1
|
Hong X, Chen T, Liu Y, Li J, Huang D, Ye K, Liao W, Wang Y, Liu M, Luan P. Design, current states, and challenges of nanomaterials in anti-neuroinflammation: A perspective on Alzheimer's disease. Ageing Res Rev 2025; 105:102669. [PMID: 39864562 DOI: 10.1016/j.arr.2025.102669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Alzheimer's disease (AD), an age-related neurodegenerative disease, brings huge damage to the society, to the whole family and even to the patient himself. However, until now, the etiological factor of AD is still unknown and there is no effective treatment for it. Massive deposition of amyloid-beta peptide(Aβ) and hyperphosphorylation of Tau proteins are acknowledged pathological features of AD. Recent studies have revealed that neuroinflammation plays a pivotal role in the pathology of AD. With the rise of nanomaterials in the biomedical field, researchers are exploring how the unique properties of these materials can be leveraged to develop effective treatments for AD. This article has summarized the influence of neuroinflammation in AD, the design of nanoplatforms, and the current research status and inadequacy of nanomaterials in improving neuroinflammation in AD.
Collapse
Affiliation(s)
- Xinyang Hong
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Yunyun Liu
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Neurology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jun Li
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Dongqing Huang
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Kaiyu Ye
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Wanchen Liao
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Yulin Wang
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Mengling Liu
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Ping Luan
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China; School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
2
|
Asante I, Louie S, Yassine HN. Uncovering mechanisms of brain inflammation in Alzheimer's disease with APOE4: Application of single cell-type lipidomics. Ann N Y Acad Sci 2022; 1518:84-105. [PMID: 36200578 PMCID: PMC10092192 DOI: 10.1111/nyas.14907] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A chronic state of unresolved inflammation in Alzheimer's disease (AD) is intrinsically involved with the remodeling of brain lipids. This review highlights the effect of carrying the apolipoprotein E ε4 allele (APOE4) on various brain cell types in promoting an unresolved inflammatory state. Among its pleotropic effects on brain lipids, we focus on APOE4's activation of Ca2+ -dependent phospholipase A2 (cPLA2) and its effects on arachidonic acid, eicosapentaenoic acid, and docosahexaenoic acid signaling cascades in the brain. During the process of neurodegeneration, various brain cell types, such as astrocytes, microglia, and neurons, together with the neurovascular unit, develop distinct inflammatory phenotypes that impact their functions and have characteristic lipidomic fingerprints. We propose that lipidomic phenotyping of single cell-types harvested from brains differing by age, sex, disease severity stage, and dietary and genetic backgrounds can be employed to probe mechanisms of neurodegeneration. A better understanding of the brain cellular inflammatory/lipidomic response promises to guide the development of nutritional and drug interventions for AD dementia.
Collapse
Affiliation(s)
- Isaac Asante
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Stan Louie
- School of Pharmacy, University of Southern California, Los Angeles, California, USA
| | - Hussein N Yassine
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
3
|
Cheng N, Xiong Y, Zhang W, Wu X, Sun Z, Zhang L, Wu H, Tang Y, Peng Y. Astrocytes promote the proliferation of oligodendrocyte precursor cells through connexin 47-mediated LAMB2 secretion in exosomes. Mol Biol Rep 2022; 49:7263-7273. [PMID: 35596050 DOI: 10.1007/s11033-022-07508-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/22/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Oligodendrocyte precursor cells (OPCs) can proliferate and differentiate into oligodendrocytes, the only myelin-forming cells in the central nervous system. Proliferating OPCs promotes remyelination in neurodegenerative diseases. Astrocytes (ASTs) are the most widespread cells in the brain and play a beneficial role in the proliferation of OPCs. Connexin 47 (Cx47) is the main component of AST-OPC gap junctions to regulate OPC proliferation. Nonetheless, the specific mechanism remains unclear. METHODS AND RESULTS This study investigates the proliferation mechanism of OPCs connected to ASTs via Cx47. Cx47 siRNA significantly inhibited OPCs from entering the proliferation cycle. Transcriptome sequencing of OPCs and gene ontology enrichment analysis revealed that ASTs enhanced the exosome secretion by OPCs via Cx47. Transmission electron microscopy, Western blot, and nanoparticle tracking analysis indicated that the OPC proliferation was related to extracellular exosomes. Cx47 siRNA decreased the OPC proliferation and exosome secretion in AST-OPC cocultures. Exogenous exosome supplementation alleviated the inhibitory effect of Cx47 siRNA and significantly improved OPC proliferation. Mass spectrometry revealed that LAMB2 was abundant in exosomes. The administration of exogenous LAMB2 induced DNA replication in the S phase in OPCs by activating cyclin D1. CONCLUSIONS Collectively, ASTs induce the secretion of exosomes that carry LAMB2 by OPCs via Cx47 to upregulate cyclin D1 thereby accelerating OPC proliferation.
Collapse
Affiliation(s)
- Nannan Cheng
- Laboratory of Tissue Engineering and Stem Cell, Department of Histology and Embryology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yuanfeng Xiong
- Laboratory of Tissue Engineering and Stem Cell, Department of Histology and Embryology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Wenjin Zhang
- Laboratory of Tissue Engineering and Stem Cell, Department of Histology and Embryology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xiaohong Wu
- Laboratory of Tissue Engineering and Stem Cell, Department of Histology and Embryology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhongxiang Sun
- Laboratory of Tissue Engineering and Stem Cell, Department of Histology and Embryology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Lei Zhang
- Laboratory of Tissue Engineering and Stem Cell, Department of Histology and Embryology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Hong Wu
- Laboratory of Tissue Engineering and Stem Cell, Department of Histology and Embryology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yong Tang
- Laboratory of Tissue Engineering and Stem Cell, Department of Histology and Embryology, Chongqing Medical University, Chongqing, People's Republic of China.
| | - Yan Peng
- Laboratory of Tissue Engineering and Stem Cell, Department of Histology and Embryology, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
4
|
De Nuccio C, Bernardo A, Troiano C, Brignone MS, Falchi M, Greco A, Rosini M, Basagni F, Lanni C, Serafini MM, Minghetti L, Visentin S. NRF2 and PPAR-γ Pathways in Oligodendrocyte Progenitors: Focus on ROS Protection, Mitochondrial Biogenesis and Promotion of Cell Differentiation. Int J Mol Sci 2020; 21:E7216. [PMID: 33003644 PMCID: PMC7583077 DOI: 10.3390/ijms21197216] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/18/2020] [Accepted: 09/26/2020] [Indexed: 12/15/2022] Open
Abstract
An adequate protection from oxidative and inflammatory reactions, together with the promotion of oligodendrocyte progenitor (OP) differentiation, is needed to recover from myelin damage in demyelinating diseases. Mitochondria are targets of inflammatory and oxidative insults and are essential in oligodendrocyte differentiation. It is known that nuclear factor-erythroid 2-related factor/antioxidant responsive element (NRF2/ARE) and peroxisome proliferator-activated receptor gamma/PPAR-γ response element (PPAR-γ/PPRE) pathways control inflammation and overcome mitochondrial impairment. In this study, we analyzed the effects of activators of these pathways on mitochondrial features, protection from inflammatory/mitochondrial insults and cell differentiation in OP cultures, to depict the specificities and similarities of their actions. We used dimethyl-fumarate (DMF) and pioglitazone (pio) as agents activating NRF2 and PPAR-γ, respectively, and two synthetic hybrids acting differently on the NRF2/ARE pathway. Only DMF and compound 1 caused early effects on the mitochondria. Both DMF and pio induced mitochondrial biogenesis but different antioxidant repertoires. Moreover, pio induced OP differentiation more efficiently than DMF. Finally, DMF, pio and compound 1 protected from tumor necrosis factor-alpha (TNF-α) insult, with pio showing faster kinetics of action and compound 1 a higher activity than DMF. In conclusion, NRF2 and PPAR-γ by inducing partially overlapping pathways accomplish complementary functions aimed at the preservation of mitochondrial function, the defense against oxidative stress and the promotion of OP differentiation.
Collapse
Affiliation(s)
- Chiara De Nuccio
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.D.N.); (L.M.)
| | - Antonietta Bernardo
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (A.G.)
| | - Carmen Troiano
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | | | - Mario Falchi
- National Research Center on HIV/AIDS, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Anita Greco
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (A.G.)
| | - Michela Rosini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (M.R.); (F.B.)
| | - Filippo Basagni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (M.R.); (F.B.)
| | - Cristina Lanni
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (C.L.); (M.M.S.)
| | | | - Luisa Minghetti
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.D.N.); (L.M.)
| | - Sergio Visentin
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (A.G.)
| |
Collapse
|
5
|
Duffy CP, McCoy CE. The Role of MicroRNAs in Repair Processes in Multiple Sclerosis. Cells 2020; 9:cells9071711. [PMID: 32708794 PMCID: PMC7408558 DOI: 10.3390/cells9071711] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disorder characterised by demyelination of central nervous system neurons with subsequent damage, cell death and disability. While mechanisms exist in the CNS to repair this damage, they are disrupted in MS and currently there are no treatments to address this deficit. In recent years, increasing attention has been paid to the influence of the small, non-coding RNA molecules, microRNAs (miRNAs), in autoimmune disorders, including MS. In this review, we examine the role of miRNAs in remyelination in the different cell types that contribute to MS. We focus on key miRNAs that have a central role in mediating the repair process, along with several more that play either secondary or inhibitory roles in one or more aspects. Finally, we consider the current state of miRNAs as therapeutic targets in MS, acknowledging current challenges and potential strategies to overcome them in developing effective novel therapeutics to enhance repair mechanisms in MS.
Collapse
|
6
|
Evolutionary genomics analysis of human nucleus-encoded mitochondrial genes: implications for the roles of energy production and metabolic pathways in the pathogenesis and pathophysiology of demyelinating diseases. Neurosci Lett 2019; 715:134600. [PMID: 31726178 DOI: 10.1016/j.neulet.2019.134600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/19/2019] [Accepted: 10/28/2019] [Indexed: 02/02/2023]
Abstract
The myelin sheath is a plasma membrane extension that lines nerve fibers to protect, support and insulate neurons. The myelination of axons in vertebrates enables fast, saltatory impulse propagation, and this process relies on organelles, especially on mitochondria to supply energy. Approximately 99% of mitochondrial proteins are encoded in the nucleus. Since mitochondria play a central role in the energy production and metabolic pathways, which are essential for myelinogenesis, studying these nucleus-encoded genes (nMGs) may provide new insights into the roles of energy metabolism in demyelinating diseases. In this work, a multiomics-based approach was employed to 1) construct a 1,740 human nMG subset with mitochondrial localization evidence obtained from the Integrated Mitochondrial Protein Index (IMPI) and MitoCarta databases, 2) conduct an evolutionary genomics analysis across mouse, rat, monkey, chimp, and human models, 3) examine dysmyelination phenotype-related genes (nMG subset genes with oligodendrocyte- and myelin-related phenotypes, OMP-nMGs) in MGI mouse lines and human patients, 4) determine the expression discrepancy of OMP-nMGs in brain tissues of cuprizone-treated mice, multiple sclerosis patients, and normal controls, and 5) conduct literature data mining to explore OMP-nMG-associated disease impacts. By contrasting OMP-nMGs with other genes, OMP-nMGs were found to be more ubiquitously expressed (59.1% vs. 16.1%), disease-associated (67.3% vs. 20.2%), and evolutionarily conserved within the human populations. Our multiomics-based analysis identified 110 OMP-nMGs implicated in energy production and lipid and glycan biosynthesis in the pathogenesis and pathophysiology of demyelinating disorders. Future targeted characterization of OMP-nMGs in abnormal myelination conditions may allow the discovery of novel nMG mediated mechanisms underlying myelinogenesis and related diseases.
Collapse
|
7
|
Rafiee Zadeh A, Ghadimi K, Mohammadi B, Hatamian H, Naghibi SN, Danaeiniya A. Effects of Estrogen and Progesterone on Different Immune Cells Related to Multiple Sclerosis. CASPIAN JOURNAL OF NEUROLOGICAL SCIENCES 2018. [DOI: 10.29252/cjns.4.13.83] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
8
|
Bansal R, Singh R. Exploring the potential of natural and synthetic neuroprotective steroids against neurodegenerative disorders: A literature review. Med Res Rev 2017; 38:1126-1158. [PMID: 28697282 DOI: 10.1002/med.21458] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022]
Abstract
Neurodegeneration is a complex process, which leads to progressive brain damage due to loss of neurons. Despite exhaustive research, the cause of neuronal loss in various degenerative disorders is not entirely understood. Neuroprotective steroids constitute an important line of attack, which could play a major role against the common mechanisms associated with various neurodegenerative disorders like Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Natural endogenous steroids induce the neuroprotection by protecting the nerve cells from neuronal injury through multiple mechanisms, therefore the structural modifications of the endogenous steroids could be helpful in the generation of new therapeutically useful neuroprotective agents. The review article will keep the readers apprised of the detailed description of natural as well as synthetic neuroprotective steroids from the medicinal chemistry point of view, which would be helpful in drug discovery efforts aimed toward neurodegenerative diseases.
Collapse
Affiliation(s)
- Ranju Bansal
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Ranjit Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
9
|
Sirt1 regulates glial progenitor proliferation and regeneration in white matter after neonatal brain injury. Nat Commun 2016; 7:13866. [PMID: 27991597 PMCID: PMC5187440 DOI: 10.1038/ncomms13866] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/07/2016] [Indexed: 11/15/2022] Open
Abstract
Regenerative processes in brain pathologies require the production of distinct neural cell populations from endogenous progenitor cells. We have previously demonstrated that oligodendrocyte progenitor cell (OPC) proliferation is crucial for oligodendrocyte (OL) regeneration in a mouse model of neonatal hypoxia (HX) that reproduces diffuse white matter injury (DWMI) of premature infants. Here we identify the histone deacetylase Sirt1 as a Cdk2 regulator in OPC proliferation and response to HX. HX enhances Sirt1 and Sirt1/Cdk2 complex formation through HIF1α activation. Sirt1 deacetylates retinoblastoma (Rb) in the Rb/E2F1 complex, leading to dissociation of E2F1 and enhanced OPC proliferation. Sirt1 knockdown in culture and its targeted ablation in vivo suppresses basal and HX-induced OPC proliferation. Inhibition of Sirt1 also promotes OPC differentiation after HX. Our results indicate that Sirt1 is an essential regulator of OPC proliferation and OL regeneration after neonatal brain injury. Therefore, enhancing Sirt1 activity may promote OL recovery after DWMI.
Oligodendrocyte progenitor cell (OPC) proliferation is crucial for regeneration after hypoxic lesions in mice, a model of diffuse white matter injury of premature infants. Here, the authors show that the histone deacetylase Sirt1 is a Cdk2-dependent mediator of OPC proliferation and OPC response to hypoxia.
Collapse
|
10
|
Bistolfi F. Extremely Low-Frequency Pulsed Magnetic Fields and Multiple Sclerosis: Effects on Neurotransmission Alone or Also on Immunomodulation? Building a Working Hypothesis. Neuroradiol J 2016; 20:676-93. [DOI: 10.1177/197140090702000612] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2007] [Accepted: 09/17/2007] [Indexed: 11/15/2022] Open
Abstract
This paper outlines the current state of knowledge on the pathology and treatment of multiple sclerosis (MS) and critically analyses the vast clinical experience of Sandyk in the use of pulsed magnetic fields of 5 Hz at 7.5 pT to treat many symptoms of MS. A complete regression of symptoms, or at least a major improvement, is sometimes so rapid as to suggest that ELF fields exert a greater effect on axonal and synaptic neurotransmission than on the processes leading to demyelination. Pulsed magnetic fields of 50–100 Hz and a few mT (whose flux intensity is 109 times greater than that of the fields used by Sandyk) have been seen to induce profound morphological changes (the Marinozzi effect) in the plasma membrane of several cell types, including Raji human lymphoblastoid cells. These observations underlie the author's hypothesis on the possible use of such fields in the treatment of MS. Indeed, these fields should induce the functional arrest of the cells (B- and T-lymphocytes, macrophages, microglia, dendritic cells) of the MS plaque, thereby providing an ‘electromagnetic immunomodulatory boost’ to the effects of drug therapy. To test this working hypothesis, it is suggested that preliminary experimental research be carried out to ascertain: 1) the Marinozzi effect in vivo; 2) the Marinozzi effect on microglia and dendritic cells; and 3) the duration of the membrane changes and their relaxation rate. ELF magnetic fields in the picotesla and millitesla ranges are aimed at improving neurotransmission and correcting local immune pathology, respectively. Both types of field might find application in the treatment of MS patients who no longer respond to or tolerate currently used drugs.
Collapse
Affiliation(s)
- F. Bistolfi
- Radiotherapy Department, Galliera Hospital, Genoa, Italy
| |
Collapse
|
11
|
Lopez Juarez A, He D, Richard Lu Q. Oligodendrocyte progenitor programming and reprogramming: Toward myelin regeneration. Brain Res 2016; 1638:209-220. [PMID: 26546966 PMCID: PMC5119932 DOI: 10.1016/j.brainres.2015.10.051] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 10/05/2015] [Accepted: 10/27/2015] [Indexed: 01/26/2023]
Abstract
Demyelinating diseases such as multiple sclerosis (MS) are among the most disabling and cost-intensive neurological disorders. The loss of myelin in the central nervous system, produced by oligodendrocytes (OLs), impairs saltatory nerve conduction, leading to motor and cognitive deficits. Immunosuppression therapy has a limited efficacy in MS patients, arguing for a paradigm shift to strategies that target OL lineage cells to achieve myelin repair. The inhibitory microenvironment in MS lesions abrogates the expansion and differentiation of resident OL precursor cells (OPCs) into mature myelin-forming OLs. Recent studies indicate that OPCs display a highly plastic ability to differentiate into alternative cell lineages under certain circumstances. Thus, understanding the mechanisms that maintain and control OPC fate and differentiation into mature OLs in a hostile, non-permissive lesion environment may open new opportunities for regenerative therapies. In this review, we will focus on 1) the plasticity of OPCs in terms of their developmental origins, distribution, and differentiation potentials in the normal and injured brain; 2) recent discoveries of extrinsic and intrinsic factors and small molecule compounds that control OPC specification and differentiation; and 3) therapeutic potential for motivation of neural progenitor cells and reprogramming of differentiated cells into OPCs and their likely impacts on remyelination. OL-based therapies through activating regenerative potentials of OPCs or cell replacement offer exciting opportunities for innovative strategies to promote remyelination and neuroprotection in devastating demyelinating diseases like MS. This article is part of a Special Issue entitled SI:NG2-glia(Invited only).
Collapse
Affiliation(s)
- Alejandro Lopez Juarez
- Department of Pediatrics, Divisions of Experimental Hematology and Cancer Biology & Developmental Biology, Cincinnati Children׳s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Danyang He
- Department of Pediatrics, Divisions of Experimental Hematology and Cancer Biology & Developmental Biology, Cincinnati Children׳s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Q Richard Lu
- Department of Pediatrics, Divisions of Experimental Hematology and Cancer Biology & Developmental Biology, Cincinnati Children׳s Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
12
|
Alizadeh A, Karimi-Abdolrezaee S. Microenvironmental regulation of oligodendrocyte replacement and remyelination in spinal cord injury. J Physiol 2016; 594:3539-52. [PMID: 26857216 DOI: 10.1113/jp270895] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/26/2015] [Indexed: 01/29/2023] Open
Abstract
Myelin is a proteolipid sheath enwrapping axons in the nervous system that facilitates signal transduction along the axons. In the central nervous system (CNS), oligodendrocytes are specialized glial cells responsible for myelin formation and maintenance. Following spinal cord injury (SCI), oligodendroglia cell death and myelin damage (demyelination) cause chronic axonal damage and irreparable loss of sensory and motor functions. Accumulating evidence shows that replacement of damaged oligodendrocytes and renewal of myelin (remyelination) are promising approaches to prevent axonal degeneration and restore function following SCI. Neural precursor cells (NPCs) and oligodendrocyte progenitor cells (OPCs) are two main resident cell populations in the spinal cord with innate capacities to foster endogenous oligodendrocyte replacement and remyelination. However, due to the hostile microenvironment of SCI, the regenerative capacity of these endogenous precursor cells is conspicuously restricted. Activated resident glia, along with infiltrating immune cells, are among the key modulators of secondary injury mechanisms that create a milieu impermissible to oligodendrocyte differentiation and remyelination. Recent studies have uncovered inhibitory roles for astrocyte-associated molecules such as matrix chondroitin sulfate proteoglycans (CSPGs), and a plethora of pro-inflammatory cytokines and neurotoxic factors produced by activated microglia/macrophages. The quality of axonal remyelination is additionally challenged by dysregulation of the supportive growth factors required for maturation of new oligodendrocytes and axo-oligodendrocyte signalling. Careful understanding of factors that modulate the activity of endogenous precursor cells in the injury microenvironment is a key step in developing efficient repair strategies for remyelination and functional recovery following SCI.
Collapse
Affiliation(s)
- Arsalan Alizadeh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
13
|
Quantification of [(11)C]PIB PET for imaging myelin in the human brain: a test-retest reproducibility study in high-resolution research tomography. J Cereb Blood Flow Metab 2015; 35:1771-82. [PMID: 26058700 PMCID: PMC4635232 DOI: 10.1038/jcbfm.2015.120] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 04/01/2015] [Accepted: 05/05/2015] [Indexed: 01/12/2023]
Abstract
An accurate in vivo measure of myelin content is essential to deepen our insight into the mechanisms underlying demyelinating and dysmyelinating neurological disorders, and to evaluate the effects of emerging remyelinating treatments. Recently [(11)C]PIB, a positron emission tomography (PET) tracer originally conceived as a beta-amyloid marker, has been shown to be sensitive to myelin changes in preclinical models and humans. In this work, we propose a reference-region methodology for the voxelwise quantification of brain white-matter (WM) binding for [(11)C]PIB. This methodology consists of a supervised procedure for the automatic extraction of a reference region and the application of the Logan graphical method to generate distribution volume ratio (DVR) maps. This approach was assessed on a test-retest group of 10 healthy volunteers using a high-resolution PET tomograph. The [(11)C]PIB PET tracer binding was shown to be up to 23% higher in WM compared with gray matter, depending on the image reconstruction. The DVR estimates were characterized by high reliability (outliers <1%) and reproducibility (intraclass correlation coefficient (ICC) >0.95). [(11)C]PIB parametric maps were also found to be significantly correlated (R(2)>0.50) to mRNA expressions of the most represented proteins in the myelin sheath. On the contrary, no correlation was found between [(11)C]PIB imaging and nonmyelin-associated proteins.
Collapse
|
14
|
Sedel F, Bernard D, Mock DM, Tourbah A. Targeting demyelination and virtual hypoxia with high-dose biotin as a treatment for progressive multiple sclerosis. Neuropharmacology 2015; 110:644-653. [PMID: 26327679 DOI: 10.1016/j.neuropharm.2015.08.028] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/24/2015] [Accepted: 08/18/2015] [Indexed: 12/30/2022]
Abstract
Progressive multiple sclerosis (MS) is a severely disabling neurological condition, and an effective treatment is urgently needed. Recently, high-dose biotin has emerged as a promising therapy for affected individuals. Initial clinical data have shown that daily doses of biotin of up to 300 mg can improve objective measures of MS-related disability. In this article, we review the biology of biotin and explore the properties of this ubiquitous coenzyme that may explain the encouraging responses seen in patients with progressive MS. The gradual worsening of neurological disability in patients with progressive MS is caused by progressive axonal loss or damage. The triggers for axonal loss in MS likely include both inflammatory demyelination of the myelin sheath and primary neurodegeneration caused by a state of virtual hypoxia within the neuron. Accordingly, targeting both these pathological processes could be effective in the treatment of progressive MS. Biotin is an essential co-factor for five carboxylases involved in fatty acid synthesis and energy production. We hypothesize that high-dose biotin is exerting a therapeutic effect in patients with progressive MS through two different and complementary mechanisms: by promoting axonal remyelination by enhancing myelin production and by reducing axonal hypoxia through enhanced energy production. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
Affiliation(s)
- Frédéric Sedel
- MedDay Pharmaceuticals, ICM-Brain and Spine Institute-IPEPs, Groupe Hospitalier Pitié Salpêtrière, 47 Boulevard de l'Hopital, 75013 Paris, France.
| | - Delphine Bernard
- MedDay Pharmaceuticals, ICM-Brain and Spine Institute-IPEPs, Groupe Hospitalier Pitié Salpêtrière, 47 Boulevard de l'Hopital, 75013 Paris, France.
| | - Donald M Mock
- Department of Biochemistry & Molecular Biology, University of Arkansas for Medical Sciences, 4301 W Markham Street, Little Rock, AR 72205, USA; Department of Pediatrics, University of Arkansas for Medical Sciences, 4301 W Markham Street, Little Rock, AR 72205, USA.
| | - Ayman Tourbah
- Department of Neurology and Faculté de Médecine de Reims, CHU de Reims, URCA, 45 Rue Cognacq Jay, 51092 Reims Cedex, France.
| |
Collapse
|
15
|
Verma V, Samanthapudi K, Raviprakash R. Classic Studies on the Potential of Stem Cell Neuroregeneration. JOURNAL OF THE HISTORY OF THE NEUROSCIENCES 2015; 25:123-141. [PMID: 26308908 DOI: 10.1080/0964704x.2015.1039904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The 1990s and 2000s were the beginning of an exciting time period for developmental neuroscience and neural stem cell research. By better understanding brain plasticity and the birth of new neurons in the adult brain, contrary to established dogma, hope for therapy from devastating neurological diseases was generated. The potential for stem cells to provide functional recovery in humans remains to be further tested and to further move into the clinical trial realm. The future certainly has great promise on stem cells to assist in alleviation of difficult-to-treat neurologic disorders. This article reviews classic studies of the 1990s and 2000s that paved the way for the advances of today, which can in turn lead to tomorrow's therapies.
Collapse
Affiliation(s)
- Vivek Verma
- a Department of Neuroscience , University of Pittsburgh , Pittsburgh , PA , USA
| | | | - Ratujit Raviprakash
- a Department of Neuroscience , University of Pittsburgh , Pittsburgh , PA , USA
| |
Collapse
|
16
|
Abstract
Extensive evidence has indicated that the breakdown of myelin is associated with Alzheimer's disease (AD) since the vulnerability of oligodendrocytes under Alzheimer's pathology easily induces the myelin breakdown and the loss of the myelin sheath which might be the initiating step in the changes of the earliest stage of AD prior to appearance of amyloid and tau pathology. Considerable research implicated that beta-amyloid (Aβ)-mediated oligodendrocyte dysfunction and myelin breakdown may be via neuroinflammation, oxidative stress and/or apoptosis. It also seems that the oligodendrocyte dysfunction is triggered by the formation of neurofibrillary tangles (NFTs) through inflammation and oxidative stress as the common pathophysiological base. Impaired repair of oligodendrocyte precursor cells (OPCs) might possibly enhance the disease progress under decreased self-healing ability from aging process and pathological factors including Aβ pathology and/or NFTs. Thus, these results have suggested that targeting oligodendrocytes may be a novel therapeutic intervention for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Zhiyou Cai
- a Department of Neurology, Renmin Hospital , Hubei University of Medicine, Shiyan Renmin Hospital , Shiyan , Hubei Province , China
| | - Ming Xiao
- b Department of Anatomy , Nanjing Medical University , Nanjing , Jiangsu , China
| |
Collapse
|
17
|
Blaber EA, Dvorochkin N, Torres ML, Yousuf R, Burns BP, Globus RK, Almeida EAC. Mechanical unloading of bone in microgravity reduces mesenchymal and hematopoietic stem cell-mediated tissue regeneration. Stem Cell Res 2014; 13:181-201. [PMID: 25011075 DOI: 10.1016/j.scr.2014.05.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 05/30/2014] [Accepted: 05/31/2014] [Indexed: 12/12/2022] Open
Abstract
Mechanical loading of mammalian tissues is a potent promoter of tissue growth and regeneration, whilst unloading in microgravity can cause reduced tissue regeneration, possibly through effects on stem cell tissue progenitors. To test the specific hypothesis that mechanical unloading alters differentiation of bone marrow mesenchymal and hematopoietic stem cell lineages, we studied cellular and molecular aspects of how bone marrow in the mouse proximal femur responds to unloading in microgravity. Trabecular and cortical endosteal bone surfaces in the femoral head underwent significant bone resorption in microgravity, enlarging the marrow cavity. Cells isolated from the femoral head marrow compartment showed significant down-regulation of gene expression markers for early mesenchymal and hematopoietic differentiation, including FUT1(-6.72), CSF2(-3.30), CD90(-3.33), PTPRC(-2.79), and GDF15(-2.45), but not stem cell markers, such as SOX2. At the cellular level, in situ histological analysis revealed decreased megakaryocyte numbers whilst erythrocytes were increased 2.33 fold. Furthermore, erythrocytes displayed elevated fucosylation and clustering adjacent to sinuses forming the marrow-blood barrier, possibly providing a mechanistic basis for explaining spaceflight anemia. Culture of isolated bone marrow cells immediately after microgravity exposure increased the marrow progenitor's potential for mesenchymal differentiation into in-vitro mineralized bone nodules, and hematopoietic differentiation into osteoclasts, suggesting an accumulation of undifferentiated progenitors during exposure to microgravity. These results support the idea that mechanical unloading of mammalian tissues in microgravity is a strong inhibitor of tissue growth and regeneration mechanisms, acting at the level of early mesenchymal and hematopoietic stem cell differentiation.
Collapse
Affiliation(s)
- E A Blaber
- School of Biotechnology and Bimolecular Sciences, University of New South Wales, Sydney, Australia; Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - N Dvorochkin
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - M L Torres
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA; Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - R Yousuf
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - B P Burns
- School of Biotechnology and Bimolecular Sciences, University of New South Wales, Sydney, Australia
| | - R K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - E A C Almeida
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
| |
Collapse
|
18
|
Bernardo A, De Simone R, De Nuccio C, Visentin S, Minghetti L. The nuclear receptor peroxisome proliferator-activated receptor-γ promotes oligodendrocyte differentiation through mechanisms involving mitochondria and oscillatory Ca2+ waves. Biol Chem 2014; 394:1607-14. [PMID: 23770533 DOI: 10.1515/hsz-2013-0152] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 06/13/2013] [Indexed: 11/15/2022]
Abstract
Peroxisome proliferator-activated receptor-γ (PPAR-γ) is one of the most studied nuclear receptor since its identification as a target to treat metabolic and neurological diseases. In addition to exerting anti-inflammatory and neuroprotective effects, PPAR-γ agonists, such as the insulin-sensitizing drug pioglitazone, promote the differentiation of oligodendrocytes (OLs), the myelin-forming cells of the central nervous system (CNS). In addition, PPAR-γ agonists increase OL mitochondrial respiratory chain activity and OL's ability to respond to environmental signals with oscillatory Ca2+ waves. Both OL maturation and oscillatory Ca2+ waves are prevented by the mitochondrial inhibitor rotenone and restored by PPAR-γ agonists, suggesting that PPAR-γ promotes myelination through mechanisms involving mitochondria.
Collapse
|
19
|
Phosphodiesterase 5 inhibition at disease onset prevents experimental autoimmune encephalomyelitis progression through immunoregulatory and neuroprotective actions. Exp Neurol 2014; 251:58-71. [DOI: 10.1016/j.expneurol.2013.10.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/25/2013] [Accepted: 10/30/2013] [Indexed: 12/15/2022]
|
20
|
Yin W, Hu B. Knockdown of Lingo1b protein promotes myelination and oligodendrocyte differentiation in zebrafish. Exp Neurol 2013; 251:72-83. [PMID: 24262204 DOI: 10.1016/j.expneurol.2013.11.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 10/29/2013] [Accepted: 11/08/2013] [Indexed: 12/23/2022]
Abstract
Demyelinating diseases include multiple sclerosis, which is a neurodegenerative disease characterized by immune attacks on the central nervous system (CNS), resulting in myelin sheath damage and axonal loss. Leucine-rich repeat and immunoglobulin domain-containing neurite outgrowth inhibitory protein (Nogo) receptor-interacting protein-1 (LINGO-1) have been identified as a negative regulator of oligodendrocytes differentiation. Targeted LINGO-1 inhibition promotes neuron survival, axon regeneration, oligodendrocyte differentiation, and remyelination in diverse animal models. Although studies in rodent models have extended our understanding of LINGO-1, its roles in neural development and myelination in zebrafish (Danio rerio) are not yet clear. In this study, we cloned the zebrafish homolog of the human LINGO-1 and found that lingo1b regulated myelination and oligodendrocyte differentiation. The expression of lingo1b started 1 (mRNA) and 2 (protein) days post-fertilization (dpf) in the CNS. Morpholino oligonucleotide knockdown of lingo1b resulted in developmental abnormalities, including less dark pigment, small eyes, and a curly spinal cord. The lack of lingo1b enhanced myelination and oligodendrocyte differentiation during embryogenesis. Furthermore, immunohistochemistry and movement analysis showed that lingo1b was involved in the axon development of primary motor neurons. These results suggested that Lingo1b protein functions as a negative regulator of myelination and oligodendrocyte differentiation during zebrafish development.
Collapse
Affiliation(s)
- Wu Yin
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Bing Hu
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
21
|
Li Y, Zhang Y, Han W, Hu F, Qian Y, Chen Q. TRO19622 promotes myelin repair in a rat model of demyelination. Int J Neurosci 2013; 123:810-22. [DOI: 10.3109/00207454.2013.804523] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
22
|
Harauz G, Boggs JM. Myelin management by the 18.5-kDa and 21.5-kDa classic myelin basic protein isoforms. J Neurochem 2013; 125:334-61. [PMID: 23398367 DOI: 10.1111/jnc.12195] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 02/05/2013] [Accepted: 02/05/2013] [Indexed: 12/15/2022]
Abstract
The classic myelin basic protein (MBP) splice isoforms range in nominal molecular mass from 14 to 21.5 kDa, and arise from the gene in the oligodendrocyte lineage (Golli) in maturing oligodendrocytes. The 18.5-kDa isoform that predominates in adult myelin adheres the cytosolic surfaces of oligodendrocyte membranes together, and forms a two-dimensional molecular sieve restricting protein diffusion into compact myelin. However, this protein has additional roles including cytoskeletal assembly and membrane extension, binding to SH3-domains, participation in Fyn-mediated signaling pathways, sequestration of phosphoinositides, and maintenance of calcium homeostasis. Of the diverse post-translational modifications of this isoform, phosphorylation is the most dynamic, and modulates 18.5-kDa MBP's protein-membrane and protein-protein interactions, indicative of a rich repertoire of functions. In developing and mature myelin, phosphorylation can result in microdomain or even nuclear targeting of the protein, supporting the conclusion that 18.5-kDa MBP has significant roles beyond membrane adhesion. The full-length, early-developmental 21.5-kDa splice isoform is predominantly karyophilic due to a non-traditional P-Y nuclear localization signal, with effects such as promotion of oligodendrocyte proliferation. We discuss in vitro and recent in vivo evidence for multifunctionality of these classic basic proteins of myelin, and argue for a systematic evaluation of the temporal and spatial distributions of these protein isoforms, and their modified variants, during oligodendrocyte differentiation.
Collapse
Affiliation(s)
- George Harauz
- Department of Molecular and Cellular Biology, Biophysics Interdepartmental Group and Collaborative Program in Neuroscience, University of Guelph, Guelph, Ontario, Canada.
| | | |
Collapse
|
23
|
Martinez NE, Sato F, Omura S, Minagar A, Alexander JS, Tsunoda I. Immunopathological patterns from EAE and Theiler's virus infection: Is multiple sclerosis a homogenous 1-stage or heterogenous 2-stage disease? PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2013; 20:71-84. [PMID: 22633747 PMCID: PMC3430756 DOI: 10.1016/j.pathophys.2012.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Multiple sclerosis (MS) is a disease which can presents in different clinical courses. The most common form of MS is the relapsing-remitting (RR) course, which in many cases evolves into secondary progressive (SP) disease. Autoimmune models such as experimental autoimmune encephalomyelitis (EAE) have been developed to represent the various clinical forms of MS. These models along with clinico-pathological evidence obtained from MS patients have allowed us to propose '1-stage' and '2-stage' disease theories to explain the transition in the clinical course of MS from RR to SP. Relapses in MS are associated with pro-inflammatory T helper (Th) 1/Th17 immune responses, while remissions are associated with anti-inflammatory Th2/regulatory T (Treg) immune responses. Based on the '1-stage disease' theory, the transition from RR to SP disease occurs when the inflammatory immune response overwhelms the anti-inflammatory immune response. The '2-stage disease' theory proposes that the transition from RR to SP-MS occurs when the Th2 response or some other responses overwhelm the inflammatory response resulting in the sustained production of anti-myelin antibodies, which cause continuing demyelination, neurodegeneration, and axonal loss. The Theiler's virus model is also a 2-stage disease, where axonal degeneration precedes demyelination during the first stage, followed by inflammatory demyelination during the second stage.
Collapse
Affiliation(s)
- Nicholas E Martinez
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, LSU Health, School of Medicine, Shreveport, LA 71130, USA
| | | | | | | | | | | |
Collapse
|
24
|
Li X, Liu X, Cui L, Brunson C, Zhao W, Bhat NR, Zhang N, Wen X. Engineering an in situ crosslinkable hydrogel for enhanced remyelination. FASEB J 2012; 27:1127-36. [PMID: 23239823 DOI: 10.1096/fj.12-211151] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Remyelination has to occur to fully regenerate injured spinal cords or brain tissues. A growing body of evidence has suggested that exogenous cell transplantation is one promising strategy to promote remyelination. However, direct injection of neural stem cells or oligodendrocyte progenitor cells (OPCs) to the lesion site may not be an optimal therapeutic strategy due to poor viability and functionality of transplanted cells resulted from the local hostile tissue environment. The overall objective of this study was to engineer an injectable biocompatible hydrogel system as a supportive niche to provide a regeneration permissive microenvironment for transplanted OPCs to survive, functionally differentiate, and remyelinate central nervous system (CNS) lesions. A highly biocompatible hydrogel, based on thiol-functionalized hyaluronic acid and thiol-functionalized gelatin, which can be crosslinked by poly-(ethylene glycol) diacrylate (PEGDA), was used. These hydrogels were optimized first regarding cell adhesive properties and mechanical properties to best support the growth properties of OPCs in culture. Transplanted OPCs with the hydrogels optimized in vitro exhibited enhanced survival and oligodendrogenic differentiation and were able to remyelinate demyelinated axons inside ethidium bromide (EB) demyelination lesion in adult spinal cord. This study provides a new possible therapeutic approach to treat CNS injuries in which cell therapies may be essential.
Collapse
Affiliation(s)
- Xiaowei Li
- Clemson-MUSC Bioengineering Program, Department of Bioengineering, Clemson University, 68 President St., BEB 313, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kaufmann W, Bolon B, Bradley A, Butt M, Czasch S, Garman RH, George C, Gröters S, Krinke G, Little P, McKay J, Narama I, Rao D, Shibutani M, Sills R. Proliferative and nonproliferative lesions of the rat and mouse central and peripheral nervous systems. Toxicol Pathol 2012; 40:87S-157S. [PMID: 22637737 DOI: 10.1177/0192623312439125] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Harmonization of diagnostic nomenclature used in the pathology analysis of tissues from rodent toxicity studies will enhance the comparability and consistency of data sets from different laboratories worldwide. The INHAND Project (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions in Rats and Mice) is a joint initiative of four major societies of toxicologic pathology to develop a globally recognized nomenclature for proliferative and nonproliferative lesions in rodents. This article recommends standardized terms for classifying changes observed in tissues of the mouse and rat central (CNS) and peripheral (PNS) nervous systems. Sources of material include academic, government, and industrial histopathology databases from around the world. Covered lesions include frequent, spontaneous, and aging-related changes as well as principal toxicant-induced findings. Common artifacts that might be confused with genuine lesions are also illustrated. The neural nomenclature presented in this document is also available electronically on the Internet at the goRENI website (http://www.goreni.org/).
Collapse
|
26
|
Magalon K, Zimmer C, Cayre M, Khaldi J, Bourbon C, Robles I, Tardif G, Viola A, Pruss RM, Bordet T, Durbec P. Olesoxime accelerates myelination and promotes repair in models of demyelination. Ann Neurol 2012; 71:213-26. [PMID: 22367994 DOI: 10.1002/ana.22593] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Multiple sclerosis is a neurodegenerative disease characterized by episodes of immune attack of oligodendrocytes leading to demyelination and progressive functional deficit. One therapeutic strategy to address disease progression could consist in stimulating the spontaneous regenerative process observed in some patients. Myelin regeneration requires endogenous oligodendrocyte progenitor migration and activation of the myelination program at the lesion site. In this study, we have tested the ability of olesoxime, a neuroprotective and neuroregenerative agent, to promote remyelination in the rodent central nervous system in vivo. METHODS The effect of olesoxime on oligodendrocyte progenitor cell (OPC) differentiation and myelin synthesis was tested directly in organotypic slice cultures and OPC-neuron cocultures. Using naive animals and different mouse models of demyelination, we morphologically and functionally assessed the effect of the compound on myelination in vivo. RESULTS Olesoxime accelerated oligodendrocyte maturation and enhanced myelination in vitro and in vivo in naive animals during development and also in the adult brain without affecting oligodendrocyte survival or proliferation. In mouse models of demyelination and remyelination, olesoxime favored the repair process, promoting myelin formation with consequent functional improvement. INTERPRETATION Our observations support the strategy of promoting oligodendrocyte maturation and myelin synthesis to enhance myelin repair and functional recovery. We also provide proof of concept that olesoxime could be useful for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Karine Magalon
- Developmental Biology Institute of Marseille-Luminy, French National Center for Scientific Research Joint Research Unit, Universite de la Mediterranee, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Regulation of Glial Cell Functions by PPAR-gamma Natural and Synthetic Agonists. PPAR Res 2011; 2008:864140. [PMID: 18464925 PMCID: PMC2367430 DOI: 10.1155/2008/864140] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Accepted: 03/12/2008] [Indexed: 11/18/2022] Open
Abstract
In the recent years, the peroxisome proliferator-activated receptor-γ (PPAR-γ), a well known target for type II diabetes treatment, has received an increasing attention for its therapeutic potential in inflammatory and degenerative brain disorders. PPAR-γ agonists, which include naturally occurring compounds (such as long chain fatty acids and the cyclopentenone prostaglandin 15-deoxy Δ12,14 prostaglandin J2), and synthetic agonists (among which the thiazolidinediones and few nonsteroidal anti-inflammatory drugs) have shown anti-inflammatory and protective effects in several experimental models of Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, multiple sclerosis and stroke, as well as in few clinical studies. The pleiotropic effects of PPAR-γ agonists are likely to be mediated by several mechanisms involving anti-inflammatory activities on peripheral immune cells (macrophages and lymphocytes), as well as direct effects on neural cells including cerebral vascular endothelial cells, neurons, and glia. In the present article, we will review the recent findings supporting a major role for PPAR-γ agonists in controlling neuroinflammation and neurodegeneration through their activities on glial cells, with a particular emphasis on microglial cells as major macrophage population of the brain parenchyma and main actors in brain inflammation.
Collapse
|
28
|
Ontaneda D, Rae-Grant AD. Management of acute exacerbations in multiple sclerosis. Ann Indian Acad Neurol 2011; 12:264-72. [PMID: 20182574 PMCID: PMC2824954 DOI: 10.4103/0972-2327.58283] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Revised: 04/10/2009] [Accepted: 07/06/2009] [Indexed: 11/04/2022] Open
Abstract
A key component of multiple sclerosis is the occurrence of episodes of clinical worsening with either new symptoms or an increase in older symptoms over a few days or weeks. These are known as exacerbations of multiple sclerosis. In this review, we summarize the pathophysiology and treatment of exacerbations and describe how they are related to the overall management of this disease.
Collapse
Affiliation(s)
- Daniel Ontaneda
- Neurological Institute, Cleveland Clinic, Cleveland, Ohio, 44195 USA
| | | |
Collapse
|
29
|
Wang T, Wang W, Jia L, Bu X, Wang F, Yao L, Zhang B. An improved method for rapid preparation of oligodendrocyte-specific rabbit polyclonal antibody. BIOTECHNOL BIOPROC E 2011; 16:470-476. [DOI: 10.1007/s12257-010-0361-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
30
|
Stankoff B, Freeman L, Aigrot MS, Chardain A, Dollé F, Williams A, Galanaud D, Armand L, Lehericy S, Lubetzki C, Zalc B, Bottlaender M. Imaging central nervous system myelin by positron emission tomography in multiple sclerosis using [methyl-¹¹C]-2-(4'-methylaminophenyl)- 6-hydroxybenzothiazole. Ann Neurol 2011; 69:673-80. [PMID: 21337603 DOI: 10.1002/ana.22320] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 09/13/2010] [Accepted: 09/17/2010] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Imaging of myelin tracts in vivo would greatly improve the monitoring of demyelinating diseases such as multiple sclerosis (MS). To date, no imaging technique specifically targets demyelination and remyelination. Recently, amyloid markers related to Congo red have been shown to bind to central nervous system (CNS) myelin. Here we questioned whether the thioflavine-T derivative 2-(4'-methylaminophenyl)-6-hydroxybenzothiazole (PIB), which also binds to amyloid plaques, could serve as a myelin marker. METHODS PIB fixation to myelin was studied by fluorescence in the normal and dysmyelinating mouse brain, as well as in the postmortem brain of MS patients. Positron emission tomography (PET) experiments were conducted using [¹¹C]PIB in baboons and in a proof of concept clinical study in 2 MS patients. RESULTS Applied directly on tissue sections or after intraperitoneal injection, PIB stained CNS myelin, and the decrease in the level of fixation paralleled the amount of myelin loss in a dysmyelinating mutant. In normally myelinated areas of postmortem MS brain, demyelinated and remyelinated lesions were clearly distinguishable by the differential intensity of labeling observed with PIB. PET using intravenously injected radiolabeled [¹¹C]PIB imaged CNS myelin in baboons and humans. In MS patients, the dynamic analysis of PET acquisitions allowed quantitative assessment of demyelination. INTERPRETATION PIB could be used as an imaging marker to quantify myelin loss and repair in demyelinating diseases.
Collapse
Affiliation(s)
- Bruno Stankoff
- Inserm, U975, Paris, France; Faculty of Medicine, Pierre and Marie Curie University, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Pickard MR, Barraud P, Chari DM. The transfection of multipotent neural precursor/stem cell transplant populations with magnetic nanoparticles. Biomaterials 2011; 32:2274-84. [PMID: 21193228 DOI: 10.1016/j.biomaterials.2010.12.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 12/01/2010] [Indexed: 01/18/2023]
Abstract
Multipotent neural precursor/stem cells (NPCs) are a major transplant population with key properties to promote repair in several neuropathological conditions. Magnetic nanoparticle (MNP)-based vector systems, in turn, offer a combination of key benefits for cell therapies including (i) safety (ii) delivery of therapeutic biomolecules (DNA/siRNA) enhanceable by 'magnetofection' approaches (iii) magnetic cell targeting of MNP-labelled cells to injury sites and (iv) non-invasive imaging of MNP-labelled transplant populations for cell tracking. However, the applications of the versatile MNP platform for NPC transplantation therapies have received limited attention so far. We have evaluated the potential of MNP vectors for gene transfer to NPCs using a neurosphere culture model system; we also assessed repeat transfection ("multifection") and repeat transfection plus applied magnetic field ("magneto-multifection") strategies [to enhance transfection efficiency]. We show for the first time that MNPs can safely mediate single/combinatorial gene delivery to NPCs. Multifection approaches significantly enhanced transfection with negligible toxicity; no adverse effects were observed on stem cell proliferation/differentiation. "Multifected" NPCs survived and differentiated in 3D neural tissue arrays post-transplantation. Our findings demonstrate that MNPs offer a simple and robust alternative to the viral vector systems currently used widely to transfect neural stem cells in neurobiology/neural transplantation research.
Collapse
Affiliation(s)
- Mark R Pickard
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
| | | | | |
Collapse
|
32
|
Yang Y, Lewis R, Miller RH. Interactions between oligodendrocyte precursors control the onset of CNS myelination. Dev Biol 2011; 350:127-38. [PMID: 21144846 PMCID: PMC3032606 DOI: 10.1016/j.ydbio.2010.11.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 10/29/2010] [Accepted: 11/24/2010] [Indexed: 01/13/2023]
Abstract
The formation of CNS myelin is dependent on the differentiation of oligodendrocyte precursor cells (OPCs) and oligodendrocyte maturation. How the initiation of myelination is regulated is unclear, but it is likely to depend on the development of competence by oligodendrocytes and receptivity by target axons. Here we identify an additional level of control of oligodendrocyte maturation mediated by interactions between the different cellular components of the oligodendrocyte lineage. During development oligodendrocyte precursors mature through a series of stages defined by labeling with monoclonal antibodies A2B5 and O4. Newly differentiated oligodendrocytes begin to express galactocerebroside recognized by O1 antibodies and subsequently mature to myelin basic protein (MBP)-positive cells prior to formation of compact myelin. Using an in vitro brain slice culture system that supports robust myelination, the consequences of ablating cells at different stages of the oligodendrocyte lineage on myelination have been assayed. Elimination of all OPC lineage cells through A2B5+, O4+, and O1+ complement-mediated cell lysis resulted in a delay in development of MBP cells and myelination. Selective elimination of early OPCs (A2B5+) also unexpectedly resulted in delayed MBP expression compared to controls suggesting that early OPCs contribute to the timing of myelination onset. By contrast, elimination of differentiated (O1+) immature oligodendrocytes permanently inhibited the appearance of MBP+ cells suggesting that oligodendrocytes are critical to facilitate the maturation of OPCs. These data illuminate that the presence of intra-lineage feed-forward and feedback cues are important for timely myelination by oligodendrocytes.
Collapse
Affiliation(s)
- Yan Yang
- Department of Neurology, Case Western Reserve University, school of Medicine 10900 Euclid Ave., Cleveland, OH 44106
- Center for Translational Neurosciences, Case Western Reserve University, school of Medicine 10900 Euclid Ave., Cleveland, OH 44106
| | - Rebecca Lewis
- Department of Neurosciences, Case Western Reserve University, school of Medicine 10900 Euclid Ave., Cleveland, OH 44106
- Center for Translational Neurosciences, Case Western Reserve University, school of Medicine 10900 Euclid Ave., Cleveland, OH 44106
| | - Robert H. Miller
- Department of Neurosciences, Case Western Reserve University, school of Medicine 10900 Euclid Ave., Cleveland, OH 44106
- Center for Translational Neurosciences, Case Western Reserve University, school of Medicine 10900 Euclid Ave., Cleveland, OH 44106
| |
Collapse
|
33
|
Hunt DP, Sajic M, Phillips H, Henderson D, Compston A, Smith K, Chandran S. Origins of gliogenic stem cell populations within adult skin and bone marrow. Stem Cells Dev 2010; 19:1055-65. [PMID: 20102260 DOI: 10.1089/scd.2009.0371] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The generation of Schwann cells from precursors within adult skin and bone marrow is of significant clinical interest because of the opportunities for disease modelling and strategies for remyelination. Recent evidence has suggested that glial cells can be generated from (i) mesenchymal stem cells (MSCs) within adult bone marrow and (ii) skin-derived precursor cells (SKPs) within adult skin. However, there is a need to clarify the developmental mechanism whereby such multipotent adult stem cell populations generate glia. We used Wnt1-Cre/Rosa26R(LacZ) and Wnt1-Cre/Rosa26R(YFP) neural crest reporter mice to test the hypothesis that (i) MSCs and (ii) SKPs represent adult gliogenic precursor cells of neural crest origin. We demonstrate that, although labeled cells can be identified within long bone preparation, such cells are rarely found in marrow plugs. Moreover, we did not find evidence of a neural crest origin of bone marrow-derived MSCs and were not able to provide a developmental rationale for the derivation of glial cells from MSCs using this approach. In contrast, we provide robust evidence for the neural crest origin of SKPs derived from adult skin. These precursor cells reliably generate cells with a Schwann cell phenotype, expressing appropriate transcription factors and Schwann cell markers. We demonstrate multiple anatomical origins of gliogenic SKPs within adult skin. We conclude that SKPs, rather than bone marrow-derived MSCs, represent a more defined and developmentally rational source for the study and generation of Schwann cells from readily accessible adult tissues.
Collapse
Affiliation(s)
- David P Hunt
- Anne Maclaren Laboratory for Regenerative Medicine, MRC Centre for Stem Cell Biology and Regenerative Medicine, Forvie Site, University of Cambridge , Cambridge, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
34
|
Miron VE, Kuhlmann T, Antel JP. Cells of the oligodendroglial lineage, myelination, and remyelination. Biochim Biophys Acta Mol Basis Dis 2010; 1812:184-93. [PMID: 20887785 DOI: 10.1016/j.bbadis.2010.09.010] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 09/08/2010] [Accepted: 09/20/2010] [Indexed: 12/21/2022]
Abstract
Myelin is critical in maintaining electrical impulse conduction in the central nervous system. The oligodendrocyte is the cell type responsible for myelin production within this compartment. The mutual supply of trophic support between oligodendrocytes and the underlying axons may indicate why demyelinated axons undergo degeneration more readily; the latter contributes to the neural decline in multiple sclerosis (MS). Myelin repair, termed remyelination, occurs in acute inflammatory lesions in MS and is associated with functional recovery and clinical remittances. Animal models have demonstrated that remyelination is mediated by oligodendrocyte progenitor cells (OPCs) which have responded to chemotactic cues, migrated into the lesion, proliferated, differentiated into mature oligodendrocytes, and ensheathed demyelinated axons. The limited remyelination observed in more chronic MS lesions may reflect intrinsic properties of neural cells or extrinsic deterrents. Therapeutic strategies currently under development include transplantation of exogenous OPCs and promotion of remyelination by endogenous OPCs. All currently approved MS therapies are aimed at dampening the immune response and are not directly targeting neural processes.
Collapse
Affiliation(s)
- Veronique E Miron
- Center for Regenerative Medicine, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | | | | |
Collapse
|
35
|
Lin W, Lin Y. Interferon-γ inhibits central nervous system myelination through both STAT1-dependent and STAT1-independent pathways. J Neurosci Res 2010; 88:2569-77. [PMID: 20648647 PMCID: PMC2911948 DOI: 10.1002/jnr.22425] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The immune cytokine interferon-gamma (IFN-gamma) plays a crucial role in immune-mediated demyelinating diseases such as multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Our previous studies have shown that enforced expression of IFN-gamma in the central nervous system (CNS) inhibits developmental myelination or remyelination in EAE demyelinated lesions. Although many of the cellular actions of IFN-gamma result from its activation of the signal transducer and activator of transcription 1 (STAT1) pathway, recent studies have shown that STAT1-independent pathways regulate some facets of IFN-gamma biology. In this study, we dissected the role ofSTAT1-dependent and STAT1-independent pathways in IFN-gamma-induced hypomyelination using a genetic approach. We found that the induction of STAT1-dependent, IFN-gamma-responsive genes in response to this cytokine was abolished in the CNS of STAT1 null mice. Moreover, STAT1 deletion diminished oligodendrocyte loss, reduction of myelinated axons, and the inflammatory response in the CNS of transgenic mice that ectopically expressed IFN-gamma in the CNS. Nevertheless, IFN-gamma-induced reduction of myelin sheath thickness in the CNS of these mice was not altered by STAT1 deletion. Collectively, these data demonstrate that both STAT1-dependent and STAT1-independent pathways are involved in the detrimental effects of IFN-gamma on the myelination process.
Collapse
Affiliation(s)
- Wensheng Lin
- Department of Cell Biology and Neuroscience, University of South Alabama College of Medicine, Mobile, Alabama 36688, USA.
| | | |
Collapse
|
36
|
Linker RA, Lee DH. Models of autoimmune demyelination in the central nervous system: on the way to translational medicine. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2009; 1:5. [PMID: 20142992 PMCID: PMC2816864 DOI: 10.1186/2040-7378-1-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Accepted: 10/21/2009] [Indexed: 11/10/2022]
Abstract
Multiple sclerosis (MS) is the most common neurologic disease of young adults. In the recent years, our understanding on disease pathomechanisms has considerably improved and new therapies have emerged. Yet a cure for this devastating disorder is still a far cry away and human resources on ex vivo specimens are limited. More than 70 years after its first description, experimental autoimmune encephalomyelitis (EAE) remains an important tool to understand concepts of T cell mediated autoimmunity as well as the roles of the innate and the humoral immune systems. Some EAE models also well reflect mechanisms of tissue damage including demyelination, axonal injury and also cortical changes. A limitation of the classical EAE model is a neglect of CD8 T cell mediated immune mechanisms. Moreover, well characterized models for primary progressive MS or demyelination patterns involving primary oligodendrocyte dystrophy are still not available. Yet many current therapeutic concepts including glatiramer acetate or natalizumab stem from their successful first application in EAE models. New strategies include the widespread use of conditional knockout mice to understand the cell-type specific function of single genes, innovative approaches to establish models on the roles of B cells and CD8 T cells as well as on the relation of inflammation to primary degeneration. In summary, EAE models continue to play an important role in neuroimmunology thereby also stimulating research in other fields of the neurosciences and immunobiology.
Collapse
Affiliation(s)
- Ralf A Linker
- Department of Neurology, St Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | | |
Collapse
|
37
|
Wang PS, Wang J, Xiao ZC, Pallen CJ. Protein-tyrosine phosphatase alpha acts as an upstream regulator of Fyn signaling to promote oligodendrocyte differentiation and myelination. J Biol Chem 2009; 284:33692-702. [PMID: 19812040 DOI: 10.1074/jbc.m109.061770] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The tyrosine kinase Fyn plays a key role in oligodendrocyte differentiation and myelination in the central nervous system, but the molecules responsible for regulating Fyn activation in these processes remain poorly defined. Here we show that receptor-like protein-tyrosine phosphatase alpha (PTPalpha) is an important positive regulator of Fyn activation and signaling that is required for the differentiation of oligodendrocyte progenitor cells (OPCs). PTPalpha is expressed in OPCs and is up-regulated during differentiation. We used two model systems to investigate the role of PTPalpha in OPC differentiation: the rat CG4 cell line where PTPalpha expression was silenced by small interfering RNA, and oligosphere-derived primary OPCs isolated from wild-type and PTPalpha-null mouse embryos. In both cell systems, the ablation of PTPalpha inhibited differentiation and morphological changes that accompany this process. Although Fyn was activated upon induction of differentiation, the level of activation was severely reduced in cells lacking PTPalpha, as was the activation of Fyn effector molecules focal adhesion kinase, Rac1, and Cdc42, and inactivation of Rho. Interestingly, another downstream effector of Fyn, p190RhoGAP, which is responsible for Rho inactivation during differentiation, was not affected by PTPalpha ablation. In vivo studies revealed defective myelination in the PTPalpha(-/-) mouse brain. Together, our findings demonstrate that PTPalpha is a critical regulator of Fyn activation and of specific Fyn signaling events during differentiation, and is essential for promoting OPC differentiation and central nervous system myelination.
Collapse
Affiliation(s)
- Pei-Shan Wang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | | | | | | |
Collapse
|
38
|
Peroxisome proliferator-activated receptor-gamma agonists promote differentiation and antioxidant defenses of oligodendrocyte progenitor cells. J Neuropathol Exp Neurol 2009; 68:797-808. [PMID: 19535992 DOI: 10.1097/nen.0b013e3181aba2c1] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Several lines of evidence suggest that peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists may control brain inflammation and, therefore, may be useful for the treatment of human CNS inflammatory conditions. The PPAR-gamma agonists delay the onset and ameliorate clinical manifestations in animal demyelinating disease models, in which the beneficial effects are thought to be mainly related to anti-inflammatory effects on peripheral and brain immune cells. Direct effects on neurons, oligodendrocytes, and other CNS resident cells cannot be excluded, however. To analyze potential direct actions of PPAR-gamma agonists on oligodendrocytes, we investigated the effects of both natural (15-deoxy Delta prostaglandin J2) and synthetic (pioglitazone) PPAR-gamma agonists in primary cultures of rat oligodendrocyte progenitor cells. The PPAR-gamma agonists promoted oligodendrocyte progenitor cell differentiation and enhanced their antioxidant defenses by increasing levels of catalase and copper-zinc superoxide dismutase while maintaining the overall homeostasis of the glutathione system. Protective effects were abolished in the presence of the specific PPAR-gamma antagonist GW9662, indicating that they are specifically dependent on PPAR-gamma. These observations suggest that in addition to their known anti-inflammatory effects, PPAR-gamma agonists may protect oligodendrocyte progenitor cells by preserving their integrity and favoring their differentiation into myelin-forming cells. Thus, PPAR-gamma may promote recovery from demyelination by direct effects on oligodendrocytes.
Collapse
|
39
|
Garay L, Deniselle MCG, Meyer M, Costa JJL, Lima A, Roig P, DeNicola AF. Protective effects of progesterone administration on axonal pathology in mice with experimental autoimmune encephalomyelitis. Brain Res 2009; 1283:177-85. [DOI: 10.1016/j.brainres.2009.04.057] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 04/16/2009] [Accepted: 04/16/2009] [Indexed: 11/24/2022]
|
40
|
Buckley CE, Goldsmith P, Franklin RJM. Zebrafish myelination: a transparent model for remyelination? Dis Model Mech 2009; 1:221-8. [PMID: 19093028 DOI: 10.1242/dmm.001248] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
There is currently an unmet need for a therapy that promotes the regenerative process of remyelination in central nervous system diseases, notably multiple sclerosis (MS). A high-throughput model is, therefore, required to screen potential therapeutic drugs and to refine genomic and proteomic data from MS lesions. Here, we review the value of the zebrafish (Danio rerio) larva as a model of the developmental process of myelination, describing the powerful applications of zebrafish for genetic manipulation and genetic screens, as well as some of the exciting imaging capabilities of this model. Finally, we discuss how a model of zebrafish myelination can be used as a high-throughput screening model to predict the effect of compounds on remyelination. We conclude that zebrafish provide a highly versatile myelination model. As more complex transgenic zebrafish lines are developed, it might soon be possible to visualise myelination, or even remyelination, in real time. However, experimental outputs must be designed carefully for such visual and temporal techniques.
Collapse
Affiliation(s)
- Clare E Buckley
- Department of Veterinary Medicine, University of Cambridge, Cambridge, CB3 0ES, UK
| | | | | |
Collapse
|
41
|
Cellular remyelinating therapy in multiple sclerosis. J Neurol Sci 2009; 276:1-5. [DOI: 10.1016/j.jns.2008.08.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Revised: 08/14/2008] [Accepted: 08/19/2008] [Indexed: 11/19/2022]
|
42
|
Sloane E, Ledeboer A, Seibert W, Coats B, van Strien M, Maier SF, Johnson KW, Chavez R, Watkins LR, Leinwand L, Milligan ED, Van Dam AM. Anti-inflammatory cytokine gene therapy decreases sensory and motor dysfunction in experimental Multiple Sclerosis: MOG-EAE behavioral and anatomical symptom treatment with cytokine gene therapy. Brain Behav Immun 2009; 23:92-100. [PMID: 18835435 PMCID: PMC2631931 DOI: 10.1016/j.bbi.2008.09.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 09/02/2008] [Accepted: 09/03/2008] [Indexed: 11/20/2022] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune inflammatory disease that presents clinically with a range of symptoms including motor, sensory, and cognitive dysfunction as well as demyelination and lesion formation in brain and spinal cord. A variety of animal models of MS have been developed that share many of the pathological hallmarks of MS including motor deficits (ascending paralysis), demyelination and axonal damage of central nervous system (CNS) tissue. In recent years, neuropathic pain has been recognized as a prevalent symptom of MS in a majority of patients. To date, there have been very few investigations into sensory disturbances in animal models of MS. The current work contains the first assessment of hind paw mechanical allodynia (von Frey test) over the course of a relapsing-remitting myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis (MOG-EAE) rat model of MS and establishes the utility of this model in examining autoimmune induced sensory dysfunction. We demonstrate periods of both decreased responsiveness to touch that precedes the onset of hind limb paralysis, and increased responsiveness (allodynia) that occurs during the period of motor deficit amelioration traditionally referred to as symptom remission. Furthermore, we tested the ability of our recently characterized anti-inflammatory IL-10 gene therapy to treat the autoimmune inflammation induced behavioral symptoms and tissue histopathological changes. This therapy is shown here to reverse inflammation induced paralysis, to reduce disease associated reduction in sensitivity to touch, to prevent the onset of allodynia, to reverse disease associated loss of body weight, and to suppress CNS glial activation associated with disease progression in this model.
Collapse
Affiliation(s)
- Evan Sloane
- Department of Psychology & Center for Neuroscience, University of Colorado, CU-Boulder 345, CO 80305, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Remyelination involves reinvesting demyelinated axons with new myelin sheaths. In stark contrast to the situation that follows loss of neurons or axonal damage, remyelination in the CNS can be a highly effective regenerative process. It is mediated by a population of precursor cells called oligodendrocyte precursor cells (OPCs), which are widely distributed throughout the adult CNS. However, despite its efficiency in experimental models and in some clinical diseases, remyelination is often inadequate in demyelinating diseases such as multiple sclerosis (MS), the most common demyelinating disease and a cause of neurological disability in young adults. The failure of remyelination has profound consequences for the health of axons, the progressive and irreversible loss of which accounts for the progressive nature of these diseases. The mechanisms of remyelination therefore provide critical clues for regeneration biologists that help them to determine why remyelination fails in MS and in other demyelinating diseases and how it might be enhanced therapeutically.
Collapse
Affiliation(s)
- Robin J M Franklin
- Department of Veterinary Medicine and Cambridge Centre for Brain Repair, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK.
| | | |
Collapse
|
44
|
Franco-Pons N, Tomàs J, Roig B, Auladell C, Martorell L, Vilella E. Discoidin domain receptor 1, a tyrosine kinase receptor, is upregulated in an experimental model of remyelination and during oligodendrocyte differentiation in vitro. J Mol Neurosci 2008; 38:2-11. [PMID: 18836851 DOI: 10.1007/s12031-008-9151-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Accepted: 09/16/2008] [Indexed: 12/23/2022]
Abstract
The discoidin domain receptor (DDR1) is highly expressed in oligodendrocytes during the neurodevelopmental myelination process and is genetically associated to schizophrenia. In this study, we aimed to further assess the involvement of DDR1 in both remyelination and oligodendrocyte differentiation. In the mouse model of demyelination-remyelination induced by oral administration of cuprizone, in situ hybridization showed an upregulation of the DDR1 gene in three different white matter areas (corpus callosum, dorsal fornix, and external capsule) during the remyelination period. Moreover, real time reverse transcriptase polymerase chain reaction showed that the increase in DDR1 messenger RNA (mRNA) was strongly correlated with the number of DDR1-positive cells in the corpus callosum (Spearman coefficient = 0.987, P = 0.013). Cells positive for DDR1 mRNA were also positive for oligodendrocyte markers (OLIG2, carnosine, and APC) but not for markers of oligodendrocyte precursors (NG2), myelin markers (CNPase), microglia (CD11b), or reactive glia (GFAP). Differentiation of a human oligodendroglial cell line, HOG16, was associated with an increase in mRNA expression of DDR1 and several myelin proteins (MBP and MOBP) but not other proteins (APC and CNPase). Here, we demonstrate that DDR1 is upregulated in vitro and in vivo when oligodendrocyte myelinating machinery is activated. Further studies are needed to identify the specific molecular pathway.
Collapse
Affiliation(s)
- Neus Franco-Pons
- Unitat de Psiquiatria i Psicologia Mèdica, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, C/Sant Llorenç 21, 43201, Reus, Spain
| | | | | | | | | | | |
Collapse
|
45
|
Zhao C, Fancy SPJ, ffrench-Constant C, Franklin RJM. Osteopontin is extensively expressed by macrophages following CNS demyelination but has a redundant role in remyelination. Neurobiol Dis 2008; 31:209-17. [PMID: 18539470 DOI: 10.1016/j.nbd.2008.04.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 04/17/2008] [Accepted: 04/19/2008] [Indexed: 11/19/2022] Open
Abstract
Osteopontin (OPN) is a key immunoregulator in the autoimmune-mediated demyelinating disease multiple sclerosis. OPN may also play a role in the remyelination since it is 1) a ligand for alpha V integrins, several of which regulate the properties of the oligodendrocyte precursor cells (OPCs) primarily responsible for remyelination, and 2) enhances myelin membrane formation in OPC lines. Here we show that OPN is expressed at high levels during remyelination of toxin-induced demyelination. The increased expression is due to mRNA expression in macrophages and follows differences in macrophage responses to demyelination in young and old adult animals. To identify the role of OPN in remyelination focal demyelination was induced in wild-type and OPN(-/-) mice. There was no difference in the rate of remyelination between the two groups indicating that OPN is not a critical component of remyelination.
Collapse
Affiliation(s)
- Chao Zhao
- Department of Veterinary Medicine and Cambridge Centre for Brain Repair, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | | | | | | |
Collapse
|
46
|
Dubois-Dalcq M, Williams A, Stadelmann C, Stankoff B, Zalc B, Lubetzki C. From fish to man: understanding endogenous remyelination in central nervous system demyelinating diseases. Brain 2008; 131:1686-700. [PMID: 18474520 PMCID: PMC2516372 DOI: 10.1093/brain/awn076] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 03/18/2008] [Accepted: 03/26/2008] [Indexed: 11/12/2022] Open
Abstract
In the central nervous system (CNS) of man, evolutionary pressure has preserved some capability for remyelination while axonal regeneration is very limited. In contrast, two efficient programmes of regeneration exist in the adult fish CNS, neurite regrowth and remyelination. The rapidity of CNS remyelination is critical since it not only restores fast conduction of nerve impulses but also maintains axon integrity. If myelin repair fails, axons degenerate, leading to increased disability. In the human CNS demyelinating disease multiple sclerosis (MS), remyelination often takes place in the midst of inflammation. Here, we discuss recent studies that address the innate repair capabilities of the axon-glia unit from fish to man. We propose that expansion of this research field will help find ways to maintain or enhance spontaneous remyelination in man.
Collapse
Affiliation(s)
- Monique Dubois-Dalcq
- National Institute of Neurological Disorders and Stroke, Porter Neuroscience Research Center, Bethesda, MD 20892-3706, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Kulbatski I, Mothe AJ, Parr AM, Kim H, Kang CE, Bozkurt G, Tator CH. Glial precursor cell transplantation therapy for neurotrauma and multiple sclerosis. ACTA ACUST UNITED AC 2008; 43:123-76. [PMID: 18706353 DOI: 10.1016/j.proghi.2008.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2008] [Accepted: 04/07/2008] [Indexed: 12/18/2022]
Abstract
Traumatic injury to the brain or spinal cord and multiple sclerosis (MS) share a common pathophysiology with regard to axonal demyelination. Despite advances in central nervous system (CNS) repair in experimental animal models, adequate functional recovery has yet to be achieved in patients in response to any of the current strategies. Functional recovery is dependent, in large part, upon remyelination of spared or regenerating axons. The mammalian CNS maintains an endogenous reservoir of glial precursor cells (GPCs), capable of generating new oligodendrocytes and astrocytes. These GPCs are upregulated following traumatic or demyelinating lesions, followed by their differentiation into oligodendrocytes. However, this innate response does not adequately promote remyelination. As a result, researchers have been focusing their efforts on harvesting, culturing, characterizing, and transplanting GPCs into injured regions of the adult mammalian CNS in a variety of animal models of CNS trauma or demyelinating disease. The technical and logistic considerations for transplanting GPCs are extensive and crucial for optimizing and maintaining cell survival before and after transplantation, promoting myelination, and tracking the fate of transplanted cells. This is especially true in trials of GPC transplantation in combination with other strategies such as neutralization of inhibitors to axonal regeneration or remyelination. Overall, such studies improve our understanding and approach to developing clinically relevant therapies for axonal remyelination following traumatic brain injury (TBI) or spinal cord injury (SCI) and demyelinating diseases such as MS.
Collapse
Affiliation(s)
- Iris Kulbatski
- Krembil Neuroscience Centre, Toronto Western Research Institute, 399 Bathurst Street, McLaughlin Pavilion #12-423, Toronto, Ontario, Canada M5T-2S8.
| | | | | | | | | | | | | |
Collapse
|
48
|
Cantarella C, Cayre M, Magalon K, Durbec P. Intranasal HB-EGF administration favors adult SVZ cell mobilization to demyelinated lesions in mouse corpus callosum. Dev Neurobiol 2008; 68:223-36. [PMID: 18000828 DOI: 10.1002/dneu.20588] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In the adult rodent brain, the subventricular zone (SVZ) represents a special niche for neural stem cells; these cells proliferate and generate neural progenitors. Most of these migrate along the rostral migratory stream to the olfactory bulb, where they differentiate into interneurons. SVZ-derived progenitors can also be recruited spontaneously to damaged brain areas to replace lost cells, including oligodendrocytes in demyelinated lesions. In this study, we searched for factors able to enhance this spontaneous recruitment of endogenous progenitors. Previous studies have suggested that epidermal growth factor (EGF) could stimulate proliferation, migration, and glial differentiation of SVZ progenitors. In the present study we examined EGF influence on endogenous SVZ cell participation to brain repair in the context of demyelinated lesions. We induced a focal demyelinated lesion in the corpus callosum by lysolecithin injection and showed that intranasal heparin-binding epidermal growth factor (HB-EGF) administration induces a significant increase in SVZ cell proliferation together with a stronger SVZ cell mobilization toward the lesions. Besides, HB-EGF causes a shift of SVZ-derived progenitor cell differentiation toward the astrocytic lineage. However, due to the threefold increase in cell recruitment by EGF treatment, the absolute number of SVZ-derived oligodendrocytes in the lesion of treated mice is higher than in controls. These results suggest that enhancing SVZ cell proliferation could be part of future strategies to promote SVZ progenitor cell mobilization toward brain lesions.
Collapse
Affiliation(s)
- Cristina Cantarella
- Université de la Méditerranée, CNRS-UMR 6216, Institute for Developmental Biology of Marseille-Luminy, Case 907, Campus de Luminy, 13288 Marseille Cedex 9, France
| | | | | | | |
Collapse
|
49
|
Insulin-like growth factor system regulates oligodendroglial cell behavior: therapeutic potential in CNS. J Mol Neurosci 2008; 35:81-90. [PMID: 18299999 DOI: 10.1007/s12031-008-9041-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2008] [Revised: 01/11/2008] [Accepted: 01/13/2008] [Indexed: 12/13/2022]
Abstract
Amongst the many soluble extracellular factors stimulating intracellular signal transduction pathways and driving cellular processes such as proliferation, differentiation and survival, insulin-like growth factors (IGFs) stand out as indispensable factors for proper oligodendrocyte differentiation and accompanying myelin production. Owing to its potent myelinogenic capacity and its neuroprotective properties, IGFs hold therapeutic potential in demyelinating and neurodengenerative diseases. However, the IGF system is comprised of a complex molecular network involving regulatory binding proteins, proteases, cell surface and extracellular matrix components which orchestrate IGF-specific functions. Thus, the complexity by which these factors are tightly regulated makes a simplistic therapeutic approach towards treating demyelinating conditions unfeasible. In the present review, we address these issues and consider current therapeutic prospects of oligodendrocyte-targeted IGF-based therapies.
Collapse
|
50
|
Armstrong RC. Growth factor regulation of remyelination: behind the growing interest in endogenous cell repair of the CNS. FUTURE NEUROLOGY 2007; 2:689-697. [PMID: 19079759 PMCID: PMC2601644 DOI: 10.2217/14796708.2.6.689] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Remyelination facilitates recovery of saltatory conduction along demyelinated axons and may help prevent axon damage in patients with demyelinating diseases, such as multiple sclerosis. The extent of remyelination in multiple sclerosis lesions varies dramatically, indicating a capacity for repair that is not fulfilled in lesions with poor remyelination. In experimental models of demyelinating disease, remyelination is limited by chronic disease that depletes the oligodendrocyte progenitor (OP) population, inhibits OP differentiation into remyelinating oligodendrocytes and/or perturbs cell survival in the lesion environment. Manipulating the activity of growth factor signaling pathways significantly improves the ability of endogenous OP cells to accomplish extensive remyelination. Specifically, growth factors have been identified that can regulate OP proliferation, differentiation and survival in demyelinated lesions. Therefore, growth factors may be key signals for strategies to improve conditions with poor remyelination.
Collapse
Affiliation(s)
- Regina C Armstrong
- Uniformed Services University of the Health Sciences, Department of Anatomy, Physiology & Genetics, 4301 Jones Bridge Rd, Bethesda, MD 20814-4799, USA, Tel.: +1 301 295 3205; ;
| |
Collapse
|