oligodendrocytes lack detectable expression of LDH. Even in neurodegenerative disease models and in mice under metabolic stress LDH was not increased. In contrast, at early development and in the remyelinating brain, LDHA was readily detectable in immature oligodendrocytes. Interestingly, by immunoelectron microscopy LDHA was particularly enriched at gap junctions formed between adjacent astrocytes and at junctions between astrocytes and oligodendrocytes. Our data suggest that oligodendrocytes metabolize lactate during development and remyelination. In contrast, for metabolic support of axons mature oligodendrocytes may export their own glycolysis products as pyruvate rather than lactate. Lacking LDH, these oligodendrocytes can also "funnel" lactate through their "myelinic" channels between gap junction-coupled astrocytes and axons without metabolizing it. We suggest a working model, in which the unequal cellular distribution of LDH in white matter tracts facilitates a rapid and efficient transport of glycolysis products among glial and axonal compartments.
Collapse
Affiliation(s)
- Erik Späte
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Baoyu Zhou
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ting Sun
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Laboratory of Molecular Neurobiology, Department of Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Ebrahim Asadollahi
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sophie B Siems
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Constanze Depp
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Johannes Hirrlinger
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
26
|
Shen X, Caverzasi E, Yang Y, Liu X, Green A, Henry RG, Emir U, Larson PEZ. 3D balanced SSFP UTE MRI for multiple contrasts whole brain imaging. Magn Reson Med 2024; 92:702-714. [PMID: 38525680 DOI: 10.1002/mrm.30093] [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: 08/18/2023] [Revised: 02/14/2024] [Accepted: 03/05/2024] [Indexed: 03/26/2024]
Abstract
PURPOSE This study aimed to develop a new high-resolution MRI sequence for the imaging of the ultra-short transverse relaxation time (uT2) components in the brain, while simultaneously providing proton density (PD) contrast for reference and quantification. THEORY The sequence combines low flip angle balanced SSFP (bSSFP) and UTE techniques, together with a 3D dual-echo rosette k-space trajectory for readout. METHODS The expected image contrast was evaluated by simulations. A study cohort of six healthy volunteers and eight multiple sclerosis (MS) patients was recruited to test the proposed sequence. Subtraction between two TEs was performed to extract uT2 signals. In addition, conventional longitudinal relaxation time (T1) weighted, T2-weighted, and PD-weighted MRI sequences were also acquired for comparison. RESULTS Typical PD-contrast was found in the second TE images, while uT2 signals were selectively captured in the first TE images. The subtraction images presented signals primarily originating from uT2 components, but only if the first TE is short enough. Lesions in the MS subjects showed hyperintense signals in the second TE images but were hypointense signals in the subtraction images. The lesions had significantly lower signal intensity in subtraction images than normal white matter (WM), which indicated a reduction of uT2 components likely associated with myelin. CONCLUSION 3D isotropic sub-millimeter (0.94 mm) spatial resolution images were acquired with the novel bSSFP UTE sequence within 3 min. It provided easy extraction of uT2 signals and PD-contrast for reference within a single acquisition.
Collapse
Affiliation(s)
- Xin Shen
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Eduardo Caverzasi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Department of Neuroradiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Yang Yang
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Xiaoxi Liu
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Ari Green
- Neurology, University of California San Francisco, San Francisco, California, USA
| | - Roland G Henry
- Neurology, University of California San Francisco, San Francisco, California, USA
| | - Uzay Emir
- School of Health Science, Purdue University, West Lafayette, Indiana, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Peder E Z Larson
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
27
|
Sawaguchi S, Ishida M, Miyamoto Y, Yamauchi J. Hypomyelination Leukodystrophy 16 (HLD16)-Associated Mutation p.Asp252Asn of TMEM106B Blunts Cell Morphological Differentiation. Curr Issues Mol Biol 2024; 46:8088-8103. [PMID: 39194695 DOI: 10.3390/cimb46080478] [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: 05/22/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Transmembrane protein 106B (TMEM106B), which is a type II transmembrane protein, is believed to be involved in intracellular dynamics and morphogenesis in the lysosome. TMEM106B is known to be a risk factor for frontotemporal lobar degeneration and has been recently identified as the receptor needed for the entry of SARS-CoV-2, independently of angiotensin-converting enzyme 2 (ACE2). A missense mutation, p.Asp252Asn, of TMEM106B is associated with hypomyelinating leukodystrophy 16 (HLD16), which is an oligodendroglial cell-related white matter disorder causing thin myelin sheaths or myelin deficiency in the central nervous system (CNS). However, it remains to be elucidated how the mutated TMEM106B affects oligodendroglial cells. Here, we show that the TMEM106B mutant protein fails to exhibit lysosome distribution in the FBD-102b cell line, an oligodendroglial precursor cell line undergoing differentiation. In contrast, wild-type TMEM106B was indeed localized in the lysosome. Cells harboring wild-type TMEM106B differentiated into ones with widespread membranes, whereas cells harboring mutated TMEM106B failed to differentiate. It is of note that the output of signaling through the lysosome-resident mechanistic target of rapamycin (mTOR) was greatly decreased in cells harboring mutated TMEM106B. Furthermore, treatment with hesperetin, a citrus flavonoid known as an activator of mTOR signaling, restored the molecular and cellular phenotypes induced by the TMEM106B mutant protein. These findings suggest the potential pathological mechanisms underlying HLD16 and their amelioration.
Collapse
Affiliation(s)
- Sui Sawaguchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Miki Ishida
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
28
|
Marangon D, Castro e Silva JH, Cerrato V, Boda E, Lecca D. Oligodendrocyte Progenitors in Glial Scar: A Bet on Remyelination. Cells 2024; 13:1024. [PMID: 38920654 PMCID: PMC11202012 DOI: 10.3390/cells13121024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) represent a subtype of glia, giving rise to oligodendrocytes, the myelin-forming cells in the central nervous system (CNS). While OPCs are highly proliferative during development, they become relatively quiescent during adulthood, when their fate is strictly influenced by the extracellular context. In traumatic injuries and chronic neurodegenerative conditions, including those of autoimmune origin, oligodendrocytes undergo apoptosis, and demyelination starts. Adult OPCs become immediately activated; they migrate at the lesion site and proliferate to replenish the damaged area, but their efficiency is hampered by the presence of a glial scar-a barrier mainly formed by reactive astrocytes, microglia and the deposition of inhibitory extracellular matrix components. If, on the one hand, a glial scar limits the lesion spreading, it also blocks tissue regeneration. Therapeutic strategies aimed at reducing astrocyte or microglia activation and shifting them toward a neuroprotective phenotype have been proposed, whereas the role of OPCs has been largely overlooked. In this review, we have considered the glial scar from the perspective of OPCs, analysing their behaviour when lesions originate and exploring the potential therapies aimed at sustaining OPCs to efficiently differentiate and promote remyelination.
Collapse
Affiliation(s)
- Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| | - Juliana Helena Castro e Silva
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| | - Valentina Cerrato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; (V.C.); (E.B.)
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; (V.C.); (E.B.)
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| |
Collapse
|
29
|
Kawaguchi S, Kan H, Uchida Y, Kasai H, Hiwatashi A, Ueki Y. Anisotropy of the R1/T2* value dependent on white matter fiber orientation with respect to the B0 field. Magn Reson Imaging 2024; 109:83-90. [PMID: 38387713 DOI: 10.1016/j.mri.2024.02.010] [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: 12/26/2023] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
The R1 (1/T1) map divided by the T2* map (R1/T2* map) draws attention as a high-resolution myelin-related map. However, both R1 and R2* (1/T2*) values demonstrate anisotropy dependent on the white matter (WM) fiber orientation with respect to the static magnetic (B0) field. Therefore, this study primarily aimed to investigate the comprehensive impact of these angular-dependent anisotropies on the R1/T2* value. This study enrolled 10 healthy human volunteers (age = 25 ± 1.3) on the 3.0 T MRI system. For R1/T2* map calculation, whole brain R1 and T2* maps were repeatedly obtained in three head tilt positions by magnetization-prepared two rapid gradient echoes and multiple spoiled gradient echo sequences, respectively. Afterward, all maps were spatially normalized and registered to the Johns Hopkins University WM atlas. R1/T2*, R1, and R2* values were binned for fiber orientation related to the B0 field, which was estimated from diffusion-weighted echo-planar imaging data with 3° intervals, to investigate angular-dependent anisotropies in vivo. A larger change in the R1/T2* value in the global WM region as a function of fiber orientation with respect to the B0 field was observed compared to the R1 and R2* values alone. The minimum R1/T2* value at the near magic-angle range was 18.86% lower than the maximum value at the perpendicular angle range. Furthermore, R1/T2* values in the corpus callosum tract and the right and left cingulum cingulate gyrus tracts changed among the three head tilt positions due to fiber orientation changes. In conclusion, the R1/T2* value demonstrates distinctive and complicated angular-dependent anisotropy indicating the trends of both R1 and R2* values and may provide supplemental information for detecting slight changes in the microstructure of myelin and axons.
Collapse
Affiliation(s)
| | - Hirohito Kan
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Japan; Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Japan.
| | - Yuto Uchida
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Harumasa Kasai
- Department of Radiology, Nagoya City University Hospital, Japan
| | - Akio Hiwatashi
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Yoshino Ueki
- Department of Rehabilitation Medicine, Nagoya City University Graduate School of Medical Sciences, Japan
| |
Collapse
|
30
|
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
|
31
|
Huang Z, Jordan JD, Zhang Q. Myelin Pathology in Alzheimer's Disease: Potential Therapeutic Opportunities. Aging Dis 2024; 15:698-713. [PMID: 37548935 PMCID: PMC10917545 DOI: 10.14336/ad.2023.0628] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by memory loss and cognitive decline. Despite significant efforts over several decades, our understanding of the pathophysiology of this disease is still incomplete. Myelin is a multi-layered membrane structure ensheathing neuronal axons, which is essential for the fast and effective propagation of action potentials along the axons. Recent studies highlight the critical involvement of myelin in memory consolidation and reveal its vulnerability in various pathological conditions. Notably, apart from the classic amyloid hypothesis, myelin degeneration has been proposed as another critical pathophysiological feature of AD, which could occur prior to the development of amyloid pathology. Here, we review recent works supporting the critical role of myelin in cognition and myelin pathology during AD progression, with a focus on the mechanisms underlying myelin degeneration in AD. We also discuss the complex intersections between myelin pathology and typical AD pathophysiology, as well as the therapeutic potential of pro-myelinating approaches for this disease. Overall, these findings implicate myelin degeneration as a critical contributor to AD-related cognitive deficits and support targeting myelin repair as a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| | - J. Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| |
Collapse
|
32
|
Trimarco A, Audano M, Marca RL, Cariello M, Falco M, Pedretti S, Imperato G, Cestaro A, Podini P, Dina G, Quattrini A, Massimino L, Caruso D, Mitro N, Taveggia C. Prostaglandin D2 synthase controls Schwann cells metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582775. [PMID: 38496560 PMCID: PMC10942270 DOI: 10.1101/2024.02.29.582775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
We previously reported that in the absence of Prostaglandin D2 synthase (L-PGDS) peripheral nerves are hypomyelinated in development and that with aging they present aberrant myelin sheaths. We now demonstrate that L-PGDS expressed in Schwann cells is part of a coordinated program aiming at preserving myelin integrity. In vivo and in vitro lipidomic, metabolomic and transcriptomic analyses confirmed that myelin lipids composition, Schwann cells energetic metabolism and key enzymes controlling these processes are altered in the absence of L-PGDS. Moreover, Schwann cells undergo a metabolic rewiring and turn to acetate as the main energetic source. Further, they produce ketone bodies to ensure glial cell and neuronal survival. Importantly, we demonstrate that all these changes correlate with morphological myelin alterations and describe the first physiological pathway implicated in preserving PNS myelin. Collectively, we posit that myelin lipids serve as a reservoir to provide ketone bodies, which together with acetate represent the adaptive substrates Schwann cells can rely on to sustain the axo-glial unit and preserve the integrity of the PNS.
Collapse
|
33
|
Selim MK, Harel M, De Santis S, Perini I, Sommer WH, Heilig M, Zangen A, Canals S. Repetitive deep TMS in alcohol dependent patients halts progression of white matter changes in early abstinence. Psychiatry Clin Neurosci 2024; 78:176-185. [PMID: 38085120 PMCID: PMC11488632 DOI: 10.1111/pcn.13624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/04/2023] [Accepted: 11/15/2023] [Indexed: 03/13/2024]
Abstract
AIM Alcohol use disorder (AUD) is the most prevalent form of addiction, with a great burden on society and limited treatment options. A recent clinical trial reported significant clinical benefits of deep transcranial magnetic stimulations (Deep TMS) targeting midline frontocortical areas. However, the underlying biological substrate remained elusive. Here, we report the effect of Deep TMS on the microstructure of white matter. METHODS A total of 37 (14 females) AUD treatment-seeking patients were randomized to sham or active Deep TMS. Twenty (six females) age-matched healthy controls were included. White matter integrity was evaluated by fractional anisotropy (FA). Secondary measures included brain functional connectivity and self-reports of craving and drinking units in the 3 months of follow-up period. RESULTS White matter integrity was compromised in patients with AUD relative to healthy controls, as reflected by the widespread reduction in FA. This alteration progressed during early abstinence (3 weeks) in the absence of Deep TMS. However, stimulation of midline frontocortical areas arrested the progression of FA changes in association with decreased craving and relapse scores. Reconstruction of axonal tracts from white-matter regions showing preserved FA values identified cortical regions in the posterior cingulate and dorsomedial prefrontal cortices where functional connectivity was persistently modulated. These effects were absent in the sham-stimulated group. CONCLUSIONS By integrating brain structure and function to characterize the alcohol-dependent brain, this study provides mechanistic insights into the TMS effect, pointing to myelin plasticity as a possible mediator.
Collapse
Affiliation(s)
- Mohamed Kotb Selim
- Instituto de NeurocienciasConsejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernández (UMH)Sant Joan d'AlacantSpain
| | - Maayan Harel
- Department of Life SciencesBen‐Gurion UniversityBeer ShevaIsrael
- Zlotowski Center for NeuroscienceBen‐Gurion UniversityBeer ShevaIsrael
| | - Silvia De Santis
- Instituto de NeurocienciasConsejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernández (UMH)Sant Joan d'AlacantSpain
| | - Irene Perini
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical SciencesLinköping University HospitalLinköpingSweden
| | - Wolfgang H. Sommer
- Department of Addiction Medicine, Department of Clinical PsychologyMedical Faculty Mannheim, Central Institute of Mental Health, University of HeidelbergMannheimGermany
| | - Markus Heilig
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical SciencesLinköping University HospitalLinköpingSweden
| | - Abraham Zangen
- Department of Life SciencesBen‐Gurion UniversityBeer ShevaIsrael
- Zlotowski Center for NeuroscienceBen‐Gurion UniversityBeer ShevaIsrael
| | - Santiago Canals
- Instituto de NeurocienciasConsejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernández (UMH)Sant Joan d'AlacantSpain
| |
Collapse
|
34
|
Oligodendroglial metabolic support of myelinated axons driven by K + dynamics. Nat Neurosci 2024; 27:397-398. [PMID: 38267525 DOI: 10.1038/s41593-023-01559-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
|
35
|
Looser ZJ, Faik Z, Ravotto L, Zanker HS, Jung RB, Werner HB, Ruhwedel T, Möbius W, Bergles DE, Barros LF, Nave KA, Weber B, Saab AS. Oligodendrocyte-axon metabolic coupling is mediated by extracellular K + and maintains axonal health. Nat Neurosci 2024; 27:433-448. [PMID: 38267524 PMCID: PMC10917689 DOI: 10.1038/s41593-023-01558-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
The integrity of myelinated axons relies on homeostatic support from oligodendrocytes (OLs). To determine how OLs detect axonal spiking and how rapid axon-OL metabolic coupling is regulated in the white matter, we studied activity-dependent calcium (Ca2+) and metabolite fluxes in the mouse optic nerve. We show that fast axonal spiking triggers Ca2+ signaling and glycolysis in OLs. OLs detect axonal activity through increases in extracellular potassium (K+) concentrations and activation of Kir4.1 channels, thereby regulating metabolite supply to axons. Both pharmacological inhibition and OL-specific inactivation of Kir4.1 reduce the activity-induced axonal lactate surge. Mice lacking oligodendroglial Kir4.1 exhibit lower resting lactate levels and altered glucose metabolism in axons. These early deficits in axonal energy metabolism are associated with late-onset axonopathy. Our findings reveal that OLs detect fast axonal spiking through K+ signaling, making acute metabolic coupling possible and adjusting the axon-OL metabolic unit to promote axonal health.
Collapse
Affiliation(s)
- Zoe J Looser
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Zainab Faik
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Henri S Zanker
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Ramona B Jung
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
36
|
Lombardi M, Scaroni F, Gabrielli M, Raffaele S, Bonfanti E, Filipello F, Giussani P, Picciolini S, de Rosbo NK, Uccelli A, Golia MT, D’Arrigo G, Rubino T, Hooshmand K, Legido-Quigley C, Fenoglio C, Gualerzi A, Fumagalli M, Verderio C. Extracellular vesicles released by microglia and macrophages carry endocannabinoids which foster oligodendrocyte differentiation. Front Immunol 2024; 15:1331210. [PMID: 38464529 PMCID: PMC10921360 DOI: 10.3389/fimmu.2024.1331210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/01/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction Microglia and macrophages can influence the evolution of myelin lesions through the production of extracellular vesicles (EVs). While microglial EVs promote in vitro differentiation of oligodendrocyte precursor cells (OPCs), whether EVs derived from macrophages aid or limit OPC maturation is unknown. Methods Immunofluorescence analysis for the myelin protein MBP was employed to evaluate the impact of EVs from primary rat macrophages on cultured OPC differentiation. Raman spectroscopy and liquid chromatography-mass spectrometry was used to define the promyelinating lipid components of myelin EVs obtained in vitro and isolated from human plasma. Results and discussion Here we show that macrophage-derived EVs do not promote OPC differentiation, and those released from macrophages polarized towards an inflammatory state inhibit OPC maturation. However, their lipid cargo promotes OPC maturation in a similar manner to microglial EVs. We identify the promyelinating endocannabinoids anandamide and 2-arachidonoylglycerol in EVs released by both macrophages and microglia in vitro and circulating in human plasma. Analysis of OPC differentiation in the presence of the endocannabinoid receptor antagonists SR141716A and AM630 reveals a key role of vesicular endocannabinoids in OPC maturation. From this study, EV-associated endocannabinoids emerge as important mediators in microglia/macrophage-oligodendrocyte crosstalk, which may be exploited to enhance myelin repair.
Collapse
Affiliation(s)
- Marta Lombardi
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Federica Scaroni
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
| | - Martina Gabrielli
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - Elisabetta Bonfanti
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - Fabia Filipello
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Segrate, Italy
| | - Silvia Picciolini
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Nicole Kerlero de Rosbo
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
- TomaLab, Institute of Nanotechnology, CNR, Rome, Italy
| | - Antonio Uccelli
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Maria Teresa Golia
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Giulia D’Arrigo
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences (DBSV) and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - Kourosh Hooshmand
- System Medicine, Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Cristina Legido-Quigley
- System Medicine, Steno Diabetes Center Copenhagen, Copenhagen, Denmark
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Chiara Fenoglio
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
- Fondazione Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Alice Gualerzi
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - Claudia Verderio
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
37
|
Dejbakht M, Akhzari M, Jalili S, Faraji F, Barazesh M. Multiple Sclerosis: New Insights into Molecular Pathogenesis and Novel Platforms for Disease Treatment. Curr Drug Res Rev 2024; 16:175-197. [PMID: 37724675 DOI: 10.2174/2589977516666230915103730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Multiple sclerosis (MS), a chronic inflammatory disorder, affects the central nervous system via myelin degradation. The cause of MS is not fully known, but during recent years, our knowledge has deepened significantly regarding the different aspects of MS, including etiology, molecular pathophysiology, diagnosis and therapeutic options. Myelin basic protein (MBP) is the main myelin protein that accounts for maintaining the stability of the myelin sheath. Recent evidence has revealed that MBP citrullination or deamination, which is catalyzed by Ca2+ dependent peptidyl arginine deiminase (PAD) enzyme leads to the reduction of positive charge, and subsequently proteolytic cleavage of MBP. The overexpression of PAD2 in the brains of MS patients plays an essential role in new epitope formation and progression of the autoimmune disorder. Some drugs have recently entered phase III clinical trials with promising efficacy and will probably obtain approval in the near future. As different therapeutic platforms develop, finding an optimal treatment for each individual patient will be more challenging. AIMS This review provides a comprehensive insight into MS with a focus on its pathogenesis and recent advances in diagnostic methods and its present and upcoming treatment modalities. CONCLUSION MS therapy alters quickly as research findings and therapeutic options surrounding MS expand. McDonald's guidelines have created different criteria for MS diagnosis. In recent years, ever-growing interest in the development of PAD inhibitors has led to the generation of many reversible and irreversible PAD inhibitors against the disease with satisfactory therapeutic outcomes.
Collapse
Affiliation(s)
- Majid Dejbakht
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Morteza Akhzari
- School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Sajad Jalili
- Department of Orthopedics, School of Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Fouziyeh Faraji
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Mahdi Barazesh
- Department of Biotechnology, Cellular and Molecular Research Center, School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
38
|
Shao J, Lang Y, Ding M, Yin X, Cui L. Transcription Factor EB: A Promising Therapeutic Target for Ischemic Stroke. Curr Neuropharmacol 2024; 22:170-190. [PMID: 37491856 PMCID: PMC10788889 DOI: 10.2174/1570159x21666230724095558] [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: 05/08/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 07/27/2023] Open
Abstract
Transcription factor EB (TFEB) is an important endogenous defensive protein that responds to ischemic stimuli. Acute ischemic stroke is a growing concern due to its high morbidity and mortality. Most survivors suffer from disabilities such as numbness or weakness in an arm or leg, facial droop, difficulty speaking or understanding speech, confusion, impaired balance or coordination, or loss of vision. Although TFEB plays a neuroprotective role, its potential effect on ischemic stroke remains unclear. This article describes the basic structure, regulation of transcriptional activity, and biological roles of TFEB relevant to ischemic stroke. Additionally, we explore the effects of TFEB on the various pathological processes underlying ischemic stroke and current therapeutic approaches. The information compiled here may inform clinical and basic studies on TFEB, which may be an effective therapeutic drug target for ischemic stroke.
Collapse
Affiliation(s)
- Jie Shao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yue Lang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Manqiu Ding
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiang Yin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Cui
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
39
|
Wang J, Ding X, Li C, Huang C, Ke C, Xu C, Wan C. Early exercise intervention promotes myelin repair in the brains of ischemic rats by inhibiting the MEK/ERK pathway. Transl Neurosci 2024; 15:20220335. [PMID: 38511170 PMCID: PMC10951688 DOI: 10.1515/tnsci-2022-0335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/22/2024] Open
Abstract
Our previous studies have shown that early exercise intervention after stroke increases neural activity and synaptic plasticity and promotes the recovery of nerve fiber bundle integrity in the brain. However, the effect of exercise on the repair of myelin in the brain and the related mechanism are still unclear. In this study, we randomly divided the rats into three groups. Before and after 28 days of intervention, body weight, nerve function, the infarct size, white matter fiber bundle integrity, and nerve myelin structure and function were observed by measuring body weight, analysis of modified neurological severity score, CatWalk gait analysis, MRI, luxol fast blue staining, immunofluorescence, and transmission electron microscopy. Changes in the expression of proteins in the MEK/ERK pathway were assessed. The results showed that early exercise intervention resulted in neurological recovery, decreased the infarct volume and increased nerve fiber integrity, the myelin coverage area, myelin basic protein (MBP) fluorescence intensity expression, and myelin thickness. Furthermore, the expression level of MBP was significantly increased after early exercise intervention, while the expression levels of p-MEK1/2 and p-ERK1/2 were significantly reduced. In the cell study, MBP expression levels were significantly higher in the oxygen and glucose deprivation and administration group.In summary, early exercise intervention after stroke can promote myelin repair by inhibiting the MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Junyi Wang
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xinyu Ding
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chen Li
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chuan Huang
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Changkai Ke
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chunlei Xu
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chunxiao Wan
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| |
Collapse
|
40
|
Bellanca CM, Augello E, Mariottini A, Bonaventura G, La Cognata V, Di Benedetto G, Cantone AF, Attaguile G, Di Mauro R, Cantarella G, Massacesi L, Bernardini R. Disease Modifying Strategies in Multiple Sclerosis: New Rays of Hope to Combat Disability? Curr Neuropharmacol 2024; 22:1286-1326. [PMID: 38275058 PMCID: PMC11092922 DOI: 10.2174/1570159x22666240124114126] [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: 05/04/2023] [Revised: 08/21/2023] [Accepted: 09/22/2023] [Indexed: 01/27/2024] Open
Abstract
Multiple sclerosis (MS) is the most prevalent chronic autoimmune inflammatory- demyelinating disorder of the central nervous system (CNS). It usually begins in young adulthood, mainly between the second and fourth decades of life. Usually, the clinical course is characterized by the involvement of multiple CNS functional systems and by different, often overlapping phenotypes. In the last decades, remarkable results have been achieved in the treatment of MS, particularly in the relapsing- remitting (RRMS) form, thus improving the long-term outcome for many patients. As deeper knowledge of MS pathogenesis and respective molecular targets keeps growing, nowadays, several lines of disease-modifying treatments (DMT) are available, an impressive change compared to the relative poverty of options available in the past. Current MS management by DMTs is aimed at reducing relapse frequency, ameliorating symptoms, and preventing clinical disability and progression. Notwithstanding the relevant increase in pharmacological options for the management of RRMS, research is now increasingly pointing to identify new molecules with high efficacy, particularly in progressive forms. Hence, future efforts should be concentrated on achieving a more extensive, if not exhaustive, understanding of the pathogenetic mechanisms underlying this phase of the disease in order to characterize novel molecules for therapeutic intervention. The purpose of this review is to provide a compact overview of the numerous currently approved treatments and future innovative approaches, including neuroprotective treatments as anti-LINGO-1 monoclonal antibody and cell therapies, for effective and safe management of MS, potentially leading to a cure for this disease.
Collapse
Affiliation(s)
- Carlo Maria Bellanca
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, 95123 Catania, Italy
- Clinical Toxicology Unit, University Hospital, University of Catania, 95123 Catania, Italy
| | - Egle Augello
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, 95123 Catania, Italy
- Clinical Toxicology Unit, University Hospital, University of Catania, 95123 Catania, Italy
| | - Alice Mariottini
- Department of Neurosciences Drugs and Child Health, University of Florence, Florence, Italy
| | - Gabriele Bonaventura
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council, 95126 Catania, Italy
| | - Valentina La Cognata
- Institute for Biomedical Research and Innovation (IRIB), Italian National Research Council, 95126 Catania, Italy
| | - Giulia Di Benedetto
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, 95123 Catania, Italy
- Clinical Toxicology Unit, University Hospital, University of Catania, 95123 Catania, Italy
| | - Anna Flavia Cantone
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Giuseppe Attaguile
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Rosaria Di Mauro
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Giuseppina Cantarella
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Luca Massacesi
- Department of Neurosciences Drugs and Child Health, University of Florence, Florence, Italy
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, 95123 Catania, Italy
- Clinical Toxicology Unit, University Hospital, University of Catania, 95123 Catania, Italy
| |
Collapse
|
41
|
Yu F, Iacono D, Perl DP, Lai C, Gill J, Le TQ, Lee P, Sukumar G, Armstrong RC. Neuronal tau pathology worsens late-phase white matter degeneration after traumatic brain injury in transgenic mice. Acta Neuropathol 2023; 146:585-610. [PMID: 37578550 PMCID: PMC10499978 DOI: 10.1007/s00401-023-02622-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Traumatic brain injury (TBI) causes diffuse axonal injury which can produce chronic white matter pathology and subsequent post-traumatic neurodegeneration with poor patient outcomes. Tau modulates axon cytoskeletal functions and undergoes phosphorylation and mis-localization in neurodegenerative disorders. The effects of tau pathology on neurodegeneration after TBI are unclear. We used mice with neuronal expression of human mutant tau to examine effects of pathological tau on white matter pathology after TBI. Adult male and female hTau.P301S (Tg2541) transgenic and wild-type (Wt) mice received either moderate single TBI (s-TBI) or repetitive mild TBI (r-mTBI; once daily × 5), or sham procedures. Acutely, s-TBI produced more extensive axon damage in the corpus callosum (CC) as compared to r-mTBI. After s-TBI, significant CC thinning was present at 6 weeks and 4 months post-injury in Wt and transgenic mice, with homozygous tau expression producing additional pathology of late demyelination. In contrast, r-mTBI did not produce significant CC thinning except at the chronic time point of 4 months in homozygous mice, which exhibited significant CC atrophy (- 29.7%) with increased microgliosis. Serum neurofilament light quantification detected traumatic axonal injury at 1 day post-TBI in Wt and homozygous mice. At 4 months, high tau and neurofilament in homozygous mice implicated tau in chronic axon pathology. These findings did not have sex differences detected. Conclusions: Neuronal tau pathology differentially exacerbated CC pathology based on injury severity and chronicity. Ongoing CC atrophy from s-TBI became accompanied by late demyelination. Pathological tau significantly worsened CC atrophy during the chronic phase after r-mTBI.
Collapse
Affiliation(s)
- Fengshan Yu
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Diego Iacono
- Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Defense-Uniformed Services University Brain Tissue Repository, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Daniel P Perl
- Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Defense-Uniformed Services University Brain Tissue Repository, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Chen Lai
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - Tuan Q Le
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Patricia Lee
- Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Defense-Uniformed Services University Brain Tissue Repository, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Gauthaman Sukumar
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Regina C Armstrong
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA.
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
42
|
Moura D, Parvathaneni A, Sahagun A, Noguchi H, Garcia J, Brennan E, Brock R, Tilton I, Halladay L, Pleasure S, Cocas L. Neuronal Activity Changes the Number of Neurons That Are Synaptically Connected to OPCs. eNeuro 2023; 10:ENEURO.0126-23.2023. [PMID: 37813563 PMCID: PMC10598642 DOI: 10.1523/eneuro.0126-23.2023] [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: 04/20/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 10/17/2023] Open
Abstract
The timing and specificity of oligodendrocyte myelination during development, as well as remyelination after injury or immune attack, remain poorly understood. Recent work has shown that oligodendrocyte progenitors receive synapses from neurons, providing a potential mechanism for neuronal-glial communication. In this study, we investigated the importance of these neuroglial connections in myelination during development and during neuronal plasticity in the mouse hippocampus. We used chemogenetic tools and viral monosynaptic circuit tracing to analyze these connections and to examine oligodendrocyte progenitor cells (OPCs) proliferation, myelination, synapse formation, and neuronal-glial connectivity in vivo after increasing or decreasing neuronal activity levels. We found that increasing neuronal activity led to greater OPC activation and proliferation. Modulation of neuronal activity also altered the organization of neuronal-glial connections: while it did not impact the total number of RabV-labeled neuronal inputs, or the number of RabV-labeled inhibitory neuronal (IN) inputs, it did alter the number of RabV-labeled excitatory neuron to OPC connections. Overall, our findings support the idea that neuronal activity plays a crucial role in regulating OPC proliferation and activation as well as the types of neuronal inputs to OPCs, indicating that neuronal activity is important for OPC circuit composition and function.
Collapse
Affiliation(s)
- Daniela Moura
- Biology Department, Neuroscience Program, Santa Clara University, Santa Clara, CA 95053
- Neurology Department, University of California, San Francisco, San Francisco, CA 94110
| | - Alekhya Parvathaneni
- Biology Department, Neuroscience Program, Santa Clara University, Santa Clara, CA 95053
| | - Atehsa Sahagun
- Biology Department, Neuroscience Program, Santa Clara University, Santa Clara, CA 95053
| | - Hirofumi Noguchi
- Neurology Department, University of California, San Francisco, San Francisco, CA 94110
| | - Jesse Garcia
- Neurology Department, University of California, San Francisco, San Francisco, CA 94110
| | - Emma Brennan
- Biology Department, Neuroscience Program, Santa Clara University, Santa Clara, CA 95053
| | - Robert Brock
- Biology Department, Neuroscience Program, Santa Clara University, Santa Clara, CA 95053
| | - Iris Tilton
- Biology Department, Neuroscience Program, Santa Clara University, Santa Clara, CA 95053
| | - Lindsay Halladay
- Biology Department, Neuroscience Program, Santa Clara University, Santa Clara, CA 95053
| | - Samuel Pleasure
- Neurology Department, University of California, San Francisco, San Francisco, CA 94110
| | - Laura Cocas
- Biology Department, Neuroscience Program, Santa Clara University, Santa Clara, CA 95053
- Neurology Department, University of California, San Francisco, San Francisco, CA 94110
| |
Collapse
|
43
|
Fujiwara M, Ferdousi F, Isoda H. Investigation into Molecular Brain Aging in Senescence-Accelerated Mouse (SAM) Model Employing Whole Transcriptomic Analysis in Search of Potential Molecular Targets for Therapeutic Interventions. Int J Mol Sci 2023; 24:13867. [PMID: 37762170 PMCID: PMC10530366 DOI: 10.3390/ijms241813867] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
With the progression of an aging society, cognitive aging has emerged as a pressing concern necessitating attention. The senescence-accelerated mouse-prone 8 (SAMP8) model has proven instrumental in investigating the early stages of cognitive aging. Through an extensive examination of molecular changes in the brain cortex, utilizing integrated whole-genome transcriptomics, our principal aim was to uncover potential molecular targets with therapeutic applications and relevance to drug screening. Our investigation encompassed four distinct conditions, comparing the same strain at different time points (1 year vs. 16 weeks) and the same time point across different strains (SAMP8 vs. SAMR1), namely: physiological aging, accelerated aging, early events in accelerated aging, and late events in accelerated aging. Focusing on key functional alterations associated with aging in the brain, including neurogenesis, synapse dynamics, neurometabolism, and neuroinflammation, we identified candidate genes linked to these processes. Furthermore, employing protein-protein interaction (PPI) analysis, we identified pivotal hub genes involved in interactions within these functional domains. Additionally, gene-set perturbation analysis allowed us to uncover potential upstream genes or transcription factors that exhibited activation or inhibition across the four conditions. In summary, our comprehensive analysis of the SAMP8 mouse brain through whole-genome transcriptomics not only deepens our understanding of age-related changes but also lays the groundwork for a predictive model to facilitate drug screening for cognitive aging.
Collapse
Affiliation(s)
- Michitaka Fujiwara
- Graduate School of Environmental Science Program, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
- Open Innovation Laboratory for Food and Medicinal Resource Engineering, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Tennodai, Tsukuba 305-8572, Japan
| | - Farhana Ferdousi
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
| | - Hiroko Isoda
- Open Innovation Laboratory for Food and Medicinal Resource Engineering, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Tennodai, Tsukuba 305-8572, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
| |
Collapse
|
44
|
Castaldo F, Páscoa Dos Santos F, Timms RC, Cabral J, Vohryzek J, Deco G, Woolrich M, Friston K, Verschure P, Litvak V. Multi-modal and multi-model interrogation of large-scale functional brain networks. Neuroimage 2023; 277:120236. [PMID: 37355200 PMCID: PMC10958139 DOI: 10.1016/j.neuroimage.2023.120236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
Existing whole-brain models are generally tailored to the modelling of a particular data modality (e.g., fMRI or MEG/EEG). We propose that despite the differing aspects of neural activity each modality captures, they originate from shared network dynamics. Building on the universal principles of self-organising delay-coupled nonlinear systems, we aim to link distinct features of brain activity - captured across modalities - to the dynamics unfolding on a macroscopic structural connectome. To jointly predict connectivity, spatiotemporal and transient features of distinct signal modalities, we consider two large-scale models - the Stuart Landau and Wilson and Cowan models - which generate short-lived 40 Hz oscillations with varying levels of realism. To this end, we measure features of functional connectivity and metastable oscillatory modes (MOMs) in fMRI and MEG signals - and compare them against simulated data. We show that both models can represent MEG functional connectivity (FC), functional connectivity dynamics (FCD) and generate MOMs to a comparable degree. This is achieved by adjusting the global coupling and mean conduction time delay and, in the WC model, through the inclusion of balance between excitation and inhibition. For both models, the omission of delays dramatically decreased the performance. For fMRI, the SL model performed worse for FCD and MOMs, highlighting the importance of balanced dynamics for the emergence of spatiotemporal and transient patterns of ultra-slow dynamics. Notably, optimal working points varied across modalities and no model was able to achieve a correlation with empirical FC higher than 0.4 across modalities for the same set of parameters. Nonetheless, both displayed the emergence of FC patterns that extended beyond the constraints of the anatomical structure. Finally, we show that both models can generate MOMs with empirical-like properties such as size (number of brain regions engaging in a mode) and duration (continuous time interval during which a mode appears). Our results demonstrate the emergence of static and dynamic properties of neural activity at different timescales from networks of delay-coupled oscillators at 40 Hz. Given the higher dependence of simulated FC on the underlying structural connectivity, we suggest that mesoscale heterogeneities in neural circuitry may be critical for the emergence of parallel cross-modal functional networks and should be accounted for in future modelling endeavours.
Collapse
Affiliation(s)
- Francesca Castaldo
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, London, United Kingdom.
| | - Francisco Páscoa Dos Santos
- Eodyne Systems SL, Barcelona, Spain; Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ryan C Timms
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Joana Cabral
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - Portuguese Government Associate Laboratory, Braga/Guimarães, Portugal; Centre for Eudaimonia and Human Flourishing, Linacre College, University of Oxford, United United Kingdom
| | - Jakub Vohryzek
- Centre for Eudaimonia and Human Flourishing, Linacre College, University of Oxford, United United Kingdom; Centre for Brain and Cognition, Computational Neuroscience Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Gustavo Deco
- Centre for Brain and Cognition, Computational Neuroscience Group, Universitat Pompeu Fabra, Barcelona, Spain; Institució Catalana de la Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Department of Neuropsychology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; School of Psychological Sciences, Monash University, Melbourne, Australia
| | - Mark Woolrich
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Karl Friston
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Paul Verschure
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Vladimir Litvak
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
45
|
Xu J, Zhang B, Cai J, Peng Q, Hu J, Askar P, Shangguan J, Su W, Zhu C, Sun H, Zhou S, Chen G, Yang X, Gu Y. The transcription factor Stat-1 is essential for Schwann cell differentiation, myelination and myelin sheath regeneration. Mol Med 2023; 29:79. [PMID: 37365519 DOI: 10.1186/s10020-023-00667-w] [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: 12/08/2022] [Accepted: 05/21/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Myelin sheath is a crucial accessory to the functional nerve-fiber unit, its disruption or loss can lead to axonal degeneration and subsequent neurodegenerative diseases (NDs). Notwithstanding of substantial progress in possible molecular mechanisms underlying myelination, there is no therapeutics that prevent demyelination in NDs. Therefore, it is crucial to seek for potential intervention targets. Here, we focused on the transcriptional factor, signal transducer and activator of transcription 1 (Stat1), to explore its effects on myelination and its potential as a drug target. METHODS By analyzing the transcriptome data obtained from Schwann cells (SCs) at different stages of myelination, it was found that Stat1 might be involved in myelination. To test this, we used the following experiments: (1) In vivo, the effect of Stat1 on remyelination was observed in an in vivo myelination mode with Stat1 knockdown in sciatic nerves or specific knockdown in SCs. (2) In vitro, the RNA interference combined with cell proliferation assay, scratch assay, SC aggregate sphere migration assay, and a SC differentiation model, were used to assess the effects of Stat1 on SC proliferation, migration and differentiation. Chromatin immunoprecipitation sequencing (ChIP-Seq), RNA-Seq, ChIP-qPCR and luciferase activity reporter assay were performed to investigate the possible mechanisms of Stat1 regulating myelination. RESULTS Stat1 is important for myelination. Stat1 knockdown in nerve or in SCs reduces the axonal remyelination in the injured sciatic nerve of rats. Deletion of Stat1 in SCs blocks SC differentiation thereby inhibiting the myelination program. Stat1 interacts with the promoter of Rab11-family interacting protein 1 (Rab11fip1) to initiate SC differentiation. CONCLUSION Our findings demonstrate that Stat1 regulates SC differentiation to control myelinogenic programs and repair, uncover a novel function of Stat1, providing a candidate molecule for clinical intervention in demyelinating diseases.
Collapse
Affiliation(s)
- Jinghui Xu
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Bin Zhang
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Jieyi Cai
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Qianqian Peng
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Junxia Hu
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Parizat Askar
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Jianghong Shangguan
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Wenfeng Su
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Changlai Zhu
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Hualin Sun
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Songlin Zhou
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Gang Chen
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Xiaoming Yang
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.
| | - Yun Gu
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.
| |
Collapse
|
46
|
Kujawa MJ, Marcinkowska AB, Grzywińska M, Waśkow M, Romanowski A, Szurowska E, Winklewski PJ, Szarmach A. Physical activity and the brain myelin content in humans. Front Cell Neurosci 2023; 17:1198657. [PMID: 37342769 PMCID: PMC10277468 DOI: 10.3389/fncel.2023.1198657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/19/2023] [Indexed: 06/23/2023] Open
Abstract
New imaging sequences and biophysical models allow adopting magnetic resonance imaging (MRI) for in vivo myelin mapping in humans. Understanding myelination and remyelination processes in the brain is fundamental from the perspective of proper design of physical exercise and rehabilitation schemes that aim to slow down demyelination in the aging population and to induce remyelination in patients with neurodegenerative diseases. Therefore, in this review we strive to provide a state-of-the art summary of the existing MRI studies in humans focused on the effects of physical activity on myelination/remyelination. We present and discuss four cross-sectional and four longitudinal studies and one case report. Physical activity and an active lifestyle have a beneficial effect on the myelin content in humans. Myelin expansion can be induced in humans throughout the entire lifespan by intensive aerobic exercise. Additional research is needed to determine (1) what exercise intensity (and cognitive novelty, which is embedded in the exercise scheme) is the most beneficial for patients with neurodegenerative diseases, (2) the relationship between cardiorespiratory fitness and myelination, and (3) how exercise-induced myelination affect cognitive abilities.
Collapse
Affiliation(s)
- Mariusz J. Kujawa
- 2nd Department of Radiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Anna B. Marcinkowska
- 2nd Department of Radiology, Medical University of Gdańsk, Gdańsk, Poland
- Applied Cognitive Neuroscience Lab, Department of Neurophysiology, Neuropsychology and Neuroinformatics, Medical University of Gdańsk, Gdańsk, Poland
| | - Małgorzata Grzywińska
- Neuroinformatics and Artificial Intelligence Lab, Department of Neurophysiology, Neuropsychology and Neuroinformatics, Medical University of Gdańsk, Gdańsk, Poland
| | - Monika Waśkow
- Institute of Health Sciences, Pomeranian University in Słupsk, Słupsk, Poland
| | | | - Edyta Szurowska
- 2nd Department of Radiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Paweł J. Winklewski
- 2nd Department of Radiology, Medical University of Gdańsk, Gdańsk, Poland
- Department of Neurophysiology, Neuropsychology and Neuroinformatics, Medical University of Gdańsk, Gdańsk, Poland
| | - Arkadiusz Szarmach
- 2nd Department of Radiology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
47
|
Krämer-Albers EM, Werner HB. Mechanisms of axonal support by oligodendrocyte-derived extracellular vesicles. Nat Rev Neurosci 2023:10.1038/s41583-023-00711-y. [PMID: 37258632 DOI: 10.1038/s41583-023-00711-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/02/2023]
Abstract
Extracellular vesicles (EVs) have recently emerged as versatile elements of cell communication in the nervous system, mediating tissue homeostasis. EVs influence the physiology of their target cells via horizontal transfer of molecular cargo between cells and by triggering signalling pathways. In this Review, we discuss recent work revealing that EVs mediate interactions between oligodendrocytes and neurons, which are relevant for maintaining the structural integrity of axons. In response to neuronal activity, myelinating oligodendrocytes release EVs, which are internalized by neurons and provide axons with key factors that improve axonal transport, stress resistance and energy homeostasis. Glia-to-neuron transfer of EVs is thus a crucial facet of axonal preservation. When glial support is impaired, axonal integrity is progressively lost, as observed in myelin-related disorders, other neurodegenerative diseases and with normal ageing. We highlight the mechanisms that oligodendroglial EVs use to sustain axonal integrity and function.
Collapse
Affiliation(s)
- Eva-Maria Krämer-Albers
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
48
|
Dustin E, Suarez-Pozos E, Stotesberry C, Qiu S, Palavicini JP, Han X, Dupree JL. Compromised Myelin and Axonal Molecular Organization Following Adult-Onset Sulfatide Depletion. Biomedicines 2023; 11:1431. [PMID: 37239102 PMCID: PMC10216104 DOI: 10.3390/biomedicines11051431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
3-O-sulfogalactosylceramide, or sulfatide, is a prominent myelin glycosphingolipid reduced in the normal appearing white matter (NAWM) in Multiple Sclerosis (MS), indicating that sulfatide reduction precedes demyelination. Using a mouse model that is constitutively depleted of sulfatide, we previously demonstrated that sulfatide is essential during development for the establishment and maintenance of myelin and axonal integrity and for the stable tethering of certain myelin proteins in the sheath. Here, using an adult-onset depletion model of sulfatide, we employ a combination of ultrastructural, immunohistochemical and biochemical approaches to analyze the consequence of sulfatide depletion from the adult CNS. Our findings show a progressive loss of axonal protein domain organization, which is accompanied by axonal degeneration, with myelin sparing. Similar to our previous work, we also observe differential myelin protein anchoring stabilities that are both sulfatide dependent and independent. Most notably, stable anchoring of neurofascin155, a myelin paranodal protein that binds the axonal paranodal complex of contactin/Caspr1, requires sulfatide. Together, our findings show that adult-onset sulfatide depletion, independent of demyelination, is sufficient to trigger progressive axonal degeneration. Although the pathologic mechanism is unknown, we propose that sulfatide is required for maintaining myelin organization and subsequent myelin-axon interactions and disruptions in these interactions results in compromised axon structure and function.
Collapse
Affiliation(s)
- Elizabeth Dustin
- Research Service, Richmond Veterans Affairs Medical Center, Central Virginia Veterans Affairs Health Care System, Richmond, VA 23249, USA; (E.D.)
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond VA 23298, USA
| | - Edna Suarez-Pozos
- Research Service, Richmond Veterans Affairs Medical Center, Central Virginia Veterans Affairs Health Care System, Richmond, VA 23249, USA; (E.D.)
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond VA 23298, USA
| | - Camryn Stotesberry
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Shulan Qiu
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Juan Pablo Palavicini
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jeffrey L. Dupree
- Research Service, Richmond Veterans Affairs Medical Center, Central Virginia Veterans Affairs Health Care System, Richmond, VA 23249, USA; (E.D.)
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond VA 23298, USA
| |
Collapse
|
49
|
Wu H, Chen X, Yu B, Zhang J, Gu X, Liu W, Mei F, Ye J, Xiao L. Deficient deposition of new myelin impairs adult optic nerve function in a murine model of diabetes. Glia 2023; 71:1333-1345. [PMID: 36661098 DOI: 10.1002/glia.24341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/21/2023]
Abstract
Visual impairment in diabetes is a growing public health concern. Apart from the well-defined diabetic retinopathy, disturbed optic nerve function, which is dependent on the myelin sheath, has recently been recognized as an early feature of visual impairment in diabetes. However, the underlying cellular mechanisms remain unclear. Using a streptozotocin-induced diabetic mouse model, we observed a myelin deficiency along with a disturbed composition of oligodendroglial lineage cells in diabetic optic nerve. We found that new myelin deposition, a continuous process that lasts throughout adulthood, was diminished during pathogenesis. Genetically dampening newly generated myelin by conditionally deleting olig2 in oligodendrocyte precursor cells within this short time window extensively delayed the signal transmission of the adult optic nerve. In addition, clemastine, an antimuscarinic compound that enhances myelination, significantly restored oligodendroglia, and promoted the functional recovery of the optic nerve in diabetic mice. Together, our results point to the role of new myelin deposition in optic neuropathy under diabetic insult and provide a promising therapeutic target for restoring visual function.
Collapse
Affiliation(s)
- Haoqian Wu
- Department of Ophthalmology, Daping Hospital, Army Medical Center, Army Medical University, Chongqing, China
| | - Xianjun Chen
- Department of Physiology, Research Center of Neuroscience, College of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Bin Yu
- Department of Neurosurgery, 2nd Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jieqiong Zhang
- Department of Ophthalmology, Daping Hospital, Army Medical Center, Army Medical University, Chongqing, China
| | - Xingmei Gu
- Department of Histology and Embryology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Army Medical University, Chongqing, China
| | - Wei Liu
- Department of Ophthalmology, Daping Hospital, Army Medical Center, Army Medical University, Chongqing, China
| | - Feng Mei
- Department of Histology and Embryology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Army Medical University, Chongqing, China
| | - Jian Ye
- Department of Ophthalmology, Daping Hospital, Army Medical Center, Army Medical University, Chongqing, China
| | - Lan Xiao
- Department of Neurosurgery, 2nd Affiliated Hospital, Army Medical University, Chongqing, China
- Department of Histology and Embryology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Army Medical University, Chongqing, China
| |
Collapse
|
50
|
Masson MA, Nait-Oumesmar B. Emerging concepts in oligodendrocyte and myelin formation, inputs from the zebrafish model. Glia 2023; 71:1147-1163. [PMID: 36645033 DOI: 10.1002/glia.24336] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/17/2023]
Abstract
Oligodendrocytes (OLs) are the myelinating cells of the central nervous system (CNS), which are derived from OL precursor cells. Myelin insulates axons allowing the saltatory conduction of action potentials and also provides trophic and metabolic supports to axons. Interestingly, oligodendroglial cells have the capacity to sense neuronal activity, which regulates myelin sheath formation via the vesicular release of neurotransmitters. Neuronal activity-dependent regulation of myelination is mediated by specialized interaction between axons and oligodendroglia, involving both synaptic and extra-synaptic modes of communications. The zebrafish has provided key advantages for the study of the myelination process in the CNS. External development and transparent larval stages of this vertebrate specie combined with the existence of several transgenic reporter lines provided key advances in oligodendroglial cell biology, axo-glial interactions and CNS myelination. In this publication, we reviewed and discussed the most recent knowledge on OL development and myelin formation, with a focus on mechanisms regulating these fundamental biological processes in the zebrafish. Especially, we highlighted the critical function of axons and oligodendroglia modes of communications and calcium signaling in myelin sheath formation and growth. Finally, we reviewed the relevance of these knowledge's in demyelinating diseases and drug discovery of pharmacological compounds favoring myelin regeneration.
Collapse
Affiliation(s)
- Mary-Amélie Masson
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Brahim Nait-Oumesmar
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| |
Collapse
|