1
|
Pascual-Guerra J, Torres-Rico M, Marín-Rodríguez B, Arasmou-Idrovo MS, García AG, Rodríguez-Navarro JA, Paíno CL. Repurposed Drugs to Enhance the Therapeutic Potential of Oligodendrocyte Precursor Cells Derived from Adult Rat Adipose Tissue. Cells 2025; 14:533. [PMID: 40214487 PMCID: PMC11988185 DOI: 10.3390/cells14070533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Failure in the proliferation, recruitment, mobilization, and/or differentiation of oligodendrocyte precursor cells (OPCs) impedes remyelination in central nervous system (CNS) demyelinating diseases. Our group has recently achieved the generation of functional oligodendroglia through direct lineage conversion by expressing Sox10, Olig2, and Zfp536 genes in adult rat adipose tissue-derived stromal cells. The present study aimed to determine whether various repurposed drugs or molecules could enhance the myelinating capacities of these induced OPCs (iOPCs). We report that kainate, benztropine, miconazole, clobetasol, and baclofen promote in vitro iOPCs migration, differentiation, and ensheathing abilities through mechanisms similar to those observed in rat neural stem cell-derived OPCs. This research supports the potential use of iOPCs as they provide an alternative and reliable cell source for testing the effects of in vitro promyelinating repurposed drugs and for assessing the molecular and cellular mechanisms involved in therapeutic strategies for demyelinating diseases.
Collapse
Affiliation(s)
- J. Pascual-Guerra
- Servicio de Neurobiología-Investigación, IRYCIS, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain (C.L.P.)
- Fundación Teófilo Hernando, 28290 Madrid, Spain
| | | | | | | | - A. G. García
- Fundación Teófilo Hernando, 28290 Madrid, Spain
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28023 Madrid, Spain
| | - J. A. Rodríguez-Navarro
- Servicio de Neurobiología-Investigación, IRYCIS, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain (C.L.P.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto Universitario de Investigación Neuroquímica (IUIN), Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - C. L. Paíno
- Servicio de Neurobiología-Investigación, IRYCIS, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain (C.L.P.)
| |
Collapse
|
2
|
Hamwi M, Thebault S, Melkus G, Auriat AM, Pham A, Carrington A, Thornhill R, Walker LAS, Chakraborty S, Torres C, Zhang L, Atkins HL, Freedman MS, Aviv RI. MRI graph parameters are longitudinal markers of neuronal integrity in multiple sclerosis. Mult Scler Relat Disord 2023; 80:105066. [PMID: 39491411 DOI: 10.1016/j.msard.2023.105066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 11/05/2024]
Abstract
PURPOSE We sought to determine if structural network parameters add to traditional markers of MS treatment response following immunoablation and autologous haemopoietic stem cell transplantation (IAHSCT). The post-IAHSCT paradigm afforded us the opportunity to study MS patients after relapsing biology had been effectively suppressed, enabling us to study the cortical substrate of progressive MS in a less confounded manner. METHODS In this analysis of data from a phase 2 prospective study, associations between magnetic resonance graph parameters, N-acetylaspartate to creatine ratio (NAA/Cr), and serum neurofilament light chain (sNfL), amongst other markers, were assessed at 3 months pre-and 12 months post-IAHSCT. Correlations between graph parameter score changes and markers of brain health were calculated. Predictive factors of NAA/Cr or sNfL levels were calculated, adjusting for reference models. Model improvements were evaluated using the G2 likelihood-ratio test. RESULTS 24 patients (aged 18-38) were evaluated. Post-IAHSCT, high NAA/Cr and low sNfL (both measures of neuronal injury) were respectively associated with more favourable degree, density, clustering and path lengths, and degree, γ, and path length. Post-IAHSCT, absolute change in degree, path length and γ were associated with NAA/Cr and sNfL. Multivariate analysis demonstrated that the relative change in network parameters after IAHSCT accounted for 14% and 35% more variance in NAA/Cr and sNfL levels respectively than the reference model alone. CONCLUSIONS Cross-sectionally and longitudinally, network parameters demonstrate added utility as markers of disease severity in MS. These measures have the potential to capture cortical changes relevant to progressive non-relapsing biology in MS.
Collapse
Affiliation(s)
- Milad Hamwi
- Ottawa Hospital Research Institute; University of Ottawa, Department of Radiology, Radiation Oncology and Medical Physics
| | | | - Gerd Melkus
- Ottawa Hospital Research Institute; University of Ottawa, Department of Radiology, Radiation Oncology and Medical Physics; University of Ottawa, Brain and Mind Research Institute
| | - Angela M Auriat
- Ottawa Hospital Research Institute; University of Ottawa, Department of Radiology, Radiation Oncology and Medical Physics; University of Ottawa, Brain and Mind Research Institute; University of Ottawa, Faculty of Medicine.
| | - Alex Pham
- Ottawa Hospital Research Institute; University of Ottawa, Department of Radiology, Radiation Oncology and Medical Physics
| | - André Carrington
- Ottawa Hospital Research Institute; University of Ottawa, Department of Radiology, Radiation Oncology and Medical Physics; University of Waterloo, Department of Systems Design Engineering
| | - Rebecca Thornhill
- Ottawa Hospital Research Institute; University of Ottawa, Department of Radiology, Radiation Oncology and Medical Physics
| | - Lisa A S Walker
- Ottawa Hospital Research Institute; Ottawa Hospital, Department of Psychology; University of Ottawa, Brain and Mind Research Institute; University of Ottawa, Faculty of Medicine
| | - Santanu Chakraborty
- Ottawa Hospital, Department of Medical Imaging; University of Ottawa, Department of Radiology, Radiation Oncology and Medical Physics; University of Ottawa, Brain and Mind Research Institute
| | - Carlos Torres
- Ottawa Hospital Research Institute; Ottawa Hospital, Department of Medical Imaging; University of Ottawa, Department of Radiology, Radiation Oncology and Medical Physics; University of Ottawa, Brain and Mind Research Institute
| | - Liying Zhang
- Department of Medical Imaging Sunnybrook Health Sciences Centre
| | - Harold L Atkins
- Ottawa Hospital Research Institute; Ottawa Hospital Blood and Marrow Transplant Program; University of Ottawa, Faculty of Medicine
| | - Mark S Freedman
- Ottawa Hospital, Department of Neurology; University of Ottawa, Brain and Mind Research Institute
| | - Richard I Aviv
- Ottawa Hospital, Department of Medical Imaging; University of Ottawa, Department of Radiology, Radiation Oncology and Medical Physics; University of Ottawa, Brain and Mind Research Institute.
| |
Collapse
|
3
|
Wang CY, Zuo Z, Jo J, Kim KI, Madamba C, Ye Q, Jung SY, Bellen HJ, Lee HK. Daam2 phosphorylation by CK2α negatively regulates Wnt activity during white matter development and injury. Proc Natl Acad Sci U S A 2023; 120:e2304112120. [PMID: 37607236 PMCID: PMC10469030 DOI: 10.1073/pnas.2304112120] [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: 03/13/2023] [Accepted: 07/20/2023] [Indexed: 08/24/2023] Open
Abstract
Wnt signaling plays an essential role in developmental and regenerative myelination in the central nervous system. The Wnt signaling pathway is composed of multiple regulatory layers; thus, how these processes are coordinated to orchestrate oligodendrocyte (OL) development remains unclear. Here, we show CK2α, a Wnt/β-catenin signaling Ser/Thr kinase, phosphorylates Daam2, inhibiting its function and Wnt activity during OL development. Intriguingly, we found Daam2 phosphorylation differentially impacts distinct stages of OL development, accelerating early differentiation followed by decelerating maturation and myelination. Application toward white matter injury revealed CK2α-mediated Daam2 phosphorylation plays a protective role for developmental and behavioral recovery after neonatal hypoxia, while promoting myelin repair following adult demyelination. Together, our findings identify a unique regulatory node in the Wnt pathway that regulates OL development via protein phosphorylation-induced signaling complex instability and highlights a new biological mechanism for myelin restoration.
Collapse
Affiliation(s)
- Chih-Yen Wang
- Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX77030
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan70101, Taiwan
| | - Zhongyuan Zuo
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
| | - Juyeon Jo
- Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX77030
| | - Kyoung In Kim
- Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX77030
| | - Christine Madamba
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX77030
- Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX77030
| | - Qi Ye
- Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX77030
| | - Sung Yun Jung
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
| | - Hugo J. Bellen
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
| | - Hyun Kyoung Lee
- Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX77030
- Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
| |
Collapse
|
4
|
Wang CY, Zuo Z, Kim KI, Bellen HJ, Lee HK. CK2α-dependent regulation of Wnt activity governs white matter development and repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536369. [PMID: 37090554 PMCID: PMC10120613 DOI: 10.1101/2023.04.11.536369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Wnt signaling plays an essential role in developmental and regenerative myelination in the CNS. The Wnt signaling pathway is comprised of multiple regulatory layers; thus, how these processes are coordinated to orchestrate oligodendrocyte development remains unclear. Here we show CK2α, a Wnt/β-catenin signaling Ser/Thr kinase, phosphorylates Daam2, inhibiting its function and Wnt-activity during oligodendrocyte development. Intriguingly, we found Daam2 phosphorylation differentially impacts distinct stages of oligodendrocyte development, accelerating early differentiation followed by decelerating maturation and myelination. Application towards white matter injury revealed CK2α-mediated Daam2 phosphorylation plays a protective role for developmental and behavioral recovery after neonatal hypoxia, while promoting myelin repair following adult demyelination. Together, our findings identify a novel regulatory node in the Wnt pathway that regulates oligodendrocyte development via protein phosphorylation-induced signaling complex instability and highlights a new biological mechanism for myelin restoration. Significance Wnt signaling plays a vital role in OL development and has been implicated as an adverse event for myelin repair after white matter injury. Emerging studies have shed light on multi-modal roles of Wnt effectors in the OL lineage, but the underlying molecular mechanisms and modifiable targets in OL remyelination remain unclear. Using genetic mouse development and injury model systems, we delineate a novel stage-specific function of Daam2 in Wnt signaling and OL development via a S704/T7-5 phosphorylation mechanism, and determine a new role of the kinase CK2α in contributing to OL development. In-depth understanding of CK2α-Daam2 pathway regulation will allow us to precisely modulate its activity in conjunction with Wnt signaling and harness its biology for white matter pathology.
Collapse
|
5
|
Kremsky I, Ma Q, Li B, Dasgupta C, Chen X, Ali S, Angeloni S, Wang C, Zhang L. Fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes. Cell Biosci 2023; 13:58. [PMID: 36932456 PMCID: PMC10022003 DOI: 10.1186/s13578-023-01012-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/10/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Fetal hypoxia causes vital, systemic, developmental malformations in the fetus, particularly in the brain, and increases the risk of diseases in later life. We previously demonstrated that fetal hypoxia exposure increases the susceptibility of the neonatal brain to hypoxic-ischemic insult. Herein, we investigate the effect of fetal hypoxia on programming of cell-specific transcriptomes in the brain of neonatal rats. RESULTS We obtained RNA sequencing (RNA-seq) data from neurons, microglia, oligodendrocytes, A2B5+ oligodendrocyte precursor cells, and astrocytes from male and female neonatal rats subjected either to fetal hypoxia or control conditions. Substantial transcriptomic responses to fetal hypoxia occurred in neurons, microglia, oligodendrocytes, and A2B5+ cells. Not only were the transcriptomic responses unique to each cell type, but they also occurred with a great deal of sexual dimorphism. We validated differential expression of several genes related to inflammation and cell death by Real-time Quantitative Polymerase Chain Reaction (qRT-PCR). Pathway and transcription factor motif analyses suggested that the NF-kappa B (NFκB) signaling pathway was enriched in the neonatal male brain due to fetal hypoxia, and we verified this result by transcription factor assay of NFκB-p65 in whole brain. CONCLUSIONS Our study reveals a significant impact of fetal hypoxia on the transcriptomes of neonatal brains in a cell-specific and sex-dependent manner, and provides mechanistic insights that may help explain the development of hypoxic-ischemic sensitive phenotypes in the neonatal brain.
Collapse
Affiliation(s)
- Isaac Kremsky
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Qingyi Ma
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Bo Li
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Chiranjib Dasgupta
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Xin Chen
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Samir Ali
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Shawnee Angeloni
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Charles Wang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Lubo Zhang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA. .,Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
6
|
Ahmed T. Neural stem cell engineering for the treatment of multiple sclerosis. BIOMEDICAL ENGINEERING ADVANCES 2022. [DOI: 10.1016/j.bea.2022.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
7
|
Beyer BA, Lairson LL. Promoting remyelination: A case study in regenerative medicine. Curr Opin Chem Biol 2022; 70:102201. [PMID: 36037558 DOI: 10.1016/j.cbpa.2022.102201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/03/2022]
Abstract
Therapeutics that modulate regenerative mechanisms by targeting the activity of endogenous (adult) stem cell populations have the potential to revolutionize medicine. In many human disease states, capacity to repair damaged tissue underlies progressive decline and disease progression. Recent insights derived from efforts aimed at promoting remyelination for the treatment of multiple sclerosis (MS) highlight the importance of considering the limiting factors and underlying mechanisms associated with all aspects of disease onset, progression and recovery, during both the discovery and clinical stages of developing a regenerative medicine. This perspective presents general considerations for the development of regenerative therapies, using remyelination as a case study.
Collapse
Affiliation(s)
- Brittney A Beyer
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Luke L Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
8
|
Cannabinoid CB 1 receptor gene inactivation in oligodendrocyte precursors disrupts oligodendrogenesis and myelination in mice. Cell Death Dis 2022; 13:585. [PMID: 35798697 PMCID: PMC9263142 DOI: 10.1038/s41419-022-05032-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 01/21/2023]
Abstract
Cannabinoids are known to modulate oligodendrogenesis and developmental CNS myelination. However, the cell-autonomous action of these compounds on oligodendroglial cells in vivo, and the molecular mechanisms underlying these effects have not yet been studied. Here, by using oligodendroglial precursor cell (OPC)-targeted genetic mouse models, we show that cannabinoid CB1 receptors exert an essential role in modulating OPC differentiation at the critical periods of postnatal myelination. We found that selective genetic inactivation of CB1 receptors in OPCs in vivo perturbs oligodendrogenesis and postnatal myelination by altering the RhoA/ROCK signaling pathway, leading to hypomyelination, and motor and cognitive alterations in young adult mice. Conversely, pharmacological CB1 receptor activation, by inducing E3 ubiquitin ligase-dependent RhoA proteasomal degradation, promotes oligodendrocyte development and CNS myelination in OPCs, an effect that was not evident in OPC-specific CB1 receptor-deficient mice. Moreover, pharmacological inactivation of ROCK in vivo overcomes the defects in oligodendrogenesis and CNS myelination, and behavioral alterations found in OPC-specific CB1 receptor-deficient mice. Overall, this study supports a cell-autonomous role for CB1 receptors in modulating oligodendrogenesis in vivo, which may have a profound impact on the scientific knowledge and therapeutic manipulation of CNS myelination by cannabinoids.
Collapse
|
9
|
Tong LY, Deng YB, Du WH, Zhou WZ, Liao XY, Jiang X. Clemastine Promotes Differentiation of Oligodendrocyte Progenitor Cells Through the Activation of ERK1/2 via Muscarinic Receptors After Spinal Cord Injury. Front Pharmacol 2022; 13:914153. [PMID: 35865954 PMCID: PMC9294397 DOI: 10.3389/fphar.2022.914153] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
The recovery of spinal cord injury (SCI) is closely associated with the obstruction of oligodendrocyte progenitor cell (OPC) differentiation, which ultimately induces the inability to generate newly formed myelin. To address the concern, drug-based methods may be the most practical and feasible way, possibly applying to clinical therapies for patients with SCI. In our previous study, we found that clemastine treatment preserves myelin integrity, decreases the loss of axons, and improves functional recovery in the SCI model. Clemastine acts as an antagonist of the muscarinic acetylcholine receptor (muscarinic receptor, MR) identified from a string of anti-muscarinic drugs that can enhance oligodendrocyte differentiation and myelin wrapping. However, the effects of clemastine on OPC differentiation through MRs in SCI and the underlying mechanism remain unclear. To explore the possibility, a rat model of SCI was established. To investigate if clemastine could promote the differentiation of OPCs in SCI via MR, the expressions of OPC and mature OL were detected at 7 days post injury (dpi) or at 14 dpi. The significant effect of clemastine on encouraging OPC differentiation was revealed at 14 dpi rather than 7 dpi. Under pre-treatment with the MR agonist cevimeline, the positive role of clemastine on OPC differentiation was partially disrupted. Further studies indicated that clemastine increased the phosphorylation level of extracellular signal–regulated kinase 1/2 (p-ERK1/2) and the expressions of transcription factors, Myrf and Olig2. To determine the relationship among clemastine, ERK1/2 signaling, specified transcription factors, and OPC differentiation, the ERK1/2 signaling was disturbed by U0126. The inhibition of ERK1/2 in SCI rats treated with clemastine decreased the expressions of p-ERK 1/2, Myrf, Olig2, and mature OLs, suggesting that ERK1/2 is required for clemastine on promoting OPC differentiation and that specified transcription factors may be affected by the activity of ERK1/2. Moreover, the impact of clemastine on modulating the level of p-ERK 1/2 was restricted following cevimeline pre-injecting, which provides further evidence that the role of clemastine was mediated by MRs. Altogether, our data demonstrated that clemastine, mediated by MRs, promotes OPC differentiation under the enhancement of Myrf and Olig2 by activating ERK1/2 signaling and suggests a novel therapeutic prospect for SCI recovery.
Collapse
Affiliation(s)
- Lu-Yao Tong
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Yong-Bing Deng
- Department of Chongqing Emergency Medical Center, Chongqing University Center Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Wei-Hong Du
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Wen-Zhu Zhou
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Xin-Yu Liao
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Xue Jiang
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- *Correspondence: Xue Jiang, ,
| |
Collapse
|
10
|
Tran V, Carpo N, Shaka S, Zamudio J, Choi S, Cepeda C, Espinosa-Jeffrey A. Delayed Maturation of Oligodendrocyte Progenitors by Microgravity: Implications for Multiple Sclerosis and Space Flight. Life (Basel) 2022; 12:797. [PMID: 35743828 PMCID: PMC9224676 DOI: 10.3390/life12060797] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
In previous studies, we examined the effects of space microgravity on human neural stem cells. To date, there are no studies on a different type of cell that is critical for myelination and electrical signals transmission, oligodendrocyte progenitors (OLPs). The purpose of the present study was to examine the behavior of space-flown OLPs (SPC-OLPs) as they were adapting to Earth's gravity. We found that SPC-OLPs survived, and most of them proliferated normally. Nonetheless, some of them displayed incomplete cytokinesis. Both morphological and ontogenetic analyses showed that they remained healthy and expressed the immature OLP markers Sox2, PDGFR-α, and transferrin (Tf) after space flight, which confirmed that SPC-OLPs displayed a more immature phenotype than their ground control (GC) counterparts. In contrast, GC OLPs expressed markers that usually appear later (GPDH, O4, and ferritin), indicating a delay in SPC-OLPs' development. These cells remained immature even after treatment with culture media designed to support oligodendrocyte (OL) maturation. The most remarkable and surprising finding was that the iron carrier glycoprotein Tf, previously described as an early marker for OLPs, was expressed ectopically in the nucleus of all SPC-OLPs. In contrast, their GC counterparts expressed it exclusively in the cytoplasm, as previously described. In addition, analysis of the secretome demonstrated that SPC-OLPs contained 3.5 times more Tf than that of GC cells, indicating that Tf is gravitationally regulated, opening two main fields of study to understand the upregulation of the Tf gene and secretion of the protein that keep OLPs at a progenitor stage rather than moving forward to more mature phenotypes. Alternatively, because Tf is an autocrine and paracrine factor in the central nervous system (CNS), in the absence of neurons, it accumulated in the secretome collected after space flight. We conclude that microgravity is becoming a novel platform to study why in some myelin disorders OLPs are present but do not mature.
Collapse
Affiliation(s)
- Victoria Tran
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, The University of California Los Angeles, Los Angeles, CA 90095, USA; (V.T.); (N.C.); (S.S.); (J.Z.); (C.C.)
| | - Nicholas Carpo
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, The University of California Los Angeles, Los Angeles, CA 90095, USA; (V.T.); (N.C.); (S.S.); (J.Z.); (C.C.)
| | - Sophia Shaka
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, The University of California Los Angeles, Los Angeles, CA 90095, USA; (V.T.); (N.C.); (S.S.); (J.Z.); (C.C.)
| | - Joile Zamudio
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, The University of California Los Angeles, Los Angeles, CA 90095, USA; (V.T.); (N.C.); (S.S.); (J.Z.); (C.C.)
| | - Sungshin Choi
- KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA;
| | - Carlos Cepeda
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, The University of California Los Angeles, Los Angeles, CA 90095, USA; (V.T.); (N.C.); (S.S.); (J.Z.); (C.C.)
| | - Araceli Espinosa-Jeffrey
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, The University of California Los Angeles, Los Angeles, CA 90095, USA; (V.T.); (N.C.); (S.S.); (J.Z.); (C.C.)
| |
Collapse
|
11
|
Scalabrino G. Epidermal Growth Factor in the CNS: A Beguiling Journey from Integrated Cell Biology to Multiple Sclerosis. An Extensive Translational Overview. Cell Mol Neurobiol 2022; 42:891-916. [PMID: 33151415 PMCID: PMC8942922 DOI: 10.1007/s10571-020-00989-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
This article reviews the wealth of papers dealing with the different effects of epidermal growth factor (EGF) on oligodendrocytes, astrocytes, neurons, and neural stem cells (NSCs). EGF induces the in vitro and in vivo proliferation of NSCs, their migration, and their differentiation towards the neuroglial cell line. It interacts with extracellular matrix components. NSCs are distributed in different CNS areas, serve as a reservoir of multipotent cells, and may be increased during CNS demyelinating diseases. EGF has pleiotropic differentiative and proliferative effects on the main CNS cell types, particularly oligodendrocytes and their precursors, and astrocytes. EGF mediates the in vivo myelinotrophic effect of cobalamin on the CNS, and modulates the synthesis and levels of CNS normal prions (PrPCs), both of which are indispensable for myelinogenesis and myelin maintenance. EGF levels are significantly lower in the cerebrospinal fluid and spinal cord of patients with multiple sclerosis (MS), which probably explains remyelination failure, also because of the EGF marginal role in immunology. When repeatedly administered, EGF protects mouse spinal cord from demyelination in various experimental models of autoimmune encephalomyelitis. It would be worth further investigating the role of EGF in the pathogenesis of MS because of its multifarious effects.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences, University of Milan, Via Mangiagalli 31, 20133, Milan, Italy.
| |
Collapse
|
12
|
Bu Shen Yi Sui Capsules Promote Remyelination by Regulating MicroRNA-219 and MicroRNA-338 in Exosomes to Promote Oligodendrocyte Precursor Cell Differentiation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3341481. [PMID: 35463062 PMCID: PMC9020954 DOI: 10.1155/2022/3341481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/18/2022] [Accepted: 03/18/2022] [Indexed: 11/18/2022]
Abstract
Remyelination is a refractory feature of demyelinating diseases such as multiple sclerosis (MS). Studies have shown that promoting oligodendrocyte precursor cell (OPC) differentiation, which cannot be achieved by currently available therapeutic agents, is the key to enhancing remyelination. Bu Shen Yi Sui capsule (BSYSC) is a traditional Chinese herbal medicine over many years of clinical practice. We have found that BSYSC can effectively treat MS. In this study, the effects of BSYSC in promoting OPCs differentiation and remyelination were assessed using an experimental autoimmune encephalomyelitis (EAE) model in vivo and cultured OPCs in vitro. The results showed that BSYSC reduced clinical function scores and increased neuroprotection. The expression of platelet-derived growth factor receptor α (PDGFR-α) was decreased and the level of 2′,3′-cyclic nucleotide 3′-phosphodiesterase (CNPase) was increased in the brains and spinal cords of mice as well as in OPCs after treatment with BSYSC. We further found that BSYSC elevated the expression of miR-219 or miR-338 in the serum exosomes of mice with EAE, thereby suppressing the expression of Sox6, Lingo1, and Hes5, which negatively regulate OPCs differentiation. Therefore, serum exosomes of BSYSC-treated mice (exos-BSYSC) were extracted and administered to OPCs in which miR-219 or miR-338 expression was knocked down by adenovirus, and the results showed that Sox6, Lingo1, and Hes5 expression was downregulated, MBP expression was upregulated, OPCs differentiation was increased, and the ability of OPCs to wrap around neuronal axons was improved. In conclusion, BSYSC may exert clinically relevant effects by regulating microRNA (miR) levels in exosomes and thus promoting the differentiation and maturation of OPCs.
Collapse
|
13
|
Phenytoin promotes the proliferation of oligodendrocytes and enhances the expression of myelin basic protein in the corpus callosum of mice demyelinated by cuprizone. Exp Brain Res 2022; 240:1617-1627. [PMID: 35362723 DOI: 10.1007/s00221-022-06356-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 03/21/2022] [Indexed: 11/04/2022]
Abstract
Oligodendrocyte loss and myelin sheet destruction are crucial characteristics of demyelinating diseases. Phenytoin promotes the proliferation of endogenous neural precursor cells in the ventricular-subventricular zone in the postnatal brain that help restore the oligodendroglial population. This study aimed to evaluate whether phenytoin promotes myelin recovery of the corpus callosum of demyelinated adult mice. CD1 male mice were exposed to a demyelinating agent (0.2% cuprizone) for 8 weeks. We assembled two groups: the phenytoin-treated group and the control-vehicle group. The treated group received oral phenytoin (10 mg/kg) for 4 weeks. We quantified the number of Olig2 + and NG2 + oligodendrocyte precursor cells (OPCs), Rip + oligodendrocytes, the expression level of myelin basic protein (MBP), and the muscle strength and motor coordination. The oligodendroglial lineage (Olig2 + cells, NG2 + cells, and RIP + cells) significantly increases by the phenytoin administration when compared to the control-vehicle group. The phenytoin-treated group also showed an increased expression of MBP in the corpus callosum and better functional scores in the horizontal bar test. These findings suggest that phenytoin stimulates the proliferation of OPCs, re-establishes the oligodendroglial population, promotes myelin recovery in the corpus callosum, and improves motor coordination and muscle strength.
Collapse
|
14
|
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
|
15
|
Beyrampour-Basmenj H, Rahmati M, Moghamddam MP, Kalan ME, Alivand M, Aliyari-Serej Z, Nastarin P, Omrani M, Khodakarimi S, Ebrahimi-Kalan A. Association between miRNAs expression and multiple sclerosis pathogenesis: A novel therapeutic approach. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2021.101457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
16
|
Psenicka MW, Smith BC, Tinkey RA, Williams JL. Connecting Neuroinflammation and Neurodegeneration in Multiple Sclerosis: Are Oligodendrocyte Precursor Cells a Nexus of Disease? Front Cell Neurosci 2021; 15:654284. [PMID: 34234647 PMCID: PMC8255483 DOI: 10.3389/fncel.2021.654284] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
The pathology in neurodegenerative diseases is often accompanied by inflammation. It is well-known that many cells within the central nervous system (CNS) also contribute to ongoing neuroinflammation, which can promote neurodegeneration. Multiple sclerosis (MS) is both an inflammatory and neurodegenerative disease in which there is a complex interplay between resident CNS cells to mediate myelin and axonal damage, and this communication network can vary depending on the subtype and chronicity of disease. Oligodendrocytes, the myelinating cell of the CNS, and their precursors, oligodendrocyte precursor cells (OPCs), are often thought of as the targets of autoimmune pathology during MS and in several animal models of MS; however, there is emerging evidence that OPCs actively contribute to inflammation that directly and indirectly contributes to neurodegeneration. Here we discuss several contributors to MS disease progression starting with lesion pathology and murine models amenable to studying particular aspects of disease. We then review how OPCs themselves can play an active role in promoting neuroinflammation and neurodegeneration, and how other resident CNS cells including microglia, astrocytes, and neurons can impact OPC function. Further, we outline the very complex and pleiotropic role(s) of several inflammatory cytokines and other secreted factors classically described as solely deleterious during MS and its animal models, but in fact, have many neuroprotective functions and promote a return to homeostasis, in part via modulation of OPC function. Finally, since MS affects patients from the onset of disease throughout their lifespan, we discuss the impact of aging on OPC function and CNS recovery. It is becoming clear that OPCs are not simply a bystander during MS progression and uncovering the active roles they play during different stages of disease will help uncover potential new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Morgan W. Psenicka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brandon C. Smith
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Rachel A. Tinkey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Jessica L. Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Brain Health Research Institute, Kent State University, Kent, OH, United States
| |
Collapse
|
17
|
Coppi E, Cencetti F, Cherchi F, Venturini M, Donati C, Bruni P, Pedata F, Pugliese AM. A 2 B Adenosine Receptors and Sphingosine 1-Phosphate Signaling Cross-Talk in Oligodendrogliogenesis. Front Neurosci 2021; 15:677988. [PMID: 34135730 PMCID: PMC8202686 DOI: 10.3389/fnins.2021.677988] [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: 03/08/2021] [Accepted: 04/22/2021] [Indexed: 11/13/2022] Open
Abstract
Oligodendrocyte-formed myelin sheaths allow fast synaptic transmission in the brain. Impairments in the process of myelination, or demyelinating insults, might cause chronic diseases such as multiple sclerosis (MS). Under physiological conditions, remyelination is an ongoing process throughout adult life consisting in the differentiation of oligodendrocyte progenitor cells (OPCs) into mature oligodendrocytes (OLs). During pathological events, this process fails due to unfavorable environment. Adenosine and sphingosine kinase/sphingosine 1-phosphate signaling axes (SphK/S1P) play important roles in remyelination processes. Remarkably, fingolimod (FTY720), a sphingosine analog recently approved for MS treatment, plays important roles in OPC maturation. We recently demonstrated that the selective stimulation of A2 B adenosine receptors (A2 B Rs) inhibit OPC differentiation in vitro and reduce voltage-dependent outward K+ currents (I K ) necessary to OPC maturation, whereas specific SphK1 or SphK2 inhibition exerts the opposite effect. During OPC differentiation A2 B R expression increases, this effect being prevented by SphK1/2 blockade. Furthermore, selective silencing of A2 B R in OPC cultures prompts maturation and, intriguingly, enhances the expression of S1P lyase, the enzyme responsible for irreversible S1P catabolism. Finally, the existence of an interplay between SphK1/S1P pathway and A2 B Rs in OPCs was confirmed since acute stimulation of A2 B Rs activates SphK1 by increasing its phosphorylation. Here the role of A2 B R and SphK/S1P signaling during oligodendrogenesis is reviewed in detail, with the purpose to shed new light on the interaction between A2 B Rs and S1P signaling, as eventual innovative targets for the treatment of demyelinating disorders.
Collapse
Affiliation(s)
- Elisabetta Coppi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Federica Cherchi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Martina Venturini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Paola Bruni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Felicita Pedata
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Anna Maria Pugliese
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| |
Collapse
|
18
|
Zhang Z, Zhou H, Zhou J. Heterogeneity and Proliferative and Differential Regulators of NG2-glia in Physiological and Pathological States. Curr Med Chem 2021; 27:6384-6406. [PMID: 31333083 DOI: 10.2174/0929867326666190717112944] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/12/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022]
Abstract
NG2-glia, also called Oligodendrocyte Precursor Cells (OPCs), account for approximately 5%-10% of the cells in the developing and adult brain and constitute the fifth major cell population in the central nervous system. NG2-glia express receptors and ion channels involved in rapid modulation of neuronal activities and signaling with neuronal synapses, which have functional significance in both physiological and pathological states. NG2-glia participate in quick signaling with peripheral neurons via direct synaptic touches in the developing and mature central nervous system. These distinctive glia perform the unique function of proliferating and differentiating into oligodendrocytes in the early developing brain, which is critical for axon myelin formation. In response to injury, NG2-glia can proliferate, migrate to the lesions, and differentiate into oligodendrocytes to form new myelin sheaths, which wrap around damaged axons and result in functional recovery. The capacity of NG2-glia to regulate their behavior and dynamics in response to neuronal activity and disease indicate their critical role in myelin preservation and remodeling in the physiological state and in repair in the pathological state. In this review, we provide a detailed summary of the characteristics of NG2-glia, including their heterogeneity, the regulators of their proliferation, and the modulators of their differentiation into oligodendrocytes.
Collapse
Affiliation(s)
- Zuo Zhang
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Hongli Zhou
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Jiyin Zhou
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
19
|
Abstract
In the twentieth century, neuropsychiatric disorders have been perceived solely from a neurone-centric point of view, which considers neurones as the key cellular elements of pathological processes. This dogma has been challenged thanks to the better comprehension of the brain functioning, which, even if far from being complete, has revealed the complexity of interactions that exist between neurones and neuroglia. Glial cells represent a highly heterogeneous population of cells of neural (astroglia and oligodendroglia) and non-neural (microglia) origin populating the central nervous system. The variety of glia reflects the innumerable functions that glial cells perform to support functions of the nervous system. Aberrant execution of glial functions contributes to the development of neuropsychiatric pathologies. Arguably, all types of glial cells are implicated in the neuropathology; however, astrocytes have received particular attention in recent years because of their pleiotropic functions that make them decisive in maintaining cerebral homeostasis. This chapter describes the multiple roles of astrocytes in the healthy central nervous system and discusses the diversity of astroglial responses in neuropsychiatric disorders suggesting that targeting astrocytes may represent an effective therapeutic strategy.
Collapse
Affiliation(s)
- Caterina Scuderi
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy.
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
| |
Collapse
|
20
|
Huerga-Gómez A, Aguado T, Sánchez-de la Torre A, Bernal-Chico A, Matute C, Mato S, Guzmán M, Galve-Roperh I, Palazuelos J. Δ 9 -Tetrahydrocannabinol promotes oligodendrocyte development and CNS myelination in vivo. Glia 2020; 69:532-545. [PMID: 32956517 PMCID: PMC7821226 DOI: 10.1002/glia.23911] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022]
Abstract
Δ9‐Tetrahydrocannabinol (THC), the main bioactive compound found in the plant Cannabis sativa, exerts its effects by activating cannabinoid receptors present in many neural cells. Cannabinoid receptors are also physiologically engaged by endogenous cannabinoid compounds, the so‐called endocannabinoids. Specifically, the endocannabinoid 2‐arachidonoylglycerol has been highlighted as an important modulator of oligodendrocyte (OL) development at embryonic stages and in animal models of demyelination. However, the potential impact of THC exposure on OL lineage progression during the critical periods of postnatal myelination has never been explored. Here, we show that acute THC administration at early postnatal ages in mice enhanced OL development and CNS myelination in the subcortical white matter by promoting oligodendrocyte precursor cell cycle exit and differentiation. Mechanistically, THC‐induced‐myelination was mediated by CB1 and CB2 cannabinoid receptors, as demonstrated by the blockade of THC actions by selective receptor antagonists. Moreover, the THC‐mediated modulation of oligodendroglial differentiation relied on the activation of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, as mTORC1 pharmacological inhibition prevented the THC effects. Our study identifies THC as an effective pharmacological strategy to enhance oligodendrogenesis and CNS myelination in vivo.
Collapse
Affiliation(s)
- Alba Huerga-Gómez
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| | - Tania Aguado
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| | - Aníbal Sánchez-de la Torre
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| | - Ana Bernal-Chico
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain.,Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Carlos Matute
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain.,Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Susana Mato
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain.,Achucarro Basque Center for Neuroscience, Leioa, Spain.,Biocruces Bizkaia, Multiple Sclerosis and Other Demyelinating Diseases Unit, Barakaldo, Spain
| | - Manuel Guzmán
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| | - Ismael Galve-Roperh
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| | - Javier Palazuelos
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain
| |
Collapse
|
21
|
iPS-Derived Early Oligodendrocyte Progenitor Cells from SPMS Patients Reveal Deficient In Vitro Cell Migration Stimulation. Cells 2020; 9:cells9081803. [PMID: 32751289 PMCID: PMC7463559 DOI: 10.3390/cells9081803] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
The most challenging aspect of secondary progressive multiple sclerosis (SPMS) is the lack of efficient regenerative response for remyelination, which is carried out by the endogenous population of adult oligoprogenitor cells (OPCs) after proper activation. OPCs must proliferate and migrate to the lesion and then differentiate into mature oligodendrocytes. To investigate the OPC cellular component in SPMS, we developed induced pluripotent stem cells (iPSCs) from SPMS-affected donors and age-matched controls (CT). We confirmed their efficient and similar OPC differentiation capacity, although we reported SPMS-OPCs were transcriptionally distinguishable from their CT counterparts. Analysis of OPC-generated conditioned media (CM) also evinced differences in protein secretion. We further confirmed SPMS-OPC CM presented a deficient capacity to stimulate OPC in vitro migration that can be compensated by exogenous addition of specific components. Our results provide an SPMS-OPC cellular model and encouraging venues to study potential cell communication deficiencies in the progressive form of multiple sclerosis (MS) for future treatment strategies.
Collapse
|
22
|
Duffy CP, McCoy CE. The Role of MicroRNAs in Repair Processes in Multiple Sclerosis. Cells 2020; 9:cells9071711. [PMID: 32708794 PMCID: PMC7408558 DOI: 10.3390/cells9071711] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disorder characterised by demyelination of central nervous system neurons with subsequent damage, cell death and disability. While mechanisms exist in the CNS to repair this damage, they are disrupted in MS and currently there are no treatments to address this deficit. In recent years, increasing attention has been paid to the influence of the small, non-coding RNA molecules, microRNAs (miRNAs), in autoimmune disorders, including MS. In this review, we examine the role of miRNAs in remyelination in the different cell types that contribute to MS. We focus on key miRNAs that have a central role in mediating the repair process, along with several more that play either secondary or inhibitory roles in one or more aspects. Finally, we consider the current state of miRNAs as therapeutic targets in MS, acknowledging current challenges and potential strategies to overcome them in developing effective novel therapeutics to enhance repair mechanisms in MS.
Collapse
|
23
|
Tyrosine Kinase Receptors Axl and MerTK Mediate the Beneficial Effect of Electroacupuncture in a Cuprizone-Induced Demyelinating Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3205176. [PMID: 32714402 PMCID: PMC7355344 DOI: 10.1155/2020/3205176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/16/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022]
Abstract
Electroacupuncture has been shown to promote remyelination in a demyelinating model of multiple sclerosis (MS) through enhanced microglial clearance of degraded myelin debris. However, the mechanisms involved in this process are yet to be clearly elucidated. It has been revealed that TAM receptor tyrosine kinases (Tyro3, Axl, and MerTK) play pivotal roles in regulating multiple features of microglia, including the phagocytic function and myelin clearance. Therefore, the aim of this study is to further confirm whether electroacupuncture improves functional recovery in this model and to characterise the involvement of the TAM receptor during this process. In addition to naive control mice, a cuprizone-induced demyelinating model was established, and long-term electroacupuncture treatment was administrated. To evaluate the efficiency of functional recovery following demyelination, we performed beam-walking test and rotarod performance test; to objectify the degree of remyelination, we performed transmission electron microscopy and protein quantification of mature oligodendrocyte markers. Oil Red O staining was used to evaluate the deposit of myelin debris. We confirmed that, in cuprizone-treated mice, electroacupuncture significantly ameliorates motor-coordinative dysfunction and counteracts demyelinating processes, with less deposit of myelin debris accumulating in the corpus callosum. Surprisingly, mRNA expression of TAM receptors was significantly upregulated after electroacupuncture treatment, and we further confirmed an increased protein expression of Axl and MerTK after electroacupuncture treatment, indicating their involvement during electroacupuncture treatment. Finally, LDC1267, a selective TAM kinase inhibitor, abolished the therapeutic effect of electroacupuncture on motor-coordinative dysfunction. Overall, our data demonstrate that electroacupuncture could mitigate the progression of demyelination by enhancing the TAM receptor expression to facilitate the clearance of myelin debris. Our results also suggest that electroacupuncture may be a potential curative treatment for MS patients.
Collapse
|
24
|
Effects of EHP-101 on inflammation and remyelination in murine models of Multiple sclerosis. Neurobiol Dis 2020; 143:104994. [PMID: 32599064 DOI: 10.1016/j.nbd.2020.104994] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/05/2020] [Accepted: 06/20/2020] [Indexed: 01/26/2023] Open
Abstract
Multiple Sclerosis (MS) is characterized by a combination of inflammatory and neurodegenerative processes in the spinal cord and the brain. Natural and synthetic cannabinoids such as VCE-004.8 have been studied in preclinical models of MS and represent promising candidates for drug development. VCE-004.8 is a multitarget synthetic cannabidiol (CBD) derivative acting as a dual Peroxisome proliferator-activated receptor-gamma/Cannabinoid receptor type 2 (PPARγ/CB2) ligand agonist that also activates the Hypoxia-inducible factor (HIF) pathway. EHP-101 is an oral lipidic formulation of VCE-004.8 that has shown efficacy in several preclinical models of autoimmune, inflammatory, fibrotic, and neurodegenerative diseases. EHP-101 alleviated clinical symptomatology in EAE and transcriptomic analysis demonstrated that EHP-101 prevented the expression of many inflammatory genes closely associated with MS pathophysiology in the spinal cord. EHP-101 normalized the expression of several genes associated with oligodendrocyte function such as Teneurin 4 (Tenm4) and Gap junction gamma-3 (Gjc3) that were downregulated in EAE. EHP-101 treatment prevented microglia activation and demyelination in both the spinal cord and the brain. Moreover, EAE was associated with a loss in the expression of Oligodendrocyte transcription factor 2 (Olig2) in the corpus callosum, a marker for oligodendrocyte differentiation, which was restored by EHP-101 treatment. In addition, EHP-101 enhanced the expression of glutathione S-transferase pi (GSTpi), a marker for mature oligodendrocytes in the brain. We also found that a diet containing 0.2% cuprizone for six weeks induced a clear loss of myelin in the brain measured by Cryomyelin staining and Myelin basic protein (MBP) expression. Moreover, EHP-101 also prevented cuprizone-induced microglial activation, astrogliosis and reduced axonal damage. Our results provide evidence that EHP-101 showed potent anti-inflammatory activity, prevented demyelination, and enhanced remyelination. Therefore, EHP-101 represents a promising drug candidate for the potential treatment of different forms of MS.
Collapse
|
25
|
Dai X, Chen J, Xu F, Zhao J, Cai W, Sun Z, Hitchens TK, Foley LM, Leak RK, Chen J, Hu X. TGFα preserves oligodendrocyte lineage cells and improves white matter integrity after cerebral ischemia. J Cereb Blood Flow Metab 2020; 40:639-655. [PMID: 30834805 PMCID: PMC7026842 DOI: 10.1177/0271678x19830791] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transforming growth factor α (TGF-α) has been reported to play important roles in neurogenesis and angiogenesis in the injured brain. The present study characterizes a novel role for TGFα in oligodendrocyte lineage cell survival and white matter integrity after ischemic stroke. Three days after transient (60 min) middle cerebral artery occlusion (tMCAO), TGFα expression was significantly increased in microglia/macrophages and neurons. Expression of the receptor of TGFα-epidermal growth factor receptor (EGFR)-was increased in glial cells after ischemia, including in oligodendrocyte lineage cells. TGFα knockout enlarged brain infarct volumes and exacerbated cell death in oligodendrocyte precursor cells (OPCs) and oligodendrocytes three days after tMCAO. TGFα-deficient mice displayed long-term exacerbation of sensorimotor deficits after tMCAO, and these functional impairments were accompanied by loss of white matter integrity and impaired oligodendrocyte replacement. In vitro studies confirmed that 5 or 10 ng/mL TGFα directly protected OPCs and oligodendrocytes against oxygen and glucose deprivation (OGD)-induced cell death, but exerted no effects on OPC differentiation. Further studies identified STAT3 as a key transcription factor mediating the effects of TGFα on OPCs and oligodendrocytes. In conclusion, TGFα provides potent oligodendrocyte protection against cerebral ischemia, thereby maintaining white matter integrity and improving neurological recovery after stroke.
Collapse
Affiliation(s)
- Xuejiao Dai
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Jie Chen
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Fei Xu
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, USA
| | - Jingyan Zhao
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Wei Cai
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Zeyu Sun
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - T Kevin Hitchens
- Animal Imaging Center, School of Medicine, University of Pittsburgh, Pittsburgh, USA.,Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Lesley M Foley
- Animal Imaging Center, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, USA
| | - Jun Chen
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, USA
| | - Xiaoming Hu
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, USA
| |
Collapse
|
26
|
Ijomone OK, Shallie PD, Naicker T. Oligodendrocytes Death Induced Sensorimotor and Cognitive Deficit in N-nitro-L-arginine methyl Rat Model of Pre-eclampsia. Neurochem Res 2020; 45:902-914. [PMID: 31983010 DOI: 10.1007/s11064-020-02969-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 12/17/2019] [Accepted: 01/20/2020] [Indexed: 12/17/2022]
Abstract
Pre-eclampsia (PE) is a pregnancy complicated syndrome that affects multiple organs including the brain that continue post- delivery in both mother and the offspring. We evaluated the expression of oligodendrocytes in the brain of PE rat model through development as well as the cognitive changes and other behavioural modifications that may occur later in the life of offspring of PE-like rat model. Pregnant rats divided into early-onset and late-onset groups were administered with N-nitro- L-arginine methyl (L-NAME) through drinking water at gestational days (GD) 8-17. Rats were allowed free access to water throughout the pregnancy. At GD 19, post-natal day (PND) 1 and 60, rats were sacrificed and brain excised for further analysis. The offspring were subjected to behavioural studies for cognitive and sensorimotor impairments before sacrificed at PND 60. Results showed significant down-regulation in the expression of OLIG2 in PE at GD 19 brain which persists till PND 60. Likewise, there was a significant increase in the latency to locate the platform in Morris water maze, time to traverse the balance beam and reduced hanging time on the wire test between the control and the PE treated. PE could lead to impaired neuronal signalling through demyelination which may contributes significantly to long-term sensorimotor and cognitive deficit.
Collapse
Affiliation(s)
- Olayemi K Ijomone
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Philemon Dauda Shallie
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
27
|
Baldassarro VA, Marchesini A, Giardino L, Calzà L. Differential effects of glucose deprivation on the survival of fetal versus adult neural stem cells-derived oligodendrocyte precursor cells. Glia 2019; 68:898-917. [PMID: 31755592 DOI: 10.1002/glia.23750] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/27/2022]
Abstract
Impaired myelination is a key feature in neonatal hypoxia/ischemia (HI), the most common perinatal/neonatal cause of death and permanent disabilities, which is triggered by the establishment of an inflammatory and hypoxic environment during the most critical period of myelin development. This process is dependent on oligodendrocyte precursor cells (OPCs) and their capability to differentiate into mature oligodendrocytes. In this study, we investigated the vulnerability of fetal and adult OPCs derived from neural stem cells (NSCs) to inflammatory and HI insults. The resulting OPCs/astrocytes cultures were exposed to cytokines to mimic inflammation, or to oxygen-glucose deprivation (OGD) to mimic an HI condition. The differentiation of both fetal and adult OPCs is completely abolished following exposure to inflammatory cytokines, while only fetal-derived OPCs degenerate when exposed to OGD. We then investigated possible mechanisms involved in OGD-mediated toxicity: (a) T3-mediated maturation induction; (b) glutamate excitotoxicity; (c) glucose metabolism. We found that while no substantial differences were observed in T3 intracellular content regulation and glutamate-mediated toxicity, glucose deprivation lead to selective OPC cell death and impaired differentiation in fetal cultures only. These results indicate that the biological response of OPCs to inflammation and demyelination is different in fetal and adult cells, and that the glucose metabolism perturbation in fetal central nervous system (CNS) may significantly contribute to neonatal pathologies. An understanding of the underlying molecular mechanism will contribute greatly to differentiating myelination enhancing and neuroprotective therapies for neonatal and adult CNS white matter lesions.
Collapse
Affiliation(s)
- Vito Antonio Baldassarro
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
| | | | - Luciana Giardino
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy.,Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Laura Calzà
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy
| |
Collapse
|
28
|
Li G, Fan ZK, Gu GF, Jia ZQ, Zhang QQ, Dai JY, He SS. Epidural Spinal Cord Stimulation Promotes Motor Functional Recovery by Enhancing Oligodendrocyte Survival and Differentiation and by Protecting Myelin after Spinal Cord Injury in Rats. Neurosci Bull 2019; 36:372-384. [PMID: 31732865 DOI: 10.1007/s12264-019-00442-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
Epidural spinal cord stimulation (ESCS) markedly improves motor and sensory function after spinal cord injury (SCI), but the underlying mechanisms are unclear. Here, we investigated whether ESCS affects oligodendrocyte differentiation and its cellular and molecular mechanisms in rats with SCI. ESCS improved hindlimb motor function at 7 days, 14 days, 21 days, and 28 days after SCI. ESCS also significantly increased the myelinated area at 28 days, and reduced the number of apoptotic cells in the spinal white matter at 7 days. SCI decreased the expression of 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNPase, an oligodendrocyte marker) at 7 days and that of myelin basic protein at 28 days. ESCS significantly upregulated these markers and increased the percentage of Sox2/CNPase/DAPI-positive cells (newly differentiated oligodendrocytes) at 7 days. Recombinant human bone morphogenetic protein 4 (rhBMP4) markedly downregulated these factors after ESCS. Furthermore, ESCS significantly decreased BMP4 and p-Smad1/5/9 expression after SCI, and rhBMP4 reduced this effect of ESCS. These findings indicate that ESCS enhances the survival and differentiation of oligodendrocytes, protects myelin, and promotes motor functional recovery by inhibiting the BMP4-Smad1/5/9 signaling pathway after SCI.
Collapse
Affiliation(s)
- Gang Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Spinal Pain Research Institute, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhong-Kai Fan
- Department of Orthopaedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121001, China
| | - Guang-Fei Gu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Spinal Pain Research Institute, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhi-Qiang Jia
- Department of Spinal Surgery, The Second Affiliated Hospital, Henan University of Science and Technology, Luoyang, 471003, China
| | - Qiang-Qiang Zhang
- Department of Orthopaedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121001, China
| | - Jun-Yu Dai
- Department of Orthopaedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121001, China
| | - Shi-Sheng He
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
- Spinal Pain Research Institute, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
29
|
Bankston AN, Forston MD, Howard RM, Andres KR, Smith AE, Ohri SS, Bates ML, Bunge MB, Whittemore SR. Autophagy is essential for oligodendrocyte differentiation, survival, and proper myelination. Glia 2019; 67:1745-1759. [PMID: 31162728 DOI: 10.1002/glia.23646] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/01/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022]
Abstract
Deficient myelination, the spiral wrapping of highly specialized membrane around axons, causes severe neurological disorders. Maturation of oligodendrocyte progenitor cells (OPC) to myelinating oligodendrocytes (OL), the sole providers of central nervous system (CNS) myelin, is tightly regulated and involves extensive morphological changes. Here, we present evidence that autophagy, the targeted isolation of cytoplasm and organelles by the double-membrane autophagosome for lysosomal degradation, is essential for OPC/OL differentiation, survival, and proper myelin development. A marked increase in autophagic activity coincides with OL differentiation, with OL processes having the greatest increase in autophagic flux. Multiple lines of evidence indicate that autophagosomes form in developing myelin sheathes before trafficking from myelin to the OL soma. Mice with conditional OPC/OL-specific deletion of the essential autophagy gene Atg5 beginning on postnatal Day 5 develop a rapid tremor and die around postnatal Day 12. Further analysis revealed apoptotic death of OPCs, reduced differentiation, and reduced myelination. Surviving Atg5-/- OLs failed to produce proper myelin structure. In vitro, pharmacological inhibition of autophagy in OPC/dorsal root ganglion (DRG) co-cultures blocked myelination, producing OLs surrounded by many short processes. Conversely, autophagy stimulation enhanced myelination. These results implicate autophagy as a key regulator of OPC survival, maturation, and proper myelination. Autophagy may provide an attractive target to promote both OL survival and subsequent myelin repair after injury.
Collapse
Affiliation(s)
- Andrew N Bankston
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Michael D Forston
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Russell M Howard
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Allison E Smith
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Margaret L Bates
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Mary B Bunge
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida.,Department of Cell Biology and Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
30
|
Mecha M, Yanguas-Casás N, Feliú A, Mestre L, Carrillo-Salinas F, Azcoitia I, Yong VW, Guaza C. The endocannabinoid 2-AG enhances spontaneous remyelination by targeting microglia. Brain Behav Immun 2019; 77:110-126. [PMID: 30582962 DOI: 10.1016/j.bbi.2018.12.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/29/2022] Open
Abstract
Remyelination is an endogenous process by which functional recovery of damaged neurons is achieved by reinstating the myelin sheath around axons. Remyelination has been documented in multiple sclerosis (MS) lesions and experimental models, although it is often incomplete or fails to affect the integrity of the axon, thereby leading to progressive disability. Microglia play a crucial role in the clearance of the myelin debris produced by demyelination and in inflammation-dependent OPC activation, two processes necessary for remyelination to occur. We show here that following corpus callosum demyelination in the TMEV-IDD viral murine model of MS, there is spontaneous and partial remyelination that involves a temporal discordance between OPC mobilization and microglia activation. Pharmacological treatment with the endocannabinoid 2-AG enhances the clearance of myelin debris by microglia and OPC differentiation, resulting in complete remyelination and a thickening of the myelin sheath. These results highlight the importance of targeting microglia during the repair processes in order to enhance remyelination.
Collapse
Affiliation(s)
- M Mecha
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Spain.
| | - N Yanguas-Casás
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Spain; CIBER de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - A Feliú
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Spain
| | - L Mestre
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Spain
| | - F Carrillo-Salinas
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Spain
| | - I Azcoitia
- CIBER de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Biología Celular, Facultad de Biología, Universidad Complutense, Madrid 28040, Spain
| | - V W Yong
- Hotchkiss Brain Institute, and the Departments of Clinical Neurosciences and Oncology, University of Calgary, 3330 Hospital Drive, Calgary, Alberta T2N 4N1, Canada
| | - C Guaza
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Spain.
| |
Collapse
|
31
|
MicroRNA-219 Inhibits Proliferation and Induces Differentiation of Oligodendrocyte Precursor Cells after Contusion Spinal Cord Injury in Rats. Neural Plast 2019; 2019:9610687. [PMID: 30911293 PMCID: PMC6398016 DOI: 10.1155/2019/9610687] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/09/2018] [Accepted: 11/19/2018] [Indexed: 01/02/2023] Open
Abstract
MicroRNA-219 (miR-219) regulates the proliferation and differentiation of oligodendrocyte precursor cells (OPCs) during central nervous system (CNS) development. OPCs only differentiate into oligodendrocytes (OLs) in the healthy CNS, but can generate astrocytes (As) after injury. We hypothesized that miR-219 may modulate OPC proliferation and differentiation in a cervical C5 contusion spinal cord injury (SCI) model. After injury, we observed a decrease in the miR-219 level and quantity of OLs and an increase in the number of OPCs and As. Silencing of miR-219 by its antagomir in vivo produced similar results, but of greater magnitude. Overexpression of miR-219 by its agomir in vivo increased the number of OLs and suppressed generation of OPCs and As. Luxol fast blue staining confirmed that SCI caused demyelination and that the extent of demyelination was attenuated by miR-219 overexpression, but aggravated by miR-219 reduction. Monocarboxylate transporter 1 (MCT-1) may be implicated in the regulation of OPC proliferation and differentiation mediated by miR-219 following contusion SCI. Collectively, our data suggest that miR-219 may mediate SCI-induced OPC proliferation and differentiation, and MCT-1 may participate in this process as a target of miR-219.
Collapse
|
32
|
Abstract
Oligodendrocytes are the myelinating cells of the CNS, producing the insulating myelin sheath that facilitates rapid electrical conduction of axonal action potentials. Oligodendrocytes arise from oligodendrocyte progenitor cells (OPCs) under the control of multiple factors, including neurotransmitters and other neuron-derived factors. A significant population of OPCs persists in the adult CNS, where they are often referred to as NG2-glia, because they are identified by their expression of the NG2 chondroitin sulphate proteoglycan (CSPG4). In the adult brain, the primary function of NG2-glia is the life-long generation of oligodendrocytes to replace myelin lost through natural 'wear and tear' and pathology, as well as to provide new oligodendrocytes to myelinate new connections formed in response to new life experiences. NG2-glia contact synapses and respond to neurotransmitters and potassium released during neuronal transmission; to this end, NG2-glia (OPCs) express multiple neurotransmitter receptors and ion channels, with prominent roles being identified for glutamatergic signalling and potassium channels in oligodendrocyte differentiation. Myelinating oligodendrocytes also express a wide range of neurotransmitter receptors and ion channels, together with transporters and gap junctions; together, these have critical functions in cellular ion and water homeostasis, as well as metabolism, which is essential for maintaining myelin and axon integrity. An overriding theme is that oligodendrocyte function and myelination is not only essential for rapid axonal conduction, but is essential for learning and the long-term integrity of axons and neurones. Hence, myelination underpins cognitive function and the massive computing power of the human brain and myelin loss has devastating effects on CNS function. This chapter focuses on normal oligodendrocyte physiology.
Collapse
|
33
|
Promising neuroprotective effects of β-caryophyllene against LPS-induced oligodendrocyte toxicity: A mechanistic study. Biochem Pharmacol 2018; 159:154-171. [PMID: 30529211 DOI: 10.1016/j.bcp.2018.12.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 12/04/2018] [Indexed: 01/09/2023]
Abstract
Myelin loss subsequent to oligodendrocyte death has been reported in a variety of myelin-associated disorders such as multiple sclerosis (MS). Lipopolysaccharide (LPS) has been shown to elicit cellular responses in the central nervous system (CNS) and trigger immune infiltrates and glial cells to release a variety of inflammatory cytokines and mediators. LPS-induced oligodendrocytes toxicity may be chosen as an efficient model to evaluate the role of oligodendrocytes in neuroprotective activities of compounds. β-Caryophyllene (BCP) is a selective type 2 cannabinoid (CB2) receptor agonist. However, the mechanisms underlying the anti-inflammatory effects of BCP are not completely understood. On this basis, we aimed to investigate the protective effects of a wide range of BCP concentrations against LPS-induced toxicity in a proliferative oligodendrocyte cell line (OLN-93) and evaluate the possible correlation between BCP concentration and selective modulation of CB2, Nrf2, sphingomyelinase (SMase) and peroxisome proliferator-activated receptors (PPAR)-γ signaling pathways. We found that LPS significantly increases the levels of reactive oxygen species (ROS), nitric oxide (NO) metabolite and tumor necrosis factor (TNF)-α production while decreases the level of GSH. BCP could prevent LPS-induced cytotoxicity and excessive production of NO, ROS, and TNF-α. Also, we demonstrated that BCP's protective effects against LPS-induced oligodendrocytes toxicity were mediated via the CB2 receptor through different pathways including Nrf2/HO-1/anti-oxidant axis, and PPAR-γ, at low (0.2 and 1 µM), and high (10-50 µM) concentrations, respectively. Additionally, we observed that the addition of SMase inhibitors imipramine (IMP) and fluoxetine (FLX) synergistically increased the protective effects of BCP. Finally, BCP at low concentrations exerted promising protective effects that could be considered for the treatment of neurodegenerative disorders such as MS. However, more studies using other models of neurodegenerative diseases should be undertaken to assess different parameters such as the activity or expression of SMase.
Collapse
|
34
|
Del Giovane A, Ragnini-Wilson A. Targeting Smoothened as a New Frontier in the Functional Recovery of Central Nervous System Demyelinating Pathologies. Int J Mol Sci 2018; 19:E3677. [PMID: 30463396 PMCID: PMC6274747 DOI: 10.3390/ijms19113677] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022] Open
Abstract
Myelin sheaths on vertebrate axons provide protection, vital support and increase the speed of neuronal signals. Myelin degeneration can be caused by viral, autoimmune or genetic diseases. Remyelination is a natural process that restores the myelin sheath and, consequently, neuronal function after a demyelination event, preventing neurodegeneration and thereby neuron functional loss. Pharmacological approaches to remyelination represent a promising new frontier in the therapy of human demyelination pathologies and might provide novel tools to improve adaptive myelination in aged individuals. Recent phenotypical screens have identified agonists of the atypical G protein-coupled receptor Smoothened and inhibitors of the glioma-associated oncogene 1 as being amongst the most potent stimulators of oligodendrocyte precursor cell (OPC) differentiation in vitro and remyelination in the central nervous system (CNS) of mice. Here, we discuss the current state-of-the-art of studies on the role of Sonic Hedgehog reactivation during remyelination, referring readers to other reviews for the role of Hedgehog signaling in cancer and stem cell maintenance.
Collapse
Affiliation(s)
- Alice Del Giovane
- Department of Biology University of Rome Tor Vergata, Viale Della Ricerca Scientifica, 00133 Rome, Italy.
| | - Antonella Ragnini-Wilson
- Department of Biology University of Rome Tor Vergata, Viale Della Ricerca Scientifica, 00133 Rome, Italy.
| |
Collapse
|
35
|
Rajaei T, Farajifard H, Rezaee SA, Azarpazhooh MR, Mahmoudi M, Valizadeh N, Rafatpanah H. Different roles of CXCR1 and CXCR2 in HTLV-1 carriers and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) patients. Med Microbiol Immunol 2018; 208:641-650. [PMID: 30341468 DOI: 10.1007/s00430-018-0568-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 10/09/2018] [Indexed: 12/24/2022]
Abstract
One of the prominent features of HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is the excessive recruitment of leukocytes to the central nervous system (CNS), which leads to an inflammatory response-with chemokines and their receptors playing the main role in this recruitment. The aim of the study was to examine the relation of CXCR1 and CXCR2, both of which are involved in the trafficking of lymphocytes into the CNS, with the outcome of HTLV-1 infection. The mRNA levels of CXCR1 and CXCR2 were examined in peripheral blood mononuclear cells (PBMCs) of HAM/TSP patients, HTLV-1 asymptomatic carriers (ACs), and healthy controls (HCs). Furthermore, the frequency of CD4+ and CD8+ T cells expressing CXCR1 and CXCR2 was evaluated in the studied groups. The results of the present study showed a substantial increase in the mean mRNA expression of CXCR2 in the HAM/TSP patients compared to the HCs and ACs (p < 0.001). A positive correlation was also found between PVL and CXCR2 mRNA expression in the total population of HTLV-1-infected subjects (R = 0.526, p < 0.001). Moreover, the percentage of CD8+ CXCR2-expressing cells was higher in HAM/TSP patients compared to ACs and HCs (p < 0.05, p < 0.01, respectively). Although the percentage of CD4+ CXCR2-expressing cells was higher in HAM/TSP patients than in ACs and HCs, a significant difference was only found between HAM/TSP patients and HCs (p < 0.05). No significant difference in the CXCR1 mRNA expression was observed in the studied groups. The frequency of the CD8+ CXCR1- and CD4+ CXCR1-expressing cells was significantly lower in HAM/TSP patients than in ACs and HCs (p < 0.001 and p < 0.01, respectively). In conclusion, the high frequency of CXCR2 CD8+ T cells and the high levels of CXCR2 mRNA expression in HAM/TSP patients are associated with disease pathogenesis, while the high frequencies of CXCR1 T cells in ACs might suggest that these cells act as effector CD8 T cells and are involved in controlling the viral spread and modulation of the immune response.
Collapse
Affiliation(s)
- Taraneh Rajaei
- Immunology Research Center, Division of Inflammation and Inflammatory Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Farajifard
- Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Abdolrahim Rezaee
- Immunology Research Center, Division of Inflammation and Inflammatory Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Valizadeh
- Immunology Research Center, Division of Inflammation and Inflammatory Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Center, Division of Inflammation and Inflammatory Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
36
|
Saeb S, Azari H, Mostafavi-Pour Z, Ghanbari A, Ebrahimi S, Mokarram P. 9-cis-Retinoic Acid and 1,25-dihydroxy Vitamin D3 Improve the Differentiation of Neural Stem Cells into Oligodendrocytes through the Inhibition of the Notch and Wnt Signaling Pathways. IRANIAN JOURNAL OF MEDICAL SCIENCES 2018; 43:523-532. [PMID: 30214105 PMCID: PMC6123560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Differentiating oligodendrocyte precursor cells (OPCs) into oligodendrocytes could be improved by inhibiting signaling pathways such as Wnt and Notch. 9-cis-retinoic acid (9-cis-RA) and 1,25-dihydroxyvitamin D3 (1,25[OH]2D3) can ameliorate oligodendrogenesis. We investigated whether they could increase oligodendrogenesis by inhibiting the Wnt and Notch signaling pathways. METHODS Cortical neural stem cells were isolated from 14-day-old rat embryos and cultured using the neurosphere assay. The cells were treated in 4 different conditions for 1 week: the negative control group received only the basic fibroblast growth factor, the positive control group received only T3 without growth factors, the RA group was treated with 9-cis-RA, and the Vit D3 group was treated with 1,25(OH)2D3. The effects of 9-cis-RA and 1,25(OH)2D3 on the level of the myelin basic protein (MBP) and the gene expression of the SOX10, MBP gene, HES5, and LRP6 were studied using flow cytometry and real-time PCR. The data were analyzed using one-way ANOVA with GraphPad Prism. A P value less than 0.05 was considered significant. RESULTS The mRNA expressions of the SOX10, MBP, and MBP gene were significantly increased in the treated groups compared with the negative control group; the increase was similar in the 9-cis-RA group and the positive control group. Furthermore, 9-cis-RA significantly decreased the expression of the HES5 gene, a Notch signaling pathway transcription factor, and 1,25(OH)2D3 significantly reduced the expression of the LRP6 gene, a Wnt signaling pathway co-receptor. CONCLUSION It seems that 9-cis-RA and 1,25(OH)2D3 are good candidates to improve the differentiation of OPCs into oligodendrocytes.
Collapse
Affiliation(s)
- Saeedeh Saeb
- Department of Biochemistry, Shiraz University of Medical Sciences, Shiraz, Iran;
| | - Hassan Azari
- Neural Stem Cell and Regenerative Neuroscience Laboratory, Department of Anatomical Sciences and Shiraz Stem Cell Institute, Shiraz University of Medical Sciences, Shiraz, Iran;
| | | | - Amir Ghanbari
- Department of Anatomical Sciences, Yasuj University of Medical sciences, Yasuj, Iran
| | - Sepideh Ebrahimi
- Department of Biochemistry, Shiraz University of Medical Sciences, Shiraz, Iran;
| | - Pooneh Mokarram
- Department of Biochemistry, Shiraz University of Medical Sciences, Shiraz, Iran;
| |
Collapse
|
37
|
Santos AK, Vieira MS, Vasconcellos R, Goulart VAM, Kihara AH, Resende RR. Decoding cell signalling and regulation of oligodendrocyte differentiation. Semin Cell Dev Biol 2018; 95:54-73. [PMID: 29782926 DOI: 10.1016/j.semcdb.2018.05.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022]
Abstract
Oligodendrocytes are fundamental for the functioning of the nervous system; they participate in several cellular processes, including axonal myelination and metabolic maintenance for astrocytes and neurons. In the mammalian nervous system, they are produced through waves of proliferation and differentiation, which occur during embryogenesis. However, oligodendrocytes and their precursors continue to be generated during adulthood from specific niches of stem cells that were not recruited during development. Deficiencies in the formation and maturation of these cells can generate pathologies mainly related to myelination. Understanding the mechanisms involved in oligodendrocyte development, from the precursor to mature cell level, will allow inferring therapies and treatments for associated pathologies and disorders. Such mechanisms include cell signalling pathways that involve many growth factors, small metabolic molecules, non-coding RNAs, and transcription factors, as well as specific elements of the extracellular matrix, which act in a coordinated temporal and spatial manner according to a given stimulus. Deciphering those aspects will allow researchers to replicate them in vitro in a controlled environment and thus mimic oligodendrocyte maturation to understand the role of oligodendrocytes in myelination in pathologies and normal conditions. In this study, we review these aspects, based on the most recent in vivo and in vitro data on oligodendrocyte generation and differentiation.
Collapse
Affiliation(s)
- A K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - M S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - R Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - V A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - A H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - R R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil.
| |
Collapse
|
38
|
Marella M, Ouyang J, Zombeck J, Zhao C, Huang L, Connor RJ, Phan KB, Jorge MC, Printz MA, Paladini RD, Gelb AB, Huang Z, Frost GI, Sugarman BJ, Steinman L, Wei G, Shepard HM, Maneval DC, Lapinskas PJ. PH20 is not expressed in murine CNS and oligodendrocyte precursor cells. Ann Clin Transl Neurol 2017; 4:191-211. [PMID: 28275653 PMCID: PMC5338182 DOI: 10.1002/acn3.393] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 12/08/2016] [Accepted: 01/10/2017] [Indexed: 12/22/2022] Open
Abstract
Objective Expression of Spam1/PH20 and its modulation of high/low molecular weight hyaluronan substrate have been proposed to play an important role in murine oligodendrocyte precursor cell (OPC) maturation in vitro and in normal and demyelinated central nervous system (CNS). We reexamined this using highly purified PH20. Methods Steady‐state expression of mRNA in OPCs was evaluated by quantitative polymerase chain reaction; the role of PH20 in bovine testicular hyaluronidase (BTH) inhibition of OPC differentiation was explored by comparing BTH to a purified recombinant human PH20 (rHuPH20). Contaminants in commercial BTH were identified and their impact on OPC differentiation characterized. Spam1/PH20 expression in normal and demyelinated mouse CNS tissue was investigated using deep RNA sequencing and immunohistological methods with two antibodies directed against recombinant murine PH20. Results BTH, but not rHuPH20, inhibited OPC differentiation in vitro. Basic fibroblast growth factor (bFGF) was identified as a significant contaminant in BTH, and bFGF immunodepletion reversed the inhibitory effects of BTH on OPC differentiation. Spam1 mRNA was undetected in OPCs in vitro and in vivo; PH20 immunolabeling was undetected in normal and demyelinated CNS. Interpretation We were unable to detect Spam1/PH20 expression in OPCs or in normal or demyelinated CNS using the most sensitive methods currently available. Further, “BTH” effects on OPC differentiation are not due to PH20, but may be attributable to contaminating bFGF. Our data suggest that caution be exercised when using some commercially available hyaluronidases, and reports of Spam1/PH20 morphogenic activity in the CNS may be due to contaminants in reagents.
Collapse
Affiliation(s)
| | - Joe Ouyang
- Halozyme Therapeutics, Inc. San Diego California
| | | | - Chunmei Zhao
- Halozyme Therapeutics, Inc. San Diego California
| | - Lei Huang
- Halozyme Therapeutics, Inc. San Diego California
| | | | - Kim B Phan
- Halozyme Therapeutics, Inc. San Diego California
| | | | | | | | | | | | | | | | - Lawrence Steinman
- University School of Medicine Department of Neurology and Neurological Sciences Beckman Center for Molecular Medicine Stanford University Stanford California
| | - Ge Wei
- Halozyme Therapeutics, Inc. San Diego California
| | | | | | | |
Collapse
|
39
|
Zhang J, Zhang ZG, Lu M, Wang X, Shang X, Elias SB, Chopp M. MiR-146a promotes remyelination in a cuprizone model of demyelinating injury. Neuroscience 2017; 348:252-263. [PMID: 28237816 DOI: 10.1016/j.neuroscience.2017.02.029] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 12/30/2022]
Abstract
The death of mature oligodendrocytes (OLs) which are the sole myelinating cells of the central nervous system (CNS), leads to demyelination and functional deficits. Currently, there is lack of effective remyelination therapies for patients with demyelinating diseases. MicroRNAs (miRNAs) mediate OL function. We hypothesized that miR-146a, by inactivating interleukin-1 receptor-associated kinase 1 (IRAK1), promotes differentiation of oligodendrocyte progenitor cells (OPCs) and thereby enhances remyelination. To test this hypothesis, a demyelination model induced by a cuprizone (CPZ) diet was employed, in which C57BL/6J mice were fed with a CPZ diet for 5weeks. After termination of CPZ diet, the mice were randomly treated with continuous infusion of miR-146a mimics or mimic controls into the corpus callosum for 7days. Compared to the mimic control, infusion of miR-146a mimics facilitated remyelination assessed by increased myelin basic proteins in the corpus callosum, which was associated with augmentation of newly generated mature OLs. Infusion of miR-146a mimics also substantially elevated miR-146a levels in the corpus callosum and fluorescently tagged miR-146a mimics were mainly detected in OPCs. Western blot and double immmunofluorescent staining analysis showed that the miR-146a treatment considerably reduced IRAK1 protein levels and the number of IRAK1-positive cells, respectively. Collectively, these data indicate that exogenous miR-146a enhances remyelination, possibly by promoting OPCs to differentiate into myelinated OLs via targeting IRAK1.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, United States.
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, United States
| | - Mei Lu
- Biostatistics and Research Epidemiology, Henry Ford Health System, Detroit, MI 48202, United States
| | - Xinli Wang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, United States
| | - Xia Shang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, United States
| | - Stanton B Elias
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, United States; Department of Physics, Oakland University, Rochester, MI 48309, United States
| |
Collapse
|
40
|
Li X, Zhang Y, Yan Y, Ciric B, Ma CG, Chin J, Curtis M, Rostami A, Zhang GX. LINGO-1-Fc-Transduced Neural Stem Cells Are Effective Therapy for Chronic Stage Experimental Autoimmune Encephalomyelitis. Mol Neurobiol 2016; 54:4365-4378. [PMID: 27344330 DOI: 10.1007/s12035-016-9994-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/14/2016] [Indexed: 12/11/2022]
Abstract
The chronic stage multiple sclerosis (MS), an inflammatory demyelinating disease of the central nervous system (CNS), remains refractory to current treatments. This refractory nature may be due to the fact that current treatments are primarily immunomodulatory, which prevent further demyelination but lack the capacity to promote remyelination. Several approaches, including transplantation of neural stem cells (NSCs) or antagonists to LINGO-1, a key part of the receptor complex for neuroregeneration inhibitors, have been effective in suppressing the acute stage of experimental autoimmune encephalomyelitis (EAE), an animal model of MS. However, their effect on the chronic stage EAE is not known. Here, we show that transplantation of NSCs had only a slight therapeutic effect when treatment started at the chronic stage of EAE (e.g., injected at day 40 postimmunization). However, NSCs engineered to produce LINGO-1-Fc, a soluble LINGO-1 antagonist, significantly promoted neurological recovery as demonstrated by amelioration of clinical signs, improvement in axonal integrity, and enhancement of oligodendrocyte maturation and neuron repopulation. Significantly enhanced NAD production and Sirt2 expression were also found in the CNS of mice treated with LINGO-1-Fc-producing NSC. Moreover, differentiation of LINGO-1-Fc-producing NSCs into oligodendrocytes in vitro was largely diminished by an NAMPT inhibitor, indicating that LINGO-1-Fc enhances the NAMPT/NAD/Sirt2 pathway. Together, our study establishes a CNS-targeted, novel LINGO-1-Fc delivery system using NSCs, which represents a novel and effective NSC-based gene therapy approach for the chronic stage of MS.
Collapse
Affiliation(s)
- Xing Li
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.,College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuan Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.,College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yaping Yan
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.,College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Cun-Gen Ma
- Institute of Brain Science, Department of Neurology, Shanxi Datong University Medical School, Datong, China
| | - Jeannie Chin
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Mark Curtis
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
41
|
Goldstein EZ, Church JS, Hesp ZC, Popovich PG, McTigue DM. A silver lining of neuroinflammation: Beneficial effects on myelination. Exp Neurol 2016; 283:550-9. [PMID: 27151600 DOI: 10.1016/j.expneurol.2016.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/27/2016] [Accepted: 05/01/2016] [Indexed: 12/19/2022]
Abstract
Myelin accelerates action potential conduction velocity and provides essential energy support for axons. Unfortunately, myelin and myelinating cells are often vulnerable to injury or disease, resulting in myelin damage, which in turn can lead to axon dysfunction, overt pathology and neurological impairment. Inflammation is a common component of trauma and disease in both the CNS and PNS and therefore an active inflammatory response is often considered deleterious to myelin health. While inflammation can certainly damage myelin, inflammatory processes also can positively affect oligodendrocyte lineage progression, myelin debris clearance, oligodendrocyte metabolism and myelin repair. In the periphery, inflammatory cascades can also augment myelin repair, including processes initiated by infiltrating immune cells as well as by local Schwann cells. In this review, various aspects of inflammation beneficial to myelin repair are discussed and should be considered when designing or implementing anti-inflammatory therapies for CNS and PNS injury involving myelinating cells.
Collapse
Affiliation(s)
- Evan Z Goldstein
- Neuroscience Graduate Program, Wexner Medical Center, The Ohio State University, United States; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, United States
| | - Jamie S Church
- Neuroscience Graduate Program, Wexner Medical Center, The Ohio State University, United States; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, United States
| | - Zoe C Hesp
- Neuroscience Graduate Program, Wexner Medical Center, The Ohio State University, United States; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, United States
| | - Phillip G Popovich
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, United States; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, United States
| | - Dana M McTigue
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, United States; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, United States.
| |
Collapse
|
42
|
Zhang J, Zhang ZG, Li Y, Lu M, Zhang Y, Elias SB, Chopp M. Thymosin beta4 promotes oligodendrogenesis in the demyelinating central nervous system. Neurobiol Dis 2016; 88:85-95. [DOI: 10.1016/j.nbd.2016.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/19/2015] [Accepted: 01/09/2016] [Indexed: 02/01/2023] Open
|
43
|
Zilkha-Falb R, Kaushansky N, Kawakami N, Ben-Nun A. Post-CNS-inflammation expression of CXCL12 promotes the endogenous myelin/neuronal repair capacity following spontaneous recovery from multiple sclerosis-like disease. J Neuroinflammation 2016; 13:7. [PMID: 26747276 PMCID: PMC4706716 DOI: 10.1186/s12974-015-0468-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/26/2015] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Demyelination and axonal degeneration, hallmarks of multiple sclerosis (MS), are associated with the central nervous system (CNS) inflammation facilitated by C-X-C motif chemokine 12 (CXCL12) chemokine. Both in MS and in experimental autoimmune encephalomyelitis (EAE), the deleterious CNS inflammation has been associated with upregulation of CXCL12 expression in the CNS. We investigated the expression dynamics of CXCL12 in the CNS with progression of clinical EAE and following spontaneous recovery, with a focus on CXCL12 expression in the hippocampal neurogenic dentate gyrus (DG) and in the corpus callosum (CC) of spontaneously recovered mice, and its potential role in promoting the endogenous myelin/neuronal repair capacity. METHODS CNS tissue sections from mice with different clinical EAE phases or following spontaneous recovery and in vitro differentiated adult neural stem cell cultures were analyzed by immunofluorescent staining and confocal imaging for detecting and enumerating neuronal progenitor cells (NPCs) and oligodendrocyte precursor cells (OPCs) and for expression of CXCL12. RESULTS Our expression dynamics analysis of CXCL12 in the CNS with EAE progression revealed elevated CXCL12 expression in the DG and CC, which persistently increases following spontaneous recovery even though CNS inflammation has subsided. Correspondingly, the numbers of NPCs and OPCs in the DG and CC, respectively, of EAE-recovered mice increased compared to that of naïve mice (NPCs, p < 0.0001; OPCs, p < 0.00001) or mice with active disease (OPCs, p < 0.0005). Notably, about 30 % of the NPCs and unexpectedly also OPCs (~50 %) express CXCL12, and their numbers in DG and CC, respectively, are higher in EAE-recovered mice compared with naïve mice and also compared with mice with ongoing clinical EAE (CXCL12(+) NPCs, p < 0.005; CXCL12(+) OPCs, p < 0.0005). Moreover, a significant proportion (>20 %) of the CXCL12(+) NPCs and OPCs co-express the CXCL12 receptor, CXCR4, and their numbers significantly increase with recovery from EAE not only relative to naïve mice (p < 0.0002) but also to mice with ongoing EAE (p < 0.004). CONCLUSIONS These data link CXCL12 expression in the DG and CC of EAE-recovering mice to the promotion of neuro/oligodendrogenesis generating CXCR4(+) CXCL12(+) neuronal and oligodendrocyte progenitor cells endowed with intrinsic neuro/oligondendroglial differentiation potential. These findings highlight the post-CNS-inflammation role of CXCL12 in augmenting the endogenous myelin/neuronal repair capacity in MS-like disease, likely via CXCL12/CXCR4 autocrine signaling.
Collapse
Affiliation(s)
- Rina Zilkha-Falb
- Present address: Multiple Sclerosis Center, Neurogenomics Laboratory, Sheba Medical Center, Tel-Hashomer, Israel.
| | - Nathali Kaushansky
- Department of Immunology, The Weizmann Institute of Science, 234 Herzl Street, Rehovot, 7610001, Israel
| | - Naoto Kawakami
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians-University, 81377, Munich, Germany.
| | - Avraham Ben-Nun
- Department of Immunology, The Weizmann Institute of Science, 234 Herzl Street, Rehovot, 7610001, Israel.
| |
Collapse
|
44
|
Berry M, Ahmed Z, Morgan-Warren P, Fulton D, Logan A. Prospects for mTOR-mediated functional repair after central nervous system trauma. Neurobiol Dis 2015; 85:99-110. [PMID: 26459109 DOI: 10.1016/j.nbd.2015.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/09/2015] [Accepted: 10/08/2015] [Indexed: 02/07/2023] Open
Abstract
Recent research has suggested that the growth of central nervous system (CNS) axons during development is mediated through the PI3K/Akt/mammalian target of rapamycin (mTOR) intracellular signalling axis and that suppression of activity in this pathway occurs during maturity as levels of the phosphatase and tensin homologue (PTEN) rise and inhibit PI3K activation of mTOR, accounting for the failure of axon regeneration in the injured adult CNS. This hypothesis is supported by findings confirming that suppression of PTEN in experimental adult animals promotes impressive axon regeneration in the injured visual and corticospinal motor systems. This review focuses on these recent developments, discussing the therapeutic potential of a mTOR-based treatment aimed at promoting functional recovery in CNS trauma patients, recognising that to fulfil this ambition, the new therapy should aim to promote not only axon regeneration but also remyelination of regenerated axons, neuronal survival and re-innervation of denervated targets through accurate axonal guidance and synaptogenesis, all with minimal adverse effects. The translational challenges presented by the implementation of this new axogenic therapy are also discussed.
Collapse
Affiliation(s)
- Martin Berry
- Neurotrauma Research Group, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Zubair Ahmed
- Neurotrauma Research Group, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Peter Morgan-Warren
- Neurotrauma Research Group, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Daniel Fulton
- Neurotrauma Research Group, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Ann Logan
- Neurotrauma Research Group, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
45
|
Improved differentiation of oligodendrocyte precursor cells and neurological function after spinal cord injury in rats by oscillating field stimulation. Neuroscience 2015; 303:346-51. [PMID: 26166729 DOI: 10.1016/j.neuroscience.2015.07.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 07/03/2015] [Accepted: 07/04/2015] [Indexed: 12/29/2022]
Abstract
Oscillating field stimulation (OFS) has been used in attempts to treat spinal cord injury (SCI) and has been shown to improve remyelination after SCI in rats. However, some controversies regarding the effects of OFS have been presented in previous papers. Oligodendrocytes (OLs) are the main cell for remyelination and are derived from the differentiation of oligodendrocyte precursor cells (OPCs). To date, it has been unclear whether the differentiation of OPCs can be regulated by OFS. The goal of this study was to determine if OFS can improve the differentiation of OPCs and promote the recovery of neurological function after SCI in rats. Immature and mature OLs were observed in spinal cord slices through immunofluorescence staining. Levels of adenosine triphosphate (ATP) and the cytokine leukemia inhibitory factor (LIF) were detected by enzyme-linked immunosorbent assay (ELISA). Basso-Beattie-Bresnahan (BBB) scores and transcranial magnetic motor-evoked potentials (tcMMEPs) were used to evaluate the locomotor outcomes of rats after SCI. Our results showed a significant improvement in the differentiation of OPCs and the content of ATP and LIF in the injured spinal cord in the OFS group. Furthermore, BBB scores and tcMMEPs were significantly improved in the rats stimulated by OFS. These findings suggest that OFS can improve the differentiation of OPCs and promote the recovery of neurological function following SCI in rats.
Collapse
|
46
|
Scalabrino G, Veber D, De Giuseppe R, Roncaroli F. Low levels of cobalamin, epidermal growth factor, and normal prions in multiple sclerosis spinal cord. Neuroscience 2015; 298:293-301. [PMID: 25888933 DOI: 10.1016/j.neuroscience.2015.04.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 04/08/2015] [Accepted: 04/10/2015] [Indexed: 11/16/2022]
Abstract
We have previously demonstrated that multiple sclerosis (MS) patients have abnormal cerebrospinal fluid (CSF) levels of the key myelin-related molecules cobalamin (Cbl), epidermal growth factor (EGF), and normal cellular prions (PrP(C)s), thus confirming that some CSF abnormalities may be co-responsible for remyelination failure. We determined the levels of these three molecules in post-mortem spinal cord (SC) samples taken from MS patients and control patients. The control SC samples, almost all of which came from non-neurological patients, did not show any microscopic lesions of any type. All of the samples were supplied by the U.K. MS Tissue Bank. The Cbl, EGF, and PrP(C) levels were determined using enzyme-linked immunosorbent assays. The SC total homocysteine level was determined using a competitive immunoenzymatic assay. CSF samples, taken from a further group of MS patients, were used for the assay of holo-transcobalamin (holo-TC) levels. The Cbl, EGF, and PrP(C) levels were significantly decreased in MS SCs in comparison with controls and, paradoxically, the decreased Cbl levels were associated with decreased SC levels of homocysteine, a biochemical marker of Cbl deficiency. The trends of EGF and PrP(C) levels paralleled those previously found in CSF, whereas that of Cbl was the opposite. There was no significant difference in CSF holo-TC levels between the MS patients and the controls. Given that we have previously demonstrated that Cbl positively regulates central nervous system EGF levels, it is conceivable that the low EGF levels in the MS SC may be causally related to a local decrease in Cbl levels. Only PrP(C) levels were invariably decreased in both the SC and CSF regardless of the clinical course of the disease. These findings suggest that the simultaneous lack of Cbl, EGF, and PrP(C)s may greatly hamper the remyelination process in MS patients, because they are key molecules of the machinery for remyelination.
Collapse
Affiliation(s)
- G Scalabrino
- Department of Biomedical Sciences, Laboratory of Neuropathology, University of Milan, 20133 Milan, Italy.
| | - D Veber
- Department of Biomedical Sciences, Laboratory of Neuropathology, University of Milan, 20133 Milan, Italy
| | - R De Giuseppe
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - F Roncaroli
- Division of Brain Sciences, Imperial College, London W12 0NN, UK
| |
Collapse
|
47
|
Oligodendrocyte birth and death following traumatic brain injury in adult mice. PLoS One 2015; 10:e0121541. [PMID: 25798924 PMCID: PMC4370677 DOI: 10.1371/journal.pone.0121541] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 02/03/2015] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes are responsible for producing and maintaining myelin throughout the CNS. One of the pathological features observed following traumatic brain injury (TBI) is the progressive demyelination and degeneration of axons within white matter tracts. While the effect of TBI on axonal health has been well documented, there is limited information regarding the response of oligodendrocytes within these areas. The aim of this study was to characterize the response of both mature oligodendrocytes and immature proliferative oligodendrocyte lineage cells across a 3 month timecourse following TBI. A computer-controlled cortical impact model was used to produce a focal lesion in the left motor cortex of adult mice. Immunohistochemical analyses were performed at 48 hours, 7 days, 2 weeks, 5 weeks and 3 months following injury to assess the prevalence of mature CC-1+ oligodendrocyte cell death, immature Olig2+ cell proliferation and longer term survival in the corpus callosum and external capsule. Decreased CC-1 immunoreactivity was observed in white matter adjacent to the site of injury from 2 days to 2 weeks post TBI, with ongoing mature oligodendrocyte apoptosis after this time. Conversely, proliferation of Olig2+ cells was observed as early as 48 hours post TBI and significant numbers of these cells and their progeny survived and remained in the external capsule within the injured hemisphere until at least 3 months post injury. These findings demonstrate that immature oligodendrocyte lineage cells respond to TBI by replacing oligodendrocytes lost due to damage and that this process occurs for months after injury.
Collapse
|
48
|
Ghaffari S, Hatami H, Dehghan G. Saffron ethanolic extract attenuates oxidative stress, spatial learning, and memory impairments induced by local injection of ethidium bromide. Res Pharm Sci 2015; 10:222-32. [PMID: 26600849 PMCID: PMC4621629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Cognitive deficits have been observed in patients with multiple sclerosis (MS) because of hippocampal insults. Oxidative stress plays a key role in the pathophysiology of MS. The aim of this study was to evaluate the effects of Crocus sativus L., commonly known as saffron, on learning and memory loss and the induction of oxidative stress in the hippocampus of toxic models of MS. One week after MS induction by intrahippocampal injection of ethidium bromide (EB), animals were treated with two doses of saffron extract (5 and 10 μg/rat) for a week. Learning and spatial memory status was assessed using Morris Water Maze. After termination of behavioral testing days, animals were decapitated and the bilateral hippocampi dissected to measure some of the oxidative stress markers including the level of hippocampi thiobarbituric acid reactive substances and the activity of antioxidant enzymes such as glutathione peroxidase and superoxide dismutase. Treatment with saffron extract ameliorated spatial learning and memory impairment (P<0.05). Total antioxidant reactivity capacity, lipid peroxidation products and antioxidant enzymes activity in the hippocampus homogenates of EB treated group were significantly higher than those of all other groups (P<0.01). Indeed, treatment with a saffron extract for 7 consecutive days significantly restored the antioxidant status to the normal levels (P<0.01). These observations reveal that saffron extract can ameliorate the impairment of learning and memory as well as the disturbances in oxidative stress parameters in the hippocampus of experimental models of MS.
Collapse
Affiliation(s)
- Sh. Ghaffari
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, I.R. Iran
| | - H. Hatami
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, I.R. Iran,Corresponding author: H. Hatami Tel: 0098 413 3392740, Fax: 0098 413 3356027
| | - Gh. Dehghan
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, I.R. Iran
| |
Collapse
|
49
|
Staszewski O, Prinz M. Glial epigenetics in neuroinflammation and neurodegeneration. Cell Tissue Res 2014; 356:609-16. [PMID: 24652504 DOI: 10.1007/s00441-014-1815-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 01/14/2014] [Indexed: 01/01/2023]
Abstract
Epigenetic regulation shapes the differentiation and response to stimuli of all tissues and cells beyond what genetics would dictate. Epigenetic regulation acts through covalent modifications of DNA and histones while leaving the nucleotide code intact. However, these chromatin modifications are known to be vital components of the regulation of cell fate and response. With regards to the central nervous system (CNS), little is known about how epigenetic regulation shapes the function of neural cell types. The focus of research so far has been on epigenetic regulation of neuronal function and the role of epigenetics in tumorigenesis. However, the glial cell compartment, which makes up 90 % of all CNS cells, has so far received scant attention as to how epigenetics shape their differentiation and function. Here, we highlight current knowledge about epigenetic changes in glial cells occurring during CNS injury, neuroinflammatory conditions and neurodegenerative disease. This review offers an overview of the current understanding of epigenetic regulation in glial cells in CNS disease.
Collapse
Affiliation(s)
- Ori Staszewski
- Institute of Neuropathology, University of Freiburg, Breisacher Str. 64, D-79106, Freiburg, Germany
| | | |
Collapse
|
50
|
May VEL, Ettle B, Poehler AM, Nuber S, Ubhi K, Rockenstein E, Winner B, Wegner M, Masliah E, Winkler J. α-Synuclein impairs oligodendrocyte progenitor maturation in multiple system atrophy. Neurobiol Aging 2014; 35:2357-68. [PMID: 24698767 DOI: 10.1016/j.neurobiolaging.2014.02.028] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 01/21/2014] [Accepted: 02/05/2014] [Indexed: 11/30/2022]
Abstract
Multiple system atrophy (MSA), an atypical parkinsonian disorder, is characterized by α-synuclein (α-syn(+)) cytoplasmatic inclusions in mature oligodendrocytes. Oligodendrocyte progenitor cells (OPCs) represent a distinct cell population with the potential to replace dysfunctional oligodendrocytes. However, the role of OPCs in MSA and their potential to replace mature oligodendrocytes is still unclear. A postmortem analysis in MSA patients revealed α-syn within OPCs and an increased number of striatal OPCs. In an MSA mouse model, an age-dependent increase of dividing OPCs within the striatum and the cortex was detected. Despite of myelin loss, there was no reduction of mature oligodendrocytes in the corpus callosum or the striatum. Dissecting the underlying molecular mechanisms an oligodendroglial cell line expressing human α-syn revealed that α-syn delays OPC maturation by severely downregulating myelin-gene regulatory factor and myelin basic protein. Brain-derived neurotrophic factor was reduced in MSA models and its in vitro supplementation partially restored the phenotype. Taken together, efficacious induction of OPC maturation may open the window to restore glial and neuronal function in MSA.
Collapse
Affiliation(s)
- Verena E L May
- Department of Molecular Neurology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Benjamin Ettle
- Department of Molecular Neurology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Anne-Maria Poehler
- Department of Molecular Neurology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Silke Nuber
- Department of Neurosciences and Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kiren Ubhi
- Department of Neurosciences and Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Edward Rockenstein
- Department of Neurosciences and Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Beate Winner
- Junior Research Group III, Interdisciplinary Centre of Clinical Research, Nikolaus Fiebiger Centre for Molecular Medicine, University Hospital Erlangen, Erlangen, Germany
| | - Michael Wegner
- Institute of Biochemistry, Emil-Fischer-Zentrum, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Eliezer Masliah
- Department of Neurosciences and Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jürgen Winkler
- Department of Molecular Neurology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany; Department of Neurosciences and Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|