1
|
Ganz T, Fainstein N, Theotokis P, Elgavish S, Vardi-Yaakov O, Lachish M, Sofer L, Zveik O, Grigoriadis N, Ben-Hur T. Targeting CNS myeloid infiltrates provides neuroprotection in a progressive multiple sclerosis model. Brain Behav Immun 2024; 122:497-509. [PMID: 39179123 DOI: 10.1016/j.bbi.2024.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/30/2024] [Accepted: 08/17/2024] [Indexed: 08/26/2024] Open
Abstract
Demyelination and axonal injury in chronic-progressive Multiple Sclerosis (MS) are presumed to be driven by a neurotoxic bystander effect of meningeal-based myeloid infiltrates. There is an unmet clinical need to attenuate disease progression in such forms of CNS-compartmentalized MS. The failure of systemic immune suppressive treatments has highlighted the need for neuroprotective and repair-inducing strategies. Here, we examined whether direct targeting of CNS myeloid cells and modulating their toxicity may prevent irreversible tissue injury in chronic immune-mediated demyelinating disease. To that end, we utilized the experimental autoimmune encephalomyelitis (EAE) model in Biozzi mice, a clinically relevant MS model. We continuously delivered intracerebroventricularly (ICV) a retinoic acid receptor alpha agonist (RARα), as a potent regulator of myeloid cells, in the chronic phase of EAE. We assessed disease severity and performed pathological evaluations, functional analyses of immune cells, and single-cell RNA sequencing on isolated spinal CD11b+ cells. Although initiating treatment in the chronic phase of the disease, the RARα agonist successfully improved clinical outcomes and prevented axonal loss. ICV RARα agonist treatment inhibited pro-inflammatory pathways and shifted CNS myeloid cells toward neuroprotective phenotypes without affecting peripheral infiltrating myeloid cell phenotypes, or peripheral immunity. The treatment regulated cell-death pathways across multiple myeloid cell populations and suppressed apoptosis, resulting in paradoxically marked increased neuroinflammatory infiltrates, consisting mainly of microglia and CNS / border-associated macrophages. This work establishes the notion of bystander neurotoxicity by CNS immune infiltrates in chronic demyelinating disease. Furthermore, it shows that targeting compartmentalized neuroinflammation by selective regulation of CNS myeloid cell toxicity and survival reduces irreversible tissue injury, and may serve as a novel disease-modifying approach.
Collapse
Affiliation(s)
- Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Paschalis Theotokis
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - Sharona Elgavish
- Info-CORE, Bioinformatics Unit of the 1-CORE, Hebrew University of Jerusalem, Israel
| | - Oriya Vardi-Yaakov
- Info-CORE, Bioinformatics Unit of the 1-CORE, Hebrew University of Jerusalem, Israel; Department of Bioinformatics, Jerusalem College of Technology, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Lihi Sofer
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nikolaos Grigoriadis
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
2
|
Kreiter D, Kalincik T, Hupperts R, Patti F, Spitaleri D, Foschi M, Surcinelli A, Maimone D, Yamout B, Khoury SJ, Lechner-Scott J, Ozakbas S, Gerlach O. Effectiveness of Disease-Modifying Treatment on Spinal Cord Lesion Formation in Relapse-Onset Multiple Sclerosis: An MSBase Registry Study. CNS Drugs 2024; 38:921-930. [PMID: 39242483 PMCID: PMC11486785 DOI: 10.1007/s40263-024-01115-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/30/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Spinal cord lesions in multiple sclerosis (MS) have considerable impact on disability. High-efficacy disease-modifying treatments (hDMTs) are associated with greater reduction of relapses and new brain lesions compared to low-efficacy treatments (lDMTs). Knowledge on the impact of DMTs on cord lesion formation is limited as these outcome measures were not included in MS treatment trials. This study aims to investigate whether hDMTs reduce the formation of cord lesions more effectively than lDMTs. METHODS Patients with relapse-onset MS, a cord magnetic resonance imaging (MRI) within 6 months before/after initiation of their first DMT and ≥1 cord MRI at follow-up (interval > 6 months) were extracted from the MSBase registry (ACTRN12605000455662). Patients treated with hDMTs ≥90% or lDMTs ≥90% of follow-up duration were considered the hDMT and lDMT groups, respectively. Matching was performed using propensity scores. Cox proportional hazards models were used to estimate the hazards of new cord lesions, brain lesions and relapses. RESULTS Ninety-four and 783 satisfied hDMT and lDMT group criteria, respectively. Seventy-seven hDMT patients were matched to 184 lDMT patients. In the hDMT group there was no evidence of reduction of new cord lesions (hazard ratio [HR] 0.99 [95% CI 0.51, 1.92], p = 0.97), while there were fewer new brain lesions (HR 0.22 [95% CI 0.10, 0.49], p < 0.001) and fewer relapses (HR 0.45 [95% CI 0.28, 0.72], p = 0.004). CONCLUSION A potential discrepancy exists in the effect of hDMTs over lDMTs in preventing spinal cord lesions versus brain lesions and relapses. While hDMTs provided a significant reduction for the latter when compared to lDMTs, there was no significant reduction in new spinal cord lesions.
Collapse
Affiliation(s)
- Daniel Kreiter
- Department of Neurology, Academic MS Center Zuyd, Zuyderland MC, Sittard-Geleen, The Netherlands.
- School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands.
| | - Tomas Kalincik
- Department of Neurology, Neuroimmunology Centre, Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, CORe, University of Melbourne, Melbourne, Australia
| | - Raymond Hupperts
- Department of Neurology, Academic MS Center Zuyd, Zuyderland MC, Sittard-Geleen, The Netherlands
- School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Francesco Patti
- Department of Medical and Surgical Sciences and Advanced Technologies, GF Ingrassia, Catania, Italy
- Multiple Sclerosis Unit, AOU Policlinico G Rodolico-San Marco, University of Catania, Catania, Italy
| | - Daniele Spitaleri
- Azienda Ospedaliera di Rilievo Nazionale San Giuseppe Moscati Avellino, Avellino, Italy
| | - Matteo Foschi
- Department of Neuroscience, MS Center, Neurology Unit, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, L'Aquila, Italy
| | - Andrea Surcinelli
- Department of Neuroscience, MS Center, Neurology Unit, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
| | - Davide Maimone
- Centro Sclerosi Multipla, UOC Neurologia, Azienda Ospedaliera Cannizzaro, Catania, Italy
| | - Bassem Yamout
- Nehme and Therese Tohme Multiple Sclerosis Center, American University of Beirut Medical Center, Beirut, Lebanon
- Harley Street Medical Center, Abu Dhabi, UAE
| | - Samia J Khoury
- Nehme and Therese Tohme Multiple Sclerosis Center, American University of Beirut Medical Center, Beirut, Lebanon
| | - Jeannette Lechner-Scott
- Hunter Medical Research Institute, University Newcastle, Newcastle, Australia
- Hunter New England Health, John Hunter Hospital, New Lambton, NSW, Australia
| | - Serkan Ozakbas
- Izmir University of Economics, Medical Point Hospital, Izmir, Turkey
- Multiple Sclerosis Research Association, Izmir, Turkey
| | - Oliver Gerlach
- Department of Neurology, Academic MS Center Zuyd, Zuyderland MC, Sittard-Geleen, The Netherlands
- School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
3
|
Abstract
The inability of the mammalian central nervous system (CNS) to undergo spontaneous regeneration has long been regarded as a central tenet of neurobiology. However, while this is largely true of the neuronal elements of the adult mammalian CNS, save for discrete populations of granule neurons, the same is not true of its glial elements. In particular, the loss of oligodendrocytes, which results in demyelination, triggers a spontaneous and often highly efficient regenerative response, remyelination, in which new oligodendrocytes are generated and myelin sheaths are restored to denuded axons. Yet remyelination in humans is not without limitation, and a variety of demyelinating conditions are associated with sustained and disabling myelin loss. In this work, we will (1) review the biology of remyelination, including the cells and signals involved; (2) describe when remyelination occurs and when and why it fails, including the consequences of its failure; and (3) discuss approaches for therapeutically enhancing remyelination in demyelinating diseases of both children and adults, both by stimulating endogenous oligodendrocyte progenitor cells and by transplanting these cells into demyelinated brain.
Collapse
Affiliation(s)
- Robin J M Franklin
- Altos Labs Cambridge Institute of Science, Cambridge CB21 6GH, United Kingdom
| | - Benedetta Bodini
- Sorbonne Université, Paris Brain Institute, CNRS, INSERM, Paris 75013, France
- Saint-Antoine Hospital, APHP, Paris 75012, France
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York 14642, USA
- University of Copenhagen Faculty of Medicine, Copenhagen 2200, Denmark
| |
Collapse
|
4
|
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
|
5
|
Evonuk KS, Wang S, Mattie J, Cracchiolo CJ, Mager R, Ferenčić Ž, Sprague E, Carrier B, Schofield K, Martinez E, Stewart Z, Petrosino T, Johnson GA, Yusuf I, Plaisted W, Naiman Z, Delp T, Carter L, Marušić S. Bruton's tyrosine kinase inhibition reduces disease severity in a model of secondary progressive autoimmune demyelination. Acta Neuropathol Commun 2023; 11:115. [PMID: 37438842 PMCID: PMC10337138 DOI: 10.1186/s40478-023-01614-w] [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: 05/23/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023] Open
Abstract
Bruton's tyrosine kinase (BTK) is an emerging target in multiple sclerosis (MS). Alongside its role in B cell receptor signaling and B cell development, BTK regulates myeloid cell activation and inflammatory responses. Here we demonstrate efficacy of BTK inhibition in a model of secondary progressive autoimmune demyelination in Biozzi mice with experimental autoimmune encephalomyelitis (EAE). We show that late in the course of disease, EAE severity could not be reduced with a potent relapse inhibitor, FTY720 (fingolimod), indicating that disease was relapse-independent. During this same phase of disease, treatment with a BTK inhibitor reduced both EAE severity and demyelination compared to vehicle treatment. Compared to vehicle treatment, late therapeutic BTK inhibition resulted in fewer spinal cord-infiltrating myeloid cells, with lower expression of CD86, pro-IL-1β, CD206, and Iba1, and higher expression of Arg1, in both tissue-resident and infiltrating myeloid cells, suggesting a less inflammatory myeloid cell milieu. These changes were accompanied by decreased spinal cord axonal damage. We show similar efficacy with two small molecule inhibitors, including a novel, highly selective, central nervous system-penetrant BTK inhibitor, GB7208. These results suggest that through lymphoid and myeloid cell regulation, BTK inhibition reduced neurodegeneration and disease progression during secondary progressive EAE.
Collapse
Affiliation(s)
| | - Sen Wang
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Josh Mattie
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - C. J. Cracchiolo
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Reine Mager
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Željko Ferenčić
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Ethan Sprague
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Brandon Carrier
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Kai Schofield
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Evelyn Martinez
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Zachary Stewart
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Tara Petrosino
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | | | - Isharat Yusuf
- Gossamer Bio, 3013 Science Park Road, Suite 200, San Diego, CA 92121 USA
| | - Warren Plaisted
- Gossamer Bio, 3013 Science Park Road, Suite 200, San Diego, CA 92121 USA
| | - Zachary Naiman
- Gossamer Bio, 3013 Science Park Road, Suite 200, San Diego, CA 92121 USA
| | - Timothy Delp
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| | - Laura Carter
- Gossamer Bio, 3013 Science Park Road, Suite 200, San Diego, CA 92121 USA
| | - Suzana Marušić
- Hooke Laboratories, LLC, 439 South Union Street, Lawrence, MA 01843 USA
| |
Collapse
|
6
|
Vainchtein ID, Alsema AM, Dubbelaar ML, Grit C, Vinet J, van Weering HRJ, Al‐Izki S, Biagini G, Brouwer N, Amor S, Baker D, Eggen BJL, Boddeke EWGM, Kooistra SM. Characterizing microglial gene expression in a model of secondary progressive multiple sclerosis. Glia 2023; 71:588-601. [PMID: 36377669 PMCID: PMC10100411 DOI: 10.1002/glia.24297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022]
Abstract
Multiple sclerosis (MS) is the most common inflammatory, demyelinating and neurodegenerative disease of the central nervous system in young adults. Chronic-relapsing experimental autoimmune encephalomyelitis (crEAE) in Biozzi ABH mice is an experimental model of MS. This crEAE model is characterized by an acute phase with severe neurological disability, followed by remission of disease, relapse of neurological disease and remission that eventually results in a chronic progressive phase that mimics the secondary progressive phase (SPEAE) of MS. In both MS and SPEAE, the role of microglia is poorly defined. We used a crEAE model to characterize microglia in the different phases of crEAE phases using morphometric and RNA sequencing analyses. At the initial, acute inflammation phase, microglia acquired a pro-inflammatory phenotype. At the remission phase, expression of standard immune activation genes was decreased while expression of genes associated with lipid metabolism and tissue remodeling were increased. Chronic phase microglia partially regain inflammatory gene sets and increase expression of genes associated with proliferation. Together, the data presented here indicate that microglia obtain different features at different stages of crEAE and a particularly mixed phenotype in the chronic stage. Understanding the properties of microglia that are present at the chronic phase of EAE will help to understand the role of microglia in secondary progressive MS, to better aid the development of therapies for this phase of the disease.
Collapse
Affiliation(s)
- Ilia D. Vainchtein
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Astrid M. Alsema
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Marissa L. Dubbelaar
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Corien Grit
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Jonathan Vinet
- Department of Biomedical, Metabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Hilmar R. J. van Weering
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Sarah Al‐Izki
- Department of NeuroimmunologyBlizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Nieske Brouwer
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Sandra Amor
- Department of NeuroimmunologyBlizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
- Department of PathologyVUMCAmsterdamThe Netherlands
| | - David Baker
- Department of NeuroimmunologyBlizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| | - Bart J. L. Eggen
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Erik W. G. M. Boddeke
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Department of Cellular and Molecular MedicineCenter for Healthy Ageing, University of CopenhagenCopenhagenDenmark
| | - Susanne M. Kooistra
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
7
|
Lariosa-Willingham K, Leonoudakis D, Bragge T, Tolppanen L, Nurmi A, Flanagan M, Gibson J, Wilson D, Stratton J, Lehtimäki KK, Miszczuk D. An in vivo accelerated developmental myelination model for testing promyelinating therapeutics. BMC Neurosci 2022; 23:30. [PMID: 35614392 PMCID: PMC9134688 DOI: 10.1186/s12868-022-00714-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 05/10/2022] [Indexed: 12/13/2022] Open
Abstract
Background Therapeutic agents stimulating the process of myelination could be beneficial for the treatment of demyelinating diseases, such as multiple sclerosis. The efficient translation of compounds promoting myelination in vitro to efficacy in vivo is inherently time-consuming and expensive. Thyroid hormones accelerate the differentiation and maturation of oligodendrocytes, thereby promoting myelination. Systemic administration of the thyroid hormone thyroxine (T4) accelerates brain maturation, including myelination, during early postnatal development. The objective of this study was to validate an animal model for rapid testing of promyelinating therapeutic candidates for their effects on early postnatal development by using T4 as a reference compound. Methods Daily subcutaneous injections of T4 were given to Sprague Dawley rat pups from postnatal day (PND) 2 to PND10. Changes in white matter were determined at PND10 using diffusion tensor magnetic resonance imaging (DTI). Temporal changes in myelination from PND3 to PND11 were also assessed by quantifying myelin basic protein (MBP) expression levels in the brain using the resonance Raman spectroscopy/enzyme-linked immunosorbent assay (RRS-ELISA) and quantitative immunohistochemistry. Results DTI of white matter tracts showed significantly higher fractional anisotropy in the internal capsule of T4-treated rat pups. The distribution of total FA values in the forebrain was significantly shifted towards higher values in the T4-treated group, suggesting increased myelination. In vivo imaging data were supported by in vitro observations, as T4 administration significantly potentiated the developmental increase in MBP levels in brain lysates starting from PND8. MBP levels in the brain of animals that received treatment for 9 days correlated with the FA metric determined in the same pups in vivo a day earlier. Furthermore, accelerated developmental myelination following T4 administration was confirmed by immunohistochemical staining for MBP in coronal brain sections of treated rat pups. Conclusions T4-treated rat pups had increased MBP expression levels and higher MRI fractional anisotropy values, both indications of accelerated myelination. This simple developmental myelination model affords a rapid test of promyelinating activity in vivo within several days, which could facilitate in vivo prescreening of candidate therapeutic compounds for developmental hypomyelinating diseases. Further research will be necessary to assess the utility of this platform for screening promyelination compounds in more complex demyelination disease models, such us multiple sclerosis. Supplementary information The online version contains supplementary material available at 10.1186/s12868-022-00714-y.
Collapse
Affiliation(s)
| | | | - Timo Bragge
- Charles River Discovery Services, Neulaniementie 4, 70210, Kuopio, Finland
| | - Laura Tolppanen
- Charles River Discovery Services, Neulaniementie 4, 70210, Kuopio, Finland
| | - Antti Nurmi
- Charles River Discovery Services, Neulaniementie 4, 70210, Kuopio, Finland
| | | | | | - David Wilson
- Teva Pharmaceutical Industries Ltd, Redwood City, CA, 94063, USA
| | | | - Kimmo K Lehtimäki
- Charles River Discovery Services, Neulaniementie 4, 70210, Kuopio, Finland
| | - Diana Miszczuk
- Charles River Discovery Services, Neulaniementie 4, 70210, Kuopio, Finland
| |
Collapse
|
8
|
Goldfarb S, Fainstein N, Ganz T, Vershkov D, Lachish M, Ben-Hur T. Electric neurostimulation regulates microglial activation via retinoic acid receptor α signaling. Brain Behav Immun 2021; 96:40-53. [PMID: 33989746 DOI: 10.1016/j.bbi.2021.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/20/2021] [Accepted: 05/09/2021] [Indexed: 12/14/2022] Open
Abstract
Brain stimulation by electroconvulsive therapy is effective in neuropsychiatric disorders by unknown mechanisms. Microglial toxicity plays key role in neuropsychiatric, neuroinflammatory and degenerative diseases. We examined the mechanism by which electroconvulsive seizures (ECS) regulates microglial phenotype and response to stimuli. Microglial responses were examined by morphological analysis, Iba1 and cytokine expression. ECS did not affect resting microglial phenotype or morphology but regulated their activation by Lipopolysaccharide stimulation. Microglia were isolated after ECS or sham sessions in naïve mice for transcriptome analysis. RNA sequencing identified 141 differentially expressed genes. ECS modulated multiple immune-associated gene families and attenuated neurotoxicity-associated gene expression. Blood brain barrier was examined by injecting Biocytin-TMR tracer. There was no breakdown of the BBB, nor increase in gene-signature of peripheral monocytes, suggesting that ECS effect is mainly on resident microglia. Unbiased analysis of regulatory sequences identified the induction of microglial retinoic acid receptor α (RARα) gene expression and a putative common RARα-binding motif in multiple ECS-upregulated genes. The effects of AM580, a selective RARα agonist on microglial response to LPS was examined in vitro. AM580 prevented LPS-induced cytokine expression and reactive oxygen species production. Chronic murine experimental autoimmune encephalomyelitis (EAE) was utilized to confirm the role RARα signaling as mediator of ECS-induced transcriptional pathway in regulating microglial toxicity. Continuous intracerebroventricular delivery of AM580 attenuated effectively EAE severity. In conclusion, ECS regulates CNS innate immune system responses by activating microglial retinoic acid receptor α pathway, signifying a novel therapeutic approach for chronic neuroinflammatory, neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Smadar Goldfarb
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Dan Vershkov
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel; The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
9
|
Chu T, Shields LB, Zeng W, Zhang YP, Wang Y, Barnes GN, Shields CB, Cai J. Dynamic glial response and crosstalk in demyelination-remyelination and neurodegeneration processes. Neural Regen Res 2021; 16:1359-1368. [PMID: 33318418 PMCID: PMC8284258 DOI: 10.4103/1673-5374.300975] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/09/2020] [Accepted: 08/28/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis is an autoimmune disease in which the immune system attacks the myelin sheath in the central nervous system. It is characterized by blood-brain barrier dysfunction throughout the course of multiple sclerosis, followed by the entry of immune cells and activation of local microglia and astrocytes. Glial cells (microglia, astrocytes, and oligodendrocyte lineage cells) are known as the important mediators of neuroinflammation, all of which play major roles in the pathogenesis of multiple sclerosis. Network communications between glial cells affect the activities of oligodendrocyte lineage cells and influence the demyelination-remyelination process. A finely balanced glial response may create a favorable lesion environment for efficient remyelination and neuroregeneration. This review focuses on glial response and neurodegeneration based on the findings from multiple sclerosis and major rodent demyelination models. In particular, glial interaction and molecular crosstalk are discussed to provide insights into the potential cell- and molecule-specific therapeutic targets to improve remyelination and neuroregeneration.
Collapse
Affiliation(s)
- Tianci Chu
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Lisa B.E. Shields
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, USA
| | - Wenxin Zeng
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Yi Ping Zhang
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, USA
| | - Yuanyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Gregory N. Barnes
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Christopher B. Shields
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jun Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
10
|
Nishri Y, Fainstein N, Goldfarb S, Hampton D, Macrini C, Meinl E, Chandran S, Ben-Hur T. Modeling compartmentalized chronic immune-mediated demyelinating CNS disease in the Biozzi ABH mouse. J Neuroimmunol 2021; 356:577582. [PMID: 33910137 DOI: 10.1016/j.jneuroim.2021.577582] [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: 02/08/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
We explored whether experimental autoimmune encephalomyelitis (EAE) in Biozzi mice recapitulates temporal dynamics of tissue injury, immune-pathogenesis and CNS compartmentalization occurring in progressive multiple sclerosis (MS). Chronic EAE exhibited relapsing and progressing disease, partial closure of BBB, reduced tissue inflammatory activity, and development of meningeal ectopic lymphoid tissue, directly opposing (potentially driving) spinal subpial demyelinated plaques. A T cell predominant disease during relapses transformed into a B cell predominant disease in late chronic EAE, with high serum anti-MOG reactivity. Thus, late chronic Biozzi EAE recapitulates essential features of progressive MS, and is suitable for developing disease modifying and regenerative therapies.
Collapse
Affiliation(s)
- Yossi Nishri
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Smadar Goldfarb
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - David Hampton
- Centre for Clinical Brain Sciences, MS Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Caterina Macrini
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, MS Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
11
|
Becquart P, Vilariño-Güell C, Quandt JA. Enhanced expression of complement and microglial-specific genes prior to clinical progression in the MOG-experimental autoimmune encephalomyelitis model of multiple sclerosis. Brain Res Bull 2020; 165:63-69. [PMID: 32979467 DOI: 10.1016/j.brainresbull.2020.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022]
Abstract
Understanding the biological changes responsible for failures in repair and the development of progressive MS is paramount for therapeutic intervention. In a well characterized experimental autoimmune encephalomyelitis (EAE) model of MS the clinical phenotype features an acute attack with partial recovery followed by a chronic or progressive disease phase. Neuropathology-focused gene expression profiles were generated from spinal cord, hindbrain and forebrain of mice 25 days after the induction of EAE, the time when recovery plateaus and transitions to a chronic or worsening phase. Differences in gene expression were most pronounced in the spinal cord of EAE mice compared to sham-immunized animals, with a subset of genes also found to be differentially expressed in the hindbrain and the forebrain, albeit with smaller fold-changes in expression. Our data suggests that changes in complement components, chemoattractant cytokines and especially enrichment in microglial cells may be the primary drivers of processes that limit recovery in EAE.
Collapse
Affiliation(s)
- Pierre Becquart
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Carles Vilariño-Güell
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Jacqueline A Quandt
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
12
|
Goldfarb S, Fainstein N, Ben-Hur T. Electroconvulsive stimulation attenuates chronic neuroinflammation. JCI Insight 2020; 5:137028. [PMID: 32780728 PMCID: PMC7526446 DOI: 10.1172/jci.insight.137028] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022] Open
Abstract
Electroconvulsive therapy is highly effective in resistant depression by unknown mechanisms. Microglial toxicity was suggested to mediate depression and plays key roles in neuroinflammatory and degenerative diseases, where there is critical shortage in therapies. We examined the effects of electroconvulsive seizures (ECS) on chronic neuroinflammation and microglial neurotoxicity. Electric brain stimulation inducing full tonic-clonic seizures during chronic relapsing-progressive experimental autoimmune encephalomyelitis (EAE) reduced spinal immune cell infiltration, reduced myelin and axonal loss, and prevented clinical deterioration. Using the transfer EAE model, we examined the effect of ECS on systemic immune response in donor mice versus ECS effect on CNS innate immune activity in recipient mice. ECS did not affect encephalitogenicity of systemic T cells, but it targeted the CNS directly to inhibit T cell-induced neuroinflammation. In vivo and ex vivo assays indicated that ECS suppressed microglial neurotoxicity by reducing inducible NOS expression, nitric oxide, and reactive oxygen species (ROS) production, and by reducing CNS oxidative stress. Microglia from ECS-treated EAE mice expressed less T cell stimulatory and chemoattractant factors. Our findings indicate that electroconvulsive therapy targets the CNS innate immune system to reduce neuroinflammation by attenuating microglial neurotoxicity. These findings signify a potentially novel therapeutic approach for chronic neuroinflammatory, neuropsychiatric, and neurodegenerative diseases.
Collapse
|
13
|
Milovanovic J, Arsenijevic A, Stojanovic B, Kanjevac T, Arsenijevic D, Radosavljevic G, Milovanovic M, Arsenijevic N. Interleukin-17 in Chronic Inflammatory Neurological Diseases. Front Immunol 2020; 11:947. [PMID: 32582147 PMCID: PMC7283538 DOI: 10.3389/fimmu.2020.00947] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
A critical role for IL-17, a cytokine produced by T helper 17 (Th17) cells, has been indicated in the pathogenesis of chronic inflammatory and autoimmune diseases. A positive effect of blockade of IL-17 secreted by autoreactive T cells has been shown in various inflammatory diseases. Several cytokines, whose production is affected by environmental factors, control Th17 differentiation and its maintenance in tissues during chronic inflammation. The roles of IL-17 in the pathogenesis of chronic neuroinflammatory conditions, multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE), Alzheimer's disease, and ischemic brain injury are reviewed here. The role of environmental stimuli in Th17 differentiation is also summarized, highlighting the role of viral infection in the regulation of pathogenic T helper cells in EAE.
Collapse
Affiliation(s)
- Jelena Milovanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Bojana Stojanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Tatjana Kanjevac
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Gordana Radosavljevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Marija Milovanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
14
|
Continuous Immune-Modulatory Effects of Human Olig2+ Precursor Cells Attenuating a Chronic-Active Model of Multiple Sclerosis. Mol Neurobiol 2019; 57:1021-1034. [DOI: 10.1007/s12035-019-01802-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 07/10/2019] [Indexed: 01/17/2023]
|
15
|
Perianes-Cachero A, Lobo MVT, Hernández-Pinto AM, Busto R, Lasunción-Ripa MA, Arilla-Ferreiro E, Puebla-Jiménez L. Oxidative Stress and Lymphocyte Alterations in Chronic Relapsing Experimental Allergic Encephalomyelitis in the Rat Hippocampus and Protective Effects of an Ethanolamine Phosphate Salt. Mol Neurobiol 2019; 57:860-878. [DOI: 10.1007/s12035-019-01774-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/01/2019] [Indexed: 01/20/2023]
|
16
|
Baker D, Nutma E, O'Shea H, Cooke A, Orian JM, Amor S. Autoimmune encephalomyelitis in NOD mice is not initially a progressive multiple sclerosis model. Ann Clin Transl Neurol 2019; 6:1362-1372. [PMID: 31402611 PMCID: PMC6689692 DOI: 10.1002/acn3.792] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/12/2019] [Accepted: 04/23/2019] [Indexed: 01/31/2023] Open
Abstract
Objective Despite progress in treating relapsing multiple sclerosis (MS), effective inhibition of nonrelapsing progressive MS is an urgent, unmet, clinical need. Animal models of MS, such as experimental autoimmune encephalomyelitis (EAE), provide valuable tools to examine the mechanisms contributing to disease and may be important for developing rational therapeutic approaches for treatment of progressive MS. It has been suggested that myelin oligodendrocyte glycoprotein (MOG) peptide residues 35‐55 (MOG35‐55)‐induced EAE in nonobese diabetic (NOD) mice resembles secondary progressive MS. The objective was to determine whether the published data merits such claims. Methods Induction and monitoring of EAE in NOD mice and literature review. Results It is evident that the NOD mouse model lacks validity as a progressive MS model as the individual course seems to be an asynchronous, relapsing‐remitting neurodegenerative disease, characterized by increasingly poor recovery from relapse. The seemingly progressive course seen in group means of clinical score is an artifact of data handling and interpretation. Interpretation Although MOG35‐55‐induced EAE in NOD mice may provide some clues about approaches to block neurodegeneration associated with the inflammatory penumbra as lesions form, it should not be used to justify trials in people with nonactive, progressive MS. This adds further support to the view that drug studies in animals should universally adopt transparent raw data deposition as part of the publication process, such that claims can adequately be interrogated. This transparency is important if animal‐based science is to remain a credible part of translational research in MS.
Collapse
Affiliation(s)
- David Baker
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - Erik Nutma
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - Helen O'Shea
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, United Kingdom
| | - Anne Cooke
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, United Kingdom
| | - Jacqueline M Orian
- La Trobe Institute of Molecular Sciences La Trobe University, Bundoora, Victoria, 3086, Australia
| | - Sandra Amor
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom.,Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam, 1081HV, The Netherlands
| |
Collapse
|
17
|
SFX-01 reduces residual disability after experimental autoimmune encephalomyelitis. Mult Scler Relat Disord 2019; 30:257-261. [DOI: 10.1016/j.msard.2019.02.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/01/2018] [Accepted: 02/25/2019] [Indexed: 11/22/2022]
|
18
|
Madhu K, T P, S M. Bacoside-A inhibits inflammatory cytokines and chemokine in experimental autoimmune encephalomyelitis. Biomed Pharmacother 2018; 109:1339-1345. [PMID: 30551384 DOI: 10.1016/j.biopha.2018.10.188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/27/2018] [Accepted: 10/31/2018] [Indexed: 11/29/2022] Open
Abstract
Chronic inflammation of the myelin sheath is the crucial event behind the progression of multiple sclerosis (MS). Bacoside-A is one of the major constituents obtained from Bacopa monerii (L.) Wettst., and possess neuroprotective as well as anti-inflammatory actions. The current study explores the effect of Bacoside-A in acute and chronic models of Experimental Autoimmune Encephalomyelitis (EAE). The results indicate that the Bacoside-A treated mice produced a significant reduction in disease score compared to disease control in both models. The treatment with Bacoside-A downregulated the inflammatory cytokines (IL-6, IL-17a, and TNFα) and inflammatory chemokine CCL-5 in EAE mice. On the other hand, Bacoside-A treated mice showed a nonsignificant effect on promoting the expressions of NCAM, BDNF1, and FOXP3 in acute and chronic models of EAE. Histopathological analysis revealed that the Bacoside-A treated mice at a dose of 10 mg/kg exhibited a significant reduction in cellular infiltrations, cellular changes, and demyelination in cerebral tissues, but unable to protect at a higher dose in both models. In conclusion, Bacoside-A can able to inhibit the progression of EAE may be by the inhibition of inflammatory cytokines and chemokine evolved during active EAE.
Collapse
Affiliation(s)
- Krishnadas Madhu
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Bengaluru, India.
| | - Prakash T
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Bengaluru, India.
| | - Maya S
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Bengaluru, India
| |
Collapse
|
19
|
Burrows DJ, McGown A, Jain SA, De Felice M, Ramesh TM, Sharrack B, Majid A. Animal models of multiple sclerosis: From rodents to zebrafish. Mult Scler 2018; 25:306-324. [PMID: 30319015 DOI: 10.1177/1352458518805246] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated demyelinating disease of the central nervous system. Animal models of MS have been critical for elucidating MS pathological mechanisms and how they may be targeted for therapeutic intervention. Here we review the most commonly used animal models of MS. Although these animal models cannot fully replicate the MS disease course, a number of models have been developed to recapitulate certain stages. Experimental autoimmune encephalomyelitis (EAE) has been used to explore neuroinflammatory mechanisms and toxin-induced demyelinating models to further our understanding of oligodendrocyte biology, demyelination and remyelination. Zebrafish models of MS are emerging as a useful research tool to validate potential therapeutic candidates due to their rapid development and amenability to genetic manipulation.
Collapse
Affiliation(s)
- David John Burrows
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Alexander McGown
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Saurabh A Jain
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Milena De Felice
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Tennore M Ramesh
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Basil Sharrack
- Academic Department of Neuroscience, The Sheffield NIHR Translational Neuroscience Biomedical Research Centre, University of Sheffield, Sheffield, UK
| | - Arshad Majid
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK/Academic Department of Neuroscience, The Sheffield NIHR Translational Neuroscience Biomedical Research Centre, University of Sheffield, Sheffield, UK
| |
Collapse
|
20
|
Silva BA, Leal MC, Farías MI, Avalos JC, Besada CH, Pitossi FJ, Ferrari CC. A new focal model resembling features of cortical pathology of the progressive forms of multiple sclerosis: Influence of innate immunity. Brain Behav Immun 2018; 69:515-531. [PMID: 29378262 DOI: 10.1016/j.bbi.2018.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/10/2018] [Accepted: 01/19/2018] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory and demyelinating disease of unknown aetiology that causes neurological disabilities in young adults. MS displays different clinical patterns, including recurrent episodes with remission periods ("relapsing-remitting MS" (RRMS)), which can progress over several years to a secondary progressive form (SPMS). However, 10% of patients display persistent progression at the onset of disease ("primary progressive MS" (PPMS)). Currently, no specific therapeutic agents are available for the progressive forms, mainly because the underlying pathogenic mechanisms are not clear and because no animal models have been specifically developed for these forms. The development of MS animal models is required to clarify the pathological mechanisms and to test novel therapeutic agents. In the present work, we overexpressed interleukin 1 beta (IL-1β) in the cortex to develop an animal model reflecting the main pathological hallmarks of MS. The treated animals presented with neuroinflammation, demyelination, glial activation, and neurodegeneration along with cognitive symptoms and MRI images consistent with MS pathology. We also demonstrated the presence of meningeal inflammation close to cortical lesions, with characteristics similar to those described in MS patients. Systemic pro-inflammatory stimulation caused a flare-up of the cortical lesions and behavioural symptoms, including impairment of working memory and the appearance of anxiety-like symptoms. Our work demonstrated induced cortical lesions, reflecting the main histopathological hallmarks and cognitive impairments characterizing the cortical pathology described in MS patients with progressive forms of the disease.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Institute of Basic Science and Experimental Medicine (ICBME), University Institute, Italian Hospital, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | - María Celeste Leal
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | - María Isabel Farías
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | | | | | - Fernando Juan Pitossi
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Institute of Basic Science and Experimental Medicine (ICBME), University Institute, Italian Hospital, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina.
| |
Collapse
|
21
|
Bjelobaba I, Begovic-Kupresanin V, Pekovic S, Lavrnja I. Animal models of multiple sclerosis: Focus on experimental autoimmune encephalomyelitis. J Neurosci Res 2018; 96:1021-1042. [PMID: 29446144 DOI: 10.1002/jnr.24224] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/15/2018] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
Multiple sclerosis (MS) is a chronic, progressive disorder of the central nervous system (CNS) that affects more than two million people worldwide. Several animal models resemble MS pathology; the most employed are experimental autoimmune encephalomyelitis (EAE) and toxin- and/or virus-induced demyelination. In this review we will summarize our knowledge on the utility of different animal models in MS research. Although animal models cannot replicate the complexity and heterogeneity of the MS pathology, they have proved to be useful for the development of several drugs approved for treatment of MS patients. This review focuses on EAE because it represents both clinical and pathological features of MS. During the past decades, EAE has been effective in illuminating various pathological processes that occur during MS, including inflammation, CNS penetration, demyelination, axonopathy, and neuron loss mediated by immune cells.
Collapse
Affiliation(s)
- Ivana Bjelobaba
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | | | - Sanja Pekovic
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
22
|
McGinley AM, Edwards SC, Raverdeau M, Mills KHG. Th17 cells, γδ T cells and their interplay in EAE and multiple sclerosis. J Autoimmun 2018; 87:S0896-8411(18)30007-6. [PMID: 29395738 DOI: 10.1016/j.jaut.2018.01.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 01/09/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis (MS) that shares many features with the human disease. This review will focus on the role of IL-17-secreting CD4 and γδ T cells in EAE and MS, the plasticity of Th17 cells in vivo and the application of these findings to the understating of the pathogenesis and the development of new treatments for MS. There is convincing evidence that IL-17-secreting CD4 T cells (Th17 cells) and IL-17-secreting γδ T cells play a critical pathogenic role in central nervous system (CNS) inflammation in EAE and MS. Indeed a significant number of the major discoveries on the pathogenic role of IL-17-secreting T cells in autoimmunity were made in the EAE model. These included the first demonstration that IL-23-activated IL-17-secreting T cells are the key T cells in driving autoimmune disease pathology. Although the early studies on IL-17 focused on Th17 cells, it was later demonstrated that γδ T cells were an important early source of IL-17 and IL-21 that helped amplify IL-17 production by Th17 cells in autoimmune diseases. Furthermore, it emerged that Th1 cells can also have encephalitogenic activity and that there was considerable plasticity in these T cell responses, with Th17 cells reverting to a Th1 phenotype in vivo. This questioned the pathogenic role of IL-17 and suggested that other cytokines, such as IFN-γ, GM-CSF and TNF, may be important. Nevertheless, biological drugs that target the IL-23-IL-17 pathway are highly effective in treating human psoriasis and are showing promise in the treatment of relapsing remitting MS and other T-cell mediated autoimmune diseases.
Collapse
Affiliation(s)
- Aoife M McGinley
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Sarah C Edwards
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Mathilde Raverdeau
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
23
|
Systematic screening of generic drugs for progressive multiple sclerosis identifies clomipramine as a promising therapeutic. Nat Commun 2017; 8:1990. [PMID: 29259169 PMCID: PMC5736601 DOI: 10.1038/s41467-017-02119-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 11/08/2017] [Indexed: 01/05/2023] Open
Abstract
The treatment of progressive multiple sclerosis (MS) is unsatisfactory. One reason is that the drivers of disease, which include iron-mediated neurotoxicity, lymphocyte activity, and oxidative stress, are not simultaneously targeted. Here we present a systematic screen to identify generic, orally available medications that target features of progressive MS. Of 249 medications that cross the blood–brain barrier, 35 prevent iron-mediated neurotoxicity in culture. Of these, several antipsychotics and antidepressants strongly reduce T-cell proliferation and oxidative stress. We focus on the antidepressant clomipramine and found that it additionally inhibits B-lymphocyte activity. In mice with experimental autoimmune encephalomyelitis, a model of MS, clomipramine ameliorates clinical signs of acute and chronic phases. Histologically, clomipramine reduces inflammation and microglial activation, and preserves axonal integrity. In summary, we present a systematic approach to identify generic medications for progressive multiple sclerosis with the potential to advance rapidly into clinical trials, and we highlight clomipramine for further development. Progressive multiple sclerosis is an inflammatory and degenerative disease of the central nervous system, for which effective treatment is lacking. The authors carry out a screen to identify orally available generic medications, and show that the antidepressant clomipramine reduces pathology in mouse models.
Collapse
|
24
|
Peferoen LAN, Breur M, van de Berg S, Peferoen-Baert R, Boddeke EHWGM, van der Valk P, Pryce G, van Noort JM, Baker D, Amor S. Ageing and recurrent episodes of neuroinflammation promote progressive experimental autoimmune encephalomyelitis in Biozzi ABH mice. Immunology 2016; 149:146-56. [PMID: 27388634 DOI: 10.1111/imm.12644] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 01/02/2023] Open
Abstract
Current therapies for multiple sclerosis (MS) reduce the frequency of relapses by modulating adaptive immune responses but fail to limit the irreversible neurodegeneration driving progressive disability. Experimental autoimmune encephalomyelitis (EAE) in Biozzi ABH mice recapitulates clinical features of MS including relapsing-remitting episodes and secondary-progressive disability. To address the contribution of recurrent inflammatory events and ageing as factors that amplify progressive neurological disease, we examined EAE in 8- to 12-week-old and 12-month-old ABH mice. Compared with the relapsing-remitting (RREAE) and secondary progressive (SPEAE) EAE observed in young mice, old mice developed progressive disease from onset (PEAE) associated with pronounced axonal damage and increased numbers of CD3(+) T cells and microglia/macrophages, but not B cells. Whereas the clinical neurological features of PEAE and SPEAE were comparable, the pathology was distinct. SPEAE was associated with significantly reduced perivascular infiltrates and T-cell numbers in the central nervous system (CNS) compared with PEAE and the acute phase of RREAE. In contrast to perivascular infiltrates that declined during progression from RREAE into SPEAE, the numbers of microglia clusters remained constant. Similar to what is observed during MS, the microglia clusters emerging during EAE were associated with axonal damage and oligodendrocytes expressing heat-shock protein B5, but not lymphocytes. Taken together, our data reveal that the course of EAE is dependent on the age of the mice. Younger mice show a relapsing-remitting phase followed by progressive disease, whereas old mice immediately show progression. This indicates that recurrent episodes of inflammation in the CNS, as well as age, contribute to progressive neurological disease.
Collapse
Affiliation(s)
- Laura A N Peferoen
- Pathology Department, VU University Medical Centre, Amsterdam, the Netherlands
| | - Marjolein Breur
- Pathology Department, VU University Medical Centre, Amsterdam, the Netherlands
| | - Sarah van de Berg
- Pathology Department, VU University Medical Centre, Amsterdam, the Netherlands
| | | | - Erik H W G M Boddeke
- Department of Neuroscience, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Paul van der Valk
- Pathology Department, VU University Medical Centre, Amsterdam, the Netherlands
| | - Gareth Pryce
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - David Baker
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sandra Amor
- Pathology Department, VU University Medical Centre, Amsterdam, the Netherlands.,Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
25
|
Sevastou I, Pryce G, Baker D, Selwood DL. Characterisation of Transcriptional Changes in the Spinal Cord of the Progressive Experimental Autoimmune Encephalomyelitis Biozzi ABH Mouse Model by RNA Sequencing. PLoS One 2016; 11:e0157754. [PMID: 27355629 PMCID: PMC4927105 DOI: 10.1371/journal.pone.0157754] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/04/2016] [Indexed: 11/30/2022] Open
Abstract
Multiple sclerosis (MS) is a debilitating immune-mediated neurological disorder affecting young adults. MS is primarily relapsing-remitting, but neurodegeneration and disability accumulate from disease onset. The most commonly used mouse MS models exhibit a monophasic immune response with fast accumulation of neurological damage that does not allow the study of progressive neurodegeneration. The chronic relapsing and secondary progressive EAE (pEAE) Biozzi ABH mouse model of MS exhibits a reproducible relapsing-remitting disease course that slowly accumulates permanent neurological deficit and develops a post-relapsing progressive disease that permits the study of demyelination and neurodegeneration. RNA sequencing (RNAseq) was used to explore global gene expression in the pEAE Biozzi ABH mouse. Spinal cord tissue RNA from pEAE Biozzi ABH mice and healthy age-matched controls was sequenced. 2,072 genes were differentially expressed (q<0.05) from which 1,397 were significantly upregulated and 675 were significantly downregulated. This hypothesis-free investigation characterised the genomic changes that describe the pEAE mouse model. The differentially expressed genes revealed a persistent immunoreactant phenotype, combined with downregulation of the cholesterol biosynthesis superpathway and the LXR/RXR activation pathway. Genes differentially expressed include the myelination genes Slc17a7, Ugt8A and Opalin, the neuroprotective genes Sprr1A, Osm and Wisp2, as well as genes identified as MS risk factors, including RGs14 and Scap2. Novel genes with unestablished roles in EAE or MS were also identified. The identification of differentially expressed novel genes and genes involved in MS pathology, opens the door to their functional study in the pEAE mouse model which recapitulates some of the important clinical features of progressive MS.
Collapse
Affiliation(s)
- Ioanna Sevastou
- Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Science, London, WC1E 6BT, United Kingdom
| | - Gareth Pryce
- Neuroimmmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT, United Kingdom
| | - David Baker
- Neuroimmmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT, United Kingdom
| | - David L. Selwood
- Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Science, London, WC1E 6BT, United Kingdom
- * E-mail:
| |
Collapse
|
26
|
Duncan ID, Radcliff AB. Inherited and acquired disorders of myelin: The underlying myelin pathology. Exp Neurol 2016; 283:452-75. [PMID: 27068622 PMCID: PMC5010953 DOI: 10.1016/j.expneurol.2016.04.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 01/26/2023]
Abstract
Remyelination is a major therapeutic goal in human myelin disorders, serving to restore function to demyelinated axons and providing neuroprotection. The target disorders that might be amenable to the promotion of this repair process are diverse and increasing in number. They range primarily from those of genetic, inflammatory to toxic origin. In order to apply remyelinating strategies to these disorders, it is essential to know whether the myelin damage results from a primary attack on myelin or the oligodendrocyte or both, and whether indeed these lead to myelin breakdown and demyelination. In some disorders, myelin sheath abnormalities are prominent but demyelination does not occur. This review explores the range of human and animal disorders where myelin pathology exists and focusses on defining the myelin changes in each and their cause, to help define whether they are targets for myelin repair therapy. We reviewed myelin disorders of the CNS in humans and animals. Myelin damage results from primary attack on the oligodendrocyte or myelin sheath. All major categories of disease can affect CNS myelin. Myelin vacuolation is common, yet does not always result in demyelination.
Collapse
Affiliation(s)
- Ian D Duncan
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States.
| | - Abigail B Radcliff
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
27
|
Abstract
The inability of the mammalian central nervous system (CNS) to undergo spontaneous regeneration has long been regarded as a central tenet of neurobiology. However, although this is largely true of the neuronal elements of the adult mammalian CNS, save for discrete populations of granular neurons, the same is not true of its glial elements. In particular, the loss of oligodendrocytes, which results in demyelination, triggers a spontaneous and often highly efficient regenerative response, remyelination, in which new oligodendrocytes are generated and myelin sheaths are restored to denuded axons. Yet, remyelination in humans is not without limitation, and a variety of demyelinating conditions are associated with sustained and disabling myelin loss. In this review, we will review the biology of remyelination, including the cells and signals involved; describe when remyelination occurs and when and why it fails and the consequences of its failure; and discuss approaches for therapeutically enhancing remyelination in demyelinating diseases of both children and adults, both by stimulating endogenous oligodendrocyte progenitor cells and by transplanting these cells into demyelinated brain.
Collapse
Affiliation(s)
- Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York 14642 University of Copenhagen Faculty of Medicine, Copenhagen 2200, Denmark
| |
Collapse
|
28
|
Ramaglia V, Jackson SJ, Hughes TR, Neal JW, Baker D, Morgan BP. Complement activation and expression during chronic relapsing experimental autoimmune encephalomyelitis in the Biozzi ABH mouse. Clin Exp Immunol 2015; 180:432-41. [PMID: 25619542 DOI: 10.1111/cei.12595] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2015] [Indexed: 12/29/2022] Open
Abstract
Chronic relapsing experimental autoimmune encephalomyelitis (crEAE) in mice recapitulates many of the clinical and histopathological features of human multiple sclerosis (MS), making it a preferred model for the disease. In both, adaptive immunity and anti-myelin T cells responses are thought to be important, while in MS a role for innate immunity and complement has emerged. Here we sought to test whether complement is activated in crEAE and important for disease. Disease was induced in Biozzi ABH mice that were terminated at different stages of the disease to assess complement activation and local complement expression in the central nervous system. Complement activation products were abundant in all spinal cord areas examined in acute disease during relapse and in the progressive phase, but were absent in early disease remission, despite significant residual clinical disease. Local expression of C1q and C3 was increased at all stages of disease, while C9 expression was increased only in acute disease; expression of the complement regulators CD55, complement receptor 1-related gene/protein y (Crry) and CD59a was reduced at all stages of the disease compared to naive controls. These data show that complement is activated in the central nervous system in the model and suggest that it is a suitable candidate for exploring whether anti-complement agents might be of benefit in MS.
Collapse
Affiliation(s)
- V Ramaglia
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - S J Jackson
- Neuroinflammation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - T R Hughes
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - J W Neal
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - D Baker
- Neuroinflammation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - B P Morgan
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
29
|
Baker D, Amor S. Experimental autoimmune encephalomyelitis is a good model of multiple sclerosis if used wisely. Mult Scler Relat Disord 2014; 3:555-64. [PMID: 26265267 DOI: 10.1016/j.msard.2014.05.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 05/01/2014] [Accepted: 05/05/2014] [Indexed: 02/07/2023]
Abstract
Although multiple sclerosis is a uniquely human disease, many pathological features can be induced in experimental autoimmune encephalomyelitis (EAE) models following induction of central nervous system-directed autoimmunity. Whilst it is an imperfect set of models, EAE can be used to identify pathogenic mechanisms and therapeutics. However, the failure to translate many treatments from EAE into human benefit has led some to question the validity of the EAE model. Whilst differences in biology between humans and other species may account for this, it is suggested here that the failure to translate may be considerably influenced by human activity. Basic science contributes to failings in aspects of experimental design and over-interpretation of results and lack of transparency and reproducibility of the studies. Importantly issues in trial design by neurologists and other actions of the pharmaceutical industry destine therapeutics to failure and terminate basic science projects. However animal, particularly mechanism-orientated, studies have increasingly identified useful treatments and provided mechanistic ideas on which most hypothesis-led clinical research is based. Without EAE and other animal studies, clinical investigations will continue to be "look-see" exercises, which will most likely provide more misses than hits and will fail the people with MS that they aim to serve.
Collapse
Affiliation(s)
- David Baker
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom.
| | - Sandra Amor
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Pathology Department, VU Medical Centre, Free University of Amsterdam, The Netherlands
| |
Collapse
|
30
|
Hampton DW, Serio A, Pryce G, Al-Izki S, Franklin RJM, Giovannoni G, Baker D, Chandran S. Neurodegeneration progresses despite complete elimination of clinical relapses in a mouse model of multiple sclerosis. Acta Neuropathol Commun 2013; 1:84. [PMID: 24364862 PMCID: PMC3895761 DOI: 10.1186/2051-5960-1-84] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 12/15/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND [corrected] Multiple Sclerosis has two clinical phases reflecting distinct but inter-related pathological processes: focal inflammation drives the relapse-remitting stage and neurodegeneration represents the principal substrate of secondary progression. In contrast to the increasing number of effective anti-inflammatory disease modifying treatments for relapse-remitting disease, the absence of therapies for progressive disease represents a major unmet clinical need. This raises the unanswered question of whether elimination of clinical relapses will prevent subsequent progression and if so how early in the disease course should treatment be initiated. Experimental autoimmune encephalomyelitis in the Biozzi ABH mouse recapitulates the clinical and pathological features of multiple sclerosis including relapse-remitting episodes with inflammatory mediated demyelination and progressive disability with neurodegeneration. To address the relationship between inflammation and neurodegeneration we used an auto-immune tolerance strategy to eliminate clinical relapses in EAE in a manner analogous to the clinical effect of disease modifying treatments. RESULTS By arresting clinical relapses in EAE at two distinct stages, early and late disease, we demonstrate that halting immune driven demyelination even after the first major clinical event is insufficient to prevent long-term neurodegeneration and associated gliosis. Nonetheless, early intervention is partially neuroprotective, whereas later interventions are not. Furthermore early tolerisation is also associated with increased remyelination. CONCLUSIONS These findings are consistent with both a partial uncoupling of inflammation and neurodegeneration and that the regenerative response of remyelination is negatively correlated with inflammation. These findings strongly support the need for early combinatorial treatment of immunomodulatory therapies and neuroprotective treatments to prevent long-term neurodegeneration in multiple sclerosis.
Collapse
Affiliation(s)
- David W Hampton
- Centre for Clinical Brain Sciences, MS Centre, University of Edinburgh Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Andrea Serio
- Centre for Clinical Brain Sciences, MS Centre, University of Edinburgh Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Gareth Pryce
- Neuroimmunology Unit, Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Sarah Al-Izki
- Neuroimmunology Unit, Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Robin JM Franklin
- MRC Cambridge Centre for Stem Cell Biology and Regenerative Medicine and Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Gavin Giovannoni
- Neuroimmunology Unit, Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - David Baker
- Neuroimmunology Unit, Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, MS Centre, University of Edinburgh Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
31
|
Al-Izki S, Pryce G, Hankey DJR, Lidster K, von Kutzleben SM, Browne L, Clutterbuck L, Posada C, Edith Chan AW, Amor S, Perkins V, Gerritsen WH, Ummenthum K, Peferoen-Baert R, van der Valk P, Montoya A, Joel SP, Garthwaite J, Giovannoni G, Selwood DL, Baker D. Lesional-targeting of neuroprotection to the inflammatory penumbra in experimental multiple sclerosis. ACTA ACUST UNITED AC 2013; 137:92-108. [PMID: 24287115 DOI: 10.1093/brain/awt324] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Progressive multiple sclerosis is associated with metabolic failure of the axon and excitotoxicity that leads to chronic neurodegeneration. Global sodium-channel blockade causes side effects that can limit its use for neuroprotection in multiple sclerosis. Through selective targeting of drugs to lesions we aimed to improve the potential therapeutic window for treatment. This was assessed in the relapsing-progressive experimental autoimmune encephalomyelitis ABH mouse model of multiple sclerosis using conventional sodium channel blockers and a novel central nervous system-excluded sodium channel blocker (CFM6104) that was synthesized with properties that selectively target the inflammatory penumbra in experimental autoimmune encephalomyelitis lesions. Carbamazepine and oxcarbazepine were not immunosuppressive in lymphocyte-driven autoimmunity, but slowed the accumulation of disability in experimental autoimmune encephalomyelitis when administered during periods of the inflammatory penumbra after active lesion formation, and was shown to limit the development of neurodegeneration during optic neuritis in myelin-specific T cell receptor transgenic mice. CFM6104 was shown to be a state-selective, sodium channel blocker and a fluorescent p-glycoprotein substrate that was traceable. This compound was >90% excluded from the central nervous system in normal mice, but entered the central nervous system during the inflammatory phase in experimental autoimmune encephalomyelitis mice. This occurs after the focal and selective downregulation of endothelial p-glycoprotein at the blood-brain barrier that occurs in both experimental autoimmune encephalomyelitis and multiple sclerosis lesions. CFM6104 significantly slowed down the accumulation of disability and nerve loss in experimental autoimmune encephalomyelitis. Therapeutic-targeting of drugs to lesions may reduce the potential side effect profile of neuroprotective agents that can influence neurotransmission. This class of agents inhibit microglial activity and neural sodium loading, which are both thought to contribute to progressive neurodegeneration in multiple sclerosis and possibly other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarah Al-Izki
- 1 Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Neuroprotection in a novel mouse model of multiple sclerosis. PLoS One 2013; 8:e79188. [PMID: 24223903 PMCID: PMC3817036 DOI: 10.1371/journal.pone.0079188] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 09/24/2013] [Indexed: 01/16/2023] Open
Abstract
Multiple sclerosis is an immune-mediated, demyelinating and neurodegenerative disease that currently lacks any neuroprotective treatments. Innovative neuroprotective trial designs are required to hasten the translational process of drug development. An ideal target to monitor the efficacy of strategies aimed at treating multiple sclerosis is the visual system, which is the most accessible part of the human central nervous system. A novel C57BL/6 mouse line was generated that expressed transgenes for a myelin oligodendrocyte glycoprotein-specific T cell receptor and a retinal ganglion cell restricted-Thy1 promoter-controlled cyan fluorescent protein. This model develops spontaneous or induced optic neuritis, in the absence of paralytic disease normally associated with most rodent autoimmune models of multiple sclerosis. Demyelination and neurodegeneration could be monitored longitudinally in the living animal using electrophysiology, visual sensitivity, confocal scanning laser ophthalmoscopy and optical coherence tomography all of which are relevant to human trials. This model offers many advantages, from a 3Rs, economic and scientific perspective, over classical experimental autoimmune encephalomyelitis models that are associated with substantial suffering of animals. Optic neuritis in this model led to inflammatory damage of axons in the optic nerve and subsequent loss of retinal ganglion cells in the retina. This was inhibited by the systemic administration of a sodium channel blocker (oxcarbazepine) or intraocular treatment with siRNA targeting caspase-2. These novel approaches have relevance to the future treatment of neurodegeneration of MS, which has so far evaded treatment.
Collapse
|
33
|
Acharjee S, Nayani N, Tsutsui M, Hill MN, Ousman SS, Pittman QJ. Altered cognitive-emotional behavior in early experimental autoimmune encephalitis--cytokine and hormonal correlates. Brain Behav Immun 2013; 33:164-72. [PMID: 23886782 DOI: 10.1016/j.bbi.2013.07.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 07/09/2013] [Accepted: 07/09/2013] [Indexed: 12/16/2022] Open
Abstract
Multiple sclerosis (MS) is often associated with co-morbid behavioural and cognitive impairments; however the presence of these symptoms does not necessarily correlate with neurological damage. This suggests that an alternate mechanism may subserve these impairments relative to motor deficits. We investigated whether these abnormalities could be studied in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. In myelin oligodendrocyte glycoprotein peptide (MOG35-55)-induced EAE mice, no motor deficits were observed until d9 after immunization. This enabled us to carry out a series of neurobehavioral tests during the presymptomatic stage, between d6 and d8 post-immunization. EAE mice spent more time in the outer zone in an open field test and in the closed arms of an elevated plus maze and, showed decreased latency for immobility in the tail suspension and forced swim tests and reduced social interaction compared with controls. These results are indicative of anxiety- and depression- like behavior. In addition, EAE mice appeared to exhibit memory impairment compared to controls based on their reduced time spent in the target quadrant in the Morris water maze and their faster memory extinction in the fear conditioning test. No demyelination, microglial activation or astrogliosis was observed in the brain at this early stage. Transcript analysis by RT-PCR from d6 to d8 brain revealed elevated interleukin (IL)-1β and TNF-α in the hypothalamus but not in the amygdala or hippocampus of EAE mice. Lastly, plasma corticosterone levels increased in EAE mice compared to controls. In conclusion, emotional and cognitive deficits are observed in EAE prior to demyelination and are associated with elevated IL-1β and TNF-α in the hypothalamus and changes in the hypothalamic-pituitary-adrenal axis.
Collapse
Affiliation(s)
- Shaona Acharjee
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| | | | | | | | | | | |
Collapse
|
34
|
Simmons SB, Pierson ER, Lee SY, Goverman JM. Modeling the heterogeneity of multiple sclerosis in animals. Trends Immunol 2013; 34:410-22. [PMID: 23707039 DOI: 10.1016/j.it.2013.04.006] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 04/09/2013] [Accepted: 04/18/2013] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating disease of the central nervous system (CNS) manifested with varying clinical course, pathology, and inflammatory patterns. There are multiple animal models that reflect different aspects of this heterogeneity. Collectively, these models reveal a balance between pathogenic and regulatory CD4(+) T cells, CD8(+) T cells, and B cells that influences the incidence, timing, and severity of CNS autoimmunity. In this review we discuss experimental autoimmune encephalomyelitis (EAE) models that have been used to study the pathogenic and regulatory roles of these immune cells; models that recapitulate different aspects of the disease seen in patients with MS, and questions remaining for future studies.
Collapse
Affiliation(s)
- Sarah B Simmons
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | | | | | | |
Collapse
|
35
|
Apolipoprotein E and its mimetic peptide suppress Th1 and Th17 responses in experimental autoimmune encephalomyelitis. Neurobiol Dis 2013; 56:59-65. [PMID: 23619428 DOI: 10.1016/j.nbd.2013.04.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 03/23/2013] [Accepted: 04/12/2013] [Indexed: 12/20/2022] Open
Abstract
Apolipoprotein E (apoE) has been detected to possess anti-inflammatory properties that can contribute to protection against experimental autoimmune encephalomyelitis (EAE). However, its impact on Th1 and Th17 responses in EAE is unclear. In this study, we induced EAE in apoE-/- mice and wild-type mice. We observed that the absence of apoE resulted in the increased proportion of Th1 and Th17 cells in the spleens and brains, as well as up-regulated expressions of proinflammatory cytokines (IL-17, IFN-γ, TNF-α, IL-12, IL-1β and IL-6) and transcription factors (RORγt and T-bet) in the CNS. ApoE-/- mice also showed the increased release of proinflammatory cytokines by macrophages in vitro. In addition, we used a mimetic peptide of apoE, which mimic the functions of apoE except for lipid transport. ApoE mimetic peptide could reverse the above negative effect in EAE. Thus, apoE can modulate Th1 and Th17 responses, likely through its inhibitory effect on the secretion of cytokines by macrophages. Our result also suggests that apoE mimetic peptide might be developed into a therapeutic agent for multiple sclerosis.
Collapse
|
36
|
de Lago E, Gómez-Ruiz M, Moreno-Martet M, Fernández-Ruiz J. Cannabinoids, multiple sclerosis and neuroprotection. Expert Rev Clin Pharmacol 2012; 2:645-60. [PMID: 22112258 DOI: 10.1586/ecp.09.42] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The cannabinoid signaling system participates in the control of cell homeostasis in the CNS, which explains why, in different neurodegenerative diseases including multiple sclerosis (MS), alterations in this system have been found to serve both as a pathogenic factor (malfunctioning of this system has been found at early phases of these diseases) and as a therapeutic target (the management of this system has beneficial effects). MS is an autoimmune disease that affects the CNS and it is characterized by inflammation, demyelination, remyelination, gliosis and axonal damage. Although it has been considered mainly as an inflammatory disorder, recent studies have recognized the importance of axonal loss both in the progression of the disorder and in the appearance of neurological disability, even in early stages of the disease. In recent years, several laboratories have addressed the therapeutic potential of cannabinoids in MS, given the experience reported by some MS patients who self-medicated with marijuana. Most of these studies focused on the alleviation of symptoms (spasticity, tremor, anxiety and pain) or on the inflammatory component of the disease. However, recent data also revealed the important neuroprotective action that could be exerted by cannabinoids in this disorder. The present review will be precisely centered on this neuroprotective potential, which is based mainly on antioxidant, anti-inflammatory and anti-excitotoxic properties, exerted through the activation of CB1 or CB2 receptors or other unknown mechanisms.
Collapse
Affiliation(s)
- Eva de Lago
- Departamento de Bioquímica y Biología Molecular and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain.
| | | | | | | |
Collapse
|
37
|
Hampton DW, Innes N, Merkler D, Zhao C, Franklin RJ, Chandran S. Focal Immune-Mediated White Matter Demyelination Reveals an Age-Associated Increase in Axonal Vulnerability and Decreased Remyelination Efficiency. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1897-905. [DOI: 10.1016/j.ajpath.2012.01.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 01/16/2012] [Accepted: 01/19/2012] [Indexed: 01/18/2023]
|
38
|
Constantinescu CS, Farooqi N, O'Brien K, Gran B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 2012; 164:1079-106. [PMID: 21371012 DOI: 10.1111/j.1476-5381.2011.01302.x] [Citation(s) in RCA: 1091] [Impact Index Per Article: 83.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental model for the human inflammatory demyelinating disease, multiple sclerosis (MS). EAE is a complex condition in which the interaction between a variety of immunopathological and neuropathological mechanisms leads to an approximation of the key pathological features of MS: inflammation, demyelination, axonal loss and gliosis. The counter-regulatory mechanisms of resolution of inflammation and remyelination also occur in EAE, which, therefore can also serve as a model for these processes. Moreover, EAE is often used as a model of cell-mediated organ-specific autoimmune conditions in general. EAE has a complex neuropharmacology, and many of the drugs that are in current or imminent use in MS have been developed, tested or validated on the basis of EAE studies. There is great heterogeneity in the susceptibility to the induction, the method of induction and the response to various immunological or neuropharmacological interventions, many of which are reviewed here. This makes EAE a very versatile system to use in translational neuro- and immunopharmacology, but the model needs to be tailored to the scientific question being asked. While creating difficulties and underscoring the inherent weaknesses of this model of MS in straightforward translation from EAE to the human disease, this variability also creates an opportunity to explore multiple facets of the immune and neural mechanisms of immune-mediated neuroinflammation and demyelination as well as intrinsic protective mechanisms. This allows the eventual development and preclinical testing of a wide range of potential therapeutic interventions.
Collapse
Affiliation(s)
- Cris S Constantinescu
- Division of Clinical Neurology, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK.
| | | | | | | |
Collapse
|
39
|
Gnanapavan S, Grant D, Pryce G, Jackson S, Baker D, Giovannoni G. Neurofilament a biomarker of neurodegeneration in autoimmune encephalomyelitis. Autoimmunity 2012; 45:298-303. [DOI: 10.3109/08916934.2012.654865] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
40
|
The biology that underpins the therapeutic potential of cannabis-based medicines for the control of spasticity in multiple sclerosis. Mult Scler Relat Disord 2012; 1:64-75. [PMID: 25876933 DOI: 10.1016/j.msard.2011.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 11/17/2011] [Indexed: 12/14/2022]
Abstract
Cannabis-based medicines have recently been approved for the treatment of pain and spasticity in multiple sclerosis (MS). This supports the original perceptions of people with MS, who were using illegal street cannabis for symptom control and pre-clinical testing in animal models of MS. This activity is supported both by the biology of the disease and the biology of the cannabis plant and the endocannabinoid system. MS results from disease that impairs neurotransmission and this is controlled by cannabinoid receptors and endogenous cannabinoid ligands. This can limit spasticity and may also influence the processes that drive the accumulation of progressive disability.
Collapse
|
41
|
Practical guide to the induction of relapsing progressive experimental autoimmune encephalomyelitis in the Biozzi ABH mouse. Mult Scler Relat Disord 2012; 1:29-38. [DOI: 10.1016/j.msard.2011.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 08/12/2011] [Accepted: 09/02/2011] [Indexed: 01/04/2023]
|
42
|
Kipp M, van der Star B, Vogel DYS, Puentes F, van der Valk P, Baker D, Amor S. Experimental in vivo and in vitro models of multiple sclerosis: EAE and beyond. Mult Scler Relat Disord 2011; 1:15-28. [PMID: 25876447 DOI: 10.1016/j.msard.2011.09.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 09/05/2011] [Indexed: 12/16/2022]
Abstract
Although the primary cause of multiple sclerosis (MS) is unknown, the widely accepted view is that aberrant (auto)immune responses possibly arising following infection(s) are responsible for the destructive inflammatory demyelination and neurodegeneration in the central nervous system (CNS). This notion, and the limited access of human brain tissue early in the course of MS, has led to the development of autoimmune, viral and toxin-induced demyelination animal models as well as the development of human CNS cell and organotypic brain slice cultures in an attempt to understand events in MS. The autoimmune models, collectively known as experimental autoimmune encephalomyelitis (EAE), and viral models have shaped ideas of how environmental factors may trigger inflammation, demyelination and neurodegeneration in the CNS. Understandably, these models have also heavily influenced the development of therapies targeting the inflammatory aspect of MS. Demyelination and remyelination in the absence of overt inflammation are better studied in toxin-induced demyelination models using cuprizone and lysolecithin. The paradigm shift of MS as an autoimmune disease of myelin to a neurodegenerative disease has required more appropriate models reflecting the axonal and neuronal damage. Thus, secondary progressive EAE and spastic models have been crucial to develop neuroprotective approaches. In this review the current in vivo and in vitro experimental models to examine pathological mechanisms involved in inflammation, demyelination and neuronal degeneration, as well as remyelination and repair in MS are discussed. Since this knowledge is the basis for the development of new therapeutic approaches for MS, we particularly address whether the currently available models truly reflect the human disease, and discuss perspectives to further optimise and develop more suitable experimental models to study MS.
Collapse
Affiliation(s)
- Markus Kipp
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Baukje van der Star
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Daphne Y S Vogel
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Fabìola Puentes
- Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
| | - Paul van der Valk
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - David Baker
- Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
| | - Sandra Amor
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK.
| |
Collapse
|
43
|
Al-Izki S, Pryce G, Jackson SJ, Giovannoni G, Baker D. Immunosuppression with FTY720 is insufficient to prevent secondary progressive neurodegeneration in experimental autoimmune encephalomyelitis. Mult Scler 2011; 17:939-48. [PMID: 21459808 DOI: 10.1177/1352458511400476] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND There has been poor translation for the use of immunosuppressive agents from experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), into the treatment of MS. This may be due to the fact that most EAE studies examine prophylactic, pre-treatment regimes that prove to be therapeutically-ineffective in long-established, often progressive, MS. FTY720 (fingolimod/Gilenya) is a sphingosine-1-phosphate receptor modulator. This is a new oral agent that markedly reduces the number of relapses in people with MS, compared with currently licensed injectable agents such as the beta interferons. FTY720 has activity against lymphocytes but may also influence oligodendroglia and could therefore have the potential to influence progressive MS, by promoting remyelination. METHODS The effect of FTY720 was assessed in relapsing-progressive EAE in mice. RESULTS Early intervention during relapsing EAE could completely inhibit subsequent relapses, inhibited the accumulation of neurodegeneration, and facilitated motor recovery. However, when examined in secondary progressive EAE, that develops after the accumulation of deficit from relapsing disease, long-term treatment with FTY720 failed to slow deterioration when initiated late (4 months) into the disease course. CONCLUSIONS This study indicates that early intervention with immunosuppressive agents may inhibit the generation of the neurodegenerative microenvironment, which is no longer responsive to potent immunosuppression. However, if treatment is initiated too late, progressive, neurological-disease continues unabated. This suggests that immunosuppression is insufficient to control secondary progression in animals, as has been found so far to be the case in MS, and may warrant early intervention with FTY720 for optimal treatment benefit.
Collapse
Affiliation(s)
- Sarah Al-Izki
- Centre of Neuroscience and Trauma, Blizard Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK.
| | | | | | | | | |
Collapse
|
44
|
Aharoni R, Vainshtein A, Stock A, Eilam R, From R, Shinder V, Arnon R. Distinct pathological patterns in relapsing-remitting and chronic models of experimental autoimmune enchephalomyelitis and the neuroprotective effect of glatiramer acetate. J Autoimmun 2011; 37:228-41. [PMID: 21752599 DOI: 10.1016/j.jaut.2011.06.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 06/09/2011] [Accepted: 06/14/2011] [Indexed: 10/17/2022]
Abstract
The respective roles of inflammatory and neurodegenerative processes in the pathology of multiple sclerosis (MS) and in its animal model experimental autoimmune encephalomyelitis (EAE) are controversial. Novel treatment strategies aim to operate within the CNS to induce neuroprotection and repair processes in addition to their anti-inflammatory properties. In this study we analyzed and compared the in situ pathological manifestations of EAE utilizing two different models, namely the relapsing-remitting PLP-induced and the chronic MOG-induced diseases. To characterize pathological changes, both transmission electron microscopy (TEM) and immunohistochemistry were employed. The effect of the approved MS drug glatiramer acetate (GA, Copaxone) on myelin damage/repair and on motor neuron loss/preservation was studied in both EAE models. Ultrastructural spinal cord analysis revealed multiple white matter damage foci, with different patterns in the two EAE models. Thus, the relapsing-remitting model was characterized mainly by widespread myelin damage and by remyelinating fibers, whereas in the chronic model axonal degeneration was more prevalent. Loss of lower motor neurons was manifested only in mice with chronic MOG-induced disease. In the GA-treated mice, smaller lesions, increased axonal density and higher prevalence of normal appearing axons were observed, as well as decreased demyelination and degeneration. Furthermore, quantitative analysis of the relative remyelination versus demyelination, provides for the first time evidence of significant augmentation of remyelination after GA treatment. The loss of motor neurons in GA-treated mice was also reduced in comparison to that of EAE untreated mice. These effects were obtained even when GA treatment was applied in a therapeutic schedule, namely after the appearance of clinical symptoms. Hence, the remyelination and neuronal preservation induced by GA are in support of the neuroprotective consequences of this treatment.
Collapse
Affiliation(s)
- Rina Aharoni
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a spectrum of neurological disorders in laboratory animals that is used to model multiple sclerosis (MS). However, few agents have translated from efficacy in EAE to the treatment of human disease. Although this may reflect species differences in pathological disease mechanisms, importantly it may also relate to the practice of how drugs and models are currently used. This often bears very little resemblance to the clinical scenarios where treatments are investigated, such that lack of appreciation of the biology of disease may doom drugs to failure. The use of EAE is critically appraised with the aim of provoking thought, improving laboratory practise and aiding researchers and reviewers to address quality issues when undertaking, reporting and interpreting animal studies related to MS research. This is important as many researchers using EAE could and should do more to improve the quality of the studies.
Collapse
Affiliation(s)
- David Baker
- Neuroscience and Trauma Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK.
| | | | | | | |
Collapse
|
46
|
|
47
|
Hampton DW, Webber DJ, Bilican B, Goedert M, Spillantini MG, Chandran S. Cell-mediated neuroprotection in a mouse model of human tauopathy. J Neurosci 2010; 30:9973-83. [PMID: 20668182 PMCID: PMC6633376 DOI: 10.1523/jneurosci.0834-10.2010] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 05/20/2010] [Accepted: 05/28/2010] [Indexed: 01/19/2023] Open
Abstract
Tau protein in a hyperphosphorylated state makes up the intracellular inclusions of several neurodegenerative diseases, including Alzheimer's disease and cases of frontotemporal dementia. Mutations in Tau cause familial forms of frontotemporal dementia, establishing that dysfunction of tau protein is sufficient to cause neurodegeneration and dementia. Transgenic mice expressing human mutant tau in neurons exhibit the essential features of tauopathies, including neurodegeneration and abundant filaments composed of hyperphosphorylated tau. Here we show that a previously described mouse line transgenic for human P301S tau exhibits an age-related, layer-specific loss of superficial cortical neurons, similar to what has been observed in human frontotemporal dementias. We also show that focal neural precursor cell implantation, resulting in glial cell differentiation, leads to the sustained rescue of cortical neurons. Together with evidence indicating that astrocyte transplantation may be neuroprotective, our findings suggest a beneficial role for glial cell-based repair in neurodegenerative diseases.
Collapse
Affiliation(s)
- David W. Hampton
- Euan MacDonald Centre for Motor Neurone Disease Research, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
- Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 2PY, United Kingdom, and
| | - Daniel J. Webber
- Euan MacDonald Centre for Motor Neurone Disease Research, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
- Anne McLaren Laboratory for Regenerative Medicine, Medical Research Council Centre for Stem Cell Biology and Regenerative Medicine, University of Cambridge, Cambridge CB2 0SZ, United Kingdom
- Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 2PY, United Kingdom, and
| | - Bilada Bilican
- Euan MacDonald Centre for Motor Neurone Disease Research, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
- Anne McLaren Laboratory for Regenerative Medicine, Medical Research Council Centre for Stem Cell Biology and Regenerative Medicine, University of Cambridge, Cambridge CB2 0SZ, United Kingdom
- Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 2PY, United Kingdom, and
| | - Michel Goedert
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Maria Grazia Spillantini
- Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 2PY, United Kingdom, and
| | - Siddharthan Chandran
- Euan MacDonald Centre for Motor Neurone Disease Research, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
- Anne McLaren Laboratory for Regenerative Medicine, Medical Research Council Centre for Stem Cell Biology and Regenerative Medicine, University of Cambridge, Cambridge CB2 0SZ, United Kingdom
- Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 2PY, United Kingdom, and
| |
Collapse
|
48
|
Amor S, Puentes F, Baker D, van der Valk P. Inflammation in neurodegenerative diseases. Immunology 2010; 129:154-69. [PMID: 20561356 PMCID: PMC2814458 DOI: 10.1111/j.1365-2567.2009.03225.x] [Citation(s) in RCA: 1003] [Impact Index Per Article: 66.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Revised: 11/25/2009] [Accepted: 11/25/2009] [Indexed: 01/01/2023] Open
Abstract
Neurodegeneration, the slow and progressive dysfunction and loss of neurons and axons in the central nervous system, is the primary pathological feature of acute and chronic neurodegenerative conditions such as Alzheimer's disease and Parkinson's disease, neurotropic viral infections, stroke, paraneoplastic disorders, traumatic brain injury and multiple sclerosis. Despite different triggering events, a common feature is chronic immune activation, in particular of microglia, the resident macrophages of the central nervous system. Apart from the pathogenic role of immune responses, emerging evidence indicates that immune responses are also critical for neuroregeneration. Here, we review the impact of innate and adaptive immune responses on the central nervous system in autoimmune, viral and other neurodegenerative disorders, and discuss their contribution to either damage or repair. We also discuss potential therapies aimed at the immune responses within the central nervous system. A better understanding of the interaction between the immune and nervous systems will be crucial to either target pathogenic responses, or augment the beneficial effects of immune responses as a strategy to intervene in chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Sandra Amor
- Department of Pathology, VU University Medical Centre De Boelelaan, Amsterdam, the Netherlands.
| | | | | | | |
Collapse
|
49
|
Hosking MP, Lane TE. The Biology of Persistent Infection: Inflammation and Demyelination following Murine Coronavirus Infection of the Central Nervous System. ACTA ACUST UNITED AC 2009; 5:267-276. [PMID: 19946572 DOI: 10.2174/157339509789504005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Multiple Sclerosis (MS) is an immune-mediated demyelinating disease of humans. Although causes of MS are enigmatic, underlying elements contributing to disease development include both genetic and environmental factors. Recent epidemiological evidence has pointed to viral infection as a trigger to initiating white matter damage in humans. Mouse hepatitis virus (MHV) is a positive strand RNA virus that, following intracranial infection of susceptible mice, induces an acute encephalomyelitis that later resolves into a chronic fulminating demyelinating disease. Immune cell infiltration into the central nervous system is critical both to quell viral replication and instigate demyelination. Recent efforts by our laboratory and others have focused upon strategies capable of enhancing remyelination in response to viral-induced demyelination, both by dampening chronic inflammation and by surgical engraftment of remyelination - competent neural precursor cells.
Collapse
Affiliation(s)
- Martin P Hosking
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900 USA
| | | |
Collapse
|
50
|
Lassmann H. Axonal and neuronal pathology in multiple sclerosis: what have we learnt from animal models. Exp Neurol 2009; 225:2-8. [PMID: 19840788 DOI: 10.1016/j.expneurol.2009.10.009] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 10/08/2009] [Accepted: 10/10/2009] [Indexed: 12/21/2022]
Abstract
Axonal and neuronal injury and loss are of critical importance for permanent clinical disability in multiple sclerosis patients. Axonal injury occurs already early during the disease and accumulates with disease progression. It is not restricted to focal demyelinated lesions in the white matter, but also affects the normal appearing white matter and the grey matter. Experimental studies show that many different immunological mechanisms may lead to axonal and neuronal injury, including antigen-specific destruction by specific T-cells and auto-antibodies as well as injury induced by products of activated macrophages and microglia. They all appear to be relevant for multiple sclerosis pathogensis in different patients and at different stages of the disease. However, in MS lesions a major mechanism of axonal and neuronal damage appears to be related to the action of reactive oxygen and nitrogen species, which may induce neuronal injury through impairment of mitochondrial function and subsequent energy failure.
Collapse
Affiliation(s)
- Hans Lassmann
- Centre for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Wien, Austria.
| |
Collapse
|