1
|
Ashwani, Sharma A, Choudhary MK, Gugulothu D, Pandita D, Verma S, Vora LK, Khatri DK, Garabadu D. Epigenetic and Mitochondrial Metabolic Dysfunction in Multiple Sclerosis: A Review of Herbal Drug Approaches and Current Clinical Trials. Mol Neurobiol 2025:10.1007/s12035-025-04868-8. [PMID: 40180689 DOI: 10.1007/s12035-025-04868-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025]
Abstract
Multiple sclerosis (MS) is a complex autoimmune disease characterised by inflammation, demyelination, and neurodegeneration within the central nervous system (CNS). While the exact causes remain unclear, recent research highlights the significant role of epigenetic modifications and mitochondrial dysfunction in the disease's onset and progression. Epigenetic alterations, such as DNA methylation, histone modification, and microRNA regulation, influence gene expression without altering the DNA sequence, leading to immune dysregulation and inflammation. Similarly, mitochondrial dysfunction, marked by impaired oxidative phosphorylation, reduced adenosine triphosphate (ATP) production, and increased reactive oxygen species (ROS), contributes to neurodegeneration and impaired remyelination in MS. The growing interest in targeting these two interconnected mechanisms has opened new avenues for MS treatment. Herbal drugs, known for their multi-targeted effects, have shown potential in modulating epigenetic markers and enhancing mitochondrial function. Compounds such as resveratrol, curcumin, epigallocatechin-3-gallate (EGCG), quercetin, and omega-3 fatty acids demonstrate potential in regulating DNA methylation, histone deacetylation, and mitochondrial biogenesis. These natural agents offer dual-action therapies by reducing oxidative stress and inflammation while promoting neuronal survival and remyelination. This review explores the therapeutic potential of herbal drugs targeting epigenetic and mitochondrial pathways in MS, evaluating their mechanisms of action and highlighting their promise as novel therapeutic agents. While initial findings are encouraging, further research and clinical trials are required to validate the efficacy of these herbal treatments and fully understand their potential in slowing disease progression and improving patient outcomes in MS. Such exploration could pave the way for safer, multi-targeted therapies, offering new hope in the management of MS and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ashwani
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | | | - Mayank Kumar Choudhary
- Department of Pharmacology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Dalapathi Gugulothu
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India.
| | - Deepti Pandita
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | - Surajpal Verma
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | - Lalitkumar K Vora
- School of Pharmacy, Medical Biology Centre, Queen'S University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK.
| | - Dharmendra Kumar Khatri
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India.
| | - Debapriya Garabadu
- Department of Pharmacology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
2
|
Teekaput C, Rachbundit C, Wantaneeyawong C, Teekaput K, Thiankhaw K. Impact of air pollution on the clinical exacerbation of central demyelinating disease: A 10-year data from the Northern Thailand MS and NMOSD registry. Mult Scler Relat Disord 2025; 94:106266. [PMID: 39817949 DOI: 10.1016/j.msard.2025.106266] [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: 05/20/2024] [Revised: 12/03/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Particulate matter (PM) 2.5 (PM2.5) and PM10 are implicated in neurological diseases, yet their impact on central demyelinating diseases like multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) remains unclear. This study aimed to determine the association between the levels of PM2.5 and PM10 and the exacerbation of MS and NMOSD. METHODS Patients with clinical exacerbations of MS and NMOSD in the Northern Thailand Registry from 2013 to 2022 were enrolled. Eligible patients were categorized based on air pollution exposure (defined as PM2.5 > 15 μg/m3 and PM10 > 45 μg/m3) or no air pollution exposure. Outcomes assessed included clinical characteristics, Expanded Disability Status Scale (EDSS), and functional outcomes. RESULTS We analyzed 126 exacerbations in the PM2.5 database (mean age: 44.9±14.9 years, 114 NMOSD, 49 first exacerbations) and 135 exacerbations with the PM10 database (mean age: 44.9±14.9 years, 121 NMOSD, 54 first exacerbations), with the highest incidence four months post-peak air pollution. The PM2.5 exposure group had higher severity, showing increased rates of unfavorable EDSS at exacerbation and 90 days (56.3% vs. 23.6%, P < 0.001 and 47.9% vs. 16.4%, P < 0.001, respectively). Gadolinium enhancement in PM2.5 exposure was significantly higher (56.3% vs. 36.4%, P = 0.03). The PM2.5 exposure group also had a higher rate of second-line therapy with plasma exchange (21.1% vs. 7.3%, P = 0.03). PM2.5 exposure, not PM10, was associated with unfavorable EDSS at any time point, active radiological activity, risk of plasma exchange, and prolonged hospitalization. CONCLUSION Environmental pollution, especially PM2.5, significantly impacts MS and NMOSD patients, influencing disease severity, causing permanent disability, and prolonging hospitalization. A national policy on pollution control is imperative, and further data on long-term exposure, together with other pollutants, is still required.
Collapse
Affiliation(s)
- Chutithep Teekaput
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; The Northern Neuroscience Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chanita Rachbundit
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; The Northern Neuroscience Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chayasak Wantaneeyawong
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; The Northern Neuroscience Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kanokkarn Teekaput
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kitti Thiankhaw
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; The Northern Neuroscience Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
3
|
Li Y, Sun J, Zhuo Z, Guo M, Duan Y, Xu X, Tian D, Li K, Zhou F, Li H, Zhang N, Han X, Shi F, Li Y, Zhang X, Liu Y. Imaging Transcriptomics of Brain Functional Alterations in MS and Neuromyelitis Optica Spectrum Disorder. AJNR Am J Neuroradiol 2024; 45:1901-1909. [PMID: 39510804 PMCID: PMC11630882 DOI: 10.3174/ajnr.a8480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/17/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND AND PURPOSE The underlying transcriptomic signatures driving brain functional alterations in MS and neuromyelitis optica spectrum disorder (NMOSD) are still unclear. MATERIALS AND METHODS Regional fractional amplitude of low-frequency fluctuation (fALFF) values were obtained and compared among 209 patients with MS, 90 patients with antiaquaporin-4 antibody (AQP4)+ NMOSD, 49 with AQP4- NMOSD, and 228 healthy controls from a discovery cohort. We used partial least squares (PLS) regression to identify the gene transcriptomic signatures associated with disease-related fALFF alterations. The biologic process and cell type-specific signature of the identified PLS genes were explored by enrichment analysis. The correlation between PLS genes and clinical variables was explored. A prospective independent cohort was used to validate the brain fALFF alterations and the repeatability of identified genes. RESULTS MS, AQP4+ NMOSD, and AQP4- NMOSD showed decreased fALFF in cognition-related regions and deep gray matter, while NMOSD (both AQP4+ and AQP4-) additionally demonstrated lower fALFF in the visual region. The overlapping PLS1- genes (indicating that the genes were overexpressed as regional fALFF decreased) were enriched in response to regulation of the immune response in all diseases, and the PLS1- genes were specifically enriched in the epigenetics profile in MS, membrane disruption and cell adhesion in AQP4+ NMOSD, and leukocyte activation in AQP4- NMOSD. For the cell type transcriptional signature, microglia and astrocytes accounted for the decreased fALFF. The fALFF-associated PLS1- genes directly correlated with Expanded Disability Status Scale of MS and disease duration across disorders. CONCLUSIONS We revealed the functional activity alterations and their underlying shared and specific gene transcriptional signatures in MS, AQP4+ NMOSD, and AQP4- NMOSD.
Collapse
Affiliation(s)
- Yuna Li
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun Sun
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhizheng Zhuo
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Min Guo
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yunyun Duan
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaolu Xu
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Decai Tian
- Center for Neurology (D.T., X.Z.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kuncheng Li
- Department of Radiology (K.L.), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fuqing Zhou
- Department of Radiology (F.Z.), The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi Province, China
| | - Haiqing Li
- Department of Radiology (H.L.), Huashan Hospital, Fudan University, Shanghai, China
| | - Ningnannan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging (N.Z.), Tianjin Medical University General Hospital, Tianjin, China
| | - Xuemei Han
- Department of Neurology (X.H.), China-Japan Union Hospital of Jilin University, Changchun, China
| | - Fudong Shi
- China National Clinical Research Center for Neurological Diseases (F.S.), Beijing, China
- Department of Neurology (F.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yongmei Li
- Department of Radiology (Yongmei Li), The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinghu Zhang
- Center for Neurology (D.T., X.Z.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yaou Liu
- From the Department of Radiology (Yuna Li, J.S., Z.Z., M.G., Y.D., X.X., Y. Liu), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Woo MS, Engler JB, Friese MA. The neuropathobiology of multiple sclerosis. Nat Rev Neurosci 2024; 25:493-513. [PMID: 38789516 DOI: 10.1038/s41583-024-00823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
Chronic low-grade inflammation and neuronal deregulation are two components of a smoldering disease activity that drives the progression of disability in people with multiple sclerosis (MS). Although several therapies exist to dampen the acute inflammation that drives MS relapses, therapeutic options to halt chronic disability progression are a major unmet clinical need. The development of such therapies is hindered by our limited understanding of the neuron-intrinsic determinants of resilience or vulnerability to inflammation. In this Review, we provide a neuron-centric overview of recent advances in deciphering neuronal response patterns that drive the pathology of MS. We describe the inflammatory CNS environment that initiates neurotoxicity by imposing ion imbalance, excitotoxicity and oxidative stress, and by direct neuro-immune interactions, which collectively lead to mitochondrial dysfunction and epigenetic dysregulation. The neuronal demise is further amplified by breakdown of neuronal transport, accumulation of cytosolic proteins and activation of cell death pathways. Continuous neuronal damage perpetuates CNS inflammation by activating surrounding glia cells and by directly exerting toxicity on neighbouring neurons. Further, we explore strategies to overcome neuronal deregulation in MS and compile a selection of neuronal actuators shown to impact neurodegeneration in preclinical studies. We conclude by discussing the therapeutic potential of targeting such neuronal actuators in MS, including some that have already been tested in interventional clinical trials.
Collapse
Affiliation(s)
- Marcel S Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
5
|
Eid K, Bjørk MH, Gilhus NE, Torkildsen Ø. Adverse Childhood Experiences and the Risk of Multiple Sclerosis Development: A Review of Potential Mechanisms. Int J Mol Sci 2024; 25:1520. [PMID: 38338799 PMCID: PMC10855716 DOI: 10.3390/ijms25031520] [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: 12/20/2023] [Revised: 01/16/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Adverse childhood experiences (ACEs), such as abuse, neglect, and household dysfunction, contribute to long-term systemic toxic stress and inflammation that may last well into adulthood. Such early-life stressors have been associated with increased susceptibility to multiple sclerosis (MS) in observational studies and with the development of experimental autoimmune encephalomyelitis in animal models. In this review, we summarize the evidence for an ACE-mediated increase in MS risk, as well as the potential mechanisms for this association. ACEs dysregulate neurodevelopment, stress responses, and immune reactivity; they also alter the interplay between the immune system and neural networks. All of this may be relevant for MS risk. We further discuss how ACEs induce epigenetic changes and how the toxic stress caused by ACEs may reactivate the Epstein-Barr Virus (EBV), a key risk factor for MS. We conclude by suggesting new initiatives to obtain further insights into this topic.
Collapse
Affiliation(s)
- Karine Eid
- Department of Neurology, Haukeland University Hospital, Jonas Lies vei 71, 5053 Bergen, Norway; (M.-H.B.); (N.E.G.)
- Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway;
| | - Marte-Helene Bjørk
- Department of Neurology, Haukeland University Hospital, Jonas Lies vei 71, 5053 Bergen, Norway; (M.-H.B.); (N.E.G.)
- Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway;
- NorHead, Norwegian Center for Headache Research, 5021 Bergen, Norway
| | - Nils Erik Gilhus
- Department of Neurology, Haukeland University Hospital, Jonas Lies vei 71, 5053 Bergen, Norway; (M.-H.B.); (N.E.G.)
- Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway;
| | - Øivind Torkildsen
- Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway;
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| |
Collapse
|
6
|
Kozlenkov A, Vadukapuram R, Zhou P, Fam P, Wegner M, Dracheva S. Novel method of isolating nuclei of human oligodendrocyte precursor cells reveals substantial developmental changes in gene expression and H3K27ac histone modification. Glia 2024; 72:69-89. [PMID: 37712493 PMCID: PMC10697634 DOI: 10.1002/glia.24462] [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: 09/18/2022] [Revised: 08/01/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) generate differentiated mature oligodendrocytes (MOs) during development. In adult brain, OPCs replenish MOs in adaptive plasticity, neurodegenerative disorders, and after trauma. The ability of OPCs to differentiate to MOs decreases with age and is compromised in disease. Here we explored the cell specific and age-dependent differences in gene expression and H3K27ac histone mark in these two cell types. H3K27ac is indicative of active promoters and enhancers. We developed a novel flow-cytometry-based approach to isolate OPC and MO nuclei from human postmortem brain and profiled gene expression and H3K27ac in adult and infant OPCs and MOs genome-wide. In adult brain, we detected extensive H3K27ac differences between the two cell types with high concordance between gene expression and epigenetic changes. Notably, the expression of genes that distinguish MOs from OPCs appears to be under a strong regulatory control by the H3K27ac modification in MOs but not in OPCs. Comparison of gene expression and H3K27ac between infants and adults uncovered numerous developmental changes in each cell type, which were linked to several biological processes, including cell proliferation and glutamate signaling. A striking example was a subset of histone genes that were highly active in infant samples but fully lost activity in adult brain. Our findings demonstrate a considerable rearrangement of the H3K27ac landscape that occurs during the differentiation of OPCs to MOs and during postnatal development of these cell types, which aligned with changes in gene expression. The uncovered regulatory changes justify further in-depth epigenetic studies of OPCs and MOs in development and disease.
Collapse
Affiliation(s)
- Alexey Kozlenkov
- James J. Peters VA Medical Center, Bronx, NY, USA
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ramu Vadukapuram
- James J. Peters VA Medical Center, Bronx, NY, USA
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ping Zhou
- James J. Peters VA Medical Center, Bronx, NY, USA
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Fam
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stella Dracheva
- James J. Peters VA Medical Center, Bronx, NY, USA
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
7
|
Reyes-Mata MP, Mireles-Ramírez MA, Griñán-Ferré C, Pallàs M, Pavón L, Guerrero-García JDJ, Ortuño-Sahagún D. Global DNA Methylation and Hydroxymethylation Levels in PBMCs Are Altered in RRMS Patients Treated with IFN-β and GA-A Preliminary Study. Int J Mol Sci 2023; 24:9074. [PMID: 37240421 PMCID: PMC10219581 DOI: 10.3390/ijms24109074] [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: 03/26/2023] [Revised: 04/15/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic disease affecting the central nervous system (CNS) due to an autoimmune attack on axonal myelin sheaths. Epigenetics is an open research topic on MS, which has been investigated in search of biomarkers and treatment targets for this heterogeneous disease. In this study, we quantified global levels of epigenetic marks using an ELISA-like approach in Peripheral Blood Mononuclear Cells (PBMCs) from 52 patients with MS, treated with Interferon beta (IFN-β) and Glatiramer Acetate (GA) or untreated, and 30 healthy controls. We performed media comparisons and correlation analyses of these epigenetic markers with clinical variables in subgroups of patients and controls. We observed that DNA methylation (5-mC) decreased in treated patients compared with untreated and healthy controls. Moreover, 5-mC and hydroxymethylation (5-hmC) correlated with clinical variables. In contrast, histone H3 and H4 acetylation did not correlate with the disease variables considered. Globally quantified epigenetic DNA marks 5-mC and 5-hmC correlate with disease and were altered with treatment. However, to date, no biomarker has been identified that can predict the potential response to therapy before treatment initiation.
Collapse
Affiliation(s)
- María Paulina Reyes-Mata
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Mario Alberto Mireles-Ramírez
- Unidad Médica de Alta Especialidad (UMAE), Hospital de Especialidades (HE), Centro Médico Nacional de Occidente (CMNO), IMSS, Guadalajara 44340, Mexico
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28220 Madrid, Spain
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28220 Madrid, Spain
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - José de Jesús Guerrero-García
- Banco de Sangre Central, Unidad Médica de Alta Especialidad (UMAE), Hospital de Especialidades (HE), Centro Médico Nacional de Occidente (CMNO), IMSS, Guadalajara 44340, Mexico
- Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico
| |
Collapse
|
8
|
Liu R, Du S, Zhao L, Jain S, Sahay K, Rizvanov A, Lezhnyova V, Khaibullin T, Martynova E, Khaiboullina S, Baranwal M. Autoreactive lymphocytes in multiple sclerosis: Pathogenesis and treatment target. Front Immunol 2022; 13:996469. [PMID: 36211343 PMCID: PMC9539795 DOI: 10.3389/fimmu.2022.996469] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by destruction of the myelin sheath structure. The loss of myelin leads to damage of a neuron’s axon and cell body, which is identified as brain lesions on magnetic resonance image (MRI). The pathogenesis of MS remains largely unknown. However, immune mechanisms, especially those linked to the aberrant lymphocyte activity, are mainly responsible for neuronal damage. Th1 and Th17 populations of lymphocytes were primarily associated with MS pathogenesis. These lymphocytes are essential for differentiation of encephalitogenic CD8+ T cell and Th17 lymphocyte crossing the blood brain barrier and targeting myelin sheath in the CNS. B-lymphocytes could also contribute to MS pathogenesis by producing anti-myelin basic protein antibodies. In later studies, aberrant function of Treg and Th9 cells was identified as contributing to MS. This review summarizes the aberrant function and count of lymphocyte, and the contributions of these cell to the mechanisms of MS. Additionally, we have outlined the novel MS therapeutics aimed to amend the aberrant function or counts of these lymphocytes.
Collapse
Affiliation(s)
- Rongzeng Liu
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Shushu Du
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Lili Zhao
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Sahil Jain
- Department of Biochemistry and Molecular Biology, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Kritika Sahay
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Albert Rizvanov
- Gene and cell Department, Kazan Federal University, Kazan, Russia
| | - Vera Lezhnyova
- Gene and cell Department, Kazan Federal University, Kazan, Russia
| | - Timur Khaibullin
- Neurological Department, Republican Clinical Neurological Center, Kazan, Russia
| | | | - Svetlana Khaiboullina
- Gene and cell Department, Kazan Federal University, Kazan, Russia
- *Correspondence: Svetlana Khaiboullina, ; Manoj Baranwal, ;
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
- *Correspondence: Svetlana Khaiboullina, ; Manoj Baranwal, ;
| |
Collapse
|
9
|
Investigating the shared genetic architecture between multiple sclerosis and inflammatory bowel diseases. Nat Commun 2021; 12:5641. [PMID: 34561436 PMCID: PMC8463615 DOI: 10.1038/s41467-021-25768-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 08/31/2021] [Indexed: 12/15/2022] Open
Abstract
An epidemiological association between multiple sclerosis (MS) and inflammatory bowel disease (IBD) is well established, but whether this reflects a shared genetic aetiology, and whether consistent genetic relationships exist between MS and the two predominant IBD subtypes, ulcerative colitis (UC) and Crohn’s disease (CD), remains unclear. Here, we use large-scale genome-wide association study summary data to investigate the shared genetic architecture between MS and IBD overall and UC and CD independently. We find a significantly greater genetic correlation between MS and UC than between MS and CD, and identify three SNPs shared between MS and IBD (rs13428812), UC (rs116555563) and CD (rs13428812, rs9977672) in cross-trait meta-analyses. We find suggestive evidence for a causal effect of MS on UC and IBD using Mendelian randomization, but no or weak and inconsistent evidence for a causal effect of IBD or UC on MS. We observe largely consistent patterns of tissue-specific heritability enrichment for MS and IBDs in lung, spleen, whole blood and small intestine, and identify cell-type-specific enrichment for MS and IBDs in CD4+ T cells in lung and CD8+ cytotoxic T cells in lung and spleen. Our study sheds light on the biological basis of comorbidity between MS and IBD. An epidemiological association between multiple sclerosis (MS) and inflammatory bowel disease (IBD) is well-established, but a genetic link is unclear. Here, the authors investigate the shared genetic architecture between MS and IBD to shed light on the biological basis of comorbidity.
Collapse
|
10
|
Hurtado Rúa SM, Kaunzner UW, Pandya S, Sweeney E, Tozlu C, Kuceyeski A, Nguyen TD, Gauthier SA. Lesion features on magnetic resonance imaging discriminate multiple sclerosis patients. Eur J Neurol 2021; 29:237-246. [PMID: 34402140 DOI: 10.1111/ene.15067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Magnetic resonance imaging (MRI) provides insight into various pathological processes in multiple sclerosis (MS) and may provide insight into patterns of damage among patients. OBJECTIVE We sought to determine if MRI features have clinical discriminative power among a cohort of MS patients. METHODS Ninety-six relapsing remitting and seven progressive MS patients underwent myelin water fraction (MWF) imaging and conventional MRI for cortical thickness and thalamic volume. Patients were clustered based on lesion level MRI features using an agglomerative hierarchical clustering algorithm based on principal component analysis (PCA). RESULTS One hundred and three patients with 1689 MS lesions were analyzed. PCA on MRI features demonstrated that lesion MWF and volume distributions (characterized by 25th, 50th, and 75th percentiles) accounted for 87% of the total variability based on four principal components. The best hierarchical cluster confirmed two distinct patient clusters. The clustering features in order of importance were lesion median MWF, MWF 25th, MWF 75th, volume 75th percentiles, median individual lesion volume, total lesion volume, cortical thickness, and thalamic volume (all p values <0.01368). The clusters were associated with patient Expanded Disability Status Scale (EDSS) (n = 103, p = 0.0338) at baseline and at 5 years (n = 72, p = 0.0337). CONCLUSIONS These results demonstrate that individual MRI features can identify two patient clusters driven by lesion-based values, and our unique approach is an analysis blinded to clinical variables. The two distinct clusters exhibit MWF differences, most likely representing individual remyelination capabilities among different patient groups. These findings support the concept of patient-specific pathophysiological processes and may guide future therapeutic approaches.
Collapse
Affiliation(s)
- Sandra M Hurtado Rúa
- Department of Mathematics and Statistics, Cleveland State University, Cleveland, Ohio, USA
| | - Ulrike W Kaunzner
- Department of Neurology, Weill Cornell Medicine, New York City, New York, USA
| | - Sneha Pandya
- Department of Radiology, Weill Cornell Medicine, New York City, New York, USA
| | - Elizabeth Sweeney
- Department of Population Health Sciences, Weill Cornell Medicine, New York City, New York, USA
| | - Ceren Tozlu
- Department of Radiology, Weill Cornell Medicine, New York City, New York, USA
| | - Amy Kuceyeski
- Department of Radiology, Weill Cornell Medicine, New York City, New York, USA.,Feil Family Brain and Mind Institute, Weill Cornell Medicine, New York City, New York, USA
| | - Thanh D Nguyen
- Department of Radiology, Weill Cornell Medicine, New York City, New York, USA
| | - Susan A Gauthier
- Department of Neurology, Weill Cornell Medicine, New York City, New York, USA.,Department of Radiology, Weill Cornell Medicine, New York City, New York, USA.,Feil Family Brain and Mind Institute, Weill Cornell Medicine, New York City, New York, USA
| |
Collapse
|
11
|
LoPresti P. HDAC6 in Diseases of Cognition and of Neurons. Cells 2020; 10:E12. [PMID: 33374719 PMCID: PMC7822434 DOI: 10.3390/cells10010012] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
Central nervous system (CNS) neurodegenerative diseases are characterized by faulty intracellular transport, cognition, and aggregate regulation. Traditionally, neuroprotection exerted by histone deacetylase (HDAC) inhibitors (HDACi) has been attributed to the ability of this drug class to promote histone acetylation. However, HDAC6 in the healthy CNS functions via distinct mechanisms, due largely to its cytoplasmic localization. Indeed, in healthy neurons, cytoplasmic HDAC6 regulates the acetylation of a variety of non-histone proteins that are linked to separate functions, i.e., intracellular transport, neurotransmitter release, and aggregate formation. These three HDAC6 activities could work independently or in synergy. Of particular interest, HDAC6 targets the synaptic protein Bruchpilot and neurotransmitter release. In pathological conditions, HDAC6 becomes abundant in the nucleus, with deleterious consequences for transcription regulation and synapses. Thus, HDAC6 plays a leading role in neuronal health or dysfunction. Here, we review recent findings and novel conclusions on the role of HDAC6 in neurodegeneration. Selective studies with pan-HDACi are also included. We propose that an early alteration of HDAC6 undermines synaptic transmission, while altering transport and aggregation, eventually leading to neurodegeneration.
Collapse
Affiliation(s)
- Patrizia LoPresti
- Department of Psychology, University of Illinois at Chicago, 1007 West Harrison Street, Chicago, IL 60607, USA
| |
Collapse
|
12
|
Martins-Ferreira R, Leal B, Costa PP, Ballestar E. Microglial innate memory and epigenetic reprogramming in neurological disorders. Prog Neurobiol 2020; 200:101971. [PMID: 33309803 DOI: 10.1016/j.pneurobio.2020.101971] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/30/2020] [Accepted: 12/06/2020] [Indexed: 02/06/2023]
Abstract
Microglia are myeloid-derived cells recognized as brain-resident macrophages. They act as the first and main line of immune defense in the central nervous system (CNS). Microglia have high phenotypic plasticity and are essential for regulating healthy brain homeostasis, and their dysregulation underlies the onset and progression of several CNS pathologies through impaired inflammatory responses. Aberrant microglial activation, following an inflammatory insult, is associated with epigenetic dysregulation in various CNS pathologies. Emerging data suggest that certain stimuli to myeloid cells determine enhanced or attenuated responses to subsequent stimuli. These phenomena, generally termed innate immune memory (IIM), are highly dependent on epigenetic reprogramming. Microglial priming has been reported in several neurological diseases and corresponds to a state of increased permissiveness or exacerbated response, promoted by continuous exposure to a chronic pro-inflammatory environment. In this article, we provide extensive evidence of these epigenetic-mediated phenomena under neurological conditions and discuss their contribution to pathogenesis and their clinical implications, including those concerning potential novel therapeutic approaches.
Collapse
Affiliation(s)
- Ricardo Martins-Ferreira
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916, Badalona, Barcelona, Spain; Immunogenetics Lab, Unit for Multidisciplinary Research in Biomedicine (UMIB), Instituto De Ciências Biomédicas Abel Salazar - Universidade Do Porto (ICBAS-UPorto), Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Barbara Leal
- Immunogenetics Lab, Unit for Multidisciplinary Research in Biomedicine (UMIB), Instituto De Ciências Biomédicas Abel Salazar - Universidade Do Porto (ICBAS-UPorto), Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Paulo Pinho Costa
- Immunogenetics Lab, Unit for Multidisciplinary Research in Biomedicine (UMIB), Instituto De Ciências Biomédicas Abel Salazar - Universidade Do Porto (ICBAS-UPorto), Rua Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916, Badalona, Barcelona, Spain.
| |
Collapse
|
13
|
Petrozziello T, Mills AN, Vaine CA, Penney EB, Fernandez-Cerado C, Legarda GPA, Velasco-Andrada MS, Acuña PJ, Ang MA, Muñoz EL, Diesta CCE, Macalintal-Canlas R, Acuña-Sunshine G, Ozelius LJ, Sharma N, Bragg DC, Sadri-Vakili G. Neuroinflammation and histone H3 citrullination are increased in X-linked Dystonia Parkinsonism post-mortem prefrontal cortex. Neurobiol Dis 2020; 144:105032. [PMID: 32739252 DOI: 10.1016/j.nbd.2020.105032] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/17/2020] [Accepted: 07/26/2020] [Indexed: 12/16/2022] Open
Abstract
Neuroinflammation plays a pathogenic role in neurodegenerative diseases and recent findings suggest that it may also be involved in X-linked Dystonia-Parkinsonism (XDP) pathogenesis. Previously, fibroblasts and neuronal stem cells derived from XDP patients demonstrated hypersensitivity to TNF-α, dysregulation in NFκB signaling, and an increase in several pro-inflammatory markers. However, the role of inflammatory processes in XDP patient brain remains unknown. Here we demonstrate that there is a significant increase in astrogliosis and microgliosis in human post-mortem XDP prefrontal cortex (PFC) compared to control. Furthermore, there is a significant increase in histone H3 citrullination (H3R2R8R17cit3) with a concomitant increase in peptidylarginine deaminase 2 (PAD2) and 4 (PAD4), the enzymes catalyzing citrullination, in XDP post-mortem PFC. While there is a significant increase in myeloperoxidase (MPO) levels in XDP PFC, neutrophil elastase (NE) levels are not altered, suggesting that MPO may be released by activated microglia or reactive astrocytes in the brain. Similarly, there was an increase in H3R2R8R17cit3, PAD2 and PAD4 levels in XDP-derived fibroblasts. Importantly, treatment of fibroblasts with Cl-amidine, a pan inhibitor of PAD enzymes, reduced histone H3 citrullination and pro-inflammatory chemokine expression, without affecting cell survival. Taken together, our results demonstrate that inflammation is increased in XDP post-mortem brain and fibroblasts and unveil a new epigenetic potential therapeutic target.
Collapse
Affiliation(s)
- Tiziana Petrozziello
- NeuroEpigenetics Laboratory, Healey Center for ALS at Mass General, Massachusetts General Hospital, Boston, MA 02129, United States of America
| | - Alexandra N Mills
- NeuroEpigenetics Laboratory, Healey Center for ALS at Mass General, Massachusetts General Hospital, Boston, MA 02129, United States of America
| | - Christine A Vaine
- The Collaborative Center for X-linked Dystonia-Parkinsonism, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, United States of America
| | - Ellen B Penney
- The Collaborative Center for X-linked Dystonia-Parkinsonism, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, United States of America
| | | | | | | | - Patrick J Acuña
- The Collaborative Center for X-linked Dystonia-Parkinsonism, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, United States of America; Sunshine Care Foundation, Roxas City, 5800, Capiz, Philippines
| | - Mark A Ang
- Department of Pathology, College of Medicine, University of the Philippines, Manila, Philippines
| | - Edwin L Muñoz
- Department of Pathology, College of Medicine, University of the Philippines, Manila, Philippines
| | | | | | - Geraldine Acuña-Sunshine
- The Collaborative Center for X-linked Dystonia-Parkinsonism, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, United States of America; Sunshine Care Foundation, Roxas City, 5800, Capiz, Philippines
| | - Laurie J Ozelius
- The Collaborative Center for X-linked Dystonia-Parkinsonism, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, United States of America
| | - Nutan Sharma
- The Collaborative Center for X-linked Dystonia-Parkinsonism, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, United States of America
| | - D Cristopher Bragg
- The Collaborative Center for X-linked Dystonia-Parkinsonism, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, United States of America
| | - Ghazaleh Sadri-Vakili
- NeuroEpigenetics Laboratory, Healey Center for ALS at Mass General, Massachusetts General Hospital, Boston, MA 02129, United States of America.
| |
Collapse
|
14
|
Hashemi R, Hosseini-Asl SS, Arefhosseini SR, Morshedi M. The impact of vitamin D3 intake on inflammatory markers in multiple sclerosis patients and their first-degree relatives. PLoS One 2020; 15:e0231145. [PMID: 32251441 PMCID: PMC7135246 DOI: 10.1371/journal.pone.0231145] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 03/17/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND & AIMS In our previous study, a Seesaw model was proposed for the fluctuation of crucial anti- (IL-10) and pro-inflammatory (Il-6 & IL-17A) cytokines through vitamin D3. In this paper, however, it is intended to extend the mentioned model by assessing the expression mRNA levels of IL-27 and TGF-β1 as well as the changes of plasma levels of IL-27, TGF-β1, IL-17A, IL-10, and IL-6 after treatment by vitamin D3. METHOD Venous blood samples were drawn from Healthy Participants (HP, n = 25) and First-Degree Relative Participants (FDRP, n = 25) as control groups and Multiple Sclerosis Participants (MSP, n = 25) before and after eight weeks of supplementation with 50000 IU vitamin D3. The mRNA expression and plasma concentrations were gauged by using Real-Time PCR and ELISA assay, respectively. RESULTS The mRNA surfaces of IL-27, as well as TGF-β1, were up-regulated. However, the plasma levels of TGF-β1, IL-17A, and IL-6 were significantly different among the three groups. In addition, the plasma levels of IL-27, TGF-β1, IL-10, IL-17A, and IL-6 significantly changed following the administration of vitamin D3. CONCLUSION The findings of this paper illustrate that anti-inflammatory cytokines could have a key role in immunomodulatory functions due to their anti-inflammatory functions. To conclude, this might contribute to preventing the pathophysiological process of MS. Also, the proposed model could be used as a preventive way on disposed people to multiple sclerosis, particularly in first degree relatives of these patients.
Collapse
Affiliation(s)
- Reza Hashemi
- School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, School of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Saeed Hosseini-Asl
- Department of Genetics, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Seyed Rafie Arefhosseini
- Nutrition Research Center, School of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry and Diet Therapy, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Morshedi
- School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Kaur J, Daoud A, Eblen ST. Targeting Chromatin Remodeling for Cancer Therapy. Curr Mol Pharmacol 2020; 12:215-229. [PMID: 30767757 PMCID: PMC6875867 DOI: 10.2174/1874467212666190215112915] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/25/2019] [Accepted: 01/31/2019] [Indexed: 12/31/2022]
Abstract
Background: Epigenetic alterations comprise key regulatory events that dynamically alter gene expression and their deregulation is commonly linked to the pathogenesis of various diseases, including cancer. Unlike DNA mutations, epigenetic alterations involve modifications to proteins and nucleic acids that regulate chromatin structure without affecting the underlying DNA sequence, altering the accessibility of the transcriptional machinery to the DNA, thus modulating gene expression. In cancer cells, this often involves the silencing of tumor suppressor genes or the increased expression of genes involved in oncogenesis. Advances in laboratory medicine have made it possible to map critical epigenetic events, including histone modifications and DNA methylation, on a genome-wide scale. Like the identification of genetic mutations, mapping of changes to the epigenetic landscape has increased our understanding of cancer progression. However, in contrast to irreversible genetic mutations, epigenetic modifications are flexible and dynamic, thereby making them promising therapeutic targets. Ongoing studies are evaluating the use of epigenetic drugs in chemotherapy sensitization and immune system modulation. With the preclinical success of drugs that modify epigenetics, along with the FDA approval of epigenetic drugs including the DNA methyltransferase 1 (DNMT1) inhibitor 5-azacitidine and the histone deacetylase (HDAC) inhibitor vorinostat, there has been a rise in the number of drugs that target epigenetic modulators over recent years. Conclusion: We provide an overview of epigenetic modulations, particularly those involved in cancer, and discuss the recent advances in drug development that target these chromatin-modifying events, primarily focusing on novel strategies to regulate the epigenome.
Collapse
Affiliation(s)
- Jasmine Kaur
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Abdelkader Daoud
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Scott T Eblen
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, United States
| |
Collapse
|
16
|
Jamebozorgi K, Rostami D, Pormasoumi H, Taghizadeh E, Barreto GE, Sahebkar A. Epigenetic aspects of multiple sclerosis and future therapeutic options. Int J Neurosci 2020; 131:56-64. [PMID: 32075477 DOI: 10.1080/00207454.2020.1732974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disease accompanied by demyelination of neurons in the central nervous system that mostly affects young adults, especially women. This disease has two phases including relapsing-remitting form (RR-MS) by episodes of relapse and periods of clinical remission and secondary-progressive form (SP-MS), which causes more disability. The inheritance pattern of MS is not exactly identified and there is an agreement that it has a complex pattern with an interplay among environmental, genetic and epigenetic alternations. Epigenetic mechanisms that are identified for MS pathogenesis are DNA methylation, histone modification and some microRNAs' alternations. Several cellular processes including apoptosis, differentiation and evolution can be modified along with epigenetic changes. Some alternations are associated with epigenetic mechanisms in MS patients and these changes can become key points for MS therapy. Therefore, the aim of this review was to discuss epigenetic mechanisms that are associated with MS pathogenesis and future therapeutic approaches.
Collapse
Affiliation(s)
| | - Daryoush Rostami
- School of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Hosein Pormasoumi
- Faculty of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Eskandar Taghizadeh
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.,Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Reworking GWAS Data to Understand the Role of Nongenetic Factors in MS Etiopathogenesis. Genes (Basel) 2020; 11:genes11010097. [PMID: 31947683 PMCID: PMC7017269 DOI: 10.3390/genes11010097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/03/2020] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
Genome-wide association studies have identified more than 200 multiple sclerosis (MS)-associated loci across the human genome over the last decade, suggesting complexity in the disease etiology. This complexity poses at least two challenges: the definition of an etiological model including the impact of nongenetic factors, and the clinical translation of genomic data that may be drivers for new druggable targets. We reviewed studies dealing with single genes of interest, to understand how MS-associated single nucleotide polymorphism (SNP) variants affect the expression and the function of those genes. We then surveyed studies on the bioinformatic reworking of genome-wide association studies (GWAS) data, with aggregate analyses of many GWAS loci, each contributing with a small effect to the overall disease predisposition. These investigations uncovered new information, especially when combined with nongenetic factors having possible roles in the disease etiology. In this context, the interactome approach, defined as “modules of genes whose products are known to physically interact with environmental or human factors with plausible relevance for MS pathogenesis”, will be reported in detail. For a future perspective, a polygenic risk score, defined as a cumulative risk derived from aggregating the contributions of many DNA variants associated with a complex trait, may be integrated with data on environmental factors affecting the disease risk or protection.
Collapse
|
18
|
Hasan A, Afzal M. Gene and environment interplay in cognition: Evidence from twin and molecular studies, future directions and suggestions for effective candidate gene x environment (cGxE) research. Mult Scler Relat Disord 2019; 33:121-130. [PMID: 31185373 DOI: 10.1016/j.msard.2019.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 04/20/2019] [Accepted: 05/13/2019] [Indexed: 12/17/2022]
Abstract
Last decade of molecular research in the field of cognitive science has shown that no single approach can give satisfactory results as far as gene hunt is concerned. Cohesive theory of gene-environment interaction seems to be a rational idea for bridging the gap in our knowledge of disorders involving cognitive deficit. It may even be helpful to some extent in resolving issues of missing heritability. We review the current state of play in the area of cognition at genetic and environmental fronts. Evidence of apparent gene-environment (GxE) interactions from various studies has been mentioned with the aim of redirecting the focus of research community towards studying such interactions with the help of sensitive designs and molecular techniques. We re-evaluate candidate gene-environment research in order to emphasize its potential if carried out strategically.
Collapse
Affiliation(s)
- Anam Hasan
- Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Mohammad Afzal
- Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India.
| |
Collapse
|
19
|
Jakimovski D, Guan Y, Ramanathan M, Weinstock-Guttman B, Zivadinov R. Lifestyle-based modifiable risk factors in multiple sclerosis: review of experimental and clinical findings. Neurodegener Dis Manag 2019; 9:149-172. [PMID: 31116081 DOI: 10.2217/nmt-2018-0046] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is a lifelong inflammatory and neurodegenerative disease influenced by multiple lifestyle-based factors. We provide a narrative review of the effects of modifiable risk factors that are identified as being associated with risk to develop MS and/or influencing the future clinical disease outcomes. The emerging data regarding the beneficial effects of diet modifications and exercise are further reviewed. In contrast, obesity and comorbid cardiovascular diseases are associated with increased MS susceptibility and worse disease progression. In addition, the potential influence of smoking, coffee and alcohol consumption on MS onset and disability development are discussed. Successful management of the modifiable risk factors may lead to better long-term outcomes and improve patients' quality of life. MS specialists should participate in educating and facilitating lifestyle-based modifications as part of their neurological consults.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Yi Guan
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Murali Ramanathan
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Bianca Weinstock-Guttman
- Jacobs MS Center, Department of Neurology, Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA.,Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| |
Collapse
|
20
|
Yazdanpanahi N, Etemadifar M, Shams E. DNMT3B-579G>T (rs1569686G>T) polymorphism and the risk of multiple sclerosis in a subset of Iranian population. IRANIAN JOURNAL OF NEUROLOGY 2019; 18:70-75. [PMID: 31565203 PMCID: PMC6755508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Background: Deoxyribonucleic acid (DNA) methyltransferase 3 beta (DNMT3B) gene encodes an MT enzyme involving in de novo methylation of DNA. The present investigation aimed to explore the association of DNMT3B-579G>T (rs1569686) polymorphism with multiple sclerosis (MS). Methods: 130 Iranian patients with MS and 130 controls were genotyped for the DNMT3B-579G>T using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Results: There was no statistically significant association between DNMT3B-579G>T and susceptibility to MS. The alleles and genotypes of DNMT3B-579G>T did not have different risks of MS development under various models [T vs. G (P = 0.86); GTvs. GG (P = 0.48); TT vs. GG (P > 0.99); GT+TT vs. GG (P = 0.60), and TT vs. GG+GT (P = 0.87)]. Also, there was no statistically significant association between genotypes and clinical and demographic characteristics of patients (P > 0.05). Conclusion: The current findings suggest that DNMT3B-579G>T is probably not a crucial potential risk marker in molecular diagnostics of MS among Iranian. However, to the best of our knowledge, this is the first genetic association study about the DNMT3B polymorphisms and MS. Therefore, further surveys should be included to estimate the exact relevance of DNMT3B gene to the development of autoimmune disorders like MS.
Collapse
Affiliation(s)
- Nasrin Yazdanpanahi
- Department of Biology and Genetics, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Masoud Etemadifar
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran,Isfahan Multiple Sclerosis Research Center, Isfahan, Iran
| | - Elaheh Shams
- Young Researchers and Elite Club, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| |
Collapse
|
21
|
Wang Z, Xie J, Wu C, Xiao G. Correlation Between Smoking and Passive Smoking with Multiple Sclerosis and the Underlying Molecular Mechanisms. Med Sci Monit 2019; 25:893-902. [PMID: 30703074 PMCID: PMC6367889 DOI: 10.12659/msm.912863] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic immune-mediated disease of the spinal cord and brain. Many studies have shown that smoking and passive smoking are key environmental risk factors for MS. Here, we provide an overview of the human leukocyte antigen (HLA) gene studies on smoking and MS risk, and we discuss recent studies on between epigenetics and smoking-induced MS. In addition, in this review we also summarize current research advances in biological pathways and smoking-induced MS. This review provides an overview of studies on the association between smoking, passive smoking, and MS susceptibility, and the underlying molecular mechanism.
Collapse
Affiliation(s)
- Zhaowei Wang
- Department of Neurology, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Jianpin Xie
- Department of Neurology, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Chenglong Wu
- Department of Neurology, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Guirong Xiao
- Department of Neurology, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| |
Collapse
|
22
|
Morris G, Maes M, Murdjeva M, Puri BK. Do Human Endogenous Retroviruses Contribute to Multiple Sclerosis, and if So, How? Mol Neurobiol 2018; 56:2590-2605. [PMID: 30047100 PMCID: PMC6459794 DOI: 10.1007/s12035-018-1255-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/17/2018] [Indexed: 12/24/2022]
Abstract
The gammaretroviral human endogenous retrovirus (HERV) families MRSV/HERV-W and HERV-H (including the closely related HERV-Fc1) are associated with an increased risk of multiple sclerosis (MS). Complete HERV sequences betray their endogenous retroviral origin, with open reading frames in gag, pro, pol and env being flanked by two long terminal repeats containing promoter and enhancer sequences with the capacity to regulate HERV transactivation and the activity of host genes in spite of endogenous epigenetic repression mechanisms. HERV virions, RNA, cDNA, Gag and Env, and antibodies to HERV transcriptional products, have variously been found in the blood and/or brain and/or cerebrospinal fluid of MS patients, with the HERV expression level being associated with disease status. Furthermore, some HERV-associated single nucleotide polymorphisms (SNPs), such as rs662139 T/C in a 3-kb region of Xq22.3 containing a HERV-W env locus, and rs391745, upstream of the HERV-Fc1 locus on the X chromosome, are associated with MS susceptibility, while a negative association has been reported with SNPs in the tripartite motif-containing (TRIM) protein-encoding genes TRIM5 and TRIM22. Factors affecting HERV transcription include immune activation and inflammation, since HERV promoter regions possess binding sites for related transcription factors; oxidative stress, with oxidation of guanine to 8-oxoguanine and conversion of cytosine to 5-hydroxymethylcytosine preventing binding of methyl groups transferred by DNA methyltransferases; oxidative stress also inhibits the activity of deacetylases, thereby favouring the acetylation of histone lysine residues favouring gene expression; interferon beta; natalizumab treatment; impaired epigenetic regulation; and the sex of patients.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Marianna Murdjeva
- Department of Microbiology and Immunology, Medical University, Plovdiv, Bulgaria
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|