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
|
Kim J, Jang S, Choi J, Han K, Jung JH, Oh SY, Park KA, Min JH. Association of optic neuritis with incident depressive disorder risk in a Korean nationwide cohort. Sci Rep 2025; 15:7764. [PMID: 40044803 PMCID: PMC11882889 DOI: 10.1038/s41598-025-92370-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/27/2025] [Indexed: 03/09/2025] Open
Abstract
Studies have highlighted complex bidirectional relationships between autoimmune diseases and depressive disorders. Given that early mental health interventions have substantial public health implications, this study investigated association between optic neuritis, an autoimmune inflammatory disorder of the optic nerve, and risk of developing depressive disorders. Utilizing extensive national health insurance data encompassing almost the entire Korean population, this cohort study included 11,745 patients with optic neuritis and 58,725 age- and sex- matched controls between 2010 and 2017. The diagnosis of optic neuritis was confirmed using ICD-10 code H46 and patient medical records. The association with depression risk identified by ICD-10 codes F32 and F33 was assessed using Cox proportional hazards regression models after adjusting for demographics, lifestyle variables, and other comorbidities. Newly diagnosed optic neuritis was associated with an increased risk of depression (hazard ratio = 1.349, 95% confidence interval: 1.277-1.426), independent of potential confounding factors. Subgroup analysis revealed a stronger association for individuals under 50 years, males, current smokers, and those without hypertension. This association suggests that autoimmune neuroinflammatory responses impact mental health differently across demographics. These findings underscore the importance of implementing routine depression screening and developing targeted early intervention strategies for patients with optic neuritis, particularly for those with a high-risk of depression.
Collapse
Affiliation(s)
- Jaeryung Kim
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irown-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Seungwon Jang
- Pyeongtaek Seoul Eye Clinic, Pyeongtaek, Republic of Korea
| | - Junbae Choi
- Samsung Yangjae Forest Mental Clinic, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Jin-Hyung Jung
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Sei Yeul Oh
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irown-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Kyung-Ah Park
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irown-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| | - Ju-Hong Min
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irown-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Amoriello R, Ballerini C. Immunofluorescence Staining of Murine Spinal Cord Sections to Evaluate Microglial Activation and Astrogliosis. Methods Mol Biol 2025; 2857:159-167. [PMID: 39348064 DOI: 10.1007/978-1-0716-4128-6_15] [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: 10/01/2024]
Abstract
Microglia and astrocytes are the main components of the central nervous system (CNS). Upon activation, microglia is able to phagocyte cell debris, pathogens, and toxins; astrocytes support neuronal functions, blood-brain barrier (BBB) homeostasis, and neurotransmitter uptake and metabolism. Furthermore, both cell types can produce cytokines and chemokines. Aging impacts microglia and astrocytes by promoting the production of pro-inflammatory cytokines, impairing microglial phagocytosis and motility and astrocyte glutamate uptake. During neurodegenerative and neuroinflammatory diseases, the aging process may be accelerated contributing to the alteration of CNS glial cells functions. Multiple sclerosis (MS) is an autoimmune, demyelinating disease in which immunosenescence can promote the conversion from relapsing-remitting form to progressive disease. The murine model of experimental autoimmune encephalomyelitis (EAE) allows to investigate MS pathogenesis. Furthermore, EAE can be developed as acute or progressive, mimicking different forms of human MS. Microglia and astrocytes report morphological and functional changes during neuroinflammation that can be investigated in different ways. We here present a protocol for the study of glial cell activation in the spinal cord tissue of EAE mice.
Collapse
Affiliation(s)
- Roberta Amoriello
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Clara Ballerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
4
|
Bolton C. Review of evidence linking exposure to environmental stressors and associated alterations in the dynamics of immunosenescence (ISC) with the global increase in multiple sclerosis (MS). Immun Ageing 2024; 21:73. [PMID: 39438909 PMCID: PMC11494837 DOI: 10.1186/s12979-024-00473-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
Historical survey confirms that, over the latter part of the 20th century, autoimmune-based diseases, including multiple sclerosis (MS), have shown a worldwide increase in incidence and prevalence. Analytical population studies have established that the exponential rise in MS is not solely due to improvements in diagnosis and healthcare but relates to an increase in autoimmune risk factors. Harmful environmental exposures, including non-communicable social determinants of health, anthropogens and indigenous or transmissible microbes, constitute a group of causal determinants that have been closely linked with the global rise in MS cases. Exposure to environmental stressors has profound effects on the adaptive arm of the immune system and, in particular, the associated intrinsic process of immune ageing or immunosenescence (ISC). Stressor-related disturbances to the dynamics of ISC include immune cell-linked untimely or premature (p) alterations and an accelerated replicative (ar) change. A recognised immune-associated feature of MS is pISC and current evidence supports the presence of an arISC during the disease. Moreover, collated data illustrates the immune-associated alterations that characterise pISC and arISC are inducible by environmental stressors strongly implicated in causing duplicate changes in adaptive immune cells during MS. The close relationship between exposure to environmental risk factors and the induction of pISC and arISC during MS offers a valid mechanism through which pro-immunosenescent stressors may act and contribute to the recorded increase in the global rate and number of new cases of the disease. Confirmation of alterations to the dynamics of ISC during MS provides a rational and valuable therapeutic target for the use of senolytic drugs to either prevent accumulation and enhance ablation of less efficient untimely senescent adaptive immune cells or decelerate the dysregulated process of replicative proliferation. A range of senotherapeutics are available including kinase and transcriptase inhibitors, rapalogs, flavanols and genetically-engineered T cells and the use of selective treatments to control emerging and unspecified aspects of pISC and arISC are discussed.
Collapse
|
5
|
Orrù V, Serra V, Marongiu M, Lai S, Lodde V, Zoledziewska M, Steri M, Loizedda A, Lobina M, Piras MG, Virdis F, Delogu G, Marini MG, Mingoia M, Floris M, Masala M, Castelli MP, Mostallino R, Frau J, Lorefice L, Farina G, Fronza M, Carmagnini D, Carta E, Pilotto S, Chessa P, Devoto M, Castiglia P, Solla P, Zarbo RI, Idda ML, Pitzalis M, Cocco E, Fiorillo E, Cucca F. Implications of disease-modifying therapies for multiple sclerosis on immune cells and response to COVID-19 vaccination. Front Immunol 2024; 15:1416464. [PMID: 39076966 PMCID: PMC11284103 DOI: 10.3389/fimmu.2024.1416464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
Introduction Disease-modifying therapies (DMTs) have been shown to improve disease outcomes in multiple sclerosis (MS) patients. They may also impair the immune response to vaccines, including the SARS-CoV-2 vaccine. However, available data on both the intrinsic immune effects of DMTs and their influence on cellular response to the SARS-CoV-2 vaccine are still incomplete. Methods Here, we evaluated the immune cell effects of 3 DMTs on the response to mRNA SARS-CoV-2 vaccination by comparing MS patients treated with one specific therapy (fingolimod, dimethyl fumarate, or natalizumab) with both healthy controls and untreated patients. We profiled 23 B-cell traits, 57 T-cell traits, and 10 cytokines, both at basal level and after stimulation with a pool of SARS-CoV-2 spike peptides, in 79 MS patients, treated with DMTs or untreated, and 32 healthy controls. Measurements were made before vaccination and at three time points after immunization. Results and Discussion MS patients treated with fingolimod showed the strongest immune cell dysregulation characterized by a reduction in all measured lymphocyte cell classes; the patients also had increased immune cell activation at baseline, accompanied by reduced specific immune cell response to the SARS-CoV-2 vaccine. Also, anti-spike specific B cells progressively increased over the three time points after vaccination, even when antibodies measured from the same samples instead showed a decline. Our findings demonstrate that repeated booster vaccinations in MS patients are crucial to overcoming the immune cell impairment caused by DMTs and achieving an immune response to the SARS-CoV-2 vaccine comparable to that of healthy controls.
Collapse
Affiliation(s)
- Valeria Orrù
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Valentina Serra
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Michele Marongiu
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Sandra Lai
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Valeria Lodde
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Magdalena Zoledziewska
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Maristella Steri
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Annalisa Loizedda
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Monia Lobina
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Maria Grazia Piras
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Francesca Virdis
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Giuseppe Delogu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Maura Mingoia
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Matteo Floris
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Marco Masala
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - M. Paola Castelli
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Rafaela Mostallino
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Jessica Frau
- Regional Multiple Sclerosis Center, Azienda Sanitaria Locale (ASL) Cagliari, Cagliari, Italy
| | - Lorena Lorefice
- Regional Multiple Sclerosis Center, Azienda Sanitaria Locale (ASL) Cagliari, Cagliari, Italy
| | - Gabriele Farina
- Neurology Unit, Azienza Ospedaliera Universitaria (AOU) Sassari, Sassari, Italy
| | - Marzia Fronza
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Daniele Carmagnini
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Elisa Carta
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Silvy Pilotto
- Neurology Unit, Azienza Ospedaliera Universitaria (AOU) Sassari, Sassari, Italy
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Paola Chessa
- Neurology Unit, Azienza Ospedaliera Universitaria (AOU) Sassari, Sassari, Italy
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Marcella Devoto
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Paolo Castiglia
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Paolo Solla
- Neurology Unit, Azienza Ospedaliera Universitaria (AOU) Sassari, Sassari, Italy
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Roberto Ignazio Zarbo
- Neurology Unit, Azienza Ospedaliera Universitaria (AOU) Sassari, Sassari, Italy
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Maria Laura Idda
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Maristella Pitzalis
- Institute for Genetic and Biomedical Research, National Research Council, Monserrato, Italy
| | - Eleonora Cocco
- Regional Multiple Sclerosis Center, Azienda Sanitaria Locale (ASL) Cagliari, Cagliari, Italy
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Edoardo Fiorillo
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
| | - Francesco Cucca
- Institute for Genetic and Biomedical Research, National Research Council, Lanusei, Italy
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
6
|
Gelibter S, Saraceno L, Pirro F, Susani EL, Protti A. As time goes by: Treatment challenges in elderly people with multiple sclerosis. J Neuroimmunol 2024; 391:578368. [PMID: 38761652 DOI: 10.1016/j.jneuroim.2024.578368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/11/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
A demographic shift in multiple sclerosis (MS) is leading to an increased number of elderly people with MS (pwMS) and a rise in late-onset MS (LOMS) cases. This shift adds complexity to the treatment management of these patients, due to enhanced treatment-associated risks and the possible interplay between immunosenescence and disease-modifying therapies (DMTs). In the present paper, we performed a systematic review of the current evidence concerning the relationship between aging and treatment management in elderly pwMS. Our literature search identified 35 original studies relevant to this topic. The gathered evidence consistently indicates a diminished efficacy of DMTs in older pwMS, particularly in preventing disability accrual. Against this background, high-efficacy therapies (HETs) appear to show less benefit over moderate-low-efficacy DMTs in older patients. These data mainly derive from observational retrospective studies or meta-analyses conducted on randomized clinical trials (RCTs). RCTs, however, exclude pwMS older than 55 years, limiting our ability to acquire robust evidence regarding this patient group. Regarding treatment discontinuation in elderly pwMS with stable disease, the available data, which mainly focuses on older injectable DMTs, suggests that their suspension appears to be relatively safe in terms of disease activity. Nevertheless, the first RCT specifically targeting treatment discontinuation recently failed to demonstrate the non-inferiority of treatment discontinuation over continuation, in terms of MRI activity. On the other hand, the evidence on the impact of discontinuation on disease progression is more conflicting and less robust. Furthermore, there is an important lack of studies concerning sequestering DMTs and virtually no data on the discontinuation of anti-CD20 monoclonal antibodies. De-escalation strategy is gaining attention as a de-risking approach alternative to complete treatment discontinuation. It may be defined as the decision to shift from HETs to less potent DMTs in elderly pwMS who have a stable disease. This strategy could reduce treatment-related risks, while minimizing the risk of disease activity and progression potentially associated with treatment discontinuation. This approach, however, remains unexplored due to a lack of studies. Given these findings, the present scenario underlines the urgent need for more comprehensive and robust studies to develop optimized, data-driven treatment strategies for elderly pwMS and LOMS, addressing the unique challenges of MS treatment and aging.
Collapse
Affiliation(s)
- Stefano Gelibter
- Department of Neurosciences, Neurology and Stroke Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| | - Lorenzo Saraceno
- Department of Neurosciences, Neurology and Stroke Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Fiammetta Pirro
- Department of Neurosciences, Neurology and Stroke Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Emanuela Laura Susani
- Department of Neurosciences, Neurology and Stroke Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessandra Protti
- Department of Neurosciences, Neurology and Stroke Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
7
|
Krieger S, Cook K, Hersh CM. Understanding multiple sclerosis as a disease spectrum: above and below the clinical threshold. Curr Opin Neurol 2024; 37:189-201. [PMID: 38535979 PMCID: PMC11064902 DOI: 10.1097/wco.0000000000001262] [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: 04/04/2024]
Abstract
PURPOSE OF REVIEW Research in multiple sclerosis (MS) has long been predicated on clinical groupings that do not reflect the underlying biologic heterogeneity apparent within patient populations. This review explicates the various levels of explanation through which the spectrum of disease is described and investigated both above and below the clinical threshold of detection, as framed by the topographical model of MS, to help advance a cogent mechanistic framework. RECENT FINDINGS Contemporary evidence has amended the view of MS as consisting of sequential disease phases in favor of a spectrum of disease with an admixture of interdependent and dynamic pathobiological axes driving tissue injury and progression. Recent studies have shown the presence of acute and compartmentalized inflammation and mechanisms of neurodegeneration beginning early and evolving throughout the disease continuum. Still, the gap between the understanding of immunopathologic processes in MS and the tools used to measure relevant molecular, laboratory, radiologic, and clinical metrics needs attention to enable better prognostication of disease and monitoring for changes along specific pathologic axes and variable treatment outcomes. SUMMARY Aligning on a consistently-applied mechanistic framework at distinct levels of explanation will enable greater precision across bench and clinical research, and inform discourse on drivers of disability progression and delivery of care for individuals with MS.
Collapse
Affiliation(s)
- Stephen Krieger
- Corinne Goldsmith Dickinson Center for MS, Icahn School of Medicine at Mount Sinai
| | - Karin Cook
- Medical Education Director, Neurology at Heartbeat/Publicis Health, New York
| | - Carrie M. Hersh
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic Lou Ruvo Center for Brain Health, Cleveland Clinic Las Vegas, Nevada, USA
| |
Collapse
|
8
|
Zhong M, Salberg S, Sampangi S, van der Walt A, Butzkueven H, Mychasiuk R, Jokubaitis V. Leukocyte telomere length in multiple sclerosis: relationship between disability severity and pregnancy history. Mult Scler Relat Disord 2024; 86:105607. [PMID: 38631073 DOI: 10.1016/j.msard.2024.105607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Aging-related processes contribute to neurodegeneration and disability in multiple sclerosis (MS). Biomarkers of biological aging such as leukocyte telomere length (LTL) could help personalise prognosis. Pregnancy has been shown to be protective against disability accumulation in women with MS, though it is unclear if this effect relates to aging mechanisms or LTL. OBJECTIVES This study aimed to cross-sectionally characterise LTL in a cohort of individuals with MS, and to correlate LTL with disability severity and pregnancy history. METHODS We extracted DNA from the whole blood of 501 people with MS in Melbourne, Australia. Expanded Disability Status Scale (EDSS) score and demographic data, as well as pregnancy history for 197 females, were obtained at sample collection. Additional data were extracted from the MSBase Registry. LTL was determined in base pairs (bp) using real-time quantitative polymerase chain reaction. RESULTS A relationship between EDSS score and shorter LTL was robust to multivariable adjustment for demographic and clinical factors including chronological age, with an adjusted LTL reduction per 1.0 increase in EDSS of 97.1 bp (95 % CI = 9.7-184.5 bp, p = 0.030). Adjusted mediation analysis found chronological age accounted for 33.6 % of the relationship between LTL and EDSS score (p = 0.018). In females with pregnancy data, history of pregnancy was associated with older age (median 49.7 vs 33.0 years, p < 0.001). There were no significant relationships between adjusted LTL and any history of pregnancy (LTL increase of 65.3 bp, 95 % CI = -471.0-601.5 bp, p = 0.81) or number of completed pregnancies (LTL increase of 14.6 bp per pregnancy, 95 % CI = -170.3-199.6 bp, p = 0.87). CONCLUSIONS The correlation between LTL and disability independent of chronological age and other factors points to a link between neurological reserve in MS and biological aging, and a potential research target for pathophysiological and therapeutic mechanisms. Although LTL did not significantly differ by pregnancy history, longitudinal analyses could help identify interactions with prospectively captured pregnancy effects.
Collapse
Affiliation(s)
- Michael Zhong
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Australia.
| | - Sabrina Salberg
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Sandeep Sampangi
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Anneke van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Australia
| |
Collapse
|
9
|
Goyne CE, Fair AE, Sumowski PE, Graves JS. The Impact of Aging on Multiple Sclerosis. Curr Neurol Neurosci Rep 2024; 24:83-93. [PMID: 38416310 DOI: 10.1007/s11910-024-01333-2] [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] [Accepted: 01/17/2024] [Indexed: 02/29/2024]
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) is a chronic, immune-mediated demyelinating disorder of the central nervous system. Age is one of the most important factors in determining MS phenotype. This review provides an overview of how age influences MS clinical characteristics, pathology, and treatment. RECENT FINDINGS New methods for measuring aging have improved our understanding of the aging process in MS. New studies have characterized the molecular and cellular composition of chronic active or smoldering plaques in MS. These lesions are important contributors to disability progression in MS. These studies highlight the important role of immunosenescence and the innate immune system in sustaining chronic inflammation. Given these changes in immune function, several studies have assessed optimal treatment strategies in aging individuals with MS. MS phenotype is intimately linked with chronologic age and immunosenescence. While there are many unanswered questions, there has been much progress in understanding this relationship which may lead to more effective treatments for progressive disease.
Collapse
Affiliation(s)
- Christopher E Goyne
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Ashley E Fair
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Paige E Sumowski
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA.
| |
Collapse
|
10
|
Bufan B, Ćuruvija I, Blagojević V, Grujić-Milanović J, Prijić I, Radosavljević T, Samardžić J, Radosavljevic M, Janković R, Djuretić J. NMDA Receptor Antagonist Memantine Ameliorates Experimental Autoimmune Encephalomyelitis in Aged Rats. Biomedicines 2024; 12:717. [PMID: 38672073 PMCID: PMC11047843 DOI: 10.3390/biomedicines12040717] [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: 01/19/2024] [Revised: 03/15/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Aging is closely related to the main aspects of multiple sclerosis (MS). The average age of the MS population is increasing and the number of elderly MS patients is expected to increase. In addition to neurons, N-methyl-D-aspartate receptors (NMDARs) are also expressed on non-neuronal cells, such as immune cells. The aim of this study was to investigate the role of NMDARs in experimental autoimmune encephalomyelitis (EAE) in young and aged rats. Memantine, a non-competitive NMDAR antagonist, was administered to young and aged Dark Agouti rats from day 7 after immunization. Antagonizing NMDARs had a more favourable effect on clinical disease, reactivation, and apoptosis of CD4+ T cells in the target organ of aged EAE rats. The expression of the fractalkine receptor CX3CR1 was increased in memantine-treated rats, but to a greater extent in aged rats. Additionally, memantine increased Nrf2 and Nrf2-regulated enzymes' mRNA expression in brain tissue. The concentrations of superoxide anion radicals, malondialdehyde, and advanced oxidation protein products in brain tissue were consistent with previous results. Overall, our results suggest that NMDARs play a more important role in the pathogenesis of EAE in aged than in young rats.
Collapse
Affiliation(s)
- Biljana Bufan
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ivana Ćuruvija
- Department of Research and Development, Institute of Virology, Vaccines and Sera, Torlak, 11000 Belgrade, Serbia; (I.Ć.); (V.B.); (I.P.)
| | - Veljko Blagojević
- Department of Research and Development, Institute of Virology, Vaccines and Sera, Torlak, 11000 Belgrade, Serbia; (I.Ć.); (V.B.); (I.P.)
| | - Jelica Grujić-Milanović
- Institute for Medical Research, National Institute of the Republic of Serbia, Department of Cardiovascular Research, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ivana Prijić
- Department of Research and Development, Institute of Virology, Vaccines and Sera, Torlak, 11000 Belgrade, Serbia; (I.Ć.); (V.B.); (I.P.)
| | - Tatjana Radosavljević
- Institute of Pathological Physiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Janko Samardžić
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (J.S.); (M.R.)
| | - Milica Radosavljevic
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (J.S.); (M.R.)
| | - Radmila Janković
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Jasmina Djuretić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
11
|
Zinganell A, Göbel G, Berek K, Hofer B, Asenbaum-Nan S, Barang M, Böck K, Bsteh C, Bsteh G, Eger S, Eggers C, Fertl E, Joldic D, Khalil M, Langenscheidt D, Komposch M, Kornek B, Kraus J, Krendl R, Rauschka H, Sellner J, Auer M, Hegen H, Pauli FD, Deisenhammer F. Multiple sclerosis in the elderly: a retrospective cohort study. J Neurol 2024; 271:674-687. [PMID: 37855871 DOI: 10.1007/s00415-023-12041-1] [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: 07/07/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND There is a lack of knowledge of disease course, prognosis, comorbidities and potential treatments of elderly MS patients. OBJECTIVE To characterize the disease course including disability progression and relapses, to quantify the use of DMTs and to identify comorbidities and risk factors for progression in elderly multiple sclerosis (MS) patients. METHODS This is a retrospective study of 1200 Austrian MS patients older than 55 years as of May 1st, 2017 representing roughly one-third of all the MS patients of this age in Austria. Data were collected from 15 MS centers including demographics, first symptom at onset, number of relapses, evolvement of disability, medication, and comorbidities. RESULTS Median observation time was 17.1 years with 957 (80%) relapsing and 243 (20%) progressive onsets. Average age at diagnosis was 45 years with a female predominance of 71%. Three-hundred and twenty-six (27%) patients were never treated with a DMT, while most treated patients received interferons (496; 41%) at some point. At last follow-up, 420 (35%) patients were still treated with a DMT. No difference was found between treated and never-treated patients in terms of clinical outcome; however, patients with worse disability progression had significantly more DMT switches. Pyramidal onset, number of comorbidities, dementia, epilepsy, and psychiatric conditions as well as a higher number of relapses were associated with worse outcome. The risk of reaching EDSS 6 rose with every additional comorbidity by 22%. In late and very-late-onset MS (LOMS, VLOMS) time to diagnosis took nearly twice the time compared to adult and early onset (AEOMS). The overall annualized relapse rate (ARR) decreased over time and patients with AEOMS had significantly higher ARR compared to LOMS and VLOMS. Four percent of MS patients had five medications or more fulfilling criteria of polypharmacy and 20% of psychiatric drugs were administered without a matching diagnosis. CONCLUSIONS In this study, we identified number of comorbidities, pyramidal and cerebellar signs, and a higher number of relapses as unfavorable prognostic factors in elderly MS patients filling gaps of knowledge in patients usually underrepresented in clinical trials and may guide future therapeutic studies.
Collapse
Affiliation(s)
- Anne Zinganell
- Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Georg Göbel
- Department of Medical Statistics, Informatics and Health Economics, Medical University Innsbruck, Innsbruck, Austria
| | - Klaus Berek
- Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Barbara Hofer
- Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | | | - Matin Barang
- Department of Neurology, Hospital of St. Pölten, St. Pölten, Austria
| | - Klaus Böck
- Department of Neurology, Kepler Universitätsklinikum, Linz, Austria
| | | | - Gabriel Bsteh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Stephan Eger
- Department of Neurology, Kepler Universitätsklinikum, Linz, Austria
| | - Christian Eggers
- Department of Neurology, Kepler Universitätsklinikum, Linz, Austria
| | - Elisabeth Fertl
- Department of Neurology, Klinik Landstrasse, Vienna, Austria
| | - Damir Joldic
- Department of Neurology, Klinik Landstrasse, Vienna, Austria
| | - Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria
| | | | - Martina Komposch
- Department of Neurology, Hospital of Klagenfurt, Klagenfurt, Austria
| | - Barbara Kornek
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Jörg Kraus
- Neurologist, Zell Am See, Austria
- Department of Laboratory Medicine, Paracelsus Medical University and Salzburger Landeskliniken, Salzburg, Austria
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Reinhard Krendl
- Department of Neurology, Hospital of Villach, Villach, Austria
| | - Helmut Rauschka
- Department of Neurology, Klinik Donaustadt, Vienna, Austria
- Karl Landsteiner Institute for Neuroimmunological and Neurodegenerative Disorders, Department of Neurology, Klinik Donaustadt, Vienna, Austria
| | - Johann Sellner
- Department of Neurology, Landesklinikum Mistelbach, Mistelbach, Austria
| | - Michael Auer
- Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Harald Hegen
- Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Franziska Di Pauli
- Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Florian Deisenhammer
- Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
12
|
Xu J, Chen Y, Shi Y, Sun A, Yang Y, Boustani M, Su J, Zhang P. Associations Between Neuroinflammation-Related Conditions and Alzheimer's Disease: A Study of US Insurance Claims Data. J Alzheimers Dis 2024; 99:739-752. [PMID: 38701142 PMCID: PMC11228456 DOI: 10.3233/jad-231286] [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: 05/05/2024]
Abstract
Background Early detection of Alzheimer's disease (AD) is a key component for the success of the recently approved lecanemab and aducanumab. Patients with neuroinflammation-related conditions are associated with a higher risk for developing AD. Objective Investigate the incidence of AD among patients with neuroinflammation-related conditions including epilepsy, hemorrhage stroke, multiple sclerosis (MS), and traumatic brain injury (TBI). Methods We used Optum's de-identified Clinformatics Data Mart Database (CDM). We derived covariate-matched cohorts including patients with neuroinflammation-related conditions and controls without the corresponding condition. The matched cohorts were: 1) patients with epilepsy and controls (N = 67,825 matched pairs); 2) patients with hemorrhage stroke and controls (N = 81,510 matched pairs); 3) patients with MS and controls (N = 9,853 matched pairs); and 4) patients TBI and controls (N = 104,637 matched pairs). We used the Cox model to investigate the associations between neuroinflammation-related conditions and AD. Results We identified that epilepsy, hemorrhage stroke, and TBI were associated with increased risks of AD in both males and females (hazard ratios [HRs]≥1.74, p < 0.001), as well as in gender- and race-conscious subpopulations (HRs≥1.64, p < 0.001). We identified that MS was associated with increased risks of AD in both males and females (HRs≥1.47, p≤0.004), while gender- and race-conscious subgroup analysis shown mixed associations. Conclusions Patients with epilepsy, hemorrhage stroke, MS, and/or TBI are associated with a higher risk of developing AD. More attention on cognitive status should be given to older patients with these conditions.
Collapse
Affiliation(s)
- Jing Xu
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yao Chen
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yi Shi
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anna Sun
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yuedi Yang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Malaz Boustani
- Indiana University Center for Health Innovation and Implementation Science, School of Medicine, Indianapolis, IN, USA
| | - Jing Su
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Pengyue Zhang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
13
|
Pradeep Kumar D, Zanotto T, Cozart JS, Bruce AS, Befort C, Siengsukon C, Shook R, Lynch S, Mahmoud R, Simon S, Hibbing PR, Drees B, Huebner J, Bradish T, Robichaud J, Sosnoff JJ, Bruce JM. Association between frailty and sleep quality in people living with multiple sclerosis and obesity: An observational cross-sectional study. Mult Scler Relat Disord 2024; 81:105154. [PMID: 38043367 DOI: 10.1016/j.msard.2023.105154] [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: 08/15/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND A majority of the people with multiple sclerosis (pwMS) experience sleep disturbances. Frailty is also common in pwMS. The geriatric literature strongly suggests that frailty is associated with worse sleep outcomes in community-dwelling older adults, but this association has yet to be explored among pwMS. This study focused on examining the association between frailty and sleep quality in pwMS. METHODS Seventy-six people with both MS and obesity (mean age: 47.6 ± 10.9 years, 81.6 % female, mean body mass index (BMI): 37.10 ± 5.5 kg/m2, mean Patient Determined Disease Steps (PDDS): 0.82 ± 1.20) were included in this cross-sectional secondary analysis. A comprehensive frailty index (FI) based on 41 health deficits from various health domains was calculated based on standardized procedures. Sleep quality was determined by the Pittsburgh Sleep Quality Index questionnaire (PSQI). RESULTS Overall, 67.1 % of the participants were identified as non-frail (FI ≤ 0.25), and 32.9 % were identified as frail (FI > 0.25). A significant correlation was observed between FI scores and global PSQI scores (ρ = 0.43, p < 0.05). Cross-tabulation analyses revealed that frail participants had worse subjective sleep quality, sleep latency, habitual sleep efficiency, sleep disturbances, daytime dysfunction, and higher use of sleep medications compared to non-frail participants (p < 0.05). CONCLUSIONS The current study identified a significant association between frailty and sleep quality in people with both MS and obesity with minimal disability. These findings underscore the importance of untangling the relationship between frailty and sleep quality in pwMS. These results could lead to a more targeted approach for rehabilitation interventions aiming to improve frailty in MS.
Collapse
Affiliation(s)
- Danya Pradeep Kumar
- Department of Physical Therapy and Rehabilitation Science, School of Health Professions, University of Kansas Medical Center, Kansas City, KS, USA
| | - Tobia Zanotto
- Department of Occupational Therapy Education, School of Health Professions, University of Kansas Medical Center, Kansas City, KS, USA; Mobility Core, University of Kansas Centre for Community Access, Rehabilitation Research, Education and Service, Kansas City, KS, USA; Landon Center on Aging, University of Kansas Medical Center, Kansas City, KS, USA
| | - Julia S Cozart
- Department of Biomedical and Health Informatics, University of Missouri - Kansas City School of Medicine, University of Missouri - Kansas City, Kansas City, MO, USA
| | - Amanda S Bruce
- Center for Children's Healthy Lifestyles and Nutrition, Children's Mercy Hospital, Kansas City, MO, USA; Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Christie Befort
- Department of Population Health, University Kansas Medical Center, Kansas City, KS, USA
| | - Catherine Siengsukon
- Department of Physical Therapy and Rehabilitation Science, School of Health Professions, University of Kansas Medical Center, Kansas City, KS, USA
| | - Robin Shook
- Center for Children's Healthy Lifestyles and Nutrition, Children's Mercy Hospital, Kansas City, MO, USA; Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, USA; School of Medicine, University of Missouri-Kansas City, Kansas City, MO USA
| | - Sharon Lynch
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Rola Mahmoud
- Department of Neurology, University of Missouri, Kansas City, Saint Luke's Hospital, Kansas City, MO, USA
| | - Steve Simon
- Department of Biomedical and Health Informatics, University of Missouri - Kansas City School of Medicine, University of Missouri - Kansas City, Kansas City, MO, USA
| | - Paul R Hibbing
- Department of Kinesiology & Nutrition, University of Illinois Chicago, Chicago, IL, USA
| | - Betty Drees
- Department of Biomedical and Health Informatics, University of Missouri - Kansas City School of Medicine, University of Missouri - Kansas City, Kansas City, MO, USA; Department of Internal Medicine, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA; Graduate School of the Stowers Institute for Medical Research, USA
| | - Joanie Huebner
- UMKC Department of Community and Family Medicine, University Health Lakewood Medical Center, Kansas City, MO, USA
| | - Taylor Bradish
- Department of Biomedical and Health Informatics, University of Missouri - Kansas City School of Medicine, University of Missouri - Kansas City, Kansas City, MO, USA
| | - Jade Robichaud
- Department of Biomedical and Health Informatics, University of Missouri - Kansas City School of Medicine, University of Missouri - Kansas City, Kansas City, MO, USA
| | - Jacob J Sosnoff
- Department of Physical Therapy and Rehabilitation Science, School of Health Professions, University of Kansas Medical Center, Kansas City, KS, USA; Department of Occupational Therapy Education, School of Health Professions, University of Kansas Medical Center, Kansas City, KS, USA; Mobility Core, University of Kansas Centre for Community Access, Rehabilitation Research, Education and Service, Kansas City, KS, USA; Landon Center on Aging, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jared M Bruce
- Department of Biomedical and Health Informatics, University of Missouri - Kansas City School of Medicine, University of Missouri - Kansas City, Kansas City, MO, USA; Departments of Neurology and Psychiatry, University Health, Kansas City, MO, USA.
| |
Collapse
|
14
|
Mendes O. Inflammation and neurodegeneration in multiple sclerosis. A REVIEW ON DIVERSE NEUROLOGICAL DISORDERS 2024:321-345. [DOI: 10.1016/b978-0-323-95735-9.00023-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
15
|
Zou P, Wu C, Liu TCY, Duan R, Yang L. Oligodendrocyte progenitor cells in Alzheimer's disease: from physiology to pathology. Transl Neurodegener 2023; 12:52. [PMID: 37964328 PMCID: PMC10644503 DOI: 10.1186/s40035-023-00385-7] [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: 08/10/2023] [Accepted: 11/01/2023] [Indexed: 11/16/2023] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) play pivotal roles in myelin formation and phagocytosis, communicating with neighboring cells and contributing to the integrity of the blood-brain barrier (BBB). However, under the pathological circumstances of Alzheimer's disease (AD), the brain's microenvironment undergoes detrimental changes that significantly impact OPCs and their functions. Starting with OPC functions, we delve into the transformation of OPCs to myelin-producing oligodendrocytes, the intricate signaling interactions with other cells in the central nervous system (CNS), and the fascinating process of phagocytosis, which influences the function of OPCs and affects CNS homeostasis. Moreover, we discuss the essential role of OPCs in BBB formation and highlight the critical contribution of OPCs in forming CNS-protective barriers. In the context of AD, the deterioration of the local microenvironment in the brain is discussed, mainly focusing on neuroinflammation, oxidative stress, and the accumulation of toxic proteins. The detrimental changes disturb the delicate balance in the brain, impacting the regenerative capacity of OPCs and compromising myelin integrity. Under pathological conditions, OPCs experience significant alterations in migration and proliferation, leading to impaired differentiation and a reduced ability to produce mature oligodendrocytes. Moreover, myelin degeneration and formation become increasingly active in AD, contributing to progressive neurodegeneration. Finally, we summarize the current therapeutic approaches targeting OPCs in AD. Strategies to revitalize OPC senescence, modulate signaling pathways to enhance OPC differentiation, and explore other potential therapeutic avenues are promising in alleviating the impact of AD on OPCs and CNS function. In conclusion, this review highlights the indispensable role of OPCs in CNS function and their involvement in the pathogenesis of AD. The intricate interplay between OPCs and the AD brain microenvironment underscores the complexity of neurodegenerative diseases. Insights from studying OPCs under pathological conditions provide a foundation for innovative therapeutic strategies targeting OPCs and fostering neurodegeneration. Future research will advance our understanding and management of neurodegenerative diseases, ultimately offering hope for effective treatments and improved quality of life for those affected by AD and related disorders.
Collapse
Affiliation(s)
- Peibin Zou
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Timon Cheng-Yi Liu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Rui Duan
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
16
|
Alruwaili M, Al-kuraishy HM, Alexiou A, Papadakis M, ALRashdi BM, Elhussieny O, Saad HM, Batiha GES. Pathogenic Role of Fibrinogen in the Neuropathology of Multiple Sclerosis: A Tale of Sorrows and Fears. Neurochem Res 2023; 48:3255-3269. [PMID: 37442896 PMCID: PMC10514123 DOI: 10.1007/s11064-023-03981-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating neurodegenerative disease of the central nervous system (CNS) due to injury of the myelin sheath by immune cells. The clotting factor fibrinogen is involved in the pathogenesis of MS by triggering microglia and the progress of neuroinflammation. Fibrinogen level is correlated with MS severity; consequently, inhibition of the fibrinogen cascade may reduce MS neuropathology. Thus, this review aimed to clarify the potential role of fibrinogen in the pathogenesis of MS and how targeting of fibrinogen affects MS neuropathology. Accumulation of fibrinogen in the CNS may occur independently or due to disruption of blood-brain barrier (BBB) integrity in MS. Fibrinogen acts as transduction and increases microglia activation which induces the progression of inflammation, oxidative stress, and neuronal injury. Besides, brain fibrinogen impairs the remyelination process by inhibiting the differentiation of oligodendrocyte precursor cells. These findings proposed that fibrinogen is associated with MS neuropathology through interruption of BBB integrity, induction of neuroinflammation, and demyelination with inhibition of the remyelination process by suppressing oligodendrocytes. Therefore, targeting of fibrinogen and/or CD11b/CD18 receptors by metformin and statins might decrease MS neuropathology. In conclusion, inhibiting the expression of CD11b/CD18 receptors by metformin and statins may decrease the pro-inflammatory effect of fibrinogen on microglia which is involved in the progression of MS.
Collapse
Affiliation(s)
- Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M. Al-kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770 Australia
- AFNP Med, 1030 Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283 Wuppertal, Germany
| | - Barakat M. ALRashdi
- Biology Department, College of Science, Jouf University, Sakaka, 41412 Saudi Arabia
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744 Egypt
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744 Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Egypt
| |
Collapse
|
17
|
Thakolwiboon S, Mills EA, Yang J, Doty J, Belkin MI, Cho T, Schultz C, Mao-Draayer Y. Immunosenescence and multiple sclerosis: inflammaging for prognosis and therapeutic consideration. FRONTIERS IN AGING 2023; 4:1234572. [PMID: 37900152 PMCID: PMC10603254 DOI: 10.3389/fragi.2023.1234572] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023]
Abstract
Aging is associated with a progressive decline of innate and adaptive immune responses, called immunosenescence. This phenomenon links to different multiple sclerosis (MS) disease courses among different age groups. While clinical relapse and active demyelination are mainly related to the altered adaptive immunity, including invasion of T- and B-lymphocytes, impairment of innate immune cell (e.g., microglia, astrocyte) function is the main contributor to disability progression and neurodegeneration. Most patients with MS manifest the relapsing-remitting phenotype at a younger age, while progressive phenotypes are mainly seen in older patients. Current disease-modifying therapies (DMTs) primarily targeting adaptive immunity are less efficacious in older patients, suggesting that immunosenescence plays a role in treatment response. This review summarizes the recent immune mechanistic studies regarding immunosenescence in patients with MS and discusses the clinical implications of these findings.
Collapse
Affiliation(s)
| | - Elizabeth A. Mills
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Jennifer Yang
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Jonathan Doty
- Michigan Institute for Neurological Disorders, Farmington Hills, MI, United States
| | - Martin I. Belkin
- Michigan Institute for Neurological Disorders, Farmington Hills, MI, United States
| | - Thomas Cho
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Charles Schultz
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Michigan Institute for Neurological Disorders, Farmington Hills, MI, United States
- Autoimmune Center of Excellence, University of Michigan, Ann Arbor, MI, United States
- Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
18
|
Chalmers S, Harrall K, Wong SY, Kablan W, Clunie G. A retrospective study of patients presenting with speech and language therapy needs within multidisciplinary Long COVID services: A service evaluation describing and comparing two cohorts across two NHS Trusts. INTERNATIONAL JOURNAL OF LANGUAGE & COMMUNICATION DISORDERS 2023; 58:1424-1439. [PMID: 36916685 DOI: 10.1111/1460-6984.12868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/22/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Post-COVID Syndrome (also known as Long COVID) refers to the multi-system condition affecting individuals following COVID-19 infection. This can include speech and language therapy (SLT) needs, including voice, swallowing, communication and upper airway difficulties. There is limited published literature in this clinical area of practice, particularly for those receiving input from community SLT services. AIMS To describe and compare demand, typical SLT presentation and service delivery across two National Health Service (NHS) Long COVID multidisciplinary services. Independent retrospective service evaluation was completed for each service. Descriptive statistics were produced and compared across services. This service evaluation followed The Strengthening the Reporting of Observation Studies in Epidemiology guidelines for cohort studies. OUTCOMES & RESULTS The findings indicated similarities across the two services in SLT service need and demand, clinical presentations and intervention approaches provided within Long COVID services. There were specific differences in the service provision and delivery of intervention in cognitive communication and upper airways subspecialities. CONCLUSIONS & IMPLICATIONS This study highlights the clinical complexities of SLT needs in individuals with Long COVID and the importance for an appropriately skilled and supported workforce within effective multidisciplinary teams. We call for consensus on SLT practices and a consistent and standardized approach to evaluation for SLT needs in Long COVID. WHAT THIS PAPER ADDS What is already known on this subject SLT needs, including voice, swallowing, communication and upper airway difficulties, are present in individuals presenting with Long COVID, both in those who were or were not hospitalized. SLTs are seeing such individuals in a variety of settings, including community services and Long COVID multidisciplinary teams. There is minimal evidence of the clinical presentations and interventions provided to individuals with SLT needs compared across Long COVID services. What this study adds to existing knowledge This study compares two NHS Long COVID services providing a SLT service pathway. It highlights the similarities and differences in service demand and capacity, patient presentation, and SLT intervention to make suggestions for future practice consideration and priority evaluation. Expert consensus among SLT clinicians is a priority to ensure clinicians are delivering consistent and equitable care for patients, while new evidence and data emerge. A consistent and standardized approach to data collection and outcome measures is essential to ensure future research captures the impact and value of SLT input with individuals with Long COVID. What are the clinical implications of this work? The complexities and multifactorial SLT needs of individual with Long COVID call for appropriate SLT staffing provision, skill and training to fulfil the needs of this population. Speech and language therapists should be integrated with multidisciplinary Long COVID services to provide holistic care for patients and to support the professionals working with individuals with post-COVID voice, swallowing, communication and upper airway symptoms.
Collapse
Affiliation(s)
- Sophie Chalmers
- Long COVID Service, Long Term Conditions, Integrated Community Services Division, Bolton NHS Foundation Trust, Bolton, UK
- Allied Health Research Unit, School of Sport and Health Sciences, University of Central Lancashire, Preston, UK
| | - Kate Harrall
- Speech and Language Therapy Dept, Integrated Pathways Division, East Suffolk and North Essex NHS Foundation Trust, Colchester, UK
- Research and Development Unit, Strategy, Research and Innovation Division, East Suffold and North Essex NHS Foundation Trust, Colchester, UK
| | - Sze Yin Wong
- Speech and Language Therapy Dept, Integrated Pathways Division, East Suffolk and North Essex NHS Foundation Trust, Colchester, UK
| | - Widad Kablan
- Paediatric Speech and Language Therapy, Family Care Division, Bolton NHS Foundation Trust, Bolton, UK
| | - Gemma Clunie
- Speech and Language Therapy, Surgery and Cancer, Imperial College London Healthcare NHS Trust & Imperial College London, London, UK
| |
Collapse
|
19
|
Bridge F, Butzkueven H, Van der Walt A, Jokubaitis VG. The impact of menopause on multiple sclerosis. Autoimmun Rev 2023; 22:103363. [PMID: 37230311 DOI: 10.1016/j.autrev.2023.103363] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/21/2023] [Indexed: 05/27/2023]
Abstract
Menopause, defined as the permanent cessation of ovarian function, represents a period of significant fluctuation in sex hormone concentrations. Sex hormones including oestrogen, progesterone, testosterone and anti-Mullerian hormone are thought have neuroinflammatory effects and are implicated in both neuroprotection and neurodegeneration. Sex hormones are thought to have a role in modifying clinical trajectory in multiple sclerosis (MS) throughout the lifespan. Multiple sclerosis predominantly effects women and is typically diagnosed early in a woman's reproductive life. Most women with MS will undergo menopause. Despite this, the effect of menopause on MS disease course remains unclear. This review examines the relationship between sex hormones and MS disease activity and clinical course, particularly around the time of menopause. It will consider the role of interventions such as exogenous hormone replacement therapy in modulating clinical outcomes in this period. Understanding the impact of menopause on multiple sclerosis is fundamental for delivering optimal care to women with MS as they age and will inform treatment decisions with the aim of minimising relapses, disease accrual and improving quality of life.
Collapse
Affiliation(s)
- Francesca Bridge
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Anneke Van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Vilija G Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
20
|
Brasanac J, Chien C. A review on multiple sclerosis prognostic findings from imaging, inflammation, and mental health studies. Front Hum Neurosci 2023; 17:1151531. [PMID: 37250694 PMCID: PMC10213782 DOI: 10.3389/fnhum.2023.1151531] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
Magnetic resonance imaging (MRI) of the brain is commonly used to detect where chronic and active lesions are in multiple sclerosis (MS). MRI is also extensively used as a tool to calculate and extrapolate brain health by way of volumetric analysis or advanced imaging techniques. In MS patients, psychiatric symptoms are common comorbidities, with depression being the main one. Even though these symptoms are a major determinant of quality of life in MS, they are often overlooked and undertreated. There has been evidence of bidirectional interactions between the course of MS and comorbid psychiatric symptoms. In order to mitigate disability progression in MS, treating psychiatric comorbidities should be investigated and optimized. New research for the prediction of disease states or phenotypes of disability have advanced, primarily due to new technologies and a better understanding of the aging brain.
Collapse
Affiliation(s)
- Jelena Brasanac
- Charité – Universitätsmedizin Berlin, Klinik für Psychiatrie und Psychotherapie, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Medizinische Klinik m.S. Psychosomatik, Berlin, Germany
| | - Claudia Chien
- Charité – Universitätsmedizin Berlin, Klinik für Psychiatrie und Psychotherapie, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Medizinische Klinik m.S. Psychosomatik, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Neuroscience Clinical Research Center, Berlin, Germany
| |
Collapse
|
21
|
Torres P, Sancho-Saldaña A, Gil Sánchez A, Peralta S, Solana MJ, Bakkioui S, González-Mingot C, Quibus L, Ruiz-Fernández E, San Pedro-Murillo E, Brieva L. A prospective study of cellular immune response to booster COVID-19 vaccination in multiple sclerosis patients treated with a broad spectrum of disease-modifying therapies. J Neurol 2023; 270:2380-2391. [PMID: 36933032 PMCID: PMC10024306 DOI: 10.1007/s00415-023-11575-8] [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: 10/27/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 03/19/2023]
Abstract
BACKGROUND Most people with Multiple Sclerosis (pwMS) are subjected to immunomodulatory disease-modifying treatments (DMTs). As a result, immune responses to COVID-19 vaccinations could be compromised. There are few data on cellular immune responses to the use of COVID-19 vaccine boosters in pwMS under a broad spectrum of DMTs. METHODS In this prospective study, we analysed cellular immune responses to SARS-CoV-2 mRNA booster vaccinations in 159 pwMS with DMT, including: ocrelizumab, rituximab, fingolimod, alemtuzumab, dimethyl fumarate, glatiramer acetate, teriflunomide, natalizumab and cladribine. RESULTS DMTs, and particularly fingolimod, interact with cellular responses to COVID-19 vaccination. One booster dose does not increase cellular immunity any more than two doses, except in the cases of natalizumab and cladribine. SARS-CoV-2 infection combined with two doses of vaccine resulted in a greater cellular immune response, but this was not observed after supplementary booster jabs. Ocrelizumab-treated pwMS who had previously received fingolimod did not develop cellular immunity, even after receiving a booster. The time after MS diagnosis and disability status negatively correlated with cellular immunity in ocrelizumab-treated pwMS in a booster dose cohort. CONCLUSIONS After two doses of SARS-CoV-2 vaccination, a high response yield was achieved, except in patients who had received fingolimod. The effects of fingolimod on cellular immunity persisted for more than 2 years after a change to ocrelizumab (which, in contrast, conserved cellular immunity). Our results confirmed the need to find alternative protective measures for fingolimod-treated people and to consider the possible failure to provide protection against SARS-CoV-2 when switching from fingolimod to ocrelizumab.
Collapse
Affiliation(s)
- Pascual Torres
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida (UdL)-IRBLleida, 25198, Lleida, Spain
- Neuroimmunology Group, Department of Medicine, University of Lleida (UdL)-IRBLleida, 25198, Lleida, Spain
| | - Agustín Sancho-Saldaña
- Neuroimmunology Group, Department of Medicine, University of Lleida (UdL)-IRBLleida, 25198, Lleida, Spain
- Department of Neurology, Hospital Universitari Arnau de Vilanova, 25198, Lleida, Spain
| | - Anna Gil Sánchez
- Neuroimmunology Group, Department of Medicine, University of Lleida (UdL)-IRBLleida, 25198, Lleida, Spain
| | - Silvia Peralta
- Neuroimmunology Group, Department of Medicine, University of Lleida (UdL)-IRBLleida, 25198, Lleida, Spain
- Department of Neurology, Hospital Universitari Arnau de Vilanova, 25198, Lleida, Spain
| | - Maria José Solana
- Neuroimmunology Group, Department of Medicine, University of Lleida (UdL)-IRBLleida, 25198, Lleida, Spain
- Department of Neurology, Hospital Universitari Arnau de Vilanova, 25198, Lleida, Spain
| | - Sofian Bakkioui
- Neuroimmunology Group, Department of Medicine, University of Lleida (UdL)-IRBLleida, 25198, Lleida, Spain
| | - Cristina González-Mingot
- Neuroimmunology Group, Department of Medicine, University of Lleida (UdL)-IRBLleida, 25198, Lleida, Spain
- Department of Neurology, Hospital Universitari Arnau de Vilanova, 25198, Lleida, Spain
| | - Laura Quibus
- Neuroimmunology Group, Department of Medicine, University of Lleida (UdL)-IRBLleida, 25198, Lleida, Spain
- Department of Neurology, Hospital Universitari Arnau de Vilanova, 25198, Lleida, Spain
| | - Emilio Ruiz-Fernández
- Department of Neurology, Hospital Universitari Arnau de Vilanova, 25198, Lleida, Spain
| | | | - Luis Brieva
- Neuroimmunology Group, Department of Medicine, University of Lleida (UdL)-IRBLleida, 25198, Lleida, Spain.
- Department of Neurology, Hospital Universitari Arnau de Vilanova, 25198, Lleida, Spain.
| |
Collapse
|
22
|
Kesidou E, Theotokis P, Damianidou O, Boziki M, Konstantinidou N, Taloumtzis C, Sintila SA, Grigoriadis P, Evangelopoulos ME, Bakirtzis C, Simeonidou C. CNS Ageing in Health and Neurodegenerative Disorders. J Clin Med 2023; 12:2255. [PMID: 36983254 PMCID: PMC10054919 DOI: 10.3390/jcm12062255] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
The process of ageing is characteristic of multicellular organisms associated with late stages of the lifecycle and is manifested through a plethora of phenotypes. Its underlying mechanisms are correlated with age-dependent diseases, especially neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and multiple sclerosis (MS) that are accompanied by social and financial difficulties for patients. Over time, people not only become more prone to neurodegeneration but they also lose the ability to trigger pivotal restorative mechanisms. In this review, we attempt to present the already known molecular and cellular hallmarks that characterize ageing in association with their impact on the central nervous system (CNS)'s structure and function intensifying possible preexisting pathogenetic conditions. A thorough and elucidative study of the underlying mechanisms of ageing will be able to contribute further to the development of new therapeutic interventions to effectively treat age-dependent manifestations of neurodegenerative diseases.
Collapse
Affiliation(s)
- Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
- Laboratory of Physiology, Faculty of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Olympia Damianidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Natalia Konstantinidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Charilaos Taloumtzis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Styliani-Aggeliki Sintila
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Panagiotis Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | | | - Christos Bakirtzis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Constantina Simeonidou
- Laboratory of Physiology, Faculty of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| |
Collapse
|
23
|
Ajala A, Uzairu A, Shallangwa GA, Abechi SE. QSAR, simulation techniques, and ADMET/pharmacokinetics assessment of a set of compounds that target MAO-B as anti-Alzheimer agent. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2023. [DOI: 10.1186/s43094-022-00452-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Abstract
Background
Alzheimer’s disease (AD), the most common cause of dementia in the elderly, is a progressive neurodegenerative disorder that gradually affects cognitive function and eventually causes death. Most approved drugs can only treat the disease alleviating the disease symptoms; therefore, there is a need to develop drugs that can treat this illness holistically. The medical community is searching for new drugs and new drug targets to cure this disease. In this study, QSAR, molecular docking evaluation, and ADMET/pharmacokinetics assessment were used as modeling methods to identify the compounds with outstanding physicochemical properties.
Results
The 37 MAO-B compounds were screened using the aforementioned methods and yielded a model with the following molecular properties: AATS1v, AATS3v, GATS4m, and GATS6e. Good statistical values were R2train = 0.69, R2adj = 0.63, R2pred = 0.57, LOF = 0.23, and RMSE = 0.38. The model was validated using an evaluation set that confirmed its robustness. The molecular docking was also utilized using crystal structure of human monoamine oxidase B in complex with chlorophenylchromone-carboxamide with ID code of 6FW0, and three compounds were identified with outstanding high binding affinity (13 = − 30.51 kcal mol−1, 31 = − 31.85 kcal mol−1, and 33 = − 33.70 kcal mol−1), and better than the Eldepryl (referenced) drug (− 11.40 kcal mol−1).
Conclusions
These three compounds (13, 31, and 33) were analyzed for ADMET/pharmacokinetics evaluation and found worthy of further analysis as promising drug candidates to cure AD and could also serve as a template to design several monoamine oxidase B inhibitors in the future to cure AD.
Collapse
|
24
|
López-Armas GDC, Ramos-Márquez ME, Navarro-Meza M, Macías-Islas MÁ, Saldaña-Cruz AM, Zepeda-Moreno A, Siller-López F, Cruz-Ramos JA. Leukocyte Telomere Length Predicts Severe Disability in Relapsing-Remitting Multiple Sclerosis and Correlates with Mitochondrial DNA Copy Number. Int J Mol Sci 2023; 24:ijms24020916. [PMID: 36674427 PMCID: PMC9862686 DOI: 10.3390/ijms24020916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/12/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune inflammatory disease that affects the nervous system. Peripheral blood leukocyte telomere length (LTL) and mitochondrial DNA copy number (mtDNA-CN) are potential biomarkers of neurological disability and neural damage. Our objective was to assess the LTL and mtDNA-CN in relapsing-remitting MS (RRMS). We included 10 healthy controls, 75 patients with RRMS, 50 of whom had an Expanded Disability Status Scale (EDSS) from 0 to 3 (mild to moderate disability), and 25 had an EDSS of 3.5 to 7 (severe disability). We use the Real-Time Polymerase Chain Reaction (qPCR) technique to quantify absolute LTL and absolute mtDNA-CN. ANOVA test show differences between healthy control vs. severe disability RRMS and mild-moderate RRMS vs. severe disability RRMS (p = 0.0130). LTL and mtDNA-CN showed a linear correlation in mild-moderate disability RRMS (r = 0.378, p = 0.007). Furthermore, we analyzed LTL between RRMS groups with a ROC curve, and LTL can predict severe disability (AUC = 0.702, p = 0.0018, cut-off < 3.0875 Kb, sensitivity = 75%, specificity = 62%), whereas the prediction is improved with a logistic regression model including LTL plus age (AUC = 0.762, p = 0.0001, sensitivity = 79.17%, specificity = 80%). These results show that LTL is a biomarker of disability in RRMS and is correlated with mtDNA-CN in mild-moderate RRMS patients.
Collapse
Affiliation(s)
- Gabriela del Carmen López-Armas
- Laboratorio de Biomédica-Mecatrónica, Subdirección de Investigación y Extensión, Centro de Enseñanza Técnica Industrial Plantel Colomos, Guadalajara 44638, Mexico
| | - Martha Eloisa Ramos-Márquez
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Mónica Navarro-Meza
- Laboratorio C. de Neuronutrición y Memoria, Departamento de Promoción, Preservación y Desarrollo de la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzmán 49000, Mexico
| | - Miguel Ángel Macías-Islas
- Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Ana Miriam Saldaña-Cruz
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Abraham Zepeda-Moreno
- Departamento de Clínicas de la Reproducción Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Fernando Siller-López
- Programa de Bacteriología, Facultad de Ciencias de la Salud, Universidad Católica de Manizales, Manizales 170002, Colombia
| | - José Alfonso Cruz-Ramos
- Departamento de Clínicas Médicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
- Coordinación de Investigación, Instituto Jalisciense de Cancerología, Guadalajara 44280, Mexico
- Correspondence:
| |
Collapse
|
25
|
Taylor BK, Smith OV, Miller GE. Chronic Home Radon Exposure Is Associated with Higher Inflammatory Biomarker Concentrations in Children and Adolescents. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 20:246. [PMID: 36612568 PMCID: PMC9819293 DOI: 10.3390/ijerph20010246] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023]
Abstract
Children are particularly vulnerable to the deleterious impacts of toxic environmental exposures, though the effects of some rather ubiquitous toxins have yet to be characterized in youths. One such toxin, radon gas, is known to accumulate to hazardous levels in homes, and has been linked with the incidence of lung cancer in aging adults. However, the degree to which chronic home radon exposure may impact risk for health problems earlier in life is unknown. Herein, we explored the degree to which chronic home radon exposure relates to biomarkers of low-grade inflammation in 68 youths ages 6- to 14 years old residing in an area of the United States prone to high home radon concentrations. Parents completed a home radon test kit, and youths provided a saliva sample to assess concentrations of five biomarkers. Using a multiple regression approach, we found that greater radon exposure was specifically associated with higher levels of C-reactive protein (β = 0.31, p = 0.007) and interleukin-1β (β = 0.33, p = 0.016). The data suggested specificity in associations between chronic home radon exposure and different biomarkers of inflammatory activity and highlight a pathway which may confer risk for future mental and physical health maladies.
Collapse
Affiliation(s)
- Brittany K. Taylor
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE 68010, USA
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68178, USA
| | - OgheneTejiri V. Smith
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE 68010, USA
| | - Gregory E. Miller
- Institute for Policy Research and Department of Psychology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
26
|
Moridi T. Association between brain volume and disability over time in multiple sclerosis. Mult Scler J Exp Transl Clin 2022; 8:20552173221144230. [PMID: 36570871 PMCID: PMC9768834 DOI: 10.1177/20552173221144230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
Background Most previous multiple sclerosis (MS) brain atrophy studies using MS impact scale 29 (MSIS-29) or symbol digit modalities test (SDMT) have been cross-sectional with limited sets of clinical outcomes. Objectives To investigate which brain and lesion volume metrics show the strongest long-term associations with the expanded disability status scale (EDSS), SDMT, and MSIS-29, and whether MRI-clinical associations vary with age. Methods We acquired MRI and clinical data from a real-world Swedish MS cohort. FreeSurfer and SPM Lesion Segmentation Tool were used to obtain brain parenchymal, cortical and subcortical grey matter, thalamic and white matter fractions as well as T1- and T2-lesion volumes. Mixed-effects and rolling regression models were used in the statistical analyses. Results We included 989 persons with MS followed for a median of 9.3 (EDSS), 10.1 (SDMT), and 9.3 (MSIS-29) years, respectively. In a cross-sectional analysis, the strength of the associations of the MRI metrics with the EDSS and MSIS-29 was found to drastically increase after 40-50 years of age. Low baseline regional grey matter fractions were associated with longitudinal increase of EDSS and physical MSIS-29 scores and decrease in SDMT scores and these atrophy measures were stronger predictors than the lesion volumes. Conclusions The strength of MRI-clinical associations increase with age. Grey matter volume fractions are stronger predictors of long-term disability measures than lesion volumes.
Collapse
Affiliation(s)
- Thomas Moridi
- Thomas Moridi, Department of Clinical Neuroscience, Karolinska Institutet, K8 Klinisk neurovetenskap, K8 Neuro Kockum, Stockholm, 171 77, Sweden.
| |
Collapse
|
27
|
Murgia F, Giagnoni F, Lorefice L, Caria P, Dettori T, D’Alterio MN, Angioni S, Hendren AJ, Caboni P, Pibiri M, Monni G, Cocco E, Atzori L. Sex Hormones as Key Modulators of the Immune Response in Multiple Sclerosis: A Review. Biomedicines 2022; 10:biomedicines10123107. [PMID: 36551863 PMCID: PMC9775368 DOI: 10.3390/biomedicines10123107] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND A variety of autoimmune diseases, including MS, amplify sex-based physiological differences in immunological responsiveness. Female MS patients experience pathophysiological changes during reproductive phases (pregnancy and menopause). Sex hormones can act on immune cells, potentially enabling them to modify MS risk, activity, and progression, and to play a role in treatment. METHODS Scientific papers (published between 1998 and 2021) were selected through PubMed, Google Scholar, and Web of Science literature repositories. The search was limited to publications analyzing the hormonal profile of male and female MS patients during different life phases, in particular focusing on sex hormone treatment. RESULTS Both men and women with MS have lower testosterone levels compared to healthy controls. The levels of estrogens and progesterone increase during pregnancy and then rapidly decrease after delivery, possibly mediating an immune-stabilizing process. The literature examined herein evidences the neuroprotective effect of testosterone and estrogens in MS, supporting further examinations of their potential therapeutic uses. CONCLUSIONS A correlation has been identified between sex hormones and MS clinical activity. The combination of disease-modifying therapies with estrogen or estrogen plus a progestin receptor modulator promoting myelin repair might represent an important strategy for MS treatment in the future.
Collapse
Affiliation(s)
- Federica Murgia
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
- Correspondence:
| | - Florianna Giagnoni
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Lorena Lorefice
- Multiple Sclerosis Regional Center, ASSL Cagliari, ATS Sardinia, 09126 Cagliari, Italy
| | - Paola Caria
- Department of Biomedical Sciences, Section of Biochemistry, Biology, and Genetics, University of Cagliari, Cittadella Universitaria, 09124 Cagliari, Italy
| | - Tinuccia Dettori
- Department of Biomedical Sciences, Section of Biochemistry, Biology, and Genetics, University of Cagliari, Cittadella Universitaria, 09124 Cagliari, Italy
| | - Maurizio N. D’Alterio
- Division of Gynecology and Obstetrics, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Stefano Angioni
- Division of Gynecology and Obstetrics, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Aran J. Hendren
- Sussex Neuroscience, University of Sussex, Brighton BN1 9QG, UK
| | - Pierluigi Caboni
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Monica Pibiri
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Giovanni Monni
- Department of Obstetrics and Gynecology, Prenatal and Preimplantation Genetic Diagnosis, Fetal Therapy, Microcitemico Pediatric Hospital “A. Cao”, 09121 Cagliari, Italy
| | - Eleonora Cocco
- Multiple Sclerosis Regional Center, ASSL Cagliari, ATS Sardinia, Department of Medical Sciences and Public Health, University of Cagliari, 09126 Cagliari, Italy
| | - Luigi Atzori
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
28
|
Clarkson BDS, Grund E, David K, Johnson RK, Howe CL. ISGylation is induced in neurons by demyelination driving ISG15-dependent microglial activation. J Neuroinflammation 2022; 19:258. [PMID: 36261842 PMCID: PMC9583544 DOI: 10.1186/s12974-022-02618-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/07/2022] [Indexed: 11/22/2022] Open
Abstract
The causes of grey matter pathology and diffuse neuron injury in MS remain incompletely understood. Axonal stress signals arising from white matter lesions has been suggested to play a role in initiating this diffuse grey matter pathology. Therefore, to identify the most upstream transcriptional responses in neurons arising from demyelinated axons, we analyzed the transcriptome of actively translating neuronal transcripts in mouse models of demyelinating disease. Among the most upregulated genes, we identified transcripts associated with the ISGylation pathway. ISGylation refers to the covalent attachment of the ubiquitin-like molecule interferon stimulated gene (ISG) 15 to lysine residues on substrates targeted by E1 ISG15-activating enzyme, E2 ISG15-conjugating enzymes and E3 ISG15-protein ligases. We further confirmed that ISG15 expression is increased in MS cortical and deep gray matter. Upon investigating the functional impact of neuronal ISG15 upregulation, we noted that ISG15 expression was associated changes in neuronal extracellular vesicle protein and miRNA cargo. Specifically, extracellular vesicle-associated miRNAs were skewed toward increased frequency of proinflammatory and neurotoxic miRNAs and decreased frequency of anti-inflammatory and neuroprotective miRNAs. Furthermore, we found that ISG15 directly activated microglia in a CD11b-dependent manner and that microglial activation was potentiated by treatment with EVs from neurons expressing ISG15. Further study of the role of ISG15 and ISGylation in neurons in MS and neurodegenerative diseases is warranted.
Collapse
Affiliation(s)
- Benjamin D. S. Clarkson
- grid.66875.3a0000 0004 0459 167XDepartment of Neurology, Mayo Clinic, Rochester, MN 55905 USA ,grid.66875.3a0000 0004 0459 167XDepartment of Laboratory Medicine and Pathology, Mayo Clinic, Guggenheim 1521C, 200 First Street SW, Rochester, MN 55905 USA
| | - Ethan Grund
- grid.66875.3a0000 0004 0459 167XDepartment of Neurology, Mayo Clinic, Rochester, MN 55905 USA ,grid.66875.3a0000 0004 0459 167XMayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine and Mayo Clinic Medical Scientist Training Program, MN 55905 Rochester, USA
| | - Kenneth David
- grid.418935.20000 0004 0436 053XConcordia College, Moorhead, MN USA
| | - Renee K. Johnson
- grid.66875.3a0000 0004 0459 167XDepartment of Neurology, Mayo Clinic, Rochester, MN 55905 USA
| | - Charles L. Howe
- grid.66875.3a0000 0004 0459 167XDepartment of Neurology, Mayo Clinic, Rochester, MN 55905 USA ,grid.66875.3a0000 0004 0459 167XDivision of Experimental Neurology, Mayo Clinic, Rochester, MN 55905 USA ,grid.66875.3a0000 0004 0459 167XCenter for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
29
|
Mander BA, Dave A, Lui KK, Sprecher KE, Berisha D, Chappel-Farley MG, Chen IY, Riedner BA, Heston M, Suridjan I, Kollmorgen G, Zetterberg H, Blennow K, Carlsson CM, Okonkwo OC, Asthana S, Johnson SC, Bendlin BB, Benca RM. Inflammation, tau pathology, and synaptic integrity associated with sleep spindles and memory prior to β-amyloid positivity. Sleep 2022; 45:zsac135. [PMID: 35670275 PMCID: PMC9758508 DOI: 10.1093/sleep/zsac135] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 05/17/2022] [Indexed: 01/25/2023] Open
Abstract
STUDY OBJECTIVES Fast frequency sleep spindles are reduced in aging and Alzheimer's disease (AD), but the mechanisms and functional relevance of these deficits remain unclear. The study objective was to identify AD biomarkers associated with fast sleep spindle deficits in cognitively unimpaired older adults at risk for AD. METHODS Fifty-eight cognitively unimpaired, β-amyloid-negative, older adults (mean ± SD; 61.4 ± 6.3 years, 38 female) enriched with parental history of AD (77.6%) and apolipoprotein E (APOE) ε4 positivity (25.9%) completed the study. Cerebrospinal fluid (CSF) biomarkers of central nervous system inflammation, β-amyloid and tau proteins, and neurodegeneration were combined with polysomnography (PSG) using high-density electroencephalography and assessment of overnight memory retention. Parallelized serial mediation models were used to assess indirect effects of age on fast frequency (13 to <16Hz) sleep spindle measures through these AD biomarkers. RESULTS Glial activation was associated with prefrontal fast frequency sleep spindle expression deficits. While adjusting for sex, APOE ε4 genotype, apnea-hypopnea index, and time between CSF sampling and sleep study, serial mediation models detected indirect effects of age on fast sleep spindle expression through microglial activation markers and then tau phosphorylation and synaptic degeneration markers. Sleep spindle expression at these electrodes was also associated with overnight memory retention in multiple regression models adjusting for covariates. CONCLUSIONS These findings point toward microglia dysfunction as associated with tau phosphorylation, synaptic loss, sleep spindle deficits, and memory impairment even prior to β-amyloid positivity, thus offering a promising candidate therapeutic target to arrest cognitive decline associated with aging and AD.
Collapse
Affiliation(s)
- Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
- Department of Cognitive Sciences, University of California, Irvine, CA, USA
| | - Abhishek Dave
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Department of Cognitive Sciences, University of California, Irvine, CA, USA
| | - Kitty K Lui
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- San Diego State University/University of California San Diego, Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Katherine E Sprecher
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Destiny Berisha
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Miranda G Chappel-Farley
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Ivy Y Chen
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Brady A Riedner
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Margo Heston
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital
, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital
, Mölndal, Sweden
| | - Cynthia M Carlsson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA
| | - Ruth M Benca
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Psychiatry and Behavioral Medicine, Wake Forest University, Winston-Salem, NC, USA
| |
Collapse
|
30
|
Juutinen L, Ahinko K, Tinkanen H, Rosti-Otajärvi E, Sumelahti ML. Menopausal symptoms and hormone therapy in women with multiple sclerosis: A baseline-controlled study. Mult Scler Relat Disord 2022; 67:104098. [PMID: 35994896 DOI: 10.1016/j.msard.2022.104098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/20/2022] [Accepted: 08/07/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Depression, sleep disturbances, and cognitive difficulties impair the quality of life in people with multiple sclerosis (MS). Similar symptoms are also frequent during the menopausal transition. In clinical practice, it is important to consider the multifactorial causes of these overlapping symptoms and the potential benefits of menopausal hormone therapy (MHT). The objective of this study was to evaluate vasomotor symptoms (VMS), mood, sleep, and cognition of menopausal women with and without MS at baseline and during one year of MHT. METHODS In this prospective baseline-controlled study, peri- and early postmenopausal participants with (n=14) and without (n=13) MS received MHT containing 1 or 2 mg of estradiol and cyclical 10 mg dydrogesterone for one year. VMS frequency, depressive symptoms (measured by Beck Depression Inventory), insomnia severity (Insomnia Severity Index), and cognitive performance (Paced Auditory Serial Addition Test; PASAT, Symbol Digit Modalities Test; SDMT) were evaluated at baseline and at 3 and 12 months of treatment. Differences in the outcome measures between groups at baseline were assessed using the Mann-Whitney U test. Changes during follow-up compared to baseline within groups were evaluated by Wilcoxon Signed Ranks Test. P < 0.05 was considered for statistical significance. MS activity was monitored by clinical assessment and brain MRI at baseline and at 12 months. RESULTS Depressive symptoms were more common in MS group, while vasomotor and insomnia symptoms were equally common. During follow-up with MHT, VMS frequency decreased in both groups. Depressive symptoms decreased at 3 months (p = 0.031 with MS; p = 0.024 without MS) and the reduction was sustained at 12 months (p = 0.017; p = 0.042, respectively). Alleviation in insomnia symptoms was seen in participants without MS at 3 months (p = 0.029) and in those participants with MS suffering insomnia at baseline (p = 0.016 at 3 months; p = 0.047 at 12 months). Both groups improved their performance in PASAT, but no significant change was observed in SDMT. MS activity at baseline was mainly stable, and no increase in activity was detected during MHT. CONCLUSION Improvements in vasomotor, depressive, and insomnia symptoms observed during one year of MHT are encouraging and suggest that larger placebo-controlled studies of MHT in women with MS are warranted. Cognitive implications were inconclusive because the findings in PASAT likely result from practice effect. MHT did not show any adverse effect on MS activity and increasing safety data will hopefully facilitate patient recruitment for future studies.
Collapse
Affiliation(s)
- Laura Juutinen
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Building, Arvo Ylpön katu 34, 33520 Tampere, Finland; Department of Neurosciences and Rehabilitation, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland.
| | - Katja Ahinko
- Department of Obstetrics and Gynecology, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland
| | - Helena Tinkanen
- Department of Obstetrics and Gynecology, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland
| | - Eija Rosti-Otajärvi
- Department of Neurosciences and Rehabilitation, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland; Department of Rehabilitation and psychosocial support, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland
| | - Marja-Liisa Sumelahti
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Building, Arvo Ylpön katu 34, 33520 Tampere, Finland
| |
Collapse
|
31
|
Ford CC, Cohen JA, Goodman AD, Lindsey JW, Lisak RP, Luzzio C, Pruitt A, Rose J, Rus H, Wolinsky JS, Kadosh SE, Bernstein-Hanlon E, Stark Y, Alexander JK. Early versus delayed treatment with glatiramer acetate: Analysis of up to 27 years of continuous follow-up in a US open-label extension study. Mult Scler 2022; 28:1729-1743. [PMID: 35768939 PMCID: PMC9442630 DOI: 10.1177/13524585221094239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background: Glatiramer acetate (GA) is US-approved for relapsing multiple sclerosis. Objectives: To describe GA long-term clinical profile. To compare effectiveness of early start (ES) versus delayed start (DS; up to 3 years) with GA. Methods: Phase 3 trial participants entered a randomized placebo-controlled period then an open-label extension (OLE) with GA. Results: Overall, 208 out of 251 (82.9%) randomized participants entered the OLE; 24 out of 101 (23.8%, ES) and 28 out of 107 (26.2%, DS) participants completed the OLE. Median GA treatment was 9.8 (0.1–26.3) years. Annualized change in Expanded Disability Status Scale (EDSS) score was lower with ES versus DS (p = 0.0858: full study; p = 0.002; Year 5). Participants with improved/stable EDSS was consistently higher with ES versus DS: 40.3% versus 31.6% (p = 0.1590; full study); 70.8% versus 55.6% (p = 0.015; Year 5). ES prolonged time-to-6-month confirmed disease worsening (CDW) versus DS (9.8 vs 6.7 years), time-to-12-month CDW (18.9 vs 11.6 years), and significantly reduced time-to-second-6-month CDW (p = 0.0441). No new safety concerns arose. Conclusion: GA long-term treatment maintained clinical benefit with a similar safety profile to phase 3 results; a key limitation was that only 25% of participants completed the OLE. Early initiation of GA had sustained benefits versus delayed treatment.
Collapse
Affiliation(s)
- Corey C Ford
- Department of Neurology, University of New Mexico Health Sciences Center, The University of New Mexico, Albuquerque, NM, USA
| | - Jeffrey A Cohen
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Andrew D Goodman
- Department of Neurology, University of Rochester, Rochester, NY, USA
| | - John W Lindsey
- Department of Neurology, University of Texas Health Science Center at Houston (UTHouston), Houston, TX, USA
| | - Robert P Lisak
- Department of Neurology and Department of Biochemistry, Microbiology and Immunology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Christopher Luzzio
- Departments of Neurology and Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Amy Pruitt
- Department of Neurology, University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | - John Rose
- Imaging and Neuroscience Center, School of Medicine, The University of Utah, Salt Lake City, UT, USA
| | - Horea Rus
- Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Jerry S Wolinsky
- Department of Neurology, University of Texas Health Science Center at Houston (UTHouston), Houston, TX, USA
| | - Shaul E Kadosh
- Innovative Research and Development, Teva Pharmaceuticals, Netanya, Israel
| | | | - Yafit Stark
- Global Clinical Development, Teva Pharmaceuticals, Netanya, Israel
| | - Jessica K Alexander
- Global Medical Affairs, Teva Pharmaceuticals, West Chester, PA, USA/Jazz Pharmaceuticals, Palo Alto, CA, USA
| |
Collapse
|
32
|
Jennings G, Monaghan A, Xue F, Duggan E, Romero-Ortuño R. Comprehensive Clinical Characterisation of Brain Fog in Adults Reporting Long COVID Symptoms. J Clin Med 2022; 11:3440. [PMID: 35743516 PMCID: PMC9224578 DOI: 10.3390/jcm11123440] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 02/05/2023] Open
Abstract
(1) Introduction: A subset of individuals experiencing long COVID symptoms are affected by 'brain fog', a lay term that often refers to general cognitive dysfunction but one that is still poorly characterised. In this study, a comprehensive clinical characterisation of self-reported brain fog was conducted vis-à-vis other long COVID symptoms and parameters of mental, cognitive, and physical health. (2) Methodology: Adult participants reporting long COVID symptoms were recruited from hospital clinics and as self-referrals. Participants completed a battery of questionnaires and clinical assessments, including COVID-19 history, symptomatology, self-reported scales (Chalder Fatigue Scale [CFQ], Center for Epidemiological Studies Depression Scale, and Impact of Events Scale-Revised), computer-based cognitive assessments (simple response time and choice reaction time tasks), physical performance tests (gait velocity and muscle strength assessments), and an orthostatic active stand test. A systematic comparison between participants with and without self-reported brain fog was conducted, and a backwards binary logistic regression model was computed to identify the strongest independent associations with brain fog. This was complemented by an automatic cluster analysis to rank the importance of associations. Finally, a structural equation model was postulated with a causal model of key symptomatic indicators and functional consequences of brain fog as a latent variable. (3) Results: Of 108 participants assessed, brain fog was a self-reported symptom in 71 (65.7%) participants. Those with brain fog were at a longer point in time since COVID-19 onset and reported longer duration of low activity during the acute illness. When assessed, those with brain fog had higher frequencies of subjective memory impairment, word-finding difficulties, dizziness, myalgia, arthralgia, hyperhidrosis, cough, voice weakness, throat pain, visual and hearing problems, dysosmia, paraesthesia, chest pain, skin rashes, and hair loss; mean scores in fatigue, depression, and post-traumatic stress scales were higher; performance in both computer-based cognitive tasks was poorer; and measured gait speed and grip strength were lower. The logistic regression suggested that the best independent associations with brain fog were memory impairment, CFQ, and myalgia. The cluster analysis suggested that the most important associations with brain fog were CFQ, dizziness, myalgia, reduced gait speed, word-finding difficulties, reduced grip strength, and memory impairment. The SEM was consistent with key indicators of brain fog being CFQ, dizziness, myalgia, word-finding difficulties, and memory impairment; and reduced grip strength, gait speed, and cognitive response times its functional consequences. (4) Conclusions: The findings indicate that self-reported brain fog in long COVID is a recognisable symptom cluster primarily characterised by fatigue, dizziness, myalgia, word-finding difficulties, and memory impairment and has adverse psychological and psychomotor correlates. In long COVID, brain fog should be regarded as a wide-ranging symptom and addressed holistically with medical, psychological, and rehabilitative supports as guided by individual needs.
Collapse
Affiliation(s)
- Glenn Jennings
- School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland; (A.M.); (F.X.); (E.D.); (R.R.-O.)
| | - Ann Monaghan
- School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland; (A.M.); (F.X.); (E.D.); (R.R.-O.)
| | - Feng Xue
- School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland; (A.M.); (F.X.); (E.D.); (R.R.-O.)
| | - Eoin Duggan
- School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland; (A.M.); (F.X.); (E.D.); (R.R.-O.)
- Mercer’s Institute for Successful Ageing, St. James’s Hospital, D08 N80H Dublin, Ireland
| | - Román Romero-Ortuño
- School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland; (A.M.); (F.X.); (E.D.); (R.R.-O.)
- Mercer’s Institute for Successful Ageing, St. James’s Hospital, D08 N80H Dublin, Ireland
- Global Brain Health Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
| |
Collapse
|
33
|
Cognitive Decline in Older People with Multiple Sclerosis—A Narrative Review of the Literature. Geriatrics (Basel) 2022; 7:geriatrics7030061. [PMID: 35735766 PMCID: PMC9223056 DOI: 10.3390/geriatrics7030061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 12/04/2022] Open
Abstract
Several important questions regarding cognitive aging and dementia in older people with multiple sclerosis (PwMS) are the focus of this narrative review: Do older PwMS have worse cognitive decline compared to older people without MS? Can older PwMS develop dementia or other neurodegenerative diseases such as Alzheimer’s disease (AD) that may be accelerated due to MS? Are there any potential biomarkers that can help to determine the etiology of cognitive decline in older PwMS? What are the neural and cellular bases of cognitive aging and neurodegeneration in MS? Current evidence suggests that cognitive impairment in MS is distinguishable from that due to other neurodegenerative diseases, although older PwMS may present with accelerated cognitive decline. While dementia is prevalent in PwMS, there is currently no consensus on defining it. Cerebrospinal fluid and imaging biomarkers have the potential to identify disease processes linked to MS and other comorbidities—such as AD and vascular disease—in older PwMS, although more research is required. In conclusion, one should be aware that multiple underlying pathologies can coexist in older PwMS and cause cognitive decline. Future basic and clinical research will need to consider these complex factors to better understand the underlying pathophysiology, and to improve diagnostic accuracy.
Collapse
|
34
|
Keegan AP, Joshi U, Abdullah L, Paris D, Darcey T, Niedospial D, Davis LA, Crawford F, Mullan M. Characterization of immune profile in an aging multiple sclerosis clinic population. Mult Scler Relat Disord 2022; 63:103818. [DOI: 10.1016/j.msard.2022.103818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/19/2022] [Accepted: 04/21/2022] [Indexed: 11/25/2022]
|
35
|
Shu MJ, Li J, Zhu YC. Genetically predicted telomere length and multiple sclerosis. Mult Scler Relat Disord 2022; 60:103731. [PMID: 35339005 DOI: 10.1016/j.msard.2022.103731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/22/2022] [Accepted: 03/05/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Previous epidemiological studies have indicated a role for telomere length in multiple sclerosis (MS) severity and phenotype. However, these studies failed to establish the causality between telomere length and MS susceptibility. Hence, we performed two-sample Mendelian randomization (MR) analysis to explore the causal relationship between telomere length and MS susceptibility. METHODS We used data of genetic variants associated with leukocyte telomere length as instrumental variables (IVs), which was identified from the largest and latest genome-wide association study (GWAS) from UK Biobank (UKB) with 472,174 participants. Summary data of MS was obtained from the International Multiple Sclerosis Genetics Consortium. We performed two-sample MR analyses using the inverse-variance weighted method as the primary approach. Other MR approaches, including the MR-Egger, the inverse variance weighted (multiplicative random effects), weighted median, simple median, weighted mode-based methods, and Causal Analysis Using Summary Effect estimates (CAUSE), were also conducted to detect the result robustness. RESULTS The genetic liability to longer telomere length was associated with a higher risk of MS susceptibility (odds ratio [OR] per one-SD telomere length, 1.91; 95% confidence interval [CI], 1.48-2.47; P = 8.04 × 10-7). The results remained consistent across multiple sensitivity analyses. CONCLUSIONS Our study supports the causal relationship between longer telomere length and increased risk of MS susceptibility.
Collapse
Affiliation(s)
- Mei-Jun Shu
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, No.1 Shuaifuyuan, Wangfujing, Beijing 10073, China
| | - Jiarui Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 10073, China
| | - Yi-Cheng Zhu
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, No.1 Shuaifuyuan, Wangfujing, Beijing 10073, China.
| |
Collapse
|
36
|
das Neves SP, Serre-Miranda C, Sousa JC, Costa P, Sousa N, Cerqueira JJ, Marques F. Lipocalin-2 does not influence EAE clinical score but it increases inflammation in central nervous system. J Neuroimmunol 2022; 368:577872. [DOI: 10.1016/j.jneuroim.2022.577872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 11/26/2022]
|
37
|
Martinez-Rojas VA, Juarez-Hernandez LJ, Musio C. Ion channels and neuronal excitability in polyglutamine neurodegenerative diseases. Biomol Concepts 2022; 13:183-199. [DOI: 10.1515/bmc-2022-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Polyglutamine (polyQ) diseases are a family composed of nine neurodegenerative inherited disorders (NDDs) caused by pathological expansions of cytosine-adenine-guanine (CAG) trinucleotide repeats which encode a polyQ tract in the corresponding proteins. CAG polyQ repeat expansions produce neurodegeneration via multiple downstream mechanisms; among those the neuronal activity underlying the ion channels is affected directly by specific channelopathies or indirectly by secondary dysregulation. In both cases, the altered excitability underlies to gain- or loss-of-function pathological effects. Here we summarize the repertoire of ion channels in polyQ NDDs emphasizing the biophysical features of neuronal excitability and their pathogenic role. The aim of this review is to point out the value of a deeper understanding of those functional mechanisms and processes as crucial elements for the designing and targeting of novel therapeutic avenues.
Collapse
Affiliation(s)
- Vladimir A. Martinez-Rojas
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) , Via Sommarive 18 , 38123 Trento , Italy
| | - Leon J. Juarez-Hernandez
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) , Via Sommarive 18 , 38123 Trento , Italy
| | - Carlo Musio
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) , Via Sommarive 18 , 38123 Trento , Italy
| |
Collapse
|
38
|
Song YC, Liu CT, Lee HJ, Yen HR. Cordycepin prevents and ameliorates experimental autoimmune encephalomyelitis by inhibiting leukocyte infiltration and reducing neuroinflammation. Biochem Pharmacol 2022; 197:114918. [DOI: 10.1016/j.bcp.2022.114918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/30/2022]
|
39
|
Perdaens O, van Pesch V. Molecular Mechanisms of Immunosenescene and Inflammaging: Relevance to the Immunopathogenesis and Treatment of Multiple Sclerosis. Front Neurol 2022; 12:811518. [PMID: 35281989 PMCID: PMC8913495 DOI: 10.3389/fneur.2021.811518] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/27/2021] [Indexed: 12/18/2022] Open
Abstract
Aging is characterized, amongst other features, by a complex process of cellular senescence involving both innate and adaptive immunity, called immunosenescence and associated to inflammaging, a low-grade chronic inflammation. Both processes fuel each other and partially explain increasing incidence of cancers, infections, age-related autoimmunity, and vascular disease as well as a reduced response to vaccination. Multiple sclerosis (MS) is a lifelong disease, for which considerable progress in disease-modifying therapies (DMTs) and management has improved long-term survival. However, disability progression, increasing with age and disease duration, remains. Neurologists are now involved in caring for elderly MS patients, with increasing comorbidities. Aging of the immune system therefore has relevant implications for MS pathogenesis, response to DMTs and the risks mediated by these treatments. We propose to review current evidence regarding markers and molecular mechanisms of immunosenescence and their relevance to understanding MS pathogenesis. We will focus on age-related changes in the innate and adaptive immune system in MS and other auto-immune diseases, such as systemic lupus erythematosus and rheumatoid arthritis. The consequences of these immune changes on MS pathology, in interaction with the intrinsic aging process of central nervous system resident cells will be discussed. Finally, the impact of immunosenescence on disease evolution and on the safety and efficacy of current DMTs will be presented.
Collapse
Affiliation(s)
- Océane Perdaens
- Laboratory of Neurochemistry, Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Vincent van Pesch
- Laboratory of Neurochemistry, Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Neurology, Cliniques universitaires Saint-Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- *Correspondence: Vincent van Pesch
| |
Collapse
|
40
|
Montazeri M, Eskandari N, Mansouri R. Evaluation of the expressed miR-129 and miR-549a in patients with multiple sclerosis. Adv Biomed Res 2022; 10:48. [PMID: 35127575 PMCID: PMC8781915 DOI: 10.4103/abr.abr_268_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/25/2021] [Accepted: 05/29/2021] [Indexed: 11/08/2022] Open
Abstract
Background: The expression of microRNAs (miRNAs) as circulating biomarkers has been underlined in multiple sclerosis (MS) in the last decade. Due to the presence of a possible relationship between expressed miRNAs and heterogeneous appearances of the pathological processes in MS, the present study attempts to evaluate the expression of miR-129 and miR-549a in patients with MS in comparison with healthy control (HC) group. Materials and Methods: Peripheral blood mononuclear cells were separated from fifty patients with MS (subtypes including relapsing–remitting MS and secondary progressive MS) in the Kashani Hospital, Isfahan, Iran, and fifty people as HC group. After RNA extraction and complementary DNA synthesis, the expression of miR-129 and miR-549a was evaluated in patients with MS in comparison with the HC group using a quantitative real-time polymerase chain reaction assay. The data were analyzed using the Kolmogorov–Smirnov and Mann–Whitney tests. Spearman's correlation coefficient was used to examine the relationship between miR-129 and miR-549a with age. Results: The results showed that the expression of miR-129 and miR-549a was not significant in patients with MS in comparison with the HC group. Furthermore, the relationship between such miRNAs and age and gender was not significant. Conclusion: We suggest the expression of miR-129 and miR-549a as circulating miRNAs in peripheral blood mononuclear cells could not be considered a biomarker for diagnosis and Para clinical.
Collapse
Affiliation(s)
- Mina Montazeri
- Department of Immunology, International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nahid Eskandari
- Department of Immunology, School of Medicine, Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Mansouri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
41
|
Role and therapeutic implications of protein glycosylation in neuroinflammation. Trends Mol Med 2022; 28:270-289. [PMID: 35120836 DOI: 10.1016/j.molmed.2022.01.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 12/13/2022]
Abstract
The importance of glycosylation (post-translational attachment of glycan residues to proteins) in the context of neuroinflammation is only now beginning to be understood. Although the glycome is challenging to investigate due to its complexity, this field is gaining interest because of the emergence of novel analytical methods. These investigations offer the possibility of further understanding the molecular signature of disorders with underlying neuroinflammatory cascades. In this review, we portray the clinically relevant trends in glyconeurobiology and suggest glyco-related paths that could be targeted therapeutically to decrease neuroinflammation. A combinatorial insight from glycobiology and neurology can be harnessed to better understand neuroinflammatory-related conditions to identify relevant molecular targets.
Collapse
|
42
|
Neuroinflammation Is Associated with GFAP and sTREM2 Levels in Multiple Sclerosis. Biomolecules 2022; 12:biom12020222. [PMID: 35204724 PMCID: PMC8961656 DOI: 10.3390/biom12020222] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/16/2022] [Accepted: 01/22/2022] [Indexed: 01/22/2023] Open
Abstract
Background: Astrocytes and microglia play an important role in the inflammatory process of multiple sclerosis (MS). We investigated the associations between the cerebrospinal fluid (CSF) levels of glial fibrillary acid protein (GFAP) and soluble triggering receptors expressed on myeloid cells-2 (sTREM-2), inflammatory molecules, and clinical characteristics in a group of patients with relapsing-remitting MS (RRMS). Methods: Fifty-one RRMS patients participated in the study. Clinical evaluation and CSF collection were performed at the time of diagnosis. The CSF levels of GFAP, sTREM-2, and of a large set of inflammatory and anti-inflammatory molecules were determined. MRI structural measures (cortical thickness, T2 lesion load, cerebellar volume) were examined. Results: The CSF levels of GFAP and sTREM-2 showed significant correlations with inflammatory cytokines IL-8, G-CSF, and IL-5. Both GFAP and sTREM-2 CSF levels positively correlated with age at diagnosis. GFAP was also higher in male MS patients, and was associated with an increased risk of MS progression, as evidenced by higher BREMS at the onset. Finally, a negative association was found between GFAP CSF levels and cerebellar volume in RRMS at diagnosis. Conclusions: GFAP and sTREM-2 represent suitable biomarkers of central inflammation in MS. Our results suggest that enhanced CSF expression of GFAP may characterize patients with a higher risk of progression.
Collapse
|
43
|
Variability of Objective Gait Measures across the Expanded Disability Status Scale in People Living with Multiple Sclerosis: a cross-sectional retrospective analysis. Mult Scler Relat Disord 2022; 59:103645. [DOI: 10.1016/j.msard.2022.103645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 01/14/2022] [Accepted: 01/29/2022] [Indexed: 11/23/2022]
|
44
|
Frailty and falls in people living with multiple sclerosis. Arch Phys Med Rehabil 2021; 103:952-957. [PMID: 34838587 DOI: 10.1016/j.apmr.2021.10.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/29/2021] [Accepted: 10/28/2021] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To explore the association between frailty and history of falls in people living with multiple sclerosis (MS). DESIGN Secondary analysis. SETTING University research laboratories in the United States and Israel. PARTICIPANTS 118 people with relapsing-remitting MS [age=48.9 years (SD=10.0); 74.6% female; expanded disability status scale (EDSS) range=1.0-6.0] were studied in this cross-sectional analysis. INTERVENTION Not applicable. MAIN OUTCOMES A frailty index was calculated from 40 health deficits by following standard validated procedures. The number of falls (12-month history) was recorded. RESULTS Overall, 33.9%, 29.7%, and 36.4% of participants were classified as non-frail, moderately frail, and severely frail, respectively. The frailty index was significantly correlated (ρ=0.37, p<0.001) with higher scores on the EDSS. In univariable negative binomial regression analysis, the frailty index was associated with a higher number of falls (IRR=3.33, 95%CI[1.85-5.99], p<0.001). After adjustment for age, gender and EDSS, frailty remained strongly associated with history of falls (IRR=2.78, 95%CI[1.51-5.10], p=0.001). CONCLUSION The current study identifies a significant relationship between frailty and history of falls in MS, independent of age, gender, and disease severity. These findings support the notion that frailty is a syndrome related to, but independent of, disability in MS.
Collapse
|
45
|
Zhao J, He Z, Wang J. MicroRNA-124: A Key Player in Microglia-Mediated Inflammation in Neurological Diseases. Front Cell Neurosci 2021; 15:771898. [PMID: 34795564 PMCID: PMC8593194 DOI: 10.3389/fncel.2021.771898] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/14/2021] [Indexed: 01/07/2023] Open
Abstract
Neurological disorders are mainly characterized by progressive neuron loss and neurological deterioration, which cause human disability and death. However, many types of neurological disorders have similar pathological mechanisms, including the neuroinflammatory response. Various microRNAs (miRs), such as miR-21, miR-124, miR-146a, and miR-132 were recently shown to affect a broad spectrum of biological functions in the central nervous system (CNS). Microglia are innate immune cells with important roles in the physiological and pathological activities of the CNS. Recently, abnormal expression of miR-124 was shown to be associated with the occurrence and development of various diseases in CNS via regulating microglia function. In addition, miR-124 is a promising biomarker and therapeutic target. Studies on the role of miR-124 in regulating microglia function involved in pathogenesis of neurological disorders at different stages will provide new ideas for the use of miR-124 as a therapeutic target for different CNS diseases.
Collapse
Affiliation(s)
- Jiuhan Zhao
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhenwei He
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jialu Wang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
46
|
Shao Y, Chen C, Zhu T, Sun Z, Li S, Gong L, Dong X, Shen W, Zeng L, Xie Y, Jiang P. TRPM2 contributes to neuroinflammation and cognitive deficits in a cuprizone-induced multiple sclerosis model via NLRP3 inflammasome. Neurobiol Dis 2021; 160:105534. [PMID: 34673151 DOI: 10.1016/j.nbd.2021.105534] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/23/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a disease of the central nervous system (CNS) that is characterized by demyelination, axonal injury and neurological deterioration. Few medications are available for progressive MS, which is associated with neuroinflammation confined to the CNS compartment. Transient receptor potential melastatin 2 (TRPM2) is a calcium-permeable, non-selective cation channel that plays pathological roles in a wide range of neuroinflammatory diseases; however, the underlying molecular mechanisms of TRPM2 remain elusive. Here, we established a cuprizone model that presents hallmark MS pathologies to investigate the role of TRPM2 in progressive MS. We demonstrated that genetic deletion of TRPM2 yields protection from the cuprizone-induced demyelination, synapse loss, microglial activation, NLRP3 inflammasome activation and proinflammatory cytokines production and ultimately leads to an improvement in cognitive decline. Furthermore, we showed that the pharmacological inhibition of NLRP3 ameliorated the demyelination, neuroinflammation and cognitive impairment in the model with no additive effects on the TRPM2 KO mice. Taken together, these results indicated that TRPM2 plays important roles in regulating neuroinflammation in progressive MS via NLRP3 inflammasome, and the results shed light on TRPM2's potential role as a therapeutic target for MS.
Collapse
Affiliation(s)
- Yu Shao
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Wenling First People's Hospital, Wenling 317500, China
| | - Chen Chen
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China
| | - Tao Zhu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China
| | - Zengxian Sun
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Lishui Central Hospital, Lishui 323000, China
| | - Shufen Li
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Lishui Central Hospital, Lishui 323000, China
| | - Lifen Gong
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China
| | - Xinyan Dong
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China
| | - Weida Shen
- Department of Pharmacy, Zhejiang University City College School of Medicine, Hangzhou 310015, China
| | - Linghui Zeng
- Department of Pharmacy, Zhejiang University City College School of Medicine, Hangzhou 310015, China
| | - Yicheng Xie
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China.
| | - Peifang Jiang
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China.
| |
Collapse
|
47
|
Satheesh NJ, Salloum-Asfar S, Abdulla SA. The Potential Role of COVID-19 in the Pathogenesis of Multiple Sclerosis-A Preliminary Report. Viruses 2021; 13:2091. [PMID: 34696521 PMCID: PMC8540806 DOI: 10.3390/v13102091] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/17/2022] Open
Abstract
Coronavirus 2019 (COVID-19) is an infectious respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that mainly affects the lungs. COVID-19 symptoms include the presence of fevers, dry coughs, fatigue, sore throat, headaches, diarrhea, and a loss of taste or smell. However, it is understood that SARS-CoV-2 is neurotoxic and neuro-invasive and could enter the central nervous system (CNS) via the hematogenous route or via the peripheral nerve route and causes encephalitis, encephalopathy, and acute disseminated encephalomyelitis (ADEM) in COVID-19 patients. This review discusses the possibility of SARS-CoV-2-mediated Multiple Sclerosis (MS) development in the future, comparable to the surge in Parkinson's disease cases following the Spanish Flu in 1918. Moreover, the SARS-CoV-2 infection is associated with a cytokine storm. This review highlights the impact of these modulated cytokines on glial cell interactions within the CNS and their role in potentially prompting MS development as a secondary disease by SARS-CoV-2. SARS-CoV-2 is neurotropic and could interfere with various functions of neurons leading to MS development. The influence of neuroinflammation, microglia phagocytotic capabilities, as well as hypoxia-mediated mitochondrial dysfunction and neurodegeneration, are mechanisms that may ultimately trigger MS development.
Collapse
Affiliation(s)
| | - Salam Salloum-Asfar
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 34110, Qatar;
| | - Sara A. Abdulla
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 34110, Qatar;
| |
Collapse
|
48
|
Souissi A, Mrabet S, Nasri A, Ben Djebara M, Gargouri A, Kacem I, Gouider R. Clinical predictors of disease progression in a cohort of Tunisian progressive Multiple Sclerosis. Mult Scler Relat Disord 2021; 56:103232. [PMID: 34619488 DOI: 10.1016/j.msard.2021.103232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/11/2021] [Accepted: 08/29/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Knowledge about progressive Multiple Sclerosis (MS) is mainly based on Caucasian studies. In our North-African context, MS exhibits particular characteristics that are mainly related to a more severe phenotype. Given the limited data available, there is an imminent need to characterize progressive MS in our latitudes. OBJECTIVE To describe the specificities of progressive MS and identify the inherent clinical predictors of disability accrual with a Tunisian cohort. METHODS A retrospective, hospital-based study was conducted in the department of neurology of Razi hospital. Patients, who had been diagnosed with MS, were divided into relapsing MS (RRMS), secondary progressive MS (SPMS) and primary progressive MS (PPMS). Epidemiological, clinical and paraclinical data were compared among the three groups. RESULTS Of the 504 patients, a progressive MS was described among 115 patients. This percentage of (22.8%) is divided into 13.9% SPMS and 8.9% PPMS. During the first clinical attack, motor symptoms have revealed to be predominant during PPMS (91.1%). For SPMS onset, the median time was 10 years, and was significantly delayed for patients with visual onset or full recovery from the first relapse. Patients with progressive MS exhibited a more rapid disability accumulation. CONCLUSION Compared to Caucasians, Tunisians exhibited a faster rate of conversion to SPMS. According to our natural progressive MS history, early clinical features are predictors of MS disability accrual.
Collapse
Affiliation(s)
- A Souissi
- Department of Neurology, LR 18SP03, Clinical Investigation Centre Neurosciences and Mental Health, Razi Universitary Hospital, Manouba, Tunis, Tunisia
| | - S Mrabet
- Department of Neurology, LR 18SP03, Clinical Investigation Centre Neurosciences and Mental Health, Razi Universitary Hospital, Manouba, Tunis, Tunisia; Faculty of medicine, University Tunis El Manar, 1007, Tunis, Tunisia
| | - A Nasri
- Department of Neurology, LR 18SP03, Clinical Investigation Centre Neurosciences and Mental Health, Razi Universitary Hospital, Manouba, Tunis, Tunisia; Faculty of medicine, University Tunis El Manar, 1007, Tunis, Tunisia
| | - M Ben Djebara
- Department of Neurology, LR 18SP03, Clinical Investigation Centre Neurosciences and Mental Health, Razi Universitary Hospital, Manouba, Tunis, Tunisia; Faculty of medicine, University Tunis El Manar, 1007, Tunis, Tunisia
| | - A Gargouri
- Department of Neurology, LR 18SP03, Clinical Investigation Centre Neurosciences and Mental Health, Razi Universitary Hospital, Manouba, Tunis, Tunisia; Faculty of medicine, University Tunis El Manar, 1007, Tunis, Tunisia
| | - I Kacem
- Department of Neurology, LR 18SP03, Clinical Investigation Centre Neurosciences and Mental Health, Razi Universitary Hospital, Manouba, Tunis, Tunisia; Faculty of medicine, University Tunis El Manar, 1007, Tunis, Tunisia
| | - R Gouider
- Department of Neurology, LR 18SP03, Clinical Investigation Centre Neurosciences and Mental Health, Razi Universitary Hospital, Manouba, Tunis, Tunisia; Faculty of medicine, University Tunis El Manar, 1007, Tunis, Tunisia.
| |
Collapse
|
49
|
Chan CK, Tian F, Pimentel Maldonado D, Mowry EM, Fitzgerald KC. Depression in multiple sclerosis across the adult lifespan. Mult Scler 2021; 27:1771-1780. [PMID: 33307963 PMCID: PMC8200363 DOI: 10.1177/1352458520979304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The objective of this study is to examine the burden of depressive symptoms across the adult age span in people with multiple sclerosis (MS) and test if the relationship between depressive symptoms and MS characteristics vary across age groups. METHODS In analyses of the MS Partners Advancing Technology and Health Solutions (MS PATHS) network of adults with MS, we compared the prevalence of depression in MS PATHS with non-MS controls across age and evaluated for effect modification by age in the association between depressive symptoms and clinical and neuroperformance measures via multivariable-adjusted regression models. RESULTS In total, 13,821 individuals with MS were included. The prevalence of depression was higher in MS versus non-MS controls, but was similar between men/women across age. The association between depression and processing speed (PST; p for interaction = 0.009) or walking speed (p for interaction = 0.04) varied by age. For example, younger depressed individuals had 0.45 standard deviation (SD) (95% confidence interval (CI) = -0.62, -0.29) worse PST Z-scores versus non-depressed younger participants, whereas older depressed individuals had 0.20 SD (95% CI = -0.32, -0.08) worse PST Z-scores versus non-depressed older participants. CONCLUSION Depressive symptoms and age should be considered when interpreting measures of walking speed and cognitive function; these findings may have implications for analyses of neuroperformance change.
Collapse
Affiliation(s)
- Carol K Chan
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Fan Tian
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Ellen M Mowry
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kathryn C Fitzgerald
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
50
|
Stampanoni Bassi M, Gilio L, Iezzi E, Moscatelli A, Pekmezovic T, Drulovic J, Furlan R, Finardi A, Mandolesi G, Musella A, Galifi G, Fantozzi R, Bellantonio P, Storto M, Centonze D, Buttari F. Age at Disease Onset Associates With Oxidative Stress, Neuroinflammation, and Impaired Synaptic Plasticity in Relapsing-Remitting Multiple Sclerosis. Front Aging Neurosci 2021; 13:694651. [PMID: 34566620 PMCID: PMC8461180 DOI: 10.3389/fnagi.2021.694651] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/12/2021] [Indexed: 12/02/2022] Open
Abstract
Age at onset is the main risk factor for disease progression in patients with relapsing-remitting multiple sclerosis (RR-MS). In this cross-sectional study, we explored whether older age is associated with specific disease features involved in the progression independent of relapse activity (PIRA). In 266 patients with RR-MS, the associations between age at onset, clinical characteristics, cerebrospinal fluid (CSF) levels of lactate, and that of several inflammatory molecules were analyzed. The long-term potentiation (LTP)-like plasticity was studied using transcranial magnetic stimulation (TMS). Older age was associated with a reduced prevalence of both clinical and radiological focal inflammatory disease activity. Older patients showed also increased CSF levels of lactate and that of the pro-inflammatory molecules monocyte chemoattractant protein 1 (MCP-1)/CCL2, macrophage inflammatory protein 1-alpha (MIP-1α)/CCL3, and interleukin (IL)-8. Finally, TMS evidenced a negative correlation between age and LTP-like plasticity. In newly diagnosed RR-MS, older age at onset is associated with reduced acute disease activity, increased oxidative stress, enhanced central inflammation, and altered synaptic plasticity. Independently of their age, patients with multiple sclerosis (MS) showing similar clinical, immunological, and neurophysiological characteristics may represent ideal candidates for early treatments effective against PIRA.
Collapse
Affiliation(s)
| | - Luana Gilio
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Ennio Iezzi
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Alessandro Moscatelli
- Department of Systems Medicine, Tor Vergata University, Rome, Italy.,Laboratory of Neuromotor Physiology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Tatjana Pekmezovic
- Institute of Epidemiology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Drulovic
- Clinic of Neurology, Clinical Center of Serbia, Belgrade, Serbia
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
| | - Giovanni Galifi
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Roberta Fantozzi
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Paolo Bellantonio
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Marianna Storto
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Diego Centonze
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy.,Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|