51
|
Beiter RM, Raghavan TP, Suchocki O, Ennerfelt HE, Rivet-Noor CR, Merchak AR, Phillips JL, Bathe T, Lukens JR, Prokop S, Dupree JL, Gaultier A. Oligomeric amyloid beta prevents myelination in a clusterin-dependent manner. RESEARCH SQUARE 2024:rs.3.rs-4415143. [PMID: 38853911 PMCID: PMC11160922 DOI: 10.21203/rs.3.rs-4415143/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background White matter loss is a well-documented phenomenon in Alzheimer's disease (AD) patients that has been recognized for decades. However, the underlying reasons for the failure of oligodendrocyte progenitor cells (OPCs) to repair myelin deficits in these patients remain elusive. A single nucleotide polymorphism (SNP) in Clusterin has been identified as a risk factor for late-onset Alzheimer's disease and linked to a decrease in white matter integrity in healthy adults, but its specific role in oligodendrocyte function and myelin maintenance in Alzheimer's disease pathology remains unclear. Methods To investigate the impact of Clusterin on OPCs in the context of Alzheimer's disease, we employed a combination of immunofluorescence and transmission electron microscopy techniques, primary culture of OPCs, and an animal model of Alzheimer's disease. Results Our findings demonstrate that Clusterin, a risk factor for late-onset AD, is produced by OPCs and inhibits their differentiation into oligodendrocytes. Specifically, we observed upregulation of Clusterin in OPCs in the 5xFAD mouse model of AD. We also found that the phagocytosis of debris, including amyloid beta (Aβ), myelin, and apoptotic cells leads to the upregulation of Clusterin in OPCs. In vivo experiments confirmed that Aβ oligomers stimulate Clusterin upregulation and that OPCs are capable of phagocytosing Aβ. Furthermore, we discovered that Clusterin significantly inhibits OPC differentiation and hinders the production of myelin proteins. Finally, we demonstrate that Clusterin inhibits OPC differentiation by reducing the production of IL-9 by OPCs. Conclusion Our data suggest that Clusterin may play a key role in the impaired myelin repair observed in AD and could serve as a promising therapeutic target for addressing AD-associated cognitive decline.
Collapse
|
52
|
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] [Download PDF] [Figures] [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
|
53
|
Huang R, Carter ER, Hughes EG, Welle CG. Paired vagus nerve stimulation drives precise remyelination and motor recovery after myelin loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593609. [PMID: 38766201 PMCID: PMC11100833 DOI: 10.1101/2024.05.10.593609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Myelin loss in the central nervous system can cause permanent motor or cognitive deficits in patients with multiple sclerosis (MS). While current immunotherapy treatments decrease the frequency of demyelinating episodes, they do not promote myelin repair or functional recovery. Vagus nerve stimulation (VNS) is a neuromodulation therapy which enhances neuroplasticity and the recovery of motor function after stroke, but its effects on myelin repair are not known. To determine if VNS influences myelin repair, we applied VNS following a demyelinating injury and measured longitudinal myelin dynamics and functional recovery. We found that VNS promotes remyelination by increasing the generation of myelinating oligodendrocytes. Pairing VNS with a skilled reach task leads to the regeneration of myelin sheaths on previously myelinated axon segments, enhancing the restoration of the original pattern of myelination. Moreover, the magnitude of sheath pattern restoration correlates with long-term motor functional improvement. Together, these results suggest that recovery of the myelin sheath pattern is a key factor in the restoration of motor function following myelin loss and identify paired VNS as a potential remyelination therapy to treat demyelinating diseases.
Collapse
|
54
|
Tang Y, Wu X, Li J, Li Y, Xu X, Li G, Zhang P, Qin C, Wu LJ, Tang Z, Tian DS. The Emerging Role of Microglial Hv1 as a Target for Immunomodulation in Myelin Repair. Aging Dis 2024; 15:1176-1203. [PMID: 38029392 PMCID: PMC11081154 DOI: 10.14336/ad.2023.1107] [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: 08/17/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
In the central nervous system (CNS), the myelin sheath ensures efficient interconnection between neurons and contributes to the regulation of the proper function of neuronal networks. The maintenance of myelin and the well-organized subtle process of myelin plasticity requires cooperation among myelin-forming cells, glial cells, and neural networks. The process of cooperation is fragile, and the balance is highly susceptible to disruption by microenvironment influences. Reactive microglia play a critical and complicated role in the demyelination and remyelination process. Recent studies have shown that the voltage-gated proton channel Hv1 is selectively expressed in microglia in CNS, which regulates intracellular pH and is involved in the production of reactive oxygen species, underlying multifaceted roles in maintaining microglia function. This paper begins by examining the molecular mechanisms of demyelination and emphasizes the crucial role of the microenvironment in demyelination. It focuses specifically on the role of Hv1 in myelin repair and its therapeutic potential in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoxiao Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
55
|
Islam R, White JD, Arefin TM, Mehta S, Liu X, Polis B, Giuliano L, Ahmed S, Bowers C, Zhang J, Kaffman A. Early adversity causes sex-specific deficits in perforant pathway connectivity and contextual memory in adolescent mice. Biol Sex Differ 2024; 15:39. [PMID: 38715106 PMCID: PMC11075329 DOI: 10.1186/s13293-024-00616-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Early life adversity impairs hippocampal development and function across diverse species. While initial evidence indicated potential variations between males and females, further research is required to validate these observations and better understand the underlying mechanisms contributing to these sex differences. Furthermore, most of the preclinical work in rodents was performed in adult males, with only few studies examining sex differences during adolescence when such differences appear more pronounced. To address these concerns, we investigated the impact of limited bedding (LB), a mouse model of early adversity, on hippocampal development in prepubescent and adolescent male and female mice. METHODS RNA sequencing, confocal microscopy, and electron microscopy were used to evaluate the impact of LB and sex on hippocampal development in prepubescent postnatal day 17 (P17) mice. Additional studies were conducted on adolescent mice aged P29-36, which included contextual fear conditioning, retrograde tracing, and ex vivo diffusion magnetic resonance imaging (dMRI). RESULTS More severe deficits in axonal innervation and myelination were found in the perforant pathway of prepubescent and adolescent LB males compared to LB female littermates. These sex differences were due to a failure of reelin-positive neurons located in the lateral entorhinal cortex (LEC) to innervate the dorsal hippocampus via the perforant pathway in males, but not LB females, and were strongly correlated with deficits in contextual fear conditioning. CONCLUSIONS LB impairs the capacity of reelin-positive cells located in the LEC to project and innervate the dorsal hippocampus in LB males but not female LB littermates. Given the critical role that these projections play in supporting normal hippocampal function, a failure to establish proper connectivity between the LEC and the dorsal hippocampus provides a compelling and novel mechanism to explain the more severe deficits in myelination and contextual freezing found in adolescent LB males.
Collapse
Affiliation(s)
- Rafiad Islam
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Jordon D White
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Tanzil M Arefin
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, 10016, USA
- Department of Biomedical Engineering, Center for Neurotechnology in Mental Health Research (CNMHR), The Pennsylvania State University, University Park, PA, 16802, USA
| | - Sameet Mehta
- Yale Center for Genomic Analysis, P.O. Box 27386, West Haven, CT, 06516-7386, USA
| | - Xinran Liu
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, SHM IE26, New Haven, CT, 06510, USA
- Center for Cellular and Molecular Imaging, Electron Microscopy Core Facility, Yale University School of Medicine, New Haven, CT, USA
| | - Baruh Polis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Lauryn Giuliano
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Sahabuddin Ahmed
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Christian Bowers
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Jiangyang Zhang
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA.
| |
Collapse
|
56
|
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
|
57
|
Fu C, Tang H, Liu L, Huang Y, Zhou H, Huang S, Peng T, Zeng P, Yang X, He L, Xu K. Constraint-Induced Movement Therapy Promotes Myelin Remodeling and Motor Function by Mediating Sox2/Fyn Signals in Rats With Hemiplegic Cerebral Palsy. Phys Ther 2024; 104:pzae011. [PMID: 38302073 DOI: 10.1093/ptj/pzae011] [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: 09/17/2022] [Revised: 08/27/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024]
Abstract
OBJECTIVE Hypoxic-ischemic brain injury in infants often leads to hemiplegic motor dysfunction. The mechanism of their motor dysfunction has been attributed to deficiencies of the transcription factor sex-determining region (SRY) box 2 (Sox2) or the non-receptor-type tyrosine kinase Fyn (involved in neuronal signal transduction), which causes a defect in myelin formation. Constraint-induced movement therapy (CIMT) following cerebral hypoxia-ischemia may stimulate myelin growth by regulating Sox2/Fyn, Ras homolog protein family A (RhoA), and rho-associated kinase 2 (ROCK2) expression levels. This study investigated how Sox2/Fyn regulates myelin remodeling following CIMT to improve motor function in rats with hemiplegic cerebral palsy (HCP). METHODS To investigate the mechanism of Sox2 involvement in myelin growth and neural function in rats with HCP, Lentivirus (Lenti)-Sox2 adeno-associated virus and negative control-Lenti-Sox2 (LS) adeno-associated virus were injected into the lateral ventricle. The rats were divided into a control group and an HCP group with different interventions (CIMT, LS, or negative control-LS [NS] treatment), yielding the HCP, HCP plus CIMT (HCP + CIMT), HCP + LS, HCP + LS + CIMT, HCP + NS, and HCP + NS + CIMT groups. Front-limb suspension and RotaRod tests, Golgi-Cox staining, transmission electron microscopy, immunofluorescence staining, western blotting, and quantitative polymerase chain reaction experiments were used to analyze the motor function, dendrite/axon area, myelin ultrastructure, and levels of expression of oligodendrocytes and Sox2/Fyn/RhoA/ROCK2 in the motor cortex. RESULTS The rats in the HCP + LS + CIMT group had better values for motor function, dendrite/axon area, myelin ultrastructure, oligodendrocytes, and Sox2/Fyn/RhoA/ROCK2 expression in the motor cortex than rats in the HCP and HCP + NS groups. The improvement of motor function and myelin remodeling, the expression of oligodendrocytes, and the expression of Sox2/Fyn/RhoA/ROCK2 in the HCP + LS group were similar to those in the HCP + CIMT group. CONCLUSION CIMT might overcome RhoA/ROCK2 signaling by upregulating the transcription of Sox2 to Fyn in the brain to induce the maturation and differentiation of oligodendrocytes, thereby promoting myelin remodeling and improving motor function in rats with HCP. IMPACT The pathway mediated by Sox2/Fyn could be a promising therapeutic target for HCP.
Collapse
Affiliation(s)
- Chaoqiong Fu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
- Department of Rehabilitation, School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Hongmei Tang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Liru Liu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Yuan Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
- School of Exercise and Health, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hongyu Zhou
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Shiya Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
- Department of Rehabilitation, School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Tingting Peng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Peishan Zeng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Xubo Yang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Lu He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Kaishou Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Rehabilitation, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| |
Collapse
|
58
|
Cohn EF, Clayton BLL, Madhavan M, Lee KA, Yacoub S, Fedorov Y, Scavuzzo MA, Paul Friedman K, Shafer TJ, Tesar PJ. Pervasive environmental chemicals impair oligodendrocyte development. Nat Neurosci 2024; 27:836-845. [PMID: 38528201 PMCID: PMC11088982 DOI: 10.1038/s41593-024-01599-2] [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: 11/15/2022] [Accepted: 02/05/2024] [Indexed: 03/27/2024]
Abstract
Exposure to environmental chemicals can impair neurodevelopment, and oligodendrocytes may be particularly vulnerable, as their development extends from gestation into adulthood. However, few environmental chemicals have been assessed for potential risks to oligodendrocytes. Here, using a high-throughput developmental screen in cultured cells, we identified environmental chemicals in two classes that disrupt oligodendrocyte development through distinct mechanisms. Quaternary compounds, ubiquitous in disinfecting agents and personal care products, were potently and selectively cytotoxic to developing oligodendrocytes, whereas organophosphate flame retardants, commonly found in household items such as furniture and electronics, prematurely arrested oligodendrocyte maturation. Chemicals from each class impaired oligodendrocyte development postnatally in mice and in a human 3D organoid model of prenatal cortical development. Analysis of epidemiological data showed that adverse neurodevelopmental outcomes were associated with childhood exposure to the top organophosphate flame retardant identified by our screen. This work identifies toxicological vulnerabilities for oligodendrocyte development and highlights the need for deeper scrutiny of these compounds' impacts on human health.
Collapse
Affiliation(s)
- Erin F Cohn
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Benjamin L L Clayton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Mayur Madhavan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kristin A Lee
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sara Yacoub
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yuriy Fedorov
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Marissa A Scavuzzo
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Katie Paul Friedman
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Timothy J Shafer
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
59
|
Sun Z, Shi C, Jin L. Mechanisms by Which SARS-CoV-2 Invades and Damages the Central Nervous System: Apart from the Immune Response and Inflammatory Storm, What Else Do We Know? Viruses 2024; 16:663. [PMID: 38793545 PMCID: PMC11125732 DOI: 10.3390/v16050663] [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: 01/30/2024] [Revised: 03/29/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Initially reported as pneumonia of unknown origin, COVID-19 is increasingly being recognized for its impact on the nervous system, despite nervous system invasions being extremely rare. As a result, numerous studies have been conducted to elucidate the mechanisms of nervous system damage and propose appropriate coping strategies. This review summarizes the mechanisms by which SARS-CoV-2 invades and damages the central nervous system, with a specific focus on aspects apart from the immune response and inflammatory storm. The latest research findings on these mechanisms are presented, providing new insights for further in-depth research.
Collapse
Affiliation(s)
- Zihan Sun
- Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Chunying Shi
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Lixin Jin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| |
Collapse
|
60
|
Poggi G, Klaus F, Pryce CR. Pathophysiology in cortico-amygdala circuits and excessive aversion processing: the role of oligodendrocytes and myelination. Brain Commun 2024; 6:fcae140. [PMID: 38712320 PMCID: PMC11073757 DOI: 10.1093/braincomms/fcae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/27/2023] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Stress-related psychiatric illnesses, such as major depressive disorder, anxiety and post-traumatic stress disorder, present with alterations in emotional processing, including excessive processing of negative/aversive stimuli and events. The bidirectional human/primate brain circuit comprising anterior cingulate cortex and amygdala is of fundamental importance in processing emotional stimuli, and in rodents the medial prefrontal cortex-amygdala circuit is to some extent analogous in structure and function. Here, we assess the comparative evidence for: (i) Anterior cingulate/medial prefrontal cortex<->amygdala bidirectional neural circuits as major contributors to aversive stimulus processing; (ii) Structural and functional changes in anterior cingulate cortex<->amygdala circuit associated with excessive aversion processing in stress-related neuropsychiatric disorders, and in medial prefrontal cortex<->amygdala circuit in rodent models of chronic stress-induced increased aversion reactivity; and (iii) Altered status of oligodendrocytes and their oligodendrocyte lineage cells and myelination in anterior cingulate/medial prefrontal cortex<->amygdala circuits in stress-related neuropsychiatric disorders and stress models. The comparative evidence from humans and rodents is that their respective anterior cingulate/medial prefrontal cortex<->amygdala circuits are integral to adaptive aversion processing. However, at the sub-regional level, the anterior cingulate/medial prefrontal cortex structure-function analogy is incomplete, and differences as well as similarities need to be taken into account. Structure-function imaging studies demonstrate that these neural circuits are altered in both human stress-related neuropsychiatric disorders and rodent models of stress-induced increased aversion processing. In both cases, the changes include altered white matter integrity, albeit the current evidence indicates that this is decreased in humans and increased in rodent models. At the cellular-molecular level, in both humans and rodents, the current evidence is that stress disorders do present with changes in oligodendrocyte lineage, oligodendrocytes and/or myelin in these neural circuits, but these changes are often discordant between and even within species. Nonetheless, by integrating the current comparative evidence, this review provides a timely insight into this field and should function to inform future studies-human, monkey and rodent-to ascertain whether or not the oligodendrocyte lineage and myelination are causally involved in the pathophysiology of stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Giulia Poggi
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, CH-8008 Zurich, Switzerland
| | - Federica Klaus
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA
- Desert-Pacific Mental Illness Research Education and Clinical Center, VA San Diego Healthcare System, San Diego, CA 92093, USA
| | - Christopher R Pryce
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, CH-8008 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
- URPP Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
61
|
Cisneros-Mejorado AJ, Ordaz RP, Garay E, Arellano RO. β-carbolines that enhance GABA A receptor response expressed in oligodendrocytes promote remyelination in an in vivo rat model of focal demyelination. Front Cell Neurosci 2024; 18:1369730. [PMID: 38694535 PMCID: PMC11061515 DOI: 10.3389/fncel.2024.1369730] [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: 01/12/2024] [Accepted: 04/09/2024] [Indexed: 05/04/2024] Open
Abstract
Demyelination is typically followed by a remyelination process through mature oligodendrocytes (OLs) differentiated from precursor cells (OPCs) recruited into the lesioned areas, however, this event usually results in uncompleted myelination. Potentiation of the remyelination process is an important target for designing effective therapeutic strategies against white matter loss. Here, it was evaluated the remyelinating effect of different β-carbolines that present differential allosteric modulation on the GABAA receptor expressed in OLs. For this, we used a focalized demyelination model in the inferior cerebellar peduncle (i.c.p.) of rats (DRICP model), in which, demyelination by ethidium bromide (0.05%) stereotaxic injection was confirmed histologically by staining with Black-Gold II (BGII) and toluidine blue. In addition, a longitudinal analysis with diffusion-weighted magnetic resonance imaging (dMRI) was made by computing fractional anisotropy (FA), apparent diffusion coefficient (ADC) and diffusivity parameters to infer i.c.p. microstructural changes. First, dMRI analysis revealed FA decreases together with ADC and radial diffusivity (RD) increases after demyelination, which correlates with histological BGII observations. Then, we evaluated the effect produced by three allosteric GABAA receptor modulators, the N-butyl-β-carboline-3-carboxylate (β-CCB), ethyl 9H-pyrido [3,4-b]indole-3-carboxylate (β-CCE), and 4-ethyl-6,7-dimethoxy-9H-pyrido [3,4-b]indole-3-carboxylic acid methyl ester (DMCM). The results indicated that daily systemic β-CCB (1 mg/Kg) or β-CCE (1 mg/Kg) administration for 2 weeks, but not DMCM (0.35 mg/Kg), in lesioned animals increased FA and decreased ADC or RD, suggesting myelination improvement. This was supported by BGII staining analysis that showed a recovery of myelin content. Also, it was quantified by immunohistochemistry both NG2+ and CC1+ cellular population in the different experimental sceneries. Data indicated that either β-CCB or β-CCE, but not DMCM, produced an increase in the population of CC1+ cells in the lesioned area. Finally, it was also calculated the g-ratio of myelinated axons and observed a similar value in those lesioned animals treated with β-CCB or β-CCE compared to controls. Thus, using the DRICP model, it was observed that either β-CCB or β-CCE, positive modulators of the GABAA receptor in OLs, had a potent promyelinating effect.
Collapse
Affiliation(s)
| | | | | | - Rogelio O. Arellano
- Instituto de Neurobiología, Laboratorio de Neurofisiología Celular, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| |
Collapse
|
62
|
Bae HG, Wu WC, Nip K, Gould E, Kim JH. Scn2a deletion disrupts oligodendroglia function: Implication for myelination, neural circuitry, and auditory hypersensitivity in ASD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589242. [PMID: 38659965 PMCID: PMC11042360 DOI: 10.1101/2024.04.15.589242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Autism spectrum disorder (ASD) is characterized by a complex etiology, with genetic determinants significantly influencing its manifestation. Among these, the Scn2a gene emerges as a pivotal player, crucially involved in both glial and neuronal functionality. This study elucidates the underexplored roles of Scn2a in oligodendrocytes, and its subsequent impact on myelination and auditory neural processes. The results reveal a nuanced interplay between oligodendrocytes and axons, where Scn2a deletion causes alterations in the intricate process of myelination. This disruption, in turn, instigates changes in axonal properties and neuronal activities at the single cell level. Furthermore, oligodendrocyte-specific Scn2a deletion compromises the integrity of neural circuitry within auditory pathways, leading to auditory hypersensitivity-a common sensory abnormality observed in ASD. Through transcriptional profiling, we identified alterations in the expression of myelin-associated genes, highlighting the cellular consequences engendered by Scn2a deletion. In summary, the findings provide unprecedented insights into the pathway from Scn2a deletion in oligodendrocytes to sensory abnormalities in ASD, underscoring the integral role of Scn2a -mediated myelination in auditory responses. This research thereby provides novel insights into the intricate tapestry of genetic and cellular interactions inherent in ASD.
Collapse
|
63
|
Khelfaoui H, Ibaceta-Gonzalez C, Angulo MC. Functional myelin in cognition and neurodevelopmental disorders. Cell Mol Life Sci 2024; 81:181. [PMID: 38615095 PMCID: PMC11016012 DOI: 10.1007/s00018-024-05222-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/18/2024] [Accepted: 03/30/2024] [Indexed: 04/15/2024]
Abstract
In vertebrates, oligodendrocytes (OLs) are glial cells of the central nervous system (CNS) responsible for the formation of the myelin sheath that surrounds the axons of neurons. The myelin sheath plays a crucial role in the transmission of neuronal information by promoting the rapid saltatory conduction of action potentials and providing neurons with structural and metabolic support. Saltatory conduction, first described in the peripheral nervous system (PNS), is now generally recognized as a universal evolutionary innovation to respond quickly to the environment: myelin helps us think and act fast. Nevertheless, the role of myelin in the central nervous system, especially in the brain, may not be primarily focused on accelerating conduction speed but rather on ensuring precision. Its principal function could be to coordinate various neuronal networks, promoting their synchronization through oscillations (or rhythms) relevant for specific information processing tasks. Interestingly, myelin has been directly involved in different types of cognitive processes relying on brain oscillations, and myelin plasticity is currently considered to be part of the fundamental mechanisms for memory formation and maintenance. However, despite ample evidence showing the involvement of myelin in cognition and neurodevelopmental disorders characterized by cognitive impairments, the link between myelin, brain oscillations, cognition and disease is not yet fully understood. In this review, we aim to highlight what is known and what remains to be explored to understand the role of myelin in high order brain processes.
Collapse
Affiliation(s)
- Hasni Khelfaoui
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France
| | - Cristobal Ibaceta-Gonzalez
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France
| | - Maria Cecilia Angulo
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014, Paris, France.
- GHU-PARIS Psychiatrie Et Neurosciences, Hôpital Sainte Anne, 75014, Paris, France.
| |
Collapse
|
64
|
Cheng YJ, Wang F, Feng J, Yu B, Wang B, Gao Q, Wang TY, Hu B, Gao X, Chen JF, Chen YJ, Lv SQ, Feng H, Xiao L, Mei F. Prolonged myelin deficits contribute to neuron loss and functional impairments after ischaemic stroke. Brain 2024; 147:1294-1311. [PMID: 38289861 DOI: 10.1093/brain/awae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 12/29/2023] [Accepted: 01/13/2024] [Indexed: 02/01/2024] Open
Abstract
Ischaemic stroke causes neuron loss and long-term functional deficits. Unfortunately, effective approaches to preserving neurons and promoting functional recovery remain unavailable. Oligodendrocytes, the myelinating cells in the CNS, are susceptible to oxygen and nutrition deprivation and undergo degeneration after ischaemic stroke. Technically, new oligodendrocytes and myelin can be generated by the differentiation of oligodendrocyte precursor cells (OPCs). However, myelin dynamics and their functional significance after ischaemic stroke remain poorly understood. Here, we report numerous denuded axons accompanied by decreased neuron density in sections from ischaemic stroke lesions in human brain, suggesting that neuron loss correlates with myelin deficits in these lesions. To investigate the longitudinal changes in myelin dynamics after stroke, we labelled and traced pre-existing and newly-formed myelin, respectively, using cell-specific genetic approaches. Our results indicated massive oligodendrocyte death and myelin loss 2 weeks after stroke in the transient middle cerebral artery occlusion (tMCAO) mouse model. In contrast, myelin regeneration remained insufficient 4 and 8 weeks post-stroke. Notably, neuronal loss and functional impairments worsened in aged brains, and new myelin generation was diminished. To analyse the causal relationship between remyelination and neuron survival, we manipulated myelinogenesis by conditional deletion of Olig2 (a positive regulator) or muscarinic receptor 1 (M1R, a negative regulator) in OPCs. Deleting Olig2 inhibited remyelination, reducing neuron survival and functional recovery after tMCAO. Conversely, enhancing remyelination by M1R conditional knockout or treatment with the pro-myelination drug clemastine after tMCAO preserved white matter integrity and neuronal survival, accelerating functional recovery. Together, our findings demonstrate that enhancing myelinogenesis is a promising strategy to preserve neurons and promote functional recovery after ischaemic stroke.
Collapse
Affiliation(s)
- Yong-Jie Cheng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, 1st affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fei Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jie Feng
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Yu
- Department of Neurosurgery, 2nd affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Wang
- Department of Physiology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Qing Gao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Mathematical Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Teng-Yue Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Mathematical Sciences, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Bo Hu
- Department of Physiology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Xing Gao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing-Fei Chen
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu-Jie Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, 1st affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, 2nd affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, 1st affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Xiao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Department of Neurosurgery, 2nd affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Feng Mei
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
65
|
Huang Z, Jordan JD, Zhang Q. Myelin Pathology in Alzheimer's Disease: Potential Therapeutic Opportunities. Aging Dis 2024; 15:698-713. [PMID: 37548935 PMCID: PMC10917545 DOI: 10.14336/ad.2023.0628] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by memory loss and cognitive decline. Despite significant efforts over several decades, our understanding of the pathophysiology of this disease is still incomplete. Myelin is a multi-layered membrane structure ensheathing neuronal axons, which is essential for the fast and effective propagation of action potentials along the axons. Recent studies highlight the critical involvement of myelin in memory consolidation and reveal its vulnerability in various pathological conditions. Notably, apart from the classic amyloid hypothesis, myelin degeneration has been proposed as another critical pathophysiological feature of AD, which could occur prior to the development of amyloid pathology. Here, we review recent works supporting the critical role of myelin in cognition and myelin pathology during AD progression, with a focus on the mechanisms underlying myelin degeneration in AD. We also discuss the complex intersections between myelin pathology and typical AD pathophysiology, as well as the therapeutic potential of pro-myelinating approaches for this disease. Overall, these findings implicate myelin degeneration as a critical contributor to AD-related cognitive deficits and support targeting myelin repair as a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| | - J. Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| |
Collapse
|
66
|
Mao X, Han D, Guo W, Zhang W, Wang H, Zhang G, Zhang N, Jin L, Nie B, Li H, Song Y, Wu Y, Chang L. Lateralized brunt of sleep deprivation on white matter injury in a rat model of Alzheimer's disease. GeroScience 2024; 46:2295-2315. [PMID: 37940789 PMCID: PMC10828179 DOI: 10.1007/s11357-023-01000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023] Open
Abstract
Sleep disturbance is a recognized risk factor for Alzheimer's disease (AD), but the underlying micro-pathological evidence remains limited. To bridge this gap, we established an amyloid-β oligomers (AβO)-induced rat model of AD and subjected it to intermittent sleep deprivation (SD). Diffusion tensor imaging (DTI) and transmission electron microscopy were employed to assess white matter (WM) integrity and ultrastructural changes in myelin sheaths. Our findings demonstrated that SD exacerbated AβO-induced cognitive decline. Furthermore, we found SD aggravated AβO-induced asymmetrical impairments in WM, presenting with reductions in tract integrity observed in commissural fibers and association fasciculi, particularly the right anterior commissure, right corpus callosum, and left cingulum. Ultrastructural changes in myelin sheaths within the hippocampus and corpus callosum further confirmed a lateralized effect. Moreover, SD worsened AβO-induced lateralized disruption of the brain structural network, with impairments in critical nodes of the left hemisphere strongly correlated with cognitive dysfunction. This work represents the first identification of a lateralized impact of SD on the mesoscopic network and cognitive deficits in an AD rat model. These findings could deepen our understanding of the complex interplay between sleep disturbance and AD pathology, providing valuable insights into the early progression of the disease, as well as the development of neuroimaging biomarkers for screening early AD patients with self-reported sleep disturbances. Enhanced understanding of these mechanisms may pave the way for targeted interventions to alleviate cognitive decline and improve the quality of life for individuals at risk of or affected by AD.
Collapse
Affiliation(s)
- Xin Mao
- Department of Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ding Han
- Department of Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wensheng Guo
- Department of Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wanning Zhang
- Department of Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hongqi Wang
- Department of Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Guitao Zhang
- Department of Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ning Zhang
- Department of Neuropsychiatry and Behavioral Neurology and Clinical Psychology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liangyun Jin
- Electron Microscope Room of Central Laboratory, Capital Medical University, Beijing, 100069, China
| | - Binbin Nie
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Hui Li
- Department of Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yizhi Song
- Department of Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan Wu
- Department of Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Lirong Chang
- Department of Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
67
|
Suthard RL, Senne RA, Buzharsky MD, Diep AH, Pyo AY, Ramirez S. Engram reactivation mimics cellular signatures of fear. Cell Rep 2024; 43:113850. [PMID: 38401120 DOI: 10.1016/j.celrep.2024.113850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 02/26/2024] Open
Abstract
Engrams, or the physical substrate of memory, recruit heterogeneous cell types. Targeted reactivation of neurons processing discrete memories drives the behavioral expression of memory, though the underlying landscape of recruited cells and their real-time responses remain elusive. To understand how artificial stimulation of fear affects intra-hippocampal neuron-astrocyte dynamics as well as their behavioral consequences, we express channelrhodopsin-2 in an activity-dependent manner within dentate gyrus neurons while recording both cell types with fiber photometry in hippocampal ventral CA1 across learning and memory. Both cell types exhibit shock responsiveness, with astrocytic calcium events uniquely modulated by fear conditioning. Optogenetic stimulation of a hippocampus-mediated engram recapitulates coordinated calcium signatures time locked to freezing, mirroring those observed during natural fear memory recall. Our findings reveal cell-type-specific dynamics in the hippocampus during freezing behavior, emphasizing neuronal-astrocytic coupling as a shared mechanism enabling both natural and artificially induced memory retrieval and the behavioral expression of fear.
Collapse
Affiliation(s)
- Rebecca L Suthard
- Graduate Program for Neuroscience, Boston University, Boston, MA 02215, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Ryan A Senne
- Graduate Program for Neuroscience, Boston University, Boston, MA 02215, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Michelle D Buzharsky
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Anh H Diep
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Angela Y Pyo
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Steve Ramirez
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
68
|
Hill RA, Nishiyama A, Hughes EG. Features, Fates, and Functions of Oligodendrocyte Precursor Cells. Cold Spring Harb Perspect Biol 2024; 16:a041425. [PMID: 38052500 PMCID: PMC10910408 DOI: 10.1101/cshperspect.a041425] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) are a central nervous system resident population of glia with a distinct molecular identity and an ever-increasing list of functions. OPCs generate oligodendrocytes throughout development and across the life span in most regions of the brain and spinal cord. This process involves a complex coordination of molecular checkpoints and biophysical cues from the environment that initiate the differentiation and integration of new oligodendrocytes that synthesize myelin sheaths on axons. Outside of their progenitor role, OPCs have been proposed to play other functions including the modulation of axonal and synaptic development and the participation in bidirectional signaling with neurons and other glia. Here, we review OPC identity and known functions and discuss recent findings implying other roles for these glial cells in brain physiology and pathology.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Ethan G Hughes
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
69
|
Naffaa MM. Significance of the anterior cingulate cortex in neurogenesis plasticity: Connections, functions, and disorders across postnatal and adult stages. Bioessays 2024; 46:e2300160. [PMID: 38135889 DOI: 10.1002/bies.202300160] [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: 08/25/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
The anterior cingulate cortex (ACC) is a complex and continually evolving brain region that remains a primary focus of research due to its multifaceted functions. Various studies and analyses have significantly advanced our understanding of how the ACC participates in a wide spectrum of memory and cognitive processes. However, despite its strong connections to brain areas associated with hippocampal and olfactory neurogenesis, the functions of the ACC in regulating postnatal and adult neurogenesis in these regions are still insufficiently explored. Investigating the intricate involvement of the ACC in neurogenesis could enhance our comprehension of essential aspects of brain plasticity. This involvement stems from its complex circuitry with other relevant brain regions, thereby exerting both direct and indirect impacts on the neurogenesis process. This review sheds light on the promising significance of the ACC in orchestrating postnatal and adult neurogenesis in conditions related to memory, cognitive behavior, and associated disorders.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
70
|
Xu X, Song X, Chen F, Yan W, Meng Q, Liu J, Yao R, Liu Y, Dong F. Solifenacin promotes remyelination in cuprizone mouse model by inhibiting the Wnt/β-catenin signaling pathway. J Chem Neuroanat 2024; 136:102375. [PMID: 38123002 DOI: 10.1016/j.jchemneu.2023.102375] [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: 09/23/2023] [Revised: 11/22/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023]
Abstract
Demyelinating diseases are a type of neurological disorder characterized by the damage to the myelin sheath in the central nervous system. Promoting the proliferation and differentiation of oligodendrocyte precursor cells (OPCs) is crucial for treatment. Non-selective muscarinic receptor (MR) antagonists have been shown to improve remyelination in rodent models, although the mechanisms are still unclear. In this study, we treated cuprizone (CPZ)-induced demyelination mouse model with different concentrations of Solifenacin (Sol), a selective M3 receptor antagonist, to determine the optimal concentration for promoting remyelination. Behavioral tests and Luxol fast blue (LFB) staining were used to observe the extent of remyelination, while immunofluorescence was used to measure the expression levels of myelin-related proteins, including myelin basic protein (MBP) and platelet-derived growth factor receptor alpha (PDGFR-α). Western blot analysis was employed to analyze the expression levels of molecules associated with the Wnt/β-catenin signaling pathway. The results showed that Sol treatment significantly promoted myelin regeneration and OPCs differentiation in CPZ-induced demyelination mouse model. Additionally, Sol treatment inhibited the Wnt/β-catenin signaling pathway and reversed the effects of CPZ on OPCs differentiation. In conclusion, Sol may promote the differentiation of OPCs by inhibiting the Wnt/β-catenin signaling pathway, making it a potential therapeutic option for central nervous system demyelinating diseases.
Collapse
Affiliation(s)
- Xinqi Xu
- The First Clinical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Xueli Song
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Fei Chen
- The First Clinical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Weixing Yan
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Qiqi Meng
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Jinfeng Liu
- School of Life Science, Xuzhou Medical University, Xuzhou, 221004, Jiangsu Province, China
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Yaping Liu
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, 221004, Jiangsu Province, China
| | - Fuxing Dong
- Public Experimental Research Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China.
| |
Collapse
|
71
|
Huang B, Chen Z, Huang F, Gao F, Chen J, Liu P, Lu Z, Chen W, Wu J. Demyelination in the medial prefrontal cortex by withdrawal from chronic nicotine causes impaired cognitive memory. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110901. [PMID: 38036034 DOI: 10.1016/j.pnpbp.2023.110901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Epidemiological studies revealed deficits in cognitive learning and memory in smokers who withdrawal from smoking, but the molecular mechanisms underlying it is unclear. Here, we employed the novel object recognition task (NORT) to evaluate cognitive memory and found impaired memory and motor skills after withdrawal from chronic nicotine. Myelin sheath hastens the conduction of signals along axons and thus plays a critical role in learning and memory. We found no effect of nicotine withdrawal on the myelination in both of the Ventral tegmental area (VTA) and Nucleus accumbens (NAc) regions, but unexpectedly, we observed a demyelination phenomenon in the medial prefrontal cortex (mPFC) after withdrawal from chronic nicotine. Moreover, we found a positive correlation between the impaired memory and demyelination, and pharmaceutical rescue of myelination by clemastine specifically improved the impaired recognition memory but not the decreased motor skills caused by withdrawal from chronic nicotine. We further found nicotine directly acts on oligodendrocytes with OPCs potential to decrease their myelination process. Taken together, these results demonstrate demyelination in the mPFC causes impaired recognition memory and reveal a potential of enhancing myelination as a therapeutic strategy to alleviate cognitive memory deficits caused by smoking withdrawal.
Collapse
Affiliation(s)
- Bing Huang
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Guangdong, China; Brain Function and Disease Laboratory, Shantou University Medical College, Guangdong, China; Department of Pharmacology, Shantou University Medical College, 22 Xinling Road, Shantou 515041, Guangdong Province, China.
| | - Zifei Chen
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Guangdong, China
| | - Fang Huang
- Joint Shantou International Eye Center, Shantou University and the Chinese University of Hong Kong, 515041 Shantou, Guangdong, China
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, 22 Xinling Road, Shantou 515041, Guangdong Province, China
| | - Jieling Chen
- Brain Function and Disease Laboratory, Shantou University Medical College, Guangdong, China
| | - Peipei Liu
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhijie Lu
- Brain Function and Disease Laboratory, Shantou University Medical College, Guangdong, China
| | - Weiyuan Chen
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Guangdong, China
| | - Jie Wu
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Guangdong, China; Brain Function and Disease Laboratory, Shantou University Medical College, Guangdong, China
| |
Collapse
|
72
|
Morrissey ZD, Gao J, Shetti A, Li W, Zhan L, Li W, Fortel I, Saido T, Saito T, Ajilore O, Cologna SM, Lazarov O, Leow AD. Temporal Alterations in White Matter in An App Knock-In Mouse Model of Alzheimer's Disease. eNeuro 2024; 11:ENEURO.0496-23.2024. [PMID: 38290851 PMCID: PMC10897532 DOI: 10.1523/eneuro.0496-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/05/2024] [Accepted: 01/17/2024] [Indexed: 02/01/2024] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and results in neurodegeneration and cognitive impairment. White matter (WM) is affected in AD and has implications for neural circuitry and cognitive function. The trajectory of these changes across age, however, is still not well understood, especially at earlier stages in life. To address this, we used the AppNL-G-F/NL-G-F knock-in (APPKI) mouse model that harbors a single copy knock-in of the human amyloid precursor protein (APP) gene with three familial AD mutations. We performed in vivo diffusion tensor imaging (DTI) to study how the structural properties of the brain change across age in the context of AD. In late age APPKI mice, we observed reduced fractional anisotropy (FA), a proxy of WM integrity, in multiple brain regions, including the hippocampus, anterior commissure (AC), neocortex, and hypothalamus. At the cellular level, we observed greater numbers of oligodendrocytes in middle age (prior to observations in DTI) in both the AC, a major interhemispheric WM tract, and the hippocampus, which is involved in memory and heavily affected in AD, prior to observations in DTI. Proteomics analysis of the hippocampus also revealed altered expression of oligodendrocyte-related proteins with age and in APPKI mice. Together, these results help to improve our understanding of the development of AD pathology with age, and imply that middle age may be an important temporal window for potential therapeutic intervention.
Collapse
Affiliation(s)
- Zachery D Morrissey
- Graduate Program in Neuroscience, University of Illinois Chicago, Chicago, Illinois 60612
- Department of Psychiatry, University of Illinois Chicago, Chicago, Illinois 60612
- Department of Anatomy & Cell Biology, University of Illinois Chicago, Chicago, Illinois 60612
| | - Jin Gao
- Department of Electrical & Computer Engineering, University of Illinois Chicago, Chicago, Illinois 60607
- Preclinical Imaging Core, University of Illinois Chicago, Chicago, Illinois 60612
| | - Aashutosh Shetti
- Department of Anatomy & Cell Biology, University of Illinois Chicago, Chicago, Illinois 60612
| | - Wenping Li
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607
| | - Liang Zhan
- Department of Electrical & Computer Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Weiguo Li
- Preclinical Imaging Core, University of Illinois Chicago, Chicago, Illinois 60612
- Department of Bioengineering, University of Illinois Chicago, Chicago, Illinois 60607
- Department of Radiology, Northwestern University, Chicago, Illinois 60611
| | - Igor Fortel
- Department of Bioengineering, University of Illinois Chicago, Chicago, Illinois 60607
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako 351-0198, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Nagoya 467-8601, Japan
| | - Olusola Ajilore
- Department of Psychiatry, University of Illinois Chicago, Chicago, Illinois 60612
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607
| | - Orly Lazarov
- Department of Anatomy & Cell Biology, University of Illinois Chicago, Chicago, Illinois 60612
| | - Alex D Leow
- Department of Psychiatry, University of Illinois Chicago, Chicago, Illinois 60612
- Department of Bioengineering, University of Illinois Chicago, Chicago, Illinois 60607
- Department of Computer Science, University of Illinois Chicago, Chicago, Illinois 60607
| |
Collapse
|
73
|
Dong H, Dai X, Zhou Y, Shi C, Bhuiyan P, Sun Z, Li N, Jin W. Enhanced meningeal lymphatic drainage ameliorates lipopolysaccharide-induced brain injury in aged mice. J Neuroinflammation 2024; 21:36. [PMID: 38287311 PMCID: PMC10826026 DOI: 10.1186/s12974-024-03028-4] [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: 09/16/2023] [Accepted: 01/22/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is an acute cerebral dysfunction caused by sepsis. Neuroinflammation induced by sepsis is considered a potential mechanism of SAE; however, very little is known about the role of the meningeal lymphatic system in SAE. METHODS Sepsis was established in male C57BL/6J mice by intraperitoneal injection of 5 mg/kg lipopolysaccharide, and the function of meningeal lymphatic drainage was assessed. Adeno-associated virus 1-vascular endothelial growth factor C (AAV1-VEGF-C) was injected into the cisterna magna to induce meningeal lymphangiogenesis. Ligation of deep cervical lymph nodes (dCLNs) was performed to induce pre-existing meningeal lymphatic dysfunction. Cognitive function was evaluated by a fear conditioning test, and inflammatory factors were detected by enzyme-linked immunosorbent assay. RESULTS The aged mice with SAE showed a significant decrease in the drainage of OVA-647 into the dCLNs and the coverage of the Lyve-1 in the meningeal lymphatic, indicating that sepsis impaired meningeal lymphatic drainage and morphology. The meningeal lymphatic function of aged mice was more vulnerable to sepsis in comparison to young mice. Sepsis also decreased the protein levels of caspase-3 and PSD95, which was accompanied by reductions in the activity of hippocampal neurons. Microglia were significantly activated in the hippocampus of SAE mice, which was accompanied by an increase in neuroinflammation, as indicated by increases in interleukin-1 beta, interleukin-6 and Iba1 expression. Cognitive function was impaired in aged mice with SAE. However, the injection of AAV1-VEGF-C significantly increased coverage in the lymphatic system and tracer dye uptake in dCLNs, suggesting that AAV1-VEGF-C promotes meningeal lymphangiogenesis and drainage. Furthermore, AAV1-VEGF-C reduced microglial activation and neuroinflammation and improved cognitive dysfunction. Improvement of meningeal lymphatics also reduced sepsis-induced expression of disease-associated genes in aged mice. Pre-existing lymphatic dysfunction by ligating bilateral dCLNs aggravated sepsis-induced neuroinflammation and cognitive impairment. CONCLUSION The meningeal lymphatic drainage is damaged in sepsis, and pre-existing defects in this drainage system exacerbate SAE-induced neuroinflammation and cognitive dysfunction. Promoting meningeal lymphatic drainage improves SAE. Manipulation of meningeal lymphangiogenesis could be a new strategy for the treatment of SAE.
Collapse
Affiliation(s)
- Hongquan Dong
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaonan Dai
- Department of Obstetrics, Nanjing Women and Children's Healthcare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Yin Zhou
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chonglong Shi
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Piplu Bhuiyan
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhaochu Sun
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Nana Li
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wenjie Jin
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
74
|
Hernandez DP, Cruz DM, Martinez CS, Garcia LM, Figueroa A, Villarreal M, Manoj LM, Lopez S, López-Lorenzo KD, López-Juárez A. Gender-Specific Fine Motor Skill Learning Is Impaired by Myelin-Targeted Neurofibromatosis Type 1 Gene Mutation. Cancers (Basel) 2024; 16:477. [PMID: 38339230 PMCID: PMC10854893 DOI: 10.3390/cancers16030477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) is caused by mutations in the NF1 gene. The clinical presentation of NF1 includes diverse neurological issues in pediatric and adult patients, ranging from learning disabilities, motor skill issues, and attention deficit disorder, to increased risk of depression and dementia. Preclinical research suggests that abnormal neuronal signaling mediates spatial learning and attention issues in NF1; however, drugs that improve phenotypes in models show inconclusive results in clinical trials, highlighting the need for a better understanding of NF1 pathophysiology and broader therapeutic options. Most NF1 patients show abnormalities in their brain white matter (WM) and myelin, and links with NF1 neuropathophysiology have been suggested; however, no current data can clearly support or refute this idea. We reported that myelin-targeted Nf1 mutation impacts oligodendrocyte signaling, myelin ultrastructure, WM connectivity, and sensory-motor behaviors in mice; however, any impact on learning and memory remains unknown. Here, we adapted a voluntary running test-the complex wheel (CW; a wheel with unevenly spaced rungs)-to delineate fine motor skill learning curves following induction of an Nf1 mutation in pre-existing myelinating cells (pNf1 mice). We found that pNf1 mutant females experience delayed or impaired learning in the CW, while proper learning in pNf1 males is predominantly disrupted; these phenotypes add complexity to the gender-dependent learning differences in the mouse strain used. No broad differences in memory of acquired CW skills were detected in any gender, but gene-dose effects were observed at the studied time points. Finally, nitric oxide signaling regulation differentially impacted learning in wild type (WT)/pNf1, male/female mice. Our results provide evidence for fine motor skill learning issues upon induction of an Nf1 mutation in mature myelinating cells. Together with previous connectivity, cellular, and molecular analyses, these results diversify the potential treatments for neurological issues in NF1.
Collapse
Affiliation(s)
- Daniella P. Hernandez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Daniela M. Cruz
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Celeste S. Martinez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Larisa M. Garcia
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Ashley Figueroa
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Marisol Villarreal
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Liya M. Manoj
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Saul Lopez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | | | - Alejandro López-Juárez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| |
Collapse
|
75
|
Kent SA, Miron VE. Microglia regulation of central nervous system myelin health and regeneration. Nat Rev Immunol 2024; 24:49-63. [PMID: 37452201 DOI: 10.1038/s41577-023-00907-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Microglia are resident macrophages of the central nervous system that have key functions in its development, homeostasis and response to damage and infection. Although microglia have been increasingly implicated in contributing to the pathology that underpins neurological dysfunction and disease, they also have crucial roles in neurological homeostasis and regeneration. This includes regulation of the maintenance and regeneration of myelin, the membrane that surrounds neuronal axons, which is required for axonal health and function. Myelin is damaged with normal ageing and in several neurodegenerative diseases, such as multiple sclerosis and Alzheimer disease. Given the lack of approved therapies targeting myelin maintenance or regeneration, it is imperative to understand the mechanisms by which microglia support and restore myelin health to identify potential therapeutic approaches. However, the mechanisms by which microglia regulate myelin loss or integrity are still being uncovered. In this Review, we discuss recent work that reveals the changes in white matter with ageing and neurodegenerative disease, how this relates to microglia dynamics during myelin damage and regeneration, and factors that influence the regenerative functions of microglia.
Collapse
Affiliation(s)
- Sarah A Kent
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK.
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK.
- Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada.
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, Ontario, Canada.
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
76
|
Zhu TT, Wang H, Liu PM, Gu HW, Pan WT, Zhao MM, Hashimoto K, Yang JJ. Clemastine-induced enhancement of hippocampal myelination alleviates memory impairment in mice with chronic pain. Neurobiol Dis 2024; 190:106375. [PMID: 38092269 DOI: 10.1016/j.nbd.2023.106375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023] Open
Abstract
Patients with chronic pain often experience memory impairment, but the underlying mechanisms remain elusive. The myelin sheath is crucial for rapid and accurate action potential conduction, playing a pivotal role in the development of cognitive abilities in the central nervous system. The study reveals that myelin degradation occurs in the hippocampus of chronic constriction injury (CCI) mice, which display both chronic pain and memory impairment. Using fiber photometry, we observed diminished task-related neuronal activity in the hippocampus of CCI mice. Interestingly, the repeated administration with clemastine, which promotes myelination, counteracts the CCI-induced myelin loss and reduced neuronal activity. Notably, clemastine specifically ameliorates the impaired memory without affecting chronic pain in CCI mice. Overall, our findings highlight the significant role of myelin abnormalities in CCI-induced memory impairment, suggesting a potential therapeutic approach for treating memory impairments associated with neuropathic pain.
Collapse
Affiliation(s)
- Ting-Ting Zhu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Neuroscience Research Institute, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - He Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Pan-Miao Liu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Neuroscience Research Institute, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Han-Wen Gu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wei-Tong Pan
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Neuroscience Research Institute, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Ming-Ming Zhao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Kenji Hashimoto
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Neuroscience Research Institute, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
77
|
Zuo Y, Hou Y, Wang Y, Yuan L, Cheng L, Zhang T. Circadian misalignment impairs oligodendrocyte myelination via Bmal1 overexpression leading to anxiety and depression-like behaviors. J Pineal Res 2024; 76:e12935. [PMID: 38241675 DOI: 10.1111/jpi.12935] [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: 08/31/2023] [Revised: 12/14/2023] [Accepted: 12/26/2023] [Indexed: 01/21/2024]
Abstract
Circadian misalignment (CM) caused by shift work can increase the risk of mood impairment. However, the pathological mechanisms underlying these deficits remain unclear. In the present study, we used long-term variable photoperiod (L-VP) in wild-type mice to better simulate real-life shift patterns and study its effects on the prefrontal cortex (PFC) and hippocampus, which are closely related to mood function. The results showed that exposure to L-VP altered the activity/rest rhythms of mice, by eliciting phase delay and decreased amplitude of the rhythms. Mice with CM developed anxiety and depression-like manifestations and the number of mature oligodendrocytes (OL) was reduced in the medial prefrontal cortex and hippocampal CA1 regions. Mood impairment and OL reduction worsened with increased exposure time to L-VP, while normal photoperiod restoration had no effect. Mechanistically, we identified upregulation of Bmal1 in the PFC and hippocampal regions of CM mice at night, when genes related to mature OL and myelination should be highly expressed. CM mice exhibited significant inhibition of the protein kinase B (AKT)/mTOR signaling pathway, which is directly associated to OL differentiation and maturation. Furthermore, we demonstrated in the OL precursor cell line Oli-Neu that overexpression of Bmal1 inhibits AKT/mTOR pathway and reduces the expression of genes OL differentiation. In conclusion, BMAL1 might play a critical role in CM, providing strong research evidence for BMAL1 as a potential target for CM therapy.
Collapse
Affiliation(s)
- Yao Zuo
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Neurological Rehabilitation, China Rehabilitation Research Center, Beijing Boai Hospital, Beijing, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Yuanyuan Hou
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yunlei Wang
- Department of Neurological Rehabilitation, China Rehabilitation Research Center, Beijing Boai Hospital, Beijing, China
| | - Linran Yuan
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Neurological Rehabilitation, China Rehabilitation Research Center, Beijing Boai Hospital, Beijing, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Lingna Cheng
- Department of Neurological Rehabilitation, China Rehabilitation Research Center, Beijing Boai Hospital, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Tong Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Neurological Rehabilitation, China Rehabilitation Research Center, Beijing Boai Hospital, Beijing, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| |
Collapse
|
78
|
Keil J, Kiiski H, Doherty L, Hernandez-Urbina V, Vassiliou C, Dean C, Müschenich M, Bahmani H. Artificial sharp-wave-ripples to support memory and counter neurodegeneration. Brain Res 2024; 1822:148646. [PMID: 37871674 DOI: 10.1016/j.brainres.2023.148646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
Information processed in our sensory neocortical areas is transported to the hippocampus during memory encoding, and between hippocampus and neocortex during memory consolidation, and retrieval. Short bursts of high-frequency oscillations, so called sharp-wave-ripples, have been proposed as a potential mechanism for this information transfer: They can synchronize neural activity to support the formation of local neural networks to store information, and between distant cortical sites to act as a bridge to transfer information between sensory cortical areas and hippocampus. In neurodegenerative diseases like Alzheimer's Disease, different neuropathological processes impair normal neural functioning and neural synchronization as well as sharp-wave-ripples, which impairs consolidation and retrieval of information, and compromises memory. Here, we formulate a new hypothesis, that artificially inducing sharp-wave-ripples with noninvasive high-frequency visual stimulation could potentially support memory functioning, as well as target the neuropathological processes underlying neurodegenerative diseases. We also outline key challenges for empirical tests of the hypothesis.
Collapse
Affiliation(s)
- Julian Keil
- Department of Psychology, Christian-Albrechts-University Kiel, Germany; Ababax Health GmbH, Berlin, Germany; Department of Cognitive Science, University of Potsdam, Germany.
| | - Hanni Kiiski
- Ababax Health GmbH, Berlin, Germany; Department of Cognitive Science, University of Potsdam, Germany
| | | | | | - Chrystalleni Vassiliou
- German Center for Neurodegenerative Diseases, Charité University of Medicine, Berlin, Germany
| | - Camin Dean
- German Center for Neurodegenerative Diseases, Charité University of Medicine, Berlin, Germany
| | | | - Hamed Bahmani
- Ababax Health GmbH, Berlin, Germany; Bernstein Center for Computational Neuroscience, Tuebingen, Germany
| |
Collapse
|
79
|
Sajad M, Zahoor I, Rashid F, Cerghet M, Rattan R, Giri S. Pyruvate Dehydrogenase-Dependent Metabolic Programming Affects the Oligodendrocyte Maturation and Remyelination. Mol Neurobiol 2024; 61:397-410. [PMID: 37620688 PMCID: PMC11984507 DOI: 10.1007/s12035-023-03546-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 07/21/2023] [Indexed: 08/26/2023]
Abstract
The metabolic needs of the premature/premyelinating oligodendrocytes (pre-OLs) and mature oligodendrocytes (OLs) are distinct. The metabolic control of oligodendrocyte maturation from the pre-OLs to the OLs is not fully understood. Here, we show that the terminal maturation and higher mitochondrial respiration in the OLs is an integrated process controlled through pyruvate dehydrogenase complex (Pdh). Combined bioenergetics and metabolic studies show that OLs show elevated mitochondrial respiration than the pre-OLs. Our signaling studies show that the increased mitochondrial respiration activity in the OLs is mediated by the activation of Pdh due to inhibition of the pyruvate dehydrogenase kinase-1 (Pdhk1) that phosphorylates and inhibits Pdh activity. Accordingly, when Pdhk1 is directly expressed in the pre-OLs, they fail to mature into the OLs. While Pdh converts pyruvate into the acetyl-CoA by its oxidative decarboxylation, our study shows that Pdh-dependent acetyl-CoA generation from pyruvate contributes to the acetylation of the bHLH family transcription factor, oligodendrocyte transcription factor 1 (Olig1) which is known to be involved in the OL maturation. Pdh inhibition via direct expression of Pdhk1 in the pre-OLs blocks the Olig1-acetylation and OL maturation. Using the cuprizone model of demyelination, we show that Pdh is deactivated during the demyelination phase, which is however reversed in the remyelination phase upon cuprizone withdrawal. In addition, Pdh activity status correlates with the Olig1-acetylation status in the cuprizone model. Hence, the Pdh metabolic node activation allows a robust mitochondrial respiration and activation of a molecular program necessary for the terminal maturation of oligodendrocytes. Our findings open a new dialogue in the developmental biology that links cellular development and metabolism. These findings have far-reaching implications in the development of therapies for a variety of demyelinating disorders including multiple sclerosis.
Collapse
Affiliation(s)
- M Sajad
- Department of Neurology, Henry Ford Health, Detroit, MI, 48202, USA.
| | - Insha Zahoor
- Department of Neurology, Henry Ford Health, Detroit, MI, 48202, USA
| | - Faraz Rashid
- Department of Neurology, Henry Ford Health, Detroit, MI, 48202, USA
| | - Mirela Cerghet
- Department of Neurology, Henry Ford Health, Detroit, MI, 48202, USA
| | - Ramandeep Rattan
- Gynecologic Oncology and Developmental Therapeutics Research Program, Henry Ford Health Hospital, Detroit, MI, 48202, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health, Detroit, MI, 48202, USA.
| |
Collapse
|
80
|
Kambe J, Usuda K, Inoue R, Hirayama K, Ito M, Suenaga K, Masukado S, Liu H, Miyata S, Li C, Kimura I, Yamamoto Y, Nagaoka K. Hydrogen peroxide in breast milk is crucial for gut microbiota formation and myelin development in neonatal mice. Gut Microbes 2024; 16:2359729. [PMID: 38816999 PMCID: PMC11146441 DOI: 10.1080/19490976.2024.2359729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Early life environment influences mammalian brain development, a growing area of research within the Developmental Origins of Health and Disease framework, necessitating a deeper understanding of early life factors on children's brain development. This study introduces a mouse model, LAO1 knockout mice, to investigate the relationship between breast milk, the gut microbiome, and brain development. The results reveal that breast milk's reactive oxygen species (ROS) are vital in shaping the neonatal gut microbiota. Decreased hydrogen peroxide (H2O2) levels in milk disrupt the gut microbiome and lead to abnormal metabolite production, including D-glucaric acid. This metabolite inhibits hippocampal myelin formation during infancy, potentially contributing to behavioral abnormalities observed in adulthood. These findings suggest that H2O2 in breast milk is crucial for normal gut microbiota formation and brain development, with implications for understanding and potentially treating neurodevelopmental disorders in humans.
Collapse
Affiliation(s)
- Jun Kambe
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kento Usuda
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, Osaka, Japan
| | - Kazuhiko Hirayama
- Laboratory of Veterinary Public Health, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Masahiko Ito
- Department of Virology and Parasitology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Ken Suenaga
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Sora Masukado
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hong Liu
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Shiho Miyata
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Chunmei Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ikuo Kimura
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yuki Yamamoto
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
81
|
Cho CH, Deyneko IV, Cordova-Martinez D, Vazquez J, Maguire AS, Diaz JR, Carbonell AU, Tindi JO, Cui MH, Fleysher R, Molholm S, Lipton ML, Branch CA, Hodgson L, Jordan BA. ANKS1B encoded AIDA-1 regulates social behaviors by controlling oligodendrocyte function. Nat Commun 2023; 14:8499. [PMID: 38129387 PMCID: PMC10739966 DOI: 10.1038/s41467-023-43438-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 11/09/2023] [Indexed: 12/23/2023] Open
Abstract
Heterozygous deletions in the ANKS1B gene cause ANKS1B neurodevelopmental syndrome (ANDS), a rare genetic disease characterized by autism spectrum disorder (ASD), attention deficit/hyperactivity disorder, and speech and motor deficits. The ANKS1B gene encodes for AIDA-1, a protein that is enriched at neuronal synapses and regulates synaptic plasticity. Here we report an unexpected role for oligodendroglial deficits in ANDS pathophysiology. We show that Anks1b-deficient mouse models display deficits in oligodendrocyte maturation, myelination, and Rac1 function, and recapitulate white matter abnormalities observed in ANDS patients. Selective loss of Anks1b from the oligodendrocyte lineage, but not from neuronal populations, leads to deficits in social preference and sensory reactivity previously observed in a brain-wide Anks1b haploinsufficiency model. Furthermore, we find that clemastine, an antihistamine shown to increase oligodendrocyte precursor cell maturation and central nervous system myelination, rescues deficits in social preference in 7-month-old Anks1b-deficient mice. Our work shows that deficits in social behaviors present in ANDS may originate from abnormal Rac1 activity within oligodendrocytes.
Collapse
Affiliation(s)
- Chang Hoon Cho
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., San Francisco, CA, USA
| | - Ilana Vasilisa Deyneko
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dylann Cordova-Martinez
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Juan Vazquez
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anne S Maguire
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jenny R Diaz
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abigail U Carbonell
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jaafar O Tindi
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Min-Hui Cui
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Roman Fleysher
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sophie Molholm
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael L Lipton
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Craig A Branch
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Louis Hodgson
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bryen A Jordan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
82
|
Brousse B, Mercier O, Magalon K, Gubellini P, Malapert P, Cayre M, Durbec P. Characterization of a new mouse line triggering transient oligodendrocyte progenitor depletion. Sci Rep 2023; 13:21959. [PMID: 38081969 PMCID: PMC10713661 DOI: 10.1038/s41598-023-48926-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Oligodendrocyte progenitor cells (OPC) are the main proliferative cells in the healthy adult brain. They produce new myelinating oligodendrocytes to ensure physiological myelin remodeling and regeneration after various pathological insults. Growing evidence suggests that OPC have other functions. Here, we aimed to develop an experimental model that allows the specific ablation of OPC at the adult stage to unravel possible new functions. We generated a transgenic mouse expressing a floxed human diphtheria toxin receptor under the control of the PDGFRa promoter, crossed with an Olig2Cre mouse to limit the recombination to the oligodendrocyte lineage in the central nervous system. We determined a diphtheria toxin dose to substantially decrease OPC density in the cortex and the corpus callosum without triggering side toxicity after a few daily injections. OPC density was normalized 7 days post-treatment, showing high repopulation capacity from few surviving OPC. We took advantage of this strong but transient depletion to show that OPC loss was associated with behavioral impairment, which was restored by OPC recovery, as well as disruption of the excitation/inhibition balance in the sensorimotor cortex, reinforcing the hypothesis of a neuromodulatory role of OPC in the adult brain.
Collapse
Affiliation(s)
- B Brousse
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
| | - O Mercier
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
| | - K Magalon
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
| | - P Gubellini
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
- Aix Marseille Univ, CNRS, LNC UMR7291, 3 Place Victor Hugo, 13331, Marseille Cedex 3, France
| | - P Malapert
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
| | - M Cayre
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France
- Aix Marseille Univ, CNRS, LNC UMR7291, 3 Place Victor Hugo, 13331, Marseille Cedex 3, France
| | - P Durbec
- Aix Marseille Univ, CNRS, IBDM UMR7288, Case 907, Parc Scientifique de Luminy, 13288, Marseille Cedex 09, France.
| |
Collapse
|
83
|
Fiore F, Alhalaseh K, Dereddi RR, Bodaleo Torres F, Çoban I, Harb A, Agarwal A. Norepinephrine regulates calcium signals and fate of oligodendrocyte precursor cells in the mouse cerebral cortex. Nat Commun 2023; 14:8122. [PMID: 38065932 PMCID: PMC10709653 DOI: 10.1038/s41467-023-43920-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Oligodendrocyte precursor cells (OPCs) generate oligodendrocytes, contributing to myelination and myelin repair. OPCs contact axons and respond to neuronal activity, but how the information relayed by the neuronal activity translates into OPC Ca2+ signals, which in turn influence their fate, remains unknown. We generated transgenic mice for concomitant monitoring of OPCs Ca2+ signals and cell fate using 2-photon microscopy in the somatosensory cortex of awake-behaving mice. Ca2+ signals in OPCs mainly occur within processes and confine to Ca2+ microdomains. A subpopulation of OPCs enhances Ca2+ transients while mice engaged in exploratory locomotion. We found that OPCs responsive to locomotion preferentially differentiate into oligodendrocytes, and locomotion-non-responsive OPCs divide. Norepinephrine mediates locomotion-evoked Ca2+ increases in OPCs by activating α1 adrenergic receptors, and chemogenetic activation of OPCs or noradrenergic neurons promotes OPC differentiation. Hence, we uncovered that for fate decisions OPCs integrate Ca2+ signals, and norepinephrine is a potent regulator of OPC fate.
Collapse
Affiliation(s)
- Frederic Fiore
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Khaleel Alhalaseh
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Ram R Dereddi
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Felipe Bodaleo Torres
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Ilknur Çoban
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Ali Harb
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Amit Agarwal
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
84
|
Maren S. It takes a village: Neurons partner with vascular pericytes to make memories. Neuron 2023; 111:3701-3702. [PMID: 38061328 DOI: 10.1016/j.neuron.2023.10.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 12/18/2023]
Abstract
Neurons have a central role in memory formation, but emerging work points to the critical role that non-neuronal cells play in this process. In this issue of Neuron, Pandey and colleagues1 show that hippocampal neurons communicate with vascular pericytes during memory consolidation. Through this dialogue, pericyte-derived growth factors support long-term memory.
Collapse
Affiliation(s)
- Stephen Maren
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX 77843.
| |
Collapse
|
85
|
de Blank P, Nishiyama A, López-Juárez A. A new era for myelin research in Neurofibromatosis type 1. Glia 2023; 71:2701-2719. [PMID: 37382486 PMCID: PMC10592420 DOI: 10.1002/glia.24432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023]
Abstract
Evidence for myelin regulating higher-order brain function and disease is rapidly accumulating; however, defining cellular/molecular mechanisms remains challenging partially due to the dynamic brain physiology involving deep changes during development, aging, and in response to learning and disease. Furthermore, as the etiology of most neurological conditions remains obscure, most research models focus on mimicking symptoms, which limits understanding of their molecular onset and progression. Studying diseases caused by single gene mutations represents an opportunity to understand brain dys/function, including those regulated by myelin. Here, we discuss known and potential repercussions of abnormal central myelin on the neuropathophysiology of Neurofibromatosis Type 1 (NF1). Most patients with this monogenic disease present with neurological symptoms diverse in kind, severity, and onset/decline, including learning disabilities, autism spectrum disorders, attention deficit and hyperactivity disorder, motor coordination issues, and increased risk for depression and dementia. Coincidentally, most NF1 patients show diverse white matter/myelin abnormalities. Although myelin-behavior links were proposed decades ago, no solid data can prove or refute this idea yet. A recent upsurge in myelin biology understanding and research/therapeutic tools provides opportunities to address this debate. As precision medicine moves forward, an integrative understanding of all cell types disrupted in neurological conditions becomes a priority. Hence, this review aims to serve as a bridge between fundamental cellular/molecular myelin biology and clinical research in NF1.
Collapse
Affiliation(s)
- Peter de Blank
- Department of Pediatrics, The Cure Starts Now Brain Tumor Center, University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| | - Alejandro López-Juárez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| |
Collapse
|
86
|
Taylor KR, Monje M. Neuron-oligodendroglial interactions in health and malignant disease. Nat Rev Neurosci 2023; 24:733-746. [PMID: 37857838 PMCID: PMC10859969 DOI: 10.1038/s41583-023-00744-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2023] [Indexed: 10/21/2023]
Abstract
Experience sculpts brain structure and function. Activity-dependent modulation of the myelinated infrastructure of the nervous system has emerged as a dimension of adaptive change during childhood development and in adulthood. Myelination is a richly dynamic process, with neuronal activity regulating oligodendrocyte precursor cell proliferation, oligodendrogenesis and myelin structural changes in some axonal subtypes and in some regions of the nervous system. This myelin plasticity and consequent changes to conduction velocity and circuit dynamics can powerfully influence neurological functions, including learning and memory. Conversely, disruption of the mechanisms mediating adaptive myelination can contribute to cognitive impairment. The robust effects of neuronal activity on normal oligodendroglial precursor cells, a putative cellular origin for many forms of glioma, indicates that dysregulated or 'hijacked' mechanisms of myelin plasticity could similarly promote growth in this devastating group of brain cancers. Indeed, neuronal activity promotes the pathogenesis of many forms of glioma in preclinical models through activity-regulated paracrine factors and direct neuron-to-glioma synapses. This synaptic integration of glioma into neural circuits is central to tumour growth and invasion. Thus, not only do neuron-oligodendroglial interactions modulate neural circuit structure and function in the healthy brain, but neuron-glioma interactions also have important roles in the pathogenesis of glial malignancies.
Collapse
Affiliation(s)
- Kathryn R Taylor
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
87
|
Fan C, An H, Kim D, Park Y. Uncovering oligodendrocyte enhancers that control Cnp expression. Hum Mol Genet 2023; 32:3225-3236. [PMID: 37642363 PMCID: PMC10656706 DOI: 10.1093/hmg/ddad141] [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: 06/01/2023] [Revised: 08/05/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023] Open
Abstract
Oligodendrocytes (OLs) produce myelin sheaths around axons in the central nervous system (CNS). Myelin accelerates the propagation of action potentials along axons and supports the integrity of axons. Impaired myelination has been linked to neurological and neuropsychiatric disorders. As a major component of CNS myelin, 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNP) plays an indispensable role in the axon-supportive function of myelin. Notably, this function requires a high-level expression of CNP in OLs, as evidenced by downregulated expression of CNP in mental disorders and animal models. Little is known about how CNP expression is regulated in OLs. Especially, OL enhancers that govern CNP remain elusive. We have recently developed a powerful method that links OL enhancers to target genes in a principled manner. Here, we applied it to Cnp, uncovering two OL enhancers for it (termed Cnp-E1 and Cnp-E2). Epigenome editing analysis revealed that Cnp-E1 and Cnp-E2 are dedicated to Cnp. ATAC-seq and ChIP-seq data show that Cnp-E1 and Cnp-E2 are conserved OL-specific enhancers. Single cell multi-omics data that jointly profile gene expression and chromatin accessibility suggest that Cnp-E2 plays an important role in Cnp expression in the early stage of OL differentiation while Cnp-E1 sustains it in mature OLs.
Collapse
Affiliation(s)
- Chuandong Fan
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY 14203, United States
| | - Hongjoo An
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY 14203, United States
| | - Dongkyeong Kim
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY 14203, United States
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Yungki Park
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY 14203, United States
| |
Collapse
|
88
|
Zou P, Wu C, Liu TCY, Duan R, Yang L. Oligodendrocyte progenitor cells in Alzheimer's disease: from physiology to pathology. Transl Neurodegener 2023; 12:52. [PMID: 37964328 PMCID: PMC10644503 DOI: 10.1186/s40035-023-00385-7] [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: 08/10/2023] [Accepted: 11/01/2023] [Indexed: 11/16/2023] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) play pivotal roles in myelin formation and phagocytosis, communicating with neighboring cells and contributing to the integrity of the blood-brain barrier (BBB). However, under the pathological circumstances of Alzheimer's disease (AD), the brain's microenvironment undergoes detrimental changes that significantly impact OPCs and their functions. Starting with OPC functions, we delve into the transformation of OPCs to myelin-producing oligodendrocytes, the intricate signaling interactions with other cells in the central nervous system (CNS), and the fascinating process of phagocytosis, which influences the function of OPCs and affects CNS homeostasis. Moreover, we discuss the essential role of OPCs in BBB formation and highlight the critical contribution of OPCs in forming CNS-protective barriers. In the context of AD, the deterioration of the local microenvironment in the brain is discussed, mainly focusing on neuroinflammation, oxidative stress, and the accumulation of toxic proteins. The detrimental changes disturb the delicate balance in the brain, impacting the regenerative capacity of OPCs and compromising myelin integrity. Under pathological conditions, OPCs experience significant alterations in migration and proliferation, leading to impaired differentiation and a reduced ability to produce mature oligodendrocytes. Moreover, myelin degeneration and formation become increasingly active in AD, contributing to progressive neurodegeneration. Finally, we summarize the current therapeutic approaches targeting OPCs in AD. Strategies to revitalize OPC senescence, modulate signaling pathways to enhance OPC differentiation, and explore other potential therapeutic avenues are promising in alleviating the impact of AD on OPCs and CNS function. In conclusion, this review highlights the indispensable role of OPCs in CNS function and their involvement in the pathogenesis of AD. The intricate interplay between OPCs and the AD brain microenvironment underscores the complexity of neurodegenerative diseases. Insights from studying OPCs under pathological conditions provide a foundation for innovative therapeutic strategies targeting OPCs and fostering neurodegeneration. Future research will advance our understanding and management of neurodegenerative diseases, ultimately offering hope for effective treatments and improved quality of life for those affected by AD and related disorders.
Collapse
Affiliation(s)
- Peibin Zou
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Timon Cheng-Yi Liu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Rui Duan
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
89
|
Kato D, Aoyama Y, Nishida K, Takahashi Y, Sakamoto T, Takeda I, Tatematsu T, Go S, Saito Y, Kunishima S, Cheng J, Hou L, Tachibana Y, Sugio S, Kondo R, Eto F, Sato S, Moorhouse AJ, Yao I, Kadomatsu K, Setou M, Wake H. Regulation of lipid synthesis in myelin modulates neural activity and is required for motor learning. Glia 2023; 71:2591-2608. [PMID: 37475643 DOI: 10.1002/glia.24441] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 06/11/2023] [Accepted: 07/03/2023] [Indexed: 07/22/2023]
Abstract
Brain function relies on both rapid electrical communication in neural circuitry and appropriate patterns or synchrony of neural activity. Rapid communication between neurons is facilitated by wrapping nerve axons with insulation by a myelin sheath composed largely of different lipids. Recent evidence has indicated that the extent of myelination of nerve axons can adapt based on neural activity levels and this adaptive myelination is associated with improved learning of motor tasks, suggesting such plasticity may enhance effective learning. In this study, we examined whether another aspect of myelin plasticity-changes in myelin lipid synthesis and composition-may also be associated with motor learning. We combined a motor learning task in mice with in vivo two-photon imaging of neural activity in the primary motor cortex (M1) to distinguish early and late stages of learning and then probed levels of some key myelin lipids using mass spectrometry analysis. Sphingomyelin levels were elevated in the early stage of motor learning while galactosylceramide levels were elevated in the middle and late stages of motor learning, and these changes were correlated across individual mice with both learning performance and neural activity changes. Targeted inhibition of oligodendrocyte-specific galactosyltransferase expression, the enzyme that synthesizes myelin galactosylceramide, impaired motor learning. Our results suggest regulation of myelin lipid composition could be a novel facet of myelin adaptations associated with learning.
Collapse
Affiliation(s)
- Daisuke Kato
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Yuki Aoyama
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuki Nishida
- Division of System Neuroscience, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yutaka Takahashi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takumi Sakamoto
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ikuko Takeda
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Tsuyako Tatematsu
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiori Go
- Institute for Glyco-core Research, Nagoya University, Nagoya, Japan
| | - Yutaro Saito
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiho Kunishima
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jinlei Cheng
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Lingnan Hou
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihisa Tachibana
- Division of System Neuroscience, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shouta Sugio
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Reon Kondo
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumihiro Eto
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, Japan
| | - Shumpei Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Andrew J Moorhouse
- School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ikuko Yao
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, Japan
| | - Kenji Kadomatsu
- Institute for Glyco-core Research, Nagoya University, Nagoya, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Center of Optical Scattering Image Science, Kobe University, Kobe, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies, SOKENDAI, Hayama, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
90
|
Caldwell M, Ayo-Jibunoh V, Mendoza JC, Brimblecombe KR, Reynolds LM, Zhu Jiang XY, Alarcon C, Fiore E, N Tomaio J, Phillips GR, Mingote S, Flores C, Casaccia P, Liu J, Cragg SJ, McCloskey DP, Yetnikoff L. Axo-glial interactions between midbrain dopamine neurons and oligodendrocyte lineage cells in the anterior corpus callosum. Brain Struct Funct 2023; 228:1993-2006. [PMID: 37668732 PMCID: PMC10516790 DOI: 10.1007/s00429-023-02695-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/09/2023] [Indexed: 09/06/2023]
Abstract
Oligodendrocyte progenitor cells (OPCs) receive synaptic innervation from glutamatergic and GABAergic axons and can be dynamically regulated by neural activity, resulting in activity-dependent changes in patterns of axon myelination. However, it remains unclear to what extent other types of neurons may innervate OPCs. Here, we provide evidence implicating midbrain dopamine neurons in the innervation of oligodendrocyte lineage cells in the anterior corpus callosum and nearby white matter tracts of male and female adult mice. Dopaminergic axon terminals were identified in the corpus callosum of DAT-Cre mice after injection of an eYFP reporter virus into the midbrain. Furthermore, fast-scan cyclic voltammetry revealed monoaminergic transients in the anterior corpus callosum, consistent with the anatomical findings. Using RNAscope, we further demonstrate that ~ 40% of Olig2 + /Pdfgra + cells and ~ 20% of Olig2 + /Pdgfra- cells in the anterior corpus callosum express Drd1 and Drd2 transcripts. These results suggest that oligodendrocyte lineage cells may respond to dopamine released from midbrain dopamine axons, which could affect myelination. Together, this work broadens our understanding of neuron-glia interactions with important implications for myelin plasticity by identifying midbrain dopamine axons as a potential regulator of corpus callosal oligodendrocyte lineage cells.
Collapse
Affiliation(s)
- Megan Caldwell
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Vanessa Ayo-Jibunoh
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Josue Criollo Mendoza
- Department of Biology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Katherine R Brimblecombe
- Centre for Integrative Neuroscience, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Lauren M Reynolds
- Plasticité du Cerveau, CNRS UMR8249, École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Paris, France
| | - Xin Yan Zhu Jiang
- Department of Biology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Colin Alarcon
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Elizabeth Fiore
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Jacquelyn N Tomaio
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Greg R Phillips
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Department of Biology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
- Center for Developmental Neuroscience, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Susana Mingote
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Cecilia Flores
- Department of Psychiatry and of Neurology and Neuroscience, McGill University, and Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
- Department of Neuroscience and Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Stephanie J Cragg
- Centre for Integrative Neuroscience, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Dan P McCloskey
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Leora Yetnikoff
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA.
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA.
| |
Collapse
|
91
|
Shimizu T, Nayar SG, Swire M, Jiang Y, Grist M, Kaller M, Sampaio Baptista C, Bannerman DM, Johansen-Berg H, Ogasawara K, Tohyama K, Li H, Richardson WD. Oligodendrocyte dynamics dictate cognitive performance outcomes of working memory training in mice. Nat Commun 2023; 14:6499. [PMID: 37838794 PMCID: PMC10576739 DOI: 10.1038/s41467-023-42293-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 10/04/2023] [Indexed: 10/16/2023] Open
Abstract
Previous work has shown that motor skill learning stimulates and requires generation of myelinating oligodendrocytes (OLs) from their precursor cells (OLPs) in the brains of adult mice. In the present study we ask whether OL production is also required for non-motor learning and cognition, using T-maze and radial-arm-maze tasks that tax spatial working memory. We find that maze training stimulates OLP proliferation and OL production in the medial prefrontal cortex (mPFC), anterior corpus callosum (genu), dorsal thalamus and hippocampal formation of adult male mice; myelin sheath formation is also stimulated in the genu. Genetic blockade of OL differentiation and neo-myelination in Myrf conditional-knockout mice strongly impairs training-induced improvements in maze performance. We find a strong positive correlation between the performance of individual wild type mice and the scale of OLP proliferation and OL generation during training, but not with the number or intensity of c-Fos+ neurons in their mPFC, underscoring the important role played by OL lineage cells in cognitive processing.
Collapse
Affiliation(s)
- Takahiro Shimizu
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| | - Stuart G Nayar
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| | - Matthew Swire
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| | - Yi Jiang
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| | - Matthew Grist
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| | - Malte Kaller
- Wellcome Centre for Integrative Neuroimaging, Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Cassandra Sampaio Baptista
- Wellcome Centre for Integrative Neuroimaging, Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
- Institute of Neuroscience and Psychology, University of Glasgow, 62 Hillhead Street, G12 8QB, Glasgow, UK
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, OX1 3TA, UK
| | - Heidi Johansen-Berg
- Wellcome Centre for Integrative Neuroimaging, Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Katsutoshi Ogasawara
- Technical Support Center for Life Science Research, Iwate Medical University, 1-1-1 Idaidori, Yahabacho, Shiwa-gun, Morioka, Iwate, 028-3694, Japan
| | - Koujiro Tohyama
- Department of Physiology, Iwate Medical University, 1-1-1 Idaidori, Yahabacho, Shiwa-gun, Morioka, Iwate, 028-3694, Japan
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| | - William D Richardson
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
92
|
Lahiri A, Walton JC, Zhang N, Billington N, DeVries AC, Meares GP. Astrocytic deletion of protein kinase R-like ER kinase (PERK) does not affect learning and memory in aged mice but worsens outcome from experimental stroke. J Neurosci Res 2023; 101:1586-1610. [PMID: 37314006 PMCID: PMC10524975 DOI: 10.1002/jnr.25224] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 05/22/2023] [Accepted: 05/27/2023] [Indexed: 06/15/2023]
Abstract
Aging is associated with cognitive decline and is the main risk factor for a myriad of conditions including neurodegeneration and stroke. Concomitant with aging is the progressive accumulation of misfolded proteins and loss of proteostasis. Accumulation of misfolded proteins in the endoplasmic reticulum (ER) leads to ER stress and activation of the unfolded protein response (UPR). The UPR is mediated, in part, by the eukaryotic initiation factor 2α (eIF2α) kinase protein kinase R-like ER kinase (PERK). Phosphorylation of eIF2α reduces protein translation as an adaptive mechanism but this also opposes synaptic plasticity. PERK, and other eIF2α kinases, have been widely studied in neurons where they modulate both cognitive function and response to injury. The impact of astrocytic PERK signaling in cognitive processes was previously unknown. To examine this, we deleted PERK from astrocytes (AstroPERKKO ) and examined the impact on cognitive functions in middle-aged and old mice of both sexes. Additionally, we tested the outcome following experimental stroke using the transient middle cerebral artery occlusion (MCAO) model. Tests of short-term and long-term learning and memory as well as of cognitive flexibility in middle-aged and old mice revealed that astrocytic PERK does not regulate these processes. Following MCAO, AstroPERKKO had increased morbidity and mortality. Collectively, our data demonstrate that astrocytic PERK has limited impact on cognitive function and has a more prominent role in the response to neural injury.
Collapse
Affiliation(s)
| | | | | | | | - A Courtney DeVries
- Department of Neuroscience
- Rockefeller Neuroscience Institute
- Department of Medicine, Division of Hematology and Oncology
- WVU Cancer Institute, Morgantown, WV- 26506, USA
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown, WV- 26506, USA
| | - Gordon P. Meares
- Department of Microbiology, Immunology and Cell Biology
- Department of Neuroscience
- Rockefeller Neuroscience Institute
| |
Collapse
|
93
|
Li X, Li Y, Jin Y, Zhang Y, Wu J, Xu Z, Huang Y, Cai L, Gao S, Liu T, Zeng F, Wang Y, Wang W, Yuan TF, Tian H, Shu Y, Guo F, Lu W, Mao Y, Mei X, Rao Y, Peng B. Transcriptional and epigenetic decoding of the microglial aging process. NATURE AGING 2023; 3:1288-1311. [PMID: 37697166 PMCID: PMC10570141 DOI: 10.1038/s43587-023-00479-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 08/03/2023] [Indexed: 09/13/2023]
Abstract
As important immune cells, microglia undergo a series of alterations during aging that increase the susceptibility to brain dysfunctions. However, the longitudinal characteristics of microglia remain poorly understood. In this study, we mapped the transcriptional and epigenetic profiles of microglia from 3- to 24-month-old mice. We first discovered unexpected sex differences and identified age-dependent microglia (ADEM) genes during the aging process. We then compared the features of aging and reactivity in female microglia at single-cell resolution and epigenetic level. To dissect functions of aged microglia excluding the influence from other aged brain cells, we established an accelerated microglial turnover model without directly affecting other brain cells. By this model, we achieved aged-like microglia in non-aged brains and confirmed that aged-like microglia per se contribute to cognitive decline. Collectively, our work provides a comprehensive resource for decoding the aging process of microglia, shedding light on how microglia maintain brain functions.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Yuxin Li
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Yuxiao Jin
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yuheng Zhang
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Jingchuan Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen Xu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yubin Huang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lin Cai
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuai Gao
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Taohui Liu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Fanzhuo Zeng
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yafei Wang
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Wenxu Wang
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hengli Tian
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yousheng Shu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Feifan Guo
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Wei Lu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xifan Mei
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Bo Peng
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China.
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- Co-Innovation Center of Neurodegeneration, Nantong University, Nantong, China.
| |
Collapse
|
94
|
Xin W, Kaneko M, Roth RH, Zhang A, Nocera S, Ding JB, Stryker MP, Chan JR. Adolescent oligodendrogenesis and myelination restrict experience-dependent neuronal plasticity in adult visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560231. [PMID: 37808666 PMCID: PMC10557765 DOI: 10.1101/2023.09.29.560231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
BACKGROUND Developmental myelination is a protracted process in the mammalian brain. One theory for why oligodendrocytes mature so slowly posits that myelination may stabilize neuronal circuits and temper neuronal plasticity as animals age. We tested this hypothesis in the visual cortex, which has a well-defined critical period for experience-dependent neuronal plasticity. OBJECTIVES/METHODS To prevent myelin progression, we conditionally deleted Myrf, a transcription factor necessary for oligodendrocyte maturation, from oligodendrocyte precursor cells (Myrf cKO) in adolescent mice. To induce experience-dependent plasticity, adult control and Myrf cKO mice were monocularly deprived by eyelid suture. Functional and structural neuronal plasticity in the visual cortex were assessed in vivo by intrinsic signal optical imaging and longitudinal two photon imaging of dendritic spines, respectively. RESULTS During adolescence, visual experience modulated the rate of oligodendrocyte maturation in visual cortex. Myrf deletion from oligodendrocyte precursors during adolescence led to inhibition of oligodendrocyte maturation and myelination that persisted into adulthood. Following monocular deprivation, visual cortex activity in response to visual stimulation of the deprived eye remained stable in adult control mice, as expected for post-critical period animals. By contrast, visual cortex responses to the deprived eye decreased significantly following monocular deprivation in adult Myrf cKO mice, reminiscent of the plasticity observed in adolescent mice. Furthermore, visual cortex neurons in adult Myrf cKO mice had fewer dendritic spines and a higher level of spine turnover. Finally, monocular deprivation induced spatially coordinated spine size decreases in adult Myrf cKO, but not control, mice. CONCLUSIONS These results demonstrate a critical role for oligodendrocytes in shaping the maturation and stabilization of cortical circuits and support the concept of myelin acting as a brake on neuronal plasticity during development.
Collapse
Affiliation(s)
- Wendy Xin
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco
| | - Megumi Kaneko
- Department of Physiology, Kavli Institute for Fundamental Neuroscience and Weill Institute for Neurosciences, University of California San Francisco
| | - Richard H Roth
- Departments of Neurosurgery and Neurology and Neurological Science, Stanford University
| | - Albert Zhang
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco
| | - Sonia Nocera
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco
| | - Jun B Ding
- Departments of Neurosurgery and Neurology and Neurological Science, Stanford University
| | - Michael P Stryker
- Department of Physiology, Kavli Institute for Fundamental Neuroscience and Weill Institute for Neurosciences, University of California San Francisco
| | - Jonah R Chan
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco
| |
Collapse
|
95
|
Vanrobaeys Y, Mukherjee U, Langmack L, Beyer SE, Bahl E, Lin LC, Michaelson JJ, Abel T, Chatterjee S. Mapping the spatial transcriptomic signature of the hippocampus during memory consolidation. Nat Commun 2023; 14:6100. [PMID: 37773230 PMCID: PMC10541893 DOI: 10.1038/s41467-023-41715-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023] Open
Abstract
Memory consolidation involves discrete patterns of transcriptional events in the hippocampus. Despite the emergence of single-cell transcriptomic profiling techniques, mapping the transcriptomic signature across subregions of the hippocampus has remained challenging. Here, we utilized unbiased spatial sequencing to delineate transcriptome-wide gene expression changes across subregions of the dorsal hippocampus of male mice following learning. We find that each subregion of the hippocampus exhibits distinct yet overlapping transcriptomic signatures. The CA1 region exhibited increased expression of genes related to transcriptional regulation, while the DG showed upregulation of genes associated with protein folding. Importantly, our approach enabled us to define the transcriptomic signature of learning within two less-defined hippocampal subregions, CA1 stratum radiatum, and oriens. We demonstrated that CA1 subregion-specific expression of a transcription factor subfamily has a critical functional role in the consolidation of long-term memory. This work demonstrates the power of spatial molecular approaches to reveal simultaneous transcriptional events across the hippocampus during memory consolidation.
Collapse
Affiliation(s)
- Yann Vanrobaeys
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52242, USA
| | - Utsav Mukherjee
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, 52242, USA
| | - Lucy Langmack
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Biochemistry and Molecular Biology Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Stacy E Beyer
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Ethan Bahl
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52242, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Li-Chun Lin
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Jacob J Michaelson
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
| | - Snehajyoti Chatterjee
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
96
|
Mich JK, Sunil S, Johansen N, Martinez RA, Leytze M, Gore BB, Mahoney JT, Ben-Simon Y, Bishaw Y, Brouner K, Campos J, Canfield R, Casper T, Dee N, Egdorf T, Gary A, Gibson S, Goldy J, Groce EL, Hirschstein D, Loftus L, Lusk N, Malone J, Martin NX, Monet D, Omstead V, Opitz-Araya X, Oster A, Pom CA, Potekhina L, Reding M, Rimorin C, Ruiz A, Sedeño-Cortés AE, Shapovalova NV, Taormina M, Taskin N, Tieu M, Valera Cuevas NJ, Weed N, Way S, Yao Z, McMillen DA, Kunst M, McGraw M, Thyagarajan B, Waters J, Bakken TE, Yao S, Smith KA, Svoboda K, Podgorski K, Kojima Y, Horwitz GD, Zeng H, Daigle TL, Lein ES, Tasic B, Ting JT, Levi BP. Enhancer-AAVs allow genetic access to oligodendrocytes and diverse populations of astrocytes across species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.558718. [PMID: 37790503 PMCID: PMC10542530 DOI: 10.1101/2023.09.20.558718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Proper brain function requires the assembly and function of diverse populations of neurons and glia. Single cell gene expression studies have mostly focused on characterization of neuronal cell diversity; however, recent studies have revealed substantial diversity of glial cells, particularly astrocytes. To better understand glial cell types and their roles in neurobiology, we built a new suite of adeno-associated viral (AAV)-based genetic tools to enable genetic access to astrocytes and oligodendrocytes. These oligodendrocyte and astrocyte enhancer-AAVs are highly specific (usually > 95% cell type specificity) with variable expression levels, and our astrocyte enhancer-AAVs show multiple distinct expression patterns reflecting the spatial distribution of astrocyte cell types. To provide the best glial-specific functional tools, several enhancer-AAVs were: optimized for higher expression levels, shown to be functional and specific in rat and macaque, shown to maintain specific activity in epilepsy where traditional promoters changed activity, and used to drive functional transgenes in astrocytes including Cre recombinase and acetylcholine-responsive sensor iAChSnFR. The astrocyte-specific iAChSnFR revealed a clear reward-dependent acetylcholine response in astrocytes of the nucleus accumbens during reinforcement learning. Together, this collection of glial enhancer-AAVs will enable characterization of astrocyte and oligodendrocyte populations and their roles across species, disease states, and behavioral epochs.
Collapse
|
97
|
Gerevich Z, Kovács R, Liotta A, Hasam-Henderson LA, Weh L, Wallach I, Berndt N. Metabolic implications of axonal demyelination and its consequences for synchronized network activity: An in silico and in vitro study. J Cereb Blood Flow Metab 2023; 43:1571-1587. [PMID: 37125487 PMCID: PMC10414014 DOI: 10.1177/0271678x231170746] [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: 01/04/2022] [Revised: 02/13/2023] [Accepted: 03/22/2023] [Indexed: 05/02/2023]
Abstract
Myelination enhances the conduction velocity of action potentials (AP) and increases energy efficiency. Thick myelin sheaths are typically found on large-distance axonal connections or in fast-spiking interneurons, which are critical for synchronizing neuronal networks during gamma-band oscillations. Loss of myelin sheath is associated with multiple alterations in axonal architecture leading to impaired AP propagation. While numerous studies are devoted to the effects of demyelination on conduction velocity, the metabolic effects and the consequences for network synchronization have not been investigated. Here we present a unifying computational model for electrophysiology and metabolism of the myelinated axon. The computational model suggested that demyelination not only decreases the AP speed but AP propagation in demyelinated axons requires compensatory processes like mitochondrial mass increase and a switch from saltatory to continuous propagation to rescue axon functionality at the cost of reduced AP propagation speed and increased energy expenditure. Indeed, these predictions were proven to be true in a culture model of demyelination where the pharmacologically-induced loss of myelin was associated with increased oxygen consumption rates, and a significant broadening of bandwidth as well as a decrease in the power of gamma oscillations.
Collapse
Affiliation(s)
- Zoltan Gerevich
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Richard Kovács
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Agustin Liotta
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Anesthesiology and Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Computer-assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Luisa A Hasam-Henderson
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ludwig Weh
- Institute of Biochemistry, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Iwona Wallach
- Institute of Computer-assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nikolaus Berndt
- Institute of Computer-assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
98
|
Bohlen JF, Cleary CM, Das D, Sripathy SR, Sadowski N, Shim G, Kenney RF, Buchler IP, Banerji T, Scanlan TS, Mulkey DK, Maher BJ. Promyelinating drugs promote functional recovery in an autism spectrum disorder mouse model of Pitt-Hopkins syndrome. Brain 2023; 146:3331-3346. [PMID: 37068912 PMCID: PMC10393406 DOI: 10.1093/brain/awad057] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 01/10/2023] [Accepted: 02/03/2023] [Indexed: 04/19/2023] Open
Abstract
Pitt-Hopkins syndrome is an autism spectrum disorder caused by autosomal dominant mutations in the human transcription factor 4 gene (TCF4). One pathobiological process caused by murine Tcf4 mutation is a cell autonomous reduction in oligodendrocytes and myelination. In this study, we show that the promyelinating compounds, clemastine, sobetirome and Sob-AM2 are effective at restoring myelination defects in a Pitt-Hopkins syndrome mouse model. In vitro, clemastine treatment reduced excess oligodendrocyte precursor cells and normalized oligodendrocyte density. In vivo, 2-week intraperitoneal administration of clemastine also normalized oligodendrocyte precursor cell and oligodendrocyte density in the cortex of Tcf4 mutant mice and appeared to increase the number of axons undergoing myelination, as EM imaging of the corpus callosum showed a significant increase in the proportion of uncompacted myelin and an overall reduction in the g-ratio. Importantly, this treatment paradigm resulted in functional rescue by improving electrophysiology and behaviour. To confirm behavioural rescue was achieved via enhancing myelination, we show that treatment with the thyroid hormone receptor agonist sobetirome or its brain penetrating prodrug Sob-AM2, was also effective at normalizing oligodendrocyte precursor cell and oligodendrocyte densities and behaviour in the Pitt-Hopkins syndrome mouse model. Together, these results provide preclinical evidence that promyelinating therapies may be beneficial in Pitt-Hopkins syndrome and potentially other neurodevelopmental disorders characterized by dysmyelination.
Collapse
Affiliation(s)
- Joseph F Bohlen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Colin M Cleary
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Debamitra Das
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Srinidhi Rao Sripathy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Norah Sadowski
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Gina Shim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Rakaia F Kenney
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Ingrid P Buchler
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Tapasree Banerji
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Thomas S Scanlan
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Daniel K Mulkey
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Brady J Maher
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
99
|
Terranova JI, Yokose J, Osanai H, Ogawa SK, Kitamura T. Systems consolidation induces multiple memory engrams for a flexible recall strategy in observational fear memory in male mice. Nat Commun 2023; 14:3976. [PMID: 37407567 DOI: 10.1038/s41467-023-39718-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
Observers learn to fear the context in which they witnessed a demonstrator's aversive experience, called observational contextual fear conditioning (CFC). The neural mechanisms governing whether recall of the observational CFC memory occurs from the observer's own or from the demonstrator's point of view remain unclear. Here, we show in male mice that recent observational CFC memory is recalled in the observer's context only, but remote memory is recalled in both observer and demonstrator contexts. Recall of recent memory in the observer's context requires dorsal hippocampus activity, while recall of remote memory in both contexts requires the medial prefrontal cortex (mPFC)-basolateral amygdala pathway. Although mPFC neurons activated by observational CFC are involved in remote recall in both contexts, distinct mPFC subpopulations regulate remote recall in each context. Our data provide insights into a flexible recall strategy and the functional reorganization of circuits and memory engram cells underlying observational CFC memory.
Collapse
Affiliation(s)
- Joseph I Terranova
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Anatomy, Midwestern University, Downers Grove, IL, 60615, USA
| | - Jun Yokose
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hisayuki Osanai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sachie K Ogawa
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
100
|
Sierra RO, Pedraza LK, Barcsai L, Pejin A, Li Q, Kozák G, Takeuchi Y, Nagy AJ, Lőrincz ML, Devinsky O, Buzsáki G, Berényi A. Closed-loop brain stimulation augments fear extinction in male rats. Nat Commun 2023; 14:3972. [PMID: 37407557 DOI: 10.1038/s41467-023-39546-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 06/16/2023] [Indexed: 07/07/2023] Open
Abstract
Dysregulated fear reactions can result from maladaptive processing of trauma-related memories. In post-traumatic stress disorder (PTSD) and other psychiatric disorders, dysfunctional extinction learning prevents discretization of trauma-related memory engrams and generalizes fear responses. Although PTSD may be viewed as a memory-based disorder, no approved treatments target pathological fear memory processing. Hippocampal sharp wave-ripples (SWRs) and concurrent neocortical oscillations are scaffolds to consolidate contextual memory, but their role during fear processing remains poorly understood. Here, we show that closed-loop, SWR triggered neuromodulation of the medial forebrain bundle (MFB) can enhance fear extinction consolidation in male rats. The modified fear memories became resistant to induced recall (i.e., 'renewal' and 'reinstatement') and did not reemerge spontaneously. These effects were mediated by D2 receptor signaling-induced synaptic remodeling in the basolateral amygdala. Our results demonstrate that SWR-triggered closed-loop stimulation of the MFB reward system enhances extinction of fearful memories and reducing fear expression across different contexts and preventing excessive and persistent fear responses. These findings highlight the potential of neuromodulation to augment extinction learning and provide a new avenue to develop treatments for anxiety disorders.
Collapse
Affiliation(s)
- Rodrigo Ordoñez Sierra
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
| | - Lizeth Katherine Pedraza
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
| | - Lívia Barcsai
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
- HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged, 6720, Hungary
- Neunos Inc, Boston, MA, 02108, USA
| | - Andrea Pejin
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
- HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged, 6720, Hungary
- Neunos Inc, Boston, MA, 02108, USA
| | - Qun Li
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
| | - Gábor Kozák
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
| | - Yuichi Takeuchi
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
- Department of Biopharmaceutical Sciences and Pharmacy, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Anett J Nagy
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
- HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged, 6720, Hungary
- Neunos Inc, Boston, MA, 02108, USA
| | - Magor L Lőrincz
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
- Department of Physiology, Anatomy and Neuroscience, Faculty of Sciences University of Szeged, Szeged, 6726, Hungary
- Neuroscience Division, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Orrin Devinsky
- Department of Neurology, NYU Langone Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - György Buzsáki
- Neuroscience Institute, New York University, New York, NY, 10016, USA
- Center for Neural Science, New York University, New York, NY, 10016, USA
| | - Antal Berényi
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary.
- HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged, 6720, Hungary.
- Neunos Inc, Boston, MA, 02108, USA.
- Neuroscience Institute, New York University, New York, NY, 10016, USA.
| |
Collapse
|