1
|
Serneels PJ, De Schutter JD, De Groef L, Moons L, Bergmans S. Oligodendroglial heterogeneity in health, disease, and recovery: deeper insights into myelin dynamics. Neural Regen Res 2025; 20:3179-3192. [PMID: 39665821 DOI: 10.4103/nrr.nrr-d-24-00694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/17/2024] [Indexed: 12/13/2024] Open
Abstract
Decades of research asserted that the oligodendroglial lineage comprises two cell types: oligodendrocyte precursor cells and oligodendrocytes. However, recent studies employing single-cell RNA sequencing techniques have uncovered novel cell states, prompting a revision of the existing terminology. Going forward, the oligodendroglial lineage should be delineated into five distinct cell states: oligodendrocyte precursor cells, committed oligodendrocyte precursor cells, newly formed oligodendrocytes, myelin-forming oligodendrocytes, and mature oligodendrocytes. This new classification system enables a deeper understanding of the oligodendroglia in both physiological and pathological contexts. Adopting this uniform terminology will facilitate comparison and integration of data across studies. This, including the consolidation of findings from various demyelinating models, is essential to better understand the pathogenesis of demyelinating diseases. Additionally, comparing injury models across species with varying regenerative capacities can provide insights that may lead to new therapeutic strategies to overcome remyelination failure. Thus, by standardizing terminology and synthesizing data from diverse studies across different animal models, we can enhance our understanding of myelin pathology in central nervous system disorders such as multiple sclerosis, Alzheimer's disease, and amyotrophic lateral sclerosis, all of which involve oligodendroglial and myelin dysfunction.
Collapse
Affiliation(s)
- Pieter-Jan Serneels
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Julie D De Schutter
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Lies De Groef
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Cellular Communication & Neurodegeneration Research Group, Leuven, Belgium
| | - Lieve Moons
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Steven Bergmans
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| |
Collapse
|
2
|
Wang Z, Wang X, Li P, Xia H, Yang X. Genetic associations between immune-related plasma proteins and neurodegenerative diseases. Neurol Res 2025:1-10. [PMID: 39743746 DOI: 10.1080/01616412.2024.2448745] [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: 05/28/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Immune dysregulation is commonly associated with neurodegenerative diseases (NDs), yet the underlying causes and mechanisms still require further investigation. OBJECTIVE This study investigates the correlation between immune-related plasma proteins and the risk of NDs by integrating genome-wide association study (GWAS) data for Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS) with plasma proteome analysis. METHODS By analyzing GWAS data for 4907 immune-related plasma proteins, this research evaluates the direct impact of plasma proteins on the risk of four NDs: AD, PD, ALS, and MS. Additionally, the study conducts an analysis of protein expression levels using single-cell RNA sequencing data. RESULTS We have identified plasma proteins that are closely associated with the risk of NDs. Using stringent criteria, we identified 88 proteins associated with AD, 115 with PD, 100 with ALS, and 87 with MS. Additionally, single-cell sequencing analyzed the protein expression and its distribution within different cell types in the brain. CONCLUSIONS Our research has demonstrated that plasma proteins may contribute to the risk of NDs, and it has also provided concrete evidence linking genetic susceptibility for these diseases to immune mechanisms. Furthermore, we found that specific proteins influence genetic variations linked to NDs risk via plasma-mediated regulation, emphasizing the importance of interactions between the brain and circulatory system.
Collapse
Affiliation(s)
- Zihao Wang
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Medical Research, Xinjiang Key Laboratory of Neurological Disease Research, Urumqi, Xinjiang, China
| | - Xiaobei Wang
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Medical Research, Xinjiang Key Laboratory of Neurological Disease Research, Urumqi, Xinjiang, China
| | - Peishan Li
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Medical Research, Xinjiang Key Laboratory of Neurological Disease Research, Urumqi, Xinjiang, China
| | - Huan Xia
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Medical Research, Xinjiang Key Laboratory of Neurological Disease Research, Urumqi, Xinjiang, China
| | - Xinling Yang
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Medical Research, Xinjiang Key Laboratory of Neurological Disease Research, Urumqi, Xinjiang, China
| |
Collapse
|
3
|
Noor L, Upadhyay A, Joshi V. Role of T Lymphocytes in Glioma Immune Microenvironment: Two Sides of a Coin. BIOLOGY 2024; 13:846. [PMID: 39452154 PMCID: PMC11505600 DOI: 10.3390/biology13100846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
Glioma is known for its immunosuppressive microenvironment, which makes it challenging to target through immunotherapies. Immune cells like macrophages, microglia, myeloid-derived suppressor cells, and T lymphocytes are known to infiltrate the glioma tumor microenvironment and regulate immune response distinctively. Among the variety of immune cells, T lymphocytes have highly complex and multifaceted roles in the glioma immune landscape. T lymphocytes, which include CD4+ helper and CD8+ cytotoxic T cells, are known for their pivotal roles in anti-tumor responses. However, these cells may behave differently in the highly dynamic glioma microenvironment, for example, via an immune invasion mechanism enforced by tumor cells. Therefore, T lymphocytes play dual roles in glioma immunity, firstly by their anti-tumor responses, and secondly by exploiting gliomas to promote immune invasion. As an immunosuppression strategy, glioma induces T-cell exhaustion and suppression of effector T cells by regulatory T cells (Tregs) or by altering their signaling pathways. Further, the expression of immune checkpoint inhibitors on the glioma cell surface leads to T cell anergy and dysfunction. Overall, this dynamic interplay between T lymphocytes and glioma is crucial for designing more effective immunotherapies. The current review provides detailed knowledge on the roles of T lymphocytes in the glioma immune microenvironment and helps to explore novel therapeutic approaches to reinvigorate T lymphocytes.
Collapse
Affiliation(s)
- Laiba Noor
- Department of Biotechnology, Bennett University, Greater Noida 201310, Uttar Pradesh, India
| | - Arun Upadhyay
- Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India
| | - Vibhuti Joshi
- Department of Biotechnology, Bennett University, Greater Noida 201310, Uttar Pradesh, India
| |
Collapse
|
4
|
Mallardo M, Mazzeo F, Lus G, Signoriello E, Daniele A, Nigro E. Impact of Lifestyle Interventions on Multiple Sclerosis: Focus on Adipose Tissue. Nutrients 2024; 16:3100. [PMID: 39339700 PMCID: PMC11434938 DOI: 10.3390/nu16183100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder characterized by demyelination in the central nervous system (CNS), affecting individuals globally. The pathological mechanisms underlying MS remain unclear, but current evidence suggests that inflammation and immune dysfunction play a critical role in the pathogenesis of MS disease. Adipose tissue (AT) is a dynamic multifunctional organ involved in various immune diseases, including MS, due to its endocrine function and the secretion of adipokines, which can influence inflammation and immune responses. Physical activity represents an efficacious non-pharmacological strategy for the management of a spectrum of conditions that not only improves inflammatory and immune functions but also directly affects the status and function of AT. Additionally, the exploration of nutritional supplementation represents an important field of MS research aimed at enhancing clinical symptoms and is closely tied to the regulation of metabolic responses, including adipokine secretion. This review, therefore, aims to elucidate the intricate relationship between lifestyle and MS by providing an overview of the latest published data about the involvement of AT and the main adipokines, such as adiponectin, leptin, and tumor necrosis factor α (TNFα) in the pathogenesis of MS. Furthermore, we explore whether physical activity and dietary management could serve as useful strategies to improve the quality of life of MS patients.
Collapse
Affiliation(s)
- Marta Mallardo
- Department of Molecular and Biotechnological Medicine, University of Naples "Federico II", 80138 Naples, Italy
- CEINGE-Biotechnologies Advances S.c.a r.l., Via G. Salvatore 486, 80145 Naples, Italy
| | - Filomena Mazzeo
- Department of Economics, Law, Cybersecurity and Sports Sciences (DiSEGIM), University of Naples "Parthenope", 80035 Naples, Italy
| | - Giacomo Lus
- Multiple Sclerosis Center, II Neurological Clinic, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
- Department of Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Elisabetta Signoriello
- Multiple Sclerosis Center, II Neurological Clinic, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
- Department of Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Aurora Daniele
- Department of Molecular and Biotechnological Medicine, University of Naples "Federico II", 80138 Naples, Italy
- CEINGE-Biotechnologies Advances S.c.a r.l., Via G. Salvatore 486, 80145 Naples, Italy
| | - Ersilia Nigro
- CEINGE-Biotechnologies Advances S.c.a r.l., Via G. Salvatore 486, 80145 Naples, Italy
- Department of Pharmaceutical, Biological, Environmental Sciences and Technologies, University of Campania "Luigi Vanvitelli", Via G. Vivaldi 42, 81100 Caserta, Italy
| |
Collapse
|
5
|
Chanpura A, Gupta RK, Sriwastava SK, Rahmig J. Diagnostic value of soluble Interleukin-2 receptor in patients suffering neurosarcoidosis: A systematic review. J Cent Nerv Syst Dis 2024; 16:11795735241274186. [PMID: 39193252 PMCID: PMC11348353 DOI: 10.1177/11795735241274186] [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: 02/19/2024] [Revised: 06/16/2024] [Accepted: 07/17/2024] [Indexed: 08/29/2024] Open
Abstract
Background Neurosarcoidosis is an inflammatory granulomatous disease. Up to 25% of occult sarcoidosis affecting the nervous system are only detected by autopsy. In addition, in recent years the suspicion arose that the soluble Interleukin-2 Receptor (sIL-2R) might be useful in differentiating between neurosarcoidosis and neurosarcoidosis-like diseases such as neurotuberculosis, multiple sclerosis, or cerebral lymphoma. Objectives Therefore, we aimed to systematically review randomized controlled trials (RCT), observational studies, and case-control studies evaluating sIL-2R levels in neurosarcoidosis patients. Design For this systematic review, a comprehensive literature search of electronic databases including EMBASE, The Web Of Science, The Cochrane Library, MEDLINE, and Google Scholar was conducted. The search was limited to the English language and publication date up to January 08th, 2024. Data Sources and Methods As part of the search strategy conducted, 6 articles met the inclusion criteria. Two independent reviewers extracted the relevant data from each article. In addition, 2 independent reviewers assessed the quality of each study using the Newcastle-Ottawa Scale (NOS). Results We included 6 studies comprising 98 patients suffering from neurosarcoidosis, 525 non-sarcoidosis patients, and 118 healthy controls. Included studies were published between 2010 and 2023. Cerebrospinal fluid (CSF) sIL-2R levels differed significantly between neurosarcoidosis patients and multiple sclerosis, vasculitis, and healthy controls whereas serum sIL-2R levels did not reveal sufficient discriminative power. sIL-2R index was able to discriminate neurosarcoidosis from neurotuberculosis, bacterial/viral meningitis, and healthy controls. Conclusions In this systematic review, we found indications that sIL-2R may be a useful biomarker for the diagnosis of neurosarcoidosis. To determine an additional diagnostic value of sIL-2R, large prospective studies are needed that not only examine absolute sIL-2R levels in serum or CSF but also the dynamic changes as well as the implications of renal function on sIL-2R levels.
Collapse
Affiliation(s)
- Aditya Chanpura
- Department of Neurology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Rajesh K. Gupta
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shitiz K. Sriwastava
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jan Rahmig
- Department of Neurology, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| |
Collapse
|
6
|
Arcas VC, Fratila AM, Moga DFC, Roman-Filip I, Arcas AMC, Roman-Filip C, Sava M. A Literature Review and Meta-Analysis on the Potential Use of miR-150 as a Novel Biomarker in the Detection and Progression of Multiple Sclerosis. J Pers Med 2024; 14:815. [PMID: 39202006 PMCID: PMC11355600 DOI: 10.3390/jpm14080815] [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: 07/14/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND MicroRNA-150 (miR-150) plays a critical role in immune regulation and has been implicated in autoimmune diseases like Multiple Sclerosis (MS). This review aims to evaluate miR-150's potential as a biomarker for MS, necessitating this review to consolidate current evidence and highlight miR-150's utility in improving diagnostic accuracy and monitoring disease progression. METHODS A comprehensive literature search was conducted in databases like PubMed, Scopus, Google Scholar, SciSpace, MDPI and Web of Science, adhering to PRISMA guidelines. Studies focusing on miR-150 implications in MS were included. Data extraction was conducted, while quality assessment was done using the NOS and AMSTAR 2 tools. With the extracted data a statistical analyses conducted. RESULTS 10 eligible articles were included in review. Findings show that miR-150 levels were consistently deregulated in MS patients compared to healthy controls, correlating with disease severity and clinical parameters such as (EDSS) scores and disease activity. Additionally, miR-150 is implicated in the inflammatory pathogenesis of MS, affecting immune cell regulation and inflammatory pathways. CONCLUSIONS MiR-150 is a promising biomarker for MS, showing significant potential for improving diagnostic accuracy and monitoring disease progression. Its consistent deregulation in MS patients and correlation with clinical parameters underscore its clinical utility. Further research should validate miR-150's salivary presence and its possible usage as a novel biomarker and therapeutic potential in the development of MS.
Collapse
Affiliation(s)
- Vasile Calin Arcas
- Doctoral School, Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania;
| | - Anca Maria Fratila
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (C.R.-F.); (M.S.)
- Military Clinical Emergency Hospital of Sibiu, 550024 Sibiu, Romania
| | - Doru Florian Cornel Moga
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (C.R.-F.); (M.S.)
- Military Clinical Emergency Hospital of Sibiu, 550024 Sibiu, Romania
| | - Iulian Roman-Filip
- Department of Neurology, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, 540136 Targu Mures, Romania;
| | - Ana-Maria Cristina Arcas
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy of Cluj-Napoca, 400012 Cluj-Napoca, Romania;
| | - Corina Roman-Filip
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (C.R.-F.); (M.S.)
- Emergency County Clinical Hospital Sibiu, 550245 Sibiu, Romania
| | - Mihai Sava
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (C.R.-F.); (M.S.)
- Emergency County Clinical Hospital Sibiu, 550245 Sibiu, Romania
| |
Collapse
|
7
|
Bergsland N, Dwyer MG, Jakimovski D, Tavazzi E, Weinstock-Guttman B, Zivadinov R. Choroid plexus enlargement is associated with future periventricular neurodegeneration in multiple sclerosis. Mult Scler Relat Disord 2024; 87:105668. [PMID: 38744032 DOI: 10.1016/j.msard.2024.105668] [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: 02/02/2024] [Revised: 04/10/2024] [Accepted: 05/05/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND The choroid plexus (CP), located within the ventricles of the brain and the primary producer of cerebrospinal fluid, has been shown to be enlarged in patients with multiple sclerosis (MS) and linked to periventricular remyelination failure. Atrophied T2-lesion volume (aT2-LV), a promising neurodegenerative imaging marker in progressive MS (PMS), reflects the volume of periventricular lesions subsumed into cerebrospinal fluid over the follow-up. METHODS In a cohort of 143 people with relapsing-remitting MS (RRMS) and 53 with PMS, we used 3T magnetic resonance imaging (MRI) to quantify CP volume (CPV) at baseline and aT2-LV over an average of 5.4 years of follow-up. Partial correlations, adjusting for age and sex, and linear regression analyses were used to assess the relationships between imaging measures. RESULTS In both cohorts, CPV was associated with aT2-LV in both the RRMS group (r = 0.329, p < 0.001) as well as the PMS group (r = 0.522, p < 0.001). In regression analyses predicting aT2-LV, ventricular volume (final adjusted R2 = 0.407, p < 0.001) explained additional variance beyond age, sex, and T2-lesion volume in the RRMS group while CPV (final adjusted R2 = 0.446, p = 0.009) was retained in the PMS group. CONCLUSION Findings from this study suggest that the CP enlargement is associated with future neurodegeneration, with a particularly relevant role in PMS.
Collapse
Affiliation(s)
- Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.
| | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Eleonora Tavazzi
- Multiple Sclerosis Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Bianca Weinstock-Guttman
- Department of Neurology, Jacobs Comprehensive MS Treatment and Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging at the Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
8
|
Khan Z, Mehan S, Gupta GD, Narula AS. Immune System Dysregulation in the Progression of Multiple Sclerosis: Molecular Insights and Therapeutic Implications. Neuroscience 2024; 548:9-26. [PMID: 38692349 DOI: 10.1016/j.neuroscience.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 05/03/2024]
Abstract
Multiple sclerosis (MS), a prevalent neurological disorder, predominantly affects young adults and is characterized by chronic autoimmune activity. The study explores the immune system dysregulation in MS, highlighting the crucial roles of immune and non-neuronal cells in the disease's progression. This review examines the dual role of cytokines, with some like IL-6, TNF-α, and interferon-gamma (IFN-γ) promoting inflammation and CNS tissue injury, and others such as IL-4, IL-10, IL-37, and TGF-β fostering remyelination and protecting against MS. Elevated chemokine levels in the cerebrospinal fluid (CSF), including CCL2, CCL5, CXCL10, CXCL13, and fractalkine, are analyzed for their role in facilitating immune cell migration across the blood-brain barrier (BBB), worsening inflammation and neurodegeneration. The study also delves into the impact of auto-antibodies targeting myelin components like MOG and AQP4, which activate complement cascades leading to further myelin destruction. The article discusses how compromised BBB integrity allows immune cells and inflammatory mediators to infiltrate the CNS, intensifying MS symptoms. It also examines the involvement of astrocytes, microglia, and oligodendrocytes in the disease's progression. Additionally, the effectiveness of immunomodulatory drugs such as IFN-β and CD20-targeting monoclonal antibodies (e.g., rituximab) in modulating immune responses is reviewed, highlighting their potential to reduce relapse rates and delaying MS progression. These insights emphasize the importance of immune system dysfunction in MS development and progression, guiding the development of new therapeutic strategies. The study underscores recent advancements in understanding MS's molecular pathways, opening avenues for more targeted and effective treatments.
Collapse
Affiliation(s)
- Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga 142001, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga 142001, Punjab, India.
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga, Punjab, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
9
|
Tian H, Huang D, Wang J, Li H, Gao J, Zhong Y, Xia L, Zhang A, Lin Z, Ke X. The role of the "gut microbiota-mitochondria" crosstalk in the pathogenesis of multiple sclerosis. Front Microbiol 2024; 15:1404995. [PMID: 38741740 PMCID: PMC11089144 DOI: 10.3389/fmicb.2024.1404995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 04/11/2024] [Indexed: 05/16/2024] Open
Abstract
Multiple Sclerosis (MS) is a neurologic autoimmune disease whose exact pathophysiologic mechanisms remain to be elucidated. Recent studies have shown that the onset and progression of MS are associated with dysbiosis of the gut microbiota. Similarly, a large body of evidence suggests that mitochondrial dysfunction may also have a significant impact on the development of MS. Endosymbiotic theory has found that human mitochondria are microbial in origin and share similar biological characteristics with the gut microbiota. Therefore, gut microbiota and mitochondrial function crosstalk are relevant in the development of MS. However, the relationship between gut microbiota and mitochondrial function in the development of MS is not fully understood. Therefore, by synthesizing previous relevant literature, this paper focuses on the changes in gut microbiota and metabolite composition in the development of MS and the possible mechanisms of the crosstalk between gut microbiota and mitochondrial function in the progression of MS, to provide new therapeutic approaches for the prevention or reduction of MS based on this crosstalk.
Collapse
Affiliation(s)
- Huan Tian
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dunbing Huang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiaqi Wang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huaqiang Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiaxin Gao
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Zhong
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Libin Xia
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhonghua Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Rehabilitation Medicine Center, Fujian Provincial Hospital, Fuzhou, China
- Fujian Provincial Center for Geriatrics, Fujian Provincia Hospital, Fuzhou, China
| | - Xiaohua Ke
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
10
|
Bruzaite A, Gedvilaite G, Balnyte R, Kriauciuniene L, Liutkeviciene R. Influence of STAT4 Genetic Variants and Serum Levels on Multiple Sclerosis Occurrence in the Lithuanian Population. J Clin Med 2024; 13:2385. [PMID: 38673659 PMCID: PMC11050845 DOI: 10.3390/jcm13082385] [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: 03/20/2024] [Revised: 04/10/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Background: Multiple sclerosis (MS) is an autoimmune disease involving demyelination, inflammation, gliosis, and the loss of neurons. MS is a growing global health problem most likely caused by genetic, immunological, and environmental factors. However, the exact etiology of the disease is still unknown. Since MS is related to a dysregulation of the immune system, it could be linked to signal transducer and activator of transcription 4 (STAT4). To fully comprehend the significance of the STAT4 gene and STAT4 serum levels in MS, further research is required. Methods: A total of 200 MS patients and 200 healthy controls participated in the study. Deoxyribonucleic acid (DNA) was extracted using silica-based membrane technology. Polymerase chain reaction was used in real time for genotyping. Using the ELISA technique, serum levels were measured. Results:STAT4 rs7601754 AA genotype and the A allele were statistically significantly less frequent in MS patients (p = 0.003). Also, rs7601754 was associated with 1.9-fold increased odds of MS occurrence (p = 0.004). The rs7601754 AG genotype was more common in males with MS (p = 0.011) and was associated with 2.5-fold increased odds of MS occurrence in males (p = 0.012). STAT4 serum levels were statistically significantly lower in MS patients compared to the control group (p = 0.007). Conclusions:STAT4 rs7601754 increases the odds of MS occurrence. STAT4 serum levels were statistically significantly lower in MS patients compared to the control group.
Collapse
Affiliation(s)
- Akvile Bruzaite
- Ophthalmology Laboratory, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu Street 2, LT-50161 Kaunas, Lithuania; (G.G.); (L.K.); (R.L.)
| | - Greta Gedvilaite
- Ophthalmology Laboratory, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu Street 2, LT-50161 Kaunas, Lithuania; (G.G.); (L.K.); (R.L.)
| | - Renata Balnyte
- Department of Neurology, Medical Academy, Lithuanian University of Health Sciences, Eiveniu Street 2, LT-50161 Kaunas, Lithuania;
| | - Loresa Kriauciuniene
- Ophthalmology Laboratory, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu Street 2, LT-50161 Kaunas, Lithuania; (G.G.); (L.K.); (R.L.)
| | - Rasa Liutkeviciene
- Ophthalmology Laboratory, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu Street 2, LT-50161 Kaunas, Lithuania; (G.G.); (L.K.); (R.L.)
| |
Collapse
|
11
|
Li J, Qi H, Chen Y, Zhu X. Epilepsy and demyelination: Towards a bidirectional relationship. Prog Neurobiol 2024; 234:102588. [PMID: 38378072 DOI: 10.1016/j.pneurobio.2024.102588] [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: 01/14/2024] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Demyelination stands out as a prominent feature in individuals with specific types of epilepsy. Concurrently, individuals with demyelinating diseases, such as multiple sclerosis (MS) are at a greater risk of developing epilepsy compared to non-MS individuals. These bidirectional connections raise the question of whether both pathological conditions share common pathogenic mechanisms. This review focuses on the reciprocal relationship between epilepsy and demyelination diseases. We commence with an overview of the neurological basis of epilepsy and demyelination diseases, followed by an exploration of how our comprehension of these two disorders has evolved in tandem. Additionally, we discuss the potential pathogenic mechanisms contributing to the interactive relationship between these two diseases. A more nuanced understanding of the interplay between epilepsy and demyelination diseases has the potential to unveiling the molecular intricacies of their pathological relationships, paving the way for innovative directions in future clinical management and treatment strategies for these diseases.
Collapse
Affiliation(s)
- Jiayi Li
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China; Clinical Medicine, Medical School of Southeast University, Nanjing, China
| | - Honggang Qi
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Yuzhou Chen
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China; Clinical Medicine, Medical School of Southeast University, Nanjing, China
| | - Xinjian Zhu
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| |
Collapse
|
12
|
Min H, O’Neil SM, Xu L, Moseman EA, Kurtzberg J, Filiano AJ. Mural cells interact with macrophages in the dura mater to regulate CNS immune surveillance. J Exp Med 2024; 221:e20230326. [PMID: 38193859 PMCID: PMC10783178 DOI: 10.1084/jem.20230326] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 09/07/2023] [Accepted: 12/13/2023] [Indexed: 01/10/2024] Open
Abstract
The central nervous system (CNS) tightly regulates access of circulating immune cells. Immunosurveillance is therefore managed in the meninges at the borders of the CNS. Here, we demonstrated that mural cells, which include pericytes and smooth muscle cells, decreased coverage around blood vessels in the dura, the outermost layer of the meninges, and upregulated gene pathways involved in leukocyte migration in presymptomatic experimental autoimmune encephalomyelitis (EAE). Partially depleting mural cells promoted the trafficking of CNS antigen-specific T cells to the dura in a process that depended on resident antigen-presenting cells, thereby increasing susceptibility to passive EAE. Mechanistically, mural cells physically contacted macrophages in the dura and transferred cytoplasmic components, including processing bodies (RNA granules shown to reprogram transcriptomes), which were critical to suppress antigen-dependent T helper (TH) cell activation and TH17 differentiation. Our study revealed a mechanism by which mural cell-macrophage interactions regulate the trafficking of CNS antigen-specific T cells to the dura.
Collapse
Affiliation(s)
- Hyunjung Min
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Shane M. O’Neil
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
| | - Li Xu
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
| | - E. Ashley Moseman
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Anthony J. Filiano
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
13
|
Chen X, Wang Y, Ji J, Li C, Zhuang W, Luo J, Shi Y, Lin Q, Wu J, Li A, Wang J, Meng Y, Zhang S, Lang X, Liu X, Sun B, Li H, Liu Y. Electroacupuncture at ST36 acupoint regulates stem cells during experimental autoimmune encephalomyelitis. Int Immunopharmacol 2023; 124:110856. [PMID: 37647680 DOI: 10.1016/j.intimp.2023.110856] [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: 05/16/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Electroacupuncture (EA) is given to assist in the treatment of MS, which is an effective therapeutic method. However, the therapy mechanism of EA related to stem cells in the treatment of MS is not yet known. In this study, we used a classic animal model of multiple sclerosis: experimental autoimmune encephalomyelitis (EAE) to evaluate the therapeutic effect of EA at Zusanli (ST36) acupoint in EAE and shed light on its potential roles in the effects of stem cells in vivo. METHODS The EAE animal models were established. From the first day after immunization, EAE model mice received EA at ST36 acupoint, named the EA group. The weight and clinical score of the three groups were recorded for 28 days. The demyelination, inflammatory cell infiltration, and markers of neural stem cells (NSCs), hematopoietic stem cells (HSCs), and mesenchymal stem cells (MSCs) were compared. RESULTS We showed that EAE mice treated with EA at ST36 acupoint, were suppressed in demyelination and inflammatory cell infiltration, and thus decreased clinical score and weight loss and mitigated the development of EAE when compared with the EAE group. Moreover, our data revealed that the proportions of NSCs, HSCs, and MSCs increased in the EA group compared with the EAE group. CONCLUSIONS Our study suggested that EA at ST36 acupoint was an effective nonpharmacological therapeutic protocol that not only reduced the CNS demyelination and inflammatory cell infiltration in EAE disease but also increased the proportions of various stem cells. Further study is necessary to better understand how EA at the ST36 acupoint affects EAE.
Collapse
Affiliation(s)
- Xin Chen
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Yanping Wang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Jiayu Ji
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Changyu Li
- Department of Neurosurgery, Hainan Cancer Hospital, Haikou, China
| | - Wei Zhuang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Jingyu Luo
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Yu Shi
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Qian Lin
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Junfeng Wu
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Anqi Li
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Jing Wang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Yanting Meng
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Xiujuan Lang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Xijun Liu
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Bo Sun
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin, China
| | - Yumei Liu
- Department of Neurobiology, Harbin Medical University, Harbin, China.
| |
Collapse
|
14
|
Jafari Karegar S, Aryaeian N, Hajiluian G, Suzuki K, Shidfar F, Salehi M, Ashtiani BH, Farhangnia P, Delbandi AA. Ellagic acid effects on disease severity, levels of cytokines and T-bet, RORγt, and GATA3 genes expression in multiple sclerosis patients: a multicentral-triple blind randomized clinical trial. Front Nutr 2023; 10:1238846. [PMID: 37794975 PMCID: PMC10546207 DOI: 10.3389/fnut.2023.1238846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/28/2023] [Indexed: 10/06/2023] Open
Abstract
Background Multiple sclerosis (MS) is a chronic autoimmune disease. Ellagic acid is a natural polyphenol and affects the fate of neurons through its anti-inflammatory and antioxidant properties. The present study aimed to investigate ellagic acid effects on disease severity, the expression of involved genes in the pathogenesis of MS, and the levels of related cytokines. Methods The present study was a triple-blind clinical trial. Eligible patients were randomly assigned to two groups: Ellagic acid (25 subjects) for 12 weeks, receiving 180 mg of Ellagic acid (Axenic, Australia) and the control group (25 subjects) receiving a placebo, before the main meals. Before and after the study, the data including general information, foods intake, physical activity, anthropometric data, expanded disability status scale (EDSS), general health questionnaire (GHQ) and pain rating index (PRI), fatigue severity scale (FSS) were assessed, as well as serum levels of interferon-gamma (IFNγ), interleukin-17 (IL-17), interleukin-4 (IL-4) and transforming growth factor-beta (TGF-β), nitric-oxide (NO) using enzyme-linked immunoassay (ELISA) method and expression of T-box transcription factor (Tbet), GATA Binding Protein 3 (GATA3), retinoic acid-related orphan receptor-γt (RORγt) and Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) genes were determined using Real-Time Quantitative Reverse Transcription PCR (RT-qPCR) method. Findings Ellagic acid supplementation led to a reduction in IFNγ, IL-17, NO and increased IL-4 in the ellagic acid group, however in the placebo group no such changes were observed (-24.52 ± 3.79 vs. -0.05 ± 0.02, p < 0.01; -5.37 ± 0.92 vs. 2.03 ± 1.03, p < 0.01; -18.03 ± 1.02 vs. -0.06 ± 0.05, p < 0.01, 14.69 ± 0.47 vs. -0.09 ± 0.14, p < 0.01, respectively). Ellagic acid supplementation had no effect on TGF-β in any of the study groups (p > 0.05). Also, the Tbet and RORγt genes expression decreased, and the GATA3 gene expression in the group receiving ellagic acid compared to control group significantly increased (0.52 ± 0.29 vs. 1.51 ± 0.18, p < 0.01, 0.49 ± 0.18 vs. 1.38 ± 0.14, p < 0.01, 1.71 ± 0.39 vs. 0.27 ± 0.10, p < 0.01). Also, ellagic acid supplementation led to significant decrease in EDSS, FSS and GHQ scores (p < 0.05), and no significant changes observed in PRI score (p > 0.05). Conclusion Ellagic acid supplementation can improve the health status of MS patients by reduction of the inflammatory cytokines and Tbet and RORγt gene expression, and increment of anti-inflammatory cytokines and GATA3 gene expression.Clinical trial registration: (https://en.irct.ir/trial/53020), IRCT20120415009472N22.
Collapse
Affiliation(s)
- Sahar Jafari Karegar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Naheed Aryaeian
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Hajiluian
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | | | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Salehi
- Department of Statistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | | | - Pooya Farhangnia
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Disease, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Brandl S, Reindl M. Blood-Brain Barrier Breakdown in Neuroinflammation: Current In Vitro Models. Int J Mol Sci 2023; 24:12699. [PMID: 37628879 PMCID: PMC10454051 DOI: 10.3390/ijms241612699] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The blood-brain barrier, which is formed by tightly interconnected microvascular endothelial cells, separates the brain from the peripheral circulation. Together with other central nervous system-resident cell types, including pericytes and astrocytes, the blood-brain barrier forms the neurovascular unit. Upon neuroinflammation, this barrier becomes leaky, allowing molecules and cells to enter the brain and to potentially harm the tissue of the central nervous system. Despite the significance of animal models in research, they may not always adequately reflect human pathophysiology. Therefore, human models are needed. This review will provide an overview of the blood-brain barrier in terms of both health and disease. It will describe all key elements of the in vitro models and will explore how different compositions can be utilized to effectively model a variety of neuroinflammatory conditions. Furthermore, it will explore the existing types of models that are used in basic research to study the respective pathologies thus far.
Collapse
Affiliation(s)
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
16
|
Alsbrook DL, Di Napoli M, Bhatia K, Biller J, Andalib S, Hinduja A, Rodrigues R, Rodriguez M, Sabbagh SY, Selim M, Farahabadi MH, Jafarli A, Divani AA. Neuroinflammation in Acute Ischemic and Hemorrhagic Stroke. Curr Neurol Neurosci Rep 2023; 23:407-431. [PMID: 37395873 PMCID: PMC10544736 DOI: 10.1007/s11910-023-01282-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE OF REVIEW This review aims to provide an overview of neuroinflammation in ischemic and hemorrhagic stroke, including recent findings on the mechanisms and cellular players involved in the inflammatory response to brain injury. RECENT FINDINGS Neuroinflammation is a crucial process following acute ischemic stroke (AIS) and hemorrhagic stroke (HS). In AIS, neuroinflammation is initiated within minutes of the ischemia onset and continues for several days. In HS, neuroinflammation is initiated by blood byproducts in the subarachnoid space and/or brain parenchyma. In both cases, neuroinflammation is characterized by the activation of resident immune cells, such as microglia and astrocytes, and infiltration of peripheral immune cells, leading to the release of pro-inflammatory cytokines, chemokines, and reactive oxygen species. These inflammatory mediators contribute to blood-brain barrier disruption, neuronal damage, and cerebral edema, promoting neuronal apoptosis and impairing neuroplasticity, ultimately exacerbating the neurologic deficit. However, neuroinflammation can also have beneficial effects by clearing cellular debris and promoting tissue repair. The role of neuroinflammation in AIS and ICH is complex and multifaceted, and further research is necessary to develop effective therapies that target this process. Intracerebral hemorrhage (ICH) will be the HS subtype addressed in this review. Neuroinflammation is a significant contributor to brain tissue damage following AIS and HS. Understanding the mechanisms and cellular players involved in neuroinflammation is essential for developing effective therapies to reduce secondary injury and improve stroke outcomes. Recent findings have provided new insights into the pathophysiology of neuroinflammation, highlighting the potential for targeting specific cytokines, chemokines, and glial cells as therapeutic strategies.
Collapse
Affiliation(s)
- Diana L Alsbrook
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mario Di Napoli
- Neurological Service, SS Annunziata Hospital, Sulmona, L'Aquila, Italy
| | - Kunal Bhatia
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA
| | - José Biller
- Department of Neurology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Sasan Andalib
- Research Unit of Neurology, Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Archana Hinduja
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Roysten Rodrigues
- Department of Neurology, University of Louisville, Louisville, KY, USA
| | - Miguel Rodriguez
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sara Y Sabbagh
- Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Magdy Selim
- Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Alibay Jafarli
- Department of Neurology, Tufts Medical Center, Boston, MA, USA
| | - Afshin A Divani
- Department of Neurology, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
17
|
Qi J, Liu C, Bai Z, Li X, Yao G. T follicular helper cells and T follicular regulatory cells in autoimmune diseases. Front Immunol 2023; 14:1178792. [PMID: 37187757 PMCID: PMC10175690 DOI: 10.3389/fimmu.2023.1178792] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
T follicular helper (Tfh) cells are heterogeneous and mainly characterized by expressing surface markers CXCR5, ICOS, and PD-1; cytokine IL-21; and transcription factor Bcl6. They are crucial for B-cell differentiation into long-lived plasma cells and high-affinity antibody production. T follicular regulatory (Tfr) cells were described to express markers of conventional T regulatory (Treg) cells and Tfh cells and were able to suppress Tfh-cell and B-cell responses. Evidence has revealed that the dysregulation of Tfh and Tfr cells is positively associated with the pathogenic processes of autoimmune diseases. Herein, we briefly introduce the phenotype, differentiation, and function of Tfh and Tfr cells, and review their potential roles in autoimmune diseases. In addition, we discuss perspectives to develop novel therapies targeting Tfh/Tfr balance.
Collapse
Affiliation(s)
- Jingjing Qi
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
- *Correspondence: Genhong Yao, ; Jingjing Qi,
| | - Chang Liu
- Department of Rheumatology and Immunology, Dalian Municipal Central Hospital, Dalian, Liaoning, China
| | - Ziran Bai
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Xia Li
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
- *Correspondence: Genhong Yao, ; Jingjing Qi,
| |
Collapse
|