1
|
Weng C, Groh AM, Yaqubi M, Cui QL, Stratton JA, Moore GRW, Antel JP. Heterogeneity of mature oligodendrocytes in the central nervous system. Neural Regen Res 2025; 20:1336-1349. [PMID: 38934385 PMCID: PMC11624867 DOI: 10.4103/nrr.nrr-d-24-00055] [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: 01/15/2024] [Revised: 03/26/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Mature oligodendrocytes form myelin sheaths that are crucial for the insulation of axons and efficient signal transmission in the central nervous system. Recent evidence has challenged the classical view of the functionally static mature oligodendrocyte and revealed a gamut of dynamic functions such as the ability to modulate neuronal circuitry and provide metabolic support to axons. Despite the recognition of potential heterogeneity in mature oligodendrocyte function, a comprehensive summary of mature oligodendrocyte diversity is lacking. We delve into early 20 th -century studies by Robertson and Río-Hortega that laid the foundation for the modern identification of regional and morphological heterogeneity in mature oligodendrocytes. Indeed, recent morphologic and functional studies call into question the long-assumed homogeneity of mature oligodendrocyte function through the identification of distinct subtypes with varying myelination preferences. Furthermore, modern molecular investigations, employing techniques such as single cell/nucleus RNA sequencing, consistently unveil at least six mature oligodendrocyte subpopulations in the human central nervous system that are highly transcriptomically diverse and vary with central nervous system region. Age and disease related mature oligodendrocyte variation denotes the impact of pathological conditions such as multiple sclerosis, Alzheimer's disease, and psychiatric disorders. Nevertheless, caution is warranted when subclassifying mature oligodendrocytes because of the simplification needed to make conclusions about cell identity from temporally confined investigations. Future studies leveraging advanced techniques like spatial transcriptomics and single-cell proteomics promise a more nuanced understanding of mature oligodendrocyte heterogeneity. Such research avenues that precisely evaluate mature oligodendrocyte heterogeneity with care to understand the mitigating influence of species, sex, central nervous system region, age, and disease, hold promise for the development of therapeutic interventions targeting varied central nervous system pathology.
Collapse
Affiliation(s)
- Chao Weng
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Adam M.R. Groh
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Moein Yaqubi
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Qiao-Ling Cui
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Jo Anne Stratton
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - G. R. Wayne Moore
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Jack P. Antel
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
2
|
Hodebourg R, Scofield MD, Kalivas PW, Kuhn BN. Nonneuronal contributions to synaptic function. Neuron 2025:S0896-6273(25)00260-0. [PMID: 40311612 DOI: 10.1016/j.neuron.2025.04.004] [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: 01/20/2025] [Revised: 03/09/2025] [Accepted: 04/04/2025] [Indexed: 05/03/2025]
Abstract
Synapses are elegantly integrated signaling hubs containing the canonical synaptic elements, neuronal pre- and postsynapses, along with other components of the neuropil, including perisynaptic astroglia and extracellular matrix proteins, as well as microglia and oligodendrocytes. Signaling within these multipartite hubs is essential for synaptic function and is often disrupted in neuropsychiatric disorders. We review data that have refined our understanding of how environmental stimuli shape signaling and synaptic plasticity within synapses. We propose working models that integrate what is known about how different cell types within the perisynaptic neuropil regulate synaptic functions and dysfunctions that are elicited by addictive drugs. While these working models integrate existing findings, they are constrained by a need for new technology. Accordingly, we propose directions for improving reagents and experimental approaches to better probe how signaling between cell types within perisynaptic ecosystems creates the synaptic plasticity necessary to establish and maintain adaptive and maladaptive behaviors.
Collapse
Affiliation(s)
- Ritchy Hodebourg
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Anesthesiology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, SC 29401, USA.
| | - Brittany N Kuhn
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
3
|
Jia H, Wang K, Zhang P, Zhang M, Mai Y, Chu C, Yin X, Fan L, Zhang L. Cortical thickness deviations as biomarker for subtyping and prognosis in pediatric brainstem tumors. Sci Rep 2025; 15:13132. [PMID: 40240399 PMCID: PMC12003633 DOI: 10.1038/s41598-025-95017-7] [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: 09/24/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Brainstem tumors exert profound effects on cortical organization and functionality across the whole brain. However, the precise implications of changes in cortical thickness (CTh) for patient stratification and prognostic assessment remain unclear. Our study seeks to address these gaps and provide clearer insights into the distant impact of brainstem tumors. This study involved 124 pediatric patients with brainstem tumors and 849 healthy controls. Using CAT12 segmentation on 3D T1-weighted MRI scans and Gaussian process regression modeling, we established a normative CTh model from healthy data. CTh deviations of patients were quantified and clustered, revealing two distinct subtypes: Subtype 1 with extremely positive deviations and Subtype 2 with extremely negative deviations, correlating with better survival. Kaplan-Meier analysis confirmed significant survival differences between these subtypes. Additionally, a greater number of brain regions with positive CTh deviations was found to correlate with larger tumor volumes. These findings suggest that CTh deviation is a non-invasive imaging marker, facilitating patient subtyping and survival prediction. These insights equip clinicians to tailor treatment plans and establishes a valuable precision medical tool for clinical evaluation and monitoring.
Collapse
Affiliation(s)
- Heyuan Jia
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China
- Institute of Large-scale Scientific Facility and Centre for Zero Magnetic Field Science, Beihang University, Beijing, China
| | - Kaikai Wang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mingxin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yiying Mai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Congying Chu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xuntao Yin
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Lingzhong Fan
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266000, China.
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
4
|
Huang Q, Tang J, Xiang Y, Shang X, Li K, Chen L, Hu J, Li H, Pi Y, Yang H, Zhang H, Tan H, Xiyang Y, Jin H, Li X, Chen M, Mao R, Wang Q. 4-Benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione rescues oligodendrocytes ferroptosis leading to myelin loss and ameliorates neuronal injury facilitating memory in neonatal hypoxic-ischemic brain damage. Exp Neurol 2025; 390:115262. [PMID: 40246011 DOI: 10.1016/j.expneurol.2025.115262] [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: 11/28/2024] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
Neonatal brain hypoxia-ischemia (HI) is proved to cause white matter injury (WMI), which resulted in behavioral disturbance. Myelin formed by oligodendrocytes vulnerable to hypoxia-ischemia (HI), regulating motor and cognitive function, is easily damaged by HI causing myelin loss. 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8) has a potential rescue role in neuronal death post HI. Studies reported that neuronal ferroptosis could be induced by HI and linked to behavioral abnormalities. However, the effect of TDZD-8 on WMI and its involvement in memory recovery remains unclear. In this study, our HIBD model showed impaired memory function caused by neuronal injury and myelin loss. TDZD-8 effectively reversed this pathology. Underlying mechanistic exploration implied that TDZD-8 ameliorating myelin loss via ferroptosis pathway was involved in the process of TDZD-8 treating neonatal HIBD. In conclusion, our data demonstrated that combined effect of white matter repairment and neuronal protection achieved the therapeutic role of TDZD-8 in neonatal HIBD, and suggested that white matter repairment also could be a considerable clinical therapy for neonatal HIBD.
Collapse
Affiliation(s)
- Qiyi Huang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Jiahang Tang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - You Xiang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Xinying Shang
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650500, China
| | - Kunlin Li
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Lijia Chen
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Junnan Hu
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Han Li
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Yanxiong Pi
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Haiyan Yang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Huijia Zhang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Heng Tan
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Yanbin Xiyang
- Institution of Neuroscience, Kunming Medical University, Kunming 650500, China
| | - Huiyan Jin
- Department of Functional Experiment, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Xia Li
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Manjun Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650500, China
| | - Rongrong Mao
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China.
| | - Qian Wang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China.
| |
Collapse
|
5
|
Huang M, Di J, He L, Li N, Tian Z, Xiao L, Zhu R, He T, Pang M, Liu B, Rong L. Double-target magnetic stimulation attenuates oligodendrocyte apoptosis and oxidative stress impairment after spinal cord injury via GAP43. Spine J 2025; 25:820-842. [PMID: 39701305 DOI: 10.1016/j.spinee.2024.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/29/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND CONTEXT Spinal cord injury (SCI) causes neural circuit interruption and permanent functional damage. Magnetic stimulation in humans with SCI aims to engage residual neural networks to improve neurological functional, but the detailed mechanism remains unknown. PURPOSE This study evaluates functional recovery and neural circuitry improvements in rodent with double-target (brain and spinal cord) magnetic stimulation (DTMS) treatment and explores the effect of DTMS on the modulation of glial cells in vivo and in vitro. STUDY DESIGN In vivo animal study. METHODS SCI model rats at T10 level were induced via a weight-drop method and underwent long-time DTMS treatment. A series of behavioral assessments and tissue staining were used to evaluate neurological function and neural circuitry improvements. More importantly, single-cell RNA sequencing was conducted to identify the most significant glial cells after DTMS treatment. Furthermore, transmission electron microscopy, western blotting, immunofluorescence staining, TUNEL staining, Annexin V-FITC apoptosis kit and Lipid ROS kit were used to explore the mechanism underlying the observed changes. Study funding sources: National Natural Science Foundation of China (Grant number: U22A20297; Dollar amount: 62500); Key Research and Development Program of Guangzhou (Grant number: 202206060003; Dollar amount: 63750). There are no conflicts of interest or disclosures to report. RESULTS DTMS promoted the improvements of motor and sensory neural circuitry by modulating remyelination and neuronal survival, while silencing growth-associated protein 43 (GAP43) in oligodendrocytes suppressed these effects of DTMS in vivo. Mechanically, GAP43 played a crucial part to promote the branching and mature of oligodendrocytes and axonal regeneration via anti-apoptotic and antioxidative stress effects. Furthermore, oligodendrocytes subjected to magnetic stimulation exerted neuroprotective effects on neurons by secreting exosomes containing GAP43. CONCLUSIONS Our study revealed the neuroprotection of DTMS on SCI. The GAP43 in oligodendrocytes were associated with this relationship between magnetic stimulation and myelin and neuronal regeneration after SCI. CLINICAL SIGNIFICANCE The current study demonstrated the beneficial effects of DTMS on SCI based on functional, electrophysiological, cellular and histological evidence. According to these findings, we expect DTMS to make a positive and significant difference for SCI therapeutic screening.
Collapse
Affiliation(s)
- Mudan Huang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Jiawei Di
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Lei He
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Na Li
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Zhenming Tian
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Longyou Xiao
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Ruijue Zhu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Tianwei He
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Mao Pang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
6
|
Liu H, Wen S, Xu C, Kang X, Kong E. Mechanisms and functional implications of ZDHHC5 in cellular physiology and disease. J Lipid Res 2025; 66:100793. [PMID: 40180214 DOI: 10.1016/j.jlr.2025.100793] [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: 02/05/2025] [Revised: 03/27/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025] Open
Abstract
Post-translational lipid modification by palmitoylation is a reversible process crucial for maintaining cellular functionality. The palmitoyl acyltransferase zinc finger Asp-His-His-Cys motif-containing 5 (ZDHHC5) has garnered significant attention due to its roles in neurodegenerative diseases, oncogenesis, and cardiac function. ZDHHC5 recognizes substrates through diverse mechanisms and its activity is regulated by multiple factors. Highly expressed in the brain, liver, and heart, ZDHHC5 exerts regulatory functions in various cellular processes through self-regulation and substrate palmitoylation. This review summarizes ZDHHC5's regulatory roles in the nervous system, lipid metabolism and oncogenesis, highlighting its potential as a therapeutic target for neurological, lipid metabolic diseases, and cancer due to its involvement in diverse cellular processes and disease-associated dysfunctions.
Collapse
Affiliation(s)
- Huicong Liu
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, China; Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China.
| | - Shuo Wen
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Chang Xu
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Xiaohong Kang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, China
| | - Eryan Kong
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, China; Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
7
|
Wang Y, Jiang A, Yan J, Wen D, Gu N, Li Z, Sun X, Wu Y, Guo Z. Inhibition of GPR17/ID2 Axis Improve Remyelination and Cognitive Recovery after SAH by Mediating OPC Differentiation in Rat Model. Transl Stroke Res 2025; 16:178-193. [PMID: 37935878 DOI: 10.1007/s12975-023-01201-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/21/2023] [Accepted: 10/13/2023] [Indexed: 11/09/2023]
Abstract
Myelin sheath injury contributes to cognitive deficits following subarachnoid hemorrhage (SAH). G protein-coupled receptor 17 (GPR17), a membrane receptor, negatively regulates oligodendrocyte precursor cell (OPC) differentiation in both developmental and pathological contexts. Nonetheless, GPR17's role in modulating OPC differentiation, facilitating remyelination post SAH, and its interaction with downstream molecules remain elusive. In a rat SAH model induced by arterial puncture, OPCs expressing GPR17 proliferated prominently by day 14 post-onset, coinciding with compromised myelin sheath integrity and cognitive deficits. Selective Gpr17 knockdown in oligodendrocytes (OLs) via adeno-associated virus (AAV) administration revealed that reduced GPR17 levels promoted OPC differentiation, restored myelin sheath integrity, and improved cognitive deficits by day 14 post-SAH. Moreover, GPR17 knockdown attenuated the elevated expression of the inhibitor of DNA binding 2 (ID2) post-SAH, suggesting a GPR17-ID2 regulatory axis. Bi-directional modulation of ID2 expression in OLs using AAV unveiled that elevated ID2 counteracted the restorative effects of GPR17 knockdown. This resulted in hindered differentiation, exacerbated myelin sheath impairment, and worsened cognitive deficits. These findings highlight the pivotal roles of GPR17 and ID2 in governing OPC differentiation and axonal remyelination post-SAH. This study positions GPR17 as a potential therapeutic target for SAH intervention. The interplay between GPR17 and ID2 introduces a novel avenue for ameliorating cognitive deficits post-SAH.
Collapse
Affiliation(s)
- Yingwen Wang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Anan Jiang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Yan
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Daochen Wen
- Department of Neurosurgery, Xuanhan County People's Hospital, Dazhou, China
| | - Nina Gu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Zhao Li
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Yue Wu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China.
| | - Zongduo Guo
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China.
| |
Collapse
|
8
|
Wang Y, Zhang M, Zhang T, Zhang S, Ji F, Qin J, Li H, Jiao J. PD-L1/PD-1 checkpoint pathway regulates astrocyte morphogenesis and myelination during brain development. Mol Psychiatry 2025:10.1038/s41380-025-02969-3. [PMID: 40164696 DOI: 10.1038/s41380-025-02969-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Programmed cell death protein 1 (PD-1) and its primary ligand PD-L1 are integral components of a significant immune checkpoint pathway, widely recognized for its central role in cancer immunotherapy. However, emerging evidence highlights their broader involvement in both the central and peripheral nervous systems. In this study, we demonstrate that PD-L1/PD-1 signaling in astrocytes during mouse brain development regulates astrocyte maturation and morphogenesis via the MEK/ERK pathway by targeting the downstream effector cysteine and glycine rich protein 1 (CSRP1). This enhanced astrocyte morphological complexity results in increased end-foot coverage of blood vessels. Additionally, aberrant secretion of CSRP1 by astrocytes interacts with oligodendrocyte precursor cells (OPCs) membrane proteins annexin A1 (ANXA1) and annexin A2 (ANXA2), leading to the exclusion of migrating OPCs from blood vessels. This disruption in OPC migration and differentiation results in abnormal myelination and is associated with cognitive deficits in the mice. Our results provide critical insights into the function of PD-L1/PD-1 signaling in astrocyte-OPC interactions and underscore its relevance to glial cell development and pathogenesis in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yanyan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengtian Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianyu Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shukui Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fen Ji
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jie Qin
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Li
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianwei Jiao
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
9
|
Li J, Fiore F, Monk KR, Agarwal A. Spatiotemporal calcium dynamics orchestrate oligodendrocyte development and myelination. Trends Neurosci 2025:S0166-2236(25)00052-9. [PMID: 40155271 DOI: 10.1016/j.tins.2025.02.010] [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: 08/31/2024] [Revised: 01/13/2025] [Accepted: 02/28/2025] [Indexed: 04/01/2025]
Abstract
Oligodendrocyte lineage cells (OLCs), comprising oligodendrocyte precursor cells (OPCs) and oligodendrocytes, are pivotal in sculpting central nervous system (CNS) architecture and function. OPCs mature into oligodendrocytes, which enwrap axons with myelin sheaths that are critical for enhancing neural transmission. Notably, OLCs actively respond to neuronal activity, modulating neural circuit functions. Understanding neuron-OLC interactions is key to unraveling how OLCs contribute to CNS health and pathology. This review highlights insights from zebrafish and mouse models, revealing how synaptic and extrasynaptic pathways converge to shape spatiotemporal calcium (Ca2+) dynamics within OLCs. We explore how Ca2+ signal integration across spatial and temporal scales acts as a master regulator of OLC fate determination and myelin plasticity.
Collapse
Affiliation(s)
- Jiaxing Li
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Frederic Fiore
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Kelly R Monk
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| | - Amit Agarwal
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
10
|
Lin ML, Lin W. Thinning of originally-existing, mature myelin represents a nondestructive form of myelin loss in the adult CNS. Front Cell Neurosci 2025; 19:1565913. [PMID: 40134707 PMCID: PMC11933062 DOI: 10.3389/fncel.2025.1565913] [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: 01/23/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
The main function of oligodendrocytes is to assemble and maintain myelin that wraps and insulates axons in the central nervous system (CNS). Traditionally, myelin structure, particularly its thickness, was believed to remain remarkably stable in adulthood (including early and middle adulthood, but not late adulthood or aging). However, emerging evidence reveals that the thickness of originally-existing, mature myelin (OEM) can undergo dynamic changes in the adult CNS. This overview highlights recent findings on the alteration of OEM thickness in the adult CNS, explores the underlying mechanisms, and proposes that progressive thinning of OEM represents a novel, nondestructive form of myelin loss in myelin disorders of the CNS.
Collapse
Affiliation(s)
- Min Li Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
11
|
Zou Y, Zhang X, Chen XY, Ma XF, Feng XY, Sun Y, Ma T, Ma QH, Zhao XD, Xu DE. Contactin -Associated protein1 Regulates Autophagy by Modulating the PI3K/AKT/mTOR Signaling Pathway and ATG4B Levels in Vitro and in Vivo. Mol Neurobiol 2025; 62:2764-2780. [PMID: 39164481 PMCID: PMC11790771 DOI: 10.1007/s12035-024-04425-9] [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: 04/19/2024] [Accepted: 08/06/2024] [Indexed: 08/22/2024]
Abstract
Contactin-associated protein1 (Caspr1) plays an important role in the formation and stability of myelinated axons. In Caspr1 mutant mice, autophagy-related structures accumulate in neurons, causing axonal degeneration; however, the mechanism by which Caspr1 regulates autophagy remains unknown. To illustrate the mechanism of Caspr1 in autophagy process, we demonstrated that Caspr1 knockout in primary neurons from mice along with human cell lines, HEK-293 and HeLa, induced autophagy by downregulating the PI3K/AKT/mTOR signaling pathway to promote the conversion of microtubule-associated protein light chain 3 I (LC3-I) to LC3-II. In contrast, Caspr1 overexpression in cells contributed to the upregulation of this signaling pathway. We also demonstrated that Caspr1 knockout led to increased LC3-I protein expression in mice. In addition, Caspr1 could inhibit the expression of autophagy-related 4B cysteine peptidase (ATG4B) protein by directly binding to ATG4B in overexpressed Caspr1 cells. Intriguingly, we found an accumulation of ATG4B in the Golgi apparatuses of cells overexpressing Caspr1; therefore, we speculate that Caspr1 may restrict ATG4 secretion from the Golgi apparatus to the cytoplasm. Collectively, our results indicate that Caspr1 may regulate autophagy by modulating the PI3K/AKT/mTOR signaling pathway and the levels of ATG4 protein, both in vitro and in vivo. Thus, Caspr1 can be a potential therapeutic target in axonal damage and demyelinating diseases.
Collapse
Affiliation(s)
- Yan Zou
- Department of Neurosurgery, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Xiao Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Xin-Yi Chen
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Xiao-Fang Ma
- Hong Shan Hospital, Wuxi, 214000, Jiangsu, China
| | - Xiao-Yan Feng
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Yang Sun
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Tao Ma
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215004, Jiangsu, China
| | - Xu-Dong Zhao
- Department of Neurosurgery, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China.
- Wuxi Neurosurgical Institute, Wuxi, 214122, Jiangsu, China.
| | - De-En Xu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China.
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China.
| |
Collapse
|
12
|
Oatman SR, Reddy JS, Atashgaran A, Wang X, Min Y, Quicksall Z, Vanelderen F, Carrasquillo MM, Liu CC, Yamazaki Y, Nguyen TT, Heckman M, Zhao N, DeTure M, Murray ME, Bu G, Kanekiyo T, Dickson DW, Allen M, Ertekin-Taner N. Integrative Epigenomic Landscape of Alzheimer's Disease Brains Reveals Oligodendrocyte Molecular Perturbations Associated with Tau. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637140. [PMID: 40027794 PMCID: PMC11870448 DOI: 10.1101/2025.02.12.637140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Alzheimer's disease (AD) brains are characterized by neuropathologic and biochemical changes that are highly variable across individuals. Capturing epigenetic factors that associate with this variability can reveal novel biological insights into AD pathophysiology. We conducted an epigenome-wide association study of DNA methylation (DNAm) in 472 AD brains with neuropathologic measures (Braak stage, Thal phase, and cerebral amyloid angiopathy score) and brain biochemical levels of five proteins (APOE, amyloid-β (Aβ)40, Aβ42, tau, and p-tau) core to AD pathogenesis. Using a novel regional methylation (rCpGm) approach, we identified 5,478 significant associations, 99.7% of which were with brain tau biochemical measures. Of the tau-associated rCpGms, 93 had concordant associations in external datasets comprising 1,337 brain samples. Integrative transcriptome-methylome analyses uncovered 535 significant gene expression associations for these 93 rCpGms. Genes with concurrent transcriptome-methylome perturbations were enriched in oligodendrocyte marker genes, including known AD risk genes such as BIN1 , myelination genes MYRF, MBP and MAG previously implicated in AD, as well as novel genes like LDB3 . We further annotated the top oligodendrocyte genes in an additional 6 brain single cell and 2 bulk transcriptome datasets from AD and two other tauopathies, Pick's disease and progressive supranuclear palsy (PSP). Our findings support consistent rCpGm and gene expression associations with these tauopathies and tau-related phenotypes in both bulk brain tissue and oligodendrocyte clusters. In summary, we uncover the integrative epigenomic landscape of AD and demonstrate tau-related oligodendrocyte gene perturbations as a common potential pathomechanism across different tauopathies.
Collapse
|
13
|
Liu H, Su B, Zhang Z, Jia S, Wang J, Zhou F, Liu Y, Cao Q, Tang J, Ou Z, Zhang MM, Chen Y, Dong H, Zhong H. Neonatal sevoflurane exposures inhibits DHHC5-mediated palmitoylation of TfR1 in oligodendrocytes, leading to hypomyelination and neurological impairments. J Adv Res 2025:S2090-1232(25)00107-9. [PMID: 39929269 DOI: 10.1016/j.jare.2025.02.009] [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: 09/28/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025] Open
Abstract
INTRODUCTION Neonatal anesthesia-related neurological impairments are of significant concern, closely linked to oligodendrocyte dysfunction. However, there is a notable temporal discrepancy between the sustained development of oligodendrocytes (myelination) and the short-term vulnerability to anesthesia exposures. OBJECTIVES Given the significant rise in iron demand by oligodendrocytes during neonatal period, our objective was to clarify the potential roles and underlying mechanisms of iron homeostasis, particularly focusing on transferrin receptor 1 (TfR1), in governing the transient susceptibility to anesthesia. METHODS Sevoflurane (3 %, 2 h/day) was administered to wildtype or Pdgfrα-CreERT mice from postnatal day (P)6 to P8. Subsequently, behavioral tests, genetic modulation, co-immunoprecipitation assays, Acyl-resin assisted capture assay and single-cell RNA sequencing were employed on P8 and/or P32. RESULTS Following neonatal exposure to sevoflurane, the observed cognitive impairments and hypomyelination at P32 were attributed to iron accumulation and ferroptosis, particularly within oligodendrocytes of the corpus callosum (CC). This ferroptosis was mediated by enhanced endocytosis of transiently expressed TfR1, rather than its overexpression, due to inhibited palmitoylation. Among the 21 palmitoyltransferases, only Asp-His-His-Cys5 (DHHC5) was down-regulated in oligodendrocytes, reducing palmitoylation of TfR1 at the C98 cysteine site. Furthermore, specific overexpression of DHHC5 in oligodendrocytes significantly restored TfR1 endocytosis, hypomyelination, and ferroptosis, thereby preventing neuronal ferroptosis across multiple brain regions by decreasing iron transport, ultimately mitigating neurological impairments. CONCLUSION We discovered that decreased DHHC5 in oligodendrocytes promotes TfR1 associated ferroptosis, resulting in hypomyelination and initiating neuronal ferroptosis, thereby impairing cognition following neonatal sevoflurane exposures. The transiently expressed TfR1 may mediate the critical period for neonatal anesthesia vulnerability. These findings highlight the pivotal role of TfR1-associated ferroptosis in neonatal anesthesia-associated neurotoxicity and oligodendrocyte-neuron interaction, while providing new perspect to understand temporary neurotoxicity of anesthesia. DHHC5 may represent promising therapeutic target to enhance the safety of neonatal anesthesia and iron-related oligodendrocytes disorders.
Collapse
Affiliation(s)
- Huiqing Liu
- Key Laboratory of Anesthesiology (The Fourth Military Medical University), Ministry of Education, Xi'an 710032, China
| | - Binxiao Su
- Key Laboratory of Anesthesiology (The Fourth Military Medical University), Ministry of Education, Xi'an 710032, China
| | - Zhihao Zhang
- Department of Anesthesiology, Binzhou Medical University Hospital, Huanghe, Binzhou 256600, China
| | - Sansan Jia
- Key Laboratory of Anesthesiology (The Fourth Military Medical University), Ministry of Education, Xi'an 710032, China
| | - Jiajia Wang
- Key Laboratory of Anesthesiology (The Fourth Military Medical University), Ministry of Education, Xi'an 710032, China
| | - Fang Zhou
- Key Laboratory of Anesthesiology (The Fourth Military Medical University), Ministry of Education, Xi'an 710032, China
| | - Yang Liu
- Key Laboratory of Anesthesiology (The Fourth Military Medical University), Ministry of Education, Xi'an 710032, China
| | - Qiuxia Cao
- Key Laboratory of Anesthesiology (The Fourth Military Medical University), Ministry of Education, Xi'an 710032, China
| | - Jun Tang
- Key Laboratory of Anesthesiology (The Fourth Military Medical University), Ministry of Education, Xi'an 710032, China
| | - Zhimin Ou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361000, China
| | - Ming-Ming Zhang
- Department of Human Anatomy, Histology and Embryology & K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an 710032, China
| | - Ying Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361000, China.
| | - Hailong Dong
- Key Laboratory of Anesthesiology (The Fourth Military Medical University), Ministry of Education, Xi'an 710032, China.
| | - Haixing Zhong
- Key Laboratory of Anesthesiology (The Fourth Military Medical University), Ministry of Education, Xi'an 710032, China.
| |
Collapse
|
14
|
Yan X, Jiang C, Han Z, Huang D, Cheng L, Han W, Jiang L. Effects of ascorbic acid on myelination in offspring of advanced maternal age. Neuroscience 2025; 566:218-226. [PMID: 39571959 DOI: 10.1016/j.neuroscience.2024.11.019] [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: 07/30/2024] [Revised: 10/17/2024] [Accepted: 11/07/2024] [Indexed: 01/22/2025]
Abstract
Myelination is the process by which oligodendrocytes ensheathe axons to form myelin sheaths. Myelination is a crucial aspect of brain development and is closely associated with central nervous system abnormalities. However, previous studies have found that advanced maternal age might affect the myelination of offspring, potentially through the pathway of disrupting DNA methylation levels in the offspring's hippocampus. Current research has demonstrated that ascorbic acid can promote hydroxymethylation to reduce methylation levels in vivo. This study aims to verify the relationship between ascorbic acid and myelination, as well as the specific mechanism involved. Initially, oligodendrocyte differentiation was observed using immunofluorescence and Western blot. Myelination was assessed through Luxol Fast Blue staining, Glycine silver staining, immunofluorescence, and transmission electron microscopy. The demethylation level of oligodendrocyte progenitor cells was detected by immunofluorescence co-expression of OLIG2 and DNA hydroxylase ten-eleven translocation 1 (TET1), TET2, and TET3. Our study found that advanced maternal age could impair myelination in the hippocampus and corpus callosum of offspring. Ascorbic acid intervention may induce TET1 and TET2-mediated hydroxymethylation to ameliorate myelination disorders, promote myelination and maturation, and reverse the effects of advanced maternal age on offspring.
Collapse
Affiliation(s)
- Xinru Yan
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, China
| | - Chunxue Jiang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, China
| | - Ziyao Han
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, China
| | - Dishu Huang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, China
| | - Li Cheng
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, China
| | - Wei Han
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, China.
| | - Li Jiang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, China.
| |
Collapse
|
15
|
Zhang H, Yang S, Lu YL, Zhou LQ, Dong MH, Chu YH, Pang XW, Chen L, Xu LL, Zhang LY, Zhu LF, Xu T, Wang W, Shang K, Tian DS, Qin C. Microglial Nrf2-mediated lipid and iron metabolism reprogramming promotes remyelination during white matter ischemia. Redox Biol 2025; 79:103473. [PMID: 39718294 PMCID: PMC11728325 DOI: 10.1016/j.redox.2024.103473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/06/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Oxidative stress and microglial activation are critical pathomechanisms in ischemic white matter injury. Microglia, as resident immune cells in the brain, are the main cells undergoing oxidative stress response. However, the role and molecular mechanism of oxidative stress in microglia have not been clearly elucidated during white matter ischemia. METHODS Extensive histological analysis of the corpus callosum was performed in BCAS mice at different time points to assess white matter injury, oxidative stress and microglial activation. Flow cytometric sorting and transcriptomic sequencing were combined to explore the underlying mechanisms regulating microglial oxidative stress and functional phenotypes. The expression of critical molecule in microglia was regulated using Cx3cr1CreER mice and clinical-stage drugs to assess its effect on white matter injury and cognitive function. RESULTS Our study identified nuclear factor erythroid-2 related factor 2 (Nrf2) as a key transcription factor regulating oxidative stress and functional phenotype in microglia. Interestingly, we found that the sustained decrease in transiently upregulated expression of Nrf2 following chronic cerebral hypoperfusion resulted in abnormal microglial activation and white matter injury. In addition, high loads of myelin debris promoted lipid peroxidation and ferroptosis in microglia with diminished antioxidant function. Microglia with pharmacologically or genetically stimulated Nrf2 expression exhibited enhanced resistance to ferroptosis and pro-regenerative properties to myelination due to lipid and iron metabolism reprogramming. CONCLUSION Weakened Nrf2-mediated antioxidant responses in microglia induced metabolic disturbances and ferroptosis during chronic cerebral hypoperfusion. Targeted enhancement of Nrf2 expression in microglia may be a potential therapeutic strategy for ischemic white matter injury.
Collapse
Affiliation(s)
- Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Yi-Lin Lu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Lian Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Lu-Lu Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Lu-Yang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Li-Fang Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Ting Xu
- Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou, 510080, PR China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Ke Shang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| |
Collapse
|
16
|
Talidou A, Lefebvre J. Spatial Heterogeneity in Myelin Sheathing Impacts Signaling Reliability and Susceptibility to Injury. eNeuro 2025; 12:ENEURO.0402-24.2025. [PMID: 39870523 PMCID: PMC11839277 DOI: 10.1523/eneuro.0402-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/29/2025] Open
Abstract
Axons in the mammalian brain show significant diversity in myelination motifs, displaying spatial heterogeneity in sheathing along individual axons and across brain regions. However, its impact on neural signaling and susceptibility to injury remains poorly understood. To address this, we leveraged cable theory and developed model axons replicating the myelin sheath distributions observed experimentally in different regions of the mouse central nervous system. We examined how the spatial arrangement of myelin affects propagation and predisposition to conduction failure in axons with cortical versus callosal myelination motifs. Our results indicate that regional differences in myelination significantly influence conduction timing and signaling reliability. Sensitivity of action potential propagation to the specific positioning, lengths, and ordering of myelinated and exposed segments reveals non-linear and path-dependent conduction. Furthermore, myelination motifs impact signaling vulnerability to demyelination, with callosal motifs being particularly sensitive to myelin changes. These findings highlight the crucial role of myelinating glia in brain function and disease.
Collapse
Affiliation(s)
- Afroditi Talidou
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta T2N 4N1, Canada
- Department of Biology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Jérémie Lefebvre
- Department of Biology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
- Department of Mathematics, University of Toronto, Toronto, Ontario M5S 2E4, Canada
| |
Collapse
|
17
|
Sydnor VJ, Petrie D, McKeon SD, Famalette A, Foran W, Calabro FJ, Luna B. Heterochronous laminar maturation in the human prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635751. [PMID: 39975178 PMCID: PMC11838308 DOI: 10.1101/2025.01.30.635751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The human prefrontal cortex (PFC) exhibits markedly protracted developmental plasticity, yet whether reductions in plasticity occur synchronously across prefrontal cortical layers is unclear. Animal studies have shown that intracortical myelin consolidates neural circuits to close periods of plasticity. Here, we use quantitative myelin imaging collected from youth (ages 10-32 years) at ultra-high field (7T) to investigate whether deep and superficial PFC layers exhibit different timeframes of plasticity. We find that myelin matures along a deep-to-superficial axis in the PFC; this axis of maturational timing is expressed to a different extent in cytoarchitecturally distinct regions along the frontal cortical hierarchy. By integrating myelin mapping with electroencephalogram and cognitive phenotyping, we provide evidence that deep and superficial prefrontal myelin dissociably impact timescales of neural activity, task learning rates, and cognitive processing speed. Heterochronous maturation across deep and superficial layers is an underrecognized mechanism through which association cortex balances cognitively-relevant increases in circuit stability and efficiency with extended neuroplasticity.
Collapse
Affiliation(s)
- Valerie J. Sydnor
- Department of Psychiatry, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
- The Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, USA
| | - Daniel Petrie
- Department of Psychiatry, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
- The Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, USA
| | - Shane D. McKeon
- Department of Psychiatry, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
- The Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alyssa Famalette
- Department of Psychiatry, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Will Foran
- Department of Psychiatry, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Finnegan J. Calabro
- Department of Psychiatry, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beatriz Luna
- Department of Psychiatry, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
- The Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
18
|
Chen H, Wang W, Cui W, Tu C, Han Y, Zhang C, Yang L, Huang X, Zhang Q, Lu L. Seipin Deficiency Impairs Motor Coordination in Mice by Compromising Spinal Cord Myelination. Neuromolecular Med 2025; 27:12. [PMID: 39869141 DOI: 10.1007/s12017-025-08834-4] [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: 12/17/2024] [Accepted: 01/17/2025] [Indexed: 01/28/2025]
Abstract
The integrity of the myelin sheath of the spinal cord (SC) is essential for motor coordination. Seipin is an endoplasmic reticulum transmembrane protein highly expressed in adipose tissue and motor neurons in the SC. It was reported Seipin deficiency induced lipid dysregulation and neurobehavioral deficits, but the underlying mechanism, especially in SC, remains to be elucidated. In present study, we found that Seipin and myelin basic protein (MBP) increased synchronously in SC of developmental stage of mice. Demyelination impaired motor coordination as well as MBP and Seipin expression, which were alleviated by remyelination. Moreover, Seipin deficiency impaired motor coordination of mice, accompanied by hypomyelination in spinal cord. Mechanistically, we further demonstrated that myelin content as labeled by Fluormyelin, myelin basic protein (MBP) was down-regulated by Seipin deficiency. Seipin deficiency led to reduction of myelin-forming oligodendrocytes (OLs) density in spinal cord. Notably, administration of rosiglitazone (RG), a classic PPARγ activator, successfully restored the phenotypes manifested by Seipin deficiency including reduced OLs density, hypomyelination, as well as motor dyscoordination. In summary, present study revealed that Seipin deficiency disrupted motor coordination by compromising myelination in SC, and RG treatment could rescue the phenotypes. This study throws light on the mechanism underlying Seipin deficiency associated disorders and paves ways for developing therapeutics toward those diseases.
Collapse
Affiliation(s)
- Hong Chen
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Department of Spine Surgery, Yuncheng Central Hospital of Shanxi Province, Shanxi Medical University, No. 3690, Hedong East Road, Yanhu District, Yuncheng, 044020, Shanxi, China
| | - Wenru Wang
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Wenli Cui
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chuanyun Tu
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yuanyuan Han
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chengwu Zhang
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Liu Yang
- First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xintao Huang
- First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qin Zhang
- Department of Spine Surgery, Yuncheng Central Hospital of Shanxi Province, Shanxi Medical University, No. 3690, Hedong East Road, Yanhu District, Yuncheng, 044020, Shanxi, China.
| | - Li Lu
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China.
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
19
|
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
|
20
|
Sharma G, Wade Q, Graziane NM. Evaluating remyelination compounds for new applications in opioid use disorder management. J Addict Dis 2025:1-17. [PMID: 39834150 DOI: 10.1080/10550887.2025.2452691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Opioid use disorder (OUD) is associated with a reduction in brain white matter, affecting critical areas involved in decision-making, impulse control, and reward processing. The FDA has approved several drugs and natural compounds that enhance myelination, targeting oligodendrocyte progenitor cells (OPCs), directly enhancing oligodendrocyte (OL) function, or acting as cofactors for myelin production. This retrospective case study aimed to assess whether current clinical evidence supports the use of myelin-enhancing agents to promote remission in OUD. We evaluated a range of compounds with demonstrated effects on myelination, including muscarinic antagonists, cholesterol and lipid homeostatic agents, anti-aging drugs, immunomodulatory agents, anti-inflammatory medications, and others (25 medications in total), as well as 17 vitamins and supplements. Buprenorphine and methadone were used as positive controls. Sequential analyses were performed to identify individual drugs driving significant changes in remission rates (p ≤ 0.01; N ≥ 3,000) and their effects across age, sex, and Body Mass Index (BMI) categories. Three key findings emerged: (1) melatonin improved remission rates in males but showed no effect in females; (2) ibuprofen significantly increased remission rates, particularly in individuals aged 20-39 and 40-59 years; and (3) thiamin was associated with decreased remission rates in males and individuals with a BMI ranging from normal weight to obese. Additionally, buprenorphine and methadone were confirmed as effective in promoting remission. These findings highlight the importance of personalized medicine in treating OUD and suggest that further research is needed to explore individualized treatment strategies based on sex, age, and BMI.
Collapse
Affiliation(s)
- Gouri Sharma
- Department of Anesthesiology and Perioperative Medicine, Hershey, PA, USA
| | - Quinn Wade
- Department of Anesthesiology and Perioperative Medicine, Hershey, PA, USA
| | - Nicholas M Graziane
- Departments of Anesthesiology and Perioperative Medicine and Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
21
|
Quattrini G, Carcione A, Lanfredi M, Nicolò G, Pedrini L, Corbo D, Magni LR, Geviti A, Ferrari C, Gasparotti R, Semerari A, Pievani M, Rossi R. Effect of metacognitive interpersonal therapy on brain structural connectivity in borderline personality disorder: Results from the CLIMAMITHE randomized clinical trial. J Affect Disord 2025; 369:1145-1152. [PMID: 39454963 DOI: 10.1016/j.jad.2024.10.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/16/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Recently, we showed that Metacognitive Interpersonal Therapy (MIT) is effective in improving clinical symptoms in borderline personality disorder (BPD). Here, we investigated whether the effect of MIT on clinical features is associated to microstructural changes in brain circuits supporting core BPD symptoms. METHODS Forty-seven BPD were randomized to MIT or structured clinical management, and underwent a clinical assessment and diffusion-weighted imaging before and after the intervention. Fractional anisotropy (FA), mean, radial, and axial diffusivities maps were computed using FSL toolbox. Microstructural changes were assessed (i) voxel-wise, with tract based spatial statistics (TBSS) and (ii) ROI-wise, in the triple network system (default mode, salience, and executive control networks). The effect of MIT on brain microstructure was assessed with paired tests using FSL PALM (voxel-wise), Linear Mixed-Effect Models or Generalized Linear Mixed Models (ROI-wise). Associations between microstructural and clinical changes were explored with linear regression (voxel-wise) and correlations (ROI-wise). RESULTS The voxel-wise analysis showed that MIT was associated with increased FA in the bilateral thalamic radiation and left associative tracts (p < .050, family-wise error rate corrected). At network system level, MIT increased FA and both interventions reduced AD in the executive control network (p = .05, uncorrected). LIMITATIONS The DTI metrics can't clarify the nature of axonal changes. CONCLUSIONS Our results indicate that MIT modulates brain structural connectivity in circuits related to associative and executive control functions. These microstructural changes may denote activity-dependent plasticity, possibly representing a neurobiological mechanism underlying MIT effects. TRIAL REGISTRATION ClinicalTrials.govNCT02370316 (https://clinicaltrials.gov/study/NCT02370316).
Collapse
Affiliation(s)
- Giulia Quattrini
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Mariangela Lanfredi
- Unit of Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Laura Pedrini
- Unit of Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Daniele Corbo
- Neuroradiology Unit, Department of Medical and Surgical Specialities, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Laura R Magni
- Clinical Psychology Unit, Mental Health and Addiction Department, ASST Brianza, Vimercate, MB, Italy
| | - Andrea Geviti
- Unit of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Clarissa Ferrari
- Unit of Research and Clinical Trials, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Roberto Gasparotti
- Neuroradiology Unit, Department of Medical and Surgical Specialities, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | | | - Michela Pievani
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - Roberta Rossi
- Unit of Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
22
|
Xin Y, Zhou S, Chu T, Zhou Y, Xu A. Protective Role of Electroacupuncture Against Cognitive Impairment in Neurological Diseases. Curr Neuropharmacol 2025; 23:145-171. [PMID: 38379403 PMCID: PMC11793074 DOI: 10.2174/1570159x22999240209102116] [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: 08/12/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 02/22/2024] Open
Abstract
Many neurological diseases can lead to cognitive impairment in patients, which includes dementia and mild cognitive impairment and thus create a heavy burden both to their families and public health. Due to the limited effectiveness of medications in treating cognitive impairment, it is imperative to develop alternative treatments. Electroacupuncture (EA), a required method for Traditional Chinese Medicine, has the potential treatment of cognitive impairment. However, the molecular mechanisms involved have not been fully elucidated. Considering the current research status, preclinical literature published within the ten years until October 2022 was systematically searched through PubMed, Web of Science, MEDLINE, Ovid, and Embase. By reading the titles and abstracts, a total of 56 studies were initially included. It is concluded that EA can effectively ameliorate cognitive impairment in preclinical research of neurological diseases and induce potentially beneficial changes in molecular pathways, including Alzheimer's disease, vascular cognitive impairment, chronic pain, and Parkinson's disease. Moreover, EA exerts beneficial effects through the same or diverse mechanisms for different disease types, including but not limited to neuroinflammation, neuronal apoptosis, neurogenesis, synaptic plasticity, and autophagy. However, these findings raise further questions that need to be elucidated. Overall, EA therapy for cognitive impairment is an area with great promise, even though more research regarding its detailed mechanisms is warranted.
Collapse
Affiliation(s)
- Yueyang Xin
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siqi Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tiantian Chu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaqun Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aijun Xu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
McCaig CD. Electrical Forces Improve Memory in Old Age. Rev Physiol Biochem Pharmacol 2025; 187:453-520. [PMID: 39838022 DOI: 10.1007/978-3-031-68827-0_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
This penultimate chapter is based on a single paper published in Nature in 2022. I have used it specifically as an exemplar, in this case to show that memory improvement in old age may be regulated by a multiplicity of electrical forces. However, I include it because I believe that one could pick almost any other substantial single paper and show that a completely disparate set of biological mechanisms similarly depend crucially on multiple electrical forces.
Collapse
Affiliation(s)
- Colin D McCaig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
24
|
Zhang A. Revisiting the role of ErbBs in oligodendrocyte development. Proc Natl Acad Sci U S A 2024; 121:e2422181121. [PMID: 39680783 DOI: 10.1073/pnas.2422181121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Affiliation(s)
- Albert Zhang
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158
| |
Collapse
|
25
|
Wu J, Kislinger G, Duschek J, Durmaz AD, Wefers B, Feng R, Nalbach K, Wurst W, Behrends C, Schifferer M, Simons M. Nonvesicular lipid transfer drives myelin growth in the central nervous system. Nat Commun 2024; 15:9756. [PMID: 39528474 PMCID: PMC11554831 DOI: 10.1038/s41467-024-53511-y] [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: 02/29/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Oligodendrocytes extend numerous cellular processes that wrap multiple times around axons to generate lipid-rich myelin sheaths. Myelin biogenesis requires an enormously productive biosynthetic machinery for generating and delivering these large amounts of newly synthesized lipids. Yet, a complete understanding of this process remains elusive. Utilizing volume electron microscopy, we demonstrate that the oligodendroglial endoplasmic reticulum (ER) is enriched in developing myelin, extending into and making contact with the innermost myelin layer where growth occurs. We explore the possibility of transfer of lipids from the ER to myelin, and find that the glycolipid transfer protein (GLTP), implicated in nonvesicular lipid transport, is highly enriched in the growing myelin sheath. Mice with a specific knockout of Gltp in oligodendrocytes exhibit ER pathology, hypomyelination and a decrease in myelin glycolipid content. In summary, our results demonstrate a role for nonvesicular lipid transport in CNS myelin growth, revealing a cellular pathway in developmental myelination.
Collapse
Affiliation(s)
- Jianping Wu
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Georg Kislinger
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Jerome Duschek
- Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ayşe Damla Durmaz
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Benedikt Wefers
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Ruoqing Feng
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Karsten Nalbach
- Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Developmental Genetics, Helmholtz Center Munich, Neuherberg, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Christian Behrends
- Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
26
|
Huang G, Li Z, Liu X, Guan M, Zhou S, Zhong X, Zheng T, Xin D, Gu X, Mu D, Guo Y, Zhang L, Zhang L, Lu QR, He X. DOR activation in mature oligodendrocytes regulates α-ketoglutarate metabolism leading to enhanced remyelination in aged mice. Nat Neurosci 2024; 27:2073-2085. [PMID: 39266660 DOI: 10.1038/s41593-024-01754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/07/2024] [Indexed: 09/14/2024]
Abstract
The decreased ability of mature oligodendrocytes to produce myelin negatively affects remyelination in demyelinating diseases and aging, but the underlying mechanisms are incompletely understood. In the present study, we identify a mature oligodendrocyte-enriched transcriptional coregulator diabetes- and obesity-related gene (DOR)/tumor protein p53-inducible nuclear protein 2 (TP53INP2), downregulated in demyelinated lesions of donors with multiple sclerosis and in aged oligodendrocyte-lineage cells. Dor ablation in mice of both sexes results in defective myelinogenesis and remyelination. Genomic occupancy in oligodendrocytes and transcriptome profiling of the optic nerves of wild-type and Dor conditional knockout mice reveal that DOR and SOX10 co-occupy enhancers of critical myelinogenesis-associated genes including Prr18, encoding an oligodendrocyte-enriched, proline-rich factor. We show that DOR targets regulatory elements of genes responsible for α-ketoglutarate biosynthesis in mature oligodendrocytes and is essential for α-ketoglutarate production and lipid biosynthesis. Supplementation with α-ketoglutarate restores oligodendrocyte-maturation defects in Dor-deficient adult mice and improves remyelination after lysolecithin-induced demyelination and cognitive function in 17-month-old wild-type mice. Our data suggest that activation of α-ketoglutarate metabolism in mature oligodendrocytes can promote myelin production during demyelination and aging.
Collapse
Affiliation(s)
- Guojiao Huang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhidan Li
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xuezhao Liu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Menglong Guan
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaowen Zhong
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tao Zheng
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dazhuan Xin
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Dezhi Mu
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yingkun Guo
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lin Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liguo Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xuelian He
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
27
|
Grouza V, Bagheri H, Liu H, Tuznik M, Wu Z, Robinson N, Siminovitch KA, Peterson AC, Rudko DA. Ultra-high-resolution mapping of myelin and g-ratio in a panel of Mbp enhancer-edited mouse strains using microstructural MRI. Neuroimage 2024; 300:120850. [PMID: 39260782 DOI: 10.1016/j.neuroimage.2024.120850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/27/2024] [Accepted: 09/09/2024] [Indexed: 09/13/2024] Open
Abstract
Non-invasive myelin water fraction (MWF) and g-ratio mapping using microstructural MRI have the potential to offer critical insights into brain microstructure and our understanding of neuroplasticity and neuroinflammation. By leveraging a unique panel of variably hypomyelinating mouse strains, we validated a high-resolution, model-free image reconstruction method for whole-brain MWF mapping. Further, by employing a bipolar gradient echo MRI sequence, we achieved high spatial resolution and robust mapping of MWF and g-ratio across the whole mouse brain. Our regional white matter-tract specific analyses demonstrated a graded decrease in MWF in white matter tracts which correlated strongly with myelin basic protein gene (Mbp) mRNA levels. Using these measures, we derived the first sensitive calibrations between MWF and Mbp mRNA in the mouse. Minimal changes in axonal density supported our hypothesis that observed MWF alterations stem from hypomyelination. Overall, our work strongly emphasizes the potential of non-invasive, MRI-derived MWF and g-ratio modeling for both preclinical model validation and ultimately translation to humans.
Collapse
Affiliation(s)
- Vladimir Grouza
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Hooman Bagheri
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Hanwen Liu
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Marius Tuznik
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Zhe Wu
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Nicole Robinson
- Histology Innovation Platform, Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Katherine A Siminovitch
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Mount Sinai Hospital, Lunenfeld-Tanenbaum and Toronto General Hospital Research Institutes, Toronto, Ontario, Canada
| | - Alan C Peterson
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Department of Human Genetics, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - David A Rudko
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
28
|
Faust TE, Devlin BA, Farhy-Tselnicker I, Ferro A, Postolache M, Xin W. Glial Control of Cortical Neuronal Circuit Maturation and Plasticity. J Neurosci 2024; 44:e1208242024. [PMID: 39358028 PMCID: PMC11450532 DOI: 10.1523/jneurosci.1208-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 10/04/2024] Open
Abstract
The brain is a highly adaptable organ that is molded by experience throughout life. Although the field of neuroscience has historically focused on intrinsic neuronal mechanisms of plasticity, there is growing evidence that multiple glial populations regulate the timing and extent of neuronal plasticity, particularly over the course of development. This review highlights recent discoveries on the role of glial cells in the establishment of cortical circuits and the regulation of experience-dependent neuronal plasticity during critical periods of neurodevelopment. These studies provide strong evidence that neuronal circuit maturation and plasticity are non-cell autonomous processes that require both glial-neuronal and glial-glial cross talk to proceed. We conclude by discussing open questions that will continue to guide research in this nascent field.
Collapse
Affiliation(s)
- Travis E Faust
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605
| | - Benjamin A Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina 27708
| | | | - Austin Ferro
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724
| | - Maggie Postolache
- Brain Immunology & Glia Center, Washington University School of Medicine, St. Louis, Missouri 63110
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Wendy Xin
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California 94158
| |
Collapse
|
29
|
Kaller MS, Lazari A, Feng Y, van der Toorn A, Rühling S, Thomas CW, Shimizu T, Bannerman D, Vyazovskiy V, Richardson WD, Sampaio-Baptista C, Johansen-Berg H. Ablation of oligodendrogenesis in adult mice alters brain microstructure and activity independently of behavioral deficits. Glia 2024; 72:1728-1745. [PMID: 38982743 DOI: 10.1002/glia.24576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 07/11/2024]
Abstract
Oligodendrocytes continue to differentiate from their precursor cells even in adulthood, a process that can be modulated by neuronal activity and experience. Previous work has indicated that conditional ablation of oligodendrogenesis in adult mice leads to learning and memory deficits in a range of behavioral tasks. The current study replicated and re-evaluated evidence for a role of oligodendrogenesis in motor learning, using a complex running wheel task. Further, we found that ablating oligodendrogenesis alters brain microstructure (ex vivo MRI) and brain activity (in vivo EEG) independent of experience with the task. This suggests a role for adult oligodendrocyte formation in the maintenance of brain function and indicates that task-independent changes due to oligodendrogenesis ablation need to be considered when interpreting learning and memory deficits in this model.
Collapse
Affiliation(s)
- Malte S Kaller
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Alberto Lazari
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Yingshi Feng
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Annette van der Toorn
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht & Utrecht University, Utrecht, The Netherlands
| | - Sebastian Rühling
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christopher W Thomas
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Takahiro Shimizu
- The Wolfson Institute for Biomedical Research, University College London, London, UK
| | - David Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Vladyslav Vyazovskiy
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, UK
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - William D Richardson
- The Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Cassandra Sampaio-Baptista
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, UK
| | - Heidi Johansen-Berg
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
30
|
Chen S, Ding S, Pang Y, Jin Y, Sun P, Li Y, Cao M, Wang Y, Wang Z, Wang T, Zou Y, Zhang Y, Xiao M. Dysregulated miR-124 mediates impaired social memory behavior caused by paternal early social isolation. Transl Psychiatry 2024; 14:392. [PMID: 39341799 PMCID: PMC11438908 DOI: 10.1038/s41398-024-03109-1] [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: 01/02/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
Early social isolation (SI) leads to various abnormalities in emotion and behavior during adulthood. However, the negative impact of SI on offspring remains unclear. This study has discovered that paternal early SI causes social memory deficits and anxiety-like behavior in F1 young adult mice, with alterations of myelin and synapses in the medial prefrontal cortex (mPFC). The 2-week SI in the F1 progeny exacerbates social memory impairment and hypomyelination in the mPFC. Furthermore, the down-regulation of miR-124, a key inhibitor of myelinogenesis, or over-expression of its target gene Nr4a1 in the mPFC of the F1 mice improves social interaction ability and enhances oligodendrocyte maturation and myelin formation. Mechanistically, elevated levels of miR-124 in the sperm of paternal SI mice are transmitted epigenetically to offspring, altering the expression levels of miR-124/Nr4a1/glucocorticoid receptors in mPFC oligodendrocytes. This, in turn, impedes the establishment of myelinogenesis-dependent social behavior. This study unveils a novel mechanism through which miR-124 mediates the intergenerational effects of early isolation stress, ultimately impairing the establishment of social behavior and neurodevelopment.
Collapse
Affiliation(s)
- Sijia Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Shixin Ding
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yingting Pang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Yuxi Jin
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Peng Sun
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yue Li
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Min Cao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yimiao Wang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ze Wang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Tianqi Wang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Ying Zou
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Yanli Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China.
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213003, China.
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou, 213000, China.
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
- Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213003, China
| |
Collapse
|
31
|
Yang L, Xing W, Shi Y, Hu M, Li B, Hu Y, Zhang G. Stress-induced NLRP3 inflammasome activation and myelin alterations in the hippocampus of PTSD rats. Neuroscience 2024; 555:156-166. [PMID: 39043314 DOI: 10.1016/j.neuroscience.2024.07.028] [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: 01/31/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
Inflammatory and myelin changes may contribute to the pathophysiology of post-traumatic stress disorder (PTSD). The NOD-like receptor (NLR) family, pyrin domain-containing protein 3 (NLRP3), a brain inflammasome, is activated in the hippocampus of mice with PTSD. In other psychiatric disorders, NLRP3 expression has been associated with axonal myelination and demyelination. However, the association between NLRP3 and myelin in rats with PTSD remains unclear. Therefore, this study aims to investigate the relationship between the NLRP3 inflammasome and myelin in the hippocampus of rats with PTSD. A rat model of post-traumatic stress disorder was established using the single-prolonged stress (SPS) approach. Hippocampal tissues were collected for the detection of NLRP3 inflammasome-associated proteins and myelin basic protein at 3, 7, and 14 days after SPS. To further explore the relationship between NLRP3 and myelin, the NLRP3-specific inhibitor MCC950 was administered intraperitoneally to rats starting 72 h before SPS, and then alterations in NLRP3 inflammasome-associated proteins and myelin were observed in the PTSD and control groups. We found that NLRP3 and downstream related proteins were activated in the hippocampus of rats 3 days after SPS, and the myelin content in the hippocampus increased after SPS stress. MCC950 reduced the expression of NLRP3-related pathway proteins, improved anxiety behaviour and spatial learning memory impairment, and inhibited the increase in myelin content in the hippocampal region of rats after SPS. In conclusion the study indicates that NLRP3 has a significant role in the hippocampal region of rats with PTSD. Inhibition of the NLRP3 inflammasome could be a potential target for treating PTSD.
Collapse
Affiliation(s)
- Luodong Yang
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Wenlong Xing
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Yan Shi
- Shihezi University, Shihezi, China
| | - Min Hu
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Bin Li
- Shihezi University, Shihezi, China
| | - Yuanyuan Hu
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Guiqing Zhang
- First Affiliated Hospital of Shihezi University, Shihezi, China.
| |
Collapse
|
32
|
Xin W, Kaneko M, Roth RH, Zhang A, Nocera S, Ding JB, Stryker MP, Chan JR. Oligodendrocytes and myelin limit neuronal plasticity in visual cortex. Nature 2024; 633:856-863. [PMID: 39169185 PMCID: PMC11424474 DOI: 10.1038/s41586-024-07853-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
Developmental myelination is a protracted process in the mammalian brain1. One theory for why oligodendrocytes mature so slowly posits that myelination may stabilize neuronal circuits and temper neuronal plasticity as animals age2-4. We tested this theory in the visual cortex, which has a well-defined critical period for experience-dependent neuronal plasticity5. During adolescence, visual experience modulated the rate of oligodendrocyte maturation in visual cortex. To determine whether oligodendrocyte maturation in turn regulates neuronal plasticity, we genetically blocked oligodendrocyte differentiation and myelination in adolescent mice. In adult mice lacking adolescent oligodendrogenesis, a brief period of monocular deprivation led to a significant decrease in visual cortex responses to the deprived eye, reminiscent of the plasticity normally restricted to adolescence. This enhanced functional plasticity was accompanied by a greater turnover of dendritic spines and coordinated reductions in spine size following deprivation. Furthermore, inhibitory synaptic transmission, which gates experience-dependent plasticity at the circuit level, was diminished in the absence of adolescent oligodendrogenesis. These results establish a critical role for oligodendrocytes in shaping the maturation and stabilization of cortical circuits and support the concept of developmental myelination acting as a functional brake on neuronal plasticity.
Collapse
Affiliation(s)
- Wendy Xin
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| | - Megumi Kaneko
- Department of Physiology, Kavli Institute for Fundamental Neuroscience and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Richard H Roth
- Departments of Neurosurgery and Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Albert Zhang
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Sonia Nocera
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Jun B Ding
- Departments of Neurosurgery and Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michael P Stryker
- Department of Physiology, Kavli Institute for Fundamental Neuroscience and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Jonah R Chan
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
33
|
Li J, Capuano AW, Agarwal P, Arvanitakis Z, Wang Y, De Jager PL, Schneider JA, Tasaki S, de Paiva Lopes K, Hu FB, Bennett DA, Liang L, Grodstein F. The MIND diet, brain transcriptomic alterations, and dementia. Alzheimers Dement 2024; 20:5996-6007. [PMID: 39129336 PMCID: PMC11497672 DOI: 10.1002/alz.14062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 05/01/2024] [Accepted: 05/20/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Dietary patterns are associated with dementia risk, but the underlying molecular mechanisms are largely unknown. METHODS We used RNA sequencing data from post mortem prefrontal cortex tissue and annual cognitive evaluations from 1204 participants in the Religious Orders Study and Memory and Aging Project. We identified a transcriptomic profile correlated with the MIND diet (Mediterranean-Dietary Approaches to Stop Hypertension Intervention for Neurodegenerative Delay) among 482 individuals who completed ante mortem food frequency questionnaires; and examined its associations with cognitive health in the remaining 722 participants. RESULTS We identified a transcriptomic profile, consisting of 50 genes, correlated with the MIND diet score (p = 0.001). Each standard deviation increase in the transcriptomic profile score was associated with a slower annual rate of decline in global cognition (β = 0.011, p = 0.003) and lower odds of dementia (odds ratio = 0.76, p = 0.0002). Expressions of several genes (including TCIM and IGSF5) appeared to mediate the association between MIND diet and dementia. DISCUSSION A brain transcriptomic profile for healthy diets revealed novel genes potentially associated with cognitive health. HIGHLIGHTS Why healthy dietary patterns are associated with lower dementia risk are unknown. We integrated dietary, brain transcriptomic, and cognitive data in older adults. Mediterranean-Dietary Approaches to Stop Hypertension Intervention for Neurodegenerative Delay (MIND) diet intake is correlated with a specific brain transcriptomic profile. This brain transcriptomic profile score is associated with better cognitive health. More data are needed to elucidate the causality and functionality of identified genes.
Collapse
Affiliation(s)
- Jun Li
- Division of Preventive MedicineDepartment of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Department of NutritionHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Ana W. Capuano
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Puja Agarwal
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Internal MedicineRush University Medical CenterChicagoIllinoisUSA
| | - Zoe Arvanitakis
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Yanling Wang
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Philip L. De Jager
- Center for Translational & Computational NeuroimmunologyDepartment of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Julie A. Schneider
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
| | - Shinya Tasaki
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Katia de Paiva Lopes
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Frank B. Hu
- Department of NutritionHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Channing Division of Network MedicineDepartment of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - David A Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Liming Liang
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Francine Grodstein
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Internal MedicineRush University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
34
|
Yamazaki R, Ohno N. Myosin superfamily members during myelin formation and regeneration. J Neurochem 2024; 168:2264-2274. [PMID: 39136255 DOI: 10.1111/jnc.16202] [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: 04/10/2024] [Revised: 06/10/2024] [Accepted: 07/31/2024] [Indexed: 10/04/2024]
Abstract
Myelin is an insulator that forms around axons that enhance the conduction velocity of nerve fibers. Oligodendrocytes dramatically change cell morphology to produce myelin throughout the central nervous system (CNS). Cytoskeletal alterations are critical for the morphogenesis of oligodendrocytes, and actin is involved in cell differentiation and myelin wrapping via polymerization and depolymerization, respectively. Various protein members of the myosin superfamily are known to be major binding partners of actin filaments and have been intensively researched because of their involvement in various cellular functions, including differentiation, cell movement, membrane trafficking, organelle transport, signal transduction, and morphogenesis. Some members of the myosin superfamily have been found to play important roles in the differentiation of oligodendrocytes and in CNS myelination. Interestingly, each member of the myosin superfamily expressed in oligodendrocyte lineage cells also shows specific spatial and temporal expression patterns and different distributions. In this review, we summarize previous findings related to the myosin superfamily and discuss how these molecules contribute to myelin formation and regeneration by oligodendrocytes.
Collapse
Affiliation(s)
- Reiji Yamazaki
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
35
|
Meisler SL, Kubota E, Grotheer M, Gabrieli JDE, Grill-Spector K. A practical guide for combining functional regions of interest and white matter bundles. Front Neurosci 2024; 18:1385847. [PMID: 39221005 PMCID: PMC11363198 DOI: 10.3389/fnins.2024.1385847] [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: 02/13/2024] [Accepted: 07/11/2024] [Indexed: 09/04/2024] Open
Abstract
Diffusion-weighted imaging (DWI) is the primary method to investigate macro- and microstructure of neural white matter in vivo. DWI can be used to identify and characterize individual-specific white matter bundles, enabling precise analyses on hypothesis-driven connections in the brain and bridging the relationships between brain structure, function, and behavior. However, cortical endpoints of bundles may span larger areas than what a researcher is interested in, challenging presumptions that bundles are specifically tied to certain brain functions. Functional MRI (fMRI) can be integrated to further refine bundles such that they are restricted to functionally-defined cortical regions. Analyzing properties of these Functional Sub-Bundles (FSuB) increases precision and interpretability of results when studying neural connections supporting specific tasks. Several parameters of DWI and fMRI analyses, ranging from data acquisition to processing, can impact the efficacy of integrating functional and diffusion MRI. Here, we discuss the applications of the FSuB approach, suggest best practices for acquiring and processing neuroimaging data towards this end, and introduce the FSuB-Extractor, a flexible open-source software for creating FSuBs. We demonstrate our processing code and the FSuB-Extractor on an openly-available dataset, the Natural Scenes Dataset.
Collapse
Affiliation(s)
- Steven L. Meisler
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Emily Kubota
- Department of Psychology, Stanford University, Stanford, CA, United States
| | - Mareike Grotheer
- Department of Psychology, Philipps-Universität Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior – CMBB, Philipps-Universität Marburg and Justus-Liebig-Universität Giessen, Marburg, Germany
| | - John D. E. Gabrieli
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Kalanit Grill-Spector
- Department of Psychology, Stanford University, Stanford, CA, United States
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
| |
Collapse
|
36
|
Louie AY, Drnevich J, Johnson JL, Woodard M, Kukekova AV, Johnson RW, Steelman AJ. Respiratory infection with influenza A virus delays remyelination and alters oligodendrocyte metabolism. iScience 2024; 27:110464. [PMID: 39104416 PMCID: PMC11298649 DOI: 10.1016/j.isci.2024.110464] [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: 02/09/2024] [Revised: 05/31/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Peripheral viral infection disrupts oligodendrocyte (OL) homeostasis such that endogenous remyelination may be affected. Here, we demonstrate that influenza A virus infection perpetuated a demyelination- and disease-associated OL phenotype following cuprizone-induced demyelination that resulted in delayed OL maturation and remyelination in the prefrontal cortex. Furthermore, we assessed cellular metabolism ex vivo, and found that infection altered brain OL and microglia metabolism in a manner that opposed the metabolic profile induced by remyelination. Specifically, infection increased glycolytic capacity of OLs and microglia, an effect that was recapitulated by lipopolysaccharide (LPS) stimulation of mixed glia cultures. In contrast, mitochondrial dependence was increased in OLs during remyelination, which was similarly observed in OLs of myelinating P14 mice compared to adult and aged mice. Collectively, our data indicate that respiratory viral infection is capable of suppressing remyelination, and suggest that metabolic dysfunction of OLs is implicated in remyelination impairment.
Collapse
Affiliation(s)
- Allison Y. Louie
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jenny Drnevich
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jennifer L. Johnson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Meagan Woodard
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Anna V. Kukekova
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Rodney W. Johnson
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Andrew J. Steelman
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
37
|
Bame X, Hill RA. Mitochondrial network reorganization and transient expansion during oligodendrocyte generation. Nat Commun 2024; 15:6979. [PMID: 39143079 PMCID: PMC11324877 DOI: 10.1038/s41467-024-51016-2] [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: 11/27/2023] [Accepted: 07/24/2024] [Indexed: 08/16/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) give rise to myelinating oligodendrocytes of the brain. This process persists throughout life and is essential for recovery from neurodegeneration. To better understand the cellular checkpoints that occur during oligodendrogenesis, we determined the mitochondrial distribution and morphometrics across the oligodendrocyte lineage in mouse and human cerebral cortex. During oligodendrocyte generation, mitochondrial content expands concurrently with a change in subcellular partitioning towards the distal processes. These changes are followed by an abrupt loss of mitochondria in the oligodendrocyte processes and myelin, coinciding with sheath compaction. This reorganization and extensive expansion and depletion take 3 days. Oligodendrocyte mitochondria are stationary over days while OPC mitochondrial motility is modulated by animal arousal state within minutes. Aged OPCs also display decreased mitochondrial size, volume fraction, and motility. Thus, mitochondrial dynamics are linked to oligodendrocyte generation, dynamically modified by their local microenvironment, and altered in the aging brain.
Collapse
Affiliation(s)
- Xhoela Bame
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
38
|
Roth RH, Ding JB. Cortico-basal ganglia plasticity in motor learning. Neuron 2024; 112:2486-2502. [PMID: 39002543 PMCID: PMC11309896 DOI: 10.1016/j.neuron.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 07/15/2024]
Abstract
One key function of the brain is to control our body's movements, allowing us to interact with the world around us. Yet, many motor behaviors are not innate but require learning through repeated practice. Among the brain's motor regions, the cortico-basal ganglia circuit is particularly crucial for acquiring and executing motor skills, and neuronal activity in these regions is directly linked to movement parameters. Cell-type-specific adaptations of activity patterns and synaptic connectivity support the learning of new motor skills. Functionally, neuronal activity sequences become structured and associated with learned movements. On the synaptic level, specific connections become potentiated during learning through mechanisms such as long-term synaptic plasticity and dendritic spine dynamics, which are thought to mediate functional circuit plasticity. These synaptic and circuit adaptations within the cortico-basal ganglia circuitry are thus critical for motor skill acquisition, and disruptions in this plasticity can contribute to movement disorders.
Collapse
Affiliation(s)
- Richard H Roth
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
39
|
Yin C, Luo K, Zhu X, Zheng R, Wang Y, Yu G, Wang X, She F, Chen X, Li T, Chen J, Bian B, Su Y, Niu J, Wang Y. Fluoxetine Rescues Excessive Myelin Formation and Psychological Behaviors in a Murine PTSD Model. Neurosci Bull 2024; 40:1037-1052. [PMID: 39014176 PMCID: PMC11306862 DOI: 10.1007/s12264-024-01249-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/04/2024] [Indexed: 07/18/2024] Open
Abstract
Posttraumatic stress disorder (PTSD) is a complex mental disorder notable for traumatic experience memory. Although current first-line treatments are linked with clinically important symptom reduction, a large proportion of patients retained to experience considerable residual symptoms, indicating pathogenic mechanism should be illustrated further. Recent studies reported that newly formed myelin could shape neural circuit function and be implicated in fear memory preservation. However, its role in PTSD remains to be elucidated. In this study, we adopted a restraint stress-induced PTSD mouse model and found that PTSD-related neuropsychiatric symptoms were accompanied by increased myelination in the posterior parietal cortex and hippocampus. Fluoxetine, but not risperidone or sertraline, has a more profound rescue effect on neuropsychological behaviors and myelin abnormalities. Further mechanistic experiments revealed that fluoxetine could directly interfere with oligodendroglial differentiation by upregulating Wnt signaling. Our data demonstrated the correlation between PTSD and abnormal myelination, suggesting that the oligodendroglial lineage could be a target for PTSD treatment.
Collapse
Affiliation(s)
- Chenrui Yin
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Kefei Luo
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Xinyue Zhu
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Ronghang Zheng
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Yu Wang
- Department of Respiratory Diseases, Central Medical Branch of PLA General Hospital, Beijing, 100853, China
| | - Guangdan Yu
- China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xiaorui Wang
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Fei She
- Department of Emergency, the Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100142, China
| | - Xiaoying Chen
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Tao Li
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Jingfei Chen
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Baduojie Bian
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Third Military Medical University (Army Medical University), Shigatse, 857000, China
| | - Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Jianqin Niu
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China.
| | - Yuxin Wang
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China.
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Third Military Medical University (Army Medical University), Shigatse, 857000, China.
| |
Collapse
|
40
|
Baudouin L, Adès N, Kanté K, Bachelin C, Hmidan H, Deboux C, Panic R, Ben Messaoud R, Velut Y, Hamada S, Pionneau C, Duarte K, Poëa-Guyon S, Barnier JV, Nait Oumesmar B, Bouslama-Oueghlani L. Antagonistic actions of PAK1 and NF2/Merlin drive myelin membrane expansion in oligodendrocytes. Glia 2024; 72:1518-1540. [PMID: 38794866 DOI: 10.1002/glia.24570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
In the central nervous system, the formation of myelin by oligodendrocytes (OLs) relies on the switch from the polymerization of the actin cytoskeleton to its depolymerization. The molecular mechanisms that trigger this switch have yet to be elucidated. Here, we identified P21-activated kinase 1 (PAK1) as a major regulator of actin depolymerization in OLs. Our results demonstrate that PAK1 accumulates in OLs in a kinase-inhibited form, triggering actin disassembly and, consequently, myelin membrane expansion. Remarkably, proteomic analysis of PAK1 binding partners enabled the identification of NF2/Merlin as its endogenous inhibitor. Our findings indicate that Nf2 knockdown in OLs results in PAK1 activation, actin polymerization, and a reduction in OL myelin membrane expansion. This effect is rescued by treatment with a PAK1 inhibitor. We also provide evidence that the specific Pak1 loss-of-function in oligodendroglia stimulates the thickening of myelin sheaths in vivo. Overall, our data indicate that the antagonistic actions of PAK1 and NF2/Merlin on the actin cytoskeleton of the OLs are critical for proper myelin formation. These findings have broad mechanistic and therapeutic implications in demyelinating diseases and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lucas Baudouin
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Noémie Adès
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Kadia Kanté
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Corinne Bachelin
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Hatem Hmidan
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
- Al-Quds University, Faculty of Medicine, Jerusalem, Palestine
| | - Cyrille Deboux
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Radmila Panic
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Rémy Ben Messaoud
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Yoan Velut
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Soumia Hamada
- Sorbonne Université, Inserm, UMS Production et Analyse des Données en Sciences de la vie et en Santé, PASS, Plateforme Post-génomique de la Pitié-Salpêtrière, Paris, France
| | - Cédric Pionneau
- Sorbonne Université, Inserm, UMS Production et Analyse des Données en Sciences de la vie et en Santé, PASS, Plateforme Post-génomique de la Pitié-Salpêtrière, Paris, France
| | - Kévin Duarte
- Institut des Neurosciences Paris-Saclay, UMR 9197, CNRS, Université Paris-Saclay, Saclay, France
| | - Sandrine Poëa-Guyon
- Institut des Neurosciences Paris-Saclay, UMR 9197, CNRS, Université Paris-Saclay, Saclay, France
| | - Jean-Vianney Barnier
- Institut des Neurosciences Paris-Saclay, UMR 9197, CNRS, Université Paris-Saclay, Saclay, France
| | - Brahim Nait Oumesmar
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Lamia Bouslama-Oueghlani
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| |
Collapse
|
41
|
Hettwer MD, Dorfschmidt L, Puhlmann LMC, Jacob LM, Paquola C, Bethlehem RAI, Bullmore ET, Eickhoff SB, Valk SL. Longitudinal variation in resilient psychosocial functioning is associated with ongoing cortical myelination and functional reorganization during adolescence. Nat Commun 2024; 15:6283. [PMID: 39075054 PMCID: PMC11286871 DOI: 10.1038/s41467-024-50292-2] [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: 02/09/2024] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
Adolescence is a period of dynamic brain remodeling and susceptibility to psychiatric risk factors, mediated by the protracted consolidation of association cortices. Here, we investigated whether longitudinal variation in adolescents' resilience to psychosocial stressors during this vulnerable period is associated with ongoing myeloarchitectural maturation and consolidation of functional networks. We used repeated myelin-sensitive Magnetic Transfer (MT) and resting-state functional neuroimaging (n = 141), and captured adversity exposure by adverse life events, dysfunctional family settings, and socio-economic status at two timepoints, one to two years apart. Development toward more resilient psychosocial functioning was associated with increasing myelination in the anterolateral prefrontal cortex, which showed stabilized functional connectivity. Studying depth-specific intracortical MT profiles and the cortex-wide synchronization of myeloarchitectural maturation, we further observed wide-spread myeloarchitectural reconfiguration of association cortices paralleled by attenuated functional reorganization with increasingly resilient outcomes. Together, resilient/susceptible psychosocial functioning showed considerable intra-individual change associated with multi-modal cortical refinement processes at the local and system-level.
Collapse
Affiliation(s)
- Meike D Hettwer
- Institute of Neuroscience and Medicine, Brain & Behavior (INM-7), Research Centre Jülich, Jülich, Germany.
- Max Planck School of Cognition, Leipzig, Germany.
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| | - Lena Dorfschmidt
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lara M C Puhlmann
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Leibniz Institute for Resilience Research, Mainz, Germany
| | - Linda M Jacob
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Casey Paquola
- Institute of Neuroscience and Medicine, Brain & Behavior (INM-7), Research Centre Jülich, Jülich, Germany
| | | | | | - Simon B Eickhoff
- Institute of Neuroscience and Medicine, Brain & Behavior (INM-7), Research Centre Jülich, Jülich, Germany
- Max Planck School of Cognition, Leipzig, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sofie L Valk
- Institute of Neuroscience and Medicine, Brain & Behavior (INM-7), Research Centre Jülich, Jülich, Germany.
- Max Planck School of Cognition, Leipzig, Germany.
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| |
Collapse
|
42
|
Zhou LQ, Chu YH, Dong MH, Yang S, Chen M, Tang Y, Pang XW, You YF, Wu LJ, Wang W, Qin C, Tian DS. Ldl-stimulated microglial activation exacerbates ischemic white matter damage. Brain Behav Immun 2024; 119:416-430. [PMID: 38636563 DOI: 10.1016/j.bbi.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
The role of microglia in triggering the blood-brain barrier (BBB) impairment and white matter damage after chronic cerebral hypoperfusion is unclear. Here we demonstrated that the vessel-adjacent microglia were specifically activated by the leakage of plasma low-density lipoprotein (LDL), which led to BBB breakdown and ischemic demyelination. Interestingly, we found that LDL stimulation enhanced microglial phagocytosis, causing excessive engulfment of myelin debris and resulting in an overwhelming lipid burden in microglia. Surprisingly, these lipid-laden microglia exhibited a suppressed profile of inflammatory response and compromised pro-regenerative properties. Microglia-specific knockdown of LDLR or systematic medication lowering circulating LDL-C showed protective effects against ischemic demyelination. Overall, our findings demonstrated that LDL-stimulated vessel-adjacent microglia possess a disease-specific molecular signature, characterized by suppressed regenerative properties, which is associated with the propagation of demyelination during ischemic white matter damage.
Collapse
Affiliation(s)
- Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun-Fan You
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
43
|
Takemura H, Kruper JA, Miyata T, Rokem A. Tractometry of Human Visual White Matter Pathways in Health and Disease. Magn Reson Med Sci 2024; 23:316-340. [PMID: 38866532 PMCID: PMC11234945 DOI: 10.2463/mrms.rev.2024-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Diffusion-weighted MRI (dMRI) provides a unique non-invasive view of human brain tissue properties. The present review article focuses on tractometry analysis methods that use dMRI to assess the properties of brain tissue within the long-range connections comprising brain networks. We focus specifically on the major white matter tracts that convey visual information. These connections are particularly important because vision provides rich information from the environment that supports a large range of daily life activities. Many of the diseases of the visual system are associated with advanced aging, and tractometry of the visual system is particularly important in the modern aging society. We provide an overview of the tractometry analysis pipeline, which includes a primer on dMRI data acquisition, voxelwise model fitting, tractography, recognition of white matter tracts, and calculation of tract tissue property profiles. We then review dMRI-based methods for analyzing visual white matter tracts: the optic nerve, optic tract, optic radiation, forceps major, and vertical occipital fasciculus. For each tract, we review background anatomical knowledge together with recent findings in tractometry studies on these tracts and their properties in relation to visual function and disease. Overall, we find that measurements of the brain's visual white matter are sensitive to a range of disorders and correlate with perceptual abilities. We highlight new and promising analysis methods, as well as some of the current barriers to progress toward integration of these methods into clinical practice. These barriers, such as variability in measurements between protocols and instruments, are targets for future development.
Collapse
Affiliation(s)
- Hiromasa Takemura
- Division of Sensory and Cognitive Brain Mapping, Department of System Neuroscience, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Hayama, Kanagawa, Japan
- Center for Information and Neural Networks (CiNet), Advanced ICT Research Institute, National Institute of Information and Communications Technology, Suita, Osaka, Japan
| | - John A Kruper
- Department of Psychology and eScience Institute, University of Washington, Seattle, WA, USA
| | - Toshikazu Miyata
- Division of Sensory and Cognitive Brain Mapping, Department of System Neuroscience, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Center for Information and Neural Networks (CiNet), Advanced ICT Research Institute, National Institute of Information and Communications Technology, Suita, Osaka, Japan
| | - Ariel Rokem
- Department of Psychology and eScience Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
44
|
Akter M, Fu Z, Zheng X, Iqbal Z, Zhang N, Karim A, Li Y. Astrocytic GPCR signaling in the anterior cingulate cortex modulates decision making in rats. OXFORD OPEN NEUROSCIENCE 2024; 3:kvae010. [PMID: 38915791 PMCID: PMC11194462 DOI: 10.1093/oons/kvae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/26/2024]
Abstract
Decision making is a process of selecting a course of action by assessing the worth or value of the potential consequences. Rat Gambling Task (RGT) is a well-established behavioral paradigm that allows for assessment of the decision-making performance of rats. Astrocytes are emerging as key players in modulating cognitive functions. Using repeated RGTs with short intersession time intervals (48 h), the current study demonstrates that Gi pathway activation of astrocytes in the anterior cingulate cortex (ACC) leads to impaired decision-making in consistently good decision-making rats. On the other hand, ACC astrocytic Gq pathway activation improves decision-making in a subset of rats who are not consistently good decision-makers. Furthermore, we show that astrocytic Gq activation is associated with an increase in the L-lactate level in the extracellular fluid of the ACC. Together, these results expand our knowledge of the role of astrocytic GPCR signaling in modulating cognitive functions.
Collapse
Affiliation(s)
- Mastura Akter
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| | - Zhongqi Fu
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| | - Xianlin Zheng
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| | - Zafar Iqbal
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 15 Science Park West Avenue, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong, SAR, China
- Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 17W, Science Park West Avenue, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong, SAR, China
| | - Na Zhang
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| | - Anwarul Karim
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| | - Ying Li
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 15 Science Park West Avenue, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong, SAR, China
- Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 17W, Science Park West Avenue, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong, SAR, China
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| |
Collapse
|
45
|
Zhang Y, Tian L, Chen J, Liu X, Li K, Liu H, Lai W, Shi Y, Lin B, Xi Z. Selective bioaccumulation of polystyrene nanoplastics in fetal rat brain and damage to myelin development. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116393. [PMID: 38714083 DOI: 10.1016/j.ecoenv.2024.116393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/13/2024] [Accepted: 04/22/2024] [Indexed: 05/09/2024]
Abstract
Micro(nano)plastic, as a new type of environmental pollutant, have become a potential threat to the life and health of various stages of biology. However, it is not yet clear whether they will affect brain development in the fetal stage. Therefore, this study aims to explore the potential effects of nanoplastics on the development of fetal rat brains. To assess the allocation of NPs (25 nm and 50 nm) in various regions of the fetal brain, pregnant rats were exposed to concentrations (50, 10, 2.5, and 0.5 mg/kg) of PS-NPs. Our results provided evidence of the transplacental transfer of PS-NPs to the fetal brain, with a prominent presence observed in several cerebral regions, notably the cerebellum, hippocampus, striatum, and prefrontal cortex. This distribution bias might be linked to the developmental sequence of each brain region. Additionally, we explored the influence of prenatal exposure on the myelin development of the cerebellum, given its the highest PS-NP accumulation in offspring. Compared with control rats, PS-NPs exposure caused a significant reduction in myelin basic protein (MBP) and myelin oligodendrocyte glycoprotein (MOG) expression, a decrease in myelin thickness, an increase in cell apoptosis, and a decline in the oligodendrocyte population. These effects gave rise to motor deficits. In conclusion, our results identified the specific distribution of NPs in the fetal brain following prenatal exposure and revealed that prenatal exposure to PS-NPs can suppress myelin formation in the cerebellum of the fetus.
Collapse
Affiliation(s)
- Yaping Zhang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School of Public Health and Management, Binzhou Medical University, Yantai 264003, China
| | - Lei Tian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Jiang Chen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School of Public Health, North China University of Science and Technology, Tangshan 063200, China
| | - Xuan Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Kang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Huanliang Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Wenqing Lai
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yue Shi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| |
Collapse
|
46
|
Akter M, Li Y. Does astrocytic L-lactate enhance cognition through myelination? Neural Regen Res 2024; 19:1167-1168. [PMID: 37905847 PMCID: PMC11467931 DOI: 10.4103/1673-5374.385872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/07/2023] [Accepted: 08/23/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Mastura Akter
- Department of Neuroscience, City University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ying Li
- Department of Neuroscience, City University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Special Administrative Region, China
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
47
|
Cui W, Yang J, Tu C, Zhang Z, Zhao H, Qiao Y, Li Y, Yang W, Lim KL, Ma Q, Zhang C, Lu L. Seipin deficiency-induced lipid dysregulation leads to hypomyelination-associated cognitive deficits via compromising oligodendrocyte precursor cell differentiation. Cell Death Dis 2024; 15:350. [PMID: 38773070 PMCID: PMC11109229 DOI: 10.1038/s41419-024-06737-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/23/2024]
Abstract
Seipin is one key mediator of lipid metabolism that is highly expressed in adipose tissues as well as in the brain. Lack of Seipin gene, Bscl2, leads to not only severe lipid metabolic disorders but also cognitive impairments and motor disabilities. Myelin, composed mainly of lipids, facilitates nerve transmission and is important for motor coordination and learning. Whether Seipin deficiency-leaded defects in learning and motor coordination is underlined by lipid dysregulation and its consequent myelin abnormalities remains to be elucidated. In the present study, we verified the expression of Seipin in oligodendrocytes (OLs) and their precursors, oligodendrocyte precursor cells (OPCs), and demonstrated that Seipin deficiency compromised OPC differentiation, which led to decreased OL numbers, myelin protein, myelinated fiber proportion and thickness of myelin. Deficiency of Seipin resulted in impaired spatial cognition and motor coordination in mice. Mechanistically, Seipin deficiency suppressed sphingolipid metabolism-related genes in OPCs and caused morphological abnormalities in lipid droplets (LDs), which markedly impeded OPC differentiation. Importantly, rosiglitazone, one agonist of PPAR-gamma, substantially restored phenotypes resulting from Seipin deficiency, such as aberrant LDs, reduced sphingolipids, obstructed OPC differentiation, and neurobehavioral defects. Collectively, the present study elucidated how Seipin deficiency-induced lipid dysregulation leads to neurobehavioral deficits via impairing myelination, which may pave the way for developing novel intervention strategy for treating metabolism-involved neurological disorders.
Collapse
Affiliation(s)
- Wenli Cui
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jing Yang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chuanyun Tu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Ziting Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Huifang Zhao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yan Qiao
- Analytical Instrumentation Center & State Key Laboratory of Coal Conversion, Institute of Coal Chemistry, Chinese Academy of Sciences, Taiyuan, 030001, Shanxi, China
| | - Yanqiu Li
- Analytical Instrumentation Center & State Key Laboratory of Coal Conversion, Institute of Coal Chemistry, Chinese Academy of Sciences, Taiyuan, 030001, Shanxi, China
| | - Wulin Yang
- Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Kah-Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Quanhong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China.
| | - Chengwu Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Li Lu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
48
|
Milbocker KA, Smith IF, Klintsova AY. Maintaining a Dynamic Brain: A Review of Empirical Findings Describing the Roles of Exercise, Learning, and Environmental Enrichment in Neuroplasticity from 2017-2023. Brain Plast 2024; 9:75-95. [PMID: 38993580 PMCID: PMC11234674 DOI: 10.3233/bpl-230151] [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] [Accepted: 10/19/2023] [Indexed: 07/13/2024] Open
Abstract
Brain plasticity, also termed neuroplasticity, refers to the brain's life-long ability to reorganize itself in response to various changes in the environment, experiences, and learning. The brain is a dynamic organ capable of responding to stimulating or depriving environments, activities, and circumstances from changes in gene expression, release of neurotransmitters and neurotrophic factors, to cellular reorganization and reprogrammed functional connectivity. The rate of neuroplastic alteration varies across the lifespan, creating further challenges for understanding and manipulating these processes to benefit motor control, learning, memory, and neural remodeling after injury. Neuroplasticity-related research spans several decades, and hundreds of reviews have been written and published since its inception. Here we present an overview of the empirical papers published between 2017 and 2023 that address the unique effects of exercise, plasticity-stimulating activities, and the depriving effect of social isolation on brain plasticity and behavior.
Collapse
Affiliation(s)
| | - Ian F. Smith
- Department of Psychological and Brain Sciences, University of Delaware, University of Delaware, Newark, USA
| | - Anna Y. Klintsova
- Department of Psychological and Brain Sciences, University of Delaware, University of Delaware, Newark, USA
| |
Collapse
|
49
|
Elitt CM, Ross MM, Wang J, Fahrni CJ, Rosenberg PA. Developmental regulation of zinc homeostasis in differentiating oligodendrocytes. Neurosci Lett 2024; 831:137727. [PMID: 38467270 DOI: 10.1016/j.neulet.2024.137727] [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: 07/09/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
Oligodendrocytes develop through sequential stages and understanding pathways regulating their differentiation remains an important area of investigation. Zinc is required for the function of enzymes, proteins and transcription factors, including those important in myelination and mitosis. Our previous studies using the ratiometric zinc sensor chromis-1 demonstrated a reduction in intracellular free zinc concentrations in mature MBP+ oligodendrocytes compared with earlier stages (Bourassa et al., 2018). We performed a more detailed developmental study to better understand the temporal course of zinc homeostasis across the oligodendrocyte lineage. Using chromis-1, we found a transient increase in free zinc after O4+,O1- pre-oligodendrocytes were switched from proliferation medium into terminal differentiation medium. To gather other evidence for dynamic regulation of free zinc during oligodendrocyte development, qPCR was used to evaluate mRNA expression of major zinc storage proteins metallothioneins (MTs) and metal regulatory transcription factor 1 (MTF1), which controls expression of MTs. MT1, MT2 and MTF1 mRNAs were increased several fold in mature oligodendrocytes compared to oligodendrocytes in proliferation medium. To assess the depth of the zinc buffer, we assayed zinc release from intracellular stores using the oxidizing thiol reagent 2,2'-dithiodipyridine (DTDP). Exposure to DTDP resulted in ∼ 100% increase in free zinc in pre-oligodendrocytes but, paradoxically more modest ∼ 60% increase in mature oligodendrocytes despite increased expression of MTs. These results suggest that zinc homeostasis is regulated during oligodendrocyte development, that oligodendrocytes are a useful model for studying zinc homeostasis in the central nervous system, and that regulation of zinc homeostasis may be important in oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Christopher M Elitt
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States.
| | - Madeline M Ross
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States
| | - Jianlin Wang
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States
| | - Christoph J Fahrni
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, United States; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, United States
| | - Paul A Rosenberg
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States
| |
Collapse
|
50
|
Mercier O, Quilichini PP, Magalon K, Gil F, Ghestem A, Richard F, Boudier T, Cayre M, Durbec P. Transient demyelination causes long-term cognitive impairment, myelin alteration and network synchrony defects. Glia 2024; 72:960-981. [PMID: 38363046 DOI: 10.1002/glia.24513] [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: 05/17/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024]
Abstract
In the adult brain, activity-dependent myelin plasticity is required for proper learning and memory consolidation. Myelin loss, alteration, or even subtle structural modifications can therefore compromise the network activity, leading to functional impairment. In multiple sclerosis, spontaneous myelin repair process is possible, but it is heterogeneous among patients, sometimes leading to functional recovery, often more visible at the motor level than at the cognitive level. In cuprizone-treated mouse model, massive brain demyelination is followed by spontaneous and robust remyelination. However, reformed myelin, although functional, may not exhibit the same morphological characteristics as developmental myelin, which can have an impact on the activity of neural networks. In this context, we used the cuprizone-treated mouse model to analyze the structural, functional, and cognitive long-term effects of transient demyelination. Our results show that an episode of demyelination induces despite remyelination long-term cognitive impairment, such as deficits in spatial working memory, social memory, cognitive flexibility, and hyperactivity. These deficits were associated with a reduction in myelin content in the medial prefrontal cortex (mPFC) and hippocampus (HPC), as well as structural myelin modifications, suggesting that the remyelination process may be imperfect in these structures. In vivo electrophysiological recordings showed that the demyelination episode altered the synchronization of HPC-mPFC activity, which is crucial for memory processes. Altogether, our data indicate that the myelin repair process following transient demyelination does not allow the complete recovery of the initial myelin properties in cortical structures. These subtle modifications alter network features, leading to prolonged cognitive deficits in mice.
Collapse
Affiliation(s)
- Océane Mercier
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Pascale P Quilichini
- U1106 after INS, Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Karine Magalon
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Florian Gil
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Antoine Ghestem
- U1106 after INS, Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Fabrice Richard
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Thomas Boudier
- Aix Marseille Univ, Turing Centre for Living Systems, Marseille, France
| | - Myriam Cayre
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Pascale Durbec
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| |
Collapse
|