1
|
Liu Z, Zhao Z, Du H, Zhou Q, Li M, Gui Z, Wu J, Gao Y, Zheng N, Zhang Y, Du A, Wang H, Wang J. Intermittent Fasting Enhances Motor Coordination Through Myelin Preservation in Aged Mice. Aging Cell 2025; 24:e14476. [PMID: 39780365 PMCID: PMC12074029 DOI: 10.1111/acel.14476] [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: 08/02/2024] [Revised: 11/30/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Integrating dietary interventions have been extensively studied for their health benefits, such as Alzheimer's disease, Huntington's disease, and aging. However, it is necessary to fully understand the mechanisms of long-term effects and practical applications of these dietary interventions for health. A 10-week intermittent fasting (IMF) regimen was implemented on the aging animals in the current study. The variations of cerebral functions were analyzed employing a comprehensive experimental design that includes behavioral tests, neuroimaging, and ultrastructural analysis, such as resting-state functional MRI (rsfMRI), EEG/EMG recordings, transmission electron microscopy, and immunohistochemistry. Over a 10-week regimen, IMF significantly improved locomotor activity, motor coordination, and muscle strength compared to controls (p < 0.01). Resting-state fMRI (rsfMRI) demonstrated that IMF modulates brain-wide functional connectivity, enhancing communication between key brain regions. Advanced imaging techniques revealed increased expression of myelin-related proteins, including myelin basic protein (MBP), and myelin-associated glycoprotein (MAG), indicating enhanced myelin integrity and repair, particularly in axons with diameters < 400 nm (p < 0.01). These findings suggest that IMF may mitigate age-related declines by promoting better neuronal signaling. This study highlights the potential function of IMF as a non-pharmacological intervention to promote brain health and mitigate cognitive decline in aging populations.
Collapse
Affiliation(s)
- Zhuang Liu
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective DisordersSongjiang Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and TechnologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ziyue Zhao
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective DisordersSongjiang Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and TechnologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Hongying Du
- Department of Food Science and Engineering, College of Light Industry and Food EngineeringNanjing Forestry UniversityNanjingChina
| | - Qingqing Zhou
- Department of Anesthesiology, Zhongnan HospitalWuhan UniversityWuhanChina
| | - Mei Li
- Department of AnesthesiologyFirst People Hospital of FoshanFoshanChina
| | - Zhu Gui
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective DisordersSongjiang Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and TechnologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jinfeng Wu
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective DisordersSongjiang Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yunling Gao
- Institute of Neuroscience and Brain Diseases; Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangHubeiChina
| | - Ning Zheng
- Clinical & Technical Support, Philips HealthcareShanghaiChina
| | - Yu Zhang
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective DisordersSongjiang Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ailian Du
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective DisordersSongjiang Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hongxing Wang
- Division of Neuropsychiatry and Psychosomatics, Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Jie Wang
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective DisordersSongjiang Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Neuroscience and Brain Diseases; Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangHubeiChina
| |
Collapse
|
2
|
García-Domínguez M. Pathological and Inflammatory Consequences of Aging. Biomolecules 2025; 15:404. [PMID: 40149940 PMCID: PMC11939965 DOI: 10.3390/biom15030404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Aging is a complex, progressive, and irreversible biological process that entails numerous structural and functional changes in the organism. These changes affect all bodily systems, reducing their ability to respond and adapt to the environment. Chronic inflammation is one of the key factors driving the development of age-related diseases, ultimately causing a substantial decline in the functional abilities of older individuals. This persistent inflammatory state (commonly known as "inflammaging") is characterized by elevated levels of pro-inflammatory cytokines, an increase in oxidative stress, and a perturbation of immune homeostasis. Several factors, including cellular senescence, contribute to this inflammatory milieu, thereby amplifying conditions such as cardiovascular disease, neurodegeneration, and metabolic disorders. Exploring the mechanisms of chronic inflammation in aging is essential for developing targeted interventions aimed at promoting healthy aging. This review explains the strong connection between aging and chronic inflammation, highlighting potential therapeutic approaches like pharmacological treatments, dietary strategies, and lifestyle changes.
Collapse
Affiliation(s)
- Mario García-Domínguez
- Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain;
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
3
|
Lin ML, Lin W. Thinning of originally-existing, mature myelin represents a nondestructive form of myelin loss in the adult CNS. Front Cell Neurosci 2025; 19:1565913. [PMID: 40134707 PMCID: PMC11933062 DOI: 10.3389/fncel.2025.1565913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
The main function of oligodendrocytes is to assemble and maintain myelin that wraps and insulates axons in the central nervous system (CNS). Traditionally, myelin structure, particularly its thickness, was believed to remain remarkably stable in adulthood (including early and middle adulthood, but not late adulthood or aging). However, emerging evidence reveals that the thickness of originally-existing, mature myelin (OEM) can undergo dynamic changes in the adult CNS. This overview highlights recent findings on the alteration of OEM thickness in the adult CNS, explores the underlying mechanisms, and proposes that progressive thinning of OEM represents a novel, nondestructive form of myelin loss in myelin disorders of the CNS.
Collapse
Affiliation(s)
- Min Li Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
4
|
Gadek M, Shaw CK, Abdulai-Saiku S, Saloner R, Marino F, Wang D, Bonham LW, Yokoyama JS, Panning B, Benayoun BA, Casaletto KB, Ramani V, Dubal DB. Aging activates escape of the silent X chromosome in the female mouse hippocampus. SCIENCE ADVANCES 2025; 11:eads8169. [PMID: 40043106 PMCID: PMC11881916 DOI: 10.1126/sciadv.ads8169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/31/2024] [Indexed: 03/09/2025]
Abstract
Women live longer than men and exhibit less cognitive aging. The X chromosome contributes to sex differences, as females harbor an inactive X (Xi) and active X (Xa), in contrast to males with only an Xa. Thus, reactivation of silent Xi genes may contribute to sex differences. We use allele-specific, single-nucleus RNA sequencing to show that aging remodels transcription of the Xi and Xa across hippocampal cell types. Aging preferentially changed gene expression on the X's relative to autosomes. Select genes on the Xi underwent activation, with new escape across cells including in the dentate gyrus, critical to learning and memory. Expression of the Xi escapee Plp1, a myelin component, was increased in the aging hippocampus of female mice and parahippocampus of women. AAV-mediated Plp1 elevation in the dentate gyrus of aging male and female mice improved cognition. Understanding how the Xi may confer female advantage could lead to novel targets that counter brain aging and disease in both sexes.
Collapse
Affiliation(s)
- Margaret Gadek
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Cayce K. Shaw
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Rehabilitation Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Samira Abdulai-Saiku
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Rowan Saloner
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Francesca Marino
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Neurosciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Dan Wang
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Luke W. Bonham
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jennifer S. Yokoyama
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Barbara Panning
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Bérénice A. Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA, USA
- Biochemistry and Molecular Medicine Department, USC Keck School of Medicine; USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
- USC Stem Cell Initiative, Los Angeles, CA, USA
| | - Kaitlin B. Casaletto
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Vijay Ramani
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Gladstone Institute for Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, San Francisco, CA, USA
| | - Dena B. Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Rehabilitation Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Neurosciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
5
|
Sun ED, Nagvekar R, Pogson AN, Brunet A. Brain aging and rejuvenation at single-cell resolution. Neuron 2025; 113:82-108. [PMID: 39788089 PMCID: PMC11842159 DOI: 10.1016/j.neuron.2024.12.007] [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: 08/26/2024] [Revised: 11/16/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025]
Abstract
Brain aging leads to a decline in cognitive function and a concomitant increase in the susceptibility to neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. A key question is how changes within individual cells of the brain give rise to age-related dysfunction. Developments in single-cell "omics" technologies, such as single-cell transcriptomics, have facilitated high-dimensional profiling of individual cells. These technologies have led to new and comprehensive characterizations of brain aging at single-cell resolution. Here, we review insights gleaned from single-cell omics studies of brain aging, starting with a cell-type-centric overview of age-associated changes and followed by a discussion of cell-cell interactions during aging. We highlight how single-cell omics studies provide an unbiased view of different rejuvenation interventions and comment on the promise of combinatorial rejuvenation approaches for the brain. Finally, we propose new directions, including models of brain aging and neural stem cells as a focal point for rejuvenation.
Collapse
Affiliation(s)
- Eric D Sun
- Department of Genetics, Stanford University, Stanford, CA, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA, USA; Biomedical Informatics Graduate Program, Stanford University, Stanford, CA, USA
| | - Rahul Nagvekar
- Department of Genetics, Stanford University, Stanford, CA, USA; Genetics Graduate Program, Stanford University, Stanford, CA, USA
| | - Angela N Pogson
- Department of Genetics, Stanford University, Stanford, CA, USA; Developmental Biology Graduate Program, Stanford University, Stanford, CA, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
6
|
Carlyon RP, Deeks JM, Delgutte B, Chung Y, Vollmer M, Ohl FW, Kral A, Tillein J, Litovsky RY, Schnupp J, Rosskothen-Kuhl N, Goldsworthy RL. Limitations on Temporal Processing by Cochlear Implant Users: A Compilation of Viewpoints. Trends Hear 2025; 29:23312165251317006. [PMID: 40095543 PMCID: PMC12076235 DOI: 10.1177/23312165251317006] [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: 04/28/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 03/19/2025] Open
Abstract
Cochlear implant (CI) users are usually poor at using timing information to detect changes in either pitch or sound location. This deficit occurs even for listeners with good speech perception and even when the speech processor is bypassed to present simple, idealized stimuli to one or more electrodes. The present article presents seven expert opinion pieces on the likely neural bases for these limitations, the extent to which they are modifiable by sensory experience and training, and the most promising ways to overcome them in future. The article combines insights from physiology and psychophysics in cochlear-implanted humans and animals, highlights areas of agreement and controversy, and proposes new experiments that could resolve areas of disagreement.
Collapse
Affiliation(s)
- Robert P. Carlyon
- Cambridge Hearing Group, MRC Cognition & Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - John M. Deeks
- Cambridge Hearing Group, MRC Cognition & Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Bertrand Delgutte
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
| | - Yoojin Chung
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
| | - Maike Vollmer
- Department of Experimental Audiology, University Clinic of Otolaryngology, Head and Neck Surgery, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Frank W. Ohl
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
| | - Andrej Kral
- Institute of Audio-Neuro-Technology & Department of Experimental Otology, Clinics of Otolaryngology, Head and Neck Surgery, Hannover Medical School, Hannover, Germany
| | - Jochen Tillein
- Clinics of Otolaryngology, Head and Neck Surgery, J.W.Goethe University, Frankfurt, Germany
- MedEl Company, Hannover, Germany
| | - Ruth Y. Litovsky
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jan Schnupp
- Gerald Choa Neuroscience Institute and Department of Otolaryngology, Chinese University of Hong Kong, Hong Kong (NB Hong Kong is a Special Administrative Region) of China
| | - Nicole Rosskothen-Kuhl
- Neurobiological Research Laboratory, Section for Experimental and Clinical Otology, Department of Oto-Rhino-Laryngology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Bernstein Center Freiburg & Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Raymond L. Goldsworthy
- Auditory Research Center, Caruso Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
7
|
Pun R, Kumari N, Monieb RH, Wagh S, North BJ. BubR1 and SIRT2: Insights into aneuploidy, aging, and cancer. Semin Cancer Biol 2024; 106-107:201-216. [PMID: 39490401 PMCID: PMC11625622 DOI: 10.1016/j.semcancer.2024.10.005] [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: 08/09/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Aging is a significant risk factor for cancer which is due, in part, to heightened genomic instability. Mitotic surveillance proteins such as BubR1 play a pivotal role in ensuring accurate chromosomal segregation and preventing aneuploidy. BubR1 levels have been shown to naturally decline with age and its loss is associated with various age-related pathologies. Sirtuins, a class of NAD+-dependent deacylases, are implicated in cancer and genomic instability. Among them, SIRT2 acts as an upstream regulator of BubR1, offering a critical pathway that can potentially mitigate age-related diseases, including cancer. In this review, we explore BubR1 as a key regulator of cellular processes crucial for aging-related phenotypes. We delve into the intricate mechanisms through which BubR1 influences genomic stability and cellular senescence. Moreover, we highlight the role of NAD+ and SIRT2 in modulating BubR1 expression and function, emphasizing its potential as a therapeutic target. The interaction between BubR1 and SIRT2 not only serves as a fundamental regulatory pathway in cellular homeostasis but also represents a promising avenue for developing targeted therapies against age-related diseases, particularly cancer.
Collapse
Affiliation(s)
- Renju Pun
- Biomedical Sciences Department, Creighton University School of Medicine, Omaha, NE, USA
| | - Niti Kumari
- Biomedical Sciences Department, Creighton University School of Medicine, Omaha, NE, USA
| | - Rodaina Hazem Monieb
- Biomedical Sciences Department, Creighton University School of Medicine, Omaha, NE, USA
| | - Sachin Wagh
- Biomedical Sciences Department, Creighton University School of Medicine, Omaha, NE, USA
| | - Brian J North
- Biomedical Sciences Department, Creighton University School of Medicine, Omaha, NE, USA.
| |
Collapse
|
8
|
Huang G, Li Z, Liu X, Guan M, Zhou S, Zhong X, Zheng T, Xin D, Gu X, Mu D, Guo Y, Zhang L, Zhang L, Lu QR, He X. DOR activation in mature oligodendrocytes regulates α-ketoglutarate metabolism leading to enhanced remyelination in aged mice. Nat Neurosci 2024; 27:2073-2085. [PMID: 39266660 DOI: 10.1038/s41593-024-01754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/07/2024] [Indexed: 09/14/2024]
Abstract
The decreased ability of mature oligodendrocytes to produce myelin negatively affects remyelination in demyelinating diseases and aging, but the underlying mechanisms are incompletely understood. In the present study, we identify a mature oligodendrocyte-enriched transcriptional coregulator diabetes- and obesity-related gene (DOR)/tumor protein p53-inducible nuclear protein 2 (TP53INP2), downregulated in demyelinated lesions of donors with multiple sclerosis and in aged oligodendrocyte-lineage cells. Dor ablation in mice of both sexes results in defective myelinogenesis and remyelination. Genomic occupancy in oligodendrocytes and transcriptome profiling of the optic nerves of wild-type and Dor conditional knockout mice reveal that DOR and SOX10 co-occupy enhancers of critical myelinogenesis-associated genes including Prr18, encoding an oligodendrocyte-enriched, proline-rich factor. We show that DOR targets regulatory elements of genes responsible for α-ketoglutarate biosynthesis in mature oligodendrocytes and is essential for α-ketoglutarate production and lipid biosynthesis. Supplementation with α-ketoglutarate restores oligodendrocyte-maturation defects in Dor-deficient adult mice and improves remyelination after lysolecithin-induced demyelination and cognitive function in 17-month-old wild-type mice. Our data suggest that activation of α-ketoglutarate metabolism in mature oligodendrocytes can promote myelin production during demyelination and aging.
Collapse
Affiliation(s)
- Guojiao Huang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhidan Li
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xuezhao Liu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Menglong Guan
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaowen Zhong
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tao Zheng
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dazhuan Xin
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Dezhi Mu
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yingkun Guo
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lin Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liguo Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xuelian He
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Li Y, Badawi Y, Meriney SD. Age-Related Homeostatic Plasticity at Rodent Neuromuscular Junctions. Cells 2024; 13:1684. [PMID: 39451202 PMCID: PMC11506802 DOI: 10.3390/cells13201684] [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: 09/04/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Motor ability decline remains a major threat to the quality of life of the elderly. Although the later stages of aging co-exist with degenerative pathologies, the long process of aging is more complicated than a simple and gradual degeneration. To combat senescence and the associated late-stage degeneration of the neuromuscular system, it is imperative to examine changes that occur during the long process of aging. Prior to late-stage degeneration, age-induced changes in the neuromuscular system trigger homeostatic plasticity. This unique phenomenon may be important for the maintenance of the neuromuscular system during the early stages of aging. In this review, we will focus on age-induced changes in neurotransmission at the neuromuscular junction, providing the potential mechanisms responsible for these changes. The goal is to highlight these key elements and their role in regulating neurotransmission, facilitating future research efforts to combat late-stage degeneration in the neuromuscular system by preserving the functional and structural integrity of these elements prior to the late stage of aging.
Collapse
Affiliation(s)
| | | | - Stephen D. Meriney
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; (Y.L.); (Y.B.)
| |
Collapse
|
10
|
Martín-Lopez G, Mallavibarrena PR, Villa-Gonzalez M, Vidal N, Pérez-Alvarez MJ. The dynamics of oligodendrocyte populations following permanent ischemia promotes long-term spontaneous remyelination of damaged area. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167270. [PMID: 38823461 DOI: 10.1016/j.bbadis.2024.167270] [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: 12/04/2023] [Revised: 05/10/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Stroke is a major public health concern, with limited clinically approved interventions available to enhance sensorimotor recovery beyond reperfusion. Remarkably, spontaneous recovery is observed in certain stroke patients, suggesting the existence of a brain self-repair mechanism not yet fully understood. In a rat model of permanent cerebral ischemia, we described an increase in oligodendrocytes expressing 3RTau in damaged area. Considering that restoration of myelin integrity ameliorates symptoms in many neurodegenerative diseases, here we hypothesize that this cellular response could trigger remyelination. Our results revealed after ischemia an early recruitment of OPCs to damaged area, followed by their differentiation into 3RTau+ pre-myelinating cells and subsequent into remyelinating oligodendrocytes. Using rat brain slices and mouse primary culture we confirmed the presence of 3RTau in pre-myelinating and a subset of mature oligodendrocytes. The myelin status analysis confirmed long-term remyelination in the damaged area. Postmortem samples from stroke subjects showed a reduction in oligodendrocytes, 3RTau+ cells, and myelin complexity in subcortical white matter. In conclusion, the dynamics of oligodendrocyte populations after ischemia reveals a spontaneous brain self-repair mechanism which restores the functionality of neuronal circuits long-term by remyelination of damaged area. This is evidenced by the improvement of sensorimotor functions in ischemic rats. A deep understanding of this mechanism could be valuable in the search for alternative oligodendrocyte-based, therapeutic interventions to reduce the effects of stroke.
Collapse
Affiliation(s)
- Gerardo Martín-Lopez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Paula R Mallavibarrena
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Mario Villa-Gonzalez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Noemi Vidal
- Departamento de Patología, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Maria José Pérez-Alvarez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain; Instituto Universitario de Biología Molecular (IUBM), Universidad Autónoma de Madrid, Centro de Biología Molecular Severo Ochoa (CBM), Departamento de Neuropatología Molecular UAM-CSIC, 28049 Madrid, Spain.
| |
Collapse
|
11
|
Jörg LM, Schlötzer-Schrehardt U, Lefebvre V, Sock E, Wegner M. Transcription Factors Sox8 and Sox10 Contribute with Different Importance to the Maintenance of Mature Oligodendrocytes. Int J Mol Sci 2024; 25:8754. [PMID: 39201442 PMCID: PMC11354551 DOI: 10.3390/ijms25168754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Myelin-forming oligodendrocytes in the vertebrate nervous system co-express the transcription factor Sox10 and its paralog Sox8. While Sox10 plays crucial roles throughout all stages of oligodendrocyte development, including terminal differentiation, the loss of Sox8 results in only mild and transient perturbations. Here, we aimed to elucidate the roles and interrelationships of these transcription factors in fully differentiated oligodendrocytes and myelin maintenance in adults. For that purpose, we conducted targeted deletions of Sox10, Sox8, or both in the brains of two-month-old mice. Three weeks post-deletion, none of the resulting mouse mutants exhibited significant alterations in oligodendrocyte numbers, myelin sheath counts, myelin ultrastructure, or myelin protein levels in the corpus callosum, despite efficient gene inactivation. However, differences were observed in the myelin gene expression in mice with Sox10 or combined Sox8/Sox10 deletion. RNA-sequencing analysis on dissected corpus callosum confirmed substantial alterations in the oligodendrocyte expression profile in mice with combined deletion and more subtle changes in mice with Sox10 deletion alone. Notably, Sox8 deletion did not affect any aspects of the expression profile related to the differentiated state of oligodendrocytes or myelin integrity. These findings extend our understanding of the roles of Sox8 and Sox10 in oligodendrocytes into adulthood and have important implications for the functional relationship between the paralogs and the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Lisa Mirja Jörg
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D91054 Erlangen, Germany; (L.M.J.); (E.S.)
| | | | - Véronique Lefebvre
- Department of Surgery, Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Elisabeth Sock
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D91054 Erlangen, Germany; (L.M.J.); (E.S.)
| | - Michael Wegner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D91054 Erlangen, Germany; (L.M.J.); (E.S.)
| |
Collapse
|
12
|
Khodanovich M, Svetlik M, Kamaeva D, Usova A, Kudabaeva M, Anan’ina T, Vasserlauf I, Pashkevich V, Moshkina M, Obukhovskaya V, Kataeva N, Levina A, Tumentceva Y, Vasilieva S, Schastnyy E, Naumova A. Demyelination in Patients with POST-COVID Depression. J Clin Med 2024; 13:4692. [PMID: 39200834 PMCID: PMC11355865 DOI: 10.3390/jcm13164692] [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: 06/14/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Depression is one of the most severe sequelae of COVID-19, with major depressive disorder often characterized by disruption in white matter (WM) connectivity stemming from changes in brain myelination. This study aimed to quantitatively assess brain myelination in clinically diagnosed post-COVID depression (PCD) using the recently proposed MRI method, macromolecular proton fraction (MPF) mapping. Methods: The study involved 63 recovered COVID-19 patients (52 mild, 11 moderate, and 2 severe) at 13.5 ± 10.0 months post-recovery, with matched controls without prior COVID-19 history (n = 19). A post-COVID depression group (PCD, n = 25) was identified based on psychiatric diagnosis, while a comparison group (noPCD, n = 38) included participants with neurological COVID-19 complications, excluding clinical depression. Results: Fast MPF mapping revealed extensive demyelination in PCD patients, particularly in juxtacortical WM (predominantly occipital lobe and medial surface), WM tracts (inferior fronto-occipital fasciculus (IFOF), posterior thalamic radiation, external capsule, sagittal stratum, tapetum), and grey matter (GM) structures (hippocampus, putamen, globus pallidus, and amygdala). The noPCD group also displayed notable demyelination, but with less magnitude and propagation. Multiple regression analysis highlighted IFOF demyelination as the primary predictor of Hamilton scores, PCD presence, and severity. The number of post-COVID symptoms was a significant predictor of PCD presence, while the number of acute symptoms was a significant predictor of PCD severity. Conclusions: This study, for the first time, reveals extensive demyelination in numerous WM and GM structures in PCD, outlining IFOF demyelination as a key biomarker.
Collapse
Affiliation(s)
- Marina Khodanovich
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Mikhail Svetlik
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Daria Kamaeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Anna Usova
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 12/1 Savinykh Street, Tomsk 634028, Russia
| | - Marina Kudabaeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Tatyana Anan’ina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Irina Vasserlauf
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Valentina Pashkevich
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Marina Moshkina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Victoria Obukhovskaya
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Department of Fundamental Psychology and Behavioral Medicine, Siberian State Medical University, 2 Moskovskiy Trakt, Tomsk 634050, Russia
| | - Nadezhda Kataeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Department of Neurology and Neurosurgery, Siberian State Medical University, 2 Moskovskiy Trakt, Tomsk 634028, Russia
| | - Anastasia Levina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Medica Diagnostic and Treatment Center, 86 Sovetskaya Street, Tomsk 634510, Russia
| | - Yana Tumentceva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Svetlana Vasilieva
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Evgeny Schastnyy
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Anna Naumova
- Department of Radiology, School of Medicine, South Lake Union Campus, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| |
Collapse
|
13
|
Shinsato RN, Correa CG, Herai RH. Genetic network analysis indicate that individuals affected by neurodevelopmental conditions have genetic variations associated with ophthalmologic alterations: A critical review of literature. Gene 2024; 908:148246. [PMID: 38325665 DOI: 10.1016/j.gene.2024.148246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/19/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Changes in the nervous system are related to a wide range of mental disorders, which include neurodevelopmental disorders (NDD) that are characterized by early onset mental conditions, such as schizophrenia and autism spectrum disorders and correlated conditions (ASD). Previous studies have shown distinct genetic components associated with diverse schizophrenia and ASD phenotypes, with mostly focused on rescuing neural phenotypes and brain activity, but alterations related to vision are overlooked. Thus, as the vision is composed by the eyes that itself represents a part of the brain, with the retina being formed by neurons and cells originating from the glia, genetic variations affecting the brain can also affect the vision. Here, we performed a critical systematic literature review to screen for all genetic variations in individuals presenting NDD with reported alterations in vision. Using these restricting criteria, we found 20 genes with distinct types of genetic variations, inherited or de novo, that includes SNP, SNV, deletion, insertion, duplication or indel. The variations occurring within protein coding regions have different impact on protein formation, such as missense, nonsense or frameshift. Moreover, a molecular analysis of the 20 genes found revealed that 17 shared a common protein-protein or genetic interaction network. Moreover, gene expression analysis in samples from the brain and other tissues indicates that 18 of the genes found are highly expressed in the brain and retina, indicating their potential role in adult vision phenotype. Finally, we only found 3 genes from our study described in standard public databanks of ophthalmogenetics, suggesting that the other 17 genes could be novel target for vision diseases.
Collapse
Affiliation(s)
- Rogério N Shinsato
- Unisalesiano, Araçatuba, São Paulo, Brazil; Laboratory of Bioinformatics and Neurogenetics (LaBiN/LEM), Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Paraná, 80215-901, Brazil.
| | - Camila Graczyk Correa
- Laboratory of Bioinformatics and Neurogenetics (LaBiN/LEM), Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Paraná, 80215-901, Brazil
| | - Roberto H Herai
- Laboratory of Bioinformatics and Neurogenetics (LaBiN/LEM), Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Paraná, 80215-901, Brazil; Research Division, Buko Kaesemodel Institute (IBK), Curitiba, Paraná 80240-000, Brazil; Research Division, 9p Brazil Association (A9pB), Santa Maria, Rio Grande do Sul 97060-580, Brazil.
| |
Collapse
|
14
|
Liu X, Xin DE, Zhong X, Zhao C, Li Z, Zhang L, Dourson AJ, Lee L, Mishra S, Bayat AE, Nicholson E, Seibel WL, Yan B, Mason J, Turner BJ, Gonsalvez DG, Ong W, Chew SY, Ghosh B, Yoon SO, Xin M, He Z, Tchieu J, Wegner M, Nave KA, Franklin RJM, Dutta R, Trapp BD, Hu M, Smith MA, Jankowski MP, Barton SK, He X, Lu QR. Small-molecule-induced epigenetic rejuvenation promotes SREBP condensation and overcomes barriers to CNS myelin regeneration. Cell 2024; 187:2465-2484.e22. [PMID: 38701782 PMCID: PMC11812128 DOI: 10.1016/j.cell.2024.04.005] [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: 05/31/2023] [Revised: 01/01/2024] [Accepted: 04/04/2024] [Indexed: 05/05/2024]
Abstract
Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.
Collapse
Affiliation(s)
- Xuezhao Liu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Dazhuan Eric Xin
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaowen Zhong
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, 45229, USA
| | - Chuntao Zhao
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zhidan Li
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Liguo Zhang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adam J Dourson
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lindsay Lee
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Shreya Mishra
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Arman E Bayat
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Eva Nicholson
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - William L Seibel
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bingfang Yan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, 45229, USA
| | - Joel Mason
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne 3052, Australia
| | - Bradley J Turner
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne 3052, Australia
| | - David G Gonsalvez
- Department of Anatomy and Developmental Biology, Monash University, Melbourne 3168, Australia
| | - William Ong
- School of Chemistry, Chemical Engineering, and Biotechnology Nanyang Technological University, Singapore 637459, Singapore
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering, and Biotechnology Nanyang Technological University, Singapore 637459, Singapore; Lee Kong Chian School of Medicine, School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani Hyderabad Campus, Shamirpet, Hyderabad, India, 500078
| | - Sung Ok Yoon
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, Ohio
| | - Mei Xin
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jason Tchieu
- Department of Pediatrics, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael Wegner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Robin J M Franklin
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge CB21 6GP, UK
| | - Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Case Western Reserve University School of Medicine, Cleveland, OH 44195, USA
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Case Western Reserve University School of Medicine, Cleveland, OH 44195, USA
| | - Ming Hu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Matthew A Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA; Rebecca D. Considine Research Institute, Akron Children's Hospital, Akron, OH, USA
| | - Michael P Jankowski
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Pediatric Pain Research Center, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Samantha K Barton
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne 3052, Australia
| | - Xuelian He
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China.
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
15
|
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
|
16
|
Hu M, Xu F, Liu S, Yao Y, Xia Q, Zhu C, Zhang X, Tang H, Qaiser Z, Liu S, Tang Y. Aging pattern of the brainstem based on volumetric measurement and optimized surface shape analysis. Brain Imaging Behav 2024; 18:396-411. [PMID: 38155336 DOI: 10.1007/s11682-023-00840-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2023] [Indexed: 12/30/2023]
Abstract
The brainstem, a small and crucial structure, is connected to the cerebrum, spinal cord, and cerebellum, playing a vital role in regulating autonomic functions, transmitting motor and sensory information, and modulating cognitive processes, emotions, and consciousness. While previous research has indicated that changes in brainstem anatomy can serve as a biomarker for aging and neurodegenerative diseases, the structural changes that occur in the brainstem during normal aging remain unclear. This study aimed to examine the age- and sex-related differences in the global and local structural measures of the brainstem in 187 healthy adults (ranging in age from 18 to 70 years) using structural magnetic resonance imaging. The findings showed a significant negative age effect on the volume of the two major components of the brainstem: the medulla oblongata and midbrain. The shape analysis revealed that atrophy primarily occurs in specific structures, such as the pyramid, cerebral peduncle, superior and inferior colliculi. Surface area and shape analysis showed a trend of flattening in the aging brainstem. There were no significant differences between the sexes or sex-by-age interactions in brainstem structural measures. These findings provide a systematic description of age associations with brainstem structures in healthy adults and may provide a reference for future research on brain aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Minqi Hu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Feifei Xu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Shizhou Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Yuan Yao
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Qing Xia
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Caiting Zhu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Xinwen Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Haiyan Tang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Zubair Qaiser
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Shuwei Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Yuchun Tang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
17
|
Zota I, Chanoumidou K, Charalampopoulos I, Gravanis A. Dynamics of myelin deficits in the 5xFAD mouse model for Alzheimer's disease and the protective role of BDNF. Glia 2024; 72:809-827. [PMID: 38205694 DOI: 10.1002/glia.24505] [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: 06/11/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024]
Abstract
Recent findings highlight myelin breakdown as a decisive early event in Alzheimer's Disease (AD) acting as aggravating factor of its progression. However, it is still unclear whether myelin loss is attributed to increased oligodendrocyte vulnerability, reduced repairing capacity or toxic stimuli. In the present study, we sought to clarify the starting point of myelin disruption accompanied with Oligodendrocyte Progenitor Cell (OPC) elimination in the brain of the 5xFAD mouse model of AD at 6 months of age in Dentate Gyrus of the hippocampus in relation to neurotrophin system. Prominent inflammation presence was detected since the age of 6 months playing a key role in myelin disturbance and AD progression. Expression analysis of neurotrophin receptors in OPCs was performed to identify new targets that could increase myelination in health and disease. OPCs in both control and 5xFAD mice express TrkB, TrkC and p75 receptors but not TrkA. Brain-derived neurotrophic factor (BDNF) that binds to TrkB receptor is well-known about its pro-myelination effect, promoting oligodendrocytes proliferation and differentiation, so we focused our investigation on its effects in OPCs under neurodegenerative conditions. Our in vitro results showed that BDNF rescues OPCs from death and promotes their proliferation and differentiation in presence of the toxic Amyloid-β 1-42. Collectively, our results indicate that BDNF possess an additional neuroprotective role through its actions on oligodendrocytic component and its use could be proposed as a drug-based myelin-enhancing strategy, complementary to amyloid and tau centered therapies in AD.
Collapse
Affiliation(s)
- Ioanna Zota
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| | - Konstantina Chanoumidou
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| |
Collapse
|
18
|
Chapman TW, Kamen Y, Piedra ET, Hill RA. Oligodendrocyte Maturation Alters the Cell Death Mechanisms That Cause Demyelination. J Neurosci 2024; 44:e1794232024. [PMID: 38395617 PMCID: PMC10977033 DOI: 10.1523/jneurosci.1794-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Myelinating oligodendrocytes die in human disease and early in aging. Despite this, the mechanisms that underly oligodendrocyte death are not resolved and it is also not clear whether these mechanisms change as oligodendrocyte lineage cells are undergoing differentiation and maturation. Here, we used a combination of intravital imaging, single-cell ablation, and cuprizone-mediated demyelination, in both female and male mice, to discover that oligodendrocyte maturation dictates the dynamics and mechanisms of cell death. After single-cell phototoxic damage, oligodendrocyte precursor cells underwent programmed cell death within hours, differentiating oligodendrocytes died over several days, while mature oligodendrocytes took weeks to die. Importantly cells at each maturation stage all eventually died but did so with drastically different temporal dynamics and morphological features. Consistent with this, cuprizone treatment initiated a caspase-3-dependent form of rapid cell death in differentiating oligodendrocytes, while mature oligodendrocytes never activated this executioner caspase. Instead, mature oligodendrocytes exhibited delayed cell death which was marked by DNA damage and disruption in poly-ADP-ribose subcellular localization. Thus, oligodendrocyte maturation plays a key role in determining the mechanism of death a cell undergoes in response to the same insult. This means that oligodendrocyte maturation is important to consider when designing strategies for preventing cell death and preserving myelin while also enhancing the survival of new oligodendrocytes in demyelinating conditions.
Collapse
Affiliation(s)
- Timothy W Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Yasmine Kamen
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Enrique T Piedra
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| |
Collapse
|
19
|
Khodanovich M, Naumova A, Kamaeva D, Obukhovskaya V, Vasilieva S, Schastnyy E, Kataeva N, Levina A, Kudabaeva M, Pashkevich V, Moshkina M, Tumentceva Y, Svetlik M. Neurocognitive Changes in Patients with Post-COVID Depression. J Clin Med 2024; 13:1442. [PMID: 38592295 PMCID: PMC10933987 DOI: 10.3390/jcm13051442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Depression and cognitive impairment are recognized complications of COVID-19. This study aimed to assess cognitive performance in clinically diagnosed post-COVID depression (PCD, n = 25) patients using neuropsychological testing. Methods: The study involved 71 post-COVID patients with matched control groups: recovered COVID-19 individuals without complications (n = 18) and individuals without prior COVID-19 history (n = 19). A post-COVID depression group (PCD, n = 25) was identified based on psychiatric diagnosis, and a comparison group (noPCD, n = 46) included participants with neurological COVID-19 complications, excluding clinical depression. Results: The PCD patients showed gender-dependent significant cognitive impairment in the MoCA, Word Memory Test (WMT), Stroop task (SCWT), and Trail Making Test (TMT) compared to the controls and noPCD patients. Men with PCD showed worse performances on the SCWT, in MoCA attention score, and on the WMT (immediate and delayed word recall), while women with PCD showed a decline in MoCA total score, an increased processing time with less errors on the TMT, and worse immediate recall. No differences between groups in Sniffin's stick test were found. Conclusions: COVID-related direct (post-COVID symptoms) and depression-mediated (depression itself, male sex, and severity of COVID-19) predictors of decline in memory and information processing speed were identified. Our findings may help to personalize the treatment of depression, taking a patient's gender and severity of previous COVID-19 disease into account.
Collapse
Affiliation(s)
- Marina Khodanovich
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (D.K.); (V.O.); (N.K.); (A.L.); (M.K.); (V.P.); (M.M.); (Y.T.); (M.S.)
| | - Anna Naumova
- Department of Radiology, School of Medicine, South Lake Union Campus, University of Washington, 850 Republican Street, Seattle, WA 98109, USA;
| | - Daria Kamaeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (D.K.); (V.O.); (N.K.); (A.L.); (M.K.); (V.P.); (M.M.); (Y.T.); (M.S.)
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 4 Aleutskaya Street, Tomsk 634014, Russia
| | - Victoria Obukhovskaya
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (D.K.); (V.O.); (N.K.); (A.L.); (M.K.); (V.P.); (M.M.); (Y.T.); (M.S.)
- Department of Fundamental Psychology and Behavioral Medicine, Siberian State Medical University, 2 Moskovskiy Trakt, Tomsk 6340505, Russia
| | - Svetlana Vasilieva
- Department of Affective States, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 4 Aleutskaya Street, Tomsk 634014, Russia; (S.V.); (E.S.)
| | - Evgeny Schastnyy
- Department of Affective States, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 4 Aleutskaya Street, Tomsk 634014, Russia; (S.V.); (E.S.)
| | - Nadezhda Kataeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (D.K.); (V.O.); (N.K.); (A.L.); (M.K.); (V.P.); (M.M.); (Y.T.); (M.S.)
- Department of Neurology and Neurosurgery, Siberian State Medical University, 2 Moskovskiy Trakt, Tomsk 6340505, Russia
| | - Anastasia Levina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (D.K.); (V.O.); (N.K.); (A.L.); (M.K.); (V.P.); (M.M.); (Y.T.); (M.S.)
- Medica Diagnostic and Treatment Center, 86 Sovetskaya Street, Tomsk 634510, Russia
| | - Marina Kudabaeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (D.K.); (V.O.); (N.K.); (A.L.); (M.K.); (V.P.); (M.M.); (Y.T.); (M.S.)
| | - Valentina Pashkevich
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (D.K.); (V.O.); (N.K.); (A.L.); (M.K.); (V.P.); (M.M.); (Y.T.); (M.S.)
| | - Marina Moshkina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (D.K.); (V.O.); (N.K.); (A.L.); (M.K.); (V.P.); (M.M.); (Y.T.); (M.S.)
| | - Yana Tumentceva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (D.K.); (V.O.); (N.K.); (A.L.); (M.K.); (V.P.); (M.M.); (Y.T.); (M.S.)
| | - Mikhail Svetlik
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (D.K.); (V.O.); (N.K.); (A.L.); (M.K.); (V.P.); (M.M.); (Y.T.); (M.S.)
| |
Collapse
|
20
|
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
|
21
|
Khodanovich M, Svetlik M, Naumova A, Kamaeva D, Usova A, Kudabaeva M, Anan’ina T, Wasserlauf I, Pashkevich V, Moshkina M, Obukhovskaya V, Kataeva N, Levina A, Tumentceva Y, Yarnykh V. Age-Related Decline in Brain Myelination: Quantitative Macromolecular Proton Fraction Mapping, T2-FLAIR Hyperintensity Volume, and Anti-Myelin Antibodies Seven Years Apart. Biomedicines 2023; 12:61. [PMID: 38255168 PMCID: PMC10812983 DOI: 10.3390/biomedicines12010061] [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: 11/07/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Age-related myelination decrease is considered one of the likely mechanisms of cognitive decline. The present preliminary study is based on the longitudinal assessment of global and regional myelination of the normal adult human brain using fast macromolecular fraction (MPF) mapping. Additional markers were age-related changes in white matter (WM) hyperintensities on FLAIR-MRI and the levels of anti-myelin autoantibodies in serum. Eleven healthy subjects (33-60 years in the first study) were scanned twice, seven years apart. An age-related decrease in MPF was found in global WM, grey matter (GM), and mixed WM-GM, as well as in 48 out of 82 examined WM and GM regions. The greatest decrease in MPF was observed for the frontal WM (2-5%), genu of the corpus callosum (CC) (4.0%), and caudate nucleus (5.9%). The age-related decrease in MPF significantly correlated with an increase in the level of antibodies against myelin basic protein (MBP) in serum (r = 0.69 and r = 0.63 for global WM and mixed WM-GM, correspondingly). The volume of FLAIR hyperintensities increased with age but did not correlate with MPF changes and the levels of anti-myelin antibodies. MPF mapping showed high sensitivity to age-related changes in brain myelination, providing the feasibility of this method in clinics.
Collapse
Affiliation(s)
- Marina Khodanovich
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
| | - Mikhail Svetlik
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
| | - Anna Naumova
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
- Department of Radiology, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Daria Kamaeva
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia;
| | - Anna Usova
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 12/1 Savinykh St., Tomsk 634009, Russia;
| | - Marina Kudabaeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
| | - Tatyana Anan’ina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
| | - Irina Wasserlauf
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
| | - Valentina Pashkevich
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
| | - Marina Moshkina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
| | - Victoria Obukhovskaya
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
- Department of Fundamental Psychology and Behavioral Medicine, Siberian State Medical University, 2 Moskovskiy Trakt, Tomsk 634050, Russia
| | - Nadezhda Kataeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
- Department of Neurology and Neurosurgery, Siberian State Medical University, 2 Moskovskiy Trakt, Tomsk 634050, Russia
| | - Anastasia Levina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
- Medica Diagnostic and Treatment Center, 86 Sovetskaya st., Tomsk 634510, Russia
| | - Yana Tumentceva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia; (M.S.); (A.N.); (M.K.); (T.A.); (I.W.); (N.K.); (A.L.); (Y.T.)
| | - Vasily Yarnykh
- Department of Radiology, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| |
Collapse
|
22
|
Cozzolino F, Canè L, Gatto MC, Iacobucci I, Sacchettino L, De Biase D, Di Napoli E, Paciello O, Avallone L, Monti M, d’Angelo D, Napolitano F. Proteomic signature profiling in the cortex of dairy cattle unravels the physiology of brain aging. Front Aging Neurosci 2023; 15:1277546. [PMID: 38131010 PMCID: PMC10733460 DOI: 10.3389/fnagi.2023.1277546] [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: 08/14/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction Aging is a physiological process occurring in all living organisms. It is characterized by a progressive deterioration of the physiological and cognitive functions of the organism, accompanied by a gradual impairment of mechanisms involved in the regulation of tissue and organ homeostasis, thus exacerbating the risk of developing pathologies, including cancer and neurodegenerative disorders. Methods In the present work, for the first time, the influence of aging has been investigated in the brain cortex of the Podolica cattle breed, through LC-MS/MS-based differential proteomics and the bioinformatic analysis approach (data are available via ProteomeXchange with identifier PXD044108), with the aim of identifying potential aging or longevity markers, also associated with a specific lifestyle. Results and discussion We found a significant down-regulation of proteins involved in cellular respiration, dendric spine development, synaptic vesicle transport, and myelination. On the other hand, together with a reduction of the neurofilament light chain, we observed an up-regulation of both GFAP and vimentin in the aged samples. In conclusion, our data pave the way for a better understanding of molecular mechanisms underlying brain aging in grazing cattle, which could allow strategies to be developed that are aimed at improving animal welfare and husbandry practices of dairy cattle from intensive livestock.
Collapse
Affiliation(s)
- Flora Cozzolino
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”-Via G. Salvatore, Naples, Italy
- Department of Chemical Sciences, University of Naples, Naples, Italy
| | - Luisa Canè
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”-Via G. Salvatore, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Maria Claudia Gatto
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”-Via G. Salvatore, Naples, Italy
| | - Ilaria Iacobucci
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”-Via G. Salvatore, Naples, Italy
- Department of Chemical Sciences, University of Naples, Naples, Italy
| | - Luigi Sacchettino
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Davide De Biase
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Evaristo Di Napoli
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Maria Monti
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”-Via G. Salvatore, Naples, Italy
- Department of Chemical Sciences, University of Naples, Naples, Italy
| | - Danila d’Angelo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Francesco Napolitano
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”-Via G. Salvatore, Naples, Italy
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| |
Collapse
|
23
|
Aberra AS, Wang R, Grill WM, Peterchev AV. Multi-scale model of axonal and dendritic polarization by transcranial direct current stimulation in realistic head geometry. Brain Stimul 2023; 16:1776-1791. [PMID: 38056825 PMCID: PMC10842743 DOI: 10.1016/j.brs.2023.11.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/06/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Transcranial direct current stimulation (tDCS) is a non-invasive brain stimulation modality that can alter cortical excitability. However, it remains unclear how the subcellular elements of different neuron types are polarized by specific electric field (E-field) distributions. OBJECTIVE To quantify neuronal polarization generated by tDCS in a multi-scale computational model. METHODS We embedded layer-specific, morphologically-realistic cortical neuron models in a finite element model of the E-field in a human head and simulated steady-state polarization generated by conventional primary-motor-cortex-supraorbital (M1-SO) and 4 × 1 high-definition (HD) tDCS. We quantified somatic, axonal, and dendritic polarization of excitatory pyramidal cells in layers 2/3, 5, and 6, as well as inhibitory interneurons in layers 1 and 4 of the hand knob. RESULTS Axonal and dendritic terminals were polarized more than the soma in all neurons, with peak axonal and dendritic polarization of 0.92 mV and 0.21 mV, respectively, compared to peak somatic polarization of 0.07 mV for 1.8 mA M1-SO stimulation. Both montages generated regions of depolarization and hyperpolarization beneath the M1 anode; M1-SO produced slightly stronger, more diffuse polarization peaking in the central sulcus, while 4 × 1 HD produced higher peak polarization in the gyral crown. The E-field component normal to the cortical surface correlated strongly with pyramidal neuron somatic polarization (R2>0.9), but exhibited weaker correlations with peak pyramidal axonal and dendritic polarization (R2:0.5-0.9) and peak polarization in all subcellular regions of interneurons (R2:0.3-0.6). Simulating polarization by uniform local E-field extracted at the soma approximated the spatial distribution of tDCS polarization but produced large errors in some regions (median absolute percent error: 7.9 %). CONCLUSIONS Polarization of pre- and postsynaptic compartments of excitatory and inhibitory cortical neurons may play a significant role in tDCS neuromodulation. These effects cannot be predicted from the E-field distribution alone but rather require calculation of the neuronal response.
Collapse
Affiliation(s)
- Aman S Aberra
- Dept. of Biomedical Engineering, Pratt School of Engineering, Duke University, NC, USA.
| | - Ruochen Wang
- Dept. of Biomedical Engineering, Pratt School of Engineering, Duke University, NC, USA; Dept. of Psychiatry and Behavioral Sciences, School of Medicine, Duke University, NC, USA.
| | - Warren M Grill
- Dept. of Biomedical Engineering, Pratt School of Engineering, Duke University, NC, USA; Dept. of Electrical and Computer Engineering, Pratt School of Engineering, Duke University, NC, USA; Dept. of Neurobiology, School of Medicine, Duke University, NC, USA; Dept. of Neurosurgery, School of Medicine, Duke University, NC, USA.
| | - Angel V Peterchev
- Dept. of Biomedical Engineering, Pratt School of Engineering, Duke University, NC, USA; Dept. of Psychiatry and Behavioral Sciences, School of Medicine, Duke University, NC, USA; Dept. of Electrical and Computer Engineering, Pratt School of Engineering, Duke University, NC, USA; Dept. of Neurosurgery, School of Medicine, Duke University, NC, USA.
| |
Collapse
|
24
|
Festa LK, Clyde AE, Long CC, Roth LM, Grinspan JB, Jordan-Sciutto KL. Antiretroviral treatment reveals a novel role for lysosomes in oligodendrocyte maturation. J Neurochem 2023; 165:722-740. [PMID: 36718947 PMCID: PMC10724866 DOI: 10.1111/jnc.15773] [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: 12/12/2022] [Accepted: 01/24/2023] [Indexed: 02/01/2023]
Abstract
White matter deficits are a common neuropathologic finding in neurologic disorders, including HIV-associated neurocognitive disorders (HAND). In HAND, the persistence of white matter alterations despite suppressive antiretroviral (ARV) therapy suggests that ARVs may be directly contributing to these impairments. Here, we report that a frontline ARV, bictegravir (BIC), significantly attenuates remyelination following cuprizone-mediated demyelination, a model that recapitulates acute demyelination, but has no impact on already formed mature myelin. Mechanistic studies utilizing primary rat oligodendrocyte precursor cells (OPCs) revealed that treatment with BIC leads to significant decrease in mature oligodendrocytes accompanied by lysosomal deacidification and impairment of lysosomal degradative capacity with no alterations in lysosomal membrane permeability or total lysosome number. Activation of the endolysosomal cation channel TRPML1 prevents both lysosomal deacidification and impairment of oligodendrocyte differentiation by BIC. Lastly, we show that deacidification of lysosomes by compounds that raise lysosomal pH is sufficient to prevent maturation of oligodendrocytes. Overall, this study has uncovered a critical role for lysosomal acidification in modulating oligodendrocyte function and has implications for neurologic diseases characterized by lysosomal dysfunction and white matter abnormalities.
Collapse
Affiliation(s)
- Lindsay K. Festa
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Abigail E. Clyde
- School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| | - Caela C. Long
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | | | - Judith B. Grinspan
- Department of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Kelly L. Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
25
|
Schlett JS, Mettang M, Skaf A, Schweizer P, Errerd A, Mulugeta EA, Hein TM, Tsesmelis K, Tsesmelis M, Büttner UFG, Wendt H, Abaei A, Rasche V, Prex V, Nespoli E, Alami NO, Tews D, Walther P, Yilmazer-Hanke D, Oswald F, Dimou L, Wirth T, Baumann B. NF-κB is a critical mediator of post-mitotic senescence in oligodendrocytes and subsequent white matter loss. Mol Neurodegener 2023; 18:24. [PMID: 37069623 PMCID: PMC10108549 DOI: 10.1186/s13024-023-00616-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/25/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Inflammaging represents an accepted concept where the immune system shifts to a low-grade chronic pro-inflammatory state without overt infection upon aging. In the CNS, inflammaging is mainly driven by glia cells and associated with neurodegenerative processes. White matter degeneration (WMD), a well-known process in the aging brain, manifests in myelin loss finally resulting in motor, sensory and cognitive impairments. Oligodendrocytes (OL) are responsible for homeostasis and maintenance of the myelin sheaths, which is a complex and highly energy demanding process sensitizing these cells to metabolic, oxidative and other forms of stress. Yet, the immediate impact of chronic inflammatory stress like inflammaging on OL homeostasis, myelin maintenance and WMD remains open. METHODS To functionally analyze the role of IKK/NF-κB signaling in the regulation of myelin homeostasis and maintenance in the adult CNS, we established a conditional mouse model allowing NF-κB activation in mature myelinating oligodendrocytes. IKK2-CAPLP-CreERT2 mice were characterized by biochemical, immunohistochemical, ultrastructural and behavioral analyses. Transcriptome data from isolated, primary OLs and microglia cells were explored by in silico pathway analysis and validated by complementary molecular approaches. RESULTS Chronic NF-κB activation in mature OLs leads to aggravated neuroinflammatory conditions phenocopying brain inflammaging. As a consequence, IKK2-CAPLP-CreERT2 mice showed specific neurological deficits and impaired motoric learning. Upon aging, persistent NF-κB signaling promotes WMD in these mice as ultrastructural analysis revealed myelination deficits in the corpus callosum accompanied by impaired myelin protein expression. RNA-Seq analysis of primary oligodendrocytes and microglia cells uncovers gene expression signatures associated with activated stress responses and increased post mitotic cellular senescence (PoMiCS) which was confirmed by elevated senescence-associated β-galactosidase activity and SASP gene expression profile. We identified an elevated integrated stress response (ISR) characterized by phosphorylation of eIF2α as a relevant molecular mechanism which is able to affect translation of myelin proteins. CONCLUSIONS Our findings demonstrate an essential role of IKK/NF-κB signaling in mature, post-mitotic OLs in regulating stress-induced senescence in these cells. Moreover, our study identifies PoMICS as an important driving force of age-dependent WMD as well as of traumatic brain injury induced myelin defects.
Collapse
Affiliation(s)
- Judith Stefanie Schlett
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Melanie Mettang
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Aladdin Skaf
- Molecular and Translational Neuroscience, Department of Neurology, University Medical Center Ulm, 89081, Ulm, Germany
| | - Pavel Schweizer
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Alina Errerd
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | | | - Tabea Melissa Hein
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Konstantinos Tsesmelis
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Miltiadis Tsesmelis
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Ulrike F G Büttner
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Heinrich Wendt
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Alireza Abaei
- Core Facility Small Animal Imaging (CF-SANI), Ulm University, 89081, Ulm, Germany
| | - Volker Rasche
- Core Facility Small Animal Imaging (CF-SANI), Ulm University, 89081, Ulm, Germany
| | - Vivien Prex
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Ester Nespoli
- Molecular and Translational Neuroscience, Department of Neurology, University Medical Center Ulm, 89081, Ulm, Germany
| | - Najwa Ouali Alami
- Institute of Clinical Neuroanatomy, Ulm University, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Daniel Tews
- Core Facility Extracellular Flux Analyzer, Ulm University Medical Center, 89081, Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Deniz Yilmazer-Hanke
- Institute of Clinical Neuroanatomy, Ulm University, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Franz Oswald
- Department of Internal Medicine I, Center for Internal Medicine, University Medical Center Ulm, 89081, Ulm, Germany
| | - Leda Dimou
- Molecular and Translational Neuroscience, Department of Neurology, University Medical Center Ulm, 89081, Ulm, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
26
|
Chapman TW, Olveda GE, Bame X, Pereira E, Hill RA. Oligodendrocyte death initiates synchronous remyelination to restore cortical myelin patterns in mice. Nat Neurosci 2023; 26:555-569. [PMID: 36928635 PMCID: PMC10208560 DOI: 10.1038/s41593-023-01271-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/09/2023] [Indexed: 03/18/2023]
Abstract
Myelin degeneration occurs in neurodegenerative diseases and aging. In these conditions, resident oligodendrocyte progenitor cells (OPCs) differentiate into oligodendrocytes that carry out myelin repair. To investigate the cellular dynamics underlying these events, we developed a noninflammatory demyelination model that combines intravital two-photon imaging with a single-cell ablation technique called two-photon apoptotic targeted ablation (2Phatal). Oligodendrocyte 2Phatal in both sexes results in a myelin degeneration cascade that triggers rapid forms of synchronous remyelination on defined axons. This remyelination is driven by oligodendrocytes differentiated from a subset of morphologically distinct, highly branched OPCs. Moreover, remyelination efficiency depends on the initial myelin patterns, as well as the age of the organism. In summary, using 2Phatal, we show a form of rapid synchronous remyelination, mediated by a distinct subset of OPCs, capable of restoring the original myelin patterning in adulthood but not aging.
Collapse
Affiliation(s)
- Timothy W Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Genaro E Olveda
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Xhoela Bame
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Elizabeth Pereira
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
27
|
Zhi JJ, Wu SL, Wu HQ, Ran Q, Gao X, Chen JF, Gu XM, Li T, Wang F, Xiao L, Ye J, Mei F. Insufficient Oligodendrocyte Turnover in Optic Nerve Contributes to Age-Related Axon Loss and Visual Deficits. J Neurosci 2023; 43:1859-1870. [PMID: 36725322 PMCID: PMC10027114 DOI: 10.1523/jneurosci.2130-22.2023] [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: 11/16/2022] [Revised: 01/16/2023] [Accepted: 01/27/2023] [Indexed: 02/03/2023] Open
Abstract
Age-related decline in visual functions is a prevalent health problem among elderly people, and no effective therapies are available up-to-date. Axon degeneration and myelin loss in optic nerves (ONs) are age-dependent and become evident in middle-aged (13-18 months) and old (20-22 months) mice of either sex compared with adult mice (3-8 months), accompanied by functional deficits. Oligodendrocyte (OL) turnover is actively going on in adult ONs. However, the longitudinal change and functional significance of OL turnover in aging ONs remain largely unknown. Here, using cell-lineage labeling and tracing, we reported that oligodendrogenesis displayed an age-dependent decrease in aging ONs. To understand whether active OL turnover is required for maintaining axons and visual function, we conditionally deleted the transcription factor Olig2 in the oligodendrocyte precursor cells of young mice. Genetically dampening OL turnover by Olig2 ablation resulted in accelerated axon loss and retinal degeneration, and subsequently impaired ON signal transmission, suggesting that OL turnover is an important mechanism to sustain axon survival and visual function. To test whether enhancing oligodendrogenesis can prevent age-related visual deficits, 12-month-old mice were treated with clemastine, a pro-myelination drug, or induced deletion of the muscarinic receptor 1 in oligodendrocyte precursor cells. The clemastine treatment or muscarinic receptor 1 deletion significantly increased new OL generation in the aged ONs and consequently preserved visual function and retinal integrity. Together, our data indicate that dynamic OL turnover in ONs is required for axon survival and visual function, and enhancing new OL generation represents a potential approach to reversing age-related declines of visual function.SIGNIFICANCE STATEMENT Oligodendrocyte (OL) turnover has been reported in adult optic nerves (ONs), but the longitudinal change and functional significance of OL turnover during aging remain largely unknown. Using cell-lineage tracing and oligodendroglia-specific manipulation, this study reported that OL generation was active in adult ONs and the efficiency decreased in an age-dependent manner. Genetically dampening OL generation by Olig2 ablation resulted in significant axon loss and retinal degeneration, along with delayed visual signal transmission. Conversely, pro-myelination approaches significantly increased new myelin generation in aging ONs, and consequently preserved retinal integrity and visual function. Our findings indicate that promoting OL generation might be a promising strategy to preserve visual function from age-related decline.
Collapse
Affiliation(s)
- Jun-Jie Zhi
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shuang-Ling Wu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Hao-Qian Wu
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qi Ran
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xing Gao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jing-Fei Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xing-Mei Gu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Department of Medical English Teaching and Research, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Tao Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jian Ye
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Feng Mei
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- School of Medicine, Chongqing University, Chongqing, 400030, China
| |
Collapse
|
28
|
Murray CJ, Vecchiarelli HA, Tremblay MÈ. Enhancing axonal myelination in seniors: A review exploring the potential impact cannabis has on myelination in the aged brain. Front Aging Neurosci 2023; 15:1119552. [PMID: 37032821 PMCID: PMC10073480 DOI: 10.3389/fnagi.2023.1119552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/22/2023] [Indexed: 04/11/2023] Open
Abstract
Consumption of cannabis is on the rise as public opinion trends toward acceptance and its consequent legalization. Specifically, the senior population is one of the demographics increasing their use of cannabis the fastest, but research aimed at understanding cannabis' impact on the aged brain is still scarce. Aging is characterized by many brain changes that slowly alter cognitive ability. One process that is greatly impacted during aging is axonal myelination. The slow degradation and loss of myelin (i.e., demyelination) in the brain with age has been shown to associate with cognitive decline and, furthermore, is a common characteristic of numerous neurological diseases experienced in aging. It is currently not known what causes this age-dependent degradation, but it is likely due to numerous confounding factors (i.e., heightened inflammation, reduced blood flow, cellular senescence) that impact the many cells responsible for maintaining overall homeostasis and myelin integrity. Importantly, animal studies using non-human primates and rodents have also revealed demyelination with age, providing a reliable model for researchers to try and understand the cellular mechanisms at play. In rodents, cannabis was recently shown to modulate the myelination process. Furthermore, studies looking at the direct modulatory impact cannabis has on microglia, astrocytes and oligodendrocyte lineage cells hint at potential mechanisms to prevent some of the more damaging activities performed by these cells that contribute to demyelination in aging. However, research focusing on how cannabis impacts myelination in the aged brain is lacking. Therefore, this review will explore the evidence thus far accumulated to show how cannabis impacts myelination and will extrapolate what this knowledge may mean for the aged brain.
Collapse
Affiliation(s)
- Colin J. Murray
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- *Correspondence: Colin J. Murray,
| | | | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Départment de Médicine Moléculaire, Université Laval, Québec City, QC, Canada
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
- Marie-Ève Tremblay,
| |
Collapse
|
29
|
Lee GY, Kim OH, Kim ER, Lee HJ. Biomechanical forces in the aged brain: Relationship to AD. Life Sci 2022; 312:121237. [PMID: 36436618 DOI: 10.1016/j.lfs.2022.121237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
The pathogenesis of neurodegenerative disorders, including Alzheimer's disease, has been studied with a focus on biochemical mechanisms, such as the amyloid-β plaque deposition and removal. Recently, the importance of brain microenvironmental cues, which comprise the sophisticated cellular and fluid system, has been emphasized in the aged brain or in pathological conditions. Especially, substrate rigidity and biomechanical forces of the brain microenvironment determine the function of glial cells and neurons; furthermore, these microenvironmental cues change with age. However, our understanding of role of the biomechanical cues on glial cells and neurons is relatively poor. In this review, we briefly introduce an overview of biomechanical forces that present in the aged brain and its sensations, and then examine the brain in Alzheimer's disease, which constitutes a representative neurodegenerative disorder, with regard to changes in the biomechanical forces associated with disease and aging.
Collapse
Affiliation(s)
- Gyeong Yun Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Ok-Hyeon Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Eun Ran Kim
- Division of Endocrine and Kidney Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea.
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Tang Y, Yu J, Zhou M, Li J, Long T, Li Y, Feng L, Chen D, Yang Z, Huang Y, Hu S. Cortical abnormalities of synaptic vesicle protein 2A in focal cortical dysplasia type II identified in vivo with 18F-SynVesT-1 positron emission tomography imaging. Eur J Nucl Med Mol Imaging 2022; 49:3482-3491. [PMID: 34978594 PMCID: PMC9308579 DOI: 10.1007/s00259-021-05665-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 12/19/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE The loss of synaptic vesicle glycoprotein 2A (SV2A) is well established as the major correlate of epileptogenesis in focal cortical dysplasia type II (FCD II), but this has not been directly tested in vivo. In this positron emission tomography (PET) study with the new tracer 18F-SynVesT-1, we evaluated SV2A abnormalities in patients with FCD II and compared the pattern to 18F-fluorodeoxyglucose (18F-FDG). METHODS Sixteen patients with proven FCD II and 16 healthy controls were recruited. All FCD II patients underwent magnetic resonance imaging (MRI) and static PET imaging with both 18F-SynVesT-1 and 18F-FDG, while the controls underwent MRI and PET with only 18F-SynVesT-1. Visual assessment of PET images was undertaken. The standardized uptake values (SUVs) of 18F-SynVesT-1 were computed for regions of interest (ROIs), along with SUV ratio (SUVr) between ROI and centrum semiovale (white matter). Asymmetry indices (AIs) were analyzed between the lesion and the contralateral hemisphere for intersubject comparisons. RESULTS Lesions in the brains of FCD II patients had significantly reduced 18F-SynVesT-1 uptake compared with contralateral regions, and brains of the controls. 18F-SynVesT-1 PET indicated low lesion uptake in 14 patients (87.5%), corresponding to hypometabolism detected by 18F-FDG PET, with higher accuracy for lesion localization than MRI (43.8%) (P < 0.05). AI analyses demonstrated that in the lesions, SUVr for each of the radiotracers were not significantly different (P > 0.05), and 18F-SynVesT-1 SUVr correlated with that of 18F-FDG across subjects (R2 = 0.41, P = 0.008). Subsequent visual ratings indicated that 18F-SynVesT-1 uptake had a more restricted pattern of reduction than 18F-FDG uptake in FCD II lesions (P < 0.05). CONCLUSION SV2A PET with 18F-SynVesT-1 shows a higher accuracy for the localization of FCD II lesions than MRI and a more restricted pattern of abnormality than 18F-FDG PET.
Collapse
Affiliation(s)
- Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Jie Yu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Jian Li
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Tingting Long
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yulai Li
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Li Feng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dengming Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Zhiquan Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, P.O. Box 208048, New Haven, CT, 06520-8048, USA.
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya), Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
31
|
Miyanishi H, Kitazawa A, Izuo N, Muramatsu SI, Nitta A. N-Acetyl Transferase, Shati/Nat8l, in the Dorsal Hippocampus Suppresses Aging-induced Impairment of Cognitive Function in Mice. Neurochem Res 2022; 47:2703-2714. [DOI: 10.1007/s11064-022-03594-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/02/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
|
32
|
Mohamed WMY, Yi C, Soreq L, Yamashita T. Editorial: Genes and Aging: From Bench-to-Bedside. Front Aging Neurosci 2022; 14:886967. [PMID: 35431892 PMCID: PMC9006944 DOI: 10.3389/fnagi.2022.886967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Wael M. Y. Mohamed
- Department of Basic Medical Science, International Islamic University Malaysia, Selayang, Malaysia
- Department of Clinical Pharmacology, Menoufia University, Shebeen El-Kom, Egypt
| | - Chenju Yi
- Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Lilach Soreq
- UCL Institute of Neurology, University College London (UCL), London, United Kingdom
| | | |
Collapse
|
33
|
Chen B, Linke A, Olson L, Kohli J, Kinnear M, Sereno M, Müller RA, Carper R, Fishman I. Cortical Myelination in Toddlers and Preschoolers with Autism Spectrum Disorder. Dev Neurobiol 2022; 82:261-274. [PMID: 35348301 PMCID: PMC9325547 DOI: 10.1002/dneu.22874] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 02/22/2022] [Accepted: 03/17/2022] [Indexed: 11/07/2022]
Abstract
Intracortical myelin is thought to play a significant role in the development of neural circuits and functional networks, with consistent evidence of atypical network connectivity in children with autism spectrum disorders (ASD). However, little is known about the development of intracortical myelin in the first years of life in ASD, during the critical neurodevelopmental period when autism symptoms first emerge. Using T1-weighted (T1w) and T2-weighted (T2w) structural magnetic resonance imaging (MRI) in 21 young children with ASD and 16 typically developing (TD) children, ages 1.5 to 5.5 years, we demonstrate the feasibility of estimating intracortical myelin in vivo using the T1w/T2w ratio as a proxy. The resultant T1w/T2w maps were largely comparable with those reported in prior T1w/T2w studies in typically developing children and adults, and revealed no group differences between TD children and those with ASD. However, differential associations between T1w/T2w and age were identified in several early myelinated regions (e.g., visual, posterior cingulate, precuneus cortices) in the ASD and TD groups, with age-related increase in estimated myelin content across the toddler and preschool years detected in TD children, but not in children with ASD. The atypical age-related effects in intracortical myelin, suggesting a disrupted myelination in the first years of life in ASD, may be related to the aberrant brain network connectivity reported in young children with ASD in some of the same cortical regions and circuits. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bosi Chen
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University.,Joint Doctoral Program in Clinical Psychology, San Diego State University and University of California, San Diego, USA
| | - Annika Linke
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University
| | - Lindsay Olson
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University.,Joint Doctoral Program in Clinical Psychology, San Diego State University and University of California, San Diego, USA
| | - Jiwandeep Kohli
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University.,Joint Doctoral Program in Clinical Psychology, San Diego State University and University of California, San Diego, USA
| | - Mikaela Kinnear
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University
| | - Martin Sereno
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University.,Joint Doctoral Program in Clinical Psychology, San Diego State University and University of California, San Diego, USA
| | - Ralph-Axel Müller
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University.,Joint Doctoral Program in Clinical Psychology, San Diego State University and University of California, San Diego, USA.,Center for Autism and Developmental Disorders, San Diego State University
| | - Ruth Carper
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University.,Joint Doctoral Program in Clinical Psychology, San Diego State University and University of California, San Diego, USA.,Center for Autism and Developmental Disorders, San Diego State University
| | - Inna Fishman
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University.,Joint Doctoral Program in Clinical Psychology, San Diego State University and University of California, San Diego, USA.,Center for Autism and Developmental Disorders, San Diego State University
| |
Collapse
|
34
|
Ma XR, Zhu X, Xiao Y, Gu HM, Zheng SS, Li L, Wang F, Dong ZJ, Wang DX, Wu Y, Yang C, Jiang W, Yao K, Yin Y, Zhang Y, Peng C, Gao L, Meng Z, Hu Z, Liu C, Li L, Chen HZ, Shu Y, Ju Z, Zhao JW. Restoring nuclear entry of Sirtuin 2 in oligodendrocyte progenitor cells promotes remyelination during ageing. Nat Commun 2022; 13:1225. [PMID: 35264567 PMCID: PMC8907257 DOI: 10.1038/s41467-022-28844-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 02/15/2022] [Indexed: 11/18/2022] Open
Abstract
The age-dependent decline in remyelination potential of the central nervous system during ageing is associated with a declined differentiation capacity of oligodendrocyte progenitor cells (OPCs). The molecular players that can enhance OPC differentiation or rejuvenate OPCs are unclear. Here we show that, in mouse OPCs, nuclear entry of SIRT2 is impaired and NAD+ levels are reduced during ageing. When we supplement β-nicotinamide mononucleotide (β-NMN), an NAD+ precursor, nuclear entry of SIRT2 in OPCs, OPC differentiation, and remyelination were rescued in aged animals. We show that the effects on myelination are mediated via the NAD+-SIRT2-H3K18Ac-ID4 axis, and SIRT2 is required for rejuvenating OPCs. Our results show that SIRT2 and NAD+ levels rescue the aged OPC differentiation potential to levels comparable to young age, providing potential targets to enhance remyelination during ageing.
Collapse
Affiliation(s)
- Xiao-Ru Ma
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Center for Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Xudong Zhu
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yujie Xiao
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 200032, Shanghai, China
| | - Hui-Min Gu
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Center for Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Shuang-Shuang Zheng
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Center for Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Liang Li
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, 200032, Shanghai, China
| | - Fan Wang
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Center for Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Zhao-Jun Dong
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Center for Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Di-Xian Wang
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Center for Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Yang Wu
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Center for Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Chenyu Yang
- Center of Cryo-Electron Microscopy, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Wenhong Jiang
- Zhejiang University School of Brain Science and Brain Medicine, and Department of Neurosurgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Ke Yao
- School of Pharmaceutical Sciences, Tsinghua University, 100084, Beijing, China
| | - Yue Yin
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, 201210, Shanghai, China
| | - Yang Zhang
- Department of cardiology, Zhongshan Hospital of Fudan University, 200035, Shanghai, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, 201210, Shanghai, China
| | - Lixia Gao
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, 310020, Hangzhou, China
| | - Zhuoxian Meng
- Department of Pathology and Pathophysiology and Zhejiang Provincial Key Laboratory of Pancreatic Disease of the First Affiliated Hospital, Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences and Beijing Frontier Research Center for Biological Structure, Tsinghua University, 100084, Beijing, China
| | - Chong Liu
- Zhejiang University School of Brain Science and Brain Medicine, and Department of Neurosurgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Li Li
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 100005, Beijing, China.
| | - Yousheng Shu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, 100875, Beijing, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| | - Jing-Wei Zhao
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Center for Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.
- Center of Cryo-Electron Microscopy, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
35
|
Dhiman S, Fountain-Zaragoza S, Jensen JH, Falangola MF, McKinnon ET, Moss HG, Thorn KE, Rieter WJ, Spampinato MV, Nietert PJ, Helpern JA, Benitez A. Fiber Ball White Matter Modeling Reveals Microstructural Alterations in Healthy Brain Aging. AGING BRAIN 2022; 2:100037. [PMID: 36324695 PMCID: PMC9624504 DOI: 10.1016/j.nbas.2022.100037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Age-related white matter degeneration is characterized by myelin breakdown and neuronal fiber loss that preferentially occur in regions that myelinate later in development. Conventional diffusion MRI (dMRI) has demonstrated age-related increases in diffusivity but provide limited information regarding the tissue-specific changes driving these effects. A recently developed dMRI biophysical modeling technique, Fiber Ball White Matter (FBWM) modeling, offers enhanced biological interpretability by estimating microstructural properties specific to the intra-axonal and extra-axonal spaces. We used FBWM to illustrate the biological mechanisms underlying changes throughout white matter in healthy aging using data from 63 cognitively unimpaired adults ages 45-85 with no radiological evidence of neurodegeneration or incipient Alzheimer's disease. Conventional dMRI and FBWM metrics were computed for two late-myelinating (genu of the corpus callosum and association tracts) and two early-myelinating regions (splenium of the corpus callosum and projection tracts). We examined the associations between age and these metrics in each region and tested whether age was differentially associated with these metrics in late- vs. early-myelinating regions. We found that conventional metrics replicated patterns of age-related increases in diffusivity in late-myelinating regions. FBWM additionally revealed specific intra- and extra-axonal changes suggestive of myelin breakdown and preferential loss of smaller-diameter axons, yielding in vivo corroboration of findings from histopathological studies of aged brains. These results demonstrate that advanced biophysical modeling approaches, such as FBWM, offer novel information about the microstructure-specific alterations contributing to white matter changes in healthy aging. These tools hold promise as sensitive indicators of early pathological changes related to neurodegenerative disease.
Collapse
Affiliation(s)
- Siddhartha Dhiman
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Stephanie Fountain-Zaragoza
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA.,Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
| | - Jens H Jensen
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA.,Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA.,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Maria Fatima Falangola
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA.,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Emilie T McKinnon
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA.,Department of Neurology, Medical University of South Carolina, Charleston, SC, USA.,Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA
| | - Hunter G Moss
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA.,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Kathryn E Thorn
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - William J Rieter
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA
| | - Maria Vittoria Spampinato
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA
| | - Paul J Nietert
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Joseph A Helpern
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA.,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Andreana Benitez
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA.,Department of Neurology, Medical University of South Carolina, Charleston, SC, USA.,Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
36
|
Zhang X, Huang N, Xiao L, Wang F, Li T. Replenishing the Aged Brains: Targeting Oligodendrocytes and Myelination? Front Aging Neurosci 2021; 13:760200. [PMID: 34899272 PMCID: PMC8656359 DOI: 10.3389/fnagi.2021.760200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Aging affects almost all the aspects of brain functions, but the mechanisms remain largely undefined. Increasing number of literatures have manifested the important role of glial cells in regulating the aging process. Oligodendroglial lineage cell is a major type of glia in central nervous system (CNS), composed of mature oligodendrocytes (OLs), and oligodendroglia precursor cells (OPCs). OLs produce myelin sheaths that insulate axons and provide metabolic support to meet the energy demand. OPCs maintain the population throughout lifetime with the abilities to proliferate and differentiate into OLs. Increasing evidence has shown that oligodendroglial cells display active dynamics in adult and aging CNS, which is extensively involved in age-related brain function decline in the elderly. In this review, we summarized present knowledge about dynamic changes of oligodendroglial lineage cells during normal aging and discussed their potential roles in age-related functional decline. Especially, focused on declined myelinogenesis during aging and underlying mechanisms. Clarifying those oligodendroglial changes and their effects on neurofunctional decline may provide new insights in understanding aging associated brain function declines.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Ophthalmology, The General Hospital of Western Theater Command, Chengdu, China
| | - Nanxin Huang
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lan Xiao
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fei Wang
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Tao Li
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
37
|
Porcu M, Cocco L, Puig J, Mannelli L, Yang Q, Suri JS, Defazio G, Saba L. Global Fractional Anisotropy: Effect on Resting-state Neural Activity and Brain Networking in Healthy Participants. Neuroscience 2021; 472:103-115. [PMID: 34364954 DOI: 10.1016/j.neuroscience.2021.07.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022]
Abstract
The global fractional anisotropy (gFA) is a structural marker of white matter myelination and integrity. Previous studies already evidenced that aging-related reduced integrity of specific white matter tracts is associated with decreased functional connectivity in several hubs. However, the correlations between gFA and functional brain connectivity remain unknown. In this cross-sectional study, we analyzed structural and functional MR datasets of 79 healthy participants from the Leipzig Study for Mind-Body-Emotion Interactions. DTI model-based method was used to quantify gFA values. We tested associations between gFA, age, and gender. The fractional amplitude of low-frequency fluctuations (fALFF) and ROI-to-ROI connectivity were analyzed in a regression model for evaluating the effects of gFA on brain activity and networking, respectively. A negative correlation was found between gFA and age (ρ = -0.343; p = 0.002). No statistically significant correlation as found between gFA and gender (p = 0.229). Higher values of gFA were associated with increased brain regional activity, including areas of the default mode network. There was a higher degree of correlation between some regions, particularly those that conform to the limbic system. Our study demonstrates that gFA influences regional neural activity and brain networking on resting, particularly the limbic system.
Collapse
Affiliation(s)
- Michele Porcu
- Department of Radiology, AOU Cagliari, University of Cagliari, Italy.
| | - Luigi Cocco
- Department of Radiology, AOU Cagliari, University of Cagliari, Italy
| | - Josep Puig
- Department of Radiology (IDI) and Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr Josep Trueta, Girona, Spain
| | | | - Qi Yang
- Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing, China
| | - Jasjit S Suri
- Stroke Diagnosis and Monitoring Division, AtheroPoint™, Roseville, CA, USA
| | - Giovanni Defazio
- Department of Neurology, University of Cagliari, Cagliari, Italy
| | - Luca Saba
- Department of Radiology, AOU Cagliari, University of Cagliari, Italy
| |
Collapse
|
38
|
Yamazaki Y, Abe Y, Fujii S, Tanaka KF. Oligodendrocytic Na +-K +-Cl - co-transporter 1 activity facilitates axonal conduction and restores plasticity in the adult mouse brain. Nat Commun 2021; 12:5146. [PMID: 34446732 PMCID: PMC8390751 DOI: 10.1038/s41467-021-25488-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 08/13/2021] [Indexed: 11/30/2022] Open
Abstract
The juvenile brain presents plasticity. Oligodendrocytes are the myelinating cells of the central nervous system and myelination can be adaptive. Plasticity decreases from juvenile to adulthood. The mechanisms involving oligodendrocytes underlying plasticity are unclear. Here, we show Na+-K+-Cl– co-transporter 1 (NKCC1), highly expressed in the juvenile mouse brain, regulates the oligodendrocyte activity from juvenile to adulthood in mice, as shown by optogenetic manipulation of oligodendrocytes. The reduced neuronal activity in adults was restored by Nkcc1 overexpression in oligodendrocytes. Moreover, in adult mice overexpressing Nkcc1, long-term potentiation and learning were facilitated compared to age-matched controls. These findings demonstrate that NKCC1 plays a regulatory role in the age-dependent activity of oligodendrocytes, furthermore inducing activation of NKCC1 in oligodendrocytes can restore neuronal plasticity in the adult mouse brain. Brain plasticity declines with age. Here, the authors show that NKCC1 regulates oligodendrocyte activity, facilitating neuronal plasticity during juvenile. Inducing activation of oligodendrocytic NKCC1 results in restoration of neuronal plasticity in the adult mouse brain.
Collapse
Affiliation(s)
- Yoshihiko Yamazaki
- Department of Physiology, Yamagata University School of Medicine, Yamagata, Japan.
| | - Yoshifumi Abe
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Fujii
- Department of Physiology, Yamagata University School of Medicine, Yamagata, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
39
|
Matijevic S, Ryan L. Tract Specificity of Age Effects on Diffusion Tensor Imaging Measures of White Matter Health. Front Aging Neurosci 2021; 13:628865. [PMID: 33790778 PMCID: PMC8006297 DOI: 10.3389/fnagi.2021.628865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/11/2021] [Indexed: 11/13/2022] Open
Abstract
Well-established literature indicates that older adults have poorer cerebral white matter integrity, as measured through diffusion tensor imaging (DTI). Age differences in DTI have been observed widely across white matter, although some tracts appear more sensitive to the effects of aging than others. Factors like APOE ε4 status and sex may contribute to individual differences in white matter integrity that also selectively impact certain tracts, and could influence DTI changes in aging. The present study explored the degree to which age, APOE ε4, and sex exerted global vs. tract specific effects on DTI metrics in cognitively healthy late middle-aged to older adults. Data from 49 older adults (ages 54–92) at two time-points separated by approximately 2.7 years were collected. DTI metrics, including fractional anisotropy (FA) and mean diffusivity (MD), were extracted from nine white matter tracts and global white matter. Results showed that across timepoints, FA and MD increased globally, with no tract-specific changes observed. Baseline age had a global influence on both measures, with increasing age associated with lower FA and higher MD. After controlling for global white matter FA, age additionally predicted FA for the genu, callosum body, inferior fronto-occipital fasciculus (IFOF), and both anterior and posterior cingulum. Females exhibited lower global FA on average compared to males. In contrast, MD was selectively elevated in the anterior cingulum and superior longitudinal fasciculus (SLF), for females compared to males. APOE ε4 status was not predictive of either measure. In summary, these results indicate that age and sex are associated with both global and tract-specific alterations to DTI metrics among a healthy older adult cohort. Older women have poorer white matter integrity compared to older men, perhaps related to menopause-induced metabolic changes. While age-related alterations to white matter integrity are global, there is substantial variation in the degree to which tracts are impacted, possibly as a consequence of tract anatomical variability. The present study highlights the importance of accounting for global sources of variation in DTI metrics when attempting to investigate individual differences (due to age, sex, or other factors) in specific white matter tracts.
Collapse
Affiliation(s)
- Stephanie Matijevic
- Cognition and Neuroimaging Laboratory, Department of Psychology, University of Arizona, Tucson, AZ, United States
| | - Lee Ryan
- Cognition and Neuroimaging Laboratory, Department of Psychology, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
40
|
Naffaa V, Laprévote O, Schang AL. Effects of endocrine disrupting chemicals on myelin development and diseases. Neurotoxicology 2020; 83:51-68. [PMID: 33352275 DOI: 10.1016/j.neuro.2020.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
In the central and peripheral nervous systems, myelin is essential for efficient conduction of action potentials. During development, oligodendrocytes and Schwann cells differentiate and ensure axon myelination, and disruption of these processes can contribute to neurodevelopmental disorders. In adults, demyelination can lead to important disabilities, and recovery capacities by remyelination often decrease with disease progression. Among environmental chemical pollutants, endocrine disrupting chemicals (EDCs) are of major concern for human health and are notably suspected to participate in neurodevelopmental and neurodegenerative diseases. In this review, we have combined the current knowledge on EDCs impacts on myelin including several persistent organic pollutants, bisphenol A, triclosan, heavy metals, pesticides, and nicotine. Besides, we presented several other endocrine modulators, including pharmaceuticals and the phytoestrogen genistein, some of which are candidates for treating demyelinating conditions but could also be deleterious as contaminants. The direct impacts of EDCs on myelinating cells were considered as well as their indirect consequences on myelin, particularly on immune mechanisms associated with demyelinating conditions. More studies are needed to describe the effects of these compounds and to further understand the underlying mechanisms in relation to the potential for endocrine disruption.
Collapse
Affiliation(s)
- Vanessa Naffaa
- Université de Paris, UMR 8038 (CiTCoM), CNRS, Faculté de Pharmacie de Paris, 4 avenue de l'Observatoire, 75006 Paris, France.
| | - Olivier Laprévote
- Université de Paris, UMR 8038 (CiTCoM), CNRS, Faculté de Pharmacie de Paris, 4 avenue de l'Observatoire, 75006 Paris, France; Hôpital Européen Georges Pompidou, AP-HP, Service de Biochimie, 20 rue Leblanc, 75015 Paris, France.
| | - Anne-Laure Schang
- Université de Paris, UMR 1153 (CRESS), Faculté de Pharmacie de Paris, 4 avenue de l'Observatoire, 75006 Paris, France.
| |
Collapse
|
41
|
Islam MR, Luo R, Valaris S, Haley EB, Takase H, Chen YI, Dickerson BC, Schon K, Arai K, Nguyen CT, Wrann CD. Diffusion tensor-MRI detects exercise-induced neuroplasticity in the hippocampal microstructure in mice. Brain Plast 2020; 5:147-159. [PMID: 33282678 PMCID: PMC7685674 DOI: 10.3233/bpl-190090] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Despite considerable research on exercise-induced neuroplasticity in the brain, a major ongoing challenge in translating findings from animal studies to humans is that clinical and preclinical settings employ very different techniques. Objective: Here we aim to bridge this divide by using diffusion tensor imaging MRI (DTI), an advanced imaging technique commonly applied in human studies, in a longitudinal exercise study with mice. Methods: Wild-type mice were exercised using voluntary free-wheel running, and MRI scans were at baseline and after four weeks and nine weeks of running. Results: Both hippocampal volume and fractional anisotropy, a surrogate for microstructural directionality, significantly increased with exercise. In addition, exercise levels correlated with effect size. Histological analysis showed more PDGFRα+ oligodendrocyte precursor cells in the corpus callosum of running mice. Conclusions: These results provide compelling in vivo support for the concept that similar adaptive changes occur in the brains of mice and humans in response to exercise.
Collapse
Affiliation(s)
- Mohammad R Islam
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Renhao Luo
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Sophia Valaris
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Erin B Haley
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Hajime Takase
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Yinching Iris Chen
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Bradford C Dickerson
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Karin Schon
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Christopher T Nguyen
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA.,Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Christiane D Wrann
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.,Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
42
|
Mohamedi Y, Fontanil T, Cobo T, Cal S, Obaya AJ. New Insights into ADAMTS Metalloproteases in the Central Nervous System. Biomolecules 2020; 10:biom10030403. [PMID: 32150898 PMCID: PMC7175268 DOI: 10.3390/biom10030403] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Components of the extracellular matrix (ECM) are key players in regulating cellular functions throughout the whole organism. In fact, ECM components not only participate in tissue organization but also contribute to processes such as cellular maintenance, proliferation, and migration, as well as to support for various signaling pathways. In the central nervous system (CNS), proteoglycans of the lectican family, such as versican, aggrecan, brevican, and neurocan, are important constituents of the ECM. In recent years, members of this family have been found to be involved in the maintenance of CNS homeostasis and to participate directly in processes such as the organization of perineural nets, the regulation of brain plasticity, CNS development, brain injury repair, axonal guidance, and even the altering of synaptic responses. ADAMTSs are a family of “A disintegrin and metalloproteinase with thrombospondin motifs” proteins that have been found to be involved in a multitude of processes through the degradation of lecticans and other proteoglycans. Recently, alterations in ADAMTS expression and activity have been found to be involved in neuronal disorders such as stroke, neurodegeneration, schizophrenia, and even Alzheimer’s disease, which in turn may suggest their potential use as therapeutic targets. Herein, we summarize the different roles of ADAMTSs in regulating CNS events through interactions and the degradation of ECM components (more specifically, the lectican family of proteoglycans).
Collapse
Affiliation(s)
- Yamina Mohamedi
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain; (Y.M.); (T.F.); (S.C.)
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
| | - Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain; (Y.M.); (T.F.); (S.C.)
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Departamento de Investigación, Instituto Ordóñez, 33012 Oviedo, Asturias, Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain;
- Instituto Asturiano de Odontología, 33006 Oviedo, Asturias, Spain
| | - Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain; (Y.M.); (T.F.); (S.C.)
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
| | - Alvaro J. Obaya
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Correspondence:
| |
Collapse
|