1
|
Ribeiro AR, Pereira R, Barros C, Barateiro A, Alberro A, Basto AP, Graça L, Pinto MV, Santos FMF, Gois PMP, Howlett SE, Fernandes A. Experimental autoimmune encephalomyelitis pathogenesis alters along animal age: impact of S100B expression. J Neuroimmune Pharmacol 2025; 20:37. [PMID: 40227512 PMCID: PMC11997003 DOI: 10.1007/s11481-025-10195-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
Multiple Sclerosis (MS) is the leading inflammatory and non-traumatic cause of disability in young adults, with late-onset MS emerging in middle-aged patients often resulting in poorer treatment responses and worse prognoses. The calcium-binding protein S100B is elevated in MS patients, and its targeting has shown promise in reducing disease severity in experimental autoimmune encephalomyelitis (EAE) models. However, most studies on MS pathology have focused on young animal models, leaving a gap in understanding the effects of age and S100B ablation on disease progression throughout the lifespan. This study aimed to characterize EAE in mice of different ages, examining demyelination, inflammation, and immune responses to determine whether S100B ablation could mitigate MS pathogenesis across the lifespan. EAE was induced in six cohorts of C57BL/6 mice: young adults (3 months), older adults (6 months), and middle-aged (12 months), including corresponding S100B knockout (KO) groups, followed for 23 days. Upon sacrifice, spinal cords were assessed via immunohistochemistry and Real-Time qPCR, while splenocytes were analyzed for immune cell characterization. Results indicated a more severe disease course in 12-month-old mice, marked by increased gliosis, inflammation, and impaired microglial phagocytic activity. Notably, S100B absence reduced gliosis and inflammatory markers across all ages, with 12-month-old S100B KO mice showing increased regulatory T cells. These findings highlight the exacerbating role of age and elevated S100B in MS progression, underscoring the importance of identifying age-specific MS markers and therapeutic targets.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- S100 Calcium Binding Protein beta Subunit/genetics
- S100 Calcium Binding Protein beta Subunit/biosynthesis
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Female
- Aging/metabolism
- Aging/pathology
- Age Factors
- Spinal Cord/pathology
- Spinal Cord/metabolism
- Male
Collapse
Affiliation(s)
- Ana Rita Ribeiro
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Raquel Pereira
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Catarina Barros
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Barateiro
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
- Departamento de Ciências Farmacêuticas E Do Medicamento, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Ainhoa Alberro
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
- IIS Biogipuzkoa Health Research Institute, San Sebastian, Spain
| | - Afonso P Basto
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
- Laboratório Associado Para a Ciência Animal E Veterinária (AL4AnimalS), Lisbon, Portugal
| | - Luís Graça
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Vaz Pinto
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Fábio M F Santos
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Pedro M P Gois
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
- Departamento de Ciências Farmacêuticas E Do Medicamento, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
- Department of Medicine (Geriatric Medicine), Dalhousie University, Halifax, NS, Canada
| | - Adelaide Fernandes
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal.
- Departamento de Ciências Farmacêuticas E Do Medicamento, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
2
|
Manavi Z, Melchor GS, Bullard MR, Gross PS, Ray S, Gaur P, Baydyuk M, Huang JK. Senescent cell reduction does not improve recovery in mice under experimental autoimmune encephalomyelitis (EAE) induced demyelination. J Neuroinflammation 2025; 22:101. [PMID: 40197319 PMCID: PMC11974124 DOI: 10.1186/s12974-025-03425-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by immune cell-driven demyelination and progressive neurodegeneration. Senescent cells (SCs) have recently been observed in chronic MS lesions indicating their possible involvement in disease progression. However, the role of SCs and the potential therapeutic benefit of their reduction through senolytic therapy remains to be determined in experimental autoimmune encephalomyelitis (EAE), a widely used preclinical model of MS. Here, we show that senescent-like myeloid cells accumulate in the spinal cord parenchyma and meninges in mice after myelin oligodendrocyte glycoprotein (MOG33-55) EAE induction. Treatment with the senolytic cocktail, Dasatinib and Quercetin (DQ), effectively reduces the senescent-like myeloid cells, but this does not translate into improved clinical outcomes in EAE mice. Increasing DQ dosage or using INK-ATTAC transgenic mice also failed to ameliorate EAE severity. Additionally, histopathological analysis shows no significant differences in demyelination or axonal degeneration between treated and control groups. Our findings indicate that senescent-like myeloid cells are present in an immune-mediated demyelinating model of MS and can be reduced through senolytic therapy with Dasatinib and Quercetin. However, their reduction through DQ does not significantly impact inflammation or recovery, suggesting that the therapeutic potential of senolytics as disease-modifying drugs in MS may be limited.
Collapse
Affiliation(s)
- Zeeba Manavi
- Department of Biology, Georgetown University, Washington, DC, USA
| | - George S Melchor
- Department of Biology, Georgetown University, Washington, DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
| | - Meghan R Bullard
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Phillip S Gross
- Department of Biology, Georgetown University, Washington, DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
| | - Shinjini Ray
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Pankaj Gaur
- Department of Oncology, Georgetown University, Washington, DC, USA
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, DC, USA.
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA.
| |
Collapse
|
3
|
Drake SS, Mohammadnia A, Zaman A, Gianfelice C, Heale K, Groh AMR, Hua EML, Hintermayer MA, Lu YR, Gosselin D, Zandee S, Prat A, Stratton JA, Sinclair DA, Fournier AE. Cellular rejuvenation protects neurons from inflammation-mediated cell death. Cell Rep 2025; 44:115298. [PMID: 39937646 DOI: 10.1016/j.celrep.2025.115298] [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: 11/14/2023] [Revised: 10/31/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
In multiple sclerosis (MS), inflammation of the central nervous system results in demyelination, neuroaxonal injury, and cell death. However, the molecular signals responsible for injury and cell death in neurons are not fully characterized. Here, we profile the transcriptome of retinal ganglion cells (RGCs) in experimental autoimmune encephalomyelitis (EAE) mice. Pathway analysis identifies a transcriptional signature reminiscent of aged RGCs with some senescent features, with a comparable signature present in neurons from patients with MS. This is supported by immunostaining demonstrating alterations to the nuclear envelope, modifications in chromatin marks, and accumulation of DNA damage. Transduction of RGCs with an Oct4-Sox2-Klf4 adeno-associated virus (AAV) to rejuvenate the transcriptome enhances RGC survival in EAE and improves visual acuity. Collectively, these data reveal an aging-like phenotype in neurons under pathological neuroinflammation and support the possibility that rejuvenation therapies or senotherapeutic agents could offer a direct avenue for neuroprotection in neuroimmune disorders.
Collapse
Affiliation(s)
- Sienna S Drake
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada
| | - Abdulshakour Mohammadnia
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada
| | - Aliyah Zaman
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada
| | - Christine Gianfelice
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada
| | - Kali Heale
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada
| | - Adam M R Groh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada
| | - Elizabeth M-L Hua
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada
| | - Matthew A Hintermayer
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada
| | - Yuancheng Ryan Lu
- Department of Genetics, Bavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - David Gosselin
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1V4G2, Canada
| | - Stephanie Zandee
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC H2X0A9, Canada
| | - Alexandre Prat
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC H2X0A9, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada
| | - David A Sinclair
- Department of Genetics, Bavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alyson E Fournier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A2B4, Canada.
| |
Collapse
|
4
|
Rad LM, Hughes KR, Wheeler SN, Decker JT, Orbach SM, Galvan A, Thornhill J, Griffin KV, Turkistani H, Urie RR, Irani DN, Shea LD, Morris AH. Engineered immunological niche directs therapeutic development in models of progressive multiple sclerosis. Proc Natl Acad Sci U S A 2025; 122:e2409852122. [PMID: 39937858 PMCID: PMC11848328 DOI: 10.1073/pnas.2409852122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/24/2024] [Indexed: 02/14/2025] Open
Abstract
Primary progressive multiple sclerosis (MS) is a demyelinating autoimmune disease with only a single class of FDA-approved treatment, B cell depletion. Novel treatments could emerge from a deeper understanding of the interplay between multiple cell types within diseased tissue throughout progression. We initially describe an engineered biomaterial-based immunological niche (IN) as a surrogate for diseased tissue to investigate immune cell function and phenotype dynamics throughout a chronic progressive mouse model of MS. Using these niches, we identify an array of dysregulated CC chemokine signaling as potential targets. We then develop antigen-loaded nanoparticles that reduce CC chemokine signaling, while delivering antigen. These nanoparticles serve as an antigen-specific treatment, and a single injection reduces disease burden, even if administered after symptomatic disease onset. This report demonstrates proof of principle of a biomaterial scaffold as a diseased tissue surrogate that can monitor immune function, identify potential drug targets, and guide the development of a therapeutic.
Collapse
Affiliation(s)
- Laila M. Rad
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Kevin R. Hughes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Sydney N. Wheeler
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Joseph T. Decker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Sophia M. Orbach
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Angelica Galvan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Jasmine Thornhill
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Kate V. Griffin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Hamza Turkistani
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Russell R. Urie
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - David N. Irani
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Aaron H. Morris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
5
|
Dema M, Eixarch H, Hervera A, Castillo M, Villar LM, Montalban X, Espejo C. Disease Aggravation With Age in an Experimental Model of Multiple Sclerosis: Role of Immunosenescence. Aging Cell 2025:e14491. [PMID: 39894902 DOI: 10.1111/acel.14491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
The onset of multiple sclerosis (MS) in older individuals correlates with a higher risk of developing primary progressive MS, faster progression to secondary progressive MS, and increased disability accumulation. This phenomenon can be related to age-related changes in the immune system: with age, the immune system undergoes a process called immunosenescence, characterized by a decline in the function of both the innate and adaptive immune responses. This decline can lead to a decreased ability to control inflammation and repair damaged tissue. Additionally, older individuals often experience a shift toward a more pro-inflammatory state, known as inflammaging, which can exacerbate the progression of neurodegenerative diseases like MS. Therefore, age-related alterations in the immune system could be responsible for the difference in the phenotype of MS observed in older and younger patients. In this study, we investigated the effects of age on the immunopathogenesis of experimental autoimmune encephalomyelitis (EAE). Our findings indicate that EAE is more severe in aged mice due to a more inflammatory and neurodegenerative environment in the central nervous system. Age-related changes predominantly affect adaptive immunity, characterized by altered T cell ratios, a pro-inflammatory Th1 response, increased regulatory T cells, exhaustion of T cells, altered B cell antigen presentation, and reduced NK cell maturation and cytotoxicity. Transcriptomic analysis reveals that fewer pathways and transcription factors are activated with age in EAE. These findings allow us to identify potential therapeutic targets specific to elderly MS patients and work on their development in the future.
Collapse
Affiliation(s)
- María Dema
- Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Herena Eixarch
- Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Arnau Hervera
- Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Mireia Castillo
- Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Luisa M Villar
- Departmento de Inmunología, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Xavier Montalban
- Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Carmen Espejo
- Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
6
|
Maupin EA, Adams KL. Cellular Senescence in Glial Cells: Implications for Multiple Sclerosis. J Neurochem 2025; 169:e16301. [PMID: 39831743 PMCID: PMC11745082 DOI: 10.1111/jnc.16301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Aging is the most common risk factor for Multiple Sclerosis (MS) disease progression. Cellular senescence, the irreversible state of cell cycle arrest, is the main driver of aging and has been found to accumulate prematurely in neurodegenerative diseases, including Alzheimer's and Parkinson's disease. Cellular senescence in the central nervous system of MS patients has recently gained attention, with several studies providing evidence that demyelination induces cellular senescence, with common hallmarks of p16INK4A and p21 expression, oxidative stress, and senescence-associated secreted factors. Here we discuss the current evidence of cellular senescence in animal models of MS and different glial populations in the central nervous system, highlighting the major gaps in the field that still remain. As premature senescence in MS may exacerbate demyelination and inflammation, resulting in inhibition of myelin repair, it is critical to increase understanding of cellular senescence in vivo, the functional effects of senescence on glial cells, and the impact of removing senescent cells on remyelination and MS. This emerging field holds promise for opening new avenues of treatment for MS patients.
Collapse
Affiliation(s)
- Elizabeth A. Maupin
- Department of Biological SciencesUniversity of Notre DameNotre DameIndianaUSA
| | - Katrina L. Adams
- Department of Biological SciencesUniversity of Notre DameNotre DameIndianaUSA
- The Center for Stem Cells and Regenerative MedicineUniversity of Notre DameNotre DameIndianaUSA
| |
Collapse
|
7
|
Kalia LV, Asis A, Arbour N, Bar-Or A, Bove R, Di Luca DG, Fon EA, Fox S, Gan-Or Z, Gommerman JL, Kang UJ, Klawiter EC, Koch M, Kolind S, Lang AE, Lee KK, Lincoln MR, MacDonald PA, McKeown MJ, Mestre TA, Miron VE, Ontaneda D, Rousseaux MWC, Schlossmacher MG, Schneider R, Stoessl AJ, Oh J. Disease-modifying therapies for Parkinson disease: lessons from multiple sclerosis. Nat Rev Neurol 2024; 20:724-737. [PMID: 39375563 DOI: 10.1038/s41582-024-01023-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/09/2024]
Abstract
The development of disease-modifying therapies (DMTs) for neurological disorders is an important goal in modern neurology, and the associated challenges are similar in many chronic neurological conditions. Major advances have been made in the multiple sclerosis (MS) field, with a range of DMTs being approved for relapsing MS and the introduction of the first DMTs for progressive MS. By contrast, people with Parkinson disease (PD) still lack such treatment options, relying instead on decades-old therapeutic approaches that provide only symptomatic relief. To address this unmet need, an in-person symposium was held in Toronto, Canada, in November 2022 for international researchers and experts in MS and PD to discuss strategies for advancing DMT development. In this Roadmap article, we highlight discussions from the symposium, which focused on therapeutic targets and preclinical models, disease spectra and subclassifications, and clinical trial design and outcome measures. From these discussions, we propose areas for novel or deeper exploration in PD using lessons learned from therapeutic development in MS. In addition, we identify challenges common to the PD and MS fields that need to be addressed to further advance the discovery and development of effective DMTs.
Collapse
Affiliation(s)
- Lorraine V Kalia
- Edmond J Safra Program in Parkinson's Disease, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada.
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | | | - Nathalie Arbour
- Department of Neurosciences, Université de Montreal, Montreal, Quebec, Canada
- Centre de Recherche du CHUM (CRCHUM), Montreal, Quebec, Canada
| | - Amit Bar-Or
- Division of MS and Related Disorders, Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
- Centre for Neuroinflammation and Experimental Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Riley Bove
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Daniel G Di Luca
- Edmond J Safra Program in Parkinson's Disease, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Edward A Fon
- The Neuro (Montreal Neurological Institute-Hospital), Montreal, Quebec, Canada
- Department of Neurology & Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Susan Fox
- Edmond J Safra Program in Parkinson's Disease, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ziv Gan-Or
- The Neuro (Montreal Neurological Institute-Hospital), Montreal, Quebec, Canada
- Department of Neurology & Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Jennifer L Gommerman
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Un Jung Kang
- Department of Neurology, Grossman School of Medicine, NYU Langone Health, New York, NY, USA
- Parekh Center for Interdisciplinary Neurology, Grossman School of Medicine, NYU Langone Health, New York, NY, USA
- Fresco Institute for Parkinson's and Movement Disorders, Grossman School of Medicine, NYU Langone Health, New York, NY, USA
- Department of Neuroscience and Physiology, Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Eric C Klawiter
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marcus Koch
- University of Calgary MS Clinic, Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Shannon Kolind
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anthony E Lang
- Edmond J Safra Program in Parkinson's Disease, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Matthew R Lincoln
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Barlo MS Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Penny A MacDonald
- Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Martin J McKeown
- Pacific Parkinson's Research Centre, Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tiago A Mestre
- Parkinson's Disease and Movement Disorders Clinic, Division of Neurology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada
| | - Veronique E Miron
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- The United Kingdom Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | - Maxime W C Rousseaux
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael G Schlossmacher
- Parkinson's Disease and Movement Disorders Clinic, Division of Neurology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada
| | - Raphael Schneider
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Barlo MS Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - A Jon Stoessl
- Pacific Parkinson's Research Centre, Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jiwon Oh
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Barlo MS Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Drake SS, Zaman A, Gianfelice C, Hua EML, Heale K, Afanasiev E, Klement W, Stratton JA, Prat A, Zandee S, Fournier AE. Senolytic treatment diminishes microglia and decreases severity of experimental autoimmune encephalomyelitis. J Neuroinflammation 2024; 21:283. [PMID: 39487537 PMCID: PMC11529445 DOI: 10.1186/s12974-024-03278-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND The role of senescence in disease contexts is complex, however there is considerable evidence that depletion of senescent cells improves outcomes in a variety of contexts particularly related to aging, cognition, and neurodegeneration. Much research has shown previously that inflammation can promote cellular senescence. Microglia are a central nervous system innate immune cell that undergo senescence with aging and during neurodegeneration. The contribution of senescent microglia to multiple sclerosis, an inflammatory neurodegenerative disease, is not clear, but microglia are strongly implicated in chronic active lesion pathology, tissue injury, and disease progression. Drugs that could specifically eliminate dysregulated microglia in multiple sclerosis are therefore of great interest to the field. RESULTS A single-cell analysis of brain tissue from mice subjected to experimental autoimmune encephalomyelitis (EAE), a mouse model of CNS inflammation that models aspects of multiple sclerosis (MS), identified microglia with a strong transcriptional signature of senescence including the presence of BCL2-family gene transcripts. Microglia expressing Bcl2l1 had higher expression of pro-inflammatory and senescence associated genes than their Bcl2l1 negative counterparts in EAE, suggesting they may exacerbate inflammation. Notably, in human single-nucleus sequencing from MS, BCL2L1 positive microglia were enriched in lesions with active inflammatory pathology, and likewise demonstrated increased expression of immune genes suggesting they may be proinflammatory and contribute to disease processes in chronic active lesions. Employing a small molecule BCL2-family inhibitor, Navitoclax (ABT-263), significantly reduced the presence of microglia and macrophages in the EAE spinal cord, suggesting that these cells can be targeted by senolytic treatment. ABT-263 treatment had a profound effect on EAE mice: decreasing motor symptom severity, improving visual acuity, promoting neuronal survival, and decreasing white matter inflammation. CONCLUSION These results support the hypothesis that microglia and macrophages exhibit transcriptional features of cellular senescence in EAE and MS, and that microglia expressing Bcl2l1 demonstrate a proinflammatory signature that may exacerbate inflammation resulting in negative outcomes in neuroinflammatory disease. Depleting microglia and macrophages using a senolytic results in robust improvement in EAE disease severity, including across measures of neurodegeneration, inflammation, and demyelination, and may therefore represent a novel strategy to address disease progression in multiple sclerosis.
Collapse
Affiliation(s)
- Sienna S Drake
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Aliyah Zaman
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | | | - Elizabeth M-L Hua
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Kali Heale
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Elia Afanasiev
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Wendy Klement
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l', Université de Montréal (CRCHUM), Montreal, Québec, H2X 0A9, Canada
| | - Jo Anne Stratton
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l', Université de Montréal (CRCHUM), Montreal, Québec, H2X 0A9, Canada
| | - Stephanie Zandee
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Alyson E Fournier
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
9
|
Fettig NM, Pu A, Osborne LC, Gommerman JL. The influence of aging and the microbiome in multiple sclerosis and other neurologic diseases. Immunol Rev 2024; 325:166-189. [PMID: 38890777 DOI: 10.1111/imr.13361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
The human gut microbiome is well-recognized as a key player in maintaining health. However, it is a dynamic entity that changes across the lifespan. How the microbial changes that occur in later decades of life shape host health or impact age-associated inflammatory neurological diseases such as multiple sclerosis (MS) is still unclear. Current understanding of the aging gut microbiome is largely limited to cross-sectional observational studies. Moreover, studies in humans are limited by confounding host-intrinsic and extrinsic factors that are not easily disentangled from aging. This review provides a comprehensive summary of existing literature on the aging gut microbiome and its known relationships with neurological diseases, with a specific focus on MS. We will also discuss preclinical animal models and human studies that shed light on the complex microbiota-host interactions that have the potential to influence disease pathology and progression in aging individuals. Lastly, we propose potential avenues of investigation to deconvolute features of an aging microbiota that contribute to disease, or alternatively promote health in advanced age.
Collapse
Affiliation(s)
- Naomi M Fettig
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Annie Pu
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Lisa C Osborne
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
10
|
Woo MS, Engler JB, Friese MA. The neuropathobiology of multiple sclerosis. Nat Rev Neurosci 2024; 25:493-513. [PMID: 38789516 DOI: 10.1038/s41583-024-00823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
Chronic low-grade inflammation and neuronal deregulation are two components of a smoldering disease activity that drives the progression of disability in people with multiple sclerosis (MS). Although several therapies exist to dampen the acute inflammation that drives MS relapses, therapeutic options to halt chronic disability progression are a major unmet clinical need. The development of such therapies is hindered by our limited understanding of the neuron-intrinsic determinants of resilience or vulnerability to inflammation. In this Review, we provide a neuron-centric overview of recent advances in deciphering neuronal response patterns that drive the pathology of MS. We describe the inflammatory CNS environment that initiates neurotoxicity by imposing ion imbalance, excitotoxicity and oxidative stress, and by direct neuro-immune interactions, which collectively lead to mitochondrial dysfunction and epigenetic dysregulation. The neuronal demise is further amplified by breakdown of neuronal transport, accumulation of cytosolic proteins and activation of cell death pathways. Continuous neuronal damage perpetuates CNS inflammation by activating surrounding glia cells and by directly exerting toxicity on neighbouring neurons. Further, we explore strategies to overcome neuronal deregulation in MS and compile a selection of neuronal actuators shown to impact neurodegeneration in preclinical studies. We conclude by discussing the therapeutic potential of targeting such neuronal actuators in MS, including some that have already been tested in interventional clinical trials.
Collapse
Affiliation(s)
- Marcel S Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
11
|
Gommerman JL. What Jane Jacobs has taught me about neuroimmunology. Nat Immunol 2024; 25:381. [PMID: 38429453 DOI: 10.1038/s41590-024-01764-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Affiliation(s)
- Jennifer L Gommerman
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
12
|
Zhang Y, Atkinson J, Burd CE, Graves J, Segal BM. Biological aging in multiple sclerosis. Mult Scler 2023; 29:1701-1708. [PMID: 37877740 PMCID: PMC10843499 DOI: 10.1177/13524585231204122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Multiple sclerosis (MS) is most likely to adopt a progressive clinical course during middle age or beyond, and the number of older adults with MS is steadily increasing. Developing new strategies to manage progressive forms of MS, which do not respond to currently available disease-modifying therapies (DMTs), will require a deeper understanding of the mechanisms by which biological aging interacts with pathogenic pathways to propel disability accumulation. In experimental autoimmune encephalomyelitis (EAE), a widely used preclinical mouse model of MS, middle-aged animals experience a more severe and protracted clinical course than their younger counterparts. This exacerbated disease course is accompanied by persistent neuroinflammation. Clinical studies of age-related biomarkers, such as telomere length, senescence markers, and DNA methylation, suggest that biological aging is accelerated in people with MS compared with age- and sex-matched healthy controls. Furthermore, distinguishing biological age from chronological may afford more precision in determining aging effects in MS. Here we review the current literature on aging biology and its impact on MS pathogenesis. Future research on this topic may lead to the development of novel biomarkers and senotherapy agents that slow neurological decline in people with progressive MS by targeting relevant aging-related pathways.
Collapse
Affiliation(s)
- Yinan Zhang
- Department of Neurology, The Ohio State University Wexner
Medical Center, Columbus, OH
| | - Jeffrey Atkinson
- Department of Neurology, The Ohio State University Wexner
Medical Center, Columbus, OH
| | - Christin E. Burd
- Departments of Molecular Genetics, Cancer Biology and
Genetics, The Ohio State University, Columbus, OH
| | - Jennifer Graves
- Department of Neurosciences, University of California San
Diego, San Diego, CA
| | - Benjamin M. Segal
- Department of Neurology, The Ohio State University Wexner
Medical Center, Columbus, OH
| |
Collapse
|
13
|
Macaron G, Larochelle C, Arbour N, Galmard M, Girard JM, Prat A, Duquette P. Impact of aging on treatment considerations for multiple sclerosis patients. Front Neurol 2023; 14:1197212. [PMID: 37483447 PMCID: PMC10361071 DOI: 10.3389/fneur.2023.1197212] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/18/2023] [Indexed: 07/25/2023] Open
Abstract
With a rapidly aging global population and improvement of outcomes with newer multiple sclerosis (MS)-specific disease-modifying therapies (DMTs), the epidemiology of MS has shifted to an older than previously described population, with a peak prevalence of the disease seen in the 55-65 years age group. Changes in the pathophysiology of MS appear to be age-dependent. Several studies have identified a consistent phase of disability worsening around the fifth decade of life. The latter appears to be independent of prior disease duration and inflammatory activity and concomitant to pathological changes from acute focal active demyelination to chronic smoldering plaques, slow-expanding lesions, and compartmentalized inflammation within the central nervous system (CNS). On the other hand, decreased CNS tissue reserve and poorer remyelinating capacity with aging lead to loss of relapse recovery potential. Aging with MS may imply longer exposure to DMTs, although treatment efficacy in patients >55 years has not been evaluated in pivotal randomized controlled trials and appears to decrease with age. Older individuals are more prone to adverse effects of DMTs, an important aspect of treatment individualization. Aging with MS also implies a higher global burden of comorbid illnesses that contribute to overall impairments and represent a crucial confounder in interpreting clinical worsening. Discontinuation of DMTs after age 55, when no evidence of clinical or radiological activity is detected, is currently under the spotlight. In this review, we will discuss the impact of aging on MS pathobiology, the effect of comorbidities and other confounders on clinical worsening, and focus on current therapeutic considerations in this age group.
Collapse
Affiliation(s)
- Gabrielle Macaron
- Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
- Faculté de Médecine, Université Saint-Joseph de Beyrouth, Beirut, Lebanon
| | - Catherine Larochelle
- Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
| | - Nathalie Arbour
- Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
| | - Manon Galmard
- Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
| | - Jean Marc Girard
- Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
| | - Alexandre Prat
- Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
| | - Pierre Duquette
- Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
14
|
Zuo M, Fettig NM, Bernier LP, Pössnecker E, Spring S, Pu A, Ma XI, Lee DS, Ward LA, Sharma A, Kuhle J, Sled JG, Pröbstel AK, MacVicar BA, Osborne LC, Gommerman JL, Ramaglia V. Age-dependent gray matter demyelination is associated with leptomeningeal neutrophil accumulation. JCI Insight 2022; 7:e158144. [PMID: 35536649 PMCID: PMC9309059 DOI: 10.1172/jci.insight.158144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/04/2022] [Indexed: 11/21/2022] Open
Abstract
People living with multiple sclerosis (MS) experience episodic CNS white matter lesions instigated by autoreactive T cells. With age, patients with MS show evidence of gray matter demyelination and experience devastating nonremitting symptomology. What drives progression is unclear and studying this has been hampered by the lack of suitable animal models. Here, we show that passive experimental autoimmune encephalomyelitis (EAE) induced by an adoptive transfer of young Th17 cells induced a nonremitting clinical phenotype that was associated with persistent leptomeningeal inflammation and cortical pathology in old, but not young, SJL/J mice. Although the quantity and quality of T cells did not differ in the brains of old versus young EAE mice, an increase in neutrophils and a decrease in B cells were observed in the brains of old mice. Neutrophils were also found in the leptomeninges of a subset of progressive MS patient brains that showed evidence of leptomeningeal inflammation and subpial cortical demyelination. Taken together, our data show that while Th17 cells initiate CNS inflammation, subsequent clinical symptoms and gray matter pathology are dictated by age and associated with other immune cells, such as neutrophils.
Collapse
Affiliation(s)
- Michelle Zuo
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Naomi M. Fettig
- Department of Microbiology and Immunology and Life Sciences Institute, and
| | - Louis-Philippe Bernier
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Elisabeth Pössnecker
- Multiple Sclerosis Center & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Neurology, Biomedicine, and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Shoshana Spring
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Annie Pu
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Xianjie I. Ma
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Dennis S.W. Lee
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Lesley A. Ward
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Anshu Sharma
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Jens Kuhle
- Multiple Sclerosis Center & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Neurology, Biomedicine, and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - John G. Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Anne-Katrin Pröbstel
- Multiple Sclerosis Center & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Neurology, Biomedicine, and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Brian A. MacVicar
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lisa C. Osborne
- Department of Microbiology and Immunology and Life Sciences Institute, and
| | | | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Age-related changes in multiple sclerosis and experimental autoimmune encephalomyelitis. Semin Immunol 2022; 59:101631. [PMID: 35752572 DOI: 10.1016/j.smim.2022.101631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
A better understanding of the pathological mechanisms that drive neurodegeneration in people living with multiple sclerosis (MS) is needed to design effective therapies to treat and/or prevent disease progression. We propose that CNS-intrinsic inflammation and re-modelling of the sub-arachnoid space of the leptomeninges sets the stage for neurodegeneration from the earliest stages of MS. While neurodegenerative processes are clinically silent early in disease, ageing results in neurodegenerative changes that become clinically manifest as progressive disability. Here we review pathological correlates of MS disease progression, highlight emerging mouse models that mimic key progressive changes in MS, and provide new perspectives on therapeutic approaches to protect against MS-associated neurodegeneration.
Collapse
|