1
|
Chen K, Park E, Abd-Elrahman KS. Enhancing remyelination in multiple sclerosis via M1 muscarinic acetylcholine receptor. Mol Pharmacol 2025; 107:100027. [PMID: 40158341 PMCID: PMC12060155 DOI: 10.1016/j.molpha.2025.100027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Multiple sclerosis (MS) is growing in prevalence; yet, treatments that can reverse the progression of the disease are still needed. One strategy that has shown promise for reversing MS is remyelination by inhibiting the M1 receptor, a member of the muscarinic acetylcholine receptor (mAChR) family. Antagonizing the M1 mAChR is believed to be the mechanism by which clemastine, a developing drug that has been observed to enhance myelination in animal studies and phase II clinical trials, elicits its myelination-promoting effects. Recent studies have indicated that blocking M1 mAChR may promote oligodendrocyte differentiation via the extracellular signal-regulated kinase pathway, modulating Ca2+ concentration oscillations, and cross-talking with N-methyl-d-aspartate and Notch-1 receptors. However, clemastine has recently been found to accelerate disability in patients with MS, discouraging further progress in its clinical trials. Nevertheless, the underlying mechanisms following M1 mAChR antagonism by clemastine may still be targeted using alternative antimuscarinic drugs. This review consolidates recent advancements in our understanding of the mechanisms by which antagonizing M1 mAChR promotes remyelination and summarizes alternative antimuscarinic drugs that could be leveraged to treat MS in the future. SIGNIFICANCE STATEMENT: Current treatments for multiple sclerosis are limited to disease management, and there is a need for restorative treatments that can reverse progressive forms of the disease. This review aims to summarize the potential mechanisms by which antagonizing the M1 muscarinic acetylcholine receptor could promote remyelination and elaborate on a collection of promising antimuscarinic drugs, consolidating the knowledge needed to target these mechanisms and develop therapeutics that could reverse the progress of demyelinating diseases like multiple sclerosis.
Collapse
Affiliation(s)
- Keren Chen
- Department of Anaesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Eunyoung Park
- Department of Anaesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Khaled S Abd-Elrahman
- Department of Anaesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, British Columbia, Canada; Department of Medical Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
2
|
Tian R, Zhou Y, Ren Y, Zhang Y, Tang W. Wallerian degeneration: From mechanism to disease to imaging. Heliyon 2025; 11:e40729. [PMID: 39811315 PMCID: PMC11730939 DOI: 10.1016/j.heliyon.2024.e40729] [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: 05/13/2024] [Revised: 08/12/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Wallerian degeneration (WD) was first discovered by Augustus Waller in 1850 in a transection of the glossopharyngeal and hypoglossal nerves in frogs. Initial studies suggested that the formation mechanism of WD is related to the nutrition of neuronal cell bodies to axons. However, with the wide application of transgenic mice in experiments, the latest studies have found that the mechanism of WD is related to axonal degeneration, myelin clearance and extracellular matrix. This review summarizes the discovery and research progress of WD and discusses the mechanism of WD from the perspective of molecular biology. In addition, this review combines the etiology, symptoms and imaging results of WD patients, and analyzes the clinical and imaging characteristics of WD, to provide the best perspective for future clinical research.
Collapse
Affiliation(s)
- Ruiqi Tian
- Department of Neurology, Xinhua Hospital Affiliated with Dalian University, Dalian, Liaoning Province, China
| | - Yingying Zhou
- Department of Neurology, Xinhua Hospital Affiliated with Dalian University, Dalian, Liaoning Province, China
| | - Yuan Ren
- Department of Neurology, Xinhua Hospital Affiliated with Dalian University, Dalian, Liaoning Province, China
| | - Yisen Zhang
- Department of Neurology, Xinhua Hospital Affiliated with Dalian University, Dalian, Liaoning Province, China
| | - Wei Tang
- Department of Neurology, Xinhua Hospital Affiliated with Dalian University, Dalian, Liaoning Province, China
| |
Collapse
|
3
|
Manesh SB, Kondiles BR, Wheeler S, Liu J, Zhang L, Chernoff C, Duncan GJ, Ramer MS, Tetzlaff W. Compensatory changes after spinal cord injury in a remyelination deficient mouse model. J Neurochem 2025; 169:e16220. [PMID: 39268880 DOI: 10.1111/jnc.16220] [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: 03/31/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024]
Abstract
The development of therapeutic strategies to reduce impairments following spinal cord injury (SCI) motivates an active area of research, because there are no effective therapies. One strategy is to address injury-induced demyelination of spared axons by promoting endogenous or exogenous remyelination. However, previously, we showed that new myelin was not necessary to regain hindlimb stepping following moderate thoracic spinal cord contusion in 3-month-old mice. The present analysis investigated two potential mechanisms by which animals can re-establish locomotion in the absence of remyelination: compensation through intact white matter and conduction through spared axons. We induced a severe contusion injury to reduce the spared white matter rim in the remyelination deficient model, with no differences in recovery between remyelination deficient animals and injured littermate controls. We investigated the nodal properties of the axons at the lesion and found that in the remyelination deficient model, axons express the Nav1.2 voltage-gated sodium channel, a sub-type not typically expressed at mature nodes of Ranvier. In a moderate contusion injury, conduction velocities through the lesions of remyelination deficient animals were similar to those in animals with the capacity to remyelinate after injury. Detailed gait analysis and kinematics reveal subtle differences between remyelination deficient animals and remyelination competent controls, but no worse deficits. It is possible that upregulation of Nav1.2 channels may contribute to establishing conduction through the lesion. This conduction could contribute to compensation and regained motor function in mouse models of SCI. Such compensatory mechanism may have implications for interpreting efficacy results for remyelinating interventions in mice and the development of therapies for improving recovery following SCI.
Collapse
Affiliation(s)
- S B Manesh
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - B R Kondiles
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - S Wheeler
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - J Liu
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - L Zhang
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - C Chernoff
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - G J Duncan
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - M S Ramer
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - W Tetzlaff
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
4
|
Kondiles B, Murphy R, Widman A, Perlmutter S, Horner P. Cortical stimulation leads to shortened myelin sheaths and increased axonal branching in spared axons after cervical spinal cord injury. Glia 2023; 71:1947-1959. [PMID: 37096399 PMCID: PMC10649492 DOI: 10.1002/glia.24376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 03/28/2023] [Accepted: 04/02/2023] [Indexed: 04/26/2023]
Abstract
Neural activity and learning lead to myelin sheath plasticity in the intact central nervous system (CNS), but this plasticity has not been well-studied after CNS injury. In the context of spinal cord injury (SCI), demyelination occurs at the lesion site and natural remyelination of surviving axons can take months. To determine if neural activity modulates myelin and axon plasticity in the injured, adult CNS, we electrically stimulated the contralesional motor cortex at 10 Hz to drive neural activity in the corticospinal tract of rats with sub-chronic spinal contusion injuries. We quantified myelin and axonal characteristics by tracing corticospinal axons rostral to and at the lesion epicenter and identifying nodes of Ranvier by immunohistochemistry. Three weeks of daily stimulation induced very short myelin sheaths, axon branching, and thinner axons outside of the lesion zone, where remodeling has not previously been reported. Surprisingly, remodeling was particularly robust rostral to the injury which suggests that electrical stimulation can promote white matter plasticity even in areas not directly demyelinated by the contusion. Stimulation did not alter myelin or axons at the lesion site, which suggests that neuronal activity does not contribute to myelin remodeling near the injury in the sub-chronic period. These data are the first to demonstrate wide-scale remodeling of nodal and myelin structures of a mature, long-tract motor pathway in response to electrical stimulation. This finding suggests that neuromodulation promotes white matter plasticity in intact regions of pathways after injury and raises intriguing questions regarding the interplay between axonal and myelin plasticity.
Collapse
Affiliation(s)
- B.R. Kondiles
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific St. Seattle, WA, 98105, USA
- Center for Neuroregeneration, Dept. of Neurosurgery, Houston Methodist Research Institute, 6670 Bertner, Houston, TX, 77030, USA
| | - R.L. Murphy
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific St. Seattle, WA, 98105, USA
| | - A.J. Widman
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific St. Seattle, WA, 98105, USA
| | - S.I. Perlmutter
- Department of Physiology and Biophysics, University of Washington, 1705 NE Pacific St. Seattle, WA, 98105, USA
| | - P.J. Horner
- Center for Neuroregeneration, Dept. of Neurosurgery, Houston Methodist Research Institute, 6670 Bertner, Houston, TX, 77030, USA
| |
Collapse
|
5
|
Sidoryk-Węgrzynowicz M, Dąbrowska-Bouta B, Sulkowski G, Strużyńska L. Nanosystems and exosomes as future approaches in treating multiple sclerosis. Eur J Neurosci 2021; 54:7377-7404. [PMID: 34561918 DOI: 10.1111/ejn.15478] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system which leads to neurological dysfunctions and severe disabilities. MS pathology is characterised by damage of the blood-brain barrier and infiltration of autoreactive T cells that overactivate glial cells, thereby initiating neuroinflammation accompanied by the formation of demyelinating plaques and neurodegeneration. Clinical deficits in this multifactorial disease depend on the progression of myelin loss, the stage of inflammation, the status of axons and the activity of oligodendrocyte precursor cells (OPCs). Despite significant progress in the treatment of MS, current therapies remain limited and new approaches are highly desirable. Nanosystems based on liposomes and nanoparticles are among some of the more noteworthy therapeutic strategies being investigated. Applications of nanosystems alone or as drug carriers in animal models of MS have been found to successfully alleviate the symptoms of the disease and exert anti-inflammatory potential. Exosomes are a specific type of nanosystem based on nanometre-sized extracellular vesicles released by different cells which exhibit important healing features. Exosomes contain an array of anti-inflammatory and neuroprotective agents which may contribute to modulation of the immune system as well as promoting remyelination and tissue repair. In this review, opportunities to use nanosystems against progression of MS will be discussed in context of cell-specific pathologies associated with MS.
Collapse
Affiliation(s)
- Marta Sidoryk-Węgrzynowicz
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Dąbrowska-Bouta
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Grzegorz Sulkowski
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Lidia Strużyńska
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
6
|
Zhang J, Wang H, Fan Y, Yang F. Effect of mesenchymal stem cells transplantation on the changes of oligodendrocyte lineage in rat brain with experimental autoimmune encephalomyelitis. Brain Behav 2021; 11:e01999. [PMID: 33319488 PMCID: PMC7882188 DOI: 10.1002/brb3.1999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/29/2020] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE To explore the effect of bone marrow mesenchymal stem cells (BM-MSCs) transplantation on the changes of oligodendrocyte lineage in brain of experimental autoimmune encephalomyelitis (EAE) rats. METHODS The animals were divided into normal control group, EAE model group (EAE group), cell culture medium injection treatment group (placebo treatment group), and MSCs treatment group (treatment group). The changes of A2B5-, O4-, and CNPase-positive cells in oligodendrocyte lineage in rat brain were observed after 1, 3, 7, 14, 21, and 28 days. RESULTS The number of A2B5-positive cells in rat brain of the treatment group at each time point was significantly more than that of the EAE and placebo treatment groups, and most obvious at 14 days. The O4-positive cells number at each time point in the treatment group was significantly increased compared with the EAE and placebo treatment groups, and most obvious at 14 days. The CNPase-positive cells number at each time point in the treatment group was significantly increased compared with the EAE and placebo treatment groups, and most obvious at 14 days. CONCLUSIONS MSCs treatment can increase cells expression in oligodendrocyte lineage, which laying a solid foundation for myelin regeneration.
Collapse
Affiliation(s)
- Jun‐Mei Zhang
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| | - Hua Wang
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| | - Yu‐Ying Fan
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| | - Feng‐Hua Yang
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
7
|
Huntemer-Silveira A, Patil N, Brickner MA, Parr AM. Strategies for Oligodendrocyte and Myelin Repair in Traumatic CNS Injury. Front Cell Neurosci 2021; 14:619707. [PMID: 33505250 PMCID: PMC7829188 DOI: 10.3389/fncel.2020.619707] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/07/2020] [Indexed: 12/18/2022] Open
Abstract
A major consequence of traumatic brain and spinal cord injury is the loss of the myelin sheath, a cholesterol-rich layer of insulation that wraps around axons of the nervous system. In the central nervous system (CNS), myelin is produced and maintained by oligodendrocytes. Damage to the CNS may result in oligodendrocyte cell death and subsequent loss of myelin, which can have serious consequences for functional recovery. Demyelination impairs neuronal function by decelerating signal transmission along the axon and has been implicated in many neurodegenerative diseases. After a traumatic injury, mechanisms of endogenous remyelination in the CNS are limited and often fail, for reasons that remain poorly understood. One area of research focuses on enhancing this endogenous response. Existing techniques include the use of small molecules, RNA interference (RNAi), and monoclonal antibodies that target specific signaling components of myelination for recovery. Cell-based replacement strategies geared towards replenishing oligodendrocytes and their progenitors have been utilized by several groups in the last decade as well. In this review article, we discuss the effects of traumatic injury on oligodendrocytes in the CNS, the lack of endogenous remyelination, translational studies in rodent models promoting remyelination, and finally human clinical studies on remyelination in the CNS after injury.
Collapse
Affiliation(s)
| | - Nandadevi Patil
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | - Megan A. Brickner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Ann M. Parr
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
8
|
Kieser TJ, Santschi N, Nowack L, Axer A, Kehr G, Albrecht S, Gilmour R. Total Chemical Syntheses of the GM 3 and F-GM 3 Ganglioside Epitopes and Comparative Pre-Clinical Evaluation for Non-Invasive Imaging of Oligodendrocyte Differentiation. ACS Chem Neurosci 2020; 11:2129-2136. [PMID: 32559361 DOI: 10.1021/acschemneuro.0c00319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gangliosides are intimately involved in a plenum of (neuro)inflammatory processes, yet progress in establishing structure-function interplay is frequently hindered by the availability of well-defined glycostructures. Motivated by the ubiquity of the ganglioside GM3 in chemical neurology, and in particular by its conspicuous presence in myelin, the GM3 epitope was examined with a view to preclinical validation as a tracer. The suitability of this scaffold for the noninvasive imaging of oligodendrocyte differentiation in Multiple sclerosis is disclosed. The stereocontrolled synthesis of a site-selectively fluorinated analogue (F-GM3) is also disclosed to enable a comparative analysis in oligodendrocyte (OL) differentiation. Whereas the native epitope caused a decrease in the viability in a dose-dependent manner, the addition of distinct F-GM3 concentrations over 48 h had no impact on the OL viability. This is likely a consequence of the enhanced hydrolytic stability imparted by the fluorination and highlights the potential of fluorinated glycostructures in the field of molecular imaging. Given the predominant expression of GM3 in oligodendrocytes and the capacity of GM3 to interact with myelin-associated proteins, this preclinical evaluation has revealed F-GM3 to be an intriguing candidate for neurological imaging.
Collapse
Affiliation(s)
- Tobias J. Kieser
- Institute for Organic Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstrasse 40, Münster 48149, Germany
| | - Nico Santschi
- Institute for Organic Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstrasse 40, Münster 48149, Germany
| | - Luise Nowack
- Institute for Neuropathology, University Hospital Münster, Pottkamp 2, Münster 48149, Germany
| | - Alexander Axer
- Institute for Organic Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstrasse 40, Münster 48149, Germany
| | - Gerald Kehr
- Institute for Organic Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstrasse 40, Münster 48149, Germany
| | - Stefanie Albrecht
- Institute for Neuropathology, University Hospital Münster, Pottkamp 2, Münster 48149, Germany
| | - Ryan Gilmour
- Institute for Organic Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstrasse 40, Münster 48149, Germany
| |
Collapse
|
9
|
Deems NP, Leuner B. Pregnancy, postpartum and parity: Resilience and vulnerability in brain health and disease. Front Neuroendocrinol 2020; 57:100820. [PMID: 31987814 PMCID: PMC7225072 DOI: 10.1016/j.yfrne.2020.100820] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/25/2019] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Risk and resilience in brain health and disease can be influenced by a variety of factors. While there is a growing appreciation to consider sex as one of these factors, far less attention has been paid to sex-specific variables that may differentially impact females such as pregnancy and reproductive history. In this review, we focus on nervous system disorders which show a female bias and for which there is data from basic research and clinical studies pointing to modification in disease risk and progression during pregnancy, postpartum and/or as a result of parity: multiple sclerosis (MS), depression, stroke, and Alzheimer's disease (AD). In doing so, we join others (Shors, 2016; Galea et al., 2018a) in aiming to illustrate the importance of looking beyond sex in neuroscience research.
Collapse
Affiliation(s)
- Nicholas P Deems
- The Ohio State University, Department of Psychology, Columbus, OH, USA
| | - Benedetta Leuner
- The Ohio State University, Department of Psychology, Columbus, OH, USA.
| |
Collapse
|
10
|
Cuascut FX, Hutton GJ. Stem Cell-Based Therapies for Multiple Sclerosis: Current Perspectives. Biomedicines 2019; 7:biomedicines7020026. [PMID: 30935074 PMCID: PMC6631931 DOI: 10.3390/biomedicines7020026] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory and neurodegenerative autoimmune disease of the central nervous system (CNS). Disease-modifying therapies (DMT) targeting inflammation have been shown to reduce disease activity in patients with relapsing–remitting MS (RRMS). The current therapeutic challenge is to find an effective treatment to halt disease progression and reverse established neural damage. Stem cell-based therapies have emerged to address this dilemma. Several types of stem cells have been considered for clinical use, such as autologous hematopoietic (aHSC), mesenchymal (MSC), neuronal (NSC), human embryonic (hESC), and induced pluripotent (iPSC) stem cells. There is convincing evidence that immunoablation followed by hematopoietic therapy (aHSCT) has a high efficacy for suppressing inflammatory MS activity and improving neurological disability in patients with RRMS. In addition, MSC therapy may be a safe and tolerable treatment, but its clinical value is still under evaluation. Various studies have shown early promising results with other cellular therapies for CNS repair and decreasing inflammation. In this review, we discuss the current knowledge and limitations of different stem cell-based therapies for the treatment of patients with MS.
Collapse
Affiliation(s)
- Fernando X Cuascut
- Baylor College of Medicine, Maxine Mesigner Multiple Sclerosis Center, Houston, TX 77030, USA.
| | - George J Hutton
- Baylor College of Medicine, Maxine Mesigner Multiple Sclerosis Center, Houston, TX 77030, USA.
| |
Collapse
|
11
|
Yeung MSY, Djelloul M, Steiner E, Bernard S, Salehpour M, Possnert G, Brundin L, Frisén J. Dynamics of oligodendrocyte generation in multiple sclerosis. Nature 2019; 566:538-542. [PMID: 30675058 PMCID: PMC6420067 DOI: 10.1038/s41586-018-0842-3] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 10/31/2018] [Indexed: 01/09/2023]
Abstract
Oligodendrocytes wrap nerve fibres in the central nervous system with layers of specialized cell membrane to form myelin sheaths1. Myelin is destroyed by the immune system in multiple sclerosis, but myelin is thought to regenerate and neurological function can be recovered. In animal models of demyelinating disease, myelin is regenerated by newly generated oligodendrocytes, and remaining mature oligodendrocytes do not seem to contribute to this process2-4. Given the major differences in the dynamics of oligodendrocyte generation and adaptive myelination between rodents and humans5-9, it is not clear how well experimental animal models reflect the situation in multiple sclerosis. Here, by measuring the integration of 14C derived from nuclear testing in genomic DNA10, we assess the dynamics of oligodendrocyte generation in patients with multiple sclerosis. The generation of new oligodendrocytes was increased several-fold in normal-appearing white matter in a subset of individuals with very aggressive multiple sclerosis, but not in most subjects with the disease, demonstrating an inherent potential to substantially increase oligodendrocyte generation that fails in most patients. Oligodendrocytes in shadow plaques-thinly myelinated lesions that are thought to represent remyelinated areas-were old in patients with multiple sclerosis. The absence of new oligodendrocytes in shadow plaques suggests that remyelination of lesions occurs transiently or not at all, or that myelin is regenerated by pre-existing, and not new, oligodendrocytes in multiple sclerosis. We report unexpected oligodendrocyte generation dynamics in multiple sclerosis, and this should guide the use of current, and the development of new, therapies.
Collapse
Affiliation(s)
- Maggie S Y Yeung
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mehdi Djelloul
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Embla Steiner
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Samuel Bernard
- Institut Camille Jordan, CNRS UMR 5208, University of Lyon, Villeurbanne, France
| | - Mehran Salehpour
- Department of Physics and Astronomy, Ion Physics, Uppsala University, Uppsala, Sweden
| | - Göran Possnert
- Department of Physics and Astronomy, Ion Physics, Uppsala University, Uppsala, Sweden
| | - Lou Brundin
- Department of Clinical Neuroscience, Division of Neurology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
12
|
Castoldi V, Marenna S, Santangelo R, d'Isa R, Cursi M, Chaabane L, Quattrini A, Comi G, Leocani L. Optic nerve involvement in experimental autoimmune encephalomyelitis to homologous spinal cord homogenate immunization in the dark agouti rat. J Neuroimmunol 2018; 325:1-9. [PMID: 30340030 DOI: 10.1016/j.jneuroim.2018.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 12/22/2022]
Abstract
Dark-Agouti rats were immunized with spinal cord homogenate to develop Experimental Autoimmune Encephalomyelitis, a model of multiple sclerosis. We assessed motor signs and recorded VEPs for five or eight weeks with epidural or epidermal electrodes, respectively, with final histopathology of optic nerves (ONs). Injected rats exhibited motor deficits a week after immunization. VEP delays arose from the 2nd to the 5th week, when a recovery occurred in epidermal-recorded rats. ON damage appeared in epidural-, but not in epidermal-recorded rats, probably due to a remyelination process. VEP could be exploited as neurophysiological marker to test novel treatments against neurodegeneration involving ONs.
Collapse
Affiliation(s)
- Valerio Castoldi
- San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Silvia Marenna
- San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | | | - Raffaele d'Isa
- San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Marco Cursi
- San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Linda Chaabane
- San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Angelo Quattrini
- San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Giancarlo Comi
- San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Letizia Leocani
- San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy.
| |
Collapse
|
13
|
Abstract
Endogenous remyelination of the CNS can be robust and restore function, yet in multiple sclerosis it becomes less complete with time. Promoting remyelination is a major therapeutic goal, both to restore function and to protect axons from degeneration. Remyelination is thought to depend on oligodendrocyte progenitor cells, giving rise to nascent remyelinating oligodendrocytes. Surviving, mature oligodendrocytes are largely regarded as being uninvolved. We have examined this question using two large animal models. In the first model, there is extensive demyelination and remyelination of the CNS, yet oligodendrocytes survive, and in recovered animals there is a mix of remyelinated axons interspersed between mature, thick myelin sheaths. Using 2D and 3D light and electron microscopy, we show that many oligodendrocytes are connected to mature and remyelinated myelin sheaths, which we conclude are cells that have reextended processes to contact demyelinated axons while maintaining mature myelin internodes. In the second model in vitamin B12-deficient nonhuman primates, we demonstrate that surviving mature oligodendrocytes extend processes and ensheath demyelinated axons. These data indicate that mature oligodendrocytes can participate in remyelination.
Collapse
|
14
|
Pegoretti V, Baron W, Laman JD, Eisel ULM. Selective Modulation of TNF-TNFRs Signaling: Insights for Multiple Sclerosis Treatment. Front Immunol 2018; 9:925. [PMID: 29760711 PMCID: PMC5936749 DOI: 10.3389/fimmu.2018.00925] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/13/2018] [Indexed: 12/26/2022] Open
Abstract
Autoimmunity develops when self-tolerance mechanisms are failing to protect healthy tissue. A sustained reaction to self is generated, which includes the generation of effector cells and molecules that destroy tissues. A way to restore this intrinsic tolerance is through immune modulation that aims at refurbishing this immunologically naïve or unresponsive state, thereby decreasing the aberrant immune reaction taking place. One major cytokine has been shown to play a pivotal role in several autoimmune diseases such as rheumatoid arthritis (RA) and multiple sclerosis (MS): tumor necrosis factor alpha (TNFα) modulates the induction and maintenance of an inflammatory process and it comes in two variants, soluble TNF (solTNF) and transmembrane bound TNF (tmTNF). tmTNF signals via TNFR1 and TNFR2, whereas solTNF signals mainly via TNFR1. TNFR1 is widely expressed and promotes mainly inflammation and apoptosis. Conversely, TNFR2 is restricted mainly to immune and endothelial cells and it is known to activate the pro-survival PI3K-Akt/PKB signaling pathway and to sustain regulatory T cells function. Anti-TNFα therapies are successfully used to treat diseases such as RA, colitis, and psoriasis. However, clinical studies with a non-selective inhibitor of TNFα in MS patients had to be halted due to exacerbation of clinical symptoms. One possible explanation for this failure is the non-selectivity of the treatment, which avoids TNFR2 stimulation and its immune and tissue protective properties. Thus, a receptor-selective modulation of TNFα signal pathways provides a novel therapeutic concept that might lead to new insights in MS pathology with major implications for its effective treatment.
Collapse
Affiliation(s)
- Valentina Pegoretti
- Department of Molecular Neurobiology (GELIFES), University of Groningen, Groningen, Netherlands
| | - Wia Baron
- Department of Cell Biology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology (GELIFES), University of Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
G-Protein-Coupled Receptor Gpr17 Regulates Oligodendrocyte Differentiation in Response to Lysolecithin-Induced Demyelination. Sci Rep 2018. [PMID: 29540737 PMCID: PMC5852120 DOI: 10.1038/s41598-018-22452-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Oligodendrocytes are the myelin-producing cells of the central nervous system (CNS). A variety of brain disorders from “classical” demyelinating diseases, such as multiple sclerosis, stroke, schizophrenia, depression, Down syndrome and autism, are shown myelination defects. Oligodendrocyte myelination is regulated by a complex interplay of intrinsic, epigenetic and extrinsic factors. Gpr17 (G protein-coupled receptor 17) is a G protein-coupled receptor, and has been identified to be a regulator for oligodendrocyte development. Here, we demonstrate that the absence of Gpr17 enhances remyelination in vivo with a toxin-induced model whereby focal demyelinated lesions are generated in spinal cord white matter of adult mice by localized injection of LPC(L-a-lysophosphatidylcholine). The increased expression of the activated form of Erk1/2 (phospho-Erk1/2) in lesion areas suggested the potential role of Erk1/2 activity on the Gpr17-dependent modulation of myelination. The absence of Gpr17 enhances remyelination is correlate with the activated Erk1/2 (phospho-Erk1/2).Being a membrane receptor, Gpr17 represents an ideal druggable target to be exploited for innovative regenerative approaches to acute and chronic CNS diseases.
Collapse
|
16
|
Flygt J, Clausen F, Marklund N. Diffuse traumatic brain injury in the mouse induces a transient proliferation of oligodendrocyte progenitor cells in injured white matter tracts. Restor Neurol Neurosci 2018; 35:251-263. [PMID: 27768001 DOI: 10.3233/rnn-160675] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Injury to the white matter may lead to impaired neuronal signaling and is commonly observed following traumatic brain injury (TBI). Although endogenous repair of TBI-induced white matter pathology is limited, oligodendrocyte progenitor cells (OPCs) may be stimulated to proliferate and regenerate functionally myelinating oligodendrocytes. Even though OPCs are present throughout the adult brain, little is known about their proliferative activity following axonal injury caused by TBI. OBJECTIVE We hypothesized that central fluid percussion injury (cFPI) in mice, a TBI model causing wide-spread axonal injury, results in OPC proliferation. METHODS Proliferation of OPCs was evaluated in 27 cFPI mice using 5-ethynyl-2'-deoxyuridine (EdU) labeling and a cell proliferation assay at 2 (n = 9), 7 (n = 8) and 21 (n = 10) days post injury (dpi). Sham-injured mice (n = 14) were used as controls. OPC proliferation was quantified by immunohistochemistry using the OPC markers NG2 and Olig2 in several white matter loci including the corpus callosum, external capsule, fimbriae, the internal capsule and cerebral peduncle. RESULTS The number of EdU/DAPI/Olig2-positive cells were increased in the cFPI group compared to sham-injured animals at 7 days post-injury (dpi; p≤0.05) in the majority of white matter regions. The OPC proliferation had subsided by 21 dpi. The number of EdU/DAPI/NG2 cells was also increase at 7 dpi in the external capsule and fimbriae. CONCLUSION These results suggest that traumatic axonal injury in the mouse induces a transient proliferative response of residing OPCs. These proliferating OPCs may replace dead oligodendrocytes and contribute to remyelination, which needs evaluation in future studies.
Collapse
|
17
|
Kondiles BR, Horner PJ. Myelin plasticity, neural activity, and traumatic neural injury. Dev Neurobiol 2017; 78:108-122. [PMID: 28925069 DOI: 10.1002/dneu.22540] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/01/2017] [Accepted: 09/14/2017] [Indexed: 12/12/2022]
Abstract
The possibility that adult organisms exhibit myelin plasticity has recently become a topic of great interest. Many researchers are exploring the role of myelin growth and adaptation in daily functions such as memory and motor learning. Here we consider evidence for three different potential categories of myelin plasticity: the myelination of previously bare axons, remodeling of existing sheaths, and the removal of a sheath with replacement by a new internode. We also review evidence that points to the importance of neural activity as a mechanism by which oligodendrocyte precursor cells (OPCs) are cued to differentiate into myelinating oligodendrocytes, which may potentially be an important component of myelin plasticity. Finally, we discuss demyelination in the context of traumatic neural injury and present an argument for altering neural activity as a potential therapeutic target for remyelination following injury. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 108-122, 2018.
Collapse
Affiliation(s)
- Bethany R Kondiles
- Center for Neuroregeneration, Houston Methodist Research Institute, 6670 Bertner Avenue, MSR10-112, Houston, Texas.,Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Philip J Horner
- Center for Neuroregeneration, Houston Methodist Research Institute, 6670 Bertner Avenue, MSR10-112, Houston, Texas
| |
Collapse
|
18
|
Mullin AP, Cui C, Wang Y, Wang J, Troy E, Caggiano AO, Parry TJ, Colburn RW, Pavlopoulos E. rHIgM22 enhances remyelination in the brain of the cuprizone mouse model of demyelination. Neurobiol Dis 2017; 105:142-155. [DOI: 10.1016/j.nbd.2017.05.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 05/04/2017] [Accepted: 05/29/2017] [Indexed: 02/02/2023] Open
|
19
|
Schaal SM, Kitay BM, Cho KS, Lo TP, Barakat DJ, Marcillo AE, Sanchez AR, Andrade CM, Pearse DD. Schwann Cell Transplantation Improves Reticulospinal Axon Growth and Forelimb Strength after Severe Cervical Spinal Cord Contusion. Cell Transplant 2017; 16:207-28. [PMID: 17503734 DOI: 10.3727/000000007783464768] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Schwann cell (SC) implantation alone has been shown to promote the growth of propriospinal and sensory axons, but not long-tract descending axons, after thoracic spinal cord injury (SCI). In the current study, we examined if an axotomy close to the cell body of origin (so as to enhance the intrinsic growth response) could permit supraspinal axons to grow onto SC grafts. Adult female Fischer rats received a severe (C5) cervical contusion (1.1 mm displacement, 3 KDyn). At 1 week postinjury, 2 million SCs ex vivo transduced with lentiviral vector encoding enhanced green fluorescent protein (EGFP) were implanted within media into the injury epicenter; injury-only animals served as controls. Animals were tested weekly using the BBB score for 7 weeks postimplantation and received at end point tests for upper body strength: self-supported forelimb hanging, forearm grip force, and the incline plane. Following behavioral assessment, animals were anterogradely traced bilaterally from the reticular formation using BDA-Texas Red. Stereological quantification revealed a twofold increase in the numbers of preserved NeuN+ neurons rostral and caudal to the injury/graft site in SC implanted animals, corroborating previous reports of their neuroprotective efficacy. Examination of labeled reticulospinal axon growth revealed that while rarely an axon was present within the lesion site of injury-only controls, numerous reticulospinal axons had penetrated the SC implant/lesion milieu. This has not been observed following implantation of SCs alone into the injured thoracic spinal cord. Significant behavioral improvements over injury-only controls in upper limb strength, including an enhanced grip strength (a 296% increase) and an increased self-supported forelimb hanging, accompanied SC-mediated neuroprotection and reticulospinal axon growth. The current study further supports the neuroprotective efficacy of SC implants after SCI and demonstrates that SCs alone are capable of supporting modest supraspinal axon growth when the site of axon injury is closer to the cell body of the axotomized neuron.
Collapse
Affiliation(s)
- S M Schaal
- The Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami, FL 33101, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Crawford AH, Tripathi RB, Foerster S, McKenzie I, Kougioumtzidou E, Grist M, Richardson WD, Franklin RJM. Pre-Existing Mature Oligodendrocytes Do Not Contribute to Remyelination following Toxin-Induced Spinal Cord Demyelination. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:511-6. [PMID: 26773350 DOI: 10.1016/j.ajpath.2015.11.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 10/12/2015] [Accepted: 11/03/2015] [Indexed: 10/22/2022]
Abstract
Remyelination is the regenerative response to demyelination. Although the oligodendrocyte progenitor is established as the major source of remyelinating cells, there is no conclusive evidence on whether mature, differentiated oligodendrocytes can also contribute to remyelination. Using two different inducible myelin-CreER mouse strains in which mature oligodendrocytes were prelabeled by the expression of membrane-bound Green fluorescent protein, we found that after focal spinal cord demyelination, the surrounding surviving labeled oligodendrocytes did not proliferate but remained at a consistent density. Furthermore, existing (prelabeled) oligodendrocytes showed no evidence of incorporation or migration into the lesioned area, or of process extension from the peripheral margins into the lesion. Thus, mature oligodendrocytes do not normally contribute to remyelination and are therefore not a promising target for regenerative therapy.
Collapse
Affiliation(s)
- Abbe H Crawford
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Richa B Tripathi
- Wolfson Institute for Biomedical Research and Research Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Sarah Foerster
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Ian McKenzie
- Wolfson Institute for Biomedical Research and Research Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Eleni Kougioumtzidou
- Wolfson Institute for Biomedical Research and Research Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Matthew Grist
- Wolfson Institute for Biomedical Research and Research Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - William D Richardson
- Wolfson Institute for Biomedical Research and Research Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
21
|
Thyroid Hormone Potentially Benefits Multiple Sclerosis via Facilitating Remyelination. Mol Neurobiol 2015; 53:4406-16. [PMID: 26243185 DOI: 10.1007/s12035-015-9375-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 07/22/2015] [Indexed: 01/23/2023]
Abstract
Myelin destruction due to inflammatory damage of oligodendrocytes (OLs) in conjunction with axonal degeneration is one of the major histopathological hallmarks of multiple sclerosis (MS), a common autoimmune disorder affecting the central nervous system (CNS). Therapies over the last 20 years mainly focus on the immune system and, more specifically, on the modulation of immune cell behavior. It seems to be effective in MS with relapse, while it is of little benefit to progressive MS in which neurodegeneration following demyelination outweighs inflammation. Otherwise, remyelination, as a result of oligodendrocyte production from oligodendrocyte precursor cells (OPCs), is considered to be a potential target for the treatment of progressive MS. In this review, positive effects of remyelination on MS will be discussed in view of the critical role played by thyroid hormone (TH), focusing on the following points: (1) promising treatment of TH on MS that potentially targets to remyelination; (2) the active role of TH that is able to promote remyelination; (3) the regulative role of TH that works on endogenous stem and precursor cells; (4) the effect of TH on gene transcription; and (5) a working hypothesis which is developed that TH can alleviate MS by promoting remyelination, and the mechanism of which is its regulative role in gene transcription of OPCs.
Collapse
|
22
|
Marro BS, Blanc CA, Loring JF, Cahalan MD, Lane TE. Promoting remyelination: utilizing a viral model of demyelination to assess cell-based therapies. Expert Rev Neurother 2015; 14:1169-79. [PMID: 25245576 DOI: 10.1586/14737175.2014.955854] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS. While a broad range of therapeutics effectively reduce the incidence of focal white matter inflammation and plaque formation for patients with relapse-remitting forms of MS, a challenge within the field is to develop therapies that allow for axonal protection and remyelination. In the last decade, growing interest has focused on utilizing neural precursor cells (NPCs) to promote remyelination. To understand how NPCs function in chronic demyelinating environments, several excellent pre-clinical mouse models have been developed. One well accepted model is infection of susceptible mice with neurotropic variants of mouse hepatitis virus (MHV) that undergo chronic demyelination exhibiting clinical and histopathologic similarities to MS patients. Combined with the possibility that an environmental agent such as a virus could trigger MS, the MHV model of demyelination presents a relevant mouse model to assess the therapeutic potential of NPCs transplanted into an environment in which inflammatory-mediated demyelination is established.
Collapse
Affiliation(s)
- Brett S Marro
- Department of Molecular Biology and Biochemistry, University of California, Irvine 92697, USA
| | | | | | | | | |
Collapse
|
23
|
Shi H, Hu X, Leak RK, Shi Y, An C, Suenaga J, Chen J, Gao Y. Demyelination as a rational therapeutic target for ischemic or traumatic brain injury. Exp Neurol 2015; 272:17-25. [PMID: 25819104 DOI: 10.1016/j.expneurol.2015.03.017] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 03/15/2015] [Accepted: 03/18/2015] [Indexed: 12/11/2022]
Abstract
Previous research on stroke and traumatic brain injury (TBI) heavily emphasized pathological alterations in neuronal cells within gray matter. However, recent studies have highlighted the equal importance of white matter integrity in long-term recovery from these conditions. Demyelination is a major component of white matter injury and is characterized by loss of the myelin sheath and oligodendrocyte cell death. Demyelination contributes significantly to long-term sensorimotor and cognitive deficits because the adult brain only has limited capacity for oligodendrocyte regeneration and axonal remyelination. In the current review, we will provide an overview of the major causes of demyelination and oligodendrocyte cell death following acute brain injuries, and discuss the crosstalk between myelin, axons, microglia, and astrocytes during the process of demyelination. Recent discoveries of molecules that regulate the processes of remyelination may provide novel therapeutic targets to restore white matter integrity and improve long-term neurological recovery in stroke or TBI patients.
Collapse
Affiliation(s)
- Hong Shi
- The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Department of Anesthesiology of Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Xiaoming Hu
- The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, USA
| | - Yejie Shi
- The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Chengrui An
- The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Jun Suenaga
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jun Chen
- The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA.
| | - Yanqin Gao
- The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
24
|
Neurotoxocarosis alters myelin protein gene transcription and expression. Parasitol Res 2015; 114:2175-86. [DOI: 10.1007/s00436-015-4407-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 03/02/2015] [Indexed: 02/04/2023]
|
25
|
Yeung MSY, Zdunek S, Bergmann O, Bernard S, Salehpour M, Alkass K, Perl S, Tisdale J, Possnert G, Brundin L, Druid H, Frisén J. Dynamics of oligodendrocyte generation and myelination in the human brain. Cell 2015; 159:766-74. [PMID: 25417154 DOI: 10.1016/j.cell.2014.10.011] [Citation(s) in RCA: 366] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 09/02/2014] [Accepted: 10/03/2014] [Indexed: 11/16/2022]
Abstract
The myelination of axons by oligodendrocytes has been suggested to be modulated by experience, which could mediate neural plasticity by optimizing the performance of the circuitry. We have assessed the dynamics of oligodendrocyte generation and myelination in the human brain. The number of oligodendrocytes in the corpus callosum is established in childhood and remains stable after that. Analysis of the integration of nuclear bomb test-derived (14)C revealed that myelin is exchanged at a high rate, whereas the oligodendrocyte population in white matter is remarkably stable in humans, with an annual exchange of 1/300 oligodendrocytes. We conclude that oligodendrocyte turnover contributes minimally to myelin modulation in human white matter and that this instead may be carried out by mature oligodendrocytes, which may facilitate rapid neural plasticity.
Collapse
Affiliation(s)
- Maggie S Y Yeung
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sofia Zdunek
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Olaf Bergmann
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Samuel Bernard
- Institut Camille Jordan, CNRS UMR 5208, University of Lyon, 69622 Villeurbanne, France
| | - Mehran Salehpour
- Department of Physics and Astronomy, Ion Physics, Uppsala University, 751 20 Uppsala, Sweden
| | - Kanar Alkass
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Forensic Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | | | - Göran Possnert
- Department of Physics and Astronomy, Ion Physics, Uppsala University, 751 20 Uppsala, Sweden
| | - Lou Brundin
- Department of Clinical Neuroscience, Department of Neurosurgery and Neurology, Karolinska University Hospital, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Henrik Druid
- Department of Forensic Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
26
|
Pusic AD, Mitchell HM, Kunkler PE, Klauer N, Kraig RP. Spreading depression transiently disrupts myelin via interferon-gamma signaling. Exp Neurol 2015; 264:43-54. [PMID: 25500111 PMCID: PMC4324018 DOI: 10.1016/j.expneurol.2014.12.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/24/2014] [Accepted: 12/02/2014] [Indexed: 01/31/2023]
Abstract
Multiple sclerosis and migraine with aura are clinically correlated and both show imaging changes suggestive of myelin disruption. Furthermore, cortical myelin loss in the cuprizone animal model of multiple sclerosis enhances susceptibility to spreading depression, the likely underlying cause of migraine with aura. Since multiple sclerosis pathology involves inflammatory T cell lymphocyte production of interferon-gamma and a resulting increase in oxidative stress, we tested the hypothesis that spreading depression disrupts myelin through similar signaling pathways. Rat hippocampal slice cultures were initially used to explore myelin loss in spreading depression, since they contain T cells, and allow for controlled tissue microenvironment. These experiments were then translated to the in vivo condition in neocortex. Spreading depression in slice cultures induced significant loss of myelin integrity and myelin basic protein one day later, with gradual recovery by seven days. Myelin basic protein loss was abrogated by T cell depletion, neutralization of interferon-gamma, and pharmacological inhibition of neutral sphingomyelinase-2. Conversely, one day after exposure to interferon-gamma, significant reductions in spreading depression threshold, increases in oxidative stress, and reduced levels of glutathione, an endogenous neutral sphingomyelinase-2 inhibitor, emerged. Similarly, spreading depression triggered significant T cell accumulation, sphingomyelinase activation, increased oxidative stress, and reduction of gray and white matter myelin in vivo. Myelin disruption is involved in spreading depression, thereby providing pathophysiological links between multiple sclerosis and migraine with aura. Myelin disruption may promote spreading depression by enhancing aberrant excitability. Thus, preservation of myelin integrity may provide novel therapeutic targets for migraine with aura.
Collapse
Affiliation(s)
- Aya D Pusic
- Department of Neurology, The University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637-1470, USA; The Committee on Neurobiology, The University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637-1470, USA.
| | - Heidi M Mitchell
- Department of Neurology, The University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637-1470, USA.
| | - Phillip E Kunkler
- Department of Neurology, The University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637-1470, USA.
| | - Neal Klauer
- Department of Neurology, The University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637-1470, USA.
| | - Richard P Kraig
- Department of Neurology, The University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637-1470, USA; The Committee on Neurobiology, The University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637-1470, USA.
| |
Collapse
|
27
|
Michailidou I, de Vries HE, Hol EM, van Strien ME. Activation of endogenous neural stem cells for multiple sclerosis therapy. Front Neurosci 2015; 8:454. [PMID: 25653584 PMCID: PMC4299409 DOI: 10.3389/fnins.2014.00454] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/22/2014] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system, leading to severe neurological deficits. Current MS treatment regimens, consist of immunomodulatory agents aiming to reduce the rate of relapses. However, these agents are usually insufficient to treat chronic neurological disability. A promising perspective for future therapy of MS is the regeneration of lesions with replacement of the damaged oligodendrocytes or neurons. Therapies targeting to the enhancement of endogenous remyelination, aim to promote the activation of either the parenchymal oligodendrocyte progenitor cells or the subventricular zone-derived neural stem cells (NSCs). Less studied but highly potent, is the strategy of neuronal regeneration with endogenous NSCs that although being linked to numerous limitations, is anticipated to ameliorate cognitive disability in MS. Focusing on the forebrain, this review highlights the role of NSCs in the regeneration of MS lesions.
Collapse
Affiliation(s)
- Iliana Michailidou
- Department of Astrocyte Biology and Neurodegeneration, The Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Sciences Amsterdam, Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Netherlands
| | - Elly M Hol
- Department of Astrocyte Biology and Neurodegeneration, The Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Sciences Amsterdam, Netherlands ; Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands ; Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Miriam E van Strien
- Department of Astrocyte Biology and Neurodegeneration, The Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Sciences Amsterdam, Netherlands ; Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
28
|
Abstract
Spinal cord injury is a major cause of disability with devastating neurological outcomes and limited therapeutic opportunities, even though there are thousands of publications on spinal cord injury annually. There are two major types of spinal cord injury, transaction of the spinal cord and spinal cord contusion. Both can theoretically be treated, but there is no well documented treatment in human being. As for spinal cord contusion, we have developed an operation with fabulous result.
Collapse
Affiliation(s)
- Gong Ju
- Institute of Neurosciences, Department of Neurobiology, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Jian Wang
- Institute of Neurosciences, Department of Neurobiology, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Yazhou Wang
- Institute of Neurosciences, Department of Neurobiology, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Xianghui Zhao
- Institute of Neurosciences, Department of Neurobiology, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
29
|
Boulanger JJ, Messier C. From precursors to myelinating oligodendrocytes: contribution of intrinsic and extrinsic factors to white matter plasticity in the adult brain. Neuroscience 2014; 269:343-66. [PMID: 24721734 DOI: 10.1016/j.neuroscience.2014.03.063] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/28/2014] [Accepted: 03/28/2014] [Indexed: 12/21/2022]
Abstract
Oligodendrocyte precursor cells (OPC) are glial cells that metamorphose into myelinating oligodendrocytes during embryogenesis and early stages of post-natal life. OPCs continue to divide throughout adulthood and some eventually differentiate into oligodendrocytes in response to demyelinating lesions. There is growing evidence that OPCs are also involved in activity-driven de novo myelination of previously unmyelinated axons and myelin remodeling in adulthood. In this review, we summarize the interwoven factors and cascades that promote the activation, recruitment and differentiation of OPCs into myelinating oligodendrocytes in the adult brain based mostly on results found in the study of demyelinating diseases. The goal of the review was to draw a complete picture of the transformation of OPCs into mature oligodendrocytes to facilitate the study of this transformation in both the normal and diseased adult brain.
Collapse
Affiliation(s)
| | - C Messier
- School of Psychology, University of Ottawa, Canada.
| |
Collapse
|
30
|
Gordon T. Neurotrophic factor expression in denervated motor and sensory Schwann cells: relevance to specificity of peripheral nerve regeneration. Exp Neurol 2014; 254:99-108. [PMID: 24468476 DOI: 10.1016/j.expneurol.2014.01.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/09/2014] [Accepted: 01/11/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Tessa Gordon
- Department of Surgery, Division of Plastic Reconstructive Surgery 5549A, The Hospital for Sick Children, 555 University Ave, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
31
|
Brosius Lutz A, Barres BA. Contrasting the Glial Response to Axon Injury in the Central and Peripheral Nervous Systems. Dev Cell 2014; 28:7-17. [DOI: 10.1016/j.devcel.2013.12.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
32
|
Adamo AM. Nutritional factors and aging in demyelinating diseases. GENES AND NUTRITION 2013; 9:360. [PMID: 24311441 DOI: 10.1007/s12263-013-0360-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 11/07/2013] [Indexed: 12/18/2022]
Abstract
Demyelination is a pathological process characterized by the loss of myelin around axons. In the central nervous system, oligodendroglial damage and demyelination are common pathological features characterizing white matter and neurodegenerative disorders. Remyelination is a regenerative process by which myelin sheaths are restored to demyelinated axons, resolving functional deficits. This process is often deficient in demyelinating diseases such as multiple sclerosis (MS), and the reasons for the failure of repair mechanisms remain unclear. The characterization of these mechanisms and the factors involved in the proliferation, recruitment, and differentiation of oligodendroglial progenitor cells is key in designing strategies to improve remyelination in demyelinating disorders. First, a very dynamic combination of different molecules such as growth factors, cytokines, chemokines, and different signaling pathways is tightly regulated during the remyelination process. Second, factors unrelated to this pathology, i.e., age and genetic background, may impact disease progression either positively or negatively, and in particular, age-related remyelination failure has been proven to involve oligodendroglial cells aging and their intrinsic capacities among other factors. Third, nutrients may either help or hinder disease progression. Experimental evidence supports the anti-inflammatory role of omega-6 and omega-3 polyunsaturated fatty acids through the competitive inhibition of arachidonic acid, whose metabolites participate in inflammation, and the reduction in T cell proliferation. In turn, vitamin D intake and synthesis have been associated with lower MS incidence levels, while vitamin D-gene interactions might be involved in the pathogenesis of MS. Finally, dietary polyphenols have been reported to mitigate demyelination by modulating the immune response.
Collapse
Affiliation(s)
- Ana M Adamo
- Department of Biological Chemistry, IQUIFIB (UBA-CONICET), School of Pharmacy and Biochemistry, University of Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina,
| |
Collapse
|
33
|
Salehi Z, Hadiyan SP, Navidi R. Ciliary neurotrophic factor role in myelin oligodendrocyte glycoprotein expression in Cuprizone-induced multiple sclerosis mice. Cell Mol Neurobiol 2013; 33:531-5. [PMID: 23443463 PMCID: PMC11498024 DOI: 10.1007/s10571-013-9918-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 02/11/2013] [Indexed: 01/19/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system that leads to loss of myelin and oligodendrocytes and damage to axons. Myelin oligodendrocyte glycoprotein (MOG) is a minor component of the myelin sheath, but is an important autoantigen linked to the pathogenesis of MS. Ciliary neurotrophic factor (CNTF) has been shown to enhance the generation, maturation, and survival of oligodendrocytes in culture medium. The aim of this study was to demonstrate the role of CNTF on MOG expression in the cerebral cortex of Cuprizone-induced MS mice. The mice were treated by Cuprizone for five weeks in order to induce MS. The mice were then divided into 3 groups. The first group was injected subcutaneously (SC) by CNTF in the amount of 250 μg/kg BW per day. The second group (SHAM) was injected SC by normal saline and the third group was left without injection as the control group. After four weeks the mice were killed and the cerebral cortex was harvested and the expression of MOG was studied by Western blotting. The data from this study show that the MOG expression was significantly increased in the CNTF-injected group as compared to the other groups. It is concluded that CNTF increases the MOG expression and may be important in the pathophysiology of MS. It is also concluded that CNTF may play a role in the process of remyelination by inducing the MOG expression.
Collapse
Affiliation(s)
- Zivar Salehi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran.
| | | | | |
Collapse
|
34
|
Sabo JK, Cate HS. Signalling pathways that inhibit the capacity of precursor cells for myelin repair. Int J Mol Sci 2013; 14:1031-49. [PMID: 23296277 PMCID: PMC3565305 DOI: 10.3390/ijms14011031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 12/21/2012] [Accepted: 12/31/2012] [Indexed: 12/17/2022] Open
Abstract
In demyelinating disorders such as Multiple Sclerosis (MS), targets of injury are myelin and oligodendrocytes, leading to severe neurological dysfunction. Regenerative therapies aimed at promoting oligodendrocyte maturation and remyelination are promising strategies for treatment in demyelinating disorders. Endogenous precursor cells or exogenous transplanted cells are potential sources for remyelinating oligodendrocytes in the central nervous system (CNS). Several signalling pathways have been implicated in regulating the capacity of these cell populations for myelin repair. Here, we review neural precursor cells and oligodendrocyte progenitor cells as potential sources for remyelinating oligodendrocytes and evidence for the functional role of key signalling pathways in inhibiting regeneration from these precursor cell populations.
Collapse
Affiliation(s)
- Jennifer K Sabo
- Centre for Neuroscience Research, Department of Anatomy and Neuroscience, University of Melbourne, Melbourne Brain Centre, Kenneth Myer Building, 30 Royal Parade, Parkville, Vic 3010, Australia.
| | | |
Collapse
|
35
|
Tatar C, Bessert D, Tse H, Skoff RP. Determinants of central nervous system adult neurogenesis are sex, hormones, mouse strain, age, and brain region. Glia 2012; 61:192-209. [PMID: 23027402 DOI: 10.1002/glia.22426] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 08/30/2012] [Indexed: 11/11/2022]
Abstract
Multiple sclerosis is a sexually dimorphic (SD) disease that causes oligodendrocyte death, but SD of glial cells is poorly studied. Here, we analyze SD of neural progenitors in 6-8 weeks and 6-8 months normal C57BL/6, SJL/J, and BALB/c mice in the subventricular zone (SVZ), dorsolateral horn (DLC), corpus callosum (CC), and parenchyma. With a short 2-h bromodeoxyuridine (BrdU) pulse, no gender and strain differences are present at 6-8 weeks. At 6-8 months, the number of BrdU(+) cells decreases twofold in each sex, strain, and region, indicating that a common aging mechanism regulates BrdU incorporation. Strikingly, 2× more BrdU(+) cells are found in all brain regions in 6-8 months C57BL/6 females versus males, no gender differences in 6-8 months SJL/J, and fewer BrdU(+) cells in females versus males in BALB/cs. The number of BrdU(+) cells modestly fluctuates throughout the estrous cycle in C57BL/6 and SJLs. Castration causes a dramatic increase in BrdU(+) cells in SVZ and DLC. These findings indicate that testosterone is a major regulator of adult neural proliferation. At 6-8 months, the ratio of PDGFRα(+) cells in the CC to BrdU(+) cells in the DLC of both strains, sexes, estrous cycle, and castrated mice was essentially the same, suggesting that BrdU(+) cells in the DLC differentiate into CC oligodendrocytes. The ratio of TUNEL(+) to BrdU(+) cells does not match proliferation, indicating that these events are differentially regulated. Differential regulation of these two processes leads to the variation in glial numbers between gender and strain. Explanations of neural proliferation based upon data from one sex or strain may be very misleading.
Collapse
Affiliation(s)
- Carrie Tatar
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | |
Collapse
|
36
|
Ueno T, Ito J, Hoshikawa S, Ohori Y, Fujiwara S, Yamamoto S, Ohtsuka T, Kageyama R, Akai M, Nakamura K, Ogata T. The identification of transcriptional targets of Ascl1 in oligodendrocyte development. Glia 2012; 60:1495-505. [PMID: 22714260 DOI: 10.1002/glia.22369] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 04/24/2012] [Accepted: 05/21/2012] [Indexed: 11/06/2022]
Abstract
The basic helix-loop-helix (bHLH) transcription factor Ascl1 plays crucial roles in both oligodendrocyte development and neuronal development; however, the molecular target of Ascl1 in oligodendrocyte progenitor cells (OPCs) remains elusive. To identify the downstream targets of Ascl1 in OPCs, we performed gene expression microarray analysis and identified Hes5 as a putative downstream target of Ascl1. In vivo analysis revealed that Ascl1 and Hes5 were coexpressed in early developmental oligodendrocytes in both the telencephalon and the ventral spinal cord. We also found that Hes5 expression was reduced in the OPCs of Ascl1 mutant mice. Furthermore, we demonstrated that Ascl1 directly binds to an E-box region within the Hes5 promoter and regulates Hes5 expression at the transcriptional level. Taken together, these in vivo and in vitro data suggest that Ascl1 induces Hes5 expression in a cell-autonomous manner. Considering the previously known function of Hes5 as a repressor of Ascl1, our data indicate that Hes5 is involved in the negative feedback regulation of Ascl1.
Collapse
Affiliation(s)
- Takaaki Ueno
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Galanin transgenic mice with elevated circulating galanin levels alleviate demyelination in a cuprizone-induced MS mouse model. PLoS One 2012; 7:e33901. [PMID: 22442732 PMCID: PMC3307774 DOI: 10.1371/journal.pone.0033901] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 02/19/2012] [Indexed: 01/15/2023] Open
Abstract
Multiple Sclerosis (MS) is a demyelinating autoimmune disease of the central nervous system (CNS) with a presumed autoimmune etiology. Approved treatments for MS are immunoregulatory and are able to reduce the inflammatory components of the disease. However, these treatments do not suppress progressive clinical disability. Approaches that directly protect myelin-producing oligodendrocytes and enhance remyelination are likely to improve long-term outcomes and reduce the rate of axonal damage. Galanin (GAL) is a bioactive neuropeptide that is widely distributed throughout the nervous system and has diverse neuromodulatory effects. In this study, using the cuprizone (CPZ) demyelination model of MS, we demonstrate that GAL has pronounced neuroprotective effects with respect to demyelination and remyelination. Using our GAL transgenic mouse (GAL-Tg), we identified a novel attenuation of OLs against CPZ induced demyelination, which was exerted independently of progenitor cells. Alleviation of myelin breakdown in the GAL-Tg mice was observed to be significant. Furthermore, we observed changes in the expression of the GAL receptor GalR1 during the demyelination and remyelination processes. Our data strongly indicate that GAL has the capacity to influence the outcome of primary insults that directly target OLs, as opposed to cases where immune activation is the primary pathogenic event. Taken together, these results suggest that GAL is a promising next-generation target for the treatment of MS.
Collapse
|
38
|
Katz Sand IB, Krieger S. Emerging strategies for the treatment of multiple sclerosis. FUTURE NEUROLOGY 2012. [DOI: 10.2217/fnl.12.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Despite extraordinary advances in the field of neuroimmunology, ideal treatment for patients with multiple sclerosis remains an unmet need. Existing treatments are only partially effective in preventing multiple sclerosis relapses, have a limited impact on the accrual of disability, have not been effective in progressive forms of the disease, and treatment remains preventive rather than restorative. This review provides an overview of emerging therapies and targets, and incorporates strategies for two different approaches to multiple sclerosis: prevention, through immune modulation; and repair, through neuroprotection and remyelination. Agents at all stages of development, from late-stage clinical trials of BG-12, teriflunomide, alemtuzumab, daclizumab and anti-CD20 agents, to novel approaches in preclinical testing, are discussed.
Collapse
Affiliation(s)
- Ilana B Katz Sand
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Mount Sinai School of Medicine, 5 East 98th Street, Box 1138, New York, NY, 10029, USA
| | - Stephen Krieger
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Mount Sinai School of Medicine, 5 East 98th Street, Box 1138, New York, NY, 10029, USA
| |
Collapse
|
39
|
Olah M, Amor S, Brouwer N, Vinet J, Eggen B, Biber K, Boddeke HWGM. Identification of a microglia phenotype supportive of remyelination. Glia 2011; 60:306-21. [PMID: 22072381 DOI: 10.1002/glia.21266] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 10/11/2011] [Indexed: 11/07/2022]
Abstract
In multiple sclerosis, endogenous oligodendrocyte precursor cells (OPCs) attempt to remyelinate areas of myelin damage. During disease progression, however, these attempts fail. It has been suggested that modulating the inflammatory environment of the lesion might provide a promising therapeutic approach to promote endogenous remyelination. Microglia are known to play a central role in neuroinflammatory processes. To investigate the microglia phenotype that supports remyelination, we performed genome-wide gene expression analysis of microglia from the corpus callosum during demyelination and remyelination in the mouse cuprizone model, in which remyelination spontaneously occurs after an episode of toxin-induced primary demyelination. We provide evidence for the existence of a microglia phenotype that supports remyelination already at the onset of demyelination and persists throughout the remyelination process. Our data show that microglia are involved in the phagocytosis of myelin debris and apoptotic cells during demyelination. Furthermore, they express a cytokine and chemokine repertoire enabling them to activate and recruit endogenous OPCs to the lesion site and deliver trophic support during remyelination. This study not only provides a detailed transcriptomic analysis of the remyelination-supportive microglia phenotype but also reinforces the notion that the primary function of microglia is the maintenance of tissue homeostasis and the support of regeneration already at the earliest stages in the development of demyelinating lesions.
Collapse
Affiliation(s)
- Marta Olah
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
40
|
Münzel EJ, Schaefer K, Obirei B, Kremmer E, Burton EA, Kuscha V, Becker CG, Brösamle C, Williams A, Becker T. Claudin k is specifically expressed in cells that form myelin during development of the nervous system and regeneration of the optic nerve in adult zebrafish. Glia 2011; 60:253-70. [PMID: 22020875 DOI: 10.1002/glia.21260] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 10/05/2011] [Indexed: 01/07/2023]
Abstract
The zebrafish has become an important model organism to study myelination during development and after a lesion of the adult central nervous system (CNS). Here, we identify Claudin k as a myelin-associated protein in zebrafish and determine its localization during development and adult optic nerve regeneration. We find Claudin k in subcellular compartments consistent with location in autotypic tight junctions of oligodendrocytes and myelinating Schwann cells. Expression starts in the hindbrain at 2 days (mRNA) and 3 days (protein) postfertilization and is maintained in adults. A newly generated claudin k:green fluorescent protein (GFP) reporter line allowed us to characterize oligodendrocytes in the adult retina that express Claudin k and olig2, but not P0 and uniquely only form loose wraps of membrane around axons. After a crush of the adult optic nerve, Claudin k protein levels were first reduced and then recovered within 4 weeks postlesion, concomitant with optic nerve myelin de- and regeneration. During optic nerve regeneration, oligodendrocytes, many of which were newly generated, repopulated the lesion site and exhibited increasing morphological complexity over time. Thus, Claudin k is a novel myelin-associated protein expressed by oligodendrocytes and Schwann cells from early stages of wrapping and myelin formation in zebrafish development and adult regeneration, suggesting important functions of the gene for myelin formation and maintenance. Our Claudin k antibodies and claudin k:GFP reporter line represent excellent ways to visualize oligodendrocyte and Schwann cell differentiation in vivo.
Collapse
Affiliation(s)
- Eva Jolanda Münzel
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ly L, Barnett MH, Zheng YZ, Gulati T, Prineas JW, Crossett B. Comprehensive tissue processing strategy for quantitative proteomics of formalin-fixed multiple sclerosis lesions. J Proteome Res 2011; 10:4855-68. [PMID: 21870854 DOI: 10.1021/pr200672n] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Formalin-fixed (FF) autopsy tissue comprises the bulk of existing Multiple Sclerosis (MSc) pathology archives, providing a rich pool of material for biomarker discovery and disease characterization. Here, we present the development of a heat-induced extraction protocol for the proteomic analysis of FF brain tissue, its application to the study of lesion remyelination and its failure in MSc. A 4-round extraction strategy was optimized using FF tissue leading to a 35% increase in the number of proteins identified compared to a single extraction; and a 65% increase in proteins identified with ≥4 peptides. Histological staining of sections with oil red O and luxol fast blue-periodic acid Schiff, required to characterize MSc lesions was found to have minimal effect on LC-MS/MS. The application of the optimized protocol to chronic demyelinated and remyelinated FF MSc lesions and the adjacent periplaque white matter, isolated through laser guided manual dissection from 3 patients, identified 428 unique proteins (0.2% FDR) using LC-MS/MS. Comparison of the lesion types using iTRAQ and 2-D LC-MS/MS revealed 82 differentially expressed proteins. Protein quantitation by iTRAQ and spectral counting was well-correlated (r(s)= 0.7653; p < 10(-30)). The data generated from this work illustrates the scope of the methodology and provides insights into the pathogenesis of MSc and remyelination.
Collapse
Affiliation(s)
- Linda Ly
- Central Clinical School, The University of Sydney, NSW, Australia
| | | | | | | | | | | |
Collapse
|
42
|
Carbajal KS, Miranda JL, Tsukamoto MR, Lane TE. CXCR4 signaling regulates remyelination by endogenous oligodendrocyte progenitor cells in a viral model of demyelination. Glia 2011; 59:1813-21. [PMID: 21830237 DOI: 10.1002/glia.21225] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2011] [Accepted: 07/05/2011] [Indexed: 11/09/2022]
Abstract
Following intracranial infection with the neurotropic JHM strain of mouse hepatitis virus (JHMV), susceptible mice will develop widespread myelin destruction that results in pathological and clinical outcomes similar to those seen in humans with the demyelinating disease Multiple Sclerosis (MS). Partial remyelination and clinical recovery occurs during the chronic phase following control of viral replication yet the signaling mechanisms regulating these events remain enigmatic. Here we report the kinetics of proliferation and maturation of oligodendrocyte progenitor cells (OPCs) within the spinal cord following JHMV-induced demyelination and that CXCR4 signaling contributes to the maturation state of OPCs. Following treatment with AMD3100, a specific inhibitor of CXCR4, mice recovering from widespread demyelination exhibit a significant (P < 0.01) increase in the number of OPCs and fewer (P < 0.05) mature oligodendrocytes compared with HBSS-treated animals. These results suggest that CXCR4 signaling is required for OPCs to mature and contribute to remyelination in response to JHMV-induced demyelination. To assess if this effect is reversible and has potential therapeutic benefit, we pulsed mice with AMD3100 and then allowed them to recover. This treatment strategy resulted in increased numbers of mature oligodendrocytes, enhanced remyelination, and improved clinical outcome. These findings highlight the possibility to manipulate OPCs in order to increase the pool of remyelination-competent cells that can participate in recovery.
Collapse
Affiliation(s)
- Kevin S Carbajal
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | | | | | | |
Collapse
|
43
|
K(V)7/KCNQ channels are functionally expressed in oligodendrocyte progenitor cells. PLoS One 2011; 6:e21792. [PMID: 21750731 PMCID: PMC3130044 DOI: 10.1371/journal.pone.0021792] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 06/12/2011] [Indexed: 11/26/2022] Open
Abstract
Background KV7/KCNQ channels are widely expressed in neurons and they have multiple important functions, including control of excitability, spike afterpotentials, adaptation, and theta resonance. Mutations in KCNQ genes have been demonstrated to associate with human neurological pathologies. However, little is known about whether KV7/KCNQ channels are expressed in oligodendrocyte lineage cells (OLCs) and what their functions in OLCs. Methods and Findings In this study, we characterized KV7/KCNQ channels expression in rat primary cultured OLCs by RT-PCR, immunostaining and electrophysiology. KCNQ2-5 mRNAs existed in all three developmental stages of rat primary cultured OLCs. KV7/KCNQ proteins were also detected in oligodendrocyte progenitor cells (OPCs, early developmental stages of OLCs) of rat primary cultures and cortex slices. Voltage-clamp recording revealed that the IM antagonist XE991 significantly reduced KV7/KCNQ channel current (IK(Q)) in OPCs but not in differentiated oligodendrocytes. In addition, inhibition of KV7/KCNQ channels promoted OPCs motility in vitro. Conclusions These findings showed that KV7/KCNQ channels were functionally expressed in rat primary cultured OLCs and might play an important role in OPCs functioning in physiological or pathological conditions.
Collapse
|
44
|
Zhu Q, Whittemore SR, Devries WH, Zhao X, Kuypers NJ, Qiu M. Dorsally-derived oligodendrocytes in the spinal cord contribute to axonal myelination during development and remyelination following focal demyelination. Glia 2011; 59:1612-21. [PMID: 21710609 DOI: 10.1002/glia.21203] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 05/26/2011] [Indexed: 11/10/2022]
Abstract
In the developing spinal cord, the majority of oligodendrocytes are derived from the ventral ventricular zone. Several recent studies suggested that a small number of oligodendrocyte precursor cells (OPCs) can also be generated in the dorsal spinal cord. However, it is not clear whether these dorsal oligodendrocyte precursor cells participate in myelination and remyelination. To investigate the fate and potential function of these dorsally-derived oligodendrocytes (dOLs) in the adult spinal cord, Cre-lox genetic fate mapping in transgenic mice was employed. We used the Pax3(Cre) knock-in mouse to drive Cre expression in the entire dorsal epithelium and the Rosa26-lacZ or Z/EG reporter line to trace their spatial distribution and population dynamics in the spinal cord. The dorsal OPCs generated from the Pax3-expressing domains migrate into all regions of spinal cord and subsequently undergo terminal differentiation and axonal myelination. In response to a focal demyelination injury, a large number of newly differentiated oligodendrocytes originated from dOLs, suggesting that dOLs may provide an important source of OPCs for axonal remyelination in multiple sclerosis or spinal cord injury.
Collapse
Affiliation(s)
- Qiang Zhu
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, USA
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Oligodendrocytes (OLs) are particularly susceptible to the toxicity of the acute lesion environment after spinal cord injury (SCI). They undergo both necrosis and apoptosis acutely, with apoptosis continuing at chronic time points. Loss of OLs causes demyelination and impairs axon function and survival. In parallel, a rapid and protracted OL progenitor cell proliferative response occurs, especially at the lesion borders. Proliferating and migrating OL progenitor cells differentiate into myelinating OLs, which remyelinate demyelinated axons starting at 2 weeks post-injury. The progression of OL lineage cells into mature OLs in the adult after injury recapitulates development to some degree, owing to the plethora of factors within the injury milieu. Although robust, this endogenous oligogenic response is insufficient against OL loss and demyelination. First, in this review we analyze the major spatial-temporal mechanisms of OL loss, replacement, and myelination, with the purpose of highlighting potential areas of intervention after SCI. We then discuss studies on OL protection and replacement. Growth factors have been used both to boost the endogenous progenitor response, and in conjunction with progenitor transplantation to facilitate survival and OL fate. Considerable progress has been made with embryonic stem cell-derived cells and adult neural progenitor cells. For therapies targeting oligogenesis to be successful, endogenous responses and the effects of the acute and chronic lesion environment on OL lineage cells must be understood in detail, and in relation, the optimal therapeutic window for such strategies must also be determined.
Collapse
Affiliation(s)
- Akshata Almad
- Neuroscience Graduate Studies Program, Ohio State University, Columbus, Ohio 43210 USA
- Center for Brain and Spinal Cord Repair, Ohio State University, Columbus, Ohio 43210 USA
| | - F. Rezan Sahinkaya
- Neuroscience Graduate Studies Program, Ohio State University, Columbus, Ohio 43210 USA
- Center for Brain and Spinal Cord Repair, Ohio State University, Columbus, Ohio 43210 USA
| | - Dana M. McTigue
- Center for Brain and Spinal Cord Repair, Ohio State University, Columbus, Ohio 43210 USA
- Department of Neuroscience, Ohio State University, 788 Biomedical Research Tower, 460 W. 12th Ave, Columbus, Ohio 43210 USA
| |
Collapse
|
46
|
Jennings A, Carroll W. Quantification of oligodendrocyte progenitor cells in human and cat optic nerve: implications for endogenous repair in multiple sclerosis. Glia 2010; 58:1425-36. [PMID: 20549750 DOI: 10.1002/glia.21018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In multiple sclerosis (MS), one strategy to reduce disability is enhancement of endogenous repair by remyelinating oligodendrocytes derived from oligodendrocyte progenitor cells (OP). An important prerequisite is determining the abundance of OP relative to oligodendrocytes in normal human central nervous system (CNS), which, in turn, requires reliable OP identification. To achieve this, cat and human optic nerves (ON) were subjected to varied preparation protocols, and the resultant neuroglial staining profiles correlated to generate an antigenic phenotype for OP applicable to human autopsy specimens. OP, interchangeably called NG2cells due to universal NG2 expression, were shown to comprise a separate class of neuroglial cells, related to oligodendrocytes by expression of the oligodendrocyte lineage transcription factors, Olig1 and Olig2. Despite their morphological complexity, including contact with axons and other neuroglia, NG2cells all appear capable of responding as OP to counter local oligodendrocyte loss. However, quantification revealed that NG2cells comprised less than 5% of the neuroglia and had a ratio to oligodendrocytes of about 1:10, not only in human and cat ON but also in white and gray-matter regions of cat spinal cord. The finding that NG2cells are not abundant, particularly relative to oligodendrocytes, may have implications for efforts to enhance endogenous repair in MS.
Collapse
Affiliation(s)
- Alison Jennings
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia.
| | | |
Collapse
|
47
|
Cate HS, Sabo JK, Merlo D, Kemper D, Aumann TD, Robinson J, Merson TD, Emery B, Perreau VM, Kilpatrick TJ. Modulation of bone morphogenic protein signalling alters numbers of astrocytes and oligodendroglia in the subventricular zone during cuprizone-induced demyelination. J Neurochem 2010; 115:11-22. [PMID: 20193041 DOI: 10.1111/j.1471-4159.2010.06660.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The adult subventricular zone (SVZ) is a potential source of precursor cells to replace neural cells lost during demyelination. To better understand the molecular events that regulate neural precursor cell responsiveness in this context we undertook a microarray and quantitative PCR based analysis of genes expressed within the SVZ during cuprizone-induced demyelination. We identified an up-regulation of the genes encoding bone morphogenic protein 4 (BMP4) and its receptors. Immunohistochemistry confirmed an increase in BMP4 protein levels and also showed an increase in phosphorylated SMAD 1/5/8, a key component of BMP4 signalling, during demyelination. In vitro analysis revealed that neural precursor cells isolated from demyelinated animals, as well as those treated with BMP4, produce more astrocytes. Similarly, there were increased numbers of astrocytes in vivo within the SVZ during demyelination. Intraventricular infusion of Noggin, an endogenous antagonist of BMP4, during cuprizone-induced demyelination reduced pSMAD1/5/8, decreased astrocyte numbers and increased oligodendrocyte numbers in the SVZ. Our results suggest that lineage commitment of SVZ neural precursor cells is altered during demyelination and that BMP signalling plays a role in this process.
Collapse
Affiliation(s)
- Holly S Cate
- Centre for Neuroscience, University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Response of human oligodendrocyte progenitors to growth factors and axon signals. J Neuropathol Exp Neurol 2010; 69:930-44. [PMID: 20720504 DOI: 10.1097/nen.0b013e3181ef3be4] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
We examined the effects of growth factors and axonal signals on the differentiation of human fetal and adult oligodendrocyte progenitor cells (OPCs) and determined whether these effects translated into enhanced axonal ensheathment. Only small numbers of fetal OPCs grown in defined medium expressed the oligodendroglial lineage markers Olig2 and O4. The combination of platelet-derived growth factor-AA and basic fibroblast growth factor enhanced proliferation of Olig2-positive and O4-positive cells; a combination of brain-derived neurotrophic factor and insulin-like growth factor 1 promoted O4-positive cell differentiation, galactocerebroside expression, and morphological complexity. Coculturing with rodent dorsal root ganglion neurons in defined medium alone enhanced OPC differentiation and myelin basic protein expression. The addition of brain-derived neurotrophic factor/insulin-like growth factor 1 further enhanced differentiation, axonal attachment and ensheathment, and clustering of the contactin-associated protein Caspr and Na+ channels. By contrast, most adult OPCs were O4 positive and Olig2 positive in defined medium; both brain-derived neurotrophic factor/insulin-like growth factor 1 and platelet-derived growth factor-AA/basic fibroblast growth factor promoted their myelin basic protein expression and membrane sheet formation; coculture with dorsal root ganglion neurons further increased myelin basic protein expression. Growth factors also enhanced attachment of adult OPCs to axons, but their capacity to ensheath axons was lower than that of fetal OPCs. These results demonstrate that fetal and adult OPCs show measurable responses to selected growth factors and axon signals that correlate with their capacity for axon ensheathment. The distinct properties of fetal and adult OPCs may be related to differences in their chronological age and initial differentiation states.
Collapse
|
49
|
Parratt JDE, Prineas JW. Neuromyelitis optica: a demyelinating disease characterized by acute destruction and regeneration of perivascular astrocytes. Mult Scler 2010; 16:1156-72. [PMID: 20823059 DOI: 10.1177/1352458510382324] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND A serum antibody directed against astrocytes is present in a high proportion of patients with neuromyelitis optica (NMO). The pathogenicity of the antibody is uncertain because no consistent astrocyte lesion is known to occur in NMO. OBJECTIVE To determine whether there is an astrocyte lesion in NMO and if this differs from astrocyte changes in multiple sclerosis (MS). METHODS Astrocyte pathology in early (still-myelinated) lesions and subacute NMO and MS lesions was examined immunohistochemically and in sections stained for astrocytes using routine histological techniques. RESULTS Demyelination in early NMO lesions is accompanied by oligodendrocyte apoptosis in a pattern identical to that seen in MS and this is preceded by an abrupt destruction of perivascular astrocytes. Reparative astrogliosis is effected by a population of unipolar, new astrocytes. Evidence of a different type of astrocyte lesion was found in MS. DISCUSSION The findings add to experimental evidence that the antibody is pathogenic. They also raise the possibility that demyelination in MS may be a bystander effect of an astrocyte lesion, i.e. that MS is not a disease primarily of myelin and oligodendrocytes.
Collapse
Affiliation(s)
- John D E Parratt
- The Institute of Clinical Neurosciences, Department of Medicine, The University of Sydney, NSW, Australia
| | | |
Collapse
|
50
|
Cid C, Alcazar A. Protection of oligodendrocyte precursor cells by low doses of HSP90 inhibitors in cell culture. Exp Neurol 2010; 225:29-33. [DOI: 10.1016/j.expneurol.2009.11.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 11/04/2009] [Accepted: 11/21/2009] [Indexed: 01/26/2023]
|