101
|
Abstract
The nervous system regulates tissue stem and precursor populations throughout life. Parallel to roles in development, the nervous system is emerging as a critical regulator of cancer, from oncogenesis to malignant growth and metastatic spread. Various preclinical models in a range of malignancies have demonstrated that nervous system activity can control cancer initiation and powerfully influence cancer progression and metastasis. Just as the nervous system can regulate cancer progression, cancer also remodels and hijacks nervous system structure and function. Interactions between the nervous system and cancer occur both in the local tumour microenvironment and systemically. Neurons and glial cells communicate directly with malignant cells in the tumour microenvironment through paracrine factors and, in some cases, through neuron-to-cancer cell synapses. Additionally, indirect interactions occur at a distance through circulating signals and through influences on immune cell trafficking and function. Such cross-talk among the nervous system, immune system and cancer-both systemically and in the local tumour microenvironment-regulates pro-tumour inflammation and anti-cancer immunity. Elucidating the neuroscience of cancer, which calls for interdisciplinary collaboration among the fields of neuroscience, developmental biology, immunology and cancer biology, may advance effective therapies for many of the most difficult to treat malignancies.
Collapse
Affiliation(s)
- Rebecca Mancusi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
102
|
Ma J, Chen T, Wang R. Astragaloside IV ameliorates cognitive impairment and protects oligodendrocytes from antioxidative stress via regulation of the SIRT1/Nrf2 signaling pathway. Neurochem Int 2023; 167:105535. [PMID: 37209830 DOI: 10.1016/j.neuint.2023.105535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/08/2023] [Accepted: 04/23/2023] [Indexed: 05/22/2023]
Abstract
Subcortical ischemic vascular dementia (SIVD), which is caused by chronic cerebral hypoperfusion, is a common subtype of vascular dementia, accompanied by white matter damage and cognitive impairment. Currently, there are no effective treatments for this condition. Oxidative stress is a key factor in the pathogenesis of white matter damage. Astragaloside IV (AS-IV), one of the main active components of astragaloside, has antioxidant properties and promotes cognitive improvement; however, its effect on SIVD and its potential mechanism remain unknown. We aimed to clarify whether AS-IV had a protective effect against SIVD injury caused by right unilateral common carotid artery occlusion and the underlying mechanism. The results showed that AS-IV treatment improved cognitive function and white matter damage, inhibited oxidative stress and glial cells activation, and promoted the survival of mature oligodendrocytes after chronic cerebral hypoperfusion. Moreover, the protein expression levels of NQO1, HO-1, SIRT1 and Nrf2 were increased by AS-IV treatment. However, pre-treatment with EX-527, a SIRT1-specific inhibitor, eliminated the beneficial effects of AS-IV. These results demonstrate that AS-IV plays a neuroprotective role in SIVD by suppressing oxidative stress and increasing the number of mature oligodendrocytes via the modulation of SIRT1/Nrf2 signaling. Our results support AS-IV as a potential therapeutic agent for SIVD.
Collapse
Affiliation(s)
- Jing Ma
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, PR China
| | - Ting Chen
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, PR China.
| | - Ranran Wang
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, PR China.
| |
Collapse
|
103
|
Huang Z, Jordan JD, Zhang Q. Early life adversity as a risk factor for cognitive impairment and Alzheimer's disease. Transl Neurodegener 2023; 12:25. [PMID: 37173751 PMCID: PMC10182702 DOI: 10.1186/s40035-023-00355-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
Neurological conditions, including cognitive impairment and Alzheimer's disease (AD), impose a huge burden on society, affecting millions of people globally. In addition to genetic factors, recent studies indicate that environmental and experiential factors may contribute to the pathogenesis of these diseases. Early life adversity (ELA) has a profound impact on brain function and health later in life. In rodent models, exposure to ELA results in specific cognitive deficits and aggravated AD pathology. Extensive concerns have been raised regarding the higher risk of developing cognitive impairments in people with a history of ELA. In this review, we scrutinize findings from human and animal studies focusing on the connection of ELA with cognitive impairment and AD. These discoveries suggest that ELA, especially at early postnatal stages, increases susceptibility to cognitive impairment and AD later in life. In terms of mechanisms, ELA could lead to dysregulation of the hypothalamus-pituitary-adrenal axis, altered gut microbiome, persistent inflammation, oligodendrocyte dysfunction, hypomyelination, and aberrant adult hippocampal neurogenesis. Crosstalks among these events may synergistically contribute to cognitive impairment later in life. Additionally, we discuss several interventions that may alleviate adverse consequences of ELA. Further investigation into this crucial area will help improve ELA management and reduce the burden of related neurological conditions.
Collapse
Affiliation(s)
- Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - J Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| |
Collapse
|
104
|
Winkler F, Venkatesh HS, Amit M, Batchelor T, Demir IE, Deneen B, Gutmann DH, Hervey-Jumper S, Kuner T, Mabbott D, Platten M, Rolls A, Sloan EK, Wang TC, Wick W, Venkataramani V, Monje M. Cancer neuroscience: State of the field, emerging directions. Cell 2023; 186:1689-1707. [PMID: 37059069 PMCID: PMC10107403 DOI: 10.1016/j.cell.2023.02.002] [Citation(s) in RCA: 190] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 04/16/2023]
Abstract
The nervous system governs both ontogeny and oncology. Regulating organogenesis during development, maintaining homeostasis, and promoting plasticity throughout life, the nervous system plays parallel roles in the regulation of cancers. Foundational discoveries have elucidated direct paracrine and electrochemical communication between neurons and cancer cells, as well as indirect interactions through neural effects on the immune system and stromal cells in the tumor microenvironment in a wide range of malignancies. Nervous system-cancer interactions can regulate oncogenesis, growth, invasion and metastatic spread, treatment resistance, stimulation of tumor-promoting inflammation, and impairment of anti-cancer immunity. Progress in cancer neuroscience may create an important new pillar of cancer therapy.
Collapse
Affiliation(s)
- Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg and Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Humsa S Venkatesh
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Moran Amit
- Department of Head and Neck Surgery, MD Anderson Cancer Center and The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Tracy Batchelor
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Ihsan Ekin Demir
- Department of Surgery, Technical University of Munich, Munich, Germany
| | - Benjamin Deneen
- Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX, USA
| | - David H Gutmann
- Department of Neurology, Washington University, St Louis, MO, USA
| | - Shawn Hervey-Jumper
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas Kuner
- Department of Functional Neuroanatomy, University of Heidelberg, Heidelberg, Germany
| | - Donald Mabbott
- Department of Psychology, University of Toronto and Neuroscience & Mental Health Program, Research Institute, The Hospital for Sick Children, Toronto, Canada
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Asya Rolls
- Department of Immunology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Erica K Sloan
- Monash Institute of Pharmaceutical Sciences, Drug Discovery Biology Theme, Monash University, Parkville, VIC, Australia
| | - Timothy C Wang
- Department of Medicine, Division of Digestive and Gastrointestinal Diseases, Columbia University, New York, NY, USA
| | - Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg and Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Varun Venkataramani
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg and Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Functional Neuroanatomy, University of Heidelberg, Heidelberg, Germany.
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
105
|
Fabbri R, Spennato D, Conte G, Konstantoulaki A, Lazzarini C, Saracino E, Nicchia GP, Frigeri A, Zamboni R, Spray DC, Benfenati V. The emerging science of Glioception: Contribution of glia in sensing, transduction, circuit integration of interoception. Pharmacol Ther 2023; 245:108403. [PMID: 37024060 DOI: 10.1016/j.pharmthera.2023.108403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023]
Abstract
Interoception is the process by which the nervous system regulates internal functions to achieve homeostasis. The role of neurons in interoception has received considerable recent attention, but glial cells also contribute. Glial cells can sense and transduce signals including osmotic, chemical, and mechanical status of extracellular milieu. Their ability to dynamically communicate "listening" and "talking" to neurons is necessary to monitor and regulate homeostasis and information integration in the nervous system. This review introduces the concept of "Glioception" and focuses on the process by which glial cells sense, interpret and integrate information about the inner state of the organism. Glial cells are ideally positioned to act as sensors and integrators of diverse interoceptive signals and can trigger regulatory responses via modulation of the activity of neuronal networks, both in physiological and pathological conditions. We believe that understanding and manipulating glioceptive processes and underlying molecular mechanisms provide a key path to develop new therapies for the prevention and alleviation of devastating interoceptive dysfunctions, among which pain is emphasized here with more focused details.
Collapse
Affiliation(s)
- Roberta Fabbri
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy; Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, viale del Risorgimento 2, 40136 Bologna, Italy.
| | - Diletta Spennato
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy; Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Giorgia Conte
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - Aikaterini Konstantoulaki
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy; Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi, 2, 40126 Bologna, BO, Italy
| | - Chiara Lazzarini
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - Emanuela Saracino
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - Grazia Paola Nicchia
- School of Medicine, Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Bari, BA, Italy; Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Antonio Frigeri
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Roberto Zamboni
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - David C Spray
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Valentina Benfenati
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy.
| |
Collapse
|
106
|
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
|
107
|
Single cell molecular alterations reveal target cells and pathways of conditioned fear memory. Brain Res 2023; 1807:148309. [PMID: 36870465 DOI: 10.1016/j.brainres.2023.148309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/17/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
OBJECTIVES Recent evidence indicates that hippocampus is important for conditioned fear memory (CFM). Though few studies consider the roles of various cell types' contribution to such a process, as well as the accompanying transcriptome changes during this process. The purpose of this study was to explore the transcriptional regulatory genes and the targeted cells that are altered by CFM reconsolidation. METHODS A fear conditioning experiment was established on adult male C57 mice, after day 3 tone-cued CFM reconsolidation test, hippocampus cells were dissociated. Using single cell RNA sequencing (scRNA-seq) technique, alterations of transcriptional genes expression were detected and cell cluster analysis were performed and compared with those in sham group. RESULTS Seven non-neuronal and eight neuronal cell clusters (including four known neurons and four newly identified neuronal subtypes) has been explored. Among them, CA subtype 1 has characteristic gene markers of Ttr and Ptgds, which is speculated to be the outcome of acute stress and promotes the production of CFM. The results of KEGG pathway enrichment indicate the differences in the expression of certain molecular protein functional subunits in long-term potentiation (LTP) pathway between two types of neurons (DG and CA1) and astrocytes, thus providing a new transcriptional perspective for the role of hippocampus in the CFM reconsolidation. More importantly, the correlation between the reconsolidation of CFM and neurodegenerative diseases-linked genes is substantiated by the results from cell-cell interactions and KEGG pathway enrichment. Further analysis shows that the reconsolidation of CFM inhibits the risk-factor genes App and ApoE in Alzheimer's Disease (AD) and activates the protective gene Lrp1. CONCLUSIONS This study reports the transcriptional genes expression changes of hippocampal cells driven by CFM, which confirm the involvement of LTP pathway and suggest the possibility of CFM-like behavior in preventing AD. However, the current research is limited to normal C57 mice, and further studies on AD model mice are needed to prove this preliminary conclusion.
Collapse
|
108
|
Chen YH, Jin SY, Yang JM, Gao TM. The Memory Orchestra: Contribution of Astrocytes. Neurosci Bull 2023; 39:409-424. [PMID: 36738435 PMCID: PMC10043126 DOI: 10.1007/s12264-023-01024-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023] Open
Abstract
For decades, memory research has centered on the role of neurons, which do not function in isolation. However, astrocytes play important roles in regulating neuronal recruitment and function at the local and network levels, forming the basis for information processing as well as memory formation and storage. In this review, we discuss the role of astrocytes in memory functions and their cellular underpinnings at multiple time points. We summarize important breakthroughs and controversies in the field as well as potential avenues to further illuminate the role of astrocytes in memory processes.
Collapse
Affiliation(s)
- Yi-Hua Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Shi-Yang Jin
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
109
|
Buller S, Kohnke S, Hansford R, Shimizu T, Richardson WD, Blouet C. Median eminence myelin continuously turns over in adult mice. Mol Metab 2023; 69:101690. [PMID: 36739968 PMCID: PMC9950957 DOI: 10.1016/j.molmet.2023.101690] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/16/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Oligodendrocyte progenitor cell differentiation is regulated by nutritional signals in the adult median eminence (ME), but the consequences on local myelination are unknown. The aim of this study was to characterize myelin plasticity in the ME of adult mice in health or in response to chronic nutritional challenge and determine its relevance to the regulation of energy balance. METHODS We assessed new oligodendrocyte (OL) and myelin generation and stability in the ME of healthy adult male mice using bromodeoxyuridine labelling and genetic fate mapping tools. We evaluated the contribution of microglia to ME myelin plasticity in PLX5622-treated C57BL/6J mice and in Pdgfra-Cre/ERT2;R26R-eYFP;Myrffl/fl mice, where adult oligodendrogenesis is blunted. Next, we investigated how high-fat feeding or caloric restriction impact ME OL lineage progression and myelination. Finally, we characterized the functional relevance of adult oligodendrogenesis on energy balance regulation. RESULTS We show that myelinating OLs are continuously and rapidly generated in the adult ME. Paradoxically, OL number and myelin amounts remain remarkably stable in the adult ME. In fact, the high rate of new OL and myelin generation in the ME is offset by continuous turnover of both. We show that microglia are required for continuous OL and myelin production, and that ME myelin plasticity regulates the recruitment of local immune cells. Finally, we provide evidence that ME myelination is regulated by the body's energetic status and demonstrate that ME OL and myelin plasticity are required for the regulation of energy balance and hypothalamic leptin sensitivity. CONCLUSIONS This study identifies a new mechanism modulating leptin sensitivity and the central control of energy balance and uncovers a previously unappreciated form of structural plasticity in the ME.
Collapse
Affiliation(s)
- Sophie Buller
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Disease Unit, University of Cambridge, Cambridge, UK.
| | - Sara Kohnke
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Disease Unit, University of Cambridge, Cambridge, UK.
| | - Robert Hansford
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Disease Unit, University of Cambridge, Cambridge, UK.
| | - Takahiro Shimizu
- Wolfson Institute for Biomedical Research, University College London, London, UK.
| | - William D Richardson
- Wolfson Institute for Biomedical Research, University College London, London, UK.
| | - Clemence Blouet
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Disease Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
110
|
Flounlacker KM, Hahn YK, Xu R, Simons CA, Tian T, Hauser KF, Knapp PE. Myelin regulatory factor is a target of individual and interactive effects of HIV-1 Tat and morphine in the striatum and pre-frontal cortex. J Neurovirol 2023; 29:15-26. [PMID: 36853588 DOI: 10.1007/s13365-022-01107-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 11/11/2022] [Accepted: 11/25/2022] [Indexed: 03/01/2023]
Abstract
HIV-associated neurocognitive disorders (HAND) remain pervasive even with increased efficacy/use of antiretroviral therapies. Opioid use/abuse among HIV + individuals is documented to exacerbate CNS deficits. White matter (WM) alterations, including myelin pallor, and volume/structural alterations detected by diffusion tensor imaging are common observations in HIV + individuals, and studies in non-human primates suggest that WM may harbor virus. Using transgenic mice that express the HIV-1 Tat protein, we examined in vivo effects of 2-6 weeks of Tat and morphine exposure on WM using genomic and biochemical methods. RNA sequencing of striatal tissue at 2 weeks revealed robust changes in mRNAs associated with oligodendrocyte precursor populations and myelin integrity, including those for transferrin, the atypical oligodendrocyte marker N-myc downstream regulated 1 (Ndrg1), and myelin regulatory factor (Myrf/Mrf), an oligodendrocyte-specific transcription factor with a significant role in oligodendrocyte differentiation/maturation. Western blots conducted after 6-weeks exposure in 3 brain regions (striatum, corpus callosum, pre-frontal cortex) revealed regional differences in the effect of Tat and morphine on Myrf levels, and on levels of myelin basic protein (MBP), whose transcription is regulated by Myrf. Responses included individual and interactive effects. Although baseline and post-treatment levels of Myrf and MBP differed between brain regions, post-treatment MBP levels in striatum and pre-frontal cortex were compatible with changes in Myrf activity. Additionally, the Myrf regulatory ubiquitin ligase Fbxw7 was identified as a novel target in our model. These results suggest that Myrf and Fbxw7 contribute to altered myelin gene regulation in HIV.
Collapse
Affiliation(s)
- Kelly M Flounlacker
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA.
| | - Yun Kyung Hahn
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA
| | - Ruqiang Xu
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA
| | - Chloe A Simons
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA
| | - Tao Tian
- Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Kurt F Hauser
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA.,Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.,The Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Pamela E Knapp
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA.,Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.,The Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| |
Collapse
|
111
|
Mandal AS, Brem S, Suckling J. Brain network mapping and glioma pathophysiology. Brain Commun 2023; 5:fcad040. [PMID: 36895956 PMCID: PMC9989143 DOI: 10.1093/braincomms/fcad040] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 12/23/2022] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
Adult diffuse gliomas are among the most difficult brain disorders to treat in part due to a lack of clarity regarding the anatomical origins and mechanisms of migration of the tumours. While the importance of studying networks of glioma spread has been recognized for at least 80 years, the ability to carry out such investigations in humans has emerged only recently. Here, we comprehensively review the fields of brain network mapping and glioma biology to provide a primer for investigators interested in merging these areas of inquiry for the purposes of translational research. Specifically, we trace the historical development of ideas in both brain network mapping and glioma biology, highlighting studies that explore clinical applications of network neuroscience, cells-of-origin of diffuse glioma and glioma-neuronal interactions. We discuss recent research that has merged neuro-oncology and network neuroscience, finding that the spatial distribution patterns of gliomas follow intrinsic functional and structural brain networks. Ultimately, we call for more contributions from network neuroimaging to realize the translational potential of cancer neuroscience.
Collapse
Affiliation(s)
- Ayan S Mandal
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Steven Brem
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Philadelphia, PA 19104, USA
| | - John Suckling
- Department of Psychiatry, University of Cambridge, Cambridge CB2 0SZ, UK
| |
Collapse
|
112
|
Timmermann A, Tascio D, Jabs R, Boehlen A, Domingos C, Skubal M, Huang W, Kirchhoff F, Henneberger C, Bilkei-Gorzo A, Seifert G, Steinhäuser C. Dysfunction of NG2 glial cells affects neuronal plasticity and behavior. Glia 2023; 71:1481-1501. [PMID: 36802096 DOI: 10.1002/glia.24352] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 02/20/2023]
Abstract
NG2 glia represents a distinct type of macroglial cells in the CNS and is unique among glia because they receive synaptic input from neurons. They are abundantly present in white and gray matter. While the majority of white matter NG2 glia differentiates into oligodendrocytes, the physiological impact of gray matter NG2 glia and their synaptic input are still ill defined. Here, we asked whether dysfunctional NG2 glia affect neuronal signaling and behavior. We generated mice with inducible deletion of the K+ channel Kir4.1 in NG2 glia and performed comparative electrophysiological, immunohistochemical, molecular and behavioral analyses. Kir4.1 was deleted at postnatal day 23-26 (recombination efficiency about 75%) and mice were investigated 3-8 weeks later. Notably, these mice with dysfunctional NG2 glia demonstrated improved spatial memory as revealed by testing new object location recognition while working and social memory remained unaffected. Focussing on the hippocampus, we found that loss of Kir4.1 potentiated synaptic depolarizations of NG2 glia and stimulated the expression of myelin basic protein while proliferation and differentiation of hippocampal NG2 glia remained largely unaffected. Mice with targeted deletion of the K+ channel in NG2 glia showed impaired long-term potentiation at CA3-CA1 synapses, which could be fully rescued by extracellular application of a TrkB receptor agonist. Our data demonstrate that proper NG2 glia function is important for normal brain function and behavior.
Collapse
Affiliation(s)
- Aline Timmermann
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Dario Tascio
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Ronald Jabs
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Anne Boehlen
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Catia Domingos
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Magdalena Skubal
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Wenhui Huang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute of Neurology, University College London, London, UK
| | - Andras Bilkei-Gorzo
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn, Germany
| | - Gerald Seifert
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
113
|
Cohn EF, Clayton BL, Madhavan M, Yacoub S, Federov Y, Paul-Friedman K, Shafer TJ, Tesar PJ. Pervasive environmental chemicals impair oligodendrocyte development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.528042. [PMID: 36798415 PMCID: PMC9934656 DOI: 10.1101/2023.02.10.528042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Exposure to environmental chemicals can impair neurodevelopment1-4. Oligodendrocytes that wrap around axons to boost neurotransmission may be particularly vulnerable to chemical toxicity as they develop throughout fetal development and into adulthood5,6. However, few environmental chemicals have been assessed for potential risks to oligodendrocyte development. Here, we utilized a high-throughput developmental screen and human cortical brain organoids, which revealed environmental chemicals in two classes that disrupt oligodendrocyte development through distinct mechanisms. Quaternary compounds, ubiquitous in disinfecting agents, hair conditioners, and fabric softeners, were potently and selectively cytotoxic to developing oligodendrocytes through activation of the integrated stress response. Organophosphate flame retardants, commonly found in household items such as furniture and electronics, were non-cytotoxic but prematurely arrested oligodendrocyte maturation. Chemicals from each class impaired human oligodendrocyte development in a 3D organoid model of prenatal cortical development. In analysis of epidemiological data from the CDC's National Health and Nutrition Examination Survey, adverse neurodevelopmental outcomes were associated with childhood exposure to the top organophosphate flame retardant identified by our oligodendrocyte toxicity platform. Collectively, our work identifies toxicological vulnerabilities specific to oligodendrocyte development and highlights common household chemicals with high exposure risk to children that warrant deeper scrutiny for their impact on human health.
Collapse
Affiliation(s)
- Erin F. Cohn
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Benjamin L.L. Clayton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Mayur Madhavan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Sara Yacoub
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Yuriy Federov
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Katie Paul-Friedman
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Timothy J. Shafer
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Paul J. Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| |
Collapse
|
114
|
Rawlings-Mortimer F, Gullino LS, Rühling S, Ashton A, Barkus C, Johansen-Berg H. DUSP15 expression is reduced in the hippocampus of Myrf knock-out mice but attention and object recognition memory remain intact. PLoS One 2023; 18:e0281264. [PMID: 36730342 PMCID: PMC9894471 DOI: 10.1371/journal.pone.0281264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/18/2023] [Indexed: 02/03/2023] Open
Abstract
The atypical protein tyrosine phosphatase enzyme, dual-specificity phosphate 15 (DUSP15) is thought to be activated by myelin regulatory factor (MyRF) and to have a role in oligodendrocyte differentiation. Here, we assess whether Dusp15 is reduced in the hippocampus of mice with conditional knock-out of Myrf in oligodendrocyte precursor cells. Using quantitative polymerase chain reaction (qPCR) we found that Dusp15 expression was indeed lower in these mice. Alterations in myelin have been associated with Alzheimer's disease (AD), autism spectrum disorder (ASD) and attention deficit/hyperactivity disorder (ADHD). Symptoms of these disorders can include impairments of object recognition and attention. We, therefore tested the mice in the object recognition task (ORT) and 5-choice serial reaction time task (5CSRTT). However, we did not find behavioural impairments indicating that attentional abilities and object recognition are not impacted by reduced oligodendrogenesis and hippocampal Dusp15 expression. Gaining insight into the role of newly formed oligodendrocytes and Dusp15 expression is helpful for the development of well targeted treatments for myelin dysregulation.
Collapse
Affiliation(s)
- Florence Rawlings-Mortimer
- Wellcome Centre for Integrative Neuroimaging, Nuffield Dept of Clinical Neurosciences, Oxford, United Kingdom
| | - L Sophie Gullino
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Sebastian Rühling
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Anna Ashton
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, United Kingdom
| | - Chris Barkus
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Heidi Johansen-Berg
- Wellcome Centre for Integrative Neuroimaging, Nuffield Dept of Clinical Neurosciences, Oxford, United Kingdom
| |
Collapse
|
115
|
Deng S, Shu S, Zhai L, Xia S, Cao X, Li H, Bao X, Liu P, Xu Y. Optogenetic Stimulation of mPFC Alleviates White Matter Injury-Related Cognitive Decline after Chronic Ischemia through Adaptive Myelination. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2202976. [PMID: 36529961 PMCID: PMC9929132 DOI: 10.1002/advs.202202976] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/08/2022] [Indexed: 06/07/2023]
Abstract
White matter injury (WMI), which reflects myelin loss, contributes to cognitive decline or dementia caused by cerebral vascular diseases. However, because pharmacological agents specifically for WMI are lacking, novel therapeutic strategies need to be explored. It is recently found that adaptive myelination is required for homeostatic control of brain functions. In this study, adaptive myelination-related strategies are applied to explore the treatment for ischemic WMI-related cognitive dysfunction. Here, bilateral carotid artery stenosis (BCAS) is used to model ischemic WMI-related cognitive impairment and uncover that optogenetic and chemogenetic activation of glutamatergic neurons in the medial prefrontal cortex (mPFC) promote the differentiation of oligodendrocyte precursor cells (OPCs) in the corpus callosum, leading to improvements in myelin repair and working memory. Mechanistically, these neuromodulatory techniques exert a therapeutic effect by inducing the secretion of Wnt2 from activated neuronal axons, which acts on oligodendrocyte precursor cells and drives oligodendrogenesis and myelination. Thus, this study suggests that neuromodulation is a promising strategy for directing myelin repair and cognitive recovery through adaptive myelination in the context of ischemic WMI.
Collapse
Affiliation(s)
- Shiji Deng
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Shu Shu
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Lili Zhai
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Shengnan Xia
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Xiang Cao
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Huiya Li
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Xinyu Bao
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Pinyi Liu
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Yun Xu
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjing210008China
- Jiangsu Provincial Key Discipline of NeurologyNanjing210008China
- Nanjing Neurology Medical CenterNanjing210008China
| |
Collapse
|
116
|
Bostancıklıoğlu M, Kaplan DS, Temiz E, Yiğit E. Local myelin damage in the hippocampus fluctuates gut microbiome profile and memory. J Psychiatr Res 2023; 158:392-402. [PMID: 36646037 DOI: 10.1016/j.jpsychires.2023.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/18/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023]
Abstract
The concept of the gut-brain axis has focused research on how gut dysbiosis affects myelin biology in the brain. However, this axis has not been tested to determine whether it conveys the effects of myelin damage on the gut microbiome profile. Therefore, we aimed to investigate how myelin biology is correlated with gut microbiome profile. The impact of local myelin damage in the hippocampus on gut microbiome profile was investigated with 16S rRNA metagenomic sequence and molecular analysis of myelin biology-associated proteins, and its reflections on memory performance were tested with behavioral tests. Local myelin damage in the hippocampus triggered severe gut dysbiosis, p < .05, changed memory performance, p < .05, and deviated emotional responses. Moreover, myelin treatment with clemastine improved gut dysbiosis and behavioral deviations. Our study provides animal-based evidence on the direct interaction between glial biology in the hippocampus and gut microbiome profile. This study proposes a framework for generating new hypotheses bridging different systems to the gut-brain axis.
Collapse
Affiliation(s)
| | - Davut Sinan Kaplan
- Department of Physiology, Gaziantep University Faculty of Medicine, Gaziantep, Turkey
| | - Ebru Temiz
- Program of Medical Promotion and Marketing, Health Services Vocational School, Harran University, Sanliurfa, Turkey
| | - Elif Yiğit
- Department of Physiology, Gaziantep University Faculty of Medicine, Gaziantep, Turkey
| |
Collapse
|
117
|
Myelinated axon as a plastic cable regulating brain functions. Neurosci Res 2023; 187:45-51. [PMID: 36347403 DOI: 10.1016/j.neures.2022.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022]
Abstract
Each oligodendrocyte (OC) forms myelin approximately in around 10 different axons to coordinate information transfer by regulating conduction velocity in the central nervous system (CNS). In the classical view, myelin has been considered a static structure that rarely turns over under healthy conditions because myelin tightly holds axons by their laminar complex structure. However, in recent decades, the classical views of static myelin have been renewed with pioneering studies that showed plastic changes in myelin throughout life with new experiences, such as the acquisition of new motor skills and the formation of memory. These changes in myelin regulate conduction velocity to optimize the temporal pattern of neuronal circuit activity among distinct brain regions associated with skill learning and memory. Here, we introduce pioneering studies and discuss the implications of plastic myelin on neural circuits and brain function.
Collapse
|
118
|
Konakanchi S, Raavi V, Ml HK, Shankar Ms V. Impact of chronic sleep deprivation and sleep recovery on hippocampal oligodendrocytes, anxiety-like behavior, spatial learning and memory of rats. Brain Res Bull 2023; 193:59-71. [PMID: 36494056 DOI: 10.1016/j.brainresbull.2022.12.002] [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: 06/13/2022] [Revised: 11/21/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
Sleep and its quality play an important role in memory, cognition, and quality of life. Sleep deprivation-induced changes in hippocampal neurons and behavior have been studied widely, in contrast, the extent of damage to oligodendrocytes have not been fully understood. The present study aims to investigate chronic sleep deprivation (CSD) and sleep recovery-induced changes in oligodendrocytes of the hippocampus, cognition, and behavior of rats. Male Sprague-Dawley rats (n = 48) were grouped as control, sham control (SC), CSD, and CSD+sleep recovery (CSD+SR) (n = 12/group). CSD and CSD+SR group rats were sleep deprived for 21-days. After CSD, the CSD+SR group rats sleep recovered for 21-days. Oxidative markers, CNPase+ve oligodendrocytes, CNPase intensity, and CNPase gene expression were measured in the hippocampus, and the anxiety-like behavior, spatial learning, and memory were assessed. The 21-days of CSD significantly (p < 0.001) increased oxidative stress and significantly (p < 0.001) reduced the number of CNPase+ve oligodendrocytes, CNPase intensity, and CNPase gene expression when compared to controls. The increased oxidative stress was correlated with reduced CNPase+ve oligodendrocytes, CNPase intensity, and CNPase gene expression (r = -0.9). In-line with cellular changes, an increased (p < 0.01) anxiety-like behavior and impaired spatial memory were observed in the CSD group compared to controls. The 21-days of sleep recovery significantly (p < 0.01) reduced oxidative stress and anxiety-like behavior, improved spatial memory, increased CNPase intensity and CNPase gene expression, and non-significant (p > 0.05) increase in CNPase+ve oligodendrocytes compared to CSD. Overall, the 21-days of CSD reduced the number of CNPase+ve oligodendrocytes in the hippocampus, increased anxiety, and impaired spatial memory in rats. Though the 21-day sleep recovery showed an improvement in all parameters, it was not sufficient to completely reverse the CSD-induced changes to the control level.
Collapse
Affiliation(s)
- Suresh Konakanchi
- Department of Physiology, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education and Research (Deemed to be University), Kolar 563103, Karnataka, India.
| | - Venkateswarlu Raavi
- Department of Cell Biology and Molecular Genetics, Sri Devaraj Urs Academy of Higher Education and Research (Deemed to be University), Kolar 563103, Karnataka, India.
| | - Harendra Kumar Ml
- Department of Pathology, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education and Research (Deemed to be University), Kolar 563103, Karnataka, India.
| | - Vinutha Shankar Ms
- Department of Physiology, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education and Research (Deemed to be University), Kolar 563103, Karnataka, India.
| |
Collapse
|
119
|
Vanrobeys Y, Mukherjee U, Langmack L, Bahl E, Lin LC, Michaelson JJ, Abel T, Chatterjee S. Mapping the spatial transcriptomic signature of the hippocampus during memory consolidation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524576. [PMID: 36711475 PMCID: PMC9882356 DOI: 10.1101/2023.01.18.524576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Memory consolidation involves discrete patterns of transcriptional events in the hippocampus. Despite the emergence of single-cell transcriptomic profiling techniques, defining learning-responsive gene expression across subregions of the hippocampus has remained challenging. Here, we utilized unbiased spatial sequencing to elucidate transcriptome-wide changes in gene expression in the hippocampus following learning, enabling us to define molecular signatures unique to each hippocampal subregion. We find that each subregion of the hippocampus exhibits distinct yet overlapping transcriptomic signatures. Although the CA1 region exhibited increased expression of genes related to transcriptional regulation, the DG showed upregulation of genes associated with protein folding. We demonstrate the functional relevance of subregion-specific gene expression by genetic manipulation of a transcription factor selectively in the CA1 hippocampal subregion, leading to long-term memory deficits. This work demonstrates the power of using spatial molecular approaches to reveal transcriptional events during memory consolidation.
Collapse
Affiliation(s)
- Yann Vanrobeys
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
| | - Utsav Mukherjee
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242, USA
| | - Lucy Langmack
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Biochemistry and Molecular Biology Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Ethan Bahl
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Li-Chun Lin
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Jacob J Michaelson
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Snehajyoti Chatterjee
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
120
|
Huang Z, Zhang Y, Ma X, Feng Y, Zong X, Jordan JD, Zhang Q. Photobiomodulation attenuates oligodendrocyte dysfunction and prevents adverse neurological consequences in a rat model of early life adversity. Theranostics 2023; 13:913-930. [PMID: 36793860 PMCID: PMC9925323 DOI: 10.7150/thno.78777] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/04/2023] [Indexed: 02/04/2023] Open
Abstract
Rationale: Adverse experiences in early life including abuse, trauma and neglect, have been linked to poor physical and mental health outcomes. Emerging evidence implies that those who experienced early life adversity (ELA) are more likely to develop cognitive dysfunction and depressive-like symptoms in adulthood. The molecular mechanisms responsible for the negative consequences of ELA, however, remain unclear. In the absence of effective management options, anticipatory guidance is the mainstay of ELA prevention. Furthermore, there is no available treatment that prevents or alleviates the neurologic sequelae of ELA, especially traumatic stress. Hence, the present study aims to investigate the mechanisms for these associations and evaluate whether photobiomodulation (PBM), a non-invasive therapeutic procedure, can prevent the negative cognitive and behavioral manifestations of ELA in later life. Methods: ELA was induced by repeated inescapable electric foot shock of rats from postnatal day 21 to 26. On the day immediately following the last foot shock, 2-min daily PBM treatment was applied transcranially for 7 consecutive days. Cognitive dysfunction and depression-like behaviors were measured by a battery of behavioral tests in adulthood. Subsequently, oligodendrocyte progenitor cells (OPCs) differentiation, the proliferation and apoptosis of oligodendrocyte lineage cells (OLs), mature oligodendrocyte, myelinating oligodendrocyte, the level of oxidative damage, reactive oxygen species (ROS) and total antioxidant capacity were measured and analyzed using immunofluorescence staining, capillary-based immunoassay (ProteinSimple®) and antioxidant assay kit. Results: The rats exposed to ELA exhibited obvious oligodendrocyte dysfunction, including a reduction in OPCs differentiation, diminished generation and survival of OLs, decreased OLs, and decreased matured oligodendrocyte. Furthermore, a deficit in myelinating oligodendrocytes was observed, in conjunction with an imbalance in redox homeostasis and accumulated oxidative damage. These alternations were concomitant with cognitive dysfunction and depression-like behaviors. Importantly, we found that early PBM treatment largely prevented these pathologies and reversed the neurologic sequelae resulting from ELA. Conclusions: Collectively, these findings provide new insights into the mechanism by which ELA affects neurological outcomes. Moreover, our findings support that PBM may be a promising strategy to prevent ELA-induced neurologic sequelae that develops later in life.
Collapse
Affiliation(s)
| | | | | | | | | | - J. Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA 71103 USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA 71103 USA
| |
Collapse
|
121
|
Dittmann NL, Torabi P, Watson AES, Yuzwa SA, Voronova A. Culture Protocol and Transcriptomic Analysis of Murine SVZ NPCs and OPCs. Stem Cell Rev Rep 2023; 19:983-1000. [PMID: 36617597 DOI: 10.1007/s12015-022-10492-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 01/10/2023]
Abstract
The mammalian adult brain contains two neural stem and precursor (NPC) niches: the subventricular zone [SVZ] lining the lateral ventricles and the subgranular zone [SGZ] in the hippocampus. From these, SVZ NPCs represent the largest NPC pool. While SGZ NPCs typically only produce neurons and astrocytes, SVZ NPCs produce neurons, astrocytes and oligodendrocytes throughout life. Of particular importance is the generation and replacement of oligodendrocytes, the only myelinating cells of the central nervous system (CNS). SVZ NPCs contribute to myelination by regenerating the parenchymal oligodendrocyte precursor cell (OPC) pool and by differentiating into oligodendrocytes in the developing and demyelinated brain. The neurosphere assay has been widely adopted by the scientific community to facilitate the study of NPCs in vitro. Here, we present a streamlined protocol for culturing postnatal and adult SVZ NPCs and OPCs from primary neurosphere cells. We characterize the purity and differentiation potential as well as provide RNA-sequencing profiles of postnatal SVZ NPCs, postnatal SVZ OPCs and adult SVZ NPCs. We show that primary neurospheres cells generated from postnatal and adult SVZ differentiate into neurons, astrocytes and oligodendrocytes concurrently and at comparable levels. SVZ OPCs are generated by subjecting primary neurosphere cells to OPC growth factors fibroblast growth factor (FGF) and platelet-derived growth factor-AA (PDGF-AA). We further show SVZ OPCs can differentiate into oligodendrocytes in the absence and presence of thyroid hormone T3. Transcriptomic analysis confirmed the identities of each cell population and revealed novel immune and signalling pathways expressed in an age and cell type specific manner.
Collapse
Affiliation(s)
- Nicole L Dittmann
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.,Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Pouria Torabi
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Adrianne E S Watson
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Scott A Yuzwa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada. .,Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada. .,Women and Children's Health Research Institute5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, Alberta, T6G 1C9, Canada. .,Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada. .,Multiple Sclerosis Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| |
Collapse
|
122
|
Zheng R, Qi Z, Thibaut A, Wang Z, Xu Z, Di H, Wu X, Mao Y, Laureys S. Clinical application of neuromodulation therapy in patients with disorder of consciousness: A pooled analysis of 544 participants. NeuroRehabilitation 2023; 53:491-503. [PMID: 37927281 DOI: 10.3233/nre-230103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
BACKGROUND The number of patients with disorders of consciousness (DoC) has increased dramatically with the advancement of intensive care and emergency medicine, which brings tremendous economic burdens and even ethical issues to families and society. OBJECTIVE To evaluate the effectiveness of neuromodulation therapy for patients with DoC. METHODS First, we conducted a literature review of individual patient data (IPD) on PubMed, EMBASE, and Cochrane-controlled trials following PRISMA guidelines. Then, we collected neuromodulation cases from our institution. Finally, we conducted a pooled analysis using the participants from the medical literature (n = 522) and our local institutions (n = 22). RESULTS In this pooled analysis of 544 patients with DoC with a mean age of 46.33 years, our results revealed that patients have improved CRS-R scores [1.0 points (95% CI, 0.57-1.42)] after neuromodulation. Among them, patients have better effectiveness in traumatic than non-traumatic etiology (P < 0.05). The effectiveness of consciousness improvement could be affected by the age, baseline consciousness state, and duration of stimulation. Compared with non-invasive intervention, an invasive intervention can bring more behavioral improvement (P < 0.0001) to MCS rather than UWS/VS patients. Importantly, neuromodulation is a valuable therapy even years after the onset of DoC. CONCLUSION This pooled analysis spotlights that the application of neuromodulation can improve the behavioral performance of patients with DoC. A preliminary trend is that age, etiology, baseline consciousness state, and stimulation duration could impact its effectiveness.
Collapse
Affiliation(s)
- Ruizhe Zheng
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education's (MOE) Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Zengxin Qi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education's (MOE) Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Aurore Thibaut
- GIGA Consciousness Research Unit, Coma Science Group, Liège University, Liège, Belgium
- GIGA Consciousness, Centre du Cerveau, Liège University Hospital, Liège, Belgium
| | - Zhe Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education's (MOE) Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Zeyu Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education's (MOE) Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Haibo Di
- International Consciousness Science Institute, Hangzhou Normal University, Hangzhou, China
| | - Xuehai Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education's (MOE) Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education's (MOE) Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Steven Laureys
- GIGA Consciousness Research Unit, Coma Science Group, Liège University, Liège, Belgium
- GIGA Consciousness, Centre du Cerveau, Liège University Hospital, Liège, Belgium
- International Consciousness Science Institute, Hangzhou Normal University, Hangzhou, China
- Joint International Research Unit on Consciousness, CERVO Brain Research Centre, Laval University, Quebec, QC, Canada
| |
Collapse
|
123
|
McNamara NB, Munro DAD, Bestard-Cuche N, Uyeda A, Bogie JFJ, Hoffmann A, Holloway RK, Molina-Gonzalez I, Askew KE, Mitchell S, Mungall W, Dodds M, Dittmayer C, Moss J, Rose J, Szymkowiak S, Amann L, McColl BW, Prinz M, Spires-Jones TL, Stenzel W, Horsburgh K, Hendriks JJA, Pridans C, Muramatsu R, Williams A, Priller J, Miron VE. Microglia regulate central nervous system myelin growth and integrity. Nature 2023; 613:120-129. [PMID: 36517604 PMCID: PMC9812791 DOI: 10.1038/s41586-022-05534-y] [Citation(s) in RCA: 227] [Impact Index Per Article: 113.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/05/2022] [Indexed: 12/15/2022]
Abstract
Myelin is required for the function of neuronal axons in the central nervous system, but the mechanisms that support myelin health are unclear. Although macrophages in the central nervous system have been implicated in myelin health1, it is unknown which macrophage populations are involved and which aspects they influence. Here we show that resident microglia are crucial for the maintenance of myelin health in adulthood in both mice and humans. We demonstrate that microglia are dispensable for developmental myelin ensheathment. However, they are required for subsequent regulation of myelin growth and associated cognitive function, and for preservation of myelin integrity by preventing its degeneration. We show that loss of myelin health due to the absence of microglia is associated with the appearance of a myelinating oligodendrocyte state with altered lipid metabolism. Moreover, this mechanism is regulated through disruption of the TGFβ1-TGFβR1 axis. Our findings highlight microglia as promising therapeutic targets for conditions in which myelin growth and integrity are dysregulated, such as in ageing and neurodegenerative disease2,3.
Collapse
Affiliation(s)
- Niamh B McNamara
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - David A D Munro
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Nadine Bestard-Cuche
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Akiko Uyeda
- Departments of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Alana Hoffmann
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Rebecca K Holloway
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
- Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
| | - Irene Molina-Gonzalez
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Katharine E Askew
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Stephen Mitchell
- Wellcome Trust Centre for Cell Biology, King's Buildings, The University of Edinburgh, Edinburgh, UK
| | - William Mungall
- Biological and Veterinary Services, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Michael Dodds
- Biological and Veterinary Services, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Carsten Dittmayer
- Department of Neuropathology and Neurocure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jonathan Moss
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Jamie Rose
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Stefan Szymkowiak
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Lukas Amann
- Institute of Neuropathology, Centre for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Barry W McColl
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Marco Prinz
- Institute of Neuropathology, Centre for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Tara L Spires-Jones
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Werner Stenzel
- Department of Neuropathology and Neurocure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Clare Pridans
- Centre for Inflammation Research, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK
| | - Rieko Muramatsu
- Departments of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Anna Williams
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Josef Priller
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin and DZNE, Berlin, Germany
| | - Veronique E Miron
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK.
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK.
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK.
- Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada.
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, Ontario, Canada.
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
124
|
Xiong Y, Hong H, Liu C, Zhang YQ. Social isolation and the brain: effects and mechanisms. Mol Psychiatry 2023; 28:191-201. [PMID: 36434053 PMCID: PMC9702717 DOI: 10.1038/s41380-022-01835-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/05/2022] [Accepted: 10/10/2022] [Indexed: 11/26/2022]
Abstract
An obvious consequence of the coronavirus disease (COVID-19) pandemic is the worldwide reduction in social interaction, which is associated with many adverse effects on health in humans from babies to adults. Although social development under normal or isolated environments has been studied since the 1940s, the mechanism underlying social isolation (SI)-induced brain dysfunction remains poorly understood, possibly due to the complexity of SI in humans and translational gaps in findings from animal models. Herein, we present a systematic review that focused on brain changes at the molecular, cellular, structural and functional levels induced by SI at different ages and in different animal models. SI studies in humans and animal models revealed common socioemotional and cognitive deficits caused by SI in early life and an increased occurrence of depression and anxiety induced by SI during later stages of life. Altered neurotransmission and neural circuitry as well as abnormal development and function of glial cells in specific brain regions may contribute to the abnormal emotions and behaviors induced by SI. We highlight distinct alterations in oligodendrocyte progenitor cell differentiation and oligodendrocyte maturation caused by SI in early life and later stages of life, respectively, which may affect neural circuit formation and function and result in diverse brain dysfunctions. To further bridge animal and human SI studies, we propose alternative animal models with brain structures and complex social behaviors similar to those of humans.
Collapse
Affiliation(s)
- Ying Xiong
- grid.9227.e0000000119573309State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Huilin Hong
- grid.9227.e0000000119573309State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Cirong Liu
- grid.9227.e0000000119573309Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031 China ,grid.511008.dShanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210 China
| | - Yong Q. Zhang
- grid.9227.e0000000119573309State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419University of the Chinese Academy of Sciences, Beijing, 100101 China
| |
Collapse
|
125
|
Grimaldi A, Gruel A, Besnainou C, Jérémie JN, Martinet J, Perrinet LU. Precise Spiking Motifs in Neurobiological and Neuromorphic Data. Brain Sci 2022; 13:68. [PMID: 36672049 PMCID: PMC9856822 DOI: 10.3390/brainsci13010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Why do neurons communicate through spikes? By definition, spikes are all-or-none neural events which occur at continuous times. In other words, spikes are on one side binary, existing or not without further details, and on the other, can occur at any asynchronous time, without the need for a centralized clock. This stands in stark contrast to the analog representation of values and the discretized timing classically used in digital processing and at the base of modern-day neural networks. As neural systems almost systematically use this so-called event-based representation in the living world, a better understanding of this phenomenon remains a fundamental challenge in neurobiology in order to better interpret the profusion of recorded data. With the growing need for intelligent embedded systems, it also emerges as a new computing paradigm to enable the efficient operation of a new class of sensors and event-based computers, called neuromorphic, which could enable significant gains in computation time and energy consumption-a major societal issue in the era of the digital economy and global warming. In this review paper, we provide evidence from biology, theory and engineering that the precise timing of spikes plays a crucial role in our understanding of the efficiency of neural networks.
Collapse
Affiliation(s)
- Antoine Grimaldi
- INT UMR 7289, Aix Marseille Univ, CNRS, 27 Bd Jean Moulin, 13005 Marseille, France
| | - Amélie Gruel
- SPARKS, Côte d’Azur, CNRS, I3S, 2000 Rte des Lucioles, 06900 Sophia-Antipolis, France
| | - Camille Besnainou
- INT UMR 7289, Aix Marseille Univ, CNRS, 27 Bd Jean Moulin, 13005 Marseille, France
| | - Jean-Nicolas Jérémie
- INT UMR 7289, Aix Marseille Univ, CNRS, 27 Bd Jean Moulin, 13005 Marseille, France
| | - Jean Martinet
- SPARKS, Côte d’Azur, CNRS, I3S, 2000 Rte des Lucioles, 06900 Sophia-Antipolis, France
| | - Laurent U. Perrinet
- INT UMR 7289, Aix Marseille Univ, CNRS, 27 Bd Jean Moulin, 13005 Marseille, France
| |
Collapse
|
126
|
Cristobal CD, Lee HK. Development of myelinating glia: An overview. Glia 2022; 70:2237-2259. [PMID: 35785432 PMCID: PMC9561084 DOI: 10.1002/glia.24238] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 01/07/2023]
Abstract
Myelin is essential to nervous system function, playing roles in saltatory conduction and trophic support. Oligodendrocytes (OLs) and Schwann cells (SCs) form myelin in the central and peripheral nervous systems respectively and follow different developmental paths. OLs are neural stem-cell derived and follow an intrinsic developmental program resulting in a largely irreversible differentiation state. During embryonic development, OL precursor cells (OPCs) are produced in distinct waves originating from different locations in the central nervous system, with a subset developing into myelinating OLs. OPCs remain evenly distributed throughout life, providing a population of responsive, multifunctional cells with the capacity to remyelinate after injury. SCs derive from the neural crest, are highly dependent on extrinsic signals, and have plastic differentiation states. SC precursors (SCPs) are produced in early embryonic nerve structures and differentiate into multipotent immature SCs (iSCs), which initiate radial sorting and differentiate into myelinating and non-myelinating SCs. Differentiated SCs retain the capacity to radically change phenotypes in response to external signals, including becoming repair SCs, which drive peripheral regeneration. While several transcription factors and myelin components are common between OLs and SCs, their differentiation mechanisms are highly distinct, owing to their unique lineages and their respective environments. In addition, both OLs and SCs respond to neuronal activity and regulate nervous system output in reciprocal manners, possibly through different pathways. Here, we outline their basic developmental programs, mechanisms regulating their differentiation, and recent advances in the field.
Collapse
Affiliation(s)
- Carlo D. Cristobal
- Integrative Program in Molecular and Biomedical SciencesBaylor College of MedicineHoustonTexasUSA,Jan and Dan Duncan Neurological Research InstituteTexas Children's HospitalHoustonTexasUSA
| | - Hyun Kyoung Lee
- Integrative Program in Molecular and Biomedical SciencesBaylor College of MedicineHoustonTexasUSA,Jan and Dan Duncan Neurological Research InstituteTexas Children's HospitalHoustonTexasUSA,Department of PediatricsBaylor College of MedicineHoustonTexasUSA,Department of NeuroscienceBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
127
|
Nicholson M, Wood RJ, Gonsalvez DG, Hannan AJ, Fletcher JL, Xiao J, Murray SS. Remodelling of myelinated axons and oligodendrocyte differentiation is stimulated by environmental enrichment in the young adult brain. Eur J Neurosci 2022; 56:6099-6114. [PMID: 36217300 PMCID: PMC10092722 DOI: 10.1111/ejn.15840] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 12/29/2022]
Abstract
Oligodendrocyte production and myelination continues lifelong in the central nervous system (CNS), and all stages of this process can be adaptively regulated by neuronal activity. While artificial exogenous stimulation of neuronal circuits greatly enhances oligodendrocyte progenitor cell (OPC) production and increases myelination during development, the extent to which physiological stimuli replicates this is unclear, particularly in the adult CNS when the rate of new myelin addition slows. Here, we used environmental enrichment (EE) to physiologically stimulate neuronal activity for 6 weeks in 9-week-old C57BL/six male and female mice and found no increase in compact myelin in the corpus callosum or somatosensory cortex. Instead, we observed a global increase in callosal axon diameter with thicker myelin sheaths, elongated paranodes and shortened nodes of Ranvier. These findings indicate that EE induced the dynamic structural remodelling of myelinated axons. Additionally, we observed a global increase in the differentiation of OPCs and pre-myelinating oligodendroglia in the corpus callosum and somatosensory cortex. Our findings of structural remodelling of myelinated axons in response to physiological neural stimuli during young adulthood provide important insights in understanding experience-dependent myelin plasticity throughout the lifespan and provide a platform to investigate axon-myelin interactions in a physiologically relevant context.
Collapse
Affiliation(s)
- Madeline Nicholson
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Australia
| | - Rhiannon J Wood
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Australia
| | - David G Gonsalvez
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Anthony J Hannan
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Jessica L Fletcher
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Australia.,Menzies Institute of Medical Research, University of Tasmania, Hobart, Australia
| | - Junhua Xiao
- School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia.,School of Allied Health, La Trobe University, Bundoora, Victoria, Australia
| | - Simon S Murray
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| |
Collapse
|
128
|
Dembitskaya Y, Piette C, Perez S, Berry H, Magistretti PJ, Venance L. Lactate supply overtakes glucose when neural computational and cognitive loads scale up. Proc Natl Acad Sci U S A 2022; 119:e2212004119. [PMID: 36375086 PMCID: PMC9704697 DOI: 10.1073/pnas.2212004119] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/10/2022] [Indexed: 07/23/2023] Open
Abstract
Neural computational power is determined by neuroenergetics, but how and which energy substrates are allocated to various forms of memory engram is unclear. To solve this question, we asked whether neuronal fueling by glucose or lactate scales differently upon increasing neural computation and cognitive loads. Here, using electrophysiology, two-photon imaging, cognitive tasks, and mathematical modeling, we show that both glucose and lactate are involved in engram formation, with lactate supporting long-term synaptic plasticity evoked by high-stimulation load activity patterns and high attentional load in cognitive tasks and glucose being sufficient for less demanding neural computation and learning tasks. Indeed, we show that lactate is mandatory for demanding neural computation, such as theta-burst stimulation, while glucose is sufficient for lighter forms of activity-dependent long-term potentiation (LTP), such as spike timing-dependent plasticity (STDP). We find that subtle variations of spike number or frequency in STDP are sufficient to shift the on-demand fueling from glucose to lactate. Finally, we demonstrate that lactate is necessary for a cognitive task requiring high attentional load, such as the object-in-place task, and for the corresponding in vivo hippocampal LTP expression but is not needed for a less demanding task, such as a simple novel object recognition. Overall, these results demonstrate that glucose and lactate metabolism are differentially engaged in neuronal fueling depending on the complexity of the activity-dependent plasticity and behavior.
Collapse
Affiliation(s)
- Yulia Dembitskaya
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Charlotte Piette
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Sylvie Perez
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Hugues Berry
- AIStroSight Lab, INRIA, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69603 Villeurbanne, France
- University of Lyon, LIRIS UMR5205, 69622 Villeurbanne, France
| | - Pierre J. Magistretti
- Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Saudi Arabia
- Brain Mind Institute, EPFL, 1015 Lausanne, Switzerland
| | - Laurent Venance
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, 75005 Paris, France
| |
Collapse
|
129
|
Wang Y, LeDue JM, Murphy TH. Multiscale imaging informs translational mouse modeling of neurological disease. Neuron 2022; 110:3688-3710. [PMID: 36198319 DOI: 10.1016/j.neuron.2022.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/26/2022] [Accepted: 09/06/2022] [Indexed: 11/05/2022]
Abstract
Multiscale neurophysiology reveals that simple motor actions are associated with changes in neuronal firing in virtually every brain region studied. Accordingly, the assessment of focal pathology such as stroke or progressive neurodegenerative diseases must also extend widely across brain areas. To derive mechanistic information through imaging, multiple resolution scales and multimodal factors must be included, such as the structure and function of specific neurons and glial cells and the dynamics of specific neurotransmitters. Emerging multiscale methods in preclinical animal studies that span micro- to macroscale examinations fill this gap, allowing a circuit-based understanding of pathophysiological mechanisms. Combined with high-performance computation and open-source data repositories, these emerging multiscale and large field-of-view techniques include live functional ultrasound, multi- and single-photon wide-scale light microscopy, video-based miniscopes, and tissue-penetrating fiber photometry, as well as variants of post-mortem expansion microscopy. We present these technologies and outline use cases and data pipelines to uncover new knowledge within animal models of stroke, Alzheimer's disease, and movement disorders.
Collapse
Affiliation(s)
- Yundi Wang
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological Research, Detwiller Pavilion, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Jeffrey M LeDue
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological Research, Detwiller Pavilion, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Timothy H Murphy
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological Research, Detwiller Pavilion, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
130
|
Viney TJ, Sarkany B, Ozdemir AT, Hartwich K, Schweimer J, Bannerman D, Somogyi P. Spread of pathological human Tau from neurons to oligodendrocytes and loss of high-firing pyramidal neurons in aging mice. Cell Rep 2022; 41:111646. [PMID: 36384116 PMCID: PMC9681663 DOI: 10.1016/j.celrep.2022.111646] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 08/23/2022] [Accepted: 10/20/2022] [Indexed: 11/18/2022] Open
Abstract
Intracellular aggregation of hyperphosphorylated Tau (pTau) in the brain is associated with cognitive and motor impairments, and ultimately neurodegeneration. We investigate how human pTau affects cells and network activity in the hippocampal formation of the THY-Tau22 tauopathy model mice in vivo. We find that pTau preferentially accumulates in deep-layer pyramidal neurons, leading to neurodegeneration, and we establish that pTau spreads to oligodendrocytes. During goal-directed virtual navigation in aged transgenic mice, we detect fewer high-firing prosubicular pyramidal cells, but the firing population retains its coupling to theta oscillations. Analysis of network oscillations and firing patterns of pyramidal and GABAergic neurons recorded in head-fixed and freely moving mice suggests preserved neuronal coordination. In spatial memory tests, transgenic mice have reduced short-term familiarity, but spatial working and reference memory are surprisingly normal. We hypothesize that unimpaired subcortical network mechanisms maintain cortical neuronal coordination, counteracting the widespread pTau aggregation, loss of high-firing cells, and neurodegeneration.
Collapse
Affiliation(s)
- Tim J Viney
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK.
| | - Barbara Sarkany
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - A Tugrul Ozdemir
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Katja Hartwich
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Judith Schweimer
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - David Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford OX2 6GG, UK
| | - Peter Somogyi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
131
|
Abstract
Within the past decade, multiple lines of evidence have converged to identify a critical role for activity-regulated myelination in tuning the function of neural networks. In this Review, we provide an overview of accumulating evidence that activity-regulated myelination is required for brain adaptation and learning across multiple domains. We then discuss dysregulation of activity-dependent myelination in the context of neurological disease, a novel frontier with the potential to uncover new mechanisms of disease pathogenesis and to develop new therapeutic strategies. Alterations in myelination and neural network function can result from deficient myelin plasticity that impairs neurological function or from maladaptive myelination, in which intact activity-dependent myelination contributes to the disease process by promoting pathological patterns of neuronal activity. These emerging mechanisms suggest new avenues for therapeutic intervention that could more fully address the complex interactions between neurons and oligodendroglia.
Collapse
|
132
|
Effects of Visual Deprivation on Remodeling of Nodes of Ranvier in Optic Nerve. eNeuro 2022; 9:ENEURO.0194-22.2022. [PMID: 36302632 PMCID: PMC9651206 DOI: 10.1523/eneuro.0194-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/28/2022] [Accepted: 10/05/2022] [Indexed: 12/24/2022] Open
Abstract
Oligodendrocytes, the myelinating cells of the CNS, promote rapid action potential conduction along axons. Changes in the geometry of gaps between myelin segments, known as nodes of Ranvier, affect the conduction speed of neuronal impulses and can ultimately alter neural synchronization and circuit function. In contrast to synaptic plasticity, much less is known about how neural activity may affect node of Ranvier structure. Recently, perinodal astrocytes have been shown to remodel nodes of Ranvier by regulating thrombin proteolysis, but it is not known whether neural activity influences this process. To test this hypothesis, we used transgenic mice with astrocytic expression of a dominant-negative vesicle-associated membrane protein 2 ([gfap]dnVAMP2) to reduce exocytosis of thrombin inhibitors, modulating astrocytic regulation of paranodal loop attachment to induce nodal remodeling, under normal conditions and in adult mice maintained in darkness from postnatal day 40 (P40) to P70. This mechanism of nodal lengthening proceeded normally following binocular visual deprivation (BVD). The effect of BVD on nodal plasticity in animals with unimpaired astrocyte function has not been previously investigated. We find that when exocytosis from astrocytes was unimpaired, nodal gap length was not altered by BVD in adult mice. We conclude that if perinodal astrocytes participate in activity-dependent myelin remodeling through exocytosis, then, as with synaptic plasticity in the visual system, the process must be driven by alterations in neuronal firing other than those produced by BVD.
Collapse
|
133
|
Abstract
Persistent neurological and neuropsychiatric symptoms affect a substantial fraction of people after COVID-19 and represent a major component of the post-acute COVID-19 syndrome, also known as long COVID. Here, we review what is understood about the pathobiology of post-acute COVID-19 impact on the CNS and discuss possible neurobiological underpinnings of the cognitive symptoms affecting COVID-19 survivors. We propose the chief mechanisms that may contribute to this emerging neurological health crisis.
Collapse
Affiliation(s)
- Michelle Monje
- Department of Neurology, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, USA.
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University, USA.
| |
Collapse
|
134
|
Kaya T, Mattugini N, Liu L, Ji H, Cantuti-Castelvetri L, Wu J, Schifferer M, Groh J, Martini R, Besson-Girard S, Kaji S, Liesz A, Gokce O, Simons M. CD8 + T cells induce interferon-responsive oligodendrocytes and microglia in white matter aging. Nat Neurosci 2022; 25:1446-1457. [PMID: 36280798 PMCID: PMC9630119 DOI: 10.1038/s41593-022-01183-6] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 09/14/2022] [Indexed: 01/13/2023]
Abstract
A hallmark of nervous system aging is a decline of white matter volume and function, but the underlying mechanisms leading to white matter pathology are unknown. In the present study, we found age-related alterations of oligodendrocyte cell state with a reduction in total oligodendrocyte density in aging murine white matter. Using single-cell RNA-sequencing, we identified interferon (IFN)-responsive oligodendrocytes, which localize in proximity to CD8+ T cells in aging white matter. Absence of functional lymphocytes decreased the number of IFN-responsive oligodendrocytes and rescued oligodendrocyte loss, whereas T-cell checkpoint inhibition worsened the aging response. In addition, we identified a subpopulation of lymphocyte-dependent, IFN-responsive microglia in the vicinity of the CD8+ T cells in aging white matter. In summary, we provide evidence that CD8+ T-cell-induced, IFN-responsive oligodendrocytes and microglia are important modifiers of white matter aging.
Collapse
Affiliation(s)
- Tuğberk Kaya
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Nicola Mattugini
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Lu Liu
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - Hao Ji
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - Ludovico Cantuti-Castelvetri
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Jianping Wu
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster of Systems Neurology, Munich, Germany
| | - Janos Groh
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Rudolf Martini
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Simon Besson-Girard
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Seiji Kaji
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany.
- Munich Cluster of Systems Neurology, Munich, Germany.
| | - Mikael Simons
- Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig Maximilian University (LMU) of Munich, Munich, Germany.
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases, Munich, Germany.
- Munich Cluster of Systems Neurology, Munich, Germany.
| |
Collapse
|
135
|
Oligodendroglia are emerging players in several forms of learning and memory. Commun Biol 2022; 5:1148. [PMID: 36309567 PMCID: PMC9617857 DOI: 10.1038/s42003-022-04116-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/14/2022] [Indexed: 11/15/2022] Open
Abstract
Synaptic plasticity is the fundamental cellular mechanism of learning and memory, but recent research reveals that myelin-forming glia, oligodendrocytes (OL), are also involved. They contribute in ways that synaptic plasticity cannot, and the findings have not been integrated into the established conceptual framework used in the field of learning and memory. OLs and their progenitors are involved in long-term memory, memory consolidation, working memory, and recall in associative learning. They also contribute to short-term memory and non-associative learning by affecting synaptic transmission, intrinsic excitability of axons, and neural oscillations. Oligodendroglial involvement expands the field beyond synaptic plasticity to system-wide network function, where precise spike time arrival and neural oscillations are critical in information processing, storage, and retrieval. A Perspective highlights current evidence that supports oligodendrocytes and their progenitors’ involvement in cognition and proposes that our understanding of learning and memory can be expanded beyond the classic view of synaptic plasticity to a system-wide network function.
Collapse
|
136
|
Osanai Y, Yamazaki R, Shinohara Y, Ohno N. Heterogeneity and regulation of oligodendrocyte morphology. Front Cell Dev Biol 2022; 10:1030486. [PMID: 36393856 PMCID: PMC9644283 DOI: 10.3389/fcell.2022.1030486] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/10/2022] [Indexed: 09/24/2023] Open
Abstract
Oligodendrocytes form multiple myelin sheaths in the central nervous system (CNS), which increase nerve conduction velocity and are necessary for basic and higher brain functions such as sensory function, motor control, and learning. Structures of the myelin sheath such as myelin internodal length and myelin thickness regulate nerve conduction. Various parts of the central nervous system exhibit different myelin structures and oligodendrocyte morphologies. Recent studies supported that oligodendrocytes are a heterogenous population of cells and myelin sheaths formed by some oligodendrocytes can be biased to particular groups of axons, and myelin structures are dynamically modulated in certain classes of neurons by specific experiences. Structures of oligodendrocyte/myelin are also affected in pathological conditions such as demyelinating and neuropsychiatric disorders. This review summarizes our understanding of heterogeneity and regulation of oligodendrocyte morphology concerning central nervous system regions, neuronal classes, experiences, diseases, and how oligodendrocytes are optimized to execute central nervous system functions.
Collapse
Affiliation(s)
- Yasuyuki Osanai
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Reiji Yamazaki
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Yoshiaki Shinohara
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
137
|
Talidou A, Frankland PW, Mabbott D, Lefebvre J. Homeostatic coordination and up-regulation of neural activity by activity-dependent myelination. NATURE COMPUTATIONAL SCIENCE 2022; 2:665-676. [PMID: 38177260 DOI: 10.1038/s43588-022-00315-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/10/2022] [Indexed: 01/06/2024]
Abstract
Activity-dependent myelination (ADM) is a fundamental dimension of brain plasticity through which myelin changes as a function of neural activity. Mediated by structural changes in glia, ADM notably regulates axonal conduction velocity. Yet, it remains unclear how neural activity impacts myelination to orchestrate the timing of neural signalling, and how ADM shapes neural activity. We developed a model of spiking neurons enhanced with neuron-oligodendrocyte feedback and examined the relationship between ADM and neural activity. We found that ADM implements a homeostatic gain control mechanism that enhances neural firing rates and correlations through the temporal coordination of action potentials as axon lengths increase. Stimuli engage ADM plasticity to trigger bidirectional and reversible changes in conduction delays, as may occur during learning. Furthermore, ADM was found to enhance information transmission under various types of time-varying stimuli. These results highlight the role of ADM in shaping neural activity and communication.
Collapse
Affiliation(s)
- Afroditi Talidou
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada.
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
| | - Paul W Frankland
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Donald Mabbott
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Jérémie Lefebvre
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physics, University of Ottawa, Ottawa, Ontario, Canada
- Department of Mathematics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
138
|
Bacmeister CM, Huang R, Osso LA, Thornton MA, Conant L, Chavez AR, Poleg-Polsky A, Hughes EG. Motor learning drives dynamic patterns of intermittent myelination on learning-activated axons. Nat Neurosci 2022; 25:1300-1313. [PMID: 36180791 PMCID: PMC9651929 DOI: 10.1038/s41593-022-01169-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 08/18/2022] [Indexed: 01/10/2023]
Abstract
Myelin plasticity occurs when newly formed and pre-existing oligodendrocytes remodel existing patterns of myelination. Myelin remodeling occurs in response to changes in neuronal activity and is required for learning and memory. However, the link between behavior-induced neuronal activity and circuit-specific changes in myelination remains unclear. Using longitudinal in vivo two-photon imaging and targeted labeling of learning-activated neurons in mice, we explore how the pattern of intermittent myelination is altered on individual cortical axons during learning of a dexterous reach task. We show that behavior-induced myelin plasticity is targeted to learning-activated axons and occurs in a staged response across cortical layers in the mouse primary motor cortex. During learning, myelin sheaths retract, which results in lengthening of nodes of Ranvier. Following motor learning, addition of newly formed myelin sheaths increases the number of continuous stretches of myelination. Computational modeling suggests that motor learning-induced myelin plasticity initially slows and subsequently increases axonal conduction speed. Finally, we show that both the magnitude and timing of nodal and myelin dynamics correlate with improvement of behavioral performance during motor learning. Thus, learning-induced and circuit-specific myelination changes may contribute to information encoding in neural circuits during motor learning.
Collapse
Affiliation(s)
- Clara M Bacmeister
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
- Neuroscience IDP Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Rongchen Huang
- Department of Neurosurgery, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lindsay A Osso
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael A Thornton
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lauren Conant
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Anthony R Chavez
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Alon Poleg-Polsky
- Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ethan G Hughes
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
139
|
Altered activity-regulated H3K9 acetylation at TGF-beta signaling genes during egocentric memory in Huntington's disease. Prog Neurobiol 2022; 219:102363. [PMID: 36179935 DOI: 10.1016/j.pneurobio.2022.102363] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/25/2022] [Accepted: 09/24/2022] [Indexed: 11/21/2022]
Abstract
Molecular mechanisms underlying cognitive deficits in Huntington's disease (HD), a striatal neurodegenerative disorder, are unknown. Here, we generated ChIPseq, 4Cseq and RNAseq data on striatal tissue of HD and control mice during striatum-dependent egocentric memory process. Multi-omics analyses showed altered activity-dependent epigenetic gene reprogramming of neuronal and glial genes regulating striatal plasticity in HD mice, which correlated with memory deficit. First, our data reveal that spatial chromatin re-organization and transcriptional induction of BDNF-related markers, regulating neuronal plasticity, were reduced since memory acquisition in the striatum of HD mice. Second, our data show that epigenetic memory implicating H3K9 acetylation, which established during late phase of memory process (e.g. during consolidation/recall) and contributed to glia-mediated, TGFβ-dependent plasticity, was compromised in HD mouse striatum. Specifically, memory-dependent regulation of H3K9 acetylation was impaired at genes controlling extracellular matrix and myelination. Our study investigating the interplay between epigenetics and memory identifies H3K9 acetylation and TGFβ signaling as new targets of striatal plasticity, which might offer innovative leads to improve HD.
Collapse
|
140
|
Disruptions in white matter microstructure associated with impaired visual associative memory in schizophrenia-spectrum illness. Eur Arch Psychiatry Clin Neurosci 2022; 272:971-983. [PMID: 34557990 DOI: 10.1007/s00406-021-01333-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022]
Abstract
Episodic memory ability relies on hippocampal-prefrontal connectivity. However, few studies have examined relationships between memory performance and white matter (WM) microstructure in hippocampal-prefrontal pathways in schizophrenia-spectrum disorder (SSDs). Here, we investigated these relationships in individuals with first-episode psychosis (FEP) and chronic schizophrenia-spectrum disorders (SSDs) using tractography analysis designed to interrogate the microstructure of WM tracts in the hippocampal-prefrontal pathway. Measures of WM microstructure (fractional anisotropy [FA], radial diffusivity [RD], and axial diffusivity [AD]) were obtained for 47 individuals with chronic SSDs, 28 FEP individuals, 52 older healthy controls, and 27 younger healthy controls. Tractography analysis was performed between the hippocampus and three targets involved in hippocampal-prefrontal connectivity (thalamus, amygdala, nucleus accumbens). Measures of WM microstructure were then examined in relation to episodic memory performance separately across each group. Both those with FEP and chronic SSDs demonstrated impaired episodic memory performance. However, abnormal WM microstructure was only observed in individuals with chronic SSDs. Abnormal WM microstructure in the hippocampal-thalamic pathway in the right hemisphere was associated with poorer memory performance in individuals with chronic SSDs. These findings suggest that disruptions in WM microstructure in the hippocampal-prefrontal pathway may contribute to memory impairments in individuals with chronic SSDs but not FEP.
Collapse
|
141
|
Pan Y, Monje M. Neuron-Glial Interactions in Health and Brain Cancer. Adv Biol (Weinh) 2022; 6:e2200122. [PMID: 35957525 PMCID: PMC9845196 DOI: 10.1002/adbi.202200122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/21/2022] [Indexed: 01/28/2023]
Abstract
Brain tumors are devastating diseases of the central nervous system. Brain tumor pathogenesis depends on both tumor-intrinsic oncogenic programs and extrinsic microenvironmental factors, including neurons and glial cells. Glial cells (oligodendrocytes, astrocytes, and microglia) make up half of the cells in the brain, and interact with neurons to modulate neurodevelopment and plasticity. Many brain tumor cells exhibit transcriptomic profiles similar to macroglial cells (oligodendrocytes and astrocytes) and their progenitors, making them likely to subvert existing neuron-glial interactions to support tumor pathogenesis. For example, oligodendrocyte precursor cells, a putative glioma cell of origin, can form bona fide synapses with neurons. Such synapses are recently identified in gliomas and drive glioma pathophysiology, underscoring how brain tumor cells can take advantage of neuron-glial interactions to support cancer progression. In this review, it is briefly summarized how neurons and their activity normally interact with glial cells and glial progenitors, and it is discussed how brain tumor cells utilize neuron-glial interactions to support tumor initiation and progression. Unresolved questions on these topics and potential avenues to therapeutically target neuron-glia-cancer interactions in the brain are also pointed out.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Symptom Research, University of Texas MD Anderson Cancer Center,co-corresponding: ;
| | - Michelle Monje
- Department of Neurology, Stanford University,Howard Hughes Medical Institute, Stanford University,co-corresponding: ;
| |
Collapse
|
142
|
Savchuk S, Monje M. Mini-Review: Aplastic Myelin Following Chemotherapy. Neurosci Lett 2022; 790:136861. [PMID: 36055447 DOI: 10.1016/j.neulet.2022.136861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/12/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022]
Abstract
The contribution of chemotherapy to improved outcomes for cancer patients is unquestionable. Yet as its applications broaden, so do the concerns for the long-term implications of chemotherapy on the health of cancer survivors, with chemotherapy-related cognitive impairment as a cause for particular urgency. In this mini review, we explore myelin aplasticity following chemotherapy, discussing the role of myelin plasticity in healthy cognition and failure of myelin plasticity chiefly due microenvironmental aberrations in chemotherapy-related cognitive impairment. Possible therapeutic strategies to mitigate chemotherapy-induced myelin dysfunction are also discussed.
Collapse
Affiliation(s)
- Solomiia Savchuk
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA; Department of Neurosurgery, Stanford University, Stanford, CA, 94305, USA; Department of Pathology, Stanford University, Stanford, CA, 94305, USA; Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
143
|
Hong J, Jeong Y, Heo WD. The Neurotrophic Receptor Tyrosine Kinase in MEC-mPFC Neurons Contributes to Remote Memory Consolidation. J Neurosci 2022; 42:6605-6619. [PMID: 35863892 PMCID: PMC9410758 DOI: 10.1523/jneurosci.2433-21.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 06/08/2022] [Accepted: 07/06/2022] [Indexed: 11/21/2022] Open
Abstract
The PFC is thought to be the region where remote memory is recalled. However, the neurotrophic receptors that underlie the remote memory remain largely unknown. Here, we benefited from auto-assembly split Cre to accomplish the neural projection-specific recombinase activity without spontaneous leakage. Deletion of tropomyosin receptor kinase B (TrkB) in neurons projecting from the medial entorhinal cortex to the mPFC displayed reduced remote memory recall from the male mice, but the recent recall was intact. We found that the TrkB deletion attenuates the participation of mPFC cells in the remote fear memory recall. The disruption of remote recall was attributed to reduced reactivation of cells in the mPFC. Notably, TrkB deletion seriously inhibited experience-dependent maturation of oligodendroglia in the PFC, resulting in defects in remote recall that were rescued by clemastine administration. Together, our data suggest that TrkB in intercortical circuits functions in remote memory consolidation.SIGNIFICANCE STATEMENT Retrieving the past experiences or events is essential for the ones to lead life. The investigations performed in the rodent model have disclosed that the systems consolidation of memory accompanying changes of cortical circuits and transcriptome is required for maintaining the memory for a long time. In this study, the split Cre with TrkBflox/flox mice were subjected to discover that TrkB in the neurons plays a role in remote memory consolidation. We evaluated the contextual fear memory and labeled cells, which revealed deletion of TrkB interrupts newborn oligodendrocyte and reactivation of cells in mPFC at remote recall. Our data provide the implication that remote memory is relevant to neurotrophic receptor signaling as well as its influence on non-neuronal cells.
Collapse
Affiliation(s)
- Jongryul Hong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Yeonji Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- Korea Advanced Institute of Science and Technology Institute for the BioCentury, Daejeon, 34141, Republic of Korea
| |
Collapse
|
144
|
Zhao F, Li B, Yang W, Ge T, Cui R. Brain-immune interaction mechanisms: Implications for cognitive dysfunction in psychiatric disorders. Cell Prolif 2022; 55:e13295. [PMID: 35860850 PMCID: PMC9528770 DOI: 10.1111/cpr.13295] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 04/28/2022] [Accepted: 06/07/2022] [Indexed: 12/15/2022] Open
Abstract
Objectives Cognitive dysfunction has been identified as a major symptom of a series of psychiatric disorders. Multidisciplinary studies have shown that cognitive dysfunction is monitored by a two‐way interaction between the neural and immune systems. However, the specific mechanisms of cognitive dysfunction in immune response and brain immune remain unclear. Materials and methods In this review, we summarized the relevant research to uncover our comprehension of the brain–immune interaction mechanisms underlying cognitive decline. Results The pathophysiological mechanisms of brain‐immune interactions in psychiatric‐based cognitive dysfunction involve several specific immune molecules and their associated signaling pathways, impairments in neural and synaptic plasticity, and the potential neuro‐immunological mechanism of stress. Conclusions Therefore, this review may provide a better theoretical basis for integrative therapeutic considerations for psychiatric disorders associated with cognitive dysfunction.
Collapse
Affiliation(s)
- Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
145
|
Fernández-Castañeda A, Lu P, Geraghty AC, Song E, Lee MH, Wood J, O'Dea MR, Dutton S, Shamardani K, Nwangwu K, Mancusi R, Yalçın B, Taylor KR, Acosta-Alvarez L, Malacon K, Keough MB, Ni L, Woo PJ, Contreras-Esquivel D, Toland AMS, Gehlhausen JR, Klein J, Takahashi T, Silva J, Israelow B, Lucas C, Mao T, Peña-Hernández MA, Tabachnikova A, Homer RJ, Tabacof L, Tosto-Mancuso J, Breyman E, Kontorovich A, McCarthy D, Quezado M, Vogel H, Hefti MM, Perl DP, Liddelow S, Folkerth R, Putrino D, Nath A, Iwasaki A, Monje M. Mild respiratory COVID can cause multi-lineage neural cell and myelin dysregulation. Cell 2022; 185:2452-2468.e16. [PMID: 35768006 PMCID: PMC9189143 DOI: 10.1016/j.cell.2022.06.008] [Citation(s) in RCA: 308] [Impact Index Per Article: 102.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/04/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022]
Abstract
COVID survivors frequently experience lingering neurological symptoms that resemble cancer-therapy-related cognitive impairment, a syndrome for which white matter microglial reactivity and consequent neural dysregulation is central. Here, we explored the neurobiological effects of respiratory SARS-CoV-2 infection and found white-matter-selective microglial reactivity in mice and humans. Following mild respiratory COVID in mice, persistently impaired hippocampal neurogenesis, decreased oligodendrocytes, and myelin loss were evident together with elevated CSF cytokines/chemokines including CCL11. Systemic CCL11 administration specifically caused hippocampal microglial reactivity and impaired neurogenesis. Concordantly, humans with lasting cognitive symptoms post-COVID exhibit elevated CCL11 levels. Compared with SARS-CoV-2, mild respiratory influenza in mice caused similar patterns of white-matter-selective microglial reactivity, oligodendrocyte loss, impaired neurogenesis, and elevated CCL11 at early time points, but after influenza, only elevated CCL11 and hippocampal pathology persisted. These findings illustrate similar neuropathophysiology after cancer therapy and respiratory SARS-CoV-2 infection which may contribute to cognitive impairment following even mild COVID.
Collapse
Affiliation(s)
| | - Peiwen Lu
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Anna C Geraghty
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Eric Song
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Myoung-Hwa Lee
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Jamie Wood
- Abilities Research Center, Department of Rehabilitation and Human Performance, Mount Sinai School of Medicine, New York, NY, USA
| | - Michael R O'Dea
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Selena Dutton
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Kiarash Shamardani
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Kamsi Nwangwu
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Rebecca Mancusi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Belgin Yalçın
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Kathryn R Taylor
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Lehi Acosta-Alvarez
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Karen Malacon
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michael B Keough
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Lijun Ni
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Pamelyn J Woo
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | | | | | | | - Jon Klein
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | | | - Julio Silva
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | | | - Carolina Lucas
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Tianyang Mao
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | | | | | - Robert J Homer
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Laura Tabacof
- Abilities Research Center, Department of Rehabilitation and Human Performance, Mount Sinai School of Medicine, New York, NY, USA
| | - Jenna Tosto-Mancuso
- Abilities Research Center, Department of Rehabilitation and Human Performance, Mount Sinai School of Medicine, New York, NY, USA
| | - Erica Breyman
- Abilities Research Center, Department of Rehabilitation and Human Performance, Mount Sinai School of Medicine, New York, NY, USA
| | - Amy Kontorovich
- Cardiovascular Research Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Dayna McCarthy
- Abilities Research Center, Department of Rehabilitation and Human Performance, Mount Sinai School of Medicine, New York, NY, USA
| | | | - Hannes Vogel
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Marco M Hefti
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | - Daniel P Perl
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, USA
| | - Shane Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA; Departments of Neuroscience & Physiology and of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA; Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | | | - David Putrino
- Abilities Research Center, Department of Rehabilitation and Human Performance, Mount Sinai School of Medicine, New York, NY, USA
| | - Avindra Nath
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University, New Haven, CT, USA; Howard Hughes Medical Institute, Yale University, New Haven, CT, USA.
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
146
|
Ebrahim Soltani Z, Badripour A, Haddadi NS, Elahi M, Kazemi K, Afshari K, Dehpour A. Allergic rhinitis in BALB/c mice is associated with behavioral and hippocampus changes and neuroinflammation via the TLR4/ NF-κB signaling pathway. Int Immunopharmacol 2022; 108:108725. [DOI: 10.1016/j.intimp.2022.108725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/02/2022] [Accepted: 03/18/2022] [Indexed: 11/26/2022]
|
147
|
Abstract
Acute mild respiratory SARS-CoV-2 infection can lead to a more chronic cognitive syndrome known as “COVID fog.” New findings from Fernández-Castañeda et al. reveal how glial dysregulation and consequent neural circuit dysfunction may contribute to cognitive impairments in long COVID.
Collapse
|
148
|
Ridderinkhof KR, Krugers HJ. Horizons in Human Aging Neuroscience: From Normal Neural Aging to Mental (Fr)Agility. Front Hum Neurosci 2022; 16:815759. [PMID: 35845248 PMCID: PMC9277589 DOI: 10.3389/fnhum.2022.815759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
While aging is an important risk factor for neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease, age-related cognitive decline can also manifest without apparent neurodegenerative changes. In this review, we discuss molecular, cellular, and network changes that occur during normal aging in the absence of neurodegenerative disease. Emerging findings reveal that these changes include metabolic alterations, oxidative stress, DNA damage, inflammation, calcium dyshomeostasis, and several other hallmarks of age-related neural changes that do not act on their own, but are often interconnected and together may underlie age-related alterations in brain plasticity and cognitive function. Importantly, age-related cognitive decline may not be reduced to a single neurobiological cause, but should instead be considered in terms of a densely connected system that underlies age-related cognitive alterations. We speculate that a decline in one hallmark of neural aging may trigger a decline in other, otherwise thus far stable subsystems, thereby triggering a cascade that may at some point also incur a decline of cognitive functions and mental well-being. Beyond studying the effects of these factors in isolation, considerable insight may be gained by studying the larger picture that entails a representative collection of such factors and their interactions, ranging from molecules to neural networks. Finally, we discuss some potential interventions that may help to prevent these alterations, thereby reducing cognitive decline and mental fragility, and enhancing mental well-being, and healthy aging.
Collapse
Affiliation(s)
- K. Richard Ridderinkhof
- Department of Psychology, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Center for Brain and Cognition (ABC), University of Amsterdam, Amsterdam, Netherlands
| | - Harm J. Krugers
- Amsterdam Center for Brain and Cognition (ABC), University of Amsterdam, Amsterdam, Netherlands
- SILS-CNS, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
149
|
Reduced and delayed myelination and volume of corpus callosum in an animal model of Fetal Alcohol Spectrum Disorders partially benefit from voluntary exercise. Sci Rep 2022; 12:10653. [PMID: 35739222 PMCID: PMC9226126 DOI: 10.1038/s41598-022-14752-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/13/2022] [Indexed: 11/27/2022] Open
Abstract
1 in 20 live births in the United States is affected by prenatal alcohol exposure annually, creating a major public health crisis. The teratogenic impact of alcohol on physical growth, neurodevelopment, and behavior is extensive, together resulting in clinical disorders which fall under the umbrella term of Fetal Alcohol Spectrum Disorders (FASD). FASD-related impairments to executive function and perceptual learning are prevalent among affected youth and are linked to disruptions to corpus callosum growth and myelination in adolescence. Targeted interventions that support neurodevelopment in FASD-affected youth are nonexistent. We evaluated the capacity of an adolescent exercise intervention, a stimulator of myelinogenesis, to upregulate corpus callosum myelination in a rat model of FASD (third trimester-equivalent alcohol exposure). This study employs in vivo diffusion tensor imaging (DTI) scanning to investigate the effects of: (1) neonatal alcohol exposure and (2) an adolescent exercise intervention on corpus callosum myelination in a rodent model of FASD. DTI scans were acquired twice longitudinally (pre- and post-intervention) in male and female rats using a 9.4 Tesla Bruker Biospec scanner to assess alterations to corpus callosum myelination noninvasively. Fractional anisotropy values as well as radial/axial diffusivity values were compared within-animal in a longitudinal study design. Analyses using mixed repeated measures ANOVA’s confirm that neonatal alcohol exposure in a rodent model of FASD delays the trajectory of corpus callosum growth and myelination across adolescence, with a heightened vulnerability in the male brain. Alterations to corpus callosum volume are correlated with reductions to forebrain volume which mediates an indirect relationship between body weight gain and corpus callosum growth. While we did not observe any significant effects of voluntary aerobic exercise on corpus callosum myelination immediately after completion of the 12-day intervention, we did observe a beneficial effect of exercise intervention on corpus callosum volume growth in all rats. In line with clinical findings, we have shown that prenatal alcohol exposure leads to hypomyelination of the corpus callosum in adolescence and that the severity of damage is sexually dimorphic. Further, exercise intervention improves corpus callosum growth in alcohol-exposed and control rats in adolescence.
Collapse
|
150
|
Lazari A, Salvan P, Cottaar M, Papp D, Rushworth MFS, Johansen-Berg H. Hebbian activity-dependent plasticity in white matter. Cell Rep 2022; 39:110951. [PMID: 35705046 PMCID: PMC9376741 DOI: 10.1016/j.celrep.2022.110951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/07/2022] [Accepted: 05/23/2022] [Indexed: 11/28/2022] Open
Abstract
Synaptic plasticity is required for learning and follows Hebb's rule, the computational principle underpinning associative learning. In recent years, a complementary type of brain plasticity has been identified in myelinated axons, which make up the majority of brain's white matter. Like synaptic plasticity, myelin plasticity is required for learning, but it is unclear whether it is Hebbian or whether it follows different rules. Here, we provide evidence that white matter plasticity operates following Hebb's rule in humans. Across two experiments, we find that co-stimulating cortical areas to induce Hebbian plasticity leads to relative increases in cortical excitability and associated increases in a myelin marker within the stimulated fiber bundle. We conclude that Hebbian plasticity extends beyond synaptic changes and can be observed in human white matter fibers.
Collapse
Affiliation(s)
- Alberto Lazari
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX2 6GG, UK.
| | - Piergiorgio Salvan
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX2 6GG, UK
| | - Michiel Cottaar
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX2 6GG, UK
| | - Daniel Papp
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX2 6GG, UK
| | - Matthew F S Rushworth
- Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford OX2 6GG, UK
| | - Heidi Johansen-Berg
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX2 6GG, UK
| |
Collapse
|