1
|
Zhou M, Xu Z, Zhong H, Ning G, Feng S. Spinal cord injury and inflammatory mediators: Role in "fire barrier" formation and potential for neural regeneration. Neural Regen Res 2026; 21:923-937. [PMID: 39995083 DOI: 10.4103/nrr.nrr-d-24-00792] [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: 07/18/2024] [Accepted: 01/07/2025] [Indexed: 02/26/2025] Open
Abstract
Traumatic spinal cord injury result in considerable and lasting functional impairments, triggering complex inflammatory and pathological events. Spinal cord scars, often metaphorically referred to as "fire barriers," aim to control the spread of neuroinflammation during the acute phase but later hinder axon regeneration in later stages. Recent studies have enhanced our understanding of immunomodulation, revealing that injury-associated inflammation involves various cell types and molecules with positive and negative effects. This review employs bibliometric analysis to examine the literature on inflammatory mediators in spinal cord injury, highlighting recent research and providing a comprehensive overview of the current state of research and the latest advances in studies on neuroinflammation related to spinal cord injury. We summarize the immune and inflammatory responses at different stages of spinal cord injury, offering crucial insights for future research. Additionally, we review repair strategies based on inflammatory mediators for the injured spinal cord. Finally, this review discusses the current status and future directions of translational research focused on immune-targeting strategies, including pharmaceuticals, biomedical engineering, and gene therapy. The development of a combined, precise, and multitemporal strategy for the repair of injured spinal cords represents a promising direction for future research.
Collapse
Affiliation(s)
- Mi Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Zhengyu Xu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Hao Zhong
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Guangzhi Ning
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Shandong University Center for Orthopedics, Advanced Medical Research Institute, Shandong University, Jinan, Shandong Province, China
- Orthopedic Research Center of Shandong University & Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
2
|
Ganz T, Ben-Hur T. Illusion of inactivity: Revisiting progressive multiple sclerosis treatment paradigms. Neural Regen Res 2026; 21:1134-1135. [PMID: 40145948 DOI: 10.4103/nrr.nrr-d-24-01308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/10/2025] [Indexed: 03/28/2025] Open
Affiliation(s)
- Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel (Ganz T, Ben-Hur T)
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel (Ganz T, Ben-Hur T)
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel (Ganz T, Ben-Hur T)
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel (Ganz T, Ben-Hur T)
| |
Collapse
|
3
|
Ghamangiz S, Jafari A, Maleki-Kakelar H, Azimi H, Mazloomi E. Reprogram to heal: Macrophage phenotypes as living therapeutics. Life Sci 2025; 371:123601. [PMID: 40189197 DOI: 10.1016/j.lfs.2025.123601] [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: 01/15/2025] [Revised: 03/15/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
Macrophages represent a crucial cell type within the immune system, exhibiting significant adaptability that allows for the transformation into various phenotypes in response to their surrounding environment. This review investigates the characteristics of various macrophage phenotypes and their functional roles in disease pathogenesis and resolution. The M1 phenotype, recognized for its inflammatory attributes, plays a pivotal role in combating infections and tumors; however, it may also contribute to tissue injury, persistent inflammation, and the pathogenesis of autoimmune and inflammatory diseases. Conversely, the M2 phenotype is associated with anti-inflammatory activities and tissue repair processes. But this is not the end of the story and researches illustrated novel phenotypes that may provide new approaches and therapeutic opportunities. Recent progress in characterizing distinct macrophage phenotypes has enabled the development of innovative therapeutic strategies for chronic inflammatory conditions, autoimmune disorders, and cancers. This review underscores the critical role of macrophage polarization, illustrating how various stimuli can influence macrophage fate and modify their responses. Additionally, it explores the implications of macrophage plasticity on disease progression and treatment efficacy. A comprehensive understanding of these dynamics is essential for the advancement of targeted immunotherapies, which possess the potential to transform treatment strategies for a variety of medical conditions.
Collapse
Affiliation(s)
- Sheyda Ghamangiz
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Abbas Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Maleki-Kakelar
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Azimi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ebrahim Mazloomi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
4
|
Abulaban AA, Al-Kuraishy HM, Al-Gareeb AI, Albuhadily AK, Shokr MM, Alexiou A, Papadakis M, Batiha GES. The janus face of astrocytes in multiple sclerosis: Balancing protection and pathology. Brain Res Bull 2025; 226:111356. [PMID: 40288545 DOI: 10.1016/j.brainresbull.2025.111356] [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: 02/24/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder characterized by demyelination and neurodegeneration in the central nervous system (CNS), predominantly affecting young adults with a notable female predominance. While the pathogenesis of MS involves complex interactions between peripheral immune cells and CNS glia, astrocytes-the most abundant glial cells-play a dual role in disease progression. Traditionally classified into pro-inflammatory A1 and neuroprotective A2 phenotypes, recent single-cell and spatial transcriptomics reveal that human astrocytes exhibit a continuum of states beyond this binary paradigm. In MS, reactive astrocytes contribute to neurotoxicity by disrupting the blood-brain barrier (BBB), promoting glutamate excitotoxicity, and presenting antigens to autoreactive T cells. Conversely, they also support repair through neurotrophic factor release (e.g., BDNF, CNTF) and remyelination. Emerging therapies like dimethyl fumarate (DMF) and fingolimod modulate astrocyte reactivity, targeting oxidative stress and sphingosine-1-phosphate receptors to mitigate neuroinflammation. However, challenges persist in translating murine A1/A2 concepts to human MS, as human astrocytes display heterogeneous, context-dependent responses influenced by regional microenvironments and disease stages. Advanced techniques, including spatial multi-omics, highlight astrocyte-microglia crosstalk and metabolic reprogramming as key drivers of MS pathology. This review synthesizes current evidence on astrocyte heterogeneity, their Janus-faced roles in MS, and the therapeutic potential of astrocyte-targeted strategies, advocating for precision approaches that account for human-specific astrocyte biology. Future research must priorities human-centric biomarkers and dynamic modelling to bridge the gap between experimental findings and clinical applications.
Collapse
Affiliation(s)
- Ahmad A Abulaban
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; Division of Neurology, King Abdulaziz Medical City, Ministry of the National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.
| | - Hayder M Al-Kuraishy
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine Jabir ibn Hayyan Medical University, Al-Ameer Qu., Najaf, Iraq.
| | - Ali K Albuhadily
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq.
| | - Mustafa M Shokr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish 45511, Egypt.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal 42283, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511, Egypt.
| |
Collapse
|
5
|
Lambe J, Ontaneda D. Re-defining progression in multiple sclerosis. Curr Opin Neurol 2025; 38:188-196. [PMID: 40197617 DOI: 10.1097/wco.0000000000001369] [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: 04/10/2025]
Abstract
PURPOSE OF REVIEW The purpose of this article is to provide an overview of progression in multiple sclerosis (MS), including definitions, pathological mechanisms, and evidence that progressive biology begins early in the disease course. RECENT FINDINGS Definitions of MS clinical course have been refined to acknowledge the presence of both relapse and progression biology throughout the disease. Progression independent of relapse activity represents a significant proportion of disability worsening in relapsing-remitting MS (RRMS) disease. Progression in MS appears to be caused by the complex interplay of multiple processes, including nonresolving inflammation, microglial activation, oxidative stress, mitochondrial dysfunction, energetic failure, and neuro-axonal degeneration. These processes appear to begin in the earliest disease stages and their contribution to clinical phenotypes is dynamic over time. Promising results from clinical trials of tolebrutinib, in particular, underline the utility of targeting both innate and adaptive immune mechanisms to reduce disability accumulation. SUMMARY Pathological processes that underpin MS progression are detectable early in RRMS, evolve throughout the disease course and correlate with disability accumulation. Progression in MS should not be defined dichotomously - the focus instead should be on recognizing progressive components based on clinical measures and biomarkers early in the disease to better individualize treatment strategies.
Collapse
Affiliation(s)
- Jeffrey Lambe
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurology Department, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | | |
Collapse
|
6
|
Wang X, Sun Y, Yu H, Xue C, Pei X, Chen Y, Guan Y. The regulation of microglia by aging and autophagy in multiple sclerosis. Pharmacol Res 2025; 216:107786. [PMID: 40398690 DOI: 10.1016/j.phrs.2025.107786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/19/2025] [Accepted: 05/17/2025] [Indexed: 05/23/2025]
Abstract
Multiple sclerosis (MS) is an inflammatory disease that is often characterized by the development of irreversible clinical disability. Age is a strong risk factor that is strongly associated with the clinical course and progression of MS. Several lines of evidence suggest that with aging, microglia have an aging-related gene expression signature and are close to disease-associated microglia (DAM), which exhibit decreased phagocytosis but increased production of inflammatory factors. The gene expression signatures of microglia in MS overlap with those in aging, inflammation and DAM. Moreover, the clearance of damaged myelin by microglia is impaired in the aged brain. Autophagy is a cellular process that decreases in activity with age. In this review, we provide an overview of the role of autophagy and aging in MS. We describe the impact of autophagy and aging on microglial activation in MS and the molecules involved in autophagy and aging, which are related to the phagocytosis and activation of microglia. We propose that a decrease in autophagy in microglia occurs with aging, leading to a decrease in phagocytosis. Decreases in phagocytosis and increases in the production of inflammatory factors by microglia contribute to chronic inflammation in the aged brain and disease progression in MS. Thus, the modulation of autophagy in microglia serves as a potential therapeutic target for MS.
Collapse
Affiliation(s)
- Xiying Wang
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Sun
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haojun Yu
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunran Xue
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuzhong Pei
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Chen
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangtai Guan
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Yadav SK, Chen C, Dhib-Jalbut S, Ito K. The mechanism of disease progression by aging and age-related gut dysbiosis in multiple sclerosis. Neurobiol Dis 2025; 212:106956. [PMID: 40383164 DOI: 10.1016/j.nbd.2025.106956] [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: 02/14/2025] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025] Open
Abstract
Multiple sclerosis (MS) is the most common demyelinating disease caused by a multifaceted interplay of genetic predispositions and environmental factors. Most patients initially experience the relapsing-remitting form of the disease (RRMS), which is characterized by episodes of neurological deficits followed by periods of symptom resolution. However, over time, many individuals with RRMS advance to a progressive form of the disease, known as secondary progressive MS (SPMS), marked by a gradual worsening of symptoms without periods of remission. The mechanisms underlying this transition remain largely unclear, and current disease-modifying therapies (DMTs) are partially effective in treating SPMS. Age is widely acknowledged as a risk factor for the transition from RRMS to SPMS. One factor associated with aging that may influence the progression of MS is gut dysbiosis. This review discusses how aging and age-related gut dysbiosis affect the progression of MS.
Collapse
Affiliation(s)
- Sudhir Kumar Yadav
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America
| | - Claire Chen
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America
| | - Suhayl Dhib-Jalbut
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America
| | - Kouichi Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America.
| |
Collapse
|
8
|
Hermine O, Gros L, Tran TA, Loussaief L, Flosseau K, Moussy A, Mansfield CD, Vermersch P. Tyrosine kinase inhibitor, masitinib, limits neuronal damage, as measured by serum neurofilament light chain concentration in a model of neuroimmune-driven neurodegenerative disease. PLoS One 2025; 20:e0322199. [PMID: 40367050 PMCID: PMC12077730 DOI: 10.1371/journal.pone.0322199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 03/17/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Masitinib is an orally administered tyrosine kinase inhibitor that targets activated cells of the innate neuroimmune system. We have studied the neuroprotective action of masitinib on the manifestations of experimental autoimmune encephalitis (EAE) induced axonal and neuronal damage. EAE is a model of neuroimmune-driven chronic neuroinflammation and therefore highly relevant to masitinib's mechanism of action in neurodegenerative diseases. Importantly, neuronal damage, or prevention thereof, can be rapidly assessed by measuring serum neurofilament light chain (NfL) concentration in EAE-induced mice. METHODS EAE induction was performed in healthy female C57BL/6 mice via active MOG 35-55 peptide immunization. Treatments were initiated 14 days post EAE induction. On day-0, 39 mice with established EAE symptoms were randomly assigned to 3 treatment groups (n = 13): EAE control, masitinib 50 mg/kg/day (M50), and masitinib 100 mg/kg/day (M100). The treatment started on day-1 and ended on day-15. Blood samples were collected on day-1 and day-8, via tail vein sampling, and on day-15, via intracardiac puncture. Assessments included quantification of serum NfL levels along the disease duration, cytokine quantification at day-15, and clinical assessments. RESULTS Masitinib treatment significantly (p < 0.0001) limited NfL production with respect to control; specifically, relative change in serum NfL concentration at day-8 was 43% and 60% lower for the M50 and M100 groups, respectively. Likewise, for the assessment of absolute serum NfL at day-8 and day-15, there was a significantly lower NfL concentration for masitinib treatment as compared with control. Furthermore, EAE mice treated with masitinib showed significantly lower concentrations of several well-established pro-inflammatory cytokines relative to control at day-15. A beneficial effect of masitinib on functional performance was also observed, with both M50 and M100 groups showing significantly less relative deterioration in grip strength at day-15 as compared with control (p < 0.001). CONCLUSION This study is the first demonstration that masitinib, a drug that targets the innate as opposed to the adaptive neuroimmune system, can lower serum NfL levels, and by extension therefore, neuronal damage, in a neuroimmune-driven neurodegenerative disease model. Overall, findings indicate that masitinib has a neuroprotective effect under conditions of chronic neuroinflammation and therefore plausible disease-modifying activity across a broad range of neurodegenerative diseases.
Collapse
MESH Headings
- Animals
- Female
- Mice
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/blood
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Neurofilament Proteins/blood
- Benzamides
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Mice, Inbred C57BL
- Disease Models, Animal
- Neurons/drug effects
- Neurons/pathology
- Thiazoles/pharmacology
- Thiazoles/therapeutic use
- Neurodegenerative Diseases/drug therapy
- Neurodegenerative Diseases/blood
- Neurodegenerative Diseases/pathology
- Pyridines
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Tyrosine Kinase Inhibitors
- Piperidines
Collapse
Affiliation(s)
- Olivier Hermine
- Imagine Institute, INSERM UMR 1163, University of Paris, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implication, Hôpital Necker, Paris, France
- Department of Hematology, Necker Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | | | | | | | | | | | - Patrick Vermersch
- Univ. Lille, UMR Inserm U1172, CHU Lille, FHU Precise, Lille, France
| |
Collapse
|
9
|
Ojha B, Ramazani B, Belal R, Krieger J, Bloksgaard M, Lyszczarz GT, Rusin D, Wlodarczyk A, FitzGerald U, Owens T, Khorooshi R. Laser-Induced Cortical Lesions in Mice as a Model for Progressive Multiple Sclerosis Pathology. Biomedicines 2025; 13:1195. [PMID: 40427022 PMCID: PMC12109324 DOI: 10.3390/biomedicines13051195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 05/06/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Background: The current animal models of multiple sclerosis (MS) predominantly emphasize white matter inflammation, reflecting early-stage disease. However, progressive MS (PMS) is characterized by cortical pathology, including subpial demyelination, chronic meningeal inflammation, and microglial activation, which are underrepresented in the existing models. While alternative mouse models replicate the relapsing-remitting phenotype and gray matter pathology, pathology is frequently dispersed throughout the brain, complicating the analysis of the specific lesion sites. Methods: To address this gap, we developed a novel model that integrates laser-induced focal demyelination with cytokine-driven meningeal inflammation to replicate the key aspects of PMS cortical pathology. Results: Using two-photon laser irradiation, we induced controlled subpial cortical lesions in CX3CR1-GFP mice, leading to microglial activation, astrocytosis, and focal demyelination. The addition of IFNγ-expressing adenovirus to promote meningeal inflammation which resulted in prolonged glial responses, increased immune cell infiltration, and exacerbated demyelination, mimicking the PMS-associated pathology. Conclusions: This model provides a powerful tool to investigate the mechanisms underlying the cortical lesion development and immune-mediated neurodegeneration in PMS. By capturing the critical aspects of cortical pathology, it enables the evaluation of therapeutic strategies targeting neuroinflammation and demyelination, ultimately aiding in the development of new treatments of progression in PMS patients.
Collapse
Affiliation(s)
- Bhavya Ojha
- Institute of Molecular Medicine, Neurobiology Research Unit, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Bita Ramazani
- Institute of Molecular Medicine, Neurobiology Research Unit, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Rouhin Belal
- Institute of Molecular Medicine, Neurobiology Research Unit, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Jonathan Krieger
- Institute of Molecular Medicine, Neurobiology Research Unit, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Maria Bloksgaard
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Gabriela Teresa Lyszczarz
- Institute of Molecular Medicine, Neurobiology Research Unit, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Dominika Rusin
- Institute of Molecular Medicine, Neurobiology Research Unit, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Agnieszka Wlodarczyk
- Institute of Molecular Medicine, Neurobiology Research Unit, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Una FitzGerald
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, H91 W2TY Galway, Ireland
- Galway Neuroscience Centre, University of Galway, H91 TK33 Galway, Ireland
| | - Trevor Owens
- Institute of Molecular Medicine, Neurobiology Research Unit, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Reza Khorooshi
- Institute of Molecular Medicine, Neurobiology Research Unit, University of Southern Denmark, DK-5230 Odense, Denmark
| |
Collapse
|
10
|
Albelo-Martínez M, Rizvi S. Progressive multiple sclerosis: Evaluating current therapies and exploring future treatment strategies. Neurotherapeutics 2025:e00601. [PMID: 40345951 DOI: 10.1016/j.neurot.2025.e00601] [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: 01/21/2025] [Revised: 04/13/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
Progressive forms of multiple sclerosis (MS) include primary progressive MS (PPMS) and secondary progressive MS (SPMS). Unlike relapsing-remitting MS (RRMS), progressive MS is recognized by relentless progression with accumulating disability, rare to no relapses nor new activity on MRIs. Clinically, neurologic worsening in MS can occur in the relapsing-remitting (RRMS) phase of disease due to incomplete recovery from neuroinflammatory relapses. However, a progressive disease course is the dominant factor related to accumulating disability. There is persistent central nervous system (CNS) compartmentalized inflammation, mitochondrial dysfunction and altered immune responses. Unlike in RRMS, the efficacy of disease modifying agents (DMA) in progressive MS has been limited, highlighting the need for novel therapeutic approaches that address both inflammation and neurodegeneration. This article explores current management of progressive MS, and future directions in targeting the unique pathophysiology of this complex disease.
Collapse
Affiliation(s)
- Marelisa Albelo-Martínez
- Department of Neurology, Brown University Health and Rhode Island Hospital, Alpert Medical School of Brown University, USA.
| | - Syed Rizvi
- Department of Neurology, Brown University Health and Rhode Island Hospital, Alpert Medical School of Brown University, USA.
| |
Collapse
|
11
|
Yamasaki R. Microglia/Macrophages in Autoimmune Demyelinating Encephalomyelitis (Multiple Sclerosis/Neuromyelitis Optica). Int J Mol Sci 2025; 26:3585. [PMID: 40332086 PMCID: PMC12026516 DOI: 10.3390/ijms26083585] [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: 02/17/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Microglia and macrophages are critical mediators of immune responses in the central nervous system. Their roles range from homeostatic maintenance to the pathogenesis of autoimmune demyelinating diseases such as multiple sclerosis and neuromyelitis optica spectrum disorder. This review explores the origins of microglia and macrophages, as well as their mechanisms of activation, interactions with other neural cells, and contributions to disease progression and repair processes. It also highlights the translational relevance of insights gained from animal models and the therapeutic potential of targeting microglial and macrophage activity in multiple sclerosis and neuromyelitis optica spectrum disorder.
Collapse
Affiliation(s)
- Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
12
|
Zahoor I, Nematullah M, Ahmed ME, Fatma M, Sajad M, Ayasolla K, Cerghet M, Palaniyandi S, Ceci V, Carrera G, Buttari F, Centonze D, Mao-Draayer Y, Rattan R, Chiurchiù V, Giri S. Maresin-1 promotes neuroprotection and modulates metabolic and inflammatory responses in disease-associated cell types in preclinical models of multiple sclerosis. J Biol Chem 2025; 301:108226. [PMID: 39864620 PMCID: PMC11903811 DOI: 10.1016/j.jbc.2025.108226] [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: 07/23/2024] [Revised: 12/19/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025] Open
Abstract
Multiple sclerosis (MS) is a prevalent inflammatory neurodegenerative disease in young people, causing neurological abnormalities and impairment. To investigate a novel therapeutic agent for MS, we observed the impact of maresin 1 (MaR1) on disease progression in a well-known, relapsing-remitting experimental autoimmune encephalomyelitis mouse model. Treatment with MaR1 accelerated inflammation resolution, reduced neurological impairment, and delayed disease development by reducing immune cell infiltration (CD4+IL-17+ and CD4+IFNγ+) into the central nervous system. Furthermore, MaR1 administration enhanced IL-10 production, primarily in macrophages and CD4+ cells. However, neutralizing IL-10 with an anti-IL-10 antibody eliminated the protective impact by MaR1 in relapsing-remitting experimental autoimmune encephalomyelitis model, implying the significance of IL-10 in MaR1 treatment. Metabolism has been recognized as a critical mediator of effector activity in many types of immune cells. In our investigation, MaR1 administration significantly repaired metabolic dysregulation in CD4+ cells, macrophages, and microglia in EAE mice. Furthermore, MaR1 treatment restored defective efferocytosis in treated macrophages and microglia. MaR1 also preserved myelin in EAE mice and regulated O4+ oligodendrocyte metabolism by reversing metabolic dysregulation via increased mitochondrial activity and decreased glycolysis. Overall, in a preclinical MS animal model, MaR1 therapy has anti-inflammatory and neuroprotective properties. It also induced metabolic reprogramming in disease-associated cell types, increased efferocytosis, and maintained myelination. Moreover, our data on patient-derived peripheral blood mononuclear cells substantiated the protective role of MaR1, expanding the therapeutic spectrum of specialized proresolving lipid mediators. Altogether, these findings suggest the potential of MaR1 as a novel therapeutic agent for MS and other autoimmune diseases.
Collapse
MESH Headings
- Animals
- Docosahexaenoic Acids/pharmacology
- Docosahexaenoic Acids/therapeutic use
- Mice
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/pathology
- Disease Models, Animal
- Female
- Inflammation/drug therapy
- Inflammation/metabolism
- Inflammation/pathology
- Macrophages/metabolism
- Macrophages/drug effects
- Macrophages/pathology
- Macrophages/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Humans
- Mice, Inbred C57BL
- Interleukin-10/metabolism
- Neuroprotection/drug effects
- Neuroprotective Agents/pharmacology
Collapse
Affiliation(s)
- Insha Zahoor
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | | | | | - Mena Fatma
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | - Mir Sajad
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | | | - Mirela Cerghet
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | - Suresh Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health, Detroit, Michigan, USA; Department of Physiology, Wayne State University, Detroit, Michigan, USA
| | - Veronica Ceci
- Institute of Translational Pharmacology, National Research Council, Rome, Italy; Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giulia Carrera
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Fabio Buttari
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), Italy
| | - Diego Centonze
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), Italy
| | - Yang Mao-Draayer
- Oklahoma Medical Research Foundation, Oklahoma, Farmington Hills, Michigan, USA
| | - Ramandeep Rattan
- Women's Health Services, Henry Ford Health, Detroit, Michigan, USA
| | - Valerio Chiurchiù
- Institute of Translational Pharmacology, National Research Council, Rome, Italy; Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA.
| |
Collapse
|
13
|
Rubino V, Cammarota M, Criscuolo C, Cianflone A, De Martino M, de Rosa V, Esposito F, Abbadessa G, Carriero F, Terrazzano G, Chieffi P, Bonavita S, Bresciamorra V, Annunziato L, Ruggiero G, Boscia F. Modulation of NCX1 expression in monocytes associates with multiple sclerosis progression. Heliyon 2025; 11:e42332. [PMID: 40041001 PMCID: PMC11876900 DOI: 10.1016/j.heliyon.2025.e42332] [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: 05/17/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 03/06/2025] Open
Abstract
Ionic imbalance and functional heterogeneity of monocytes play key roles in multiple sclerosis (MS) progression. A better understanding of monocyte response in the context of ionic dysregulation during MS course may have relevant implications for understanding of disease pathogenesis and treatments. The sodium calcium exchanger NCX1 influences monocyte-derived macrophages reactivity under inflammation; however, little is known about its monocyte-specific expression during MS course. By means of RT-PCR, flow cytometry, and confocal analyses, we determined the expression profiling of NCX1 exchanger in monocytes of patients during relapsing-remitting MS (RRMS) and secondary progressive MS (SPMS) course. NCX1 expression was significantly upregulated in monocytes from transitional RRMS subjects. Conversely, it was significantly reduced in all monocyte subsets after RRMS conversion to SPMS. Interestingly, NCX1 levels in monocytes significantly correlated with the percentage and growth ability of the regulatory T cell (Treg) subset, whose derangement underlies MS progression. Perturbation of transcripts encoding the Ca2+-ATPase isoform 1 and 4, the Na+/K+-ATPase α1 subunit, and the long non-coding RNA SLC8A1-AS1 associated with NCX1 changes in peripheral blood mononuclear cells (PBMC) during MS. Our findings demonstrated a stage-specific dysregulation of NCX1 exchanger in monocytes during MS progression and suggested that ionic imbalance in monocytes may influence not only their functional response but also the immune regulatory network during MS course. These data may be relevant for the identification of novel biomarkers and/or therapeutic targets in MS.
Collapse
Affiliation(s)
- Valentina Rubino
- Department of Medical Translational Sciences, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Mariarosaria Cammarota
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Chiara Criscuolo
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
- CDCD Neurology, “Federico II” University Hospital, Naples, Italy
| | - Alessandra Cianflone
- Clinical and Translational Research Unit, Santobono-Pausilipon Children's Hospital, 80129, Naples, Italy
| | - Marco De Martino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Valeria de Rosa
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Francesco Esposito
- Institute of Experimental Endocrinology and Oncology (IEOS) “G. Salvatore”, National Research Council (CNR), 80131, Naples, Italy
| | - Gianmarco Abbadessa
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
| | - Flavia Carriero
- Department of Health Science, University of Basilicata, 85100, Potenza, Italy
| | - Giuseppe Terrazzano
- Department of Health Science, University of Basilicata, 85100, Potenza, Italy
| | - Paolo Chieffi
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138, Naples, Italy
| | - Vincenzo Bresciamorra
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
- Multiple Sclerosis Unit, Policlinico “Federico II” University Hospital, 80131, Naples, Naples, Italy
| | | | - Giuseppina Ruggiero
- Department of Medical Translational Sciences, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| | - Francesca Boscia
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, “Federico II” University of Naples, 80131, Naples, Italy
| |
Collapse
|
14
|
Sun Q, Zhu J, Zhao X, Huang X, Qu W, Tang X, Ma D, Shu Q, Li X. Mettl3-m 6A-NPY axis governing neuron-microglia interaction regulates sleep amount of mice. Cell Discov 2025; 11:10. [PMID: 39905012 PMCID: PMC11794856 DOI: 10.1038/s41421-024-00756-y] [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: 05/14/2024] [Accepted: 11/13/2024] [Indexed: 02/06/2025] Open
Abstract
Sleep behavior is regulated by diverse mechanisms including genetics, neuromodulation and environmental signals. However, it remains completely unknown regarding the roles of epitranscriptomics in regulating sleep behavior. In the present study, we showed that the deficiency of RNA m6A methyltransferase Mettl3 in excitatory neurons specifically induces microglia activation, neuroinflammation and neuronal loss in thalamus of mice. Mettl3 deficiency remarkably disrupts sleep rhythm and reduces the amount of non-rapid eye movement sleep. We also showed that Mettl3 regulates neuropeptide Y (NPY) via m6A modification and Mettl3 conditional knockout (cKO) mice displayed significantly decreased expression of NPY in thalamus. In addition, the dynamic distribution pattern of NPY is observed during wake-sleep cycle in cKO mice. Ectopic expression of Mettl3 and NPY significantly inhibits microglia activation and neuronal loss in thalamus, and restores the disrupted sleep behavior of cKO mice. Collectively, our study has revealed the critical function of Mettl3-m6A-NPY axis in regulating sleep behavior.
Collapse
Affiliation(s)
- Qihang Sun
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jinpiao Zhu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Xingsen Zhao
- Institute of Biotechnology, Xianghu Laboratory, Hangzhou, Zhejiang, China
| | - Xiaoli Huang
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Wenzheng Qu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Xia Tang
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Daqing Ma
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Division of Anesthetics, Pain Medicine & Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.
| | - Qiang Shu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Xuekun Li
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Weiner HL. Immune mechanisms and shared immune targets in neurodegenerative diseases. Nat Rev Neurol 2025; 21:67-85. [PMID: 39681722 DOI: 10.1038/s41582-024-01046-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The immune system plays a major part in neurodegenerative diseases. In some, such as multiple sclerosis, it is the primary driver of the disease. In others, such as Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, it has an amplifying role. Immunotherapeutic approaches that target the adaptive and innate immune systems are being explored for the treatment of almost all neurological diseases, and the targets and approaches are often common across diseases. Microglia are the primary immune cells in the brain that contribute to disease pathogenesis, and are consequently a common immune target for therapy. Other therapeutic approaches target components of the peripheral immune system, such as regulatory T cells and monocytes, which in turn act within the CNS. This Review considers in detail how microglia, monocytes and T cells contribute to the pathogenesis of multiple sclerosis, Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, and their potential as shared therapeutic targets across these diseases. The microbiome is also highlighted as an emerging therapeutic target that indirectly modulates the immune system. Therapeutic approaches being developed to target immune function in neurodegenerative diseases are discussed, highlighting how immune-based approaches developed to treat one disease could be applicable to multiple other neurological diseases.
Collapse
Affiliation(s)
- Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Mokaram Doust Delkhah A. Identification of shared pathogenic signatures of multiple sclerosis and chronic obstructive pulmonary disease: an integrated transcriptomic analysis of blood specimens. Mol Genet Genomics 2024; 300:8. [PMID: 39725779 DOI: 10.1007/s00438-024-02215-5] [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: 08/16/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
Patients with multiple sclerosis (MS) face a heightened risk of developing chronic obstructive pulmonary disease (COPD). Despite this widely reported association, the pathogenic contributors and processes that may favor the development of COPD in MS patients have yet to be identified. Recent studies have suggested peripheral blood leukocytes as a potential link between COPD and autoimmune disorders. Therefore, this study aimed to unveil shared molecular signatures between MS and COPD using blood transcriptomes. To this end, gene expression datasets obtained from MS and COPD blood specimens were retrieved from the Gene Expression Omnibus (GEO) database. By integrating datasets belonging to each disorder, differentially expressed genes (DEGs) were determined for each disease. Then, the protein-protein interaction (PPI) network was constructed for shared DEGs between MS and COPD. Subsequently, the network was analyzed to identify hub genes and key regulatory miRNAs. The integrated data for MS encompassed 51 samples (28 from MS patients and 23 from controls), and the integrated data for COPD included 450 samples (275 from COPD patients and 175 from controls). A total of 246 genes were found to exhibit identical directions of expression in both MS and COPD. By applying a high confidence threshold (0.7), a PPI network with 74 nodes was constructed. TP53, H4C6, SNRPE, and RPS11 were identified as hub genes according to the degree measure. In addition, 8 miRNAs were identified as key regulators, each interacting with 6 mRNAs. Among these miRNAs, miR-218-5p and miR-142-5p have been previously reported to contribute to the pathogenesis of these diseases, and here they were identified as key regulators of the shared PPI network, suggesting a potential epigenetic link between MS and COPD. In conclusion, the results highlighted the potential role of peripheral blood leucocytes as a bridge between MS and COPD. These findings broaden our understanding of pathogenic contributors linking MS and COPD. While this transcriptomics study identified multiple key players, such as TP53, miR-218-5p, and miR-142-5p, the assessment of their therapeutic efficacy demands further experimental studies.
Collapse
Affiliation(s)
- Arman Mokaram Doust Delkhah
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
17
|
Kim ME, Lee JS. Mechanisms and Emerging Regulators of Neuroinflammation: Exploring New Therapeutic Strategies for Neurological Disorders. Curr Issues Mol Biol 2024; 47:8. [PMID: 39852123 PMCID: PMC11763386 DOI: 10.3390/cimb47010008] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
Neuroinflammation is a complex and dynamic response of the central nervous system (CNS) to injury, infection, and disease. While acute neuroinflammation plays a protective role by facilitating pathogen clearance and tissue repair, chronic and dysregulated inflammation contributes significantly to the progression of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and Multiple Sclerosis. This review explores the cellular and molecular mechanisms underlying neuroinflammation, focusing on the roles of microglia, astrocytes, and peripheral immune cells. Key signaling pathways, including NF-κB, JAK-STAT, and the NLRP3 inflammasome, are discussed alongside emerging regulators such as non-coding RNAs, epigenetic modifications, and the gut-brain axis. The therapeutic landscape is evolving, with traditional anti-inflammatory drugs like NSAIDs and corticosteroids offering limited efficacy in chronic conditions. Immunomodulators, gene and RNA-based therapeutics, and stem cell methods have all shown promise for more specific and effective interventions. Additionally, the modulation of metabolic states and gut microbiota has emerged as a novel strategy to regulate neuroinflammation. Despite significant progress, challenges remain in translating these findings into clinically viable therapies. Future studies should concentrate on integrated, interdisciplinary methods to reduce chronic neuroinflammation and slowing the progression of neurodegenerative disorders, providing opportunities for revolutionary advances in CNS therapies.
Collapse
Affiliation(s)
| | - Jun Sik Lee
- Immunology Research Lab & BK21-Four Educational Research Group for Age-Associated Disorder Control Technology, Department of Biological Science, Chosun University, Gwangju 61452, Republic of Korea;
| |
Collapse
|
18
|
Kadam R, Gupta M, Lazarov O, Prabhakar BS. Brain-immune interactions: implication for cognitive impairments in Alzheimer's disease and autoimmune disorders. J Leukoc Biol 2024; 116:1269-1290. [PMID: 38869088 DOI: 10.1093/jleuko/qiae134] [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: 11/13/2023] [Revised: 05/09/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024] Open
Abstract
Progressive memory loss and cognitive dysfunction, encompassing deficits in learning, memory, problem solving, spatial reasoning, and verbal expression, are characteristics of Alzheimer's disease and related dementia. A wealth of studies has described multiple roles of the immune system in the development or exacerbation of dementia. Individuals with autoimmune disorders can also develop cognitive dysfunction, a phenomenon termed "autoimmune dementia." Together, these findings underscore the pivotal role of the neuroimmune axis in both Alzheimer's disease and related dementia and autoimmune dementia. The dynamic interplay between adaptive and innate immunity, both in and outside the brain, significantly affects the etiology and progression of these conditions. Multidisciplinary research shows that cognitive dysfunction arises from a bidirectional relationship between the nervous and immune systems, though the specific mechanisms that drive cognitive impairments are not fully understood. Intriguingly, this reciprocal regulation occurs at multiple levels, where neuronal signals can modulate immune responses, and immune system-related processes can influence neuronal viability and function. In this review, we consider the implications of autoimmune responses in various autoimmune disorders and Alzheimer's disease and explore their effects on brain function. We also discuss the diverse cellular and molecular crosstalk between the brain and the immune system, as they may shed light on potential triggers of peripheral inflammation, their effect on the integrity of the blood-brain barrier, and brain function. Additionally, we assess challenges and possibilities associated with developing immune-based therapies for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Rashmi Kadam
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| |
Collapse
|
19
|
Hijal N, Fouani M, Awada B. Unveiling the fate and potential neuroprotective role of neural stem/progenitor cells in multiple sclerosis. Front Neurol 2024; 15:1438404. [PMID: 39634777 PMCID: PMC11614735 DOI: 10.3389/fneur.2024.1438404] [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: 05/25/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Chronic pathological conditions often induce persistent systemic inflammation, contributing to neuroinflammatory diseases like Multiple Sclerosis (MS). MS is known for its autoimmune-mediated damage to myelin, axonal injury, and neuronal loss which drive disability accumulation and disease progression, often manifesting as cognitive impairments. Understanding the involvement of neural stem cells (NSCs) and neural progenitor cells (NPCs) in the remediation of MS through adult neurogenesis (ANG) and gliogenesis-the generation of new neurons and glial cells, respectively is of great importance. Hence, these phenomena, respectively, termed ANG and gliogenesis, involve significant structural and functional changes in neural networks. Thus, the proper integration of these newly generated cells into existing circuits is not only key to understanding the CNS's development but also its remodeling in adulthood and recovery from diseases such as MS. Understanding how MS influences the fate of NSCs/NPCs and their possible neuroprotective role, provides insights into potential therapeutic interventions to alleviate the impact of MS on cognitive function and disease progression. This review explores MS, its pathogenesis, clinical manifestations, and its association with ANG and gliogenesis. It highlights the impact of altered NSCs and NPCs' fate during MS and delves into the potential benefits of its modifications. It also evaluates treatment regimens that influence the fate of NSCS/NPCs to counteract the pathology subsequently.
Collapse
Affiliation(s)
- Nora Hijal
- Department of Nursing, American University of Beirut Medical Center, Beirut, Lebanon
| | - Malak Fouani
- Department of Neurology, Duke University Medical Center, Durham, NC, United States
| | - Bassel Awada
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
20
|
Ganz T, Fainstein N, Theotokis P, Elgavish S, Vardi-Yaakov O, Lachish M, Sofer L, Zveik O, Grigoriadis N, Ben-Hur T. Targeting CNS myeloid infiltrates provides neuroprotection in a progressive multiple sclerosis model. Brain Behav Immun 2024; 122:497-509. [PMID: 39179123 DOI: 10.1016/j.bbi.2024.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/30/2024] [Accepted: 08/17/2024] [Indexed: 08/26/2024] Open
Abstract
Demyelination and axonal injury in chronic-progressive Multiple Sclerosis (MS) are presumed to be driven by a neurotoxic bystander effect of meningeal-based myeloid infiltrates. There is an unmet clinical need to attenuate disease progression in such forms of CNS-compartmentalized MS. The failure of systemic immune suppressive treatments has highlighted the need for neuroprotective and repair-inducing strategies. Here, we examined whether direct targeting of CNS myeloid cells and modulating their toxicity may prevent irreversible tissue injury in chronic immune-mediated demyelinating disease. To that end, we utilized the experimental autoimmune encephalomyelitis (EAE) model in Biozzi mice, a clinically relevant MS model. We continuously delivered intracerebroventricularly (ICV) a retinoic acid receptor alpha agonist (RARα), as a potent regulator of myeloid cells, in the chronic phase of EAE. We assessed disease severity and performed pathological evaluations, functional analyses of immune cells, and single-cell RNA sequencing on isolated spinal CD11b+ cells. Although initiating treatment in the chronic phase of the disease, the RARα agonist successfully improved clinical outcomes and prevented axonal loss. ICV RARα agonist treatment inhibited pro-inflammatory pathways and shifted CNS myeloid cells toward neuroprotective phenotypes without affecting peripheral infiltrating myeloid cell phenotypes, or peripheral immunity. The treatment regulated cell-death pathways across multiple myeloid cell populations and suppressed apoptosis, resulting in paradoxically marked increased neuroinflammatory infiltrates, consisting mainly of microglia and CNS / border-associated macrophages. This work establishes the notion of bystander neurotoxicity by CNS immune infiltrates in chronic demyelinating disease. Furthermore, it shows that targeting compartmentalized neuroinflammation by selective regulation of CNS myeloid cell toxicity and survival reduces irreversible tissue injury, and may serve as a novel disease-modifying approach.
Collapse
Affiliation(s)
- Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Paschalis Theotokis
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - Sharona Elgavish
- Info-CORE, Bioinformatics Unit of the 1-CORE, Hebrew University of Jerusalem, Israel
| | - Oriya Vardi-Yaakov
- Info-CORE, Bioinformatics Unit of the 1-CORE, Hebrew University of Jerusalem, Israel; Department of Bioinformatics, Jerusalem College of Technology, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Lihi Sofer
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nikolaos Grigoriadis
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
21
|
Drake SS, Zaman A, Gianfelice C, Hua EML, Heale K, Afanasiev E, Klement W, Stratton JA, Prat A, Zandee S, Fournier AE. Senolytic treatment diminishes microglia and decreases severity of experimental autoimmune encephalomyelitis. J Neuroinflammation 2024; 21:283. [PMID: 39487537 PMCID: PMC11529445 DOI: 10.1186/s12974-024-03278-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND The role of senescence in disease contexts is complex, however there is considerable evidence that depletion of senescent cells improves outcomes in a variety of contexts particularly related to aging, cognition, and neurodegeneration. Much research has shown previously that inflammation can promote cellular senescence. Microglia are a central nervous system innate immune cell that undergo senescence with aging and during neurodegeneration. The contribution of senescent microglia to multiple sclerosis, an inflammatory neurodegenerative disease, is not clear, but microglia are strongly implicated in chronic active lesion pathology, tissue injury, and disease progression. Drugs that could specifically eliminate dysregulated microglia in multiple sclerosis are therefore of great interest to the field. RESULTS A single-cell analysis of brain tissue from mice subjected to experimental autoimmune encephalomyelitis (EAE), a mouse model of CNS inflammation that models aspects of multiple sclerosis (MS), identified microglia with a strong transcriptional signature of senescence including the presence of BCL2-family gene transcripts. Microglia expressing Bcl2l1 had higher expression of pro-inflammatory and senescence associated genes than their Bcl2l1 negative counterparts in EAE, suggesting they may exacerbate inflammation. Notably, in human single-nucleus sequencing from MS, BCL2L1 positive microglia were enriched in lesions with active inflammatory pathology, and likewise demonstrated increased expression of immune genes suggesting they may be proinflammatory and contribute to disease processes in chronic active lesions. Employing a small molecule BCL2-family inhibitor, Navitoclax (ABT-263), significantly reduced the presence of microglia and macrophages in the EAE spinal cord, suggesting that these cells can be targeted by senolytic treatment. ABT-263 treatment had a profound effect on EAE mice: decreasing motor symptom severity, improving visual acuity, promoting neuronal survival, and decreasing white matter inflammation. CONCLUSION These results support the hypothesis that microglia and macrophages exhibit transcriptional features of cellular senescence in EAE and MS, and that microglia expressing Bcl2l1 demonstrate a proinflammatory signature that may exacerbate inflammation resulting in negative outcomes in neuroinflammatory disease. Depleting microglia and macrophages using a senolytic results in robust improvement in EAE disease severity, including across measures of neurodegeneration, inflammation, and demyelination, and may therefore represent a novel strategy to address disease progression in multiple sclerosis.
Collapse
Affiliation(s)
- Sienna S Drake
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Aliyah Zaman
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | | | - Elizabeth M-L Hua
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Kali Heale
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Elia Afanasiev
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Wendy Klement
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l', Université de Montréal (CRCHUM), Montreal, Québec, H2X 0A9, Canada
| | - Jo Anne Stratton
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l', Université de Montréal (CRCHUM), Montreal, Québec, H2X 0A9, Canada
| | - Stephanie Zandee
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Alyson E Fournier
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
22
|
Chen L, Lu J, Hu J, Gong X. Unveiling the multifaceted role of adropin in various diseases (Review). Int J Mol Med 2024; 54:90. [PMID: 39155866 PMCID: PMC11335353 DOI: 10.3892/ijmm.2024.5414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/31/2024] [Indexed: 08/20/2024] Open
Abstract
Adropin is a secreted peptide encoded by the energy homeostasis‑associated gene, which also functions as a membrane‑bound protein facilitating intercellular communication. This peptide has been detected in various tissues and body fluids, including the brain, liver, kidney, heart, pancreas, small intestine, endothelial cells and colostrum. Notably, the amino acid sequences of adropin are identical in humans, mice and rats. Previous studies have demonstrated that adropin levels fluctuate under different physiological and pathological conditions. Adropin plays a role in regulating carbohydrate metabolism, lipid metabolism and intercellular molecular signaling pathways, implicating its involvement in the progression of numerous diseases, such as acute myocardial infarction, lung injury, non‑alcoholic fatty liver disease/non‑alcoholic steatohepatitis, kidney disease, polycystic ovary syndrome, obesity, and diabetes, atherosclerosis, systemic sclerosis and cancer. Despite its significance, the precise role and mechanism of this protein remain inadequately understood and studied. To elucidate the function of adropin and its clinical research status, a systematic review of recent studies on adropin across various diseases was conducted. Additionally, several challenges and limitations associated with adropin research in both animal and clinical contexts were identified, aiming to offer valuable insights for future investigation.
Collapse
Affiliation(s)
- Ling Chen
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Jianrao Lu
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Jing Hu
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Xuezhong Gong
- Department of Nephrology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| |
Collapse
|
23
|
Guvenek A, Parikshak N, Zamolodchikov D, Gelfman S, Moscati A, Dobbyn L, Stahl E, Shuldiner A, Coppola G. Transcriptional profiling in microglia across physiological and pathological states identifies a transcriptional module associated with neurodegeneration. Commun Biol 2024; 7:1168. [PMID: 39294270 PMCID: PMC11411103 DOI: 10.1038/s42003-024-06684-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 08/06/2024] [Indexed: 09/20/2024] Open
Abstract
Microglia are the resident immune cells of the central nervous system and are involved in brain development, homeostasis, and disease. New imaging and genomics technologies are revealing microglial complexity across developmental and functional states, brain regions, and diseases. We curated a set of publicly available gene expression datasets from human microglia spanning disease and health to identify sets of genes reflecting physiological and pathological microglial states. We also integrated multiple human microglial single-cell RNA-seq datasets in Alzheimer's disease (AD), multiple sclerosis (MS), and Parkinson's disease, and identified a distinct microglial transcriptional signature shared across diseases. Analysis of germ-line DNA identified genes with variants associated with AD and MS that are overrepresented in microglial gene sets, including the disease-associated transcriptional signature. This work points to genes that are dysregulated in disease states and provides a resource for the analysis of diseases in which microglia are implicated by genetic evidence.
Collapse
Affiliation(s)
- Aysegul Guvenek
- Regeneron Genetics Center, Tarrytown, NY, USA
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | | | | | | | | | - Lee Dobbyn
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Eli Stahl
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | |
Collapse
|
24
|
Domínguez-Mozo MI, Casanova I, Monreal E, Costa-Frossard L, Sainz-de-la-Maza S, Sainz-Amo R, Aladro-Benito Y, Lopez-Ruiz P, De-Torres L, Abellán S, Garcia-Martinez MA, De-la-Cuesta D, Lourido D, Torrado-Carvajal A, Gomez-Barbosa C, Linares-Villavicencio C, Villar LM, López-De-Silanes C, Arroyo R, Alvarez-Lafuente R. Association of MicroRNA Expression and Serum Neurofilament Light Chain Levels with Clinical and Radiological Findings in Multiple Sclerosis. Int J Mol Sci 2024; 25:10012. [PMID: 39337499 PMCID: PMC11432459 DOI: 10.3390/ijms251810012] [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: 08/01/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
microRNAs (miRNAs) are promising biomarkers for many diseases, including multiple sclerosis (MS). The neurofilament light chain (NfL) is a biomarker that can detect axonal damage in different neurological diseases. The objective of this study was to evaluate the association of the expression profile of pre-selected miRNAs and NfL levels with clinical and radiological variables in MS patients. We conducted a 1-year longitudinal prospective study in MS patients with different clinical forms. We measured clinical disability using the expanded disability status scale (EDSS), the magnetic resonance imaging (MRI) volumetry baseline, and cognitive functioning using the processing speed test (PST) at baseline and 1 year later. Selected serum miRNAs and serum NfL (sNfL) levels were quantified. Seventy-three patients were recruited. MiR-126.3p correlated with EDSS and cognitive status at baseline and miR-126.3p and miR-9p correlated with cognitive deterioration at 1 year. Correlations with regional brain volumes were observed between miR-126.3p and the cortical gray matter, cerebellum, putamen, and pallidum; miR-146a.5p with the cerebellum and pallidum; miR-29b.3p with white matter and the pallidum; miR-138.5p with the pallidum; and miR-9.5p with the thalamus. sNfL was correlated with miR-9.5p. miR-146a.5p was also associated with the MS phenotype. These data justify future studies to further explore the utility of miRNAs (mirR-126.3p, miR-146.5p, and miR.9-5p) and sNfL levels as biomarkers of MS.
Collapse
Affiliation(s)
- María Inmaculada Domínguez-Mozo
- Research Group in Environmental Factors of Neurodegenerative Diseases, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), 28040 Madrid, Spain; (M.A.G.-M.); (D.D.-l.-C.); (R.A.-L.)
| | - Ignacio Casanova
- Department of Neurology, Hospital Universitario de Torrejón, 28850 Madrid, Spain; (I.C.); (L.D.-T.); (S.A.); (C.L.-D.-S.)
- School of Medicine, Universidad Francisco de Vitoria, 28223 Madrid, Spain
- Department of Neurology, Hospital Universitario QuironSalud Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (P.L.-R.); (R.A.)
| | - Enric Monreal
- Department of Neurology, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain; (E.M.); (L.C.-F.); (S.S.-d.-l.-M.); (R.S.-A.)
| | - Lucienne Costa-Frossard
- Department of Neurology, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain; (E.M.); (L.C.-F.); (S.S.-d.-l.-M.); (R.S.-A.)
| | - Susana Sainz-de-la-Maza
- Department of Neurology, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain; (E.M.); (L.C.-F.); (S.S.-d.-l.-M.); (R.S.-A.)
| | - Raquel Sainz-Amo
- Department of Neurology, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain; (E.M.); (L.C.-F.); (S.S.-d.-l.-M.); (R.S.-A.)
| | | | - Pedro Lopez-Ruiz
- Department of Neurology, Hospital Universitario QuironSalud Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (P.L.-R.); (R.A.)
| | - Laura De-Torres
- Department of Neurology, Hospital Universitario de Torrejón, 28850 Madrid, Spain; (I.C.); (L.D.-T.); (S.A.); (C.L.-D.-S.)
| | - Sara Abellán
- Department of Neurology, Hospital Universitario de Torrejón, 28850 Madrid, Spain; (I.C.); (L.D.-T.); (S.A.); (C.L.-D.-S.)
| | - Maria Angel Garcia-Martinez
- Research Group in Environmental Factors of Neurodegenerative Diseases, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), 28040 Madrid, Spain; (M.A.G.-M.); (D.D.-l.-C.); (R.A.-L.)
| | - David De-la-Cuesta
- Research Group in Environmental Factors of Neurodegenerative Diseases, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), 28040 Madrid, Spain; (M.A.G.-M.); (D.D.-l.-C.); (R.A.-L.)
| | - Daniel Lourido
- Department of Radiology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain;
| | - Angel Torrado-Carvajal
- Medical Image Analysis and Biometry Laboratory, Universidad Rey Juan Carlos, Móstoles, 28933 Madrid, Spain;
| | - Carol Gomez-Barbosa
- Department of Radiology, Hospital Universitario de Torrejón, 28850 Madrid, Spain; (C.G.-B.); (C.L.-V.)
| | | | - Luisa Maria Villar
- Department of Immunology, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain;
| | - Carlos López-De-Silanes
- Department of Neurology, Hospital Universitario de Torrejón, 28850 Madrid, Spain; (I.C.); (L.D.-T.); (S.A.); (C.L.-D.-S.)
| | - Rafael Arroyo
- Department of Neurology, Hospital Universitario QuironSalud Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (P.L.-R.); (R.A.)
| | - Roberto Alvarez-Lafuente
- Research Group in Environmental Factors of Neurodegenerative Diseases, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), 28040 Madrid, Spain; (M.A.G.-M.); (D.D.-l.-C.); (R.A.-L.)
| |
Collapse
|
25
|
Zhang Q, Sun W, Zheng M, Zhang N. Contribution of microglia/macrophage to the pathogenesis of TMEV infection in the central nervous system. Front Microbiol 2024; 15:1452390. [PMID: 39155988 PMCID: PMC11327027 DOI: 10.3389/fmicb.2024.1452390] [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: 06/20/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Abstract
The infection of the central nervous system (CNS) with neurotropic viruses induces neuroinflammation and an immune response, which is associated with the development of neuroinflammatory and neurodegenerative diseases, including multiple sclerosis (MS). The activation of both innate and adaptive immune responses, involving microglia, macrophages, and T and B cells, while required for efficient viral control within the CNS, is also associated with neuropathology. Under pathological events, such as CNS viral infection, microglia/macrophage undergo a reactive response, leading to the infiltration of immune cells from the periphery into the brain, disrupting CNS homeostasis and contributing to the pathogenesis of disease. The Theiler's murine encephalomyelitis virus (TMEV)-induced demyelination disease (TMEV-IDD), which serves as a mouse model of MS. This murine model made significant contributions to our understanding of the pathophysiology of MS following subsequent to infection. Microglia/macrophages could be activated into two different states, classic activated state (M1 state) and alternative activated state (M2 state) during TMEV infection. M1 possesses the capacity to initiate inflammatory response and secretes pro-inflammatory cytokines, and M2-liked microglia/macrophages are anti-inflammatory characterized by the secretion of anti-inflammatory cytokines. This review aims to discuss the roles of microglia/macrophages M1/M2-liked polarization during TMEV infection, and explore the potential therapeutic effect of balancing M1/M2-liked polarization of microglia/macrophages on MS.
Collapse
Affiliation(s)
| | | | | | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| |
Collapse
|
26
|
Prapas P, Anagnostouli M. Macrophages and HLA-Class II Alleles in Multiple Sclerosis: Insights in Therapeutic Dynamics. Int J Mol Sci 2024; 25:7354. [PMID: 39000461 PMCID: PMC11242320 DOI: 10.3390/ijms25137354] [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: 06/15/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Antigen presentation is a crucial mechanism that drives the T cell-mediated immune response and the development of Multiple Sclerosis (MS). Genetic alterations within the highly variable Major Histocompatibility Complex Class II (MHC II) have been proven to result in significant changes in the molecular basis of antigen presentation and the clinical course of patients with both Adult-Onset MS (AOMS) and Pediatric-Onset MS (POMS). Among the numerous polymorphisms of the Human Leucocyte Antigens (HLA), within MHC II complex, HLA-DRB1*15:01 has been labeled, in Caucasian ethnic groups, as a high-risk allele for MS due to the ability of its structure to increase affinity to Myelin Basic Protein (MBP) epitopes. This characteristic, among others, in the context of the trimolecular complex or immunological synapsis, provides the foundation for autoimmunity triggered by environmental or endogenous factors. As with all professional antigen presenting cells, macrophages are characterized by the expression of MHC II and are often implicated in the formation of MS lesions. Increased presence of M1 macrophages in MS patients has been associated both with progression and onset of the disease, each involving separate but similar mechanisms. In this critical narrative review, we focus on macrophages, discussing how HLA genetic alterations can promote dysregulation of this population's homeostasis in the periphery and the Central Nervous System (CNS). We also explore the potential interconnection in observed pathological macrophage mechanisms and the function of the diverse structure of HLA alleles in neurodegenerative CNS, seen in MS, by comparing available clinical with molecular data through the prism of HLA-immunogenetics. Finally, we discuss available and experimental pharmacological approaches for MS targeting the trimolecular complex that are based on cell phenotype modulation and HLA genotype involvement and try to reveal fertile ground for the potential development of novel drugs.
Collapse
Affiliation(s)
- Petros Prapas
- Research Immunogenetics Laboratory, First Department of Neurology, Aeginition University Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sofias 72-74, 11528 Athens, Greece
| | - Maria Anagnostouli
- Research Immunogenetics Laboratory, First Department of Neurology, Aeginition University Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sofias 72-74, 11528 Athens, Greece
- Multiple Sclerosis and Demyelinating Diseases Unit, Center of Expertise for Rare Demyelinating and Autoimmune Diseases of CNS, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens NKUA, Aeginition University Hospital, Vas. Sofias 72-74, 11528 Athens, Greece
| |
Collapse
|
27
|
Hamano S, Yoshimizu T, Mori M, Iida A, Yamashita T. Characterization of pathological stages in a mouse model of progressive multiple sclerosis. Neurosci Res 2024; 204:46-57. [PMID: 38307349 DOI: 10.1016/j.neures.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/10/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
The purpose of this study was to analyze and elucidate the mechanisms of non-obese diabetes-experimental autoimmune encephalomyelitis (NOD-EAE), an animal model of progressive multiple sclerosis (MS), and to compare the pathological features with those observed in human progressive MS. Pathological analysis, flow cytometry analysis, immunohistochemical staining, and transcriptome analysis were performed at each pathological stage of the NOD-EAE mice to characterize each pathological stage in the lesion. The NOD-EAE mice showed a biphasic pattern of disease progression once in remission. The longitudinal profile of demyelination and inflammatory cell infiltration in the spinal cord was consistent with the pathological score. In the chronic phase of the disease, fibrosis and lymph follicle formation, characteristic of progressive human MS, were observed. Here we describe the pathological profile and transcriptome analysis of the NOD-EAE mice and verify that this model has similar features to those of human progressive MS. Our findings suggest that this model recapitulates lymph follicle formation, a disease hallmark of progressive MS, and fibrosis, a feature complicating the pathogenesis of MS in the chronic phase. This model may be useful for evaluating the efficacy of therapeutic agents and for mechanistic analysis.
Collapse
Affiliation(s)
- Satoshi Hamano
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan.
| | - Toshiki Yoshimizu
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Mutsuki Mori
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Akio Iida
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan; Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
28
|
Shi Z, Sun H, Tian X, Song X, Fan J, Sun S, Wang J, Zhang J, Wang J. Extracellular vesicles containing miR-181a-5p as a novel therapy for experimental autoimmune encephalomyelitis-induced demyelination. Int Immunopharmacol 2024; 135:112326. [PMID: 38796967 DOI: 10.1016/j.intimp.2024.112326] [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: 08/28/2023] [Revised: 05/06/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disorder of the central nervous system. Recent research has revealed that mesenchymal stem cell-derived extracellular vesicles (MSC-EVs), containing specific miRNAs, possess immunomodulatory properties and have demonstrated therapeutic potential in the treatment of MS. This study aimed to investigate the role MSC-EVs, containing microRNA-181a-5p (miR-181a-5p) in both experimental autoimmune encephalomyelitis (EAE), an established animal model of MS, and lipopolysaccharide-stimulated BV2 microglia. We evaluated clinical symptoms and inflammatory responses in EAE mice following intrathecal injections of MSC-EVs. MSC-EVs containing miR-181a-5p were co-cultured with microglia to explore their impact on inflammation and cell pyroptosis. We validated the interaction between miR-181a-5p and its downstream regulators and conducted in vivo verification by injecting manipulated EVs containing miR-181a-5p into EAE mice. Our results demonstrated that MSC-EVs, containing miR-181a-5p reduced the clinical symptoms of EAE mice. Furthermore, we observed downregulation of miR-181a-5p in EAE model mice, and its expression was restored after treatment with MSC-EVs, which corresponded to suppressed microglial inflammation and pyroptosis. Additionally, EVs containing miR-181a-5p mitigated spinal cord injury and demyelination in EAE mice. Mechanistically, ubiquitin-specific protease 15 (USP15) exhibited high expression in EAE mice, and miR-181a-5p was specifically targeted and bound to USP15, thereby regulating the RelA/NEK7 axis. In conclusion, MSC-EVs containing miR-181a-5p inhibit microglial inflammation and pyroptosis through the USP15-mediated RelA/NEK7 axis, thus alleviating the clinical symptoms of EAE. These findings present a potential therapeutic approach for the treatment of MS.
Collapse
Affiliation(s)
- Zhong Shi
- Ophthalmology Department, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Huanhuan Sun
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Xinyi Tian
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao 266002, Shandong, China
| | - Xiujuan Song
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Jingyi Fan
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Shichao Sun
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Jinli Wang
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Jing Zhang
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Jueqiong Wang
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China.
| |
Collapse
|
29
|
Sabnis RW, Sabnis AR. Novel Compounds as S1P5 Modulators for Treating Neurodegenerative Diseases. ACS Med Chem Lett 2024; 15:750-751. [PMID: 38894913 PMCID: PMC11181510 DOI: 10.1021/acsmedchemlett.4c00178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Indexed: 06/21/2024] Open
Abstract
Provided herein are novel compounds as S1P5 modulators, pharmaceutical compositions, use of such compounds in treating neurodegenerative diseases, particularly Alzheimer's disease, multiple sclerosis, migraine and amyotrophic lateral sclerosis, and processes for preparing such compounds.
Collapse
Affiliation(s)
- Ram W. Sabnis
- Smith,
Gambrell & Russell LLP, 1105 W. Peachtree Street NE, Suite 1000, Atlanta, Georgia 30309, United States
| | - Anika R. Sabnis
- Neuroscience
Institute, Georgia State University, Atlanta, Georgia 30303, United States
| |
Collapse
|
30
|
Turner TJ, Brun P, Gruber RC, Ofengeim D. Comparative CNS Pharmacology of the Bruton's Tyrosine Kinase (BTK) Inhibitor Tolebrutinib Versus Other BTK Inhibitor Candidates for Treating Multiple Sclerosis. Drugs R D 2024; 24:263-274. [PMID: 38965189 PMCID: PMC11315827 DOI: 10.1007/s40268-024-00468-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Tolebrutinib is a covalent BTK inhibitor designed and selected for potency and CNS exposure to optimize impact on BTK-dependent signaling in CNS-resident cells. We applied a translational approach to evaluate three BTK inhibitors in Phase 3 clinical development in MS with respect to their relative potency to block BTK-dependent signaling and exposure in the CNS METHODS: We used in vitro kinase and cellular activation assays, alongside pharmacokinetic sampling of cerebrospinal fluid (CSF) in the non-human primate cynomolgus to estimate the ability of these candidates (evobrutinib, fenebrutinib, and tolebrutinib) to block BTK-dependent signaling inside the CNS. RESULTS In vitro kinase assays demonstrated that tolebrutinib reacted with BTK 65-times faster than evobrutinib, while fenebrutinib, a classical reversible antagonist with a Ki value of 4.7 nM and slow off-rate (1.54 x 10-5 s-1), also had an association rate 1760-fold slower (0.00245 μM-1 * s-1). Estimates of cellular potency were largely consistent with the in vitro kinase assays, with an estimated IC50 of 0.7 nM for tolebrutinib against 33.5 nM for evobrutinib and 2.9 nM for fenebrutinib. We then observed that evobrutinib, fenebrutinib, and tolebrutinib achieved similar levels of exposure in non-human primate CSF after oral doses of 10 mg/kg. However, tolebrutinib CSF exposure (4.8 ng/mL) (kp,uu CSF=0.40) exceeded the IC90 (the estimated concentration inhibiting 90% of kinase activity) value, while evobrutinib (3.2 ng/mL) (kp,uu CSF=0.13) and fenebrutinib (12.9 ng/mL) (kp,uu CSF=0.15) failed to reach the estimated IC90 values. CONCLUSIONS Tolebrutinib was the only candidate of the three that attained relevant CSF exposure in non-human primates.
Collapse
|
31
|
Tahmasebi F, Asl ER, Vahidinia Z, Faghihi F, Barati S. The comparative effects of bone marrow mesenchymal stem cells and supernatant transplantation on demyelination and inflammation in cuprizone model. Mol Biol Rep 2024; 51:674. [PMID: 38787497 DOI: 10.1007/s11033-024-09628-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) with inflammation and immune dysfunction. OBJECTIVES We compared the remyelination and immunomodulation properties of mesenchymal stem cells (MSCs) with their conditioned medium (CM) in the cuprizone model. METHODS Twenty-four C57BL/ 6 mice were divided into four groups. After cuprizone demyelination, MSCs and their CM were injected into the right lateral ventricle of mice. The expression level of IL-1β, TNF-α, and BDNF genes was evaluated using the qRT-PCR. APC antibody was used to assess the oligodendrocyte population using the immunofluorescent method. The remyelination and axonal repair were studied by specific staining of the LFB and electron microscopy techniques. RESULTS Transplantation of MSCs and CM increased the expression of the BDNF gene and decreased the expression of IL-1β and TNF-α genes compared to the cuprizone group, and these effects in the cell group were more than CM. Furthermore, cell transplantation resulted in a significant improvement in myelination and axonal repair, which was measured by luxol fast blue and transmission electron microscope images. The cell group had a higher number of oligodendrocytes than other groups. CONCLUSIONS According to the findings, injecting MSCs intraventricularly versus cell-conditioned medium can be a more effective approach to improving chronic demyelination in degenerative diseases like MS.
Collapse
Affiliation(s)
- Fatemeh Tahmasebi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elmira Roshani Asl
- Department of Biochemistry, Saveh University of Medical Sciences, Saveh, Iran
| | - Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Pad Nahad Tabiat Company, Ltd, Tehran, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran.
| |
Collapse
|
32
|
Bachmann H, Vandemoortele B, Vermeirssen V, Carrette E, Vonck K, Boon P, Raedt R, Laureys G. Vagus nerve stimulation enhances remyelination and decreases innate neuroinflammation in lysolecithin-induced demyelination. Brain Stimul 2024; 17:575-587. [PMID: 38648972 DOI: 10.1016/j.brs.2024.04.012] [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: 01/23/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Current treatments for Multiple Sclerosis (MS) poorly address chronic innate neuroinflammation nor do they offer effective remyelination. The vagus nerve has a strong regulatory role in inflammation and Vagus Nerve Stimulation (VNS) has potential to affect both neuroinflammation and remyelination in MS. OBJECTIVE This study investigated the effects of VNS on demyelination and innate neuroinflammation in a validated MS rodent model. METHODS Lysolecithin (LPC) was injected in the corpus callosum (CC) of 46 Lewis rats, inducing a demyelinated lesion. 33/46 rats received continuously-cycled VNS (cVNS) or one-minute per day VNS (1minVNS) or sham VNS from 2 days before LPC-injection until perfusion at 3 days post-injection (dpi) (corresponding with a demyelinated lesion with peak inflammation). 13/46 rats received cVNS or sham from 2 days before LPC-injection until perfusion at 11 dpi (corresponding with a partial remyelinated lesion). Immunohistochemistry and proteomics analyses were performed to investigate the extend of demyelination and inflammation. RESULTS Immunohistochemistry showed that cVNS significantly reduced microglial and astrocytic activation in the lesion and lesion border, and significantly reduced the Olig2+ cell count at 3 dpi. Furthermore, cVNS significantly improved remyelination with 57.4 % versus sham at 11 dpi. Proteomic gene set enrichment analyses showed increased activation of (glutamatergic) synapse pathways in cVNS versus sham, most pronounced at 3 dpi. CONCLUSION cVNS improved remyelination of an LPC-induced lesion. Possible mechanisms might include modulation of microglia and astrocyte activity, increased (glutamatergic) synapses and enhanced oligodendrocyte clearance after initial injury.
Collapse
Affiliation(s)
- Helen Bachmann
- Ghent University, 4 Brain, Department of Neurology, Ghent University Hospital, Belgium.
| | - Boris Vandemoortele
- Laboratory for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Vanessa Vermeirssen
- Laboratory for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Evelien Carrette
- Ghent University, 4 Brain, Department of Neurology, Ghent University Hospital, Belgium
| | - Kristl Vonck
- Ghent University, 4 Brain, Department of Neurology, Ghent University Hospital, Belgium
| | - Paul Boon
- Ghent University, 4 Brain, Department of Neurology, Ghent University Hospital, Belgium
| | - Robrecht Raedt
- Ghent University, 4 Brain, Department of Neurology, Ghent University Hospital, Belgium
| | - Guy Laureys
- Ghent University, 4 Brain, Department of Neurology, Ghent University Hospital, Belgium
| |
Collapse
|
33
|
Muzio L, Perego J. CNS Resident Innate Immune Cells: Guardians of CNS Homeostasis. Int J Mol Sci 2024; 25:4865. [PMID: 38732082 PMCID: PMC11084235 DOI: 10.3390/ijms25094865] [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: 03/21/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Although the CNS has been considered for a long time an immune-privileged organ, it is now well known that both the parenchyma and non-parenchymal tissue (meninges, perivascular space, and choroid plexus) are richly populated in resident immune cells. The advent of more powerful tools for multiplex immunophenotyping, such as single-cell RNA sequencing technique and upscale multiparametric flow and mass spectrometry, helped in discriminating between resident and infiltrating cells and, above all, the different spectrum of phenotypes distinguishing border-associated macrophages. Here, we focus our attention on resident innate immune players and their primary role in both CNS homeostasis and pathological neuroinflammation and neurodegeneration, two key interconnected aspects of the immunopathology of multiple sclerosis.
Collapse
Affiliation(s)
- Luca Muzio
- Neuroimmunology Lab, IRCCS San Raffaele Scientific Institute, Institute of Experimental Neurology, 20133 Milan, Italy;
| | | |
Collapse
|
34
|
Geladaris A, Torke S, Saberi D, Alankus YB, Streit F, Zechel S, Stadelmann-Nessler C, Fischer A, Boschert U, Häusler D, Weber MS. BTK inhibition limits microglia-perpetuated CNS inflammation and promotes myelin repair. Acta Neuropathol 2024; 147:75. [PMID: 38656399 PMCID: PMC11043151 DOI: 10.1007/s00401-024-02730-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/28/2024] [Accepted: 04/06/2024] [Indexed: 04/26/2024]
Abstract
In multiple sclerosis (MS), persisting disability can occur independent of relapse activity or development of new central nervous system (CNS) inflammatory lesions, termed chronic progression. This process occurs early and it is mostly driven by cells within the CNS. One promising strategy to control progression of MS is the inhibition of the enzyme Bruton's tyrosine kinase (BTK), which is centrally involved in the activation of both B cells and myeloid cells, such as macrophages and microglia. The benefit of BTK inhibition by evobrutinib was shown as we observed reduced pro-inflammatory activation of microglia when treating chronic experimental autoimmune encephalomyelitis (EAE) or following the adoptive transfer of activated T cells. Additionally, in a model of toxic demyelination, evobrutinib-mediated BTK inhibition promoted the clearance of myelin debris by microglia, leading to an accelerated remyelination. These findings highlight that BTK inhibition has the potential to counteract underlying chronic progression of MS.
Collapse
Affiliation(s)
- Anastasia Geladaris
- Institute of Neuropathology, University Medical Center, Georg August University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, Göttingen, Germany
| | - Sebastian Torke
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Darius Saberi
- Fraunhofer Institute for Translational Medicine and Pharmacology, Göttingen, Germany
- Department of Neurology, University Medical Center, Georg August University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | | | - Frank Streit
- Department of Clinical Chemistry, University Medical Center, Göttingen, Germany
| | - Sabrina Zechel
- Institute of Neuropathology, University Medical Center, Georg August University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Christine Stadelmann-Nessler
- Institute of Neuropathology, University Medical Center, Georg August University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Andreas Fischer
- Department of Clinical Chemistry, University Medical Center, Göttingen, Germany
| | - Ursula Boschert
- Ares Trading SA, Eysins, Switzerland
- Merck KGaA, Darmstadt, Germany
| | - Darius Häusler
- Institute of Neuropathology, University Medical Center, Georg August University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, Göttingen, Germany
| | - Martin S Weber
- Institute of Neuropathology, University Medical Center, Georg August University, Robert-Koch-Str. 40, 37075, Göttingen, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology, Göttingen, Germany.
- Department of Neurology, University Medical Center, Georg August University, Robert-Koch-Str. 40, 37075, Göttingen, Germany.
| |
Collapse
|
35
|
Luo M, Zhao F, Cheng H, Su M, Wang Y. Macrophage polarization: an important role in inflammatory diseases. Front Immunol 2024; 15:1352946. [PMID: 38660308 PMCID: PMC11039887 DOI: 10.3389/fimmu.2024.1352946] [Citation(s) in RCA: 73] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Macrophages are crucial cells in the human body's innate immunity and are engaged in a variety of non-inflammatory reactions. Macrophages can develop into two kinds when stimulated by distinct internal environments: pro-inflammatory M1-like macrophages and anti-inflammatory M2-type macrophages. During inflammation, the two kinds of macrophages are activated alternatively, and maintaining a reasonably steady ratio is critical for maintaining homeostasis in vivo. M1 macrophages can induce inflammation, but M2 macrophages suppress it. The imbalance between the two kinds of macrophages will have a significant impact on the illness process. As a result, there are an increasing number of research being conducted on relieving or curing illnesses by altering the amount of macrophages. This review summarizes the role of macrophage polarization in various inflammatory diseases, including autoimmune diseases (RA, EAE, MS, AIH, IBD, CD), allergic diseases (allergic rhinitis, allergic dermatitis, allergic asthma), atherosclerosis, obesity and type 2 diabetes, metabolic homeostasis, and the compounds or drugs that have been discovered or applied to the treatment of these diseases by targeting macrophage polarization.
Collapse
Affiliation(s)
| | | | | | | | - Yuanmin Wang
- The Third Affiliated Hospital of Zunyi Medical University, The First People’s Hospital of Zunyi, Zunyi, Guizhou, China
| |
Collapse
|
36
|
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
|
37
|
Sun R, Jiang H. Border-associated macrophages in the central nervous system. J Neuroinflammation 2024; 21:67. [PMID: 38481312 PMCID: PMC10938757 DOI: 10.1186/s12974-024-03059-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Tissue-resident macrophages play an important role in the local maintenance of homeostasis and immune surveillance. In the central nervous system (CNS), brain macrophages are anatomically divided into parenchymal microglia and non-parenchymal border-associated macrophages (BAMs). Among these immune cell populations, microglia have been well-studied for their roles during development as well as in health and disease. BAMs, mostly located in the choroid plexus, meningeal and perivascular spaces, are now gaining increased attention due to advancements in multi-omics technologies and genetic methodologies. Research on BAMs over the past decade has focused on their ontogeny, immunophenotypes, involvement in various CNS diseases, and potential as therapeutic targets. Unlike microglia, BAMs display mixed origins and distinct self-renewal capacity. BAMs are believed to regulate neuroimmune responses associated with brain barriers and contribute to immune-mediated neuropathology. Notably, BAMs have been observed to function in diverse cerebral pathologies, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, ischemic stroke, and gliomas. The elucidation of the heterogeneity and diverse functions of BAMs during homeostasis and neuroinflammation is mesmerizing, since it may shed light on the precision medicine that emphasizes deep insights into programming cues in the unique brain immune microenvironment. In this review, we delve into the latest findings on BAMs, covering aspects like their origins, self-renewal capacity, adaptability, and implications in different brain disorders.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave., Box 8057, St. Louis, MO, 63110, USA.
| | - Haowu Jiang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave., CB 8054, St. Louis, MO, 63110, USA.
| |
Collapse
|
38
|
Mubaraki AA, Alnemari MA, Aljuaid SO, Altalhi FM, Alamri YM, Altowairqi SO. The Prevalence and Impact of Urinary Incontinence on Multiple Sclerosis Patients in Taif City, Saudi Arabia. Cureus 2024; 16:e57010. [PMID: 38681390 PMCID: PMC11046167 DOI: 10.7759/cureus.57010] [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] [Accepted: 03/26/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction Multiple sclerosis (MS) is a chronic inflammatory disease that results in demyelination and progressive loss of nerve cells within the central nervous system. Multiple sclerosis, as well as other neurological diseases that impact brain structures and spinal pathways involved in sphincter control, may cause lower urinary tract symptoms (LUTS) Our aims are to determine the prevalence, severity, and impact on the quality of life of urinary incontinence among MS patients in Taif, Saudi Arabia, as well as its potential association with demographics and clinical features. Method A cross-sectional study included 150 of MS patients aged 18 years and older who completed the validated Arabic versions of both the International Consultation on Incontinence Questionnaire Overactive Bladder (ICIQ-OAB) and the International Consultation on Incontinence Questionnaire-Urinary Incontinence Short Form (ICIQ-UI SF). In addition, other clinical parameters were collected from the medical records of patients. The data were analyzed using Statistical Package for the Social Sciences (SPSS) version 26 (IBM Inc., Armonk, New York). Qualitative data were expressed as numbers and percentages, and the Chi-squared test (χ2) was employed. Quantitative data were expressed as means with standard deviation (mean ± SD). Result 67.3% of the participants were female; the mean ICIQ UI and ICIQ OAB scores in these MS patients were found to be 6.30 ± 6.26 and 5.32 ± 3.76, respectively. They were significantly higher in progressive MS patients compared to relapsing-remitting MS patients (p<0.05). There was a high positive correlation obtained between ICIQ UI and ICIQ OAB scores (rho=0.801, p<0.001). Conclusion The findings of this study showed that urinary incontinence was a common and distressing symptom experienced by individuals with MS. The severity of UI symptoms was significantly more in progressive multiple sclerosis compared to relapsing-remitting multiple sclerosis.
Collapse
|
39
|
Lombardi M, Scaroni F, Gabrielli M, Raffaele S, Bonfanti E, Filipello F, Giussani P, Picciolini S, de Rosbo NK, Uccelli A, Golia MT, D’Arrigo G, Rubino T, Hooshmand K, Legido-Quigley C, Fenoglio C, Gualerzi A, Fumagalli M, Verderio C. Extracellular vesicles released by microglia and macrophages carry endocannabinoids which foster oligodendrocyte differentiation. Front Immunol 2024; 15:1331210. [PMID: 38464529 PMCID: PMC10921360 DOI: 10.3389/fimmu.2024.1331210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/01/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction Microglia and macrophages can influence the evolution of myelin lesions through the production of extracellular vesicles (EVs). While microglial EVs promote in vitro differentiation of oligodendrocyte precursor cells (OPCs), whether EVs derived from macrophages aid or limit OPC maturation is unknown. Methods Immunofluorescence analysis for the myelin protein MBP was employed to evaluate the impact of EVs from primary rat macrophages on cultured OPC differentiation. Raman spectroscopy and liquid chromatography-mass spectrometry was used to define the promyelinating lipid components of myelin EVs obtained in vitro and isolated from human plasma. Results and discussion Here we show that macrophage-derived EVs do not promote OPC differentiation, and those released from macrophages polarized towards an inflammatory state inhibit OPC maturation. However, their lipid cargo promotes OPC maturation in a similar manner to microglial EVs. We identify the promyelinating endocannabinoids anandamide and 2-arachidonoylglycerol in EVs released by both macrophages and microglia in vitro and circulating in human plasma. Analysis of OPC differentiation in the presence of the endocannabinoid receptor antagonists SR141716A and AM630 reveals a key role of vesicular endocannabinoids in OPC maturation. From this study, EV-associated endocannabinoids emerge as important mediators in microglia/macrophage-oligodendrocyte crosstalk, which may be exploited to enhance myelin repair.
Collapse
Affiliation(s)
- Marta Lombardi
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Federica Scaroni
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
| | - Martina Gabrielli
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - Elisabetta Bonfanti
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - Fabia Filipello
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Segrate, Italy
| | - Silvia Picciolini
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Nicole Kerlero de Rosbo
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
- TomaLab, Institute of Nanotechnology, CNR, Rome, Italy
| | - Antonio Uccelli
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Maria Teresa Golia
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Giulia D’Arrigo
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences (DBSV) and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - Kourosh Hooshmand
- System Medicine, Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Cristina Legido-Quigley
- System Medicine, Steno Diabetes Center Copenhagen, Copenhagen, Denmark
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Chiara Fenoglio
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
- Fondazione Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Alice Gualerzi
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - Claudia Verderio
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
40
|
Sun R, Jiang H. Border-associated macrophages in the central nervous system. Clin Immunol 2024:109921. [PMID: 38316202 DOI: 10.1016/j.clim.2024.109921] [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: 12/13/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024]
Abstract
Tissue-resident macrophages play an important role in the local maintenance of homeostasis and immune surveillance. In the central nervous system (CNS), brain macrophages are anatomically divided into parenchymal microglia and non-parenchymal border-associated macrophages (BAMs). Among these immune cell populations, microglia have been well-studied for their roles in normal brain development, neurodegeneration, and brain cancers. BAMs, mostly located in the choroid plexus, meningeal and perivascular spaces, are now gaining increased attention due to advancements in multi-omics technologies and genetic methodologies. Research on BAMs over the past decade has focused on their ontogeny, immunophenotypes, involvement in various CNS diseases, and potential as therapeutic targets. Unlike microglia, BAMs display mixed origins and distinct self-renewal capacity. BAMs are believed to regulate neuroimmune responses associated with brain barriers and contribute to immune-mediated neuropathology. Notably, BAMs have been observed to function in diverse cerebral pathologies, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, ischemic stroke, and gliomas. The elucidation of the heterogeneity and diverse functions of BAMs during homeostasis and neuroinflammation is mesmerizing, since it may shed light on the precision medicine that emphasizes deep insights into programming cues in the unique brain immune microenvironment. In this review, we delve into the latest findings on BAMs, covering aspects like their origins, self-renewal capacity, adaptability, and implications in different brain disorders.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| | - Haowu Jiang
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine in St Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
41
|
Nguyen KL, Bhatt IJ, Gupta S, Showkat N, Swanson KA, Fischer R, Kontermann RE, Pfizenmaier K, Bracchi-Ricard V, Bethea JR. Tumor necrosis factor receptor 2 activation elicits sex-specific effects on cortical myelin proteins and functional recovery in a model of multiple sclerosis. Brain Res Bull 2024; 207:110885. [PMID: 38246200 PMCID: PMC10923072 DOI: 10.1016/j.brainresbull.2024.110885] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
Multiple sclerosis (MS), a demyelinating autoimmune disease of the central nervous system (CNS), predominately affects females compared to males. Tumor necrosis factor (TNF), a pro-inflammatory cytokine, signaling through TNF receptor 1 contributes to inflammatory disease pathogenesis. In contrast, TNF receptor 2 signaling is neuroprotective. Current anti-TNF MS therapies are shown to be detrimental to patients due to pleiotropic effects on both pro- and anti-inflammatory functions. Using a non-pertussis toxin (nPTX) experimental autoimmune encephalomyelitis (EAE) model in C57BL/6 mice, we systemically administered a TNFR2 agonist (p53-sc-mTNFR2) to investigate behavioral and pathophysiological changes in both female and male mice. Our data shows that TNFR2 activation alleviates motor and sensory symptoms in females. However, in males, the agonist only alleviates sensory symptoms and not motor. nPTX EAE induction in TNFR2 global knockout mice caused exacerbated motor symptoms in females along with an earlier day of onset, but not in males. Our data demonstrates that TNFR2 agonist efficacy is sex-specific for alleviation of motor symptoms, however, it effectively reduces mechanical hypersensitivity in both females and males. Altogether, these data support the therapeutic promise TNFR2 agonism holds as an MS therapeutic and, more broadly, to treat central neuropathic pain.
Collapse
MESH Headings
- Humans
- Male
- Female
- Mice
- Animals
- Multiple Sclerosis
- Receptors, Tumor Necrosis Factor, Type II/agonists
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Receptors, Tumor Necrosis Factor, Type II/therapeutic use
- Tumor Necrosis Factor Inhibitors/therapeutic use
- Mice, Inbred C57BL
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Myelin Proteins
- Tumor Necrosis Factor-alpha/metabolism
- Mice, Knockout
Collapse
Affiliation(s)
- Kayla L Nguyen
- Department of Anatomy and Cell Biology, The George Washington University, Washington, DC 20052, United States.
| | - Ishaan J Bhatt
- Department of Biology, Drexel University, Philadelphia, PA 19104, United States
| | - Shruti Gupta
- Department of Biology, Drexel University, Philadelphia, PA 19104, United States
| | - Nazaf Showkat
- Department of Biology, Drexel University, Philadelphia, PA 19104, United States
| | - Kathryn A Swanson
- Department of Biology, Drexel University, Philadelphia, PA 19104, United States
| | - Roman Fischer
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany; Stuttgart Research Center Systems Biology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany; Stuttgart Research Center Systems Biology, University of Stuttgart, 70569 Stuttgart, Germany
| | | | - John R Bethea
- Department of Anatomy and Cell Biology, The George Washington University, Washington, DC 20052, United States.
| |
Collapse
|
42
|
Ban M, Bredikhin D, Huang Y, Bonder MJ, Katarzyna K, Oliver AJ, Wilson NK, Coupland P, Hadfield J, Göttgens B, Madissoon E, Stegle O, Sawcer S. Expression profiling of cerebrospinal fluid identifies dysregulated antiviral mechanisms in multiple sclerosis. Brain 2024; 147:554-565. [PMID: 38038362 PMCID: PMC10834244 DOI: 10.1093/brain/awad404] [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: 08/04/2023] [Revised: 11/06/2023] [Accepted: 11/18/2023] [Indexed: 12/02/2023] Open
Abstract
Despite the overwhelming evidence that multiple sclerosis is an autoimmune disease, relatively little is known about the precise nature of the immune dysregulation underlying the development of the disease. Reasoning that the CSF from patients might be enriched for cells relevant in pathogenesis, we have completed a high-resolution single-cell analysis of 96 732 CSF cells collected from 33 patients with multiple sclerosis (n = 48 675) and 48 patients with other neurological diseases (n = 48 057). Completing comprehensive cell type annotation, we identified a rare population of CD8+ T cells, characterized by the upregulation of inhibitory receptors, increased in patients with multiple sclerosis. Applying a Multi-Omics Factor Analysis to these single-cell data further revealed that activity in pathways responsible for controlling inflammatory and type 1 interferon responses are altered in multiple sclerosis in both T cells and myeloid cells. We also undertook a systematic search for expression quantitative trait loci in the CSF cells. Of particular interest were two expression quantitative trait loci in CD8+ T cells that were fine mapped to multiple sclerosis susceptibility variants in the viral control genes ZC3HAV1 (rs10271373) and IFITM2 (rs1059091). Further analysis suggests that these associations likely reflect genetic effects on RNA splicing and cell-type specific gene expression respectively. Collectively, our study suggests that alterations in viral control mechanisms might be important in the development of multiple sclerosis.
Collapse
Affiliation(s)
- Maria Ban
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Danila Bredikhin
- European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Yuanhua Huang
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge CB10 1SD, UK
| | - Marc Jan Bonder
- European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Kania Katarzyna
- University of Cambridge, CRUK Cambridge Institute, Cambridge CB2 0RE, UK
| | - Amanda J Oliver
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Nicola K Wilson
- Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Paul Coupland
- University of Cambridge, CRUK Cambridge Institute, Cambridge CB2 0RE, UK
| | - James Hadfield
- University of Cambridge, CRUK Cambridge Institute, Cambridge CB2 0RE, UK
| | - Berthold Göttgens
- Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Elo Madissoon
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge CB10 1SD, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Oliver Stegle
- European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge CB10 1SD, UK
| | - Stephen Sawcer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
43
|
Miscioscia A, Treaba CA, Barletta VT, Herranz E, Sloane JA, Barbuti E, Mainero C. White matter paramagnetic rim and non-rim lesions share a periventricular gradient in multiple sclerosis: A 7-T imaging study. Mult Scler 2024; 30:166-176. [PMID: 38279672 PMCID: PMC10922980 DOI: 10.1177/13524585231224681] [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: 01/28/2024]
Abstract
BACKGROUND Paramagnetic rim white matter (WM) lesions (PRL) are thought to be a main driver of non-relapsing multiple sclerosis (MS) progression. It is unknown whether cerebrospinal fluid (CSF)-soluble factors diffusing from the ventricles contribute to PRL formation. OBJECTIVE To investigate the distribution of PRL and non-rim brain WM lesions as a function of distance from ventricular CSF, their relationship with cortical lesions, the contribution of lesion phenotype, and localization to neurological disability. METHODS Lesion count and volume of PRL, non-rim WM, leukocortical lesion (LCL), and subpial/intracortical lesions were obtained at 7-T. The brain WM was divided into 1-mm-thick concentric rings radiating from the ventricles to extract PRL and non-rim WM lesion volume from each ring. RESULTS In total, 61 MS patients with ⩾1 PRL were included in the study. Both PRL and non-rim WM lesion volumes were the highest in the periventricular WM and declined with increasing distance from ventricles. A CSF distance-independent association was found between non-rim WM lesions, PRL, and LCL, but not subpial/intracortical lesions. Periventricular non-rim WM lesion volume was the strongest predictor of neurological disability. CONCLUSIONS Non-rim and PRL share a gradient of distribution from the ventricles toward the cortex, suggesting that CSF proximity equally impacts the prevalence of both lesion phenotypes.
Collapse
Affiliation(s)
- Alessandro Miscioscia
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- University of Padova, Padova, Italy
| | - Constantina A. Treaba
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Valeria T. Barletta
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elena Herranz
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jacob A. Sloane
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Elena Barbuti
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Ospedale Sant’Andrea, University La Sapienza, Rome, Italy
| | - Caterina Mainero
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Etebar F, Harkin DG, White AR, Dando SJ. Non-invasive in vivo imaging of brain and retinal microglia in neurodegenerative diseases. Front Cell Neurosci 2024; 18:1355557. [PMID: 38348116 PMCID: PMC10859418 DOI: 10.3389/fncel.2024.1355557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 02/15/2024] Open
Abstract
Microglia play crucial roles in immune responses and contribute to fundamental biological processes within the central nervous system (CNS). In neurodegenerative diseases, microglia undergo functional changes and can have both protective and pathogenic roles. Microglia in the retina, as an extension of the CNS, have also been shown to be affected in many neurological diseases. While our understanding of how microglia contribute to pathological conditions is incomplete, non-invasive in vivo imaging of brain and retinal microglia in living subjects could provide valuable insights into their role in the neurodegenerative diseases and open new avenues for diagnostic biomarkers. This mini-review provides an overview of the current brain and retinal imaging tools for studying microglia in vivo. We focus on microglia targets, the advantages and limitations of in vivo microglia imaging approaches, and applications for evaluating the pathogenesis of neurological conditions, such as Alzheimer's disease and multiple sclerosis.
Collapse
Affiliation(s)
- Fazeleh Etebar
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Damien G. Harkin
- Centre for Vision and Eye Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Anthony R. White
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Samantha J. Dando
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Centre for Vision and Eye Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| |
Collapse
|
45
|
DePaula-Silva AB. The Contribution of Microglia and Brain-Infiltrating Macrophages to the Pathogenesis of Neuroinflammatory and Neurodegenerative Diseases during TMEV Infection of the Central Nervous System. Viruses 2024; 16:119. [PMID: 38257819 PMCID: PMC10819099 DOI: 10.3390/v16010119] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The infection of the central nervous system (CNS) with neurotropic viruses induces neuroinflammation and is associated with the development of neuroinflammatory and neurodegenerative diseases, including multiple sclerosis and epilepsy. The activation of the innate and adaptive immune response, including microglial, macrophages, and T and B cells, while required for efficient viral control within the CNS, is also associated with neuropathology. Under healthy conditions, resident microglia play a pivotal role in maintaining CNS homeostasis. However, during pathological events, such as CNS viral infection, microglia become reactive, and immune cells from the periphery infiltrate into the brain, disrupting CNS homeostasis and contributing to disease development. Theiler's murine encephalomyelitis virus (TMEV), a neurotropic picornavirus, is used in two distinct mouse models: TMEV-induced demyelination disease (TMEV-IDD) and TMEV-induced seizures, representing mouse models of multiple sclerosis and epilepsy, respectively. These murine models have contributed substantially to our understanding of the pathophysiology of MS and seizures/epilepsy following viral infection, serving as critical tools for identifying pharmacological targetable pathways to modulate disease development. This review aims to discuss the host-pathogen interaction during a neurotropic picornavirus infection and to shed light on our current understanding of the multifaceted roles played by microglia and macrophages in the context of these two complexes viral-induced disease.
Collapse
Affiliation(s)
- Ana Beatriz DePaula-Silva
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
46
|
Dias-Carvalho A, Sá SI, Carvalho F, Fernandes E, Costa VM. Inflammation as common link to progressive neurological diseases. Arch Toxicol 2024; 98:95-119. [PMID: 37964100 PMCID: PMC10761431 DOI: 10.1007/s00204-023-03628-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023]
Abstract
Life expectancy has increased immensely over the past decades, bringing new challenges to the health systems as advanced age increases the predisposition for many diseases. One of those is the burden of neurologic disorders. While many hypotheses have been placed to explain aging mechanisms, it has been widely accepted that the increasing pro-inflammatory status with advanced age or "inflammaging" is a main determinant of biological aging. Furthermore, inflammaging is at the cornerstone of many age-related diseases and its involvement in neurologic disorders is an exciting hypothesis. Indeed, aging and neurologic disorders development in the elderly seem to share some basic pathways that fundamentally converge on inflammation. Peripheral inflammation significantly influences brain function and contributes to the development of neurological disorders, including Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Understanding the role of inflammation in the pathogenesis of progressive neurological diseases is of crucial importance for developing effective treatments and interventions that can slow down or prevent disease progression, therefore, decreasing its social and economic burden.
Collapse
Affiliation(s)
- Ana Dias-Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO- Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| | - Susana Isabel Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO- Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO- Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
47
|
Gadhave DG, Sugandhi VV, Kokare CR. Potential biomaterials and experimental animal models for inventing new drug delivery approaches in the neurodegenerative disorder: Multiple sclerosis. Brain Res 2024; 1822:148674. [PMID: 37952871 DOI: 10.1016/j.brainres.2023.148674] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
The tight junction of endothelial cells in the central nervous system (CNS) has an ideal characteristic, acting as a biological barrier that can securely regulate the movement of molecules in the brain. Tightly closed astrocyte cell junctions on blood capillaries are the blood-brain barrier (BBB). This biological barrier prohibits the entry of polar drugs, cells, and ions, which protect the brain from harmful toxins. However, delivering any therapeutic agent to the brain in neurodegenerative disorders (i.e., schizophrenia, multiple sclerosis, etc.) is extremely difficult. Active immune responses such as microglia, astrocytes, and lymphocytes cross the BBB and attack the nerve cells, which causes the demyelination of neurons. Therefore, there is a hindrance in transmitting electrical signals properly, resulting in blindness, paralysis, and neuropsychiatric problems. The main objective of this article is to shed light on the performance of biomaterials, which will help researchers to create nanocarriers that can cross the blood-brain barrier and achieve a therapeutic concentration of drugs in the CNS of patients with multiple sclerosis (MS). The present review focuses on the importance of biomaterials with diagnostic and therapeutic efficacy that can help enhance multiple sclerosis therapeutic potential. Currently, the development of MS in animal models is limited by immune responses, which prevent MS induction in healthy animals. Therefore, this article also showcases animal models currently used for treating MS. A future advance in developing a novel effective strategy for treating MS is now a potential area of research.
Collapse
Affiliation(s)
- Dnyandev G Gadhave
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA; Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune 413130, Maharashtra, India.
| | - Vrashabh V Sugandhi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Chandrakant R Kokare
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India
| |
Collapse
|
48
|
Hammond BP, Panda SP, Kaushik DK, Plemel JR. Microglia and Multiple Sclerosis. ADVANCES IN NEUROBIOLOGY 2024; 37:445-456. [PMID: 39207707 DOI: 10.1007/978-3-031-55529-9_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Multiple sclerosis (MS) is a devastating autoimmune disease that leads to profound disability. This disability arises from the stochastic, regional loss of myelin-the insulating sheath surrounding neurons-in the central nervous system (CNS). The demyelinated regions are dominated by the brain's resident macrophages: microglia. Microglia perform a variety of functions in MS and are thought to initiate and perpetuate demyelination through their interactions with peripheral immune cells that traffic into the brain. However, microglia are also likely essential for recruiting and promoting the differentiation of cells that can restore lost myelin in a process known as remyelination. Given these seemingly opposing functions, an overarching beneficial or detrimental role is yet to be ascribed to these immune cells. In this chapter, we will discuss microglia dynamics throughout the MS disease course and probe the apparent dichotomy of microglia as the drivers of both demyelination and remyelination.
Collapse
Affiliation(s)
- Brady P Hammond
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Sharmistha P Panda
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Deepak K Kaushik
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
49
|
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]
|
50
|
Berek K, Hegen H, Auer M, Barket R, Di Pauli F, Hocher J, Krajnc N, Zinganell A, Deisenhammer F, Berger T, Bsteh G. Odour discrimination and identification as a biomarker of long-term disability worsening in multiple sclerosis. Mult Scler 2024; 30:55-62. [PMID: 37850472 PMCID: PMC10782652 DOI: 10.1177/13524585231201093] [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: 06/04/2023] [Revised: 08/06/2023] [Accepted: 08/23/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Odour discrimination and identification (DI) are markers associated with disability worsening and neuroaxonal damage in multiple sclerosis (MS). OBJECTIVE The main objective of this research is to investigate whether longitudinal change of DI predicts long-term MS disease course. METHODS This is a 6-year prospective longitudinal study on MS patients at the MS Clinic Innsbruck. Clinical, bi-annual visits assessed patients' history and Expanded Disability Status Scale (EDSS) score. DI and cognitive function were assessed at baseline (BL), Year 1 (Y1), Year 2 (Y2) and Year 6 (Y6) by the 'Sniffin' Sticks'/Symbol Digit Modalities Test. RESULTS Around 92 of 139 patients were available for Y6 follow-up. Mean DI scores significantly decreased over time (BL = 27.8, Y1 = 27.5, Y2 = 26.3 and Y6 = 26.3; p < 0.001) and negatively correlated with patients' age (rs = -0.120, p = 0.032) and disease duration (rs = -0.103, p = 0.041). Multivariable regression analyses revealed that lower absolute DI scores and larger DI score loss over time were associated with higher probability of EDSS worsening (per -1 point: hazard ratio (HR) = 1.40 (1.16-1.68) and 2.34 (1.27-4.21)), progression independent of relapse activity (PIRA) (HR = 1.49 (1.20-1.85) and 2.22 (1.33-3.31)) and cognitive deterioration (HR = 1.75 (1.35-2.27) and 4.29 (1.26-2.84)) at Y6, but not with time to first relapse. CONCLUSION Odour DI is an irreversible marker of neuroaxonal damage, associated with PIRA, cognitive deterioration and EDSS worsening.
Collapse
Affiliation(s)
- Klaus Berek
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Harald Hegen
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Auer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Barket
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Franziska Di Pauli
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jakob Hocher
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nik Krajnc
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Anne Zinganell
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Gabriel Bsteh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|