101
|
Jung GA, Kim JA, Park HW, Lee H, Chang MS, Cho KO, Song BW, Kim HJ, Kwon YK, Oh IH. Induction of Nanog in neural progenitor cells for adaptive regeneration of ischemic brain. Exp Mol Med 2022; 54:1955-1966. [PMID: 36376495 PMCID: PMC9722910 DOI: 10.1038/s12276-022-00880-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 11/15/2022] Open
Abstract
NANOG plays a key role in cellular plasticity and the acquisition of the stem cell state during reprogramming, but its role in the regenerative process remains unclear. Here, we show that the induction of NANOG in neuronal cells is necessary for the physiological initiation of neuronal regeneration in response to ischemic stress. Specifically, we found that NANOG was preferentially expressed in undifferentiated neuronal cells, and forced expression of Nanog in neural progenitor cells (NPCs) promoted their self-renewing expansion both in ex-vivo slice cultures and in vitro limiting dilution analysis. Notably, the upstream region of the Nanog gene contains sequence motifs for hypoxia-inducible factor-1 alpha (HIF-1α). Therefore, cerebral neurons exposed to hypoxia significantly upregulated NANOG expression selectively in primitive (CD133+) cells, but not in mature cells, leading to the expansion of NPCs. Notably, up to 80% of the neuronal expansion induced by hypoxia was attributed to NANOG-expressing neuronal cells, whereas knockdown during hypoxia abolished this expansion and was accompanied by the downregulation of other pluripotency-related genes. Moreover, the number of NANOG-expressing neuronal cells were transiently increased in response to ischemic insult, predominantly in the infarct area of brain regions undergoing neurogenesis, but not in non-neurogenic loci. Together, these findings reveal a functional effect of NANOG-induction for the initiation of adaptive neuronal regeneration among heterogeneous NPC subsets, pointing to cellular plasticity as a potential link between regeneration and reprogramming processes.
Collapse
Affiliation(s)
- Gyung-Ah Jung
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jin-A Kim
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hwan-Woo Park
- Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, Seoul, Korea
- Department of Cell Biology, Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon, Korea
| | - Hyemi Lee
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Mi-Sook Chang
- Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, Seoul, Korea
| | - Kyung-Ok Cho
- Department of Pharmacology, Department of Biomedicine & Health Sciences, Catholic Neuroscience Institute, Institute of Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byeong-Wook Song
- College of Medicine, Institute for Bio-Medical Convergence, Catholic Kwandong University, Gangneung-si, 25601, Korea
| | - Hyun-Ju Kim
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea.
| | - Yunhee Kim Kwon
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea.
| | - Il-Hoan Oh
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, Korea.
- Institute for Regenerative Medical Research, StemMeditech Inc., Seoul, Korea.
| |
Collapse
|
102
|
Manterola A, Chara JC, Aguado T, Palazuelos J, Matute C, Mato S. Cannabinoid CB1 receptor expression in oligodendrocyte progenitors of the hippocampus revealed by the NG2-EYFP-knockin mouse. Front Neuroanat 2022; 16:1030060. [DOI: 10.3389/fnana.2022.1030060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Adult oligodendrocyte progenitor cells (OPCs) give rise to myelinating oligodendrocytes through life and play crucial roles in brain homeostasis and plasticity during health and disease. Cannabinoid compounds acting through CB1 receptors promote the proliferation and differentiation of OPCs in vitro and facilitate developmental myelination and myelin repair in vivo. However, CB1 receptor expression in adult OPCs in situ has not been corroborated by anatomical studies and the contribution of this receptor population to the (re)myelination effects of cannabinoids remains a matter of debate. Using electron microscopy methods applied to NG2-EYFP reporter mice we assessed the localization of CB1 receptors in OPCs of the adult mouse hippocampus. To control for the specificity of CB1 receptor immunostaining we generated transgenic mice bearing EYFP expression in NG2 glia and wild-type (NG2-EYFP-CB1+/+) and knockout (NG2-EYFP-CB1–/–) for CB1 receptors. Double immunogold and immunoperoxidase labeling for CB1 and EYFP, respectively, revealed that CB1 receptors are present in a low proportion of NG2 positive profiles within hippocampal stratum radiatum of NG2-EYFP-CB1+/+ mice. Quantitative analysis of immunogold particles in synaptic structures and NG2 profiles showed that CB1 receptors are expressed at lower density in adult OPCs than in glutamatergic cells of the rodent hippocampus. These results highlight the presence of CB1 receptors in adult OPCs thus providing an anatomical substrate for the remyelination promoting effects of cannabinoids and open a novel perspective on the roles of the endocannabinoid system in brain physiology through the modulation of NG2 glia.
Collapse
|
103
|
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
|
104
|
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
|
105
|
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
|
106
|
Li N, Yao M, Liu J, Zhu Z, Lam TL, Zhang P, Kiang KMY, Leung GKK. Vitamin D Promotes Remyelination by Suppressing c-Myc and Inducing Oligodendrocyte Precursor Cell Differentiation after Traumatic Spinal Cord Injury. Int J Biol Sci 2022; 18:5391-5404. [PMID: 36147469 PMCID: PMC9461656 DOI: 10.7150/ijbs.73673] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/13/2022] [Indexed: 11/22/2022] Open
Abstract
Demyelination due to oligodendrocytes loss occurs after traumatic spinal cord injury (TSCI). Several studies have suggested the therapeutic potential of vitamin D (VitD) in demyelinating diseases. However, experimental evidence in the context of TSCI is limited, particularly in the presence of prior VitD-deficiency. In the present study, a contusion and a transection TSCI rat model were used, representing mild and severe injury, respectively. Motor recovery was assessed in rats with normal VitD level or with VitD-deficiency after 8 weeks' treatment post-TSCI (Cholecalciferol, 500 IU/kg/day). The impact on myelin integrity was examined by transmission electron microscopy and studied in vitro using primary culture of oligodendrocytes. We found that VitD treatment post-TSCI effectively improved hindlimb movement in rats with normal VitD level irrespective of injury severity. However, cord-transected rats with prior deficiency did not seem to benefit from VitD supplementation. Our data further suggested that having sufficient VitD was essential for persevering myelin integrity after injury. VitD rescued oligodendrocytes from apoptotic cell death in vitro and enhanced their myelinating ability towards dorsal root axons. Enhanced myelination was mediated by increased oligodendrocyte precursor cells (OPCs) differentiation into oligodendrocytes in concert with c-Myc downregulation and suppressed OPCs proliferation. Our study provides novel insights into the functioning of VitD as a regulator of OPCs differentiation as well as strong preclinical evidence supporting future clinical testing of VitD for TSCI.
Collapse
Affiliation(s)
- Ning Li
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong.,Department of Neurosurgery, Zhongda Hospital, Southeast University, Nanjing, China
| | - Min Yao
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong.,School of Pharmaceutical Sciences, Health Science Centre, Shenzhen University, Shenzhen, China
| | - Jiaxin Liu
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Zhiyuan Zhu
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong.,Department of Functional Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tsz-Lung Lam
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Pingde Zhang
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Karrie Mei-Yee Kiang
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Gilberto Ka-Kit Leung
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| |
Collapse
|
107
|
Wang Z, Zhang Y, Feng W, Pang Y, Chen S, Ding S, Chen Y, Sheng C, Marshall C, Shi J, Xiao M. Miconazole Promotes Cooperative Ability of a Mouse Model of Alzheimer Disease. Int J Neuropsychopharmacol 2022; 25:951-967. [PMID: 36112386 PMCID: PMC9670758 DOI: 10.1093/ijnp/pyac061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/10/2022] [Accepted: 09/08/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Cooperative defect is 1 of the earliest manifestations of disease patients with Alzheimer disease (AD) exhibit, but the underlying mechanism remains unclear. METHODS We evaluated the cooperative function of APP/PS1 transgenic AD model mice at ages 2, 5, and 8 months by using a cooperative drinking task. We examined neuropathologic changes in the medial prefrontal cortex (mPFC). Another experiment was designed to observe whether miconazole, which has a repairing effect on myelin sheath, could promote the cooperative ability of APP/PS1 mice in the early AD-like stage. We also investigated the protective effects of miconazole on cultured mouse cortical oligodendrocytes exposed to human amyloid β peptide (Aβ1-42). RESULTS We observed an age-dependent impairment of cooperative water drinking behavior in APP/PS1 mice. The AD mice with cooperative dysfunction showed decreases in myelin sheath thickness, oligodendrocyte nuclear heterochromatin percentage, and myelin basic protein expression levels in the mPFC. The cooperative ability was significantly improved in APP/PS1 mice treated with miconazole. Miconazole treatment increased oligodendrocyte maturation and myelin sheath thickness without reducing Aβ plaque deposition, reactive gliosis, and inflammatory factor levels in the mPFC. Miconazole also protected cultured oligodendrocytes from the toxicity of Aβ1-42. CONCLUSIONS These results demonstrate that mPFC hypomyelination is involved in the cooperative deficits of APP/PS1 mice. Improving myelination through miconazole therapy may offer a potential therapeutic approach for early intervention in AD.
Collapse
Affiliation(s)
| | | | - Weixi Feng
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yingting Pang
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Sijia Chen
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Shixin Ding
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Chen
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chengyu Sheng
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Charles Marshall
- Department of Rehabilitation Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, USA
| | - Jingping Shi
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China,Department of Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Correspondence: Ming Xiao, MD, PhD, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, No. 101 Longmian Ave, Nanjing 211166, China ()
| |
Collapse
|
108
|
The role of depolarizing activation of Na +-Ca 2+ exchanger by oligodendrocyte progenitor cells in the effect of sevoflurane on myelination. Life Sci 2022; 308:120951. [PMID: 36103958 DOI: 10.1016/j.lfs.2022.120951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 10/31/2022]
Abstract
AIMS The aim of this study was to investigate the role of depolarizing activation of Na+-Ca2+ exchanger (NCX) by oligodendrocyte progenitor cells (OPC) in the effect of sevoflurane on myelination. MAIN METHODS On postnatal days 7, 8, and 9, mice were exposed to 3 % sevoflurane for 2 h per day. The proliferation, differentiation, and myelin sheath of OPC were observed with immunofluorescence, quantitative real-time polymerase chain reaction (QRT-PCR), and transmission electron microscopy (TEM) at various time points. The open field, Y maze, and new object recognition tests were used to measure spatial learning and memory. siRNA was used for the knockdown NCX1 in human OPC (HOPC) before sevoflurane exposure; the Transwell migration assay was used to measure cell migration ability and Fluo 4-AM was used to measure intracellular Ca2+ concentration. KEY FINDINGS Pretreatment with an NCX inhibitor attenuated the proliferation and differentiation of OPC induced by sevoflurane and induced a remarkable increase in platelet-derived growth factor receptor-alpha (PDGFRα), 2, 3-cyclic nucleotide 3-phosphodiesterase (CNPase), oligodendrocyte transcription factor 2 (Olig2), and homeodomain protein NK2 homeobox 2 (NKX2.2) levels. Pretreatment with an NCX inhibitor alleviated the sevoflurane-induced myelination disorder and cognitive impairment. The decreased cell migration and increased intracellular Ca2+ concentration observed in the siRNA-negative control group was reversed in the sevoflurane plus siRNA-NCX1 group. SIGNIFICANCE This study suggests that repeated sevoflurane exposure in newborn mice leads to depolarization of OPC, which leads to Ca2+ influx through NCX and affects OPC proliferation, migration, differentiation, and myelination, ultimately leading to cognitive impairment.
Collapse
|
109
|
Tabarestani TQ, Lewis NE, Kelly-Hedrick M, Zhang N, Cellini BR, Marrotte EJ, Williamson T, Wang H, Laskowitz DT, Faw TD, Abd-El-Barr MM. Surgical Considerations to Improve Recovery in Acute Spinal Cord Injury. Neurospine 2022; 19:689-702. [PMID: 36203295 PMCID: PMC9537855 DOI: 10.14245/ns.2244616.308] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/07/2022] [Indexed: 12/14/2022] Open
Abstract
Acute traumatic spinal cord injury (SCI) can be a devastating and costly event for individuals, their families, and the health system as a whole. Prognosis is heavily dependent on the physical extent of the injury and the severity of neurological dysfunction. If not treated urgently, individuals can suffer exacerbated secondary injury cascades that may increase tissue injury and limit recovery. Initial recognition and rapid treatment of acute SCI are vital to limiting secondary injury, reducing morbidity, and providing the best chance of functional recovery. This article aims to review the pathophysiology of SCI and the most up-to-date management of the acute traumatic SCI, specifically examining the modern approaches to surgical treatments along with the ethical limitations of research in this field.
Collapse
Affiliation(s)
| | - Nicholle E. Lewis
- Doctor of Physical Therapy Division, Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | | | - Nina Zhang
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC, USA
| | - Brianna R. Cellini
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC, USA
| | - Eric J. Marrotte
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Theresa Williamson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA,Center for Bioethics, Harvard Medical School, Boston, MA, USA
| | - Haichen Wang
- Department of Neurology, Duke University, Durham, NC, USA
| | | | - Timothy D. Faw
- Doctor of Physical Therapy Division, Department of Orthopaedic Surgery, Duke University, Durham, NC, USA,Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Muhammad M. Abd-El-Barr
- Department of Neurosurgery, Duke University, Durham, NC, USA,Corresponding Author Muhammad M. Abd-El-Barr Department of Neurosurgery, Duke University Medical Center 2840, Room 5335 5th Floor, Orange Zone, Duke South, Durham, NC 27710, USA
| |
Collapse
|
110
|
Guo W, Zhang X, Zhai J, Xue J. The roles and applications of neural stem cells in spinal cord injury repair. Front Bioeng Biotechnol 2022; 10:966866. [PMID: 36105599 PMCID: PMC9465243 DOI: 10.3389/fbioe.2022.966866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/28/2022] [Indexed: 12/05/2022] Open
Abstract
Spinal cord injury (SCI), which has no current cure, places a severe burden on patients. Stem cell-based therapies are considered promising in attempts to repair injured spinal cords; such options include neural stem cells (NSCs). NSCs are multipotent stem cells that differentiate into neuronal and neuroglial lineages. This feature makes NSCs suitable candidates for regenerating injured spinal cords. Many studies have revealed the therapeutic potential of NSCs. In this review, we discuss from an integrated view how NSCs can help SCI repair. We will discuss the sources and therapeutic potential of NSCs, as well as representative pre-clinical studies and clinical trials of NSC-based therapies for SCI repair.
Collapse
Affiliation(s)
- Wen Guo
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xindan Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Jiliang Zhai
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiliang Zhai, ; Jiajia Xue,
| | - Jiajia Xue
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
- *Correspondence: Jiliang Zhai, ; Jiajia Xue,
| |
Collapse
|
111
|
Zhang Z, Li X, Zhou H, Zhou J. NG2-glia crosstalk with microglia in health and disease. CNS Neurosci Ther 2022; 28:1663-1674. [PMID: 36000202 PMCID: PMC9532922 DOI: 10.1111/cns.13948] [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: 04/05/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/30/2022] Open
Abstract
Neurodegenerative diseases are increasingly becoming a global problem. However, the pathological mechanisms underlying neurodegenerative diseases are not fully understood. NG2‐glia abnormalities and microglia activation are involved in the development and/or progression of neurodegenerative disorders, such as multiple sclerosis, Alzheimer's disease, Parkinson's disease, and cerebrovascular diseases. In this review, we summarize the present understanding of the interaction between NG2‐glia and microglia in physiological and pathological states and discuss unsolved questions concerning their fate and potential fate. First, we introduce the NG2‐glia and microglia in health and disease. Second, we formulate the interaction between NG2‐glia and microglia. NG2‐glia proliferation, migration, differentiation, and apoptosis are influenced by factors released from the microglia. On the other hand, NG2‐glia also regulate microglia actions. We conclude that NG2‐glia and microglia are important immunomodulatory cells in the brain. Understanding the interaction between NG2‐glia and microglia will help provide a novel method to modulate myelination and treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Zuo Zhang
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Xiaolong Li
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Hongli Zhou
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jiyin Zhou
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
112
|
Osanai Y, Battulga B, Yamazaki R, Kouki T, Yatabe M, Mizukami H, Kobayashi K, Shinohara Y, Yoshimura Y, Ohno N. Dark Rearing in the Visual Critical Period Causes Structural Changes in Myelinated Axons in the Adult Mouse Visual Pathway. Neurochem Res 2022; 47:2815-2825. [PMID: 35933550 DOI: 10.1007/s11064-022-03689-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 06/14/2022] [Accepted: 07/12/2022] [Indexed: 12/20/2022]
Abstract
An appropriate sensory experience during the early developmental period is important for brain maturation. Dark rearing during the visual critical period delays the maturation of neuronal circuits in the visual cortex. Although the formation and structural plasticity of the myelin sheaths on retinal ganglion cell axons modulate the visual function, the effects of dark rearing during the visual critical period on the structure of the retinal ganglion cell axons and their myelin sheaths are still unclear. To address this question, mice were reared in a dark box during the visual critical period and then normally reared to adulthood. We found that myelin sheaths on the retinal ganglion cell axons of dark-reared mice were thicker than those of normally reared mice in both the optic chiasm and optic nerve. Furthermore, whole-mount immunostaining with fluorescent axonal labeling and tissue clearing revealed that the myelin internodal length in dark-reared mice was shorter than that in normally reared mice in both the optic chiasm and optic nerve. These findings demonstrate that dark rearing during the visual critical period affects the morphology of myelin sheaths, shortens and thickens myelin sheaths in the visual pathway, despite the mice being reared in normal light/dark conditions after the dark rearing.
Collapse
Affiliation(s)
- Yasuyuki Osanai
- Division of Histology and Cell Biology, Department of Anatomy, School of Medicine, Jichi Medical University, Shimotsuke, Japan. .,Australian Regenerative Medicine Institute, Monash University, 15 Innovation Walk, Clayton, VIC, 3800, Australia.
| | - Batpurev Battulga
- Division of Histology and Cell Biology, Department of Anatomy, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Reiji Yamazaki
- Division of Histology and Cell Biology, Department of Anatomy, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Tom Kouki
- Division of Histology and Cell Biology, Department of Anatomy, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Megumi Yatabe
- Division of Histology and Cell Biology, Department of Anatomy, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Japan.,SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan
| | - Yoshiaki Shinohara
- Division of Histology and Cell Biology, Department of Anatomy, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Yumiko Yoshimura
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan.,Division of Visual Information Processing, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Nobuhiko Ohno
- Division of Histology and Cell Biology, Department of Anatomy, School of Medicine, Jichi Medical University, Shimotsuke, Japan. .,Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan.
| |
Collapse
|
113
|
Wawrzyniak A, Balawender K, Lalak R, Golan MP, Wróbel K, Boroń D, Staszkiewicz R, Grabarek BO. Distribution and Morphological Characteristics of Oligodendrocytes in Selected Areas of the Brain of Male and Female Red Kangaroos (Macropus rufus). Brain Sci 2022; 12:brainsci12081035. [PMID: 36009098 PMCID: PMC9405871 DOI: 10.3390/brainsci12081035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/27/2022] [Accepted: 08/03/2022] [Indexed: 02/04/2023] Open
Abstract
This study was carried out on six adult red kangaroos of both sexes. To determine the location of the oligodendrocytes (OLGs) of the hippocampus (Hip) and corpus callosum (CC), the method of impregnation of the neuroglia with silver salts was applied. The iron distribution in the OLGs was determined by the histochemical method. The Nissl method was used to determine the location of the brain structure and to analyze the number of OLGs. In the Hip, these cells are located one beside another, mainly in blood vessels and neurons; in the neocortex (NC), they are located in layers I–VI; and in the CC, they are arranged in characteristic rows and accompany both nerve fibers and blood vessels. The analysis of the results obtained by the chosen methods in the Hip, NC, and CC in males and females did not show statistically significant differences in the distribution and location of the red kangaroo OLGs. The involvement of these cells is a physiological process that proceeds in a similar manner throughout the life of individuals and actively influences the metabolism of neurons and myelin.
Collapse
Affiliation(s)
- Agata Wawrzyniak
- Department of Morphological Sciences, College of Medical Sciences, Institute of Medical Sciences, University of Rzeszow, 35-315 Rzeszow, Poland
| | - Krzysztof Balawender
- Department of Morphological Sciences, College of Medical Sciences, Institute of Medical Sciences, University of Rzeszow, 35-315 Rzeszow, Poland
- Correspondence:
| | - Roman Lalak
- Department of Animal Anatomy and Histology, University of Life Sciences in Lublin, 20-400 Lublin, Poland
| | - Maciej Przemysław Golan
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine in Warsaw, 04-141 Warsaw, Poland
| | - Konrad Wróbel
- Department of Morphological Sciences, College of Medical Sciences, Institute of Medical Sciences, University of Rzeszow, 35-315 Rzeszow, Poland
| | - Dariusz Boroń
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine in Zabrze, Academy of Silesia in Katowice, 41-800 Zabrze, Poland
- Department of Gynaecology and Obstetrics, Faculty of Medicine in Zabrze, Academy of Silesia in Katowice, 41-800 Zabrze, Poland
| | - Rafał Staszkiewicz
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine in Zabrze, Academy of Silesia in Katowice, 41-800 Zabrze, Poland
- Department of Neurosurgery, 5th Military Clinical Hospital with the SP ZOZ Polyclinic in Krakow, 30-901 Krakow, Poland
| | - Beniamin Oskar Grabarek
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine in Zabrze, Academy of Silesia in Katowice, 41-800 Zabrze, Poland
- Department of Gynaecology and Obstetrics, Faculty of Medicine in Zabrze, Academy of Silesia in Katowice, 41-800 Zabrze, Poland
- GynCentrum, Laboratory of Molecular Biology and Virology, 40-851 Katowice, Poland
| |
Collapse
|
114
|
Dermitzakis I, Manthou ME, Meditskou S, Miliaras D, Kesidou E, Boziki M, Petratos S, Grigoriadis N, Theotokis P. Developmental Cues and Molecular Drivers in Myelinogenesis: Revisiting Early Life to Re-Evaluate the Integrity of CNS Myelin. Curr Issues Mol Biol 2022; 44:3208-3237. [PMID: 35877446 PMCID: PMC9324160 DOI: 10.3390/cimb44070222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023] Open
Abstract
The mammalian central nervous system (CNS) coordinates its communication through saltatory conduction, facilitated by myelin-forming oligodendrocytes (OLs). Despite the fact that neurogenesis from stem cell niches has caught the majority of attention in recent years, oligodendrogenesis and, more specifically, the molecular underpinnings behind OL-dependent myelinogenesis, remain largely unknown. In this comprehensive review, we determine the developmental cues and molecular drivers which regulate normal myelination both at the prenatal and postnatal periods. We have indexed the individual stages of myelinogenesis sequentially; from the initiation of oligodendrocyte precursor cells, including migration and proliferation, to first contact with the axon that enlists positive and negative regulators for myelination, until the ultimate maintenance of the axon ensheathment and myelin growth. Here, we highlight multiple developmental pathways that are key to successful myelin formation and define the molecular pathways that can potentially be targets for pharmacological interventions in a variety of neurological disorders that exhibit demyelination.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Dimosthenis Miliaras
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC 3004, Australia;
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
- Correspondence:
| |
Collapse
|
115
|
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
|
116
|
Protocadherin 15 suppresses oligodendrocyte progenitor cell proliferation and promotes motility through distinct signalling pathways. Commun Biol 2022; 5:511. [PMID: 35637313 PMCID: PMC9151716 DOI: 10.1038/s42003-022-03470-1] [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: 10/18/2021] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) express protocadherin 15 (Pcdh15), a member of the cadherin superfamily of transmembrane proteins. Little is known about the function of Pcdh15 in the central nervous system (CNS), however, Pcdh15 expression can predict glioma aggression and promote the separation of embryonic human OPCs immediately following a cell division. Herein, we show that Pcdh15 knockdown significantly increases extracellular signal-related kinase (ERK) phosphorylation and activation to enhance OPC proliferation in vitro. Furthermore, Pcdh15 knockdown elevates Cdc42-Arp2/3 signalling and impairs actin kinetics, reducing the frequency of lamellipodial extrusion and slowing filopodial withdrawal. Pcdh15 knockdown also reduces the number of processes supported by each OPC and new process generation. Our data indicate that Pcdh15 is a critical regulator of OPC proliferation and process motility, behaviours that characterise the function of these cells in the healthy CNS, and provide mechanistic insight into the role that Pcdh15 might play in glioma progression. Protocadherin 15 promotes lamellipodial and filopodial dynamics in oligodendrocyte progenitor cells by regulating Cdc42-Arp2/3 activity, but also suppresses ERK1/2 phosphorylation to reduce proliferation.
Collapse
|
117
|
Hilscher MM, Langseth CM, Kukanja P, Yokota C, Nilsson M, Castelo-Branco G. Spatial and temporal heterogeneity in the lineage progression of fine oligodendrocyte subtypes. BMC Biol 2022; 20:122. [PMID: 35610641 PMCID: PMC9131697 DOI: 10.1186/s12915-022-01325-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 05/09/2022] [Indexed: 11/11/2022] Open
Abstract
Background Oligodendrocytes are glial cells that support and insulate axons in the central nervous system through the production of myelin. Oligodendrocytes arise throughout embryonic and early postnatal development from oligodendrocyte precursor cells (OPCs), and recent work demonstrated that they are a transcriptional heterogeneous cell population, but the regional and functional implications of this heterogeneity are less clear. Here, we apply in situ sequencing (ISS) to simultaneously probe the expression of 124 marker genes of distinct oligodendrocyte populations, providing comprehensive maps of the corpus callosum, cingulate, motor, and somatosensory cortex in the brain, as well as gray matter (GM) and white matter (WM) regions in the spinal cord, at postnatal (P10), juvenile (P20), and young adult (P60) stages. We systematically compare the abundances of these populations and investigate the neighboring preference of distinct oligodendrocyte populations. Results We observed that oligodendrocyte lineage progression is more advanced in the juvenile spinal cord compared to the brain, corroborating with previous studies. We found myelination still ongoing in the adult corpus callosum while it was more advanced in the cortex. Interestingly, we also observed a lateral-to-medial gradient of oligodendrocyte lineage progression in the juvenile cortex, which could be linked to arealization, as well as a deep-to-superficial gradient with mature oligodendrocytes preferentially accumulating in the deeper layers of the cortex. The ISS experiments also exposed differences in abundances and population dynamics over time between GM and WM regions in the brain and spinal cord, indicating regional differences within GM and WM, and we found that neighboring preferences of some oligodendroglia populations are altered from the juvenile to the adult CNS. Conclusions Overall, our ISS experiments reveal spatial heterogeneity of oligodendrocyte lineage progression in the brain and spinal cord and uncover differences in the timing of oligodendrocyte differentiation and myelination, which could be relevant to further investigate functional heterogeneity of oligodendroglia, especially in the context of injury or disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01325-z.
Collapse
Affiliation(s)
- Markus M Hilscher
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65, Solna, Sweden.
| | | | - Petra Kukanja
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, 17177, Stockholm, Sweden
| | - Chika Yokota
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65, Solna, Sweden
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65, Solna, Sweden
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, 17177, Stockholm, Sweden.
| |
Collapse
|
118
|
Marguet F, Brosolo M, Friocourt G, Sauvestre F, Marcorelles P, Lesueur C, Marret S, Gonzalez BJ, Laquerrière A. Oligodendrocyte lineage is severely affected in human alcohol-exposed foetuses. Acta Neuropathol Commun 2022; 10:74. [PMID: 35568959 PMCID: PMC9107108 DOI: 10.1186/s40478-022-01378-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/01/2022] [Indexed: 12/12/2022] Open
Abstract
Prenatal alcohol exposure is a major cause of neurobehavioral disabilities. MRI studies in humans have shown that alcohol is associated with white matter microstructural anomalies but these studies focused on myelin abnormalities only after birth. Only one of these studies evaluated oligodendrocyte lineage, but only for a short period during human foetal life. As data are lacking in humans and alcohol is known to impair oligodendrocyte differentiation in rodents, the present study aimed to compare by immunohistochemistry the oligodendrocyte precursor cells expressing PDGFR-α and immature premyelinating/mature oligodendrocytes expressing Olig2 in the ganglionic eminences and the frontal cortex of 14 human foetuses exposed to alcohol from 15 to 37 weeks' gestation with age-matched controls. The human brains used in this study were obtained at the time of foetal autopsies for medical termination of pregnancy, in utero or post-natal early death. Before birth, PDGFR-α expression was strongly increased in the ganglionic eminences and the cortex of all foetuses exposed to alcohol except at the earliest stage. No massive generation of Olig2 immunoreactive cells was identified in the ganglionic eminences until the end of pregnancy and the density of Olig2-positive cells within the cortex was consistently lower in foetuses exposed to alcohol than in controls. These antenatal data from humans provides further evidence of major oligodendrocyte lineage impairment at specific and key stages of brain development upon prenatal alcohol exposure including defective or delayed generation and maturation of oligodendrocyte precursors.
Collapse
Affiliation(s)
- Florent Marguet
- Department of Pathology, Normandy Centre for Genomic and Personalized Medicine, Laboratoire d'Anatomie Pathologique, Pavillon Jacques Delarue, CHU, Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, 1 Rue de Germont, 76031, Rouen Cedex, France.
| | - Mélanie Brosolo
- UNIROUEN, INSERM U1245 F76000, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, Rouen, France
| | - Gaëlle Friocourt
- Inserm UMR1078, Université de Bretagne Occidentale, Faculté de Médecine et Des Sciences de la Santé; Etablissement Français du Sang (EFS) Bretagne; Laboratoire de Génétique Moléculaire, CHRU Brest, Hôpital Morvan, Brest, France
| | - Fanny Sauvestre
- Department of Pathology, Bordeaux University Hospital, Bordeaux, France
| | - Pascale Marcorelles
- Pathology Laboratory, Pole Pathologie-Biologie, Centre Hospitalier Universitaire Brest, Brest, France
- Laboratory of Neurosciences of Brest, Faculté de Médecine et des Sciences de la Santé, Brest University, Brest, France
| | - Céline Lesueur
- UNIROUEN, INSERM U1245 F76000, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, Rouen, France
| | - Stéphane Marret
- Department of Neonatal Paediatrics and Intensive Care, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, 76000, Rouen, France
| | - Bruno J Gonzalez
- UNIROUEN, INSERM U1245 F76000, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, Rouen, France
| | - Annie Laquerrière
- Department of Pathology, Normandy Centre for Genomic and Personalized Medicine, Laboratoire d'Anatomie Pathologique, Pavillon Jacques Delarue, CHU, Normandie Univ, UNIROUEN, INSERM U1245 and Rouen University Hospital, 1 Rue de Germont, 76031, Rouen Cedex, France
| |
Collapse
|
119
|
Renz-Polster H, Tremblay ME, Bienzle D, Fischer JE. The Pathobiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: The Case for Neuroglial Failure. Front Cell Neurosci 2022; 16:888232. [PMID: 35614970 PMCID: PMC9124899 DOI: 10.3389/fncel.2022.888232] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
Although myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) has a specific and distinctive profile of clinical features, the disease remains an enigma because causal explanation of the pathobiological matrix is lacking. Several potential disease mechanisms have been identified, including immune abnormalities, inflammatory activation, mitochondrial alterations, endothelial and muscular disturbances, cardiovascular anomalies, and dysfunction of the peripheral and central nervous systems. Yet, it remains unclear whether and how these pathways may be related and orchestrated. Here we explore the hypothesis that a common denominator of the pathobiological processes in ME/CFS may be central nervous system dysfunction due to impaired or pathologically reactive neuroglia (astrocytes, microglia and oligodendrocytes). We will test this hypothesis by reviewing, in reference to the current literature, the two most salient and widely accepted features of ME/CFS, and by investigating how these might be linked to dysfunctional neuroglia. From this review we conclude that the multifaceted pathobiology of ME/CFS may be attributable in a unifying manner to neuroglial dysfunction. Because the two key features - post exertional malaise and decreased cerebral blood flow - are also recognized in a subset of patients with post-acute sequelae COVID, we suggest that our findings may also be pertinent to this entity.
Collapse
Affiliation(s)
- Herbert Renz-Polster
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Quebec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Dorothee Bienzle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Joachim E. Fischer
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
120
|
Smith MD, Chamling X, Gill AJ, Martinez H, Li W, Fitzgerald KC, Sotirchos ES, Moroziewicz D, Bauer L, Paull D, Gharagozloo M, Bhargava P, Zack DJ, Fossati V, Calabresi PA. Reactive Astrocytes Derived From Human Induced Pluripotent Stem Cells Suppress Oligodendrocyte Precursor Cell Differentiation. Front Mol Neurosci 2022; 15:874299. [PMID: 35600072 PMCID: PMC9120968 DOI: 10.3389/fnmol.2022.874299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/28/2022] [Indexed: 01/01/2023] Open
Abstract
Astrocytes are instrumental in maintaining central nervous system (CNS) homeostasis and responding to injury. A major limitation of studying neurodegenerative diseases like multiple sclerosis (MS) is lack of human pathological specimens obtained during the acute stages, thereby relegating research to post-mortem specimens obtained years after the initiation of pathology. Rodent reactive astrocytes have been shown to be cytotoxic to neurons and oligodendrocytes but may differ from human cells, especially in diseases with genetic susceptibility. Herein, we purified human CD49f+ astrocytes from induced pluripotent stem cells derived from individual patient and control peripheral leukocytes. We compared TNF and IL1α stimulated human reactive astrocytes from seven persons with MS and six non-MS controls and show their transcriptomes are remarkably similar to those described in rodents. The functional effect of astrocyte conditioned media (ACM) was examined in a human oligodendrocyte precursor cell (OPC) line differentiation assay. ACM was not cytotoxic to the OPCs but robustly inhibited the myelin basic protein (MBP) reporter. No differences were seen between MS and control stimulated astrocytes at either the transcript level or in ACM mediated OPC suppression assays. We next used RNAseq to interrogate differentially expressed genes in the OPC lines that had suppressed differentiation from the human ACM. Remarkably, not only was OPC differentiation and myelin gene expression suppressed, but we observed induction of several immune pathways in OPCs exposed to the ACM. These data support the notion that reactive astrocytes can inhibit OPC differentiation thereby limiting their remyelination capacity, and that OPCs take on an immune profile in the context of inflammatory cues.
Collapse
Affiliation(s)
- Matthew D. Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xitiz Chamling
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alexander J. Gill
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hector Martinez
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Weifeng Li
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kathryn C. Fitzgerald
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elias S. Sotirchos
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dorota Moroziewicz
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Lauren Bauer
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Marjan Gharagozloo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Donald J. Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
121
|
Poggi G, Albiez J, Pryce CR. Effects of chronic social stress on oligodendrocyte proliferation-maturation and myelin status in prefrontal cortex and amygdala in adult mice. Neurobiol Stress 2022; 18:100451. [PMID: 35685682 PMCID: PMC9170777 DOI: 10.1016/j.ynstr.2022.100451] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 10/26/2022] Open
Abstract
Stress-related neuropsychiatric disorders present with excessive processing of aversive stimuli. Whilst underlying pathophysiology remains poorly understood, within- and between-regional changes in oligodendrocyte (OL)-myelination status in anterior cingulate cortex and amygdala (ACC-AMY network) could be important. In adult mice, a 15-day chronic social stress (CSS) protocol leads to increased aversion responsiveness, accompanied by increased resting-state functional connectivity between, and reduced oligodendrocyte- and myelin-related transcript expression within, medial prefrontal cortex and amygdala (mPFC-AMY network), the analog of the human ACC-AMY network. In the current study, young-adult male C57BL/6 mice underwent CSS or control handling (CON). To assess OL proliferation-maturation, mice received 5-ethynyl-2'-deoxyuridine via drinking water across CSS/CON and brains were collected on day 16 or 31. In mPFC, CSS decreased the density of proliferative OL precursor cells (OPCs) at days 16 and 31. CSS increased mPFC myelin basic protein (MBP) integrated density at day 31, as well as increasing myelin thickness as determined using transmission electron microscopy, at day 16. In AMY, CSS increased the densities of total CC1+ OLs (day 31) and CC1+/ASPA+ OLs (days 16 and 31), whilst decreasing the density of proliferative OPCs at days 16 and 31. CSS was without effect on AMY MBP content and myelin thickness, at days 16 and 31. Therefore, CSS impacts on the OL lineage in mPFC and AMY and to an extent that, in mPFC at least, leads to increased myelination. This increased myelination could contribute to the excessive aversion learning and memory that occur in CSS mice and, indeed, human stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Giulia Poggi
- Preclinical Laboratory for Translational Research Into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland
| | - Jamie Albiez
- Preclinical Laboratory for Translational Research Into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland
| | - Christopher R. Pryce
- Preclinical Laboratory for Translational Research Into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Switzerland
| |
Collapse
|
122
|
DeFlitch L, Gonzalez-Fernandez E, Crawley I, Kang SH. Age and Alzheimer's Disease-Related Oligodendrocyte Changes in Hippocampal Subregions. Front Cell Neurosci 2022; 16:847097. [PMID: 35465615 PMCID: PMC9023310 DOI: 10.3389/fncel.2022.847097] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Oligodendrocytes (OLs) form myelin sheaths and provide metabolic support to axons in the CNS. Although most OLs develop during early postnatal life, OL generation continues in adulthood, and this late oligodendrogenesis may contribute to neuronal network plasticity in the adult brain. We used genetic tools for OL labeling and fate tracing of OL progenitors (OPCs), thereby determining OL population growth in hippocampal subregions with normal aging. OL numbers increased up to at least 1 year of age, but the rates and degrees of this OL change differed among hippocampal subregions. In particular, adult oligodendrogenesis was most prominent in the CA3 and CA4 subregions. In Alzheimer's disease-like conditions, OL loss was also most severe in the CA3 and CA4 of APP/PS1 mice, although the disease did not impair the rate of OPC differentiation into OLs in those regions. Such region-specific, dynamic OL changes were not correlated with those of OPCs or astrocytes, or the regional distribution of Aβ deposits. Our findings suggest subregion-dependent mechanisms for myelin plasticity and disease-associated OL vulnerability in the adult hippocampus.
Collapse
Affiliation(s)
- Leah DeFlitch
- Biology Department, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Estibaliz Gonzalez-Fernandez
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States,Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Temple University, Philadelphia, PA, United States
| | - Ilan Crawley
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States,Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Temple University, Philadelphia, PA, United States
| | - Shin H. Kang
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States,Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Temple University, Philadelphia, PA, United States,*Correspondence: Shin H. Kang,
| |
Collapse
|
123
|
Scalabrino G. Newly Identified Deficiencies in the Multiple Sclerosis Central Nervous System and Their Impact on the Remyelination Failure. Biomedicines 2022; 10:biomedicines10040815. [PMID: 35453565 PMCID: PMC9026986 DOI: 10.3390/biomedicines10040815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of multiple sclerosis (MS) remains enigmatic and controversial. Myelin sheaths in the central nervous system (CNS) insulate axons and allow saltatory nerve conduction. MS brings about the destruction of myelin sheaths and the myelin-producing oligodendrocytes (ODCs). The conundrum of remyelination failure is, therefore, crucial in MS. In this review, the roles of epidermal growth factor (EGF), normal prions, and cobalamin in CNS myelinogenesis are briefly summarized. Thereafter, some findings of other authors and ourselves on MS and MS-like models are recapitulated, because they have shown that: (a) EGF is significantly decreased in the CNS of living or deceased MS patients; (b) its repeated administration to mice in various MS-models prevents demyelination and inflammatory reaction; (c) as was the case for EGF, normal prion levels are decreased in the MS CNS, with a strong correspondence between liquid and tissue levels; and (d) MS cobalamin levels are increased in the cerebrospinal fluid, but decreased in the spinal cord. In fact, no remyelination can occur in MS if these molecules (essential for any form of CNS myelination) are lacking. Lastly, other non-immunological MS abnormalities are reviewed. Together, these results have led to a critical reassessment of MS pathogenesis, partly because EGF has little or no role in immunology.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| |
Collapse
|
124
|
Sánchez-González R, López-Mascaraque L. Lineage Relationships Between Subpallial Progenitors and Glial Cells in the Piriform Cortex. Front Neurosci 2022; 16:825969. [PMID: 35386594 PMCID: PMC8979001 DOI: 10.3389/fnins.2022.825969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/21/2022] [Indexed: 11/22/2022] Open
Abstract
The piriform cortex is a paleocortical area, located in the ventrolateral surface of the rodent forebrain, receiving direct input from the olfactory bulb. The three layers of the PC are defined by the diversity of glial and neuronal cells, marker expression, connections, and functions. However, the glial layering, ontogeny, and sibling cell relationship along the PC is an unresolved question in the field. Here, using multi-color genetic lineage tracing approaches with different StarTrack strategies, we performed a rigorous analysis of the derived cell progenies from progenitors located at the subpallium ventricular surface. First, we specifically targeted E12-progenitors with UbC-StarTrack to analyze their adult derived-cell progeny and their location within the piriform cortex layers. The vast majority of the cell progeny derived from targeted progenitors were identified as neurons, but also astrocytes and NG2 cells. Further, to specifically target single Gsx-2 subpallial progenitors and their derived cell-progeny in the piriform cortex, we used the UbC-(Gsx-2-hyPB)-StarTrack to perform an accurate analysis of their clonal relationships. Our results quantitatively delineate the adult clonal cell pattern from single subpallial E12-progenitors, focusing on glial cells. In summary, there is a temporal pattern in the assembly of the glial cell diversity in the piriform cortex, which also reveals spatio-temporal progenitor heterogeneity.
Collapse
|
125
|
Meschkat M, Steyer AM, Weil MT, Kusch K, Jahn O, Piepkorn L, Agüi-Gonzalez P, Phan NTN, Ruhwedel T, Sadowski B, Rizzoli SO, Werner HB, Ehrenreich H, Nave KA, Möbius W. White matter integrity in mice requires continuous myelin synthesis at the inner tongue. Nat Commun 2022; 13:1163. [PMID: 35246535 PMCID: PMC8897471 DOI: 10.1038/s41467-022-28720-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 01/24/2022] [Indexed: 12/18/2022] Open
Abstract
Myelin, the electrically insulating sheath on axons, undergoes dynamic changes over time. However, it is composed of proteins with long lifetimes. This raises the question how such a stable structure is renewed. Here, we study the integrity of myelinated tracts after experimentally preventing the formation of new myelin in the CNS of adult mice, using an inducible Mbp null allele. Oligodendrocytes survive recombination, continue to express myelin genes, but they fail to maintain compacted myelin sheaths. Using 3D electron microscopy and mass spectrometry imaging we visualize myelin-like membranes failing to incorporate adaxonally, most prominently at juxta-paranodes. Myelinoid body formation indicates degradation of existing myelin at the abaxonal side and the inner tongue of the sheath. Thinning of compact myelin and shortening of internodes result in the loss of about 50% of myelin and axonal pathology within 20 weeks post recombination. In summary, our data suggest that functional axon-myelin units require the continuous incorporation of new myelin membranes. Myelin is formed of proteins of long half-lives. The mechanisms of renewal of such a stable structure are unclear. Here, the authors show that myelin integrity requires continuous myelin synthesis at the inner tongue, contributing to the maintenance of a functional axon-myelin unit.
Collapse
Affiliation(s)
- Martin Meschkat
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Göttingen Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences (GGNB), Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Abberior Instruments GmbH, Göttingen, Germany
| | - Anna M Steyer
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Imaging Centre, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Marie-Theres Weil
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Olaf Jahn
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Lars Piepkorn
- Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Paola Agüi-Gonzalez
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| | - Nhu Thi Ngoc Phan
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany.,Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Boguslawa Sadowski
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Silvio O Rizzoli
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Hannelore Ehrenreich
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany. .,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany. .,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany. .,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
126
|
Furber KL, Lacombe RJS, Caine S, Thangaraj MP, Read S, Rosendahl SM, Bazinet RP, Popescu BF, Nazarali AJ. Biochemical Alterations in White Matter Tracts of the Aging Mouse Brain Revealed by FTIR Spectroscopy Imaging. Neurochem Res 2022; 47:795-810. [PMID: 34820737 DOI: 10.1007/s11064-021-03491-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 05/31/2021] [Accepted: 11/17/2021] [Indexed: 11/25/2022]
Abstract
White matter degeneration in the central nervous system (CNS) has been correlated with a decline in cognitive function during aging. Ultrastructural examination of the aging human brain shows a loss of myelin, yet little is known about molecular and biochemical changes that lead to myelin degeneration. In this study, we investigate myelination across the lifespan in C57BL/6 mice using electron microscopy and Fourier transform infrared (FTIR) spectroscopic imaging to better understand the relationship between structural and biochemical changes in CNS white matter tracts. A decrease in the number of myelinated axons was associated with altered lipid profiles in the corpus callosum of aged mice. FTIR spectroscopic imaging revealed alterations in functional groups associated with phospholipids, including the lipid acyl, lipid ester and phosphate vibrations. Biochemical changes in white matter were observed prior to structural changes and most predominant in the anterior regions of the corpus callosum. This was supported by biochemical analysis of fatty acid composition that demonstrated an overall trend towards increased monounsaturated fatty acids and decreased polyunsaturated fatty acids with age. To further explore the molecular mechanisms underlying these biochemical alterations, gene expression profiles of lipid metabolism and oxidative stress pathways were investigated. A decrease in the expression of several genes involved in glutathione metabolism suggests that oxidative damage to lipids may contribute to age-related white matter degeneration.
Collapse
Affiliation(s)
- Kendra L Furber
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada.
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
- Division of Medical Sciences, University of Northern British Columbia, Prince George, BC, Canada.
| | - R J Scott Lacombe
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sally Caine
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Merlin P Thangaraj
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Stuart Read
- Canadian Light Source, Saskatoon, SK, Canada
| | | | - Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Bogdan F Popescu
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Adil J Nazarali
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
127
|
Li C, Huang S, Zhou W, Xie Z, Xie S, Li M. Effects of the Notch Signaling Pathway on Secondary Brain Changes Caused by Spinal Cord Injury in Mice. Neurochem Res 2022; 47:1651-1663. [PMID: 35211828 DOI: 10.1007/s11064-022-03558-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/22/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) can cause secondary brain changes, leading to hypomyelination in the dorsolateral prefrontal cortex (dlPFC). Some studies have shown that notch signaling pathway activation can regulate oligodendrocyte maturation and myelination. The aim of this study was to investigate whether inhibition of the Notch signaling pathway can alleviate hypomyelination in the dlPFC caused by SCI. Moreover, we further investigated whether the changes in myelination in the dlPFC are associated with neuropathic pain following SCI. We established a mouse model of SCI and observed the changes in mechanical and thermal hyperalgesia. Western blotting and immunofluorescence were used to analyze the changes in myelination in the dlPFC. The results indicated the existence of a relationship between activation of the Notch signaling pathway and hypomyelination in the dlPFC and confirmed the existence of a relationship between hypomyelination in the dlPFC and decreases in mechanical and thermal hyperalgesia thresholds. In conclusion, these results suggested that the Notch signaling pathway is activated after SCI, leading to hypomyelination in the dlPFC, and that DAPT can inhibit the Notch signaling pathway and improve mechanical and thermal hyperalgesia thresholds. Our findings provide a new target for the treatment of neuropathic pain caused by SCI.
Collapse
Affiliation(s)
- Chengcai Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, NO17 Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Shaoxin Huang
- School of Basic Medicine, Jiujiang University, Jiujiang, 332005, Jiangxi, People's Republic of China
| | - Wu Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, NO17 Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Zhiping Xie
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, NO17 Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Shenke Xie
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, NO17 Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, NO17 Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
128
|
Ozgür-Günes Y, Chedik M, LE Stunff C, Fovet CM, Bougneres P. Long-term disease prevention with a gene therapy targeting oligodendrocytes in a mouse model of adrenomyeloneuropathy. Hum Gene Ther 2022; 33:936-949. [PMID: 35166123 DOI: 10.1089/hum.2021.293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Adrenomyeloneuropathy (AMN) is a late-onset axonopathy of spinal cord tracts caused by mutations of the ABCD1 gene that encodes ALDP, a peroxisomal transporter of very long chain fatty acids (VLCFA). Disturbed metabolic interaction between oligodendrocytes (OL) and axons is suspected to play a major role in AMN axonopathy. To develop a vector targeting OL, the human ABCD1 gene driven by a short 0.3 kb part of the human myelin-associated glycoprotein (MAG) promoter was packaged into an adeno-associated viral serotype 9 (rAAV9). An intravenous injection of this vector at postnatal day 10 (P10) in Abcd1-/- mice, a model of AMN, allowed a near normal motor performance to persist for 24 months, while age-matched untreated mice developed major defects of balance and motricity. Three weeks post vector, 50-54% of spinal cord white matter OL were expressing ALDP at the cervical level, and only 6-7% after 24 months. In addition, 29-32% of cervical spinal cord astrocytes at 3 weeks and 16-19% at 24 months also expressed ALDP. C26:0-lysoPC, a sensitive VLCFA marker of AMN, was lower by 41% and 50%, respectively in the spinal cord and brain of vector-treated compared with untreated mice. In a non-human primate (NHP), the intrathecal injection of the rAAV9-MAG vector induced abundant ALDP expression at 3 weeks in spinal cord OL (43%, 29%, 26% at cervical, thoracic and lumbar levels) and cerebellum OL (35%). In addition, 33-41 % of spinal cord astrocytes expressed hALDP, and 27% of cerebellar astrocytes. To our knowledge, OL targeting had not been obtained before in primates with other vectors or promoters. The current results thus provide a robust proof-of-concept not only for the gene therapy of AMN but for other CNS diseases where the targeting of OL with the rAAV9-MAG vector may be of interest.
Collapse
Affiliation(s)
| | - Malha Chedik
- INSERM, 27102, Le Kremlin-Bicêtre, Île-de-France, France;
| | | | | | - Pierre Bougneres
- INSERM, 27102, 80 rue du Général Leclercc, Le Kremlin Bicêtre, France, 94276;
| |
Collapse
|
129
|
Abstract
Nervous system activity regulates development, homeostasis, and plasticity of the brain as well as other organs in the body. These mechanisms are subverted in cancer to propel malignant growth. In turn, cancers modulate neural structure and function to augment growth-promoting neural signaling in the tumor microenvironment. Approaching cancer biology from a neuroscience perspective will elucidate new therapeutic strategies for presently lethal forms of cancer. In this review, we highlight the neural signaling mechanisms recapitulated in primary brain tumors, brain metastases, and solid tumors throughout the body that regulate cancer progression. Expected final online publication date for the Annual Review of Neuroscience, Volume 45 is July 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Michael B Keough
- Department of Neurology and Neurological Sciences and Howard Hughes Medical Institute, Stanford University, Stanford, California, USA;
| | - Michelle Monje
- Department of Neurology and Neurological Sciences and Howard Hughes Medical Institute, Stanford University, Stanford, California, USA;
| |
Collapse
|
130
|
Eugenin von Bernhardi J, Dimou L. Oligodendrogenesis is a key process for cognitive performance improvement induced by voluntary physical activity. Glia 2022; 70:1052-1067. [PMID: 35104015 DOI: 10.1002/glia.24155] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/13/2021] [Accepted: 01/21/2022] [Indexed: 12/13/2022]
Abstract
Physical activity (PA) promotes the proliferation of neural stem cells and enhances neurogenesis in the dentate gyrus resulting in hippocampal circuit remodeling and cognitive enhancement. Nonetheless, knowledge of other neural progenitors affected by PA and the mechanisms through which they could contribute to circuit plasticity and cognitive enhancement are still poorly understood. In this work we demonstrated that NG2-glia, also known as oligodendrocyte progenitor cells, show enhanced proliferation and differentiation in response to voluntary PA in a brain region-dependent manner in adult mice. Surprisingly, preventing NG2-glia differentiation during enhanced PA abolishes the exercise-associated cognitive improvement without affecting neurogenesis or baseline learning capacity. Thus, here we provided new evidence highlighting the requirement of oligodendrogenesis for exercise induced-cognition enhancement.
Collapse
Affiliation(s)
- Jaime Eugenin von Bernhardi
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, Ulm, Germany.,Graduate School for Systemic Neuroscience, Ludwig-Maximilians University, Planegg-Martinsried, Munich, Germany
| | - Leda Dimou
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, Ulm, Germany.,Graduate School for Systemic Neuroscience, Ludwig-Maximilians University, Planegg-Martinsried, Munich, Germany
| |
Collapse
|
131
|
Chen JF, Wang F, Huang NX, Xiao L, Mei F. Oligodendrocytes and Myelin: Active players in Neurodegenerative brains? Dev Neurobiol 2022; 82:160-174. [PMID: 35081276 DOI: 10.1002/dneu.22867] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/10/2022]
Abstract
Oligodendrocytes (OLs) are a major type of glial cells in the central nervous system that generate multiple myelin sheaths to wrap axons. Myelin ensures fast and efficient propagation of action potentials along axons and supports neurons with nourishment. The decay of OLs and myelin has been implicated in age-related neurodegenerative diseases and these changes are generally considered as an inevitable result of neuron loss and axon degeneration. Noticeably, OLs and myelin undergo dynamic changes in healthy adult brains, that is, newly formed OLs are continuously added throughout life from the differentiation of oligodendrocyte precursor cells (OPCs) and the pre-existing myelin sheaths may undergo degeneration or remodeling. Increasing evidence has shown that changes in OLs and myelin are present in the early stages of neurodegenerative diseases, and even prior to significant neuronal loss and functional deficits. More importantly, oligodendroglia-specific manipulation, by either deletion of the disease gene or enhancement of myelin renewal, can alleviate functional impairments in neurodegenerative animal models. These findings underscore the possibility that OLs and myelin are not passively but actively involved in neurodegenerative diseases and may play an important role in modulating neuronal function and survival. In this review, we summarize recent work characterizing OL and myelin changes in both healthy and neurodegenerative brains and discuss the potential of targeting oligodendroglial cells in treating neurodegenerative diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jing-Fei Chen
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Fei Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Nan-Xing Huang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Lan Xiao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Feng Mei
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
132
|
Zhao Y, Zhu W, Chen H, Yan K, Wu J, Huang Q. Glioma stem cells and their microenvironment: A narrative review on docking and transformation. GLIOMA 2022. [DOI: 10.4103/glioma.glioma_5_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
133
|
Correale J, Ysrraelit MC. Multiple Sclerosis and Aging: The Dynamics of Demyelination and Remyelination. ASN Neuro 2022; 14:17590914221118502. [PMID: 35938615 PMCID: PMC9364177 DOI: 10.1177/17590914221118502] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system
(CNS) leading to demyelination and neurodegeneration. Life expectancy and age of onset in
MS patients have been rising over the last decades, and previous studies have shown that
age affects disease progression. Therefore, age appears as one of the most important
factors in accumulating disability in MS patients. Indeed, the degeneration of
oligodendrocytes (OGDs) and OGD precursors (OPCs) increases with age, in association with
increased inflammatory activity of astrocytes and microglia. Similarly, age-related
neuronal changes such as mitochondrial alterations, an increase in oxidative stress, and
disrupted paranodal junctions can impact myelin integrity. Conversely, once myelination is
complete, the long-term integrity of axons depends on OGD supply of energy. These
alterations determine pathological myelin changes consisting of myelin outfolding,
splitting, and accumulation of multilamellar fragments. Overall, these data demonstrate
that old mature OGDs lose their ability to produce and maintain healthy myelin over time,
to induce de novo myelination, and to remodel pre-existing myelinated
axons that contribute to neural plasticity in the CNS. Furthermore, as observed in other
tissues, aging induces a general decline in regenerative processes and, not surprisingly,
progressively hinders remyelination in MS. In this context, this review will provide an
overview of the current knowledge of age-related changes occurring in cells of the
oligodendroglial lineage and how they impact myelin synthesis, axonal degeneration, and
remyelination efficiency.
Collapse
Affiliation(s)
- Jorge Correale
- Departamento de Neurología, 58782Fleni, Buenos Aires, Argentina
| | | |
Collapse
|
134
|
Abstract
The pathology of fetal alcohol syndrome and the less severe fetal alcohol spectrum disorders includes brain dysmyelination. Recent studies have shed light on the molecular mechanisms underlying these white matter abnormalities. Rodent models of fetal alcohol syndrome and human studies have shown suppressed oligodendrocyte differentiation and apoptosis of oligodendrocyte precursor cells. Ethanol exposure led to reduced expression of myelin basic protein and delayed myelin basic protein expression in rat and mouse models of fetal alcohol syndrome and in human histopathological specimens. Several studies have reported increased expression of many chemokines in dysmyelinating disorders in central nervous system, including multiple sclerosis and fetal alcohol syndrome. Acute ethanol exposure reduced levels of the neuroprotective insulin-like growth factor-1 in fetal and maternal sheep and in human fetal brain tissues, while ethanol increased the expression of tumor necrosis factor α in mouse and human neurons. White matter lesions have been induced in the developing sheep brain by alcohol exposure in early gestation. Rat fetal alcohol syndrome models have shown reduced axon diameters, with thinner myelin sheaths, as well as reduced numbers of oligodendrocytes, which were also morphologically aberrant oligodendrocytes. Expressions of markers for mature myelination, including myelin basic protein, also were reduced. The accumulating knowledge concerning the mechanisms of ethanol-induced dysmyelination could lead to the development of strategies to prevent dysmyelination in children exposed to ethanol during fetal development. Future studies using fetal oligodendrocyte- and oligodendrocyte precursor cell-derived exosomes isolated from the mother’s blood may identify biomarkers for fetal alcohol syndrome and even implicate epigenetic changes in early development that affect oligodendrocyte precursor cell and oligodendrocyte function in adulthood. By combining various imaging modalities with molecular studies, it may be possible to determine which fetuses are at risk and to intervene therapeutically early in the pregnancy.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Michael E Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
135
|
Kimura Y, Hayashi Y, Hitomi S, Ikutame D, Urata K, Shibuta I, Sakai A, Ni J, Iwata K, Tonogi M, Shinoda M. IL-33 induces orofacial neuropathic pain through Fyn-dependent phosphorylation of GluN2B in the trigeminal spinal subnucleus caudalis. Brain Behav Immun 2022; 99:266-280. [PMID: 34715301 DOI: 10.1016/j.bbi.2021.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Orofacial neuropathic pain can cause considerable disruptions in patients' daily lives, especially because of a lack of effective medications as its underlying causative mechanisms are not fully understood. Here, we found neuron-specific expression of the interleukin (IL)-33 receptor in the trigeminal spinal subnucleus caudalis (Vc), distinct from the spinal dorsal horn. Reduction in head withdrawal threshold in response to von Frey filament stimulation of the whisker pad skin was inversely correlated with the upregulation of IL-33 in the Vc after infraorbital nerve injury (IONI). Neutralization of IL-33 in the Vc alleviated mechanical allodynia in the whisker pad skin after IONI; conversely, intracisternal administration of IL-33 elicited mechanical allodynia in the whisker pad skin, which was relieved by GluN2B antagonism. Moreover, IL-33 triggered the potentiation of GluN2B-containing N-methyl-D-aspartate receptor-mediated synaptic currents and phosphorylation of synaptosomal GluN2B in the Vc, whereas IONI-induced GluN2B phosphorylation was inhibited by neutralization of IL-33 in the Vc. IL-33-induced GluN2B phosphorylation was mediated by phosphorylation of Fyn kinase, and inhibition of the Fyn kinase pathway prevented the development of IL-33-induced mechanical allodynia. Our findings provide insights into a new mechanism by which IL-33 directly regulates synaptic transmission and suggest that IL-33 signaling could be a candidate target for therapeutic interventions for orofacial neuropathic pain.
Collapse
Affiliation(s)
- Yuki Kimura
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan.
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Daisuke Ikutame
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Tokyo, Japan
| | - Kentaro Urata
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Tokyo, Japan
| | - Ikuko Shibuta
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Atsushi Sakai
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Morio Tonogi
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry, Tokyo, Japan
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
136
|
Bloom MS, Orthmann-Murphy J, Grinspan JB. Motor Learning and Physical Exercise in Adaptive Myelination and Remyelination. ASN Neuro 2022; 14:17590914221097510. [PMID: 35635130 PMCID: PMC9158406 DOI: 10.1177/17590914221097510] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
The idea that myelination is driven by both intrinsic and extrinsic cues has gained much traction in recent years. Studies have demonstrated that myelination occurs in an intrinsic manner during early development and continues through adulthood in an activity-dependent manner called adaptive myelination. Motor learning, the gradual acquisition of a specific novel motor skill, promotes adaptive myelination in both the healthy and demyelinated central nervous system (CNS). On the other hand, exercise, a physical activity that involves planned, structured and repetitive bodily movements that expend energy and benefits one's fitness, promotes remyelination in pathology, but it is less clear whether it promotes adaptive myelination in healthy subjects. Studies on these topics have also investigated whether the timing of motor learning or physical exercise is important for successful addition of myelin. Here we review our current understanding of the relationship of motor skill learning and physical exercise on myelination.
Collapse
Affiliation(s)
- Mara S. Bloom
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jennifer Orthmann-Murphy
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Judith B. Grinspan
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
137
|
Rivera AD, Pieropan F, Williams G, Calzolari F, Butt AM, Azim K. Drug connectivity mapping and functional analysis reveal therapeutic small molecules that differentially modulate myelination. Biomed Pharmacother 2022; 145:112436. [PMID: 34813998 PMCID: PMC8664715 DOI: 10.1016/j.biopha.2021.112436] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/29/2021] [Accepted: 11/12/2021] [Indexed: 12/30/2022] Open
Abstract
Disruption or loss of oligodendrocytes (OLs) and myelin has devastating effects on CNS function and integrity, which occur in diverse neurological disorders, including Multiple Sclerosis (MS), Alzheimer's disease and neuropsychiatric disorders. Hence, there is a need to develop new therapies that promote oligodendrocyte regeneration and myelin repair. A promising approach is drug repurposing, but most agents have potentially contrasting biological actions depending on the cellular context and their dose-dependent effects on intracellular pathways. Here, we have used a combined systems biology and neurobiological approach to identify compounds that exert positive and negative effects on oligodendroglia, depending on concentration. Notably, next generation pharmacogenomic analysis identified the PI3K/Akt modulator LY294002 as the most highly ranked small molecule with both pro- and anti-oligodendroglial concentration-dependent effects. We validated these in silico findings using multidisciplinary approaches to reveal a profoundly bipartite effect of LY294002 on the generation of OPCs and their differentiation into myelinating oligodendrocytes in both postnatal and adult contexts. Finally, we employed transcriptional profiling and signalling pathway activity assays to determine cell-specific mechanisms of action of LY294002 on oligodendrocytes and resolve optimal in vivo conditions required to promote myelin repair. These results demonstrate the power of multidisciplinary strategies in determining the therapeutic potential of small molecules in neurodegenerative disorders.
Collapse
Affiliation(s)
- A D Rivera
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, St Michael's Building, White Swan Road, PO1 2DT Portsmouth, UK; Section of Human Anatomy, Department of Neuroscience, University of Padua, Padua, Italy.
| | - F Pieropan
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, St Michael's Building, White Swan Road, PO1 2DT Portsmouth, UK
| | - G Williams
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - F Calzolari
- Research Group Adult Neurogenesis & Cellular Reprogramming Institute of Physiological Chemistry, University Medical Center, Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 19, 55128 Mainz, Germany
| | - A M Butt
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, St Michael's Building, White Swan Road, PO1 2DT Portsmouth, UK
| | - K Azim
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
138
|
Lespay-Rebolledo C, Tapia-Bustos A, Perez-Lobos R, Vio V, Casanova-Ortiz E, Farfan-Troncoso N, Zamorano-Cataldo M, Redel-Villarroel M, Ezquer F, Quintanilla ME, Israel Y, Morales P, Herrera-Marschitz M. Sustained Energy Deficit Following Perinatal Asphyxia: A Shift towards the Fructose-2,6-bisphosphatase (TIGAR)-Dependent Pentose Phosphate Pathway and Postnatal Development. Antioxidants (Basel) 2021; 11:74. [PMID: 35052577 PMCID: PMC8773255 DOI: 10.3390/antiox11010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Labor and delivery entail a complex and sequential metabolic and physiologic cascade, culminating in most circumstances in successful childbirth, although delivery can be a risky episode if oxygen supply is interrupted, resulting in perinatal asphyxia (PA). PA causes an energy failure, leading to cell dysfunction and death if re-oxygenation is not promptly restored. PA is associated with long-term effects, challenging the ability of the brain to cope with stressors occurring along with life. We review here relevant targets responsible for metabolic cascades linked to neurodevelopmental impairments, that we have identified with a model of global PA in rats. Severe PA induces a sustained effect on redox homeostasis, increasing oxidative stress, decreasing metabolic and tissue antioxidant capacity in vulnerable brain regions, which remains weeks after the insult. Catalase activity is decreased in mesencephalon and hippocampus from PA-exposed (AS), compared to control neonates (CS), in parallel with increased cleaved caspase-3 levels, associated with decreased glutathione reductase and glutathione peroxidase activity, a shift towards the TIGAR-dependent pentose phosphate pathway, and delayed calpain-dependent cell death. The brain damage continues long after the re-oxygenation period, extending for weeks after PA, affecting neurons and glial cells, including myelination in grey and white matter. The resulting vulnerability was investigated with organotypic cultures built from AS and CS rat newborns, showing that substantia nigra TH-dopamine-positive cells from AS were more vulnerable to 1 mM of H2O2 than those from CS animals. Several therapeutic strategies are discussed, including hypothermia; N-acetylcysteine; memantine; nicotinamide, and intranasally administered mesenchymal stem cell secretomes, promising clinical translation.
Collapse
Affiliation(s)
- Carolyne Lespay-Rebolledo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Andrea Tapia-Bustos
- School of Pharmacy, Faculty of Medicine, Universidad Andres Bello, Santiago 8370149, Chile;
| | - Ronald Perez-Lobos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Valentina Vio
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Emmanuel Casanova-Ortiz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Nancy Farfan-Troncoso
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Marta Zamorano-Cataldo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Martina Redel-Villarroel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Maria Elena Quintanilla
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Yedy Israel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| |
Collapse
|
139
|
Luan W, Qi X, Liang F, Zhang X, Jin Z, Shi L, Luo B, Dai X. Microglia Impede Oligodendrocyte Generation in Aged Brain. J Inflamm Res 2021; 14:6813-6831. [PMID: 34924766 PMCID: PMC8674668 DOI: 10.2147/jir.s338242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/18/2021] [Indexed: 01/13/2023] Open
Abstract
Purpose Age-related increase in myelin loss may be responsible for brain atrophy, and the mechanism is not completely understood. We aim to comprehensively delineate oligodendrocyte heterogeneity in young and aged mice and to reveal the underlying mechanism for myelin loss during aging. Methods Diffusion tensor imaging and immunofluorescent staining were performed to verify the demyelination in the aged brains of both rodents and human. Further, the single-cell RNA sequencing data of all brain cells from young and aged mice were deeply analyzed to identify the subsets of oligodendrocyte lineage cells. Cell-to-cell interaction analysis was performed to detect the mechanism of observed changes in oligodendrocyte generation. Results Oligodendrocytes were observed to up-regulate several senescence associated genes in aged brain. Four clusters of oligodendrocyte precursor cells (OPCs) were identified in both young and aged brains. The number of those OPCs in basal state was significantly increased, while the OPCs in the procedure of differentiation were immensely decreased in aged brain. Furthermore, it was identified that activated microglia in the aged brain released inflammatory factors to suppress OPC differentiation. Stat1 might be a potential target to transform senescent microglia into tissue repair type to promote oligodendrocyte generation. Conclusion These results provided a perspective on how age activated microglia could impede remyelination and might give a new therapeutic target for age-related remyelinating diseases.
Collapse
Affiliation(s)
- Weimin Luan
- Department of Neurology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiqian Qi
- Department of Neurology, Ningbo Municipal Hospital of T.C.M., Ningbo, Zhejiang, People's Republic of China
| | - Feng Liang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaotao Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ziyang Jin
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ligen Shi
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Benyan Luo
- Department of Neurology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xuejiao Dai
- Department of Neurology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
140
|
Scalabrino G. New Epidermal-Growth-Factor-Related Insights Into the Pathogenesis of Multiple Sclerosis: Is It Also Epistemology? Front Neurol 2021; 12:754270. [PMID: 34899572 PMCID: PMC8664554 DOI: 10.3389/fneur.2021.754270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/05/2021] [Indexed: 11/23/2022] Open
Abstract
Recent findings showing that epidermal growth factor (EGF) is significantly decreased in the cerebrospinal fluid (CSF) and spinal cord (SC) of living or deceased multiple sclerosis (MS) patients, and that its repeated administration to rodents with chemically- or virally-induced demyelination of the central nervous system (CNS) or experimental allergic encephalomyelitis (EAE) prevents demyelination and inflammatory reactions in the CNS, have led to a critical reassessment of the MS pathogenesis, partly because EGF is considered to have little or no role in immunology. EGF is the only myelinotrophic factor that has been tested in the CSF and spinal cord of MS patients, and it has been shown there is a good correspondence between liquid and tissue levels. This review: (a) briefly summarises the positive EGF effects on neural stem cells, oligodendrocyte cell lineage, and astrocytes in order to explain, at least in part, the biological basis of the myelin loss and remyelination failure in MS; and (b) after a short analysis of the evolution of the principle of cause-effect in the history of Western philosophy, highlights the lack of any experimental immune-, toxin-, or virus-mediated model that precisely reproduces the histopathological features and “clinical” symptoms of MS, thus underlining the inapplicability of Claude Bernard's crucial sequence of “observation, hypothesis, and hypothesis testing.” This is followed by a discussion of most of the putative non-immunologically-linked points of MS pathogenesis (abnormalities in myelinotrophic factor CSF levels, oligodendrocytes (ODCs), astrocytes, extracellular matrix, and epigenetics) on the basis of Popper's falsification principle, and the suggestion that autoimmunity and phologosis reactions (surely the most devasting consequences of the disease) are probably the last links in a chain of events that trigger the reactions. As it is likely that there is a lack of other myelinotrophic growth factors because myelinogenesis is controlled by various CNS and extra-CNS growth factors and other molecules within and outside ODCs, further studies are needed to investigate the role of non-immunological molecules at the time of the onset of the disease. In the words of Galilei, the human mind should be prepared to understand what nature has created.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
141
|
Faw TD, Lakhani B, Schmalbrock P, Knopp MV, Lohse KR, Kramer JLK, Liu H, Nguyen HT, Phillips EG, Bratasz A, Fisher LC, Deibert RJ, Boyd LA, McTigue DM, Basso DM. Eccentric rehabilitation induces white matter plasticity and sensorimotor recovery in chronic spinal cord injury. Exp Neurol 2021; 346:113853. [PMID: 34464653 PMCID: PMC10084731 DOI: 10.1016/j.expneurol.2021.113853] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/04/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
Experience-dependent white matter plasticity offers new potential for rehabilitation-induced recovery after neurotrauma. This first-in-human translational experiment combined myelin water imaging in humans and genetic fate-mapping of oligodendrocyte lineage cells in mice to investigate whether downhill locomotor rehabilitation that emphasizes eccentric muscle actions promotes white matter plasticity and recovery in chronic, incomplete spinal cord injury (SCI). In humans, of 20 individuals with SCI that enrolled, four passed the imaging screen and had myelin water imaging before and after a 12-week (3 times/week) downhill locomotor treadmill training program (SCI + DH). One individual was excluded for imaging artifacts. Uninjured control participants (n = 7) had two myelin water imaging sessions within the same day. Changes in myelin water fraction (MWF), a histopathologically-validated myelin biomarker, were analyzed in a priori motor learning and non-motor learning brain regions and the cervical spinal cord using statistical approaches appropriate for small sample sizes. PDGFRα-CreERT2:mT/mG mice, that express green fluorescent protein on oligodendrocyte precursor cells and subsequent newly-differentiated oligodendrocytes upon tamoxifen-induced recombination, were either naive (n = 6) or received a moderate (75 kilodyne), contusive SCI at T9 and were randomized to downhill training (n = 6) or unexercised groups (n = 6). We initiated recombination 29 days post-injury, seven days prior to downhill training. Mice underwent two weeks of daily downhill training on the same 10% decline grade used in humans. Between-group comparison of functional (motor and sensory) and histological (oligodendrogenesis, oligodendroglial/axon interaction, paranodal structure) outcomes occurred post-training. In humans with SCI, downhill training increased MWF in brain motor learning regions (postcentral, precuneus) and mixed motor and sensory tracts of the ventral cervical spinal cord compared to control participants (P < 0.05). In mice with thoracic SCI, downhill training induced oligodendrogenesis in cervical dorsal and lateral white matter, increased axon-oligodendroglial interactions, and normalized paranodal structure in dorsal column sensory tracts (P < 0.05). Downhill training improved sensorimotor recovery in mice by normalizing hip and knee motor control and reducing hyperalgesia, both of which were associated with new oligodendrocytes in the cervical dorsal columns (P < 0.05). Our findings indicate that eccentric-focused, downhill rehabilitation promotes white matter plasticity and improved function in chronic SCI, likely via oligodendrogenesis in nervous system regions activated by the training paradigm. Together, these data reveal an exciting role for eccentric training in white matter plasticity and sensorimotor recovery after SCI.
Collapse
Affiliation(s)
- Timothy D Faw
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA
| | - Bimal Lakhani
- Department of Physical Therapy, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Petra Schmalbrock
- Department of Radiology, The Ohio State University, Columbus, OH 43210, USA
| | - Michael V Knopp
- Department of Radiology, The Ohio State University, Columbus, OH 43210, USA
| | - Keith R Lohse
- Department of Health, Kinesiology, and Recreation, University of Utah, Salt Lake City, UT 84112, USA; Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT 84108, USA
| | - John L K Kramer
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Hanwen Liu
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; Department of Physics and Astronomy, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Huyen T Nguyen
- Department of Radiology, The Ohio State University, Columbus, OH 43210, USA
| | - Eileen G Phillips
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Anna Bratasz
- Small Animal Imaging Shared Resources, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Lesley C Fisher
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Rochelle J Deibert
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Lara A Boyd
- Department of Physical Therapy, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Dana M McTigue
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - D Michele Basso
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
142
|
Sex differences in myelin content of white matter tracts in adolescents with depression. Neuropsychopharmacology 2021; 46:2295-2303. [PMID: 34215842 PMCID: PMC8580976 DOI: 10.1038/s41386-021-01078-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/28/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023]
Abstract
Depression is a chronic and debilitating condition that often emerges during adolescence, a period of significant brain maturation. Few studies, however, have examined how mechanisms of neuroplasticity, including myelination, are affected by adolescent-onset depression. Here, we used multimodal MR imaging to characterize myelin, indexed by R1, in white matter tracts previously associated with depression and compare 48 adolescents with lifetime depression (45 with current depression, 3 remitted) and 35 healthy controls in R1. Compared to healthy controls, R1 was higher in adolescents with lifetime depression in the uncinate fasciculus and corpus callosum genu (all βs > 0.42; all ps < 0.037). Sex significantly moderated the association between depression and R1 in the left uncinate fasciculus and corpus callosum genu (all βs > 0.86; all ps < 0.02), such that depressed female adolescents had significantly higher R1 in these tracts than did healthy female adolescents (all βs > 0.82; all ps < 0.0012). In contrast, depressed and non-depressed male adolescents did not differ in R1 in these tracts (all ps > 0.32). While fractional anisotropy (FA), a commonly examined measure of white matter organization based on diffusion-weighted MRI, in the left uncinate was positively associated with lifetime depression in our sample (β = 0.56; p = 0.016), we found no evidence of sex-specific effects of depression in FA. Our results suggest that R1 is more sensitive to sex-specific effects of depression than FA, particularly in female adolescents. Given evidence that myelin inhibits synapse formation and reduces brain plasticity, our findings implicate experience-driven regional myelination as a mechanism underlying depression during periods of significant neural maturation such as adolescence.
Collapse
|
143
|
The oligodendrocyte-enriched orphan G protein-coupled receptor Gpr62 is dispensable for central nervous system myelination. Neural Dev 2021; 16:6. [PMID: 34844642 PMCID: PMC8630896 DOI: 10.1186/s13064-021-00156-y] [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: 06/24/2021] [Accepted: 11/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Myelination is a highly regulated process in the vertebrate central nervous system (CNS) whereby oligodendrocytes wrap axons with multiple layers of insulating myelin in order to allow rapid electrical conduction. Establishing the proper pattern of myelin in neural circuits requires communicative axo-glial interactions, however, the molecular interactions that occur between oligodendrocytes and axons during developmental myelination and myelin maintenance remain to be fully elucidated. Our previous work identified G protein-coupled receptor 62 (Gpr62), an uncharacterized orphan g-protein coupled receptor, as being selectively expressed by mature oligodendrocytes within the CNS, suggesting a potential role in myelination or axoglial interactions. However, no studies to date have assessed the functional requirement for Gpr62 in oligodendrocyte development or CNS myelination. METHODS To address this, we generated a knockout mouse strain lacking the Gpr62 gene. We assessed CNS myelination during both postnatal development and adulthood using immunohistochemistry, electron microscopy and western blot. In addition, we utilized AAV-mediated expression of a tagged Gpr62 in oligodendrocytes to determine the subcellular localization of the protein in vivo. RESULTS We find that virally expressed Gpr62 protein is selectively expressed on the adaxonal myelin layer, suggestive of a potential role for Gpr62 in axo-myelinic signaling. Nevertheless, Gpr62 knockout mice display normal oligodendrocyte numbers and apparently normal myelination within the CNS during both postnatal development and adulthood. CONCLUSIONS We conclude that in spite of being well-placed to mediate neuronal-oligodendrocyte communications, Gpr62 is overall dispensable for CNS myelination.
Collapse
|
144
|
Zhang X, Huang N, Xiao L, Wang F, Li T. Replenishing the Aged Brains: Targeting Oligodendrocytes and Myelination? Front Aging Neurosci 2021; 13:760200. [PMID: 34899272 PMCID: PMC8656359 DOI: 10.3389/fnagi.2021.760200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Aging affects almost all the aspects of brain functions, but the mechanisms remain largely undefined. Increasing number of literatures have manifested the important role of glial cells in regulating the aging process. Oligodendroglial lineage cell is a major type of glia in central nervous system (CNS), composed of mature oligodendrocytes (OLs), and oligodendroglia precursor cells (OPCs). OLs produce myelin sheaths that insulate axons and provide metabolic support to meet the energy demand. OPCs maintain the population throughout lifetime with the abilities to proliferate and differentiate into OLs. Increasing evidence has shown that oligodendroglial cells display active dynamics in adult and aging CNS, which is extensively involved in age-related brain function decline in the elderly. In this review, we summarized present knowledge about dynamic changes of oligodendroglial lineage cells during normal aging and discussed their potential roles in age-related functional decline. Especially, focused on declined myelinogenesis during aging and underlying mechanisms. Clarifying those oligodendroglial changes and their effects on neurofunctional decline may provide new insights in understanding aging associated brain function declines.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Ophthalmology, The General Hospital of Western Theater Command, Chengdu, China
| | - Nanxin Huang
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lan Xiao
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fei Wang
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Tao Li
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
145
|
Experience-dependent myelination following stress is mediated by the neuropeptide dynorphin. Neuron 2021; 109:3619-3632.e5. [PMID: 34536353 DOI: 10.1016/j.neuron.2021.08.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/14/2021] [Accepted: 08/13/2021] [Indexed: 11/22/2022]
Abstract
Emerging evidence implicates experience-dependent myelination in learning and memory. However, the specific signals underlying this process remain unresolved. We demonstrate that the neuropeptide dynorphin, which is released from neurons upon high levels of activity, promotes experience-dependent myelination. Following forced swim stress, an experience that induces striatal dynorphin release, we observe increased striatal oligodendrocyte precursor cell (OPC) differentiation and myelination, which is abolished by deleting dynorphin or blocking its endogenous receptor, kappa opioid receptor (KOR). We find that dynorphin also promotes developmental OPC differentiation and myelination and demonstrate that this effect requires KOR expression specifically in OPCs. We characterize dynorphin-expressing neurons and use genetic sparse labeling to trace their axonal projections. Surprisingly, we find that they are unmyelinated normally and following forced swim stress. We propose a new model whereby experience-dependent and developmental myelination is mediated by unmyelinated, neuropeptide-expressing neurons that promote OPC differentiation for the myelination of neighboring axons.
Collapse
|
146
|
Kang M, Yao Y. Laminin regulates oligodendrocyte development and myelination. Glia 2021; 70:414-429. [PMID: 34773273 DOI: 10.1002/glia.24117] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 11/08/2022]
Abstract
Oligodendrocytes are the cells that myelinate axons and provide trophic support to neurons in the CNS. Their dysfunction has been associated with a group of disorders known as demyelinating diseases, such as multiple sclerosis. Oligodendrocytes are derived from oligodendrocyte precursor cells, which differentiate into premyelinating oligodendrocytes and eventually mature oligodendrocytes. The development and function of oligodendrocytes are tightly regulated by a variety of molecules, including laminin, a major protein of the extracellular matrix. Accumulating evidence suggests that laminin actively regulates every aspect of oligodendrocyte biology, including survival, migration, proliferation, differentiation, and myelination. How can laminin exert such diverse functions in oligodendrocytes? It is speculated that the distinct laminin isoforms, laminin receptors, and/or key signaling molecules expressed in oligodendrocytes at different developmental stages are the reasons. Understanding molecular targets and signaling pathways unique to each aspect of oligodendrocyte biology will enable more accurate manipulation of oligodendrocyte development and function, which may have implications in the therapies of demyelinating diseases. Here in this review, we first introduce oligodendrocyte biology, followed by the expression of laminin and laminin receptors in oligodendrocytes and other CNS cells. Next, the functions of laminin in oligodendrocyte biology, including survival, migration, proliferation, differentiation, and myelination, are discussed in detail. Last, key questions and challenges in the field are discussed. By providing a comprehensive review on laminin's roles in OL lineage cells, we hope to stimulate novel hypotheses and encourage new research in the field.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
147
|
Zhao N, Huang W, Cãtãlin B, Scheller A, Kirchhoff F. L-Type Ca 2+ Channels of NG2 Glia Determine Proliferation and NMDA Receptor-Dependent Plasticity. Front Cell Dev Biol 2021; 9:759477. [PMID: 34746151 PMCID: PMC8567174 DOI: 10.3389/fcell.2021.759477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/16/2021] [Indexed: 12/25/2022] Open
Abstract
NG2 (nerve/glial antigen 2) glia are uniformly distributed in the gray and white matter of the central nervous system (CNS). They are the major proliferating cells in the brain and can differentiate into oligodendrocytes. NG2 glia do not only receive synaptic input from excitatory and inhibitory neurons, but also secrete growth factors and cytokines, modulating CNS homeostasis. They express several receptors and ion channels that play a role in rapidly responding upon synaptic signals and generating fast feedback, potentially regulating their own properties. Ca2+ influx via voltage-gated Ca2+ channels (VGCCs) induces an intracellular Ca2+ rise initiating a series of cellular activities. We confirmed that NG2 glia express L-type VGCCs in the white and gray matter during CNS development, particularly in the early postnatal stage. However, the function of L-type VGCCs in NG2 glia remains elusive. Therefore, we deleted L-type VGCC subtypes Cav1.2 and Cav1.3 genes conditionally in NG2 glia by crossbreeding NG2-CreERT2 knock-in mice to floxed Cav1.2 and flexed Cav1.3 transgenic mice. Our results showed that ablation of Cav1.2 and Cav1.3 strongly inhibited the proliferation of cortical NG2 glia, while differentiation in white and gray matter was not affected. As a consequence, no difference on myelination could be detected in various brain regions. In addition, we observed morphological alterations of the nodes of Ranvier induced by VGCC-deficient NG2 glia, i.e., shortened paired paranodes in the corpus callosum. Furthermore, deletion of Cav1.2 and Cav1.3 largely eliminated N-methyl-D-aspartate (NMDA)-dependent long-term depression (LTD) and potentiation in the hippocampus while the synaptic input to NG2 glia from axons remained unaltered. We conclude that L-type VGCCs of NG2 glia are essential for cell proliferation and proper structural organization of nodes of Ranvier, but not for differentiation and myelin compaction. In addition, L-type VGCCs of NG2 glia contribute to the regulation of long-term neuronal plasticity.
Collapse
Affiliation(s)
- Na Zhao
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Wenhui Huang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Bogdan Cãtãlin
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany.,Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Anja Scheller
- 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.,Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
148
|
Niedzwiedz-Massey VM, Douglas JC, Rafferty T, Kane CJ, Drew PD. Ethanol effects on cerebellar myelination in a postnatal mouse model of fetal alcohol spectrum disorders. Alcohol 2021; 96:43-53. [PMID: 34358666 DOI: 10.1016/j.alcohol.2021.07.003] [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: 03/10/2021] [Revised: 06/12/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022]
Abstract
Fetal alcohol spectrum disorders (FASD) are alarmingly common, result in significant personal and societal loss, and there are no effective treatments for these disorders. Cerebellar neuropathology is common in FASD and can cause impaired cognitive and motor function. The current study evaluates the effects of ethanol on oligodendrocyte-lineage cells, as well as molecules that modulate oligodendrocyte differentiation and function in the cerebellum in a postnatal mouse model of FASD. Neonatal mice were treated with ethanol from P4-P9 (postnatal day), the cerebellum was isolated at P10, and mRNAs encoding oligodendrocyte-associated molecules were quantitated by qRT-PCR. Our studies demonstrated that ethanol significantly reduced the expression of markers for multiple stages of oligodendrocyte maturation, including oligodendrocyte precursor cells, pre-myelinating oligodendrocytes, and mature myelinating oligodendrocytes. Additionally, we determined that ethanol significantly decreased the expression of molecules that play critical roles in oligodendrocyte differentiation. Interestingly, we also observed that ethanol significantly reduced the expression of myelin-associated inhibitors, which may act as a compensatory mechanism to ethanol toxicity. Furthermore, we demonstrate that ethanol alters the expression of a variety of molecules important in oligodendrocyte function and myelination. Collectively, our studies increase our understanding of specific mechanisms by which ethanol modulates myelination in the developing cerebellum, and potentially identify novel targets for FASD therapy.
Collapse
|
149
|
Periods of synchronized myelin changes shape brain function and plasticity. Nat Neurosci 2021; 24:1508-1521. [PMID: 34711959 DOI: 10.1038/s41593-021-00917-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 07/30/2021] [Indexed: 12/11/2022]
Abstract
Myelin, a lipid membrane that wraps axons, enabling fast neurotransmission and metabolic support to axons, is conventionally thought of as a static structure that is set early in development. However, recent evidence indicates that in the central nervous system (CNS), myelination is a protracted and plastic process, ongoing throughout adulthood. Importantly, myelin is emerging as a potential modulator of neuronal networks, and evidence from human studies has highlighted myelin as a major player in shaping human behavior and learning. Here we review how myelin changes throughout life and with learning. We discuss potential mechanisms of myelination at different life stages, explore whether myelin plasticity provides the regenerative potential of the CNS white matter, and question whether changes in myelin may underlie neurological disorders.
Collapse
|
150
|
Grisé KN, Coles BLK, Bautista NX, van der Kooy D. Activation of adult mammalian retinal stem cells in vivo via antagonism of BMP and sFRP2. Stem Cell Res Ther 2021; 12:560. [PMID: 34717744 PMCID: PMC8557620 DOI: 10.1186/s13287-021-02630-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/17/2021] [Indexed: 11/15/2022] Open
Abstract
Background The adult mammalian retina does not have the capacity to regenerate cells lost due to damage or disease. Therefore, retinal injuries and blinding diseases result in irreversible vision loss. However, retinal stem cells (RSCs), which participate in retinogenesis during development, persist in a quiescent state in the ciliary epithelium (CE) of the adult mammalian eye. Moreover, RSCs retain the ability to generate all retinal cell types when cultured in vitro, including photoreceptors. Therefore, it may be possible to activate endogenous RSCs to induce retinal neurogenesis in vivo and restore vision in the adult mammalian eye. Methods To investigate if endogenous RSCs can be activated, we performed combinatorial intravitreal injections of antagonists to BMP and sFRP2 proteins (two proposed mediators of RSC quiescence in vivo), with or without growth factors FGF and Insulin. We also investigated the effects of chemically-induced N-methyl-N-Nitrosourea (MNU) retinal degeneration on RSC activation, both alone and in combination withthe injected factors. Further, we employed inducible Msx1-CreERT2 genetic lineage labeling of the CE followed by stimulation paradigms to determine if activated endogenous RSCs could migrate into the retina and differentiate into retinal neurons. Results We found that in vivo antagonism of BMP and sFRP2 proteins induced CE cells in the RSC niche to proliferate and expanded the RSC population. BMP and sFRP2 antagonism also enhanced CE cell proliferation in response to exogenous growth factor stimulation and MNU-induced retinal degeneration. Furthermore, Msx1-CreERT2 genetic lineage tracing revealed that CE cells migrated into the retina following stimulation and/or injury, where they expressed markers of mature photoreceptors and retinal ganglion cells. Conclusions Together, these results indicate that endogenous adult mammalian RSCs may have latent regenerative potential that can be activated by modulating the RSC niche and hold promise as a means for endogenous retinal cell therapy to repair the retina and improve vision. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02630-0.
Collapse
Affiliation(s)
- Kenneth N Grisé
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON, M5S 3E1, Canada.
| | - Brenda L K Coles
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Nelson X Bautista
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Derek van der Kooy
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON, M5S 3E1, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A8, Canada
| |
Collapse
|