1
|
Bergner CG, van der Meer F, Franz J, Vakrakou A, Würfel T, Nessler S, Schäfer L, Nau-Gietz C, Winkler A, Lagumersindez-Denis N, Wrzos C, Damkou IA, Sergiou C, Schultz V, Knauer C, Metz I, Bahn E, Garea Rodriguez E, Merkler D, Simons M, Stadelmann C. BCAS1-positive oligodendrocytes enable efficient cortical remyelination in multiple sclerosis. Brain 2025; 148:908-920. [PMID: 39319704 PMCID: PMC11884765 DOI: 10.1093/brain/awae293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 09/26/2024] Open
Abstract
Remyelination is a crucial regenerative process in demyelinating diseases, limiting persisting damage to the CNS. It restores saltatory nerve conduction and ensures trophic support of axons. In patients with multiple sclerosis, remyelination has been observed in both white and grey matter and found to be more efficient in the cortex. Brain-enriched myelin-associated protein 1 (BCAS1) identifies oligodendrocyte lineage cells in the stage of active myelin formation in development and regeneration. Other than in the white matter, BCAS1+ oligodendrocytes are maintained at high densities in the cortex throughout life. Here, we investigated cortical lesions in human biopsy and autopsy tissue from patients with multiple sclerosis in direct comparison to demyelinating mouse models and demonstrate that following a demyelinating insult BCAS1+ oligodendrocytes in remyelinating cortical lesions shift from a quiescent to an activated, internode-forming morphology co-expressing myelin-associated glycoprotein (MAG), necessary for axonal contact formation. Of note, activated BCAS1+ oligodendrocytes are found at early time points of experimental demyelination amidst ongoing inflammation. In human tissue, activated BCAS1+ oligodendrocytes correlate with the density of myeloid cells, further supporting their involvement in an immediate regenerative response. Furthermore, studying the microscopically normal appearing non demyelinated cortex in patients with chronic multiple sclerosis, we find a shift from quiescent BCAS1+ oligodendrocytes to mature, myelin-maintaining oligodendrocytes, suggesting oligodendrocyte differentiation and limited replenishment of BCAS1+ oligodendrocytes in long-standing disease. We also demonstrate that part of perineuronal satellite oligodendrocytes are BCAS1+ and contribute to remyelination in human and experimental cortical demyelination. In summary, our results provide evidence from human tissue and experimental models that BCAS1+ cells in the adult cortex represent a population of pre-differentiated oligodendrocytes that rapidly react after a demyelinating insult thus enabling immediate myelin regeneration. In addition, our data suggest that limited replenishment of BCAS1+ oligodendrocytes may contribute to the remyelination failure observed in the cortex in chronic multiple sclerosis.
Collapse
Affiliation(s)
- Caroline Gertrud Bergner
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Department of Neurology, University Hospital Leipzig, Leipzig 04103, Germany
| | - Franziska van der Meer
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Jonas Franz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Aigli Vakrakou
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens 11528, Greece
| | - Thea Würfel
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Stefan Nessler
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Lisa Schäfer
- Department of Neurology, University Hospital Leipzig, Leipzig 04103, Germany
| | - Cora Nau-Gietz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Anne Winkler
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | | | - Claudia Wrzos
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Ioanna Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, Munich 81377, Germany
| | - Christina Sergiou
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Verena Schultz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Carolin Knauer
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Imke Metz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Erik Bahn
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | | | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, Geneva 4 1211, Switzerland
- Division of Clinical Pathology, Geneva University Hospital, Geneva 1205, Switzerland
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich 81377, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich 81377, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich 81377, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Network of Excitable Cells”(MBExC), University of Göttingen, Göttingen 37073, Germany
| |
Collapse
|
2
|
Rida Zainab S, Zeb Khan J, Khalid Tipu M, Jahan F, Irshad N. A review on multiple sclerosis: Unravelling the complexities of pathogenesis, progression, mechanisms and therapeutic innovations. Neuroscience 2025; 567:133-149. [PMID: 39709058 DOI: 10.1016/j.neuroscience.2024.12.029] [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: 10/24/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory demyelinating disorder of the central nervous system (CNS) targeting myelinated axons. Pathogenesis of MS entails an intricate genetic, environmental, and immunological interaction. Dysregulation of immune response i.e. autoreactive T & B-Cells and macrophage infiltration into the CNS leads to inflammation, demyelination, and neurodegeneration. Disease progression of MS varies among individuals transitioning from one form of relapsing-remitting to secondary progressive MS (SPMS). Research advances have unfolded various molecular targets involved in MS from oxidative stress to blood-brain barrier (BBB) disruption. Different pathways are being targeted so far such as inflammatory and cytokine signaling pathways to overcome disease progression. Therapeutic innovations have significantly transformed the management of MS, especially the use of disease-modifying therapies (DMTs) to reduce relapse rates and control disease progression. Advancements in research, neuroprotective strategies, and remyelination strategies hold promising results in reversing CNS damage. Various mice models are being adopted for testing new entities in MS research.
Collapse
Affiliation(s)
- Syeda Rida Zainab
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Jehan Zeb Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Muhammad Khalid Tipu
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Faryal Jahan
- Shifa College of Pharmaceutical Sciences, STMU, Islamabad, Pakistan.
| | - Nadeem Irshad
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
3
|
Chen JQA, Wever DD, McNamara NB, Bourik M, Smolders J, Hamann J, Huitinga I. Inflammatory microglia correlate with impaired oligodendrocyte maturation in multiple sclerosis. Front Immunol 2025; 15:1522381. [PMID: 39877374 PMCID: PMC11772157 DOI: 10.3389/fimmu.2024.1522381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Remyelination of demyelinated axons can occur as an endogenous repair mechanism in multiple sclerosis (MS), but its efficacy varies between both MS individuals and lesions. The molecular and cellular mechanisms that drive remyelination remain poorly understood. Here, we studied the relation between microglia activation and remyelination activity in MS. Methods We correlated regenerative (CD163+) and inflammatory (iNOS+) microglia with BCAS1+ oligodendrocytes, subdivided into early-stage (<3 processes) and late-stage (≥3 processes) cells in brain donors with high or low remyelinating potential in remyelinated lesions and active lesions with ramified/amoeboid (non-foamy) or foamy microglia. A cohort of MS donors categorized as efficiently remyelinating donors (ERDs; n=25) or poorly remyelinating donors (PRDs; n=17) was included, based on their proportion of remyelinated lesions at autopsy. Results and discussion We hypothesized more CD163+ microglia and BCAS1+ oligodendrocytes in remyelinated and active non-foamy lesions from ERDs and more iNOS+ microglia with fewer BCAS1+ oligodendrocytes in active foamy lesions from PRDs. For CD163+ microglia, however, no differences were observed between MS lesions and MS donor groups. In line with our hypothesis, we found that INOS+ microglia were significantly increased in PRDs compared to ERDs within remyelinated lesions. MS lesions, more late-stage BCAS1+ oligodendrocytes were detected in active lesions with non-foamy or foamy microglia in comparison with remyelinated lesions. Although no differences were found for early-stage BCAS1+ oligodendrocytes between MS lesions, we did find significantly more early-stage BCAS1+ oligodendrocytes in PRDs vs ERDs in remyelinated lesions. Interestingly, a positive correlation was identified between iNOS+ microglia and the presence of early-stage BCAS1+ oligodendrocytes. These findings suggest that impaired maturation of early-stage BCAS1+ oligodendrocytes, encountering inflammatory microglia, may underlie remyelination deficits and unsuccessful lesion repair in MS.
Collapse
Affiliation(s)
- J. Q. Alida Chen
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Dennis D. Wever
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Niamh B. McNamara
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Morjana Bourik
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Joost Smolders
- Departments of Neurology and Immunology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Jörg Hamann
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Inge Huitinga
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
4
|
Szekely-Kohn AC, Castellani M, Espino DM, Baronti L, Ahmed Z, Manifold WGK, Douglas M. Machine learning for refining interpretation of magnetic resonance imaging scans in the management of multiple sclerosis: a narrative review. ROYAL SOCIETY OPEN SCIENCE 2025; 12:241052. [PMID: 39845718 PMCID: PMC11750376 DOI: 10.1098/rsos.241052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/23/2024] [Accepted: 11/17/2024] [Indexed: 01/24/2025]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the brain and spinal cord with both inflammatory and neurodegenerative features. Although advances in imaging techniques, particularly magnetic resonance imaging (MRI), have improved the process of diagnosis, its cause is unknown, a cure remains elusive and the evidence base to guide treatment is lacking. Computational techniques like machine learning (ML) have started to be used to understand MS. Published MS MRI-based computational studies can be divided into five categories: automated diagnosis; differentiation between lesion types and/or MS stages; differential diagnosis; monitoring and predicting disease progression; and synthetic MRI dataset generation. Collectively, these approaches show promise in assisting with MS diagnosis, monitoring of disease activity and prediction of future progression, all potentially contributing to disease management. Analysis quality using ML is highly dependent on the dataset size and variability used for training. Wider public access would mean larger datasets for experimentation, resulting in higher-quality analysis, permitting for more conclusive research. This narrative review provides an outline of the fundamentals of MS pathology and pathogenesis, diagnostic techniques and data types in computational analysis, as well as collating literature pertaining to the application of computational techniques to MRI towards developing a better understanding of MS.
Collapse
Affiliation(s)
- Adam C. Szekely-Kohn
- School of Engineering, University of Birmingham, Edgbaston, BirminghamB15 2TT, UK
| | - Marco Castellani
- School of Engineering, University of Birmingham, Edgbaston, BirminghamB15 2TT, UK
| | - Daniel M. Espino
- School of Engineering, University of Birmingham, Edgbaston, BirminghamB15 2TT, UK
| | - Luca Baronti
- School of Computer Science, University of Birmingham, Edgbaston, BirminghamB15 2TT, UK
| | - Zubair Ahmed
- University Hospitals Birmingham NHS Foundation Trust, Edgbaston, BirminghamB15 2GW, UK
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, BirminghamB15 2TT, UK
| | | | - Michael Douglas
- University Hospitals Birmingham NHS Foundation Trust, Edgbaston, BirminghamB15 2GW, UK
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, BirminghamB15 2TT, UK
- Department of Neurology, Dudley Group NHS Foundation Trust, Russells Hall Hospital, BirminghamDY1 2HQ, UK
- School of Life and Health Sciences, Aston University, Birmingham, UK
| |
Collapse
|
5
|
Zveik O, Rechtman A, Ganz T, Vaknin-Dembinsky A. The interplay of inflammation and remyelination: rethinking MS treatment with a focus on oligodendrocyte progenitor cells. Mol Neurodegener 2024; 19:53. [PMID: 38997755 PMCID: PMC11245841 DOI: 10.1186/s13024-024-00742-8] [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: 05/29/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) therapeutic goals have traditionally been dichotomized into two distinct avenues: immune-modulatory-centric interventions and pro-regenerative strategies. Oligodendrocyte progenitor cells (OPCs) were regarded for many years solely in concern to their potential to generate oligodendrocytes and myelin in the central nervous system (CNS). However, accumulating data elucidate the multifaceted roles of OPCs, including their immunomodulatory functions, positioning them as cardinal constituents of the CNS's immune landscape. MAIN BODY In this review, we will discuss how the two therapeutic approaches converge. We present a model by which (1) an inflammation is required for the appropriate pro-myelinating immune function of OPCs in the chronically inflamed CNS, and (2) the immune function of OPCs is crucial for their ability to differentiate and promote remyelination. This model highlights the reciprocal interactions between OPCs' pro-myelinating and immune-modulating functions. Additionally, we review the specific effects of anti- and pro-inflammatory interventions on OPCs, suggesting that immunosuppression adversely affects OPCs' differentiation and immune functions. CONCLUSION We suggest a multi-systemic therapeutic approach, which necessitates not a unidimensional focus but a harmonious balance between OPCs' pro-myelinating and immune-modulatory functions.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel.
| |
Collapse
|
6
|
Zveik O, Rechtman A, Brill L, Vaknin-Dembinsky A. Anti- and pro-inflammatory milieu differentially regulate differentiation and immune functions of oligodendrocyte progenitor cells. Immunology 2024; 171:618-633. [PMID: 38243672 DOI: 10.1111/imm.13757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/05/2024] [Indexed: 01/21/2024] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) were regarded for years solely for their regenerative role; however, their immune-modulatory roles have gained much attention recently, particularly in the context of multiple sclerosis (MS). Despite extensive studies on OPCs, there are limited data elucidating the interactions between their intrinsic regenerative and immune functions, as well as their relationship with the inflamed central nervous system (CNS) environment, a key factor in MS pathology. We examined the effects of pro-inflammatory cytokines, represented by interferon (IFN)-γ and tumour necrosis factor (TNF)-α, as well as anti-inflammatory cytokines, represented by interleukin (IL)-4 and IL-10, on OPC differentiation and immune characteristics. Using primary cultures, enzyme-linked immunosorbent assay and immunofluorescence stainings, we assessed differentiation capacity, phagocytic activity, major histocompatibility complex (MHC)-II expression, and cytokine secretion. We observed that the anti-inflammatory milieu (IL4 and IL10) reduced both OPC differentiation and immune functions. Conversely, exposure to TNF-α led to intact differentiation, increased phagocytic activity, high levels of MHC-II expression, and cytokines secretion. Those effects were attributed to signalling via TNF-receptor-2 and counteracted the detrimental effects of IFN-γ on OPC differentiation. Our findings suggest that a pro-regenerative, permissive inflammatory environment is needed for OPCs to execute both regenerative and immune-modulatory functions.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Livnat Brill
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
7
|
Kreiter D, Postma AA, Hupperts R, Gerlach O. Hallmarks of spinal cord pathology in multiple sclerosis. J Neurol Sci 2024; 456:122846. [PMID: 38142540 DOI: 10.1016/j.jns.2023.122846] [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: 10/19/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023]
Abstract
A disparity exists between spinal cord and brain involvement in multiple sclerosis (MS), each independently contributing to disability. Underlying differences between brain and cord are not just anatomical in nature (volume, white/grey matter organization, vascularization), but also in barrier functions (differences in function and composition of the blood-spinal cord barrier compared to blood-brain barrier) and possibly in repair mechanisms. Also, immunological phenotypes seem to influence localization of inflammatory activity. Whereas the brain has gained a lot of attention in MS research, the spinal cord lags behind. Advanced imaging techniques and biomarkers are improving and providing us with tools to uncover the mechanisms of spinal cord pathology in MS. In the present review, we elaborate on the underlying anatomical and physiological factors driving differences between brain and cord involvement in MS and review current literature on pathophysiology of spinal cord involvement in MS and the observed differences to brain involvement.
Collapse
Affiliation(s)
- Daniel Kreiter
- Academic MS Center Zuyd, Department of Neurology, Zuyderland MC, Sittard-Geleen, the Netherlands; School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands.
| | - Alida A Postma
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Raymond Hupperts
- Academic MS Center Zuyd, Department of Neurology, Zuyderland MC, Sittard-Geleen, the Netherlands; School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Oliver Gerlach
- Academic MS Center Zuyd, Department of Neurology, Zuyderland MC, Sittard-Geleen, the Netherlands; School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
8
|
Subhash S, Chaurawal N, Raza K. Promises of Lipid-Based Nanocarriers for Delivery of Dimethyl Fumarate to Multiple Sclerosis Brain. Methods Mol Biol 2024; 2761:457-475. [PMID: 38427255 DOI: 10.1007/978-1-0716-3662-6_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Multiple sclerosis (MS) is a neurodegenerative autoimmune disorder of the central nervous system (CNS) infecting 2.5 million people worldwide. It is the most common nontraumatic neurological impairment in young adults. The blood-brain barrier rupture for multiple sclerosis pathogenesis has two effects: first, during the onset of the immunological attack, and second, for the CNS self-sustained "inside-out" demyelination and neurodegeneration processes. In addition to genetic variations, environmental and lifestyle variables can also significantly increase the risk of developing MS. Dimethyl fumarate (DMF) and sphingosine-1-phosphate (S1P) receptor modulators that may pass the blood-brain barrier and have positive direct effects in the CNS with quite diverse mechanisms of action raise the possibility that a combination therapy could be successful in treating MS. Lipid nanocarriers are recognized as one of the best drug delivery techniques to the brain for effective brain delivery. Numerous scientific studies have shown that lipid nanoparticles can enhance the lipid solubility, oral bioavailability, and brain availability of the drugs. Nanolipidic carriers for DMF delivery could be derived through vitamin D, tocopherol acetate, stearic acid, quercetin, cell-mimicking platelet-based, and chitosan-alginate core-shell-corona-shaped nanoparticles. Clinical and laboratory diagnosis of MS can be performed mainly through magnetic resonance imaging. The advancements in nanotechnology have enabled the clinicians to cross the blood-brain barrier and to target the brain and central nervous system of the patient with multiple sclerosis.
Collapse
Affiliation(s)
- Sreya Subhash
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, India
| | - Nishtha Chaurawal
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, India
| | - Kaisar Raza
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, India.
| |
Collapse
|
9
|
Ganz T, Zveik O, Fainstein N, Lachish M, Rechtman A, Sofer L, Brill L, Ben-Hur T, Vaknin-Dembinsky A. Oligodendrocyte progenitor cells differentiation induction with MAPK/ERK inhibitor fails to support repair processes in the chronically demyelinated CNS. Glia 2023; 71:2815-2831. [PMID: 37610097 DOI: 10.1002/glia.24453] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023]
Abstract
Remyelination failure is considered a major obstacle in treating chronic-progressive multiple sclerosis (MS). Studies have shown blockage in the differentiation of resident oligodendrocyte progenitor cells (OPC) into myelin-forming cells, suggesting that pushing OPC into a differentiation program might be sufficient to overcome remyelination failure. Others stressed the need for a permissive environment to allow proper activation, migration, and differentiation of OPC. PD0325901, a MAPK/ERK inhibitor, was previously shown to induce OPC differentiation, non-specific immunosuppression, and a significant therapeutic effect in acute demyelinating MS models. We examined PD0325901 effects in the chronically inflamed central nervous system. Treatment with PD0325901 induced OPC differentiation into mature oligodendrocytes with high morphological complexity. However, treatment of Biozzi mice with chronic-progressive experimental autoimmune encephalomyelitis with PD0325901 showed no clinical improvement in comparison to the control group, no reduction in demyelination, nor induction of OPC migration into foci of demyelination. PD0325901 induced a direct general immunosuppressive effect on various cell populations, leading to a diminished phagocytic capability of microglia and less activation of lymph-node cells. It also significantly impaired the immune-modulatory functions of OPC. Our findings suggest OPC regenerative function depends on a permissive environment, which may include pro-regenerative inflammatory elements. Furthermore, they indicate that maintaining a delicate balance between the pro-myelinating and immune functions of OPC is of importance. Thus, the highly complex mission of creating a pro-regenerative environment depends upon an appropriate immune response controlled in time, place, and intensity. We suggest the need to employ a multi-systematic therapeutic approach, which cannot be achieved through a single molecule-based therapy.
Collapse
Affiliation(s)
- Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lihi Sofer
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Livnat Brill
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
10
|
Osanai Y, Yamazaki R, Shinohara Y, Ohno N. Heterogeneity and regulation of oligodendrocyte morphology. Front Cell Dev Biol 2022; 10:1030486. [PMID: 36393856 PMCID: PMC9644283 DOI: 10.3389/fcell.2022.1030486] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/10/2022] [Indexed: 09/24/2023] Open
Abstract
Oligodendrocytes form multiple myelin sheaths in the central nervous system (CNS), which increase nerve conduction velocity and are necessary for basic and higher brain functions such as sensory function, motor control, and learning. Structures of the myelin sheath such as myelin internodal length and myelin thickness regulate nerve conduction. Various parts of the central nervous system exhibit different myelin structures and oligodendrocyte morphologies. Recent studies supported that oligodendrocytes are a heterogenous population of cells and myelin sheaths formed by some oligodendrocytes can be biased to particular groups of axons, and myelin structures are dynamically modulated in certain classes of neurons by specific experiences. Structures of oligodendrocyte/myelin are also affected in pathological conditions such as demyelinating and neuropsychiatric disorders. This review summarizes our understanding of heterogeneity and regulation of oligodendrocyte morphology concerning central nervous system regions, neuronal classes, experiences, diseases, and how oligodendrocytes are optimized to execute central nervous system functions.
Collapse
Affiliation(s)
- Yasuyuki Osanai
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Reiji Yamazaki
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Yoshiaki Shinohara
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
11
|
The Possible Role of Neural Cell Apoptosis in Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23147584. [PMID: 35886931 PMCID: PMC9316123 DOI: 10.3390/ijms23147584] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
The etiology of multiple sclerosis (MS), a demyelinating disease affecting the central nervous system (CNS), remains obscure. Although apoptosis of oligodendrocytes and neurons has been observed in MS lesions, the contribution of this cell death process to disease pathogenesis remains controversial. It is usually considered that MS-associated demyelination and axonal degeneration result from neuroinflammation and an autoimmune process targeting myelin proteins. However, experimental data indicate that oligodendrocyte and/or neuronal cell death may indeed precede the development of inflammation and autoimmunity. These findings raise the question as to whether neural cell apoptosis is the key event initiating and/or driving the pathological cascade, leading to clinical functional deficits in MS. Similarly, regarding axonal damage, a key pathological feature of MS lesions, the roles of inflammation-independent and cell autonomous neuronal processes need to be further explored. While oligodendrocyte and neuronal loss in MS may not necessarily be mutually exclusive, particular attention should be given to the role of neuronal apoptosis in the development of axonal loss. If proven, MS could be viewed primarily as a neurodegenerative disease accompanied by a secondary neuroinflammatory and autoimmune process.
Collapse
|
12
|
Janjusevic M, Gagno G, Fluca AL, Padoan L, Beltrami AP, Sinagra G, Moretti R, Aleksova A. The peculiar role of vitamin D in the pathophysiology of cardiovascular and neurodegenerative diseases. Life Sci 2022; 289:120193. [PMID: 34864062 DOI: 10.1016/j.lfs.2021.120193] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 02/07/2023]
Abstract
Vitamin D is a hormone with both genomic and non-genomic actions. It exerts its activity by binding vitamin D receptor (VDR), which belongs to the superfamily of nuclear receptors and ligand-activated transcription factors. Since VDR has been found in various tissues, it has been estimated that it regulates approximately 3% of the human genome. Several recent studies have shown pleiotropic effects of vitamin D in various processes such as cellular proliferation, differentiation, DNA repair and apoptosis and its involvement in different pathophysiological conditions as inflammation, diabetes mellitus, and anemia. It has been suggested that vitamin D could play an important role in neurodegenerative and cardiovascular disorders. Moderate to strong associations between lower serum vitamin D concentrations and stroke and cardiovascular events have been identified in different analytic approaches, even after controlling for traditional demographic and lifestyle covariates. The mechanisms behind the associations between vitamin D and cerebrovascular and cardiologic profiles have been widely examined both in animal and human studies. Optimization of vitamin D levels in human subjects may improve insulin sensitivity and beta-cell function and lower levels of inflammatory markers. Moreover, it has been demonstrated that altered gene expression of VDR and 1,25D3-membrane-associated rapid response steroid-binding (1,25D3-MARRS) receptor influences the role of vitamin D within neurons and allows them to be more prone to degeneration. This review summarizes the current understanding of the molecular mechanisms underlying vitamin D signaling and the consequences of vitamin D deficiency in neurodegenerative and cardiovascular disorders.
Collapse
Affiliation(s)
- Milijana Janjusevic
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy
| | - Giulia Gagno
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy
| | - Alessandra Lucia Fluca
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy
| | - Laura Padoan
- Cardiology and Cardiovascular Physiopathology, Azienda Ospedaliero-Universitaria S. Maria della Misericordia, 06156 Perugia, Italy
| | - Antonio Paolo Beltrami
- Clinical Pathology Department, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC) and Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy
| | - Rita Moretti
- Department of Internal Medicine and Neurology, Neurological Clinic, Complex Case Section, Trieste, Italy
| | - Aneta Aleksova
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy.
| |
Collapse
|
13
|
Wang C, Barton J, Kyle K, Ly L, Barnett Y, Hartung HP, Reddel SW, Beadnall H, Taha M, Klistorner A, Barnett MH. Multiple sclerosis: structural and functional integrity of the visual system following alemtuzumab therapy. J Neurol Neurosurg Psychiatry 2021; 92:1319-1324. [PMID: 34187865 DOI: 10.1136/jnnp-2021-326164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/02/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To investigate potential neuroprotective and pro-remyelinating effects of alemtuzumab in multiple sclerosis (MS), using the visual pathway as a model. METHODS We monitored clinical, multifocal visual evoked potential (mfVEP) and MRI outcomes in 30 patients commencing alemtuzumab for relapsing MS, and a reference group of 20 healthy controls (HCs), over 24 months. Change in mfVEP latency was the primary endpoint; change in optic radiation (OR) lesion diffusion metrics and Mars letter contrast sensitivity over the course of the study were secondary endpoints. RESULTS In patients, we observed a mean shortening of mfVEP latency of 1.21 ms over the course of the study (95% CI 0.21 to 2.21, p=0.013), not altered by correction for age, gender, disease duration or change in OR T2 lesion volume. Mean mfVEP latency in the HC group increased over the course of the study by 0.72 ms (not significant). Analysis of chronic OR T2 lesions (patients) showed an increase in normalised fractional anisotropy and axial diffusivity between baseline and 24 months (both p<0.01). Mean Mars letter contrast sensitivity was improved at 24 months vs baseline (p<0.001), and driven by an early improvement, in both patients and HC. CONCLUSION We found evidence of partial lesion remyelination after alemtuzumab therapy, indicating either natural restoration in the context of a 'permissive' local milieu; or potentially an independent, pro-reparative mechanism of action. The visual system presents a unique opportunity to study function-structure specific effects of therapy and inform the design of future phase 2 MS remyelination trials.
Collapse
Affiliation(s)
- Chenyu Wang
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Joshua Barton
- Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Kain Kyle
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Linda Ly
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia
| | - Yael Barnett
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia.,Radiology Department, St Vincent's Hospital Sydney, Darlinghurst, New South Wales, Australia
| | - Hans-Peter Hartung
- Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia.,Clinic for Neurology, Heinrich Heine University Düsseldorf, Dusseldorf, Germany
| | - Stephen W Reddel
- Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Heidi Beadnall
- Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia.,Neurology Department, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Marinda Taha
- Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Alexander Klistorner
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia.,Save Sight Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Michael Harry Barnett
- Sydney Neuroimaging Analysis Centre, Camperdown, New South Wales, Australia .,Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia.,Neurology Department, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
14
|
Cairns J, Vavasour IM, Traboulsee A, Carruthers R, Kolind SH, Li DKB, Moore GRW, Laule C. Diffusely abnormal white matter in multiple sclerosis. J Neuroimaging 2021; 32:5-16. [PMID: 34752664 DOI: 10.1111/jon.12945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023] Open
Abstract
MRI enables detailed in vivo depiction of multiple sclerosis (MS) pathology. Localized areas of MS damage, commonly referred to as lesions, or plaques, have been a focus of clinical and research MRI studies for over four decades. A nonplaque MRI abnormality which is present in at least 25% of MS patients but has received far less attention is diffusely abnormal white matter (DAWM). DAWM has poorly defined boundaries and a signal intensity that is between normal-appearing white matter and classic lesions on proton density and T2 -weighted images. All clinical phenotypes of MS demonstrate DAWM, including clinically isolated syndrome, where DAWM is associated with higher lesion volume, reduced brain volume, and earlier conversion to MS. Advanced MRI metric abnormalities in DAWM tend to be greater than those in NAWM, but not as severe as focal lesions, with myelin, axons, and water-related changes commonly reported. Histological studies demonstrate a primary lipid abnormality in DAWM, with some axonal damage and lesser involvement of myelin proteins. This review provides an overview of DAWM identification, summarizes in vivo and postmortem observations, and comments on potential pathophysiological mechanisms, which may underlie DAWM in MS. Given the prevalence and potential clinical impact of DAWM, the number of imaging studies focusing on DAWM is insufficient. Characterization of DAWM significance and microstructure would benefit from larger longitudinal and additional quantitative imaging efforts. Revisiting data from previous studies that included proton density and T2 imaging would enable retrospective DAWM identification and analysis.
Collapse
Affiliation(s)
- James Cairns
- Department of Medicine (Neurology), University of British Columbia, British Columbia, Vancouver, Canada.,Department of Radiology, University of British Columbia, British Columbia, Vancouver, Canada
| | - Irene M Vavasour
- Department of Radiology, University of British Columbia, British Columbia, Vancouver, Canada.,International Collaboration on Repair Discoveries, Blusson Spinal Cord Centre, University of British Columbia, British Columbia, Vancouver, Canada
| | - Anthony Traboulsee
- Department of Medicine (Neurology), University of British Columbia, British Columbia, Vancouver, Canada
| | - Robert Carruthers
- Department of Medicine (Neurology), University of British Columbia, British Columbia, Vancouver, Canada
| | - Shannon H Kolind
- Department of Medicine (Neurology), University of British Columbia, British Columbia, Vancouver, Canada.,Department of Radiology, University of British Columbia, British Columbia, Vancouver, Canada.,International Collaboration on Repair Discoveries, Blusson Spinal Cord Centre, University of British Columbia, British Columbia, Vancouver, Canada.,Department of Physics & Astronomy, University of British Columbia, British Columbia, Vancouver, Canada
| | - David K B Li
- Department of Medicine (Neurology), University of British Columbia, British Columbia, Vancouver, Canada.,Department of Radiology, University of British Columbia, British Columbia, Vancouver, Canada
| | - G R Wayne Moore
- Department of Medicine (Neurology), University of British Columbia, British Columbia, Vancouver, Canada.,International Collaboration on Repair Discoveries, Blusson Spinal Cord Centre, University of British Columbia, British Columbia, Vancouver, Canada.,Department of Pathology & Laboratory Medicine, University of British Columbia, British Columbia, Vancouver, Canada
| | - Cornelia Laule
- Department of Radiology, University of British Columbia, British Columbia, Vancouver, Canada.,International Collaboration on Repair Discoveries, Blusson Spinal Cord Centre, University of British Columbia, British Columbia, Vancouver, Canada.,Department of Physics & Astronomy, University of British Columbia, British Columbia, Vancouver, Canada.,Department of Pathology & Laboratory Medicine, University of British Columbia, British Columbia, Vancouver, Canada
| |
Collapse
|
15
|
Samanta J, Silva HM, Lafaille JJ, Salzer JL. Transcriptomic analysis of loss of Gli1 in neural stem cells responding to demyelination in the mouse brain. Sci Data 2021; 8:278. [PMID: 34711861 PMCID: PMC8553940 DOI: 10.1038/s41597-021-01063-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 09/10/2021] [Indexed: 12/11/2022] Open
Abstract
In the adult mammalian brain, Gli1 expressing neural stem cells reside in the subventricular zone and their progeny are recruited to sites of demyelination in the white matter where they generate new oligodendrocytes, the myelin forming cells. Remarkably, genetic loss or pharmacologic inhibition of Gli1 enhances the efficacy of remyelination by these neural stem cells. To understand the molecular mechanisms involved, we performed a transcriptomic analysis of this Gli1-pool of neural stem cells. We compared murine NSCs with either intact or deficient Gli1 expression from adult mice on a control diet or on a cuprizone diet which induces widespread demyelination. These data will be a valuable resource for identifying therapeutic targets for enhancing remyelination in demyelinating diseases like multiple sclerosis.
Collapse
Affiliation(s)
- Jayshree Samanta
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University School of Medicine, New York, NY, 10016, USA.
- Stem Cell and Regenerative Medicine Center, Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Hernandez Moura Silva
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, New York, 10016, USA
| | - Juan J Lafaille
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, New York, 10016, USA
| | - James L Salzer
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
16
|
Lehmann H, Zveik O, Levin N, Brill L, Imbar T, Vaknin-Dembinsky A. Brain MRI activity during the year before pregnancy can predict post-partum clinical relapses. Mult Scler 2021; 27:2232-2239. [PMID: 33783260 DOI: 10.1177/13524585211002719] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND There are fewer multiple sclerosis (MS) relapses during pregnancy, although relapse risk increases in the early post-partum period, as has been predicted by pre-pregnancy or pregnancy disease activity in some studies. OBJECTIVE The aim of this study was to evaluate the correlation between magnetic resonance imaging (MRI) changes in the year before pregnancy and the relapse rate in the year post-partum. METHODS An observational retrospective case-control study included 172 pregnancies in 118 females with MS. Statistical analyses were used to evaluate the correlation between MRI and post-partum relapses. Clustered logistic regression was used to investigate the predictors of early post-partum relapses. RESULTS We found a significant correlation for an active-MRI pre-pregnancy and relapses in the first 3 months post-partum (p < 0.001). Expanded Disability Status Scale (EDSS) pre-pregnancy and relapses in the first 3 months post-partum were also significantly correlated (p = 0.009). Using a multivariate model, we predicted which women will not experience post-partum relapse by EDSS and by an active-MRI pre-pregnancy (96.7% specificity; p < 0.001). CONCLUSION An active-MRI pre-pregnancy is a strong and sensitive predictor of early post-partum relapse, regardless of whether the woman had clinical evidence of disease activity prior to conception and delivery. This finding could provide clinicians with a strategy to minimize post-partum relapse risk in women with MS planning pregnancy.
Collapse
Affiliation(s)
- Hillel Lehmann
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah Medical Center, Jerusalem, Israel
| | - Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah Medical Center, Jerusalem, Israel
| | - Netta Levin
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah Medical Center, Jerusalem, Israel
| | - Livnat Brill
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah Medical Center, Jerusalem, Israel
| | - Tal Imbar
- Department of Obstetrics and Gynecology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
17
|
Gröger V, Emmer A, Staege MS, Cynis H. Endogenous Retroviruses in Nervous System Disorders. Pharmaceuticals (Basel) 2021; 14:ph14010070. [PMID: 33467098 PMCID: PMC7829834 DOI: 10.3390/ph14010070] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
Human endogenous retroviruses (HERV) have been implicated in the pathogenesis of several nervous system disorders including multiple sclerosis and amyotrophic lateral sclerosis. The toxicity of HERV-derived RNAs and proteins for neuronal cells has been demonstrated. The involvement of HERV in the pathogenesis of currently incurable diseases might offer new treatment strategies based on the inhibition of HERV activities by small molecules or therapeutic antibodies.
Collapse
Affiliation(s)
- Victoria Gröger
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120 Halle (Saale), Germany;
| | - Alexander Emmer
- Department of Neurology, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Martin S. Staege
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
- Correspondence: (M.S.S.); (H.C.); Tel.: +49-345-557-7280 (M.S.S.); +49-345-13142835 (H.C.)
| | - Holger Cynis
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120 Halle (Saale), Germany;
- Correspondence: (M.S.S.); (H.C.); Tel.: +49-345-557-7280 (M.S.S.); +49-345-13142835 (H.C.)
| |
Collapse
|
18
|
Orthmann-Murphy J, Call CL, Molina-Castro GC, Hsieh YC, Rasband MN, Calabresi PA, Bergles DE. Remyelination alters the pattern of myelin in the cerebral cortex. eLife 2020; 9:e56621. [PMID: 32459173 PMCID: PMC7292648 DOI: 10.7554/elife.56621] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/22/2020] [Indexed: 11/25/2022] Open
Abstract
Destruction of oligodendrocytes and myelin sheaths in cortical gray matter profoundly alters neural activity and is associated with cognitive disability in multiple sclerosis (MS). Myelin can be restored by regenerating oligodendrocytes from resident progenitors; however, it is not known whether regeneration restores the complex myelination patterns in cortical circuits. Here, we performed time lapse in vivo two photon imaging in somatosensory cortex of adult mice to define the kinetics and specificity of myelin regeneration after acute oligodendrocyte ablation. These longitudinal studies revealed that the pattern of myelination in cortex changed dramatically after regeneration, as new oligodendrocytes were formed in different locations and new sheaths were often established along axon segments previously lacking myelin. Despite the dramatic increase in axonal territory available, oligodendrogenesis was persistently impaired in deeper cortical layers that experienced higher gliosis. Repeated reorganization of myelin patterns in MS may alter circuit function and contribute to cognitive decline.
Collapse
Affiliation(s)
- Jennifer Orthmann-Murphy
- The Solomon Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
- Department of Neurology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Cody L Call
- The Solomon Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
| | - Gian C Molina-Castro
- The Solomon Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
| | - Yu Chen Hsieh
- The Solomon Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, One Baylor PlazaHoustonUnited States
| | - Peter A Calabresi
- Department of Neurology Johns Hopkins UniversityBaltimoreUnited States
| | - Dwight E Bergles
- The Solomon Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
- Johns Hopkins University Kavli Neuroscience Discovery InstituteBaltimoreUnited States
| |
Collapse
|
19
|
Regulatory T cells promote remyelination in the murine experimental autoimmune encephalomyelitis model of multiple sclerosis following human neural stem cell transplant. Neurobiol Dis 2020; 140:104868. [PMID: 32276110 DOI: 10.1016/j.nbd.2020.104868] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 03/05/2020] [Accepted: 04/05/2020] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory autoimmune disease that affects the central nervous system (CNS) for which there is no cure. In MS, encephalitogenic T cells infiltrate the CNS causing demyelination and neuroinflammation; however, little is known about the role of regulatory T cells (Tregs) in CNS tissue repair. Transplantation of neural stem and progenitor cells (NSCs and NPCs) is a promising therapeutic strategy to promote repair through cell replacement, although recent findings suggest transplanted NSCs also instruct endogenous repair mechanisms. We have recently described that dampened neuroinflammation and increased remyelination is correlated with emergence of Tregs following human NPC transplantation in a murine viral model of immune-mediated demyelination. In the current study we utilized the prototypic murine autoimmune model of demyelination experimental autoimmune encephalomyelitis (EAE) to test the efficacy of hNSC transplantation. Eight-week-old, male EAE mice receiving an intraspinal transplant of hNSCs during the chronic phase of disease displayed remyelination, dampened neuroinflammation, and an increase in CNS CD4+CD25+FoxP3+ regulatory T cells (Tregs). Importantly, ablation of Tregs abrogated histopathological improvement. Tregs are essential for maintenance of T cell homeostasis and prevention of autoimmunity, and an emerging role for Tregs in maintenance of tissue homeostasis through interactions with stem and progenitor cells has recently been suggested. The data presented here provide direct evidence for collaboration between CNS Tregs and hNSCs promoting remyelination.
Collapse
|
20
|
Dziedzic A, Miller E, Saluk-Bijak J, Bijak M. The GPR17 Receptor-A Promising Goal for Therapy and a Potential Marker of the Neurodegenerative Process in Multiple Sclerosis. Int J Mol Sci 2020; 21:ijms21051852. [PMID: 32182666 PMCID: PMC7084627 DOI: 10.3390/ijms21051852] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 02/29/2020] [Accepted: 03/06/2020] [Indexed: 12/28/2022] Open
Abstract
One of the most important goals in the treatment of demyelinating diseases such as multiple sclerosis (MS) is, in addition to immunomodulation, reconstruction of the lost myelin sheath. The modulator of the central nervous system myelination is the metabotropic receptor coupled to the G-protein: GPR17. GPR17 receptors are considered to be sensors of local damage to the myelin sheath, and play a role in the reconstruction and repair of demyelinating plaques caused by ongoing inflammatory processes. GPR17 receptors are present on nerve cells and precursor oligodendrocyte cells. Under physiological conditions, they are responsible for the differentiation and subsequent maturation of oligodendrocytes, while under pathological conditions (during damage to nerve cells), their expression increases to become mediators in the demyelinating processes. Moreover, they are essential not only in both the processes of inducing damage and the death of neurons, but also in the local repair of the damaged myelin sheath. Therefore, GPR17 receptors may be recognized as the potential goal in creating innovative therapies for the treatment of the neurodegenerative process in MS, based on the acceleration of the remyelination processes. This review examines the role of GRP17 in pathomechanisms of MS development.
Collapse
Affiliation(s)
- Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (J.S.-B.)
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland;
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (J.S.-B.)
| | - Michal Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
- Correspondence: ; Tel.: +48-42-635-4336
| |
Collapse
|
21
|
Albert C, Mikolajczak J, Liekfeld A, Piper SK, Scheel M, Zimmermann HG, Nowak C, Dörr J, Bellmann-Strobl J, Chien C, Brandt AU, Paul F, Hoffmann O. Fingolimod after a first unilateral episode of acute optic neuritis (MOVING) - preliminary results from a randomized, rater-blind, active-controlled, phase 2 trial. BMC Neurol 2020; 20:75. [PMID: 32126977 PMCID: PMC7052969 DOI: 10.1186/s12883-020-01645-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 02/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background Neuroprotection and promotion of remyelination represent important therapeutic gaps in multiple sclerosis (MS). Acute optic neuritis (ON) is a frequent MS manifestation. Based on the presence and properties of sphingosine-1-phosphate receptors (S1PR) on astrocytes and oligodendrocytes, we hypothesized that remyelination can be enhanced by treatment with fingolimod, a S1PR modulator currently licensed for relapsing-remitting MS. Methods MOVING was an investigator-driven, rater-blind, randomized clinical trial. Patients with acute unilateral ON, occurring as a clinically isolated syndrome or MS relapse, were randomized to 6 months of treatment with 0.5 mg oral fingolimod or subcutaneous IFN-β 1b 250 μg every other day. The change in multifocal visual evoked potential (mfVEP) latency of the qualifying eye was examined as the primary (month 6 vs. baseline) and secondary (months 3, 6 and 12 vs. baseline) outcome. In addition, full field visual evoked potentials, visual acuity, optical coherence tomography as well as clinical relapses and measures of disability, cerebral MRI, and self-reported visual quality of life were obtained for follow-up. The study was halted due to insufficient recruitment (n = 15), and available results are reported. Results Per protocol analysis of the primary endpoint revealed a significantly larger reduction of mfVEP latency at 6 months compared to baseline with fingolimod treatment (n = 5; median decrease, 15.7 ms) than with IFN-β 1b treatment (n = 4; median increase, 8.15 ms) (p < 0.001 for interaction). Statistical significance was maintained in the secondary endpoint analysis. Descriptive results are reported for other endpoints. Conclusion Preliminary results of the MOVING trial argue in support of a beneficial effect of fingolimod on optic nerve remyelination when compared to IFN-β treatment. Interpretation is limited by the small number of complete observations, an unexpected deterioration of the control group and a difference in baseline mfVEP latencies. The findings need to be confirmed in larger studies. Trial registration The trial was registered as EUDRA-CT 2011–004787-30 on October 26, 2012 and as NCT01647880 on July 24, 2012.
Collapse
Affiliation(s)
- Christian Albert
- Department of Neurology, Alexianer St. Josefs-Krankenhaus Potsdam, Allee nach Sanssouci 7, 14471, Potsdam, Germany
| | - Janine Mikolajczak
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Liekfeld
- Department of Ophthalmology, Klinikum Ernst von Bergmann, Potsdam, Germany
| | - Sophie K Piper
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Michael Scheel
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Hanna G Zimmermann
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Jan Dörr
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, Oberhavel-Kliniken Hennigsdorf, Hennigsdorf, Germany
| | | | - Claudia Chien
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander U Brandt
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, University of California, Irvine, CA, USA
| | - Friedemann Paul
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité-Universitätmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Olaf Hoffmann
- Department of Neurology, Alexianer St. Josefs-Krankenhaus Potsdam, Allee nach Sanssouci 7, 14471, Potsdam, Germany. .,Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
22
|
Sivakolundu DK, Hansen MR, West KL, Wang Y, Stanley T, Wilson A, McCreary M, Turner MP, Pinho MC, Newton BD, Guo X, Rypma B, Okuda DT. Three‐Dimensional Lesion Phenotyping and Physiologic Characterization Inform Remyelination Ability in Multiple Sclerosis. J Neuroimaging 2019; 29:605-614. [DOI: 10.1111/jon.12633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 11/29/2022] Open
Affiliation(s)
- Dinesh K. Sivakolundu
- NeuroPsychometric Research Laboratory, Center for BrainHealthUniversity of Texas at Dallas Dallas TX
| | - Madison R. Hansen
- Department of Neurology & NeurotherapeuticsUT Southwestern Medical Center Dallas TX
| | - Kathryn L. West
- NeuroPsychometric Research Laboratory, Center for BrainHealthUniversity of Texas at Dallas Dallas TX
| | - Yeqi Wang
- Department of Computer ScienceUniversity of Texas at Dallas Dallas TX
| | - Thomas Stanley
- Department of Computer ScienceUniversity of Texas at Dallas Dallas TX
| | - Andrew Wilson
- Department of Computer ScienceUniversity of Texas at Dallas Dallas TX
| | | | - Monroe P. Turner
- NeuroPsychometric Research Laboratory, Center for BrainHealthUniversity of Texas at Dallas Dallas TX
| | - Marco C. Pinho
- Department of RadiologyUT Southwestern Medical Center Dallas TX
| | | | - Xiaohu Guo
- Department of Computer ScienceUniversity of Texas at Dallas Dallas TX
| | - Bart Rypma
- NeuroPsychometric Research Laboratory, Center for BrainHealthUniversity of Texas at Dallas Dallas TX
- Department of PsychiatryUT Southwestern Medical Center Dallas TX
| | - Darin T. Okuda
- Department of Neurology & NeurotherapeuticsUT Southwestern Medical Center Dallas TX
| |
Collapse
|
23
|
Mausner-Fainberg K, Penn M, Golan M, Benhamou M, Wilf-Yarkoni A, Gertel S, Karni A. Reduced levels of Coco in sera of multiple sclerosis patients: A potential role in neuro-regeneration failure. J Neuroimmunol 2019; 327:36-40. [PMID: 30685069 DOI: 10.1016/j.jneuroim.2019.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/04/2019] [Accepted: 01/16/2019] [Indexed: 01/17/2023]
Abstract
Demyelination, axonal loss and failure of tissue repair characterize MS lesions. Bone morphogenetic proteins (BMPs) signaling is associated with remyelination failure. Coco is one of the BMP antagonists. We found reduced Coco serum levels in relapsing-remitting MS (RR-MS) and primary progressive MS (PP-MS) patients compared to matched healthy controls (HC) and patients with rheumatoid arthritis. Exposure of P19 cells, in the presence of retinoic acid, BMP-2, or BMP-4 to Coco, at average sera level of MS patients failed to induce neuronal phenotype, in contrast to the average sera level of HC. Coco may be a player in the BMP dysregulation and the tissue repair failure in MS.
Collapse
Affiliation(s)
- Karin Mausner-Fainberg
- Neuroimmunology Laboratory, Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Moran Penn
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Maya Golan
- Neuroimmunology Laboratory, Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Moshe Benhamou
- Neuroimmunology Laboratory, Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - A Wilf-Yarkoni
- Department of Neurology, Tel Aviv Sourasky Medical Center, Israel
| | - Smadar Gertel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Arnon Karni
- Neuroimmunology Laboratory, Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Segol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
24
|
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease that affects the central nervous system (CNS), particularly, in young adults. Current MS treatments aim to reduce demyelination; however, these have limited efficacy, display side effects and lack of regenerative activities. Oligodendrocyte progenitor cells (OPCs) represents the major source for new myelin. Upon demyelination, OPCs get activated, proliferate, migrate towards the lesion, and differentiate into remyelinating oligodendrocytes. Although myelin repair (remyelination) represents a robust response to myelin damage, during MS, this regenerative phenomenon decays in efficiency or even fails. CNS-resident pericytes (CNS-PCs) are essential for vascular homeostasis regulating blood-brain barrier (BBB) permeability and stability as well as endothelial cells (ECs) function during angiogenesis and neovascularization. Recent studies indicate that CNS-PCs also play a crucial role regulating OPC function during remyelination, and very importantly, these cells are substantially affected in MS. This chapter summarizes important aspects of MS and CNS remyelination as well as it provides new insights supporting the contribution of CNS-PCs to myelin regeneration and to MS pathology. Currently, there is evidence arguing in favor of CNS-PCs as novel therapeutic targets for the development of future treatments for MS.
Collapse
|
25
|
Abstract
Endogenous remyelination of the CNS can be robust and restore function, yet in multiple sclerosis it becomes less complete with time. Promoting remyelination is a major therapeutic goal, both to restore function and to protect axons from degeneration. Remyelination is thought to depend on oligodendrocyte progenitor cells, giving rise to nascent remyelinating oligodendrocytes. Surviving, mature oligodendrocytes are largely regarded as being uninvolved. We have examined this question using two large animal models. In the first model, there is extensive demyelination and remyelination of the CNS, yet oligodendrocytes survive, and in recovered animals there is a mix of remyelinated axons interspersed between mature, thick myelin sheaths. Using 2D and 3D light and electron microscopy, we show that many oligodendrocytes are connected to mature and remyelinated myelin sheaths, which we conclude are cells that have reextended processes to contact demyelinated axons while maintaining mature myelin internodes. In the second model in vitamin B12-deficient nonhuman primates, we demonstrate that surviving mature oligodendrocytes extend processes and ensheath demyelinated axons. These data indicate that mature oligodendrocytes can participate in remyelination.
Collapse
|
26
|
Vitamin D in Neurological Diseases: A Rationale for a Pathogenic Impact. Int J Mol Sci 2018; 19:ijms19082245. [PMID: 30065237 PMCID: PMC6121649 DOI: 10.3390/ijms19082245] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 12/22/2022] Open
Abstract
It is widely known that vitamin D receptors have been found in neurons and glial cells, and their highest expression is in the hippocampus, hypothalamus, thalamus and subcortical grey nuclei, and substantia nigra. Vitamin D helps the regulation of neurotrophin, neural differentiation, and maturation, through the control operation of growing factors synthesis (i.e., neural growth factor [NGF] and glial cell line-derived growth factor (GDNF), the trafficking of the septohippocampal pathway, and the control of the synthesis process of different neuromodulators (such as acetylcholine [Ach], dopamine [DA], and gamma-aminobutyric [GABA]). Based on these assumptions, we have written this review to summarize the potential role of vitamin D in neurological pathologies. This work could be titanic and the results might have been very fuzzy and even incoherent had we not conjectured to taper our first intentions and devoted our interests towards three mainstreams, demyelinating pathologies, vascular syndromes, and neurodegeneration. As a result of the lack of useful therapeutic options, apart from the disease-modifying strategies, the role of different risk factors should be investigated in neurology, as their correction may lead to the improvement of the cerebral conditions. We have explored the relationships between the gene-environmental influence and long-term vitamin D deficiency, as a risk factor for the development of different types of neurological disorders, along with the role and the rationale of therapeutic trials with vitamin D implementation.
Collapse
|
27
|
Abu-Rub M, Miller RH. Emerging Cellular and Molecular Strategies for Enhancing Central Nervous System (CNS) Remyelination. Brain Sci 2018; 8:brainsci8060111. [PMID: 29914096 PMCID: PMC6024921 DOI: 10.3390/brainsci8060111] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023] Open
Abstract
Myelination is critical for the normal functioning of the central nervous system (CNS) in vertebrates. Conditions in which the development of myelin is perturbed result in severely compromised individuals often with shorter lifespans, while loss of myelin in the adult results in a variety of functional deficits. Although some form of spontaneous remyelination often takes place, the repair process as a whole often fails. Several lines of evidence suggest it is feasible to develop strategies that enhance the capacity of the CNS to undergo remyelination and potentially reverse functional deficits. Such strategies include cellular therapies using either neural or mesenchymal stem cells as well as molecular regulators of oligodendrocyte development and differentiation. Given the prevalence of demyelinating diseases and their effects on the quality of life for affected individuals it is imperative that effective therapies are developed. Here we discuss some of the new approaches to CNS myelin repair that hold promise for reducing the burden of diseases characterized by myelin loss.
Collapse
Affiliation(s)
- Mohammad Abu-Rub
- Department of Neurology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| | - Robert H Miller
- Department of Anatomy and Regenerative Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| |
Collapse
|
28
|
Molecular Mechanisms of Oligodendrocyte Regeneration in White Matter-Related Diseases. Int J Mol Sci 2018; 19:ijms19061743. [PMID: 29895784 PMCID: PMC6032201 DOI: 10.3390/ijms19061743] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/25/2018] [Accepted: 06/06/2018] [Indexed: 12/13/2022] Open
Abstract
Even in adult brains, restorative mechanisms are still retained to maintain the microenvironment. Under the pathological conditions of central nervous system (CNS) diseases, several immature cells in the brain would be activated as a compensative response. As the concept of the neurovascular unit emphasizes, cell-cell interactions play important roles in this restorative process. White matter damage and oligodendrocyte loss are representative characteristics for many neurodegenerative diseases. In response to oligodendrocyte damage, residual oligodendrocyte precursor cells (OPCs) initiate their proliferation and differentiation for the purpose of remyelination. Although mechanisms of oligodendrogenesis and remyelination in CNS diseases are still mostly unknown and understudied, accumulated evidence now suggests that support from neighboring cells is necessary for OPC proliferation and differentiation. In this review, we first overview basic mechanisms of interaction between oligodendrocyte lineage cells and neighboring cells, and then introduce how oligodendrogenesis occurs under the conditions of neurodegenerative diseases, focusing on vascular cognitive impairment syndrome, Alzheimer’s disease, and multiple sclerosis.
Collapse
|
29
|
Matías-Guíu J, Oreja-Guevara C, Matias-Guiu J, Gomez-Pinedo U. Vitamin D and remyelination in multiple sclerosis. NEUROLOGÍA (ENGLISH EDITION) 2018. [DOI: 10.1016/j.nrleng.2016.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
30
|
The extracellular matrix: Focus on oligodendrocyte biology and targeting CSPGs for remyelination therapies. Glia 2018; 66:1809-1825. [DOI: 10.1002/glia.23333] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 12/31/2022]
|
31
|
Rezapour-Firouzi S, Shahabi S, Mohammadzadeh A, Tehrani AA, Kheradmand F, Mazloomi E. The potential effects of hemp seed/evening primrose oils on the mammalian target of rapamycin complex 1 and interferon-gamma genes expression in experimental autoimmune encephalomyelitis. Res Pharm Sci 2018; 13:523-532. [PMID: 30607150 PMCID: PMC6288989 DOI: 10.4103/1735-5362.245964] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) has a fundamental role in the metabolism, growth, and regulation of the immune system. The interferon gamma (IFN-γ)derived from T helper 1 (Th1) cells is a prominent pro-inflammatory cytokine in multiple sclerosis (MS) and its animal model, the experimental autoimmune encephalomyelitis (EAE). Due to the exclusive role of rapamycin (RAPA) in mTOR complex 1 (mTORC1) inhibition, essentially Th1 differentiation and IFN-γ production, we evaluated the potential therapeutic effects of hemp seed/evening primrose oils (HSO/EPO) in comparison with RAPA administration in EAE. To evaluate the therapeutic effects of EPO/HSO supplement in comparison with RAPA, EAE was induced using myelin oligodendrocyte glycoprotein (MOG) peptide and complete Freund's adjuvant in C57BL/6 mice. The weight, clinical score, and histological findings were evaluated. Total mRNA was extracted from local lymph nodes and qRT-PCR was used for the purpose of the genes expression level of regulatory associated protein of TORC1 (RAPTOR) and IFN-γ. Our results indicated that the relative expression of RAPTOR and IFN-γ genes were significantly reduced in HSO/EPO, RAPA, and RAPA + HSO/EPO treated groups in comparison with the untreated group. Interestingly, histological findings have shown that the HSO/EPO treated group remarkably regenerated the myelin sheath, but this did not occur in the case of RAPA or combined RAPA and HSO/EPO treated groups. Our findings suggeste that HSO/HPO can be used as a potent immunomodulator and as a good candidate for re-myelination and downregulation of immune response for treatment of MS.
Collapse
Affiliation(s)
- Soheila Rezapour-Firouzi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, I.R. Iran
| | - Shahram Shahabi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, I.R. Iran
| | - Adel Mohammadzadeh
- Departement of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, I.R. Iran
| | - Ali Asgar Tehrani
- Department of Pathobiology, Faculty of Veterinary Medicine, Urmia University, Urmia, I.R. Iran
| | - Fatemeh Kheradmand
- Department of Biochemistry, School of Medicine, Urmia University of Medical Science, Urmia, I.R. Iran
| | - Ebrahim Mazloomi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, I.R. Iran
| |
Collapse
|
32
|
Abstract
This chapter provides a brief overview of studies that combine postmortem magnetic resonance imaging (MRI) and histopathology. We touch upon the logistics of setting up a protocol that limits unwanted postmortem delays and explain how combining postmortem MRI and histopathology can elucidate the histologic substrate of signal changes that appear on MRI. This is demonstrated by exemplary studies in multiple sclerosis, and includes various histopathologic techniques and a wide range of conventional and advanced MRI sequences at various field strengths. We cover topics such as how to visualize white-matter pathology and repair with conventional and advanced MRI sequences, describe the history of visualizing pathology of the gray matter (with newly developed MRI and immunohistopathology techniques), and how advanced methods have aided research in other neurologic diseases. We conclude with several suggestions for future development, such as bridging the gap between postmortem and in vivo research and the importance of collecting non-neurological control tissue.
Collapse
Affiliation(s)
- Laura E Jonkman
- Department of Anatomy and Neurosciences, VU Medical Center, Amsterdam, The Netherlands
| | - Jeroen J G Geurts
- Department of Anatomy and Neurosciences, VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Popescu BF, Frischer JM, Webb SM, Tham M, Adiele RC, Robinson CA, Fitz-Gibbon PD, Weigand SD, Metz I, Nehzati S, George GN, Pickering IJ, Brück W, Hametner S, Lassmann H, Parisi JE, Yong G, Lucchinetti CF. Pathogenic implications of distinct patterns of iron and zinc in chronic MS lesions. Acta Neuropathol 2017; 134:45-64. [PMID: 28332093 PMCID: PMC5486634 DOI: 10.1007/s00401-017-1696-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS) in which oligodendrocytes, the CNS cells that stain most robustly for iron and myelin are the targets of injury. Metals are essential for normal CNS functioning, and metal imbalances have been linked to demyelination and neurodegeneration. Using a multidisciplinary approach involving synchrotron techniques, iron histochemistry and immunohistochemistry, we compared the distribution and quantification of iron and zinc in MS lesions to the surrounding normal appearing and periplaque white matter, and assessed the involvement of these metals in MS lesion pathogenesis. We found that the distribution of iron and zinc is heterogeneous in MS plaques, and with few remarkable exceptions they do not accumulate in chronic MS lesions. We show that brain iron tends to decrease with increasing age and disease duration of MS patients; reactive astrocytes organized in large astrogliotic areas in a subset of smoldering and inactive plaques accumulate iron and safely store it in ferritin; a subset of smoldering lesions do not contain a rim of iron-loaded macrophages/microglia; and the iron content of shadow plaques varies with the stage of remyelination. Zinc in MS lesions was generally decreased, paralleling myelin loss. Iron accumulates concentrically in a subset of chronic inactive lesions suggesting that not all iron rims around MS lesions equate with smoldering plaques. Upon degeneration of iron-loaded microglia/macrophages, astrocytes may form an additional protective barrier that may prevent iron-induced oxidative damage.
Collapse
Affiliation(s)
- Bogdan F Popescu
- Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, 701 Queen Street, Saskatoon, SK, S7N 5E5, Canada.
- Cameco MS Neuroscience Research Center, University of Saskatchewan, 701 Queen Street, Saskatoon City Hospital, Rm 5800, Saskatoon, SK, S7K 0M7, Canada.
| | - Josa M Frischer
- Department of Neurosurgery, Medical University Vienna, Vienna, Austria
| | - Samuel M Webb
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, CA, USA
| | - Mylyne Tham
- Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, 701 Queen Street, Saskatoon, SK, S7N 5E5, Canada
- Cameco MS Neuroscience Research Center, University of Saskatchewan, 701 Queen Street, Saskatoon City Hospital, Rm 5800, Saskatoon, SK, S7K 0M7, Canada
| | - Reginald C Adiele
- Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, 701 Queen Street, Saskatoon, SK, S7N 5E5, Canada
- Cameco MS Neuroscience Research Center, University of Saskatchewan, 701 Queen Street, Saskatoon City Hospital, Rm 5800, Saskatoon, SK, S7K 0M7, Canada
| | - Christopher A Robinson
- Department of Pathology and Laboratory Medicine, Saskatoon Health Region/College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Patrick D Fitz-Gibbon
- Department of Health Sciences Research, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Stephen D Weigand
- Department of Health Sciences Research, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Imke Metz
- Department of Neuropathology, University of Göttingen, Göttingen, Germany
| | - Susan Nehzati
- Molecular and Environmental Science Research Group, Department of Geological Sciences, University of Saskatchewan, Saskatoon, Canada
| | - Graham N George
- Molecular and Environmental Science Research Group, Department of Geological Sciences, University of Saskatchewan, Saskatoon, Canada
- Toxicology Center, University of Saskatchewan, Saskatoon, Canada
- Department of Chemistry, University of Saskatchewan, Saskatoon, Canada
| | - Ingrid J Pickering
- Molecular and Environmental Science Research Group, Department of Geological Sciences, University of Saskatchewan, Saskatoon, Canada
- Toxicology Center, University of Saskatchewan, Saskatoon, Canada
- Department of Chemistry, University of Saskatchewan, Saskatoon, Canada
| | - Wolfgang Brück
- Department of Neuropathology, University of Göttingen, Göttingen, Germany
| | - Simon Hametner
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Joseph E Parisi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Guo Yong
- Department of Neurology, Mayo Clinic, College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
| | - Claudia F Lucchinetti
- Department of Neurology, Mayo Clinic, College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
34
|
Rezapour-Firouzi S. Herbal Oil Supplement With Hot-Nature Diet for Multiple Sclerosis. NUTRITION AND LIFESTYLE IN NEUROLOGICAL AUTOIMMUNE DISEASES 2017:229-245. [DOI: 10.1016/b978-0-12-805298-3.00024-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
35
|
Thaler C, Faizy TD, Sedlacik J, Holst B, Stürner K, Heesen C, Stellmann JP, Fiehler J, Siemonsen S. T1 Recovery Is Predominantly Found in Black Holes and Is Associated with Clinical Improvement in Patients with Multiple Sclerosis. AJNR Am J Neuroradiol 2016; 38:264-269. [PMID: 28059711 DOI: 10.3174/ajnr.a5004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/08/2016] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Quantitative MR imaging parameters help to evaluate disease progression in multiple sclerosis and increase correlation with clinical disability. We therefore hypothesized that T1 values might be a marker for ongoing tissue damage or even remyelination and may help increase clinical correlation. MATERIALS AND METHODS MR imaging was performed in 17 patients with relapsing-remitting MS at baseline and after 12 months of starting immunotherapy with dimethyl fumarate. On baseline images, lesion segmentation was performed for normal-appearing white matter, T2 hyperintense (FLAIR lesions), T1 hypointense (black holes), and contrast-enhancing lesions, and T1 relaxation times were obtained at baseline and after 12 months. Changes in clinical status were assessed by using the Expanded Disability Status Scale and Symbol Digit Modalities Test at both dates (Expanded Disability Status Scale-difference/Symbol Digit Modalities Test-diff). RESULTS The highest T1 relaxation time at baseline was measured in black holes (1460.2 ± 209.46 ms) followed by FLAIR lesions (1400.38 ± 189.1 ms), pure FLAIR lesions (1327.5 ± 210.04 ms), contrast-enhancing lesions (1205.59 ± 199.95 ms), and normal-appearing white matter (851.34 ± 30.61 ms). After 12 months, T1 values had decreased significantly in black holes (1369.4 ± 267.81 ms), contrast-enhancing lesions (1079.57 ± 183.36 ms) (both P < .001), and normal-appearing white matter (841.98 ± 36.1 ms, P = .006). With the Jonckheere-Terpstra Test, better clinical scores were associated with decreasing T1 relaxation times in black holes (P < .05). CONCLUSIONS T1 relaxation time is a useful quantitative MR imaging technique, which helps detect changes in MS lesions with time. We assume that these changes are associated with the degree of myelination within the lesions themselves and are pronounced in black holes. Additionally, decreasing T1 values in black holes were associated with clinical improvement.
Collapse
Affiliation(s)
- C Thaler
- From the Departments of Diagnostic and Interventional Neuroradiology (C.T., T.D.F., J.S., B.H., J.F., S.S.)
| | - T D Faizy
- From the Departments of Diagnostic and Interventional Neuroradiology (C.T., T.D.F., J.S., B.H., J.F., S.S.)
| | - J Sedlacik
- From the Departments of Diagnostic and Interventional Neuroradiology (C.T., T.D.F., J.S., B.H., J.F., S.S.)
| | - B Holst
- From the Departments of Diagnostic and Interventional Neuroradiology (C.T., T.D.F., J.S., B.H., J.F., S.S.)
| | - K Stürner
- Neurology (K.S., C.H., J.-P.S.).,Institute for Neuroimmunology and Clinical MS Research (K.S., C.H., J.-P.S., S.S.), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - C Heesen
- Neurology (K.S., C.H., J.-P.S.).,Institute for Neuroimmunology and Clinical MS Research (K.S., C.H., J.-P.S., S.S.), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - J-P Stellmann
- Neurology (K.S., C.H., J.-P.S.).,Institute for Neuroimmunology and Clinical MS Research (K.S., C.H., J.-P.S., S.S.), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - J Fiehler
- From the Departments of Diagnostic and Interventional Neuroradiology (C.T., T.D.F., J.S., B.H., J.F., S.S.)
| | - S Siemonsen
- From the Departments of Diagnostic and Interventional Neuroradiology (C.T., T.D.F., J.S., B.H., J.F., S.S.).,Institute for Neuroimmunology and Clinical MS Research (K.S., C.H., J.-P.S., S.S.), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
36
|
Psachoulia K, Chamberlain KA, Heo D, Davis SE, Paskus JD, Nanescu SE, Dupree JL, Wynn TA, Huang JK. IL4I1 augments CNS remyelination and axonal protection by modulating T cell driven inflammation. Brain 2016; 139:3121-3136. [PMID: 27797811 PMCID: PMC5382940 DOI: 10.1093/brain/aww254] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/01/2016] [Accepted: 08/22/2016] [Indexed: 01/01/2023] Open
Abstract
SEE PLUCHINO AND PERUZZOTTI-JAMETTI DOI101093/AWW266 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE: Myelin regeneration (remyelination) is a spontaneous process that occurs following central nervous system demyelination. However, for reasons that remain poorly understood, remyelination fails in the progressive phase of multiple sclerosis. Emerging evidence indicates that alternatively activated macrophages in central nervous system lesions are required for oligodendrocyte progenitor differentiation into remyelinating oligodendrocytes. Here, we show that an alternatively activated macrophage secreted enzyme, interleukin-four induced one (IL4I1), is upregulated at the onset of inflammation resolution and remyelination in mouse central nervous system lesions after lysolecithin-induced focal demyelination. Focal demyelination in mice lacking IL4I1 or interleukin 4 receptor alpha (IL4Rα) results in increased proinflammatory macrophage density, remyelination impairment, and axonal injury in central nervous system lesions. Conversely, recombinant IL4I1 administration into central nervous system lesions reduces proinflammatory macrophage density, enhances remyelination, and rescues remyelination impairment in IL4Rα deficient mice. We find that IL4I1 does not directly affect oligodendrocyte differentiation, but modulates inflammation by reducing interferon gamma and IL17 expression in lesioned central nervous system tissues, and in activated T cells from splenocyte cultures. Remarkably, intravenous injection of IL4I1 into mice with experimental autoimmune encephalomyelitis at disease onset significantly reversed disease severity, resulting in recovery from hindlimb paralysis. Analysis of post-mortem tissues reveals reduced axonal dystrophy in spinal cord, and decreased CD4+ T cell populations in spinal cord and spleen tissues. These results indicate that IL4I1 modulates inflammation by regulating T cell expansion, thereby permitting the formation of a favourable environment in the central nervous system tissue for remyelination. Therefore, IL4I1 is a potentially novel therapeutic for promoting central nervous system repair in multiple sclerosis.
Collapse
Affiliation(s)
| | | | - Dongeun Heo
- 1 Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Stephanie E Davis
- 1 Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Jeremiah D Paskus
- 1 Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Sonia E Nanescu
- 1 Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Jeffrey L Dupree
- 2 Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Thomas A Wynn
- 3 Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffrey K Huang
- 1 Department of Biology, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
37
|
Matías-Guíu J, Oreja-Guevara C, Matias-Guiu JA, Gomez-Pinedo U. Vitamin D and remyelination in multiple sclerosis. Neurologia 2016; 33:177-186. [PMID: 27321170 DOI: 10.1016/j.nrl.2016.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 05/12/2016] [Indexed: 12/01/2022] Open
Abstract
INTRODUCTION Several studies have found an association between multiple sclerosis and vitamin D (VD) deficiency, which suggests that VD may play a role in the immune response. However, few studies have addressed its role in remyelination. DEVELOPMENT The VD receptor and the enzymes transforming VD into metabolites which activate the VD receptor are expressed in central nervous system (CNS) cells, which suggests a potential effect of VD on the CNS. Both in vitro and animal model studies have shown that VD may play a role in myelination by acting on factors that influence the microenvironment which promotes both proliferation and differentiation of neural stem cells into oligodendrocyte progenitor cells and oligodendrocytes. It remains unknown whether the mechanisms of internalisation of VD in the CNS are synergistic with or antagonistic to the mechanisms that facilitate the entry of VD metabolites into immune cells. CONCLUSIONS VD seems to play a role in the CNS and our hypothesis is that VD is involved in remyelination. Understanding the basic mechanisms of VD in myelination is necessary to manage multiple sclerosis patients with VD deficiency.
Collapse
Affiliation(s)
- J Matías-Guíu
- Servicio de Neurología, Hospital Clínico San Carlos, Facultad de Medicina, Universidad Complutense, IdiSSC, Madrid, España.
| | - C Oreja-Guevara
- Servicio de Neurología, Hospital Clínico San Carlos, Facultad de Medicina, Universidad Complutense, IdiSSC, Madrid, España
| | - J A Matias-Guiu
- Servicio de Neurología, Hospital Clínico San Carlos, Facultad de Medicina, Universidad Complutense, IdiSSC, Madrid, España
| | - U Gomez-Pinedo
- Servicio de Neurología, Hospital Clínico San Carlos, Facultad de Medicina, Universidad Complutense, IdiSSC, Madrid, España
| |
Collapse
|
38
|
Matías-Guiu JA, Oreja-Guevara C, Cabrera-Martín MN, Moreno-Ramos T, Carreras JL, Matías-Guiu J. Amyloid Proteins and Their Role in Multiple Sclerosis. Considerations in the Use of Amyloid-PET Imaging. Front Neurol 2016; 7:53. [PMID: 27065425 PMCID: PMC4814935 DOI: 10.3389/fneur.2016.00053] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 03/22/2016] [Indexed: 02/06/2023] Open
Abstract
Thioflavin T derivatives are used in positron-emission tomography (PET) studies to detect amyloid protein deposits in patients with Alzheimer disease. These tracers bind extensively to white matter, which suggests that they may be useful in studies of multiple sclerosis (MS), and that proteins resulting from proteolytic processing of the amyloid precursor protein (APP) may contribute to MS. This article reviews data from both clinical and preclinical studies addressing the role of these proteins, whether they are detected in CSF studies or using PET imaging. APP is widely expressed in demyelinated axons and may have a protective effect in MS and in experimental allergic encephalomyelitis in animals. Several mechanisms associated with this increased expression may affect the degree of remyelination in MS. Amyloid-PET imaging may help determine the degree of demyelination and provide information on the molecular changes linked to APP proteolytic processing experienced by patients with MS.
Collapse
Affiliation(s)
- Jordi A Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Celia Oreja-Guevara
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - María Nieves Cabrera-Martín
- Department of Nuclear Medicine, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Teresa Moreno-Ramos
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - José Luis Carreras
- Department of Nuclear Medicine, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Jorge Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| |
Collapse
|
39
|
Activation status of human microglia is dependent on lesion formation stage and remyelination in multiple sclerosis. J Neuropathol Exp Neurol 2015; 74:48-63. [PMID: 25470347 DOI: 10.1097/nen.0000000000000149] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Similar to macrophages, microglia adopt diverse activation states and contribute to repair and tissue damage in multiple sclerosis. Using reverse transcription-quantitative polymerase chain reaction and immunohistochemistry, we show that in vitro M1-polarized (proinflammatory) human adult microglia express the distinctive markers CD74, CD40, CD86, and CCR7, whereas M2 (anti-inflammatory) microglia express mannose receptor and the anti-inflammatory cytokine CCL22. The expression of these markers was assessed in clusters of activated microglia in normal-appearing white matter (preactive lesions) and areas of remyelination, representing reparative multiple sclerosis lesions. We show that activated microglia in preactive and remyelinating lesions express CD74, CD40, CD86, and the M2 markers CCL22 and CD209, but not mannose receptor. To examine whether this intermediate microglia profile is static or dynamic and thus susceptible to changes in the microenvironment, we polarized microglia into M1 or M2 phenotype in vitro and then subsequently treated them with the opposing polarization regimen. These studies revealed that expression of CD40, CXCL10, and mannose receptor is dynamic and that microglia, like macrophages, can switch between M1 and M2 phenotypic profiles. Taken together, our data define the differential activation states of microglia during lesion development in multiple sclerosis-affected CNS tissues and underscore the plasticity of human adult microglia in vitro.
Collapse
|
40
|
Ortiz GG, Pacheco-Moisés FP, Macías-Islas MÁ, Flores-Alvarado LJ, Mireles-Ramírez MA, González-Renovato ED, Hernández-Navarro VE, Sánchez-López AL, Alatorre-Jiménez MA. Role of the blood-brain barrier in multiple sclerosis. Arch Med Res 2014; 45:687-97. [PMID: 25431839 DOI: 10.1016/j.arcmed.2014.11.013] [Citation(s) in RCA: 254] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 11/18/2014] [Indexed: 12/24/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system associated with demyelination and axonal loss eventually leading to neurodegeneration. MS exhibits many of the hallmarks of an inflammatory autoimmune disorder including breakdown of the blood-brain barrier (BBB). The BBB is a complex organization of cerebral endothelial cells, pericytes and their basal lamina, which are surrounded and supported by astrocytes and perivascular macrophages. In pathological conditions, lymphocytes activated in the periphery infiltrate the central nervous system to trigger a local immune response that ultimately damages myelin and axons. Cytotoxic factors including pro-inflammatory cytokines, proteases, and reactive oxygen and nitrogen species accumulate and may contribute to myelin destruction. Dysregulation of the BBB and transendothelial migration of activated leukocytes are among the earliest cerebrovascular abnormalities seen in MS brains and parallel the release of inflammatory cytokines. In this review we establish the importance of the role of the BBB in MS. Improvements in our understanding of molecular mechanism of BBB functioning in physiological and pathological conditions could lead to improvement in the quality of life of MS patients.
Collapse
Affiliation(s)
- Genaro Gabriel Ortiz
- Laboratorio Desarrollo-Envejecimiento, Enfermedades Neurodegenerativas, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México.
| | - Fermín Paul Pacheco-Moisés
- Departamento de Química, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Miguel Ángel Macías-Islas
- Departamento de Neurología, Unidad Médica de Alta Especialidad (UMAE), Hospital de Especialidades (HE), Centro Médico de Nacional de Occidente (CMNO), IMSS, Guadalajara, Jalisco, México
| | - Luis Javier Flores-Alvarado
- Departamento de Bioquímica, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Mario A Mireles-Ramírez
- Departamento de Neurología, Unidad Médica de Alta Especialidad (UMAE), Hospital de Especialidades (HE), Centro Médico de Nacional de Occidente (CMNO), IMSS, Guadalajara, Jalisco, México
| | - Erika Daniela González-Renovato
- Laboratorio Desarrollo-Envejecimiento, Enfermedades Neurodegenerativas, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México
| | - Vanessa Elizabeth Hernández-Navarro
- Laboratorio Desarrollo-Envejecimiento, Enfermedades Neurodegenerativas, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México
| | - Angélica Lizeth Sánchez-López
- Laboratorio Desarrollo-Envejecimiento, Enfermedades Neurodegenerativas, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México
| | - Moisés Alejandro Alatorre-Jiménez
- Laboratorio Desarrollo-Envejecimiento, Enfermedades Neurodegenerativas, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México
| |
Collapse
|
41
|
Chen L, Coleman R, Leang R, Tran H, Kopf A, Walsh CM, Sears-Kraxberger I, Steward O, Macklin WB, Loring JF, Lane TE. Human neural precursor cells promote neurologic recovery in a viral model of multiple sclerosis. Stem Cell Reports 2014; 2:825-37. [PMID: 24936469 PMCID: PMC4050357 DOI: 10.1016/j.stemcr.2014.04.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 12/21/2022] Open
Abstract
Using a viral model of the demyelinating disease multiple sclerosis (MS), we show that intraspinal transplantation of human embryonic stem cell-derived neural precursor cells (hNPCs) results in sustained clinical recovery, although hNPCs were not detectable beyond day 8 posttransplantation. Improved motor skills were associated with a reduction in neuroinflammation, decreased demyelination, and enhanced remyelination. Evidence indicates that the reduced neuroinflammation is correlated with an increased number of CD4+CD25+FOXP3+ regulatory T cells (Tregs) within the spinal cords. Coculture of hNPCs with activated T cells resulted in reduced T cell proliferation and increased Treg numbers. The hNPCs acted, in part, through secretion of TGF-β1 and TGF-β2. These findings indicate that the transient presence of hNPCs transplanted in an animal model of MS has powerful immunomodulatory effects and mediates recovery. Further investigation of the restorative effects of hNPC transplantation may aid in the development of clinically relevant MS treatments. Spinal cord transplantation of hNPCs results in recovery in a viral model of MS hNPC-mediated recovery occurs in the absence of engrafted cells hNPCs are immunomodulatory through increasing the frequency of Tregs in the CNS hNPCs increase Treg frequency via a TGF-β1- and TGF-β2-dependent pathway
Collapse
Affiliation(s)
- Lu Chen
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Ronald Coleman
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ronika Leang
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Ha Tran
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Alexandra Kopf
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Craig M Walsh
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Ilse Sears-Kraxberger
- Reeve-Irvine Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Oswald Steward
- Reeve-Irvine Research Center, Departments of Anatomy & Neurobiology, Neurobiology & Behavior, and Neurosurgery, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Jeanne F Loring
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas E Lane
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
42
|
Lisak RP, Nedelkoska L, Benjamins JA. Effects of dextromethorphan on glial cell function: Proliferation, maturation, and protection from cytotoxic molecules. Glia 2014; 62:751-62. [DOI: 10.1002/glia.22639] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 01/20/2014] [Accepted: 01/22/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Robert P. Lisak
- Department of Neurology; Wayne State University School of Medicine; Detroit Missouri
- Department of Immunology/Microbiology; Wayne State University School of Medicine; Detroit Missouri
| | - Liljana Nedelkoska
- Department of Neurology; Wayne State University School of Medicine; Detroit Missouri
| | - Joyce A. Benjamins
- Department of Neurology; Wayne State University School of Medicine; Detroit Missouri
- Department of Immunology/Microbiology; Wayne State University School of Medicine; Detroit Missouri
| |
Collapse
|
43
|
Pusic AD, Kraig RP. Youth and environmental enrichment generate serum exosomes containing miR-219 that promote CNS myelination. Glia 2013; 62:284-99. [PMID: 24339157 DOI: 10.1002/glia.22606] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 11/05/2013] [Accepted: 11/09/2013] [Indexed: 12/18/2022]
Abstract
Although commonly considered a disease of white matter, gray matter demyelination is increasingly recognized as an important component of multiple sclerosis (MS) pathogenesis, particularly in the secondary progressive disease phase. Extent of damage to gray matter is strongly correlated to decline in memory and cognitive dysfunction in MS patients. Aging likewise occurs with cognitive decline from myelin loss, and age-associated failure to remyelinate significantly contributes to MS progression. However, recent evidence demonstrates that parabiotic exposure of aged animals to a youthful systemic milieu can promote oligodendrocyte precursor cell (OPC) differentiation and improve remyelination. In the current study, we focus on this potential for stimulating remyelination, and show it involves serum exosomes that increase OPCs and their differentiation into mature myelin-producing cells-both under control conditions and after acute demyelination. Environmental enrichment (EE) of aging animals produced exosomes that mimicked this promyelinating effect. Additionally, stimulating OPC differentiation via exosomes derived from environmentally enriched animals is unlikely to deplete progenitors, as EE itself promotes proliferation of neural stem cells. We found that both young and EE serum-derived exosomes were enriched in miR-219, which is necessary and sufficient for production of myelinating oligodendrocytes by reducing the expression of inhibitory regulators of differentiation. Accordingly, protein transcript levels of these miR-219 target mRNAs decreased following exosome application to slice cultures. Finally, nasal administration of exosomes to aging rats also enhanced myelination. Thus, peripheral circulating cells in young or environmentally enriched animals produce exosomes that may be a useful therapy for remyelination.
Collapse
Affiliation(s)
- Aya D Pusic
- Department of Neurology, The University of Chicago, Chicago, Illinois; Committee on Neurobiology, The University of Chicago, Chicago, Illinois
| | | |
Collapse
|
44
|
Cristofanilli M, Cymring B, Lu A, Rosenthal H, Sadiq SA. Cerebrospinal fluid derived from progressive multiple sclerosis patients promotes neuronal and oligodendroglial differentiation of human neural precursor cells in vitro. Neuroscience 2013; 250:614-21. [PMID: 23876320 DOI: 10.1016/j.neuroscience.2013.07.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 07/10/2013] [Accepted: 07/11/2013] [Indexed: 01/08/2023]
Abstract
In the adult CNS, tissue-specific germinal niches, such as the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus of the hippocampus, contain multipotent neural precursor cells (NPCs) with the capacity to self-renew and differentiate into functional brain cells (i.e. neurons, astrocytes or oligodendrocytes). Due to their intrinsic plasticity, NPCs can be considered an essential part of the cellular mechanism(s) by which the CNS tries to repair itself after an injury. In inflammatory CNS disorders, such as multiple sclerosis (MS), neurogenesis and gliogenesis occur as part of an 'intrinsic' self-repair process. However, full and long-lasting repair in progressive MS is not achieved. Recent data suggest that endogenous NPCs, while trying to repair the damaged CNS in MS, may become the target of the disease itself. It is possible that factors produced during MS, like CNS-infiltrating blood-borne inflammatory mononuclear cells, reactive CNS-resident cells, and humoral mediators, can alter the physiological properties of NPCs, ultimately impairing their ability to promote neural regeneration. Here, we investigate the effect of cerebrospinal fluid (CSF) derived from primary progressive (PPMS) and secondary progressive (SPMS) MS patients (CSF-MS) on the survival, proliferation, and differentiation of commercially available human embryonic-derived NPCs named ENStem-A. We found that PPMS derived CSF markedly reduced the proliferation of ENStem-A and increased their differentiation toward neuronal and oligodendroglial cells, compared to control CSF. Similar but less striking results were seen when ENstem-A were treated with SPMS derived CSF. Our findings suggest that in both SPMS and PPMS the CNS milieu, as determined by extrapolation from CSF findings, may stimulate the endogenous pool of NPCs to differentiate into neurons and oligodendrocytes.
Collapse
Affiliation(s)
- M Cristofanilli
- Multiple Sclerosis Research Center of New York, New York, NY 10019, USA
| | | | | | | | | |
Collapse
|
45
|
Wootla B, Watzlawik JO, Denic A, Rodriguez M. The road to remyelination in demyelinating diseases: current status and prospects for clinical treatment. Expert Rev Clin Immunol 2013; 9:535-49. [PMID: 23730884 DOI: 10.1586/eci.13.37] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Within CNS disorders, demyelinating diseases are among the most devastating and cost intensive due to long-term disabilities affecting relatively young patients. Multiple sclerosis, a chronic inflammatory demyelinating disease in which the persistent inhibitory microenvironment of the resident oligodendrocyte precursor cells abrogates regeneration of myelin sheaths, is the most prominent disease in the spectrum of demyelinating diseases. The essential goal is to stimulate creation of new myelin sheaths on the demyelinated axons, leading to restoration of saltatory conduction and resolving functional deficits. The past few decades witnessed significant efforts to understand the cellular interactions at the lesion site with studies suggesting efficient remyelination as a prerequisite for functional repair. Despite its proven efficacy in experimental models, immunosuppression has not had profound clinical consequences in multiple sclerosis, which argued for a paradigm shift in the design of therapeutics aiming to achieve remyelination. For example, targeting oligodendrocytes themselves may drive remyelination in the CNS. This group and others have demonstrated that natural autoreactive antibodies directed at oligodendrocyte progenitors participate in remyelination. Accordingly, the authors developed a recombinant autoreactive natural human IgM antibody with therapeutic potential for remyelination.
Collapse
Affiliation(s)
- Bharath Wootla
- Department of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|
46
|
Abstract
Multiple sclerosis is a chronic inflammatory demyelinating disease of the central nervous system. Both genetic and environmental factors are believed to contribute to the pathogenesis of the disease. Histopathological findings suggest that multiple sclerosis is an immune-mediated disease, involving both the cellular and humoral immune systems. Within the last 20 years, several disease-modifying therapies for the treatment of multiple sclerosis were established. Moreover, promising new substances are currently being tested in clinical trials and will likely broaden the therapeutic opportunities available within the upcoming years.
Collapse
Affiliation(s)
- Rebecca C Selter
- Department of Neurology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Competence Network on Multiple Sklerosis, Munich, Germany
| |
Collapse
|
47
|
Popescu BFG, Lucchinetti CF. Pathology of demyelinating diseases. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2013; 7:185-217. [PMID: 22313379 DOI: 10.1146/annurev-pathol-011811-132443] [Citation(s) in RCA: 267] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There has been significant progress in our understanding of the pathology and pathogenesis of central nervous system inflammatory demyelinating diseases. Neuropathological studies have provided fundamental new insights into the pathogenesis of these disorders and have led to major advances in our understanding of multiple sclerosis (MS) heterogeneity, the substrate of irreversible progressive disability in MS, the relationship between inflammation and neurodegeneration in MS, the neuroimaging correlates of MS lesions, and the pathogenesis of other central nervous system inflammatory disorders, including neuromyelitis optica, acute disseminated encephalomyelitis, and Balo's concentric sclerosis. Herein, we review the pathological features of these central nervous system inflammatory demyelinating disorders and discuss neuropathological studies that have yielded novel insights into potential mechanisms involved in the formation of the demyelinated lesion.
Collapse
Affiliation(s)
- Bogdan F Gh Popescu
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Saskatchewan S7K 0M7, Canada.
| | | |
Collapse
|
48
|
Diffusely abnormal white matter in multiple sclerosis: further histologic studies provide evidence for a primary lipid abnormality with neurodegeneration. J Neuropathol Exp Neurol 2013; 72:42-52. [PMID: 23242281 DOI: 10.1097/nen.0b013e31827bced3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Although multiple sclerosis (MS) lesions have been studied extensively using histology and magnetic resonance imaging (MRI), little is known about diffusely abnormal white matter (DAWM). Diffusely abnormal white matter, regions with reduced mild MRI hyperintensity and ill-defined boundaries, show reduced myelin water fraction, and decreased Luxol fast blue staining of myelin phospholipids, with relative preservation of myelin basic protein and 2',3'-cyclic-nucleotide 3'-phosphohydrolase. Because DAWM may be important in MS disability and progression, further histologic characterization is warranted. The MRI data were collected on 14 formalin-fixed MS brain samples that were then stained for myelin phospholipids, myelin proteins, astrocytes and axons. Diffusely abnormal white matter showed reduced myelin water fraction (-30%, p < 0.05 for 13 samples). Myelin phospholipids showed the most dramatic and consistent histologic reductions in staining optical density (-29% Luxol fast blue and -24% Weil's, p < 0.05 for 13 and 14 samples,respectively) with lesser myelin protein involvement (-11% myelin-associated glycoprotein, -10% myelin basic protein, -8% myelin-oligodendrocyte glycoprotein, -7% proteolipid protein, -5% 2',3'-cyclic-nucleotide 3'-phosphohydrolase, p < 0.05 for 3, 3, 1, 2, and 3 samples, respectively). Axonal involvement was intermediate. Diffusely abnormal white matter lipid and protein reductions occurred independently. These findings suggest a primary lipid abnormality in DAWM that exceeds protein loss and is accompanied by axonal degeneration. These phenomena may be important in MS pathogenesis and disease progression, which is prominent in individuals with DAWM.
Collapse
|
49
|
Du C, Xie X. G protein-coupled receptors as therapeutic targets for multiple sclerosis. Cell Res 2012; 22:1108-28. [PMID: 22664908 DOI: 10.1038/cr.2012.87] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) mediate most of our physiological responses to hormones, neurotransmitters and environmental stimulants. They are considered as the most successful therapeutic targets for a broad spectrum of diseases. Multiple sclerosis (MS) is an inflammatory disease that is characterized by immune-mediated demyelination and degeneration of the central nervous system (CNS). It is the leading cause of non-traumatic disability in young adults. Great progress has been made over the past few decades in understanding the pathogenesis of MS. Numerous data from animal and clinical studies indicate that many GPCRs are critically involved in various aspects of MS pathogenesis, including antigen presentation, cytokine production, T-cell differentiation, T-cell proliferation, T-cell invasion, etc. In this review, we summarize the recent findings regarding the expression or functional changes of GPCRs in MS patients or animal models, and the influences of GPCRs on disease severity upon genetic or pharmacological manipulations. Hopefully some of these findings will lead to the development of novel therapies for MS in the near future.
Collapse
Affiliation(s)
- Changsheng Du
- Laboratory of Receptor-Based BioMedicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | | |
Collapse
|
50
|
Jadasz JJ, Aigner L, Rivera FJ, Küry P. The remyelination Philosopher's Stone: stem and progenitor cell therapies for multiple sclerosis. Cell Tissue Res 2012; 349:331-47. [PMID: 22322424 DOI: 10.1007/s00441-012-1331-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 01/16/2012] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease that leads to oligodendrocyte loss and subsequent demyelination of the adult central nervous system (CNS). The pathology is characterized by transient phases of recovery during which remyelination can occur as a result of resident oligodendroglial precursor and stem/progenitor cell activation. However, myelin repair efficiency remains low urging the development of new therapeutical approaches that promote remyelination activities. Current MS treatments target primarily the immune system in order to reduce the relapse rate and the formation of inflammatory lesions, whereas no therapies exist in order to regenerate damaged myelin sheaths. During the last few years, several transplantation studies have been conducted with adult neural stem/progenitor cells and glial precursor cells to evaluate their potential to generate mature oligodendrocytes that can remyelinate axons. In parallel, modulation of the endogenous progenitor niche by neural and mesenchymal stem cell transplantation with the aim of promoting CNS progenitor differentiation and myelination has been studied. Here, we summarize these findings and discuss the properties and consequences of the various molecular and cell-mediated remyelination approaches. Moreover, we address age-associated intrinsic cellular changes that might influence the regenerative outcome. We also evaluate the extent to which these experimental treatments might increase the regeneration capacity of the demyelinated human CNS and hence be turned into future therapies.
Collapse
Affiliation(s)
- Janusz J Jadasz
- Department of Neurology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | | | | | | |
Collapse
|