1
|
Nalbant E, Akkaya-Ulum YZ. Exploring regulatory mechanisms on miRNAs and their implications in inflammation-related diseases. Clin Exp Med 2024; 24:142. [PMID: 38958690 PMCID: PMC11222192 DOI: 10.1007/s10238-024-01334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/20/2024] [Indexed: 07/04/2024]
Abstract
This comprehensive exploration delves into the pivotal role of microRNAs (miRNAs) within the intricate tapestry of cellular regulation. As potent orchestrators of gene expression, miRNAs exhibit diverse functions in cellular processes, extending their influence from the nucleus to the cytoplasm. The complex journey of miRNA biogenesis, involving transcription, processing, and integration into the RNA-induced silencing complex, showcases their versatility. In the cytoplasm, mature miRNAs finely tune cellular functions by modulating target mRNA expression, while their reach extends into the nucleus, influencing transcriptional regulation and epigenetic modifications. Dysregulation of miRNAs becomes apparent in various pathologies, such as cancer, autoimmune diseases, and inflammatory conditions. The adaptability of miRNAs to environmental signals, interactions with transcription factors, and involvement in intricate regulatory networks underscore their significance. DNA methylation and histone modifications adds depth to understanding the dynamic regulation of miRNAs. Mechanisms like competition with RNA-binding proteins, sponging, and the control of miRNA levels through degradation and editing contribute to this complex regulation process. In this review, we mainly focus on how dysregulation of miRNA expression can be related with skin-related autoimmune and autoinflammatory diseases, arthritis, cardiovascular diseases, inflammatory bowel disease, autoimmune and autoinflammatory diseases, and neurodegenerative disorders. We also emphasize the multifaceted roles of miRNAs, urging continued research to unravel their complexities. The mechanisms governing miRNA functions promise advancements in therapeutic interventions and enhanced insights into cellular dynamics in health and disease.
Collapse
Affiliation(s)
- Emre Nalbant
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, 06100, Sihhiye, Ankara, Türkiye
| | - Yeliz Z Akkaya-Ulum
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, 06100, Sihhiye, Ankara, Türkiye.
| |
Collapse
|
2
|
Saadh MJ, Mahdi MS, Allela OQB, Alazzawi TS, Ubaid M, Rakhimov NM, Athab ZH, Ramaiah P, Chinnasamy L, Alsaikhan F, Farhood B. Critical role of miR-21/exosomal miR-21 in autophagy pathway. Pathol Res Pract 2024; 257:155275. [PMID: 38643552 DOI: 10.1016/j.prp.2024.155275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/23/2024]
Abstract
Activation of autophagy, a process of cellular stress response, leads to the breakdown of proteins, organelles, and other parts of the cell in lysosomes, and can be linked to several ailments, such as cancer, neurological diseases, and rare hereditary syndromes. Thus, its regulation is very carefully monitored. Transcriptional and post-translational mechanisms domestically or in whole organisms utilized to control the autophagic activity, have been heavily researched. In modern times, microRNAs (miRNAs) are being considered to have a part in post-translational orchestration of the autophagic activity, with miR-21 as one of the best studied miRNAs, it is often more than expressed in cancer cells. This regulatory RNA is thought to play a major role in a plethora of processes and illnesses including growth, cancer, cardiovascular disease, and inflammation. Different studies have suggested that a few autophagy-oriented genes, such as PTEN, Rab11a, Atg12, SIPA1L2, and ATG5, are all targeted by miR-21, indicating its essential role in the regulation.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | | | | | - Tuqa S Alazzawi
- College of dentist, National University of Science and Technology, Dhi Qar, Iraq
| | | | - Nodir M Rakhimov
- Department of Oncology, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan; Department of Oncology, Tashkent State Dental Institute, Tashkent, Uzbekistan
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia jSchool of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
3
|
Maltby V, Xavier A, Ewing E, Campagna MP, Sampangi S, Scott RJ, Butzkueven H, Jokubaitis V, Kular L, Bos S, Slee M, van der Mei IA, Taylor BV, Ponsonby AL, Jagodic M, Lea R, Lechner-Scott J. Evaluation of Cell-Specific Epigenetic Age Acceleration in People With Multiple Sclerosis. Neurology 2023; 101:e679-e689. [PMID: 37541839 PMCID: PMC10437016 DOI: 10.1212/wnl.0000000000207489] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/20/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND AND OBJECTIVES In multiple sclerosis (MS), accelerated aging of the immune system (immunosenescence) may be associated with disease onset or drive progression. DNA methylation (DNAm) is an epigenetic factor that varies among lymphocyte subtypes, and cell-specific DNAm is associated with MS. DNAm varies across the life span and can be used to accurately estimate biological age acceleration, which has been linked to a range of morbidities. The objective of this study was to test for cell-specific epigenetic age acceleration (EAA) in people with MS. METHODS This was a case-control study of EAA using existing DNAm data from several independent previously published studies. Data were included if .idat files from Illumina 450K or EPIC arrays were available for both a case with MS and an age-matched and sex-matched control, from the same study. Multifactor statistical modeling was performed to assess the primary outcome of EAA. We explored the relationship of EAA and MS, including interaction terms to identify immune cell-specific effects. Cell-sorted DNA methylation data from 3 independent datasets were used to validate findings. RESULTS We used whole blood DNA methylation data from 583 cases with MS and 643 non-MS controls to calculate EAA using the GrimAge algorithm. The MS group exhibited an increased EAA compared with controls (approximately 9 mths, 95% CI 3.6-14.4), p = 0.001). Statistical deconvolution showed that EAA is associated with MS in a B cell-dependent manner (β int = 1.7, 95% CI 0.3-2.8), p = 0.002), irrespective of B-cell proportions. Validation analysis using 3 independent datasets enriched for B cells showed an EAA increase of 5.1 years in cases with MS compared with that in controls (95% CI 2.8-7.4, p = 5.5 × 10-5). By comparison, there was no EAA difference in MS in a T cell-enriched dataset. We found that EAA was attributed to the DNAm surrogates for Beta-2-microglobulin (difference = 47,546, 95% CI 10,067-85,026; p = 7.2 × 10-5), and smoking pack-years (difference = 8.1, 95% CI 1.9-14.2, p = 0.002). DISCUSSION This study provides compelling evidence that B cells exhibit marked EAA in MS and supports the hypothesis that premature B-cell immune senescence plays a role in MS. Future MS studies should focus on age-related molecular mechanisms in B cells.
Collapse
Affiliation(s)
- Vicki Maltby
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Alexandre Xavier
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Ewoud Ewing
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Maria-Pia Campagna
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Sandeep Sampangi
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Rodney J Scott
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia.
| | - Helmut Butzkueven
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Vilija Jokubaitis
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Lara Kular
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Steffan Bos
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Mark Slee
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Ingrid A van der Mei
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Bruce V Taylor
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Anne-Louise Ponsonby
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Maja Jagodic
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Rodney Lea
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia
| | - Jeannette Lechner-Scott
- From the School of Medicine and Public Health (V.M., R.L., J.L.-S.), University of Newcastle, University Drive, Callaghan; Immune Health Program (V.M., A.X., J.L.-S.), Hunter Medical Research Institute; Department of Neurology (V.M., J.L.-S.), John Hunter Hospital, New Lambton Heights; School of Biomedical Sciences and Pharmacy (A.X.), University of Newcastle, University Drive, Callaghan, Australia; Department of Clinical Neuroscience (E.E., L.K., M.J.), Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Neuroscience (M.-P.C., S.S., H.B., V.J.), Central Clinical School, Monash University, Victoria; Division of Molecular Genetics (R.J.S.), Pathology North, John Hunter Hospital, New Lambton Heights; MSBase Foundation (H.B.), Melbourne, Australia; Institute of Clinical Medicine (S.B.), University of Oslo,; Department of Neurology (S.B.), Oslo University Hospital, Norway; Flinders University (M.S.), Adelaide; Menzies Institute for Medical Research (I.A.M., B.V.T.), University of Tasmania, Hobart; Florey Institute of Neuroscience and Mental Health (A.-L.P.), The University of Melbourne; Centre of Epidemiology and Biostatistics (A.-L.P.), School of Population and Global Health, University of Melbourne; Murdoch Children's Research Institute (A.-L.P.), Royal Children's Hospital, Melbourne; and Centre for Genomics and Personalized Health (R.L.), School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Australia.
| |
Collapse
|
4
|
Boziki M, Theotokis P, Kesidou E, Karafoulidou E, Konstantinou C, Michailidou I, Bahar Y, Altintas A, Grigoriadis N. Sex, aging and immunity in multiple sclerosis and experimental autoimmune encephalomyelitis: An intriguing interaction. Front Neurol 2023; 13:1104552. [PMID: 36698908 PMCID: PMC9869255 DOI: 10.3389/fneur.2022.1104552] [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: 11/21/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) with a profound neurodegenerative component early in the disease pathogenesis. Age is a factor with a well-described effect on the primary disease phenotype, namely, the relapsing-remitting vs. the primary progressive disease. Moreover, aging is a prominent factor contributing to the transition from relapsing-remitting MS (RRMS) to secondary progressive disease. However, sex also seems to, at least in part, dictate disease phenotype and evolution, as evidenced in humans and in animal models of the disease. Sex-specific gene expression profiles have recently elucidated an association with differential immunological signatures in the context of experimental disease. This review aims to summarize current knowledge stemming from experimental autoimmune encephalomyelitis (EAE) models regarding the effects of sex, either independently or as a factor combined with aging, on disease phenotype, with relevance to the immune system and the CNS.
Collapse
Affiliation(s)
- Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Karafoulidou
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Chrystalla Konstantinou
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Iliana Michailidou
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Ayse Altintas
- School of Medicine, Koç University, Istanbul, Turkey
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology and Multiple Sclerosis Center, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece,*Correspondence: Nikolaos Grigoriadis ✉
| |
Collapse
|
5
|
Campagna MP, Xavier A, Lea RA, Stankovich J, Maltby VE, Butzkueven H, Lechner-Scott J, Scott RJ, Jokubaitis VG. Whole-blood methylation signatures are associated with and accurately classify multiple sclerosis disease severity. Clin Epigenetics 2022; 14:194. [PMID: 36585691 PMCID: PMC9805090 DOI: 10.1186/s13148-022-01397-2] [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: 08/08/2022] [Accepted: 12/02/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The variation in multiple sclerosis (MS) disease severity is incompletely explained by genetics, suggesting genetic and environmental interactions are involved. Moreover, the lack of prognostic biomarkers makes it difficult for clinicians to optimise care. DNA methylation is one epigenetic mechanism by which gene-environment interactions can be assessed. Here, we aimed to identify DNA methylation patterns associated with mild and severe relapse-onset MS (RMS) and to test the utility of methylation as a predictive biomarker. METHODS We conducted an epigenome-wide association study between 235 females with mild (n = 119) or severe (n = 116) with RMS. Methylation was measured with the Illumina methylationEPIC array and analysed using logistic regression. To generate hypotheses about the functional consequence of differential methylation, we conducted gene set enrichment analysis using ToppGene. We compared the accuracy of three machine learning models in classifying disease severity: (1) clinical data available at baseline (age at onset and first symptoms) built using elastic net (EN) regression, (2) methylation data using EN regression and (3) a weighted methylation risk score of differentially methylated positions (DMPs) from the main analysis using logistic regression. We used a conservative 70:30 test:train split for classification modelling. A false discovery rate threshold of 0.05 was used to assess statistical significance. RESULTS Females with mild or severe RMS had 1472 DMPs in whole blood (839 hypermethylated, 633 hypomethylated in the severe group). Differential methylation was enriched in genes related to neuronal cellular compartments and processes, and B-cell receptor signalling. Whole-blood methylation levels at 1708 correlated CpG sites classified disease severity more accurately (machine learning model 2, AUC = 0.91) than clinical data (model 1, AUC = 0.74) or the wMRS (model 3, AUC = 0.77). Of the 1708 selected CpGs, 100 overlapped with DMPs from the main analysis at the gene level. These overlapping genes were enriched in neuron projection and dendrite extension, lending support to our finding that neuronal processes, rather than immune processes, are implicated in disease severity. CONCLUSION RMS disease severity is associated with whole-blood methylation at genes related to neuronal structure and function. Moreover, correlated whole-blood methylation patterns can assign disease severity in females with RMS more accurately than clinical data available at diagnosis.
Collapse
Affiliation(s)
- Maria Pia Campagna
- grid.1002.30000 0004 1936 7857Central Clinical School, Monash University, Melbourne, VIC Australia
| | - Alexandre Xavier
- grid.266842.c0000 0000 8831 109XHunter Medical Research Institute, University of Newcastle, Newcastle, NSW Australia
| | - Rodney A. Lea
- grid.1024.70000000089150953Queensland University of Technology, Brisbane, QLD Australia ,grid.1008.90000 0001 2179 088XUniversity of Melbourne, Melbourne, VIC Australia
| | - Jim Stankovich
- grid.1002.30000 0004 1936 7857Monash University, Melbourne, VIC Australia
| | - Vicki E. Maltby
- grid.266842.c0000 0000 8831 109XHunter Medical Research Institute, University of Newcastle, Newcastle, NSW Australia
| | - Helmut Butzkueven
- grid.1002.30000 0004 1936 7857Monash University, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XUniversity of Melbourne, Melbourne, VIC Australia ,grid.416153.40000 0004 0624 1200Royal Melbourne Hospital, Melbourne, VIC Australia ,grid.414366.20000 0004 0379 3501Neurology Department, Eastern Health, Melbourne, VIC Australia ,grid.267362.40000 0004 0432 5259Neurology Department, Alfred Health, Melbourne, VIC Australia
| | - Jeannette Lechner-Scott
- grid.266842.c0000 0000 8831 109XHunter Medical Research Institute, University of Newcastle, Newcastle, NSW Australia ,grid.3006.50000 0004 0438 2042Neurology Department, John Hunter Hospital, Hunter New England Health, Newcastle, NSW Australia
| | - Rodney J. Scott
- grid.266842.c0000 0000 8831 109XSchool of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW Australia ,Division of Molecular Medicine, New South Wales Health Pathology North, Newcastle, NSW Australia
| | - Vilija G. Jokubaitis
- grid.1002.30000 0004 1936 7857Monash University, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XUniversity of Melbourne, Melbourne, VIC Australia ,grid.416153.40000 0004 0624 1200Royal Melbourne Hospital, Melbourne, VIC Australia ,grid.267362.40000 0004 0432 5259Neurology Department, Alfred Health, Melbourne, VIC Australia
| |
Collapse
|
6
|
Cappelletti C, Eriksson A, Brorson IS, Leikfoss IS, Kråbøl O, Høgestøl EA, Vitelli V, Mjaavatten O, Harbo HF, Berven F, Bos SD, Berge T. Quantitative proteomics reveals protein dysregulation during T cell activation in multiple sclerosis patients compared to healthy controls. Clin Proteomics 2022; 19:23. [PMID: 35790914 PMCID: PMC9254507 DOI: 10.1186/s12014-022-09361-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 06/22/2022] [Indexed: 12/20/2022] Open
Abstract
Background Multiple sclerosis (MS) is an autoimmune, neurodegenerative disorder with a strong genetic component that acts in a complex interaction with environmental factors for disease development. CD4+ T cells are pivotal players in MS pathogenesis, where peripherally activated T cells migrate to the central nervous system leading to demyelination and axonal degeneration. Through a proteomic approach, we aim at identifying dysregulated pathways in activated T cells from MS patients as compared to healthy controls. Methods CD4+ T cells were purified from peripheral blood from MS patients and healthy controls by magnetic separation. Cells were left unstimulated or stimulated in vitro through the TCR and costimulatory CD28 receptor for 24 h prior to sampling. Electrospray liquid chromatography-tandem mass spectrometry was used to measure protein abundances. Results Upon T cell activation the abundance of 1801 proteins was changed. Among these proteins, we observed an enrichment of proteins expressed by MS-susceptibility genes. When comparing protein abundances in T cell samples from healthy controls and MS patients, 18 and 33 proteins were differentially expressed in unstimulated and stimulated CD4+ T cells, respectively. Moreover, 353 and 304 proteins were identified as proteins exclusively induced upon T cell activation in healthy controls and MS patients, respectively and dysregulation of the Nur77 pathway was observed only in samples from MS patients. Conclusions Our study highlights the importance of CD4+ T cell activation for MS, as proteins that change in abundance upon T cell activation are enriched for proteins encoded by MS susceptibility genes. The results provide evidence for proteomic disturbances in T cell activation in MS, and pinpoint to dysregulation of the Nur77 pathway, a biological pathway known to limit aberrant effector T cell responses.
Collapse
|
7
|
Wang L, Liang Y. MicroRNAs as T Lymphocyte Regulators in Multiple Sclerosis. Front Mol Neurosci 2022; 15:865529. [PMID: 35548667 PMCID: PMC9082748 DOI: 10.3389/fnmol.2022.865529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/30/2022] [Indexed: 01/22/2023] Open
Abstract
MicroRNA (miRNA) is a class of endogenous non-coding small RNA with regulatory activities, which generally regulates the expression of target genes at the post-transcriptional level. Multiple Sclerosis (MS) is thought to be an autoimmune-mediated chronic inflammatory demyelinating disease of the central nervous system (CNS) that typically affect young adults. T lymphocytes play an important role in the pathogenesis of MS, and studies have suggested that miRNAs are involved in regulating the proliferation, differentiation, and functional maintenance of T lymphocytes in MS. Dysregulated expression of miRNAs may lead to the differentiation balance and dysfunction of T lymphocytes, and they are thus involved in the occurrence and development of MS. In addition, some specific miRNAs, such as miR-155 and miR-326, may have potential diagnostic values for MS or be useful for discriminating subtypes of MS. Moreover, miRNAs may be a promising therapeutic strategy for MS by regulating T lymphocyte function. By summarizing the recent literature, we reviewed the involvement of T lymphocytes in the pathogenesis of MS, the role of miRNAs in the pathogenesis and disease progression of MS by regulating T lymphocytes, the possibility of differentially expressed miRNAs to function as biomarkers for MS diagnosis, and the therapeutic potential of miRNAs in MS by regulating T lymphocytes.
Collapse
|
8
|
Kong X, Wang J, Cao Y, Lu X, Zhang H, Zhang X, Bo C, Bai M, Li S, Jiao Y, Wang L. Construction of miRNA-regulated drug-pathway network to screen drug repurposing candidates for multiple sclerosis. Medicine (Baltimore) 2022; 101:e29107. [PMID: 35356949 PMCID: PMC10684250 DOI: 10.1097/md.0000000000029107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 02/28/2022] [Indexed: 02/07/2023] Open
Abstract
ABSTRACT Given the high disability rate of multiple sclerosis (MS), there is a need for safer and more effective therapeutic agents. Existing literature highlights the prominent roles of miRNA in MS pathophysiology. Nevertheless, there are few studies that have explored the usefulness of existing drugs in treating MS through potential miRNA-modulating abilities.The current investigation identifies genes that may exacerbate the risk of MS due to their respective miRNA associations. These findings were then used to determine potential drug candidates through the construction of miRNA-regulated drug-pathway network through genes. We uncovered a total of 48 MS risk pathways, 133 MS risk miRNAs, and 186 drugs that can affect these pathways. Potential MS risk miRNAs that are also regulated by therapeutic candidates were hsa05215 and hsa05152. We analyzed the properties of the miRNA-regulated drug-pathway network through genes and uncovered a number of novel MS agents by assessing their respective Z-values.A total of 20 likely drug candidates were identified, including human immunoglobulin, aspirin, alemtuzumab, minocycline, abciximab, alefacept, palivizumab, bevacizumab, efalizumab, tositumomab, minocycline, etanercept, catumaxomab, and sarilumab. Each of these agents were then explored with regards to their likely mechanism of action in treating MS.The current investigation provides a fresh perspective on MS biological mechanisms as well as likely treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Lihua Wang
- Correspondence: Lihua Wang, Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin 150086, Heilongjiang Province, China(e-mail: ).
| |
Collapse
|
9
|
Kiselev I, Danilova L, Baulina N, Baturina O, Kabilov M, Boyko A, Kulakova O, Favorova O. Genome-wide DNA methylation profiling identifies epigenetic changes in CD4+ and CD14+ cells of multiple sclerosis patients. Mult Scler Relat Disord 2022; 60:103714. [PMID: 35245816 DOI: 10.1016/j.msard.2022.103714] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/29/2022] [Accepted: 02/24/2022] [Indexed: 10/19/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune and degenerative disease of the central nervous system, which develops in genetically predisposed individuals upon exposure to environmental influences. Environmental triggers of MS, such as viral infections or smoking, were demonstrated to affect DNA methylation, and thus to involve this important epigenetic mechanism in the development of pathological process. To identify MS-associated DNA methylation hallmarks, we performed genome-wide DNA methylation profiling of two cell populations (CD4+ T-lymphocytes and CD14+ monocytes), collected from the same treatment-naive relapsing-remitting MS patients and healthy subjects, using Illumina 450 K methylation arrays. We revealed significant changes in DNA methylation for both cell populations in MS. In CD4+ cells of MS patients the majority of differentially methylated positions (DMPs) were shown to be hypomethylated, while in CD14+ cells - hypermethylated. Differential methylation of HLA-DRB1 gene in CD4+ and CD14+ cells was associated with carriage of DRB1*15 allele independently from the disease status. Besides, about 20% of identified DMPs were shared between two cell populations and had the same direction of methylation changes; they may be involved in basic epigenetic processes occuring in MS. These findings suggest that the epigenetic mechanism of DNA methylation in immune cells contributes to MS; further studies are now required to validate these results and understand their functional significance.
Collapse
Affiliation(s)
- Ivan Kiselev
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Ostrovityanova st. 1, Moscow 117997, Russian Federation
| | - Ludmila Danilova
- Vavilov Institute of General Genetics, Gubkin st. 3, Moscow 119991, Russian Federation; Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Natalia Baulina
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Ostrovityanova st. 1, Moscow 117997, Russian Federation
| | - Olga Baturina
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk 630090, Russian Federation
| | - Marsel Kabilov
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk 630090, Russian Federation
| | - Alexey Boyko
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Ostrovityanova st. 1, Moscow 117997, Russian Federation
| | - Olga Kulakova
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Ostrovityanova st. 1, Moscow 117997, Russian Federation
| | - Olga Favorova
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Ostrovityanova st. 1, Moscow 117997, Russian Federation
| |
Collapse
|
10
|
Bai X, Bian Z. MicroRNA-21 Is a Versatile Regulator and Potential Treatment Target in Central Nervous System Disorders. Front Mol Neurosci 2022; 15:842288. [PMID: 35173580 PMCID: PMC8841607 DOI: 10.3389/fnmol.2022.842288] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of endogenous, non-coding, single-stranded RNAs with a length of approximately 22 nucleotides that are found in eukaryotes. miRNAs are involved in the regulation of cell differentiation, proliferation, invasion, apoptosis, and metabolism by regulating the expression of their target genes. Emerging studies have suggested that various miRNAs play key roles in the pathogenesis of central nervous system (CNS) disorders and may be viable therapeutic targets. In particular, miR-21 has prominently emerged as a focus of increasing research on the mechanisms of its involvement in CNS disorders. Herein, we reviewed recent studies on the critical roles of miR-21, including its dysregulated expression and target genes, in the regulation of pathophysiological processes of CNS disorders, with a special focus on apoptosis and inflammation. Collectively, miR-21 is a versatile regulator in the progression of CNS disorders and could be a promising biomarker and therapeutic target for these diseases. An in-depth understanding of the mechanisms by which miR-21 affects the pathogenesis of CNS disorders could pave the way for miR-21 to serve as a therapeutic target for these conditions.
Collapse
Affiliation(s)
- Xue Bai
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhigang Bian
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Zhigang Bian,
| |
Collapse
|
11
|
Xiong Y, Xiong Y, Zhang H, Zhao Y, Han K, Zhang J, Zhao D, Yu Z, Geng Z, Wang L, Wang Y, Luan X. hPMSCs-Derived Exosomal miRNA-21 Protects Against Aging-Related Oxidative Damage of CD4 + T Cells by Targeting the PTEN/PI3K-Nrf2 Axis. Front Immunol 2021; 12:780897. [PMID: 34887868 PMCID: PMC8649962 DOI: 10.3389/fimmu.2021.780897] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs)-derived exosomes were considered a novel therapeutic approach in many aging-related diseases. This study aimed to clarify the protective effects of human placenta MSCs-derived exosomes (hPMSC-Exo) in aging-related CD4+ T cell senescence and identified the underlying mechanisms using a D-gal induced mouse aging model. Senescent T cells were detected SA-β-gal stain. The degree of DNA damage was evaluated by detecting the level of 8-OH-dG. The superoxide dismutase (SOD) and total antioxidant capacity (T-AOC) activities were measured. The expression of aging-related proteins and senescence-associated secretory phenotype (SASP) were detected by Western blot and RT-PCR. We found that hPMSC-Exo treatment markedly decreased oxidative stress damage (ROS and 8-OH-dG), SA-β-gal positive cell number, aging-related protein expression (p53 and γ-H2AX), and SASP expression (IL-6 and OPN) in senescent CD4+ T cells. Additionally, hPMSC-Exo containing miR-21 effectively downregulated the expression of PTEN, increased p-PI3K and p-AKT expression, and Nrf2 nuclear translocation and the expression of downstream target genes (NQO1 and HO-1) in senescent CD4+ T cells. Furthermore, in vitro studies uncovered that hPMSC-Exo attenuated CD4+ T cell senescence by improving the PTEN/PI3K-Nrf2 axis by using the PTEN inhibitor bpV (HOpic). We also validated that PTEN was a target of miR-21 by using a luciferase reporter assay. Collectively, the obtained results suggested that hPMSC-Exo attenuates CD4+ T cells senescence via carrying miRNA-21 and activating PTEN/PI3K-Nrf2 axis mediated exogenous antioxidant defenses.
Collapse
Affiliation(s)
- Yanlian Xiong
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Yanlei Xiong
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hengchao Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Yaxuan Zhao
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Kaiyue Han
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Jiashen Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Dongmei Zhao
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Zhenhai Yu
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Ziran Geng
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Longfei Wang
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Yueming Wang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Xiying Luan
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| |
Collapse
|
12
|
Contribution of Dysregulated DNA Methylation to Autoimmunity. Int J Mol Sci 2021; 22:ijms222111892. [PMID: 34769338 PMCID: PMC8584328 DOI: 10.3390/ijms222111892] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/22/2021] [Accepted: 10/29/2021] [Indexed: 12/24/2022] Open
Abstract
Epigenetic mechanisms, such as DNA methylation, histone modifications, and non-coding RNAs are known regulators of gene expression and genomic stability in cell growth, development, and differentiation. Because epigenetic mechanisms can regulate several immune system elements, epigenetic alterations have been found in several autoimmune diseases. The purpose of this review is to discuss the epigenetic modifications, mainly DNA methylation, involved in autoimmune diseases in which T cells play a significant role. For example, Rheumatoid Arthritis and Systemic Lupus Erythematosus display differential gene methylation, mostly hypomethylated 5′-C-phosphate-G-3′ (CpG) sites that may associate with disease activity. However, a clear association between DNA methylation, gene expression, and disease pathogenesis must be demonstrated. A better understanding of the impact of epigenetic modifications on the onset of autoimmunity will contribute to the design of novel therapeutic approaches for these diseases.
Collapse
|
13
|
Wang H. MicroRNAs, Multiple Sclerosis, and Depression. Int J Mol Sci 2021; 22:ijms22157802. [PMID: 34360568 PMCID: PMC8346048 DOI: 10.3390/ijms22157802] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 12/16/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic disease of the central nervous system that affects the brain and spinal cord. There are several disease courses in MS including relapsing–remitting MS (RRMS), primary progressive MS (PPMS), and secondary progressive MS (SPMS). Up to 50% of MS patients experience depressive disorders. Major depression (MD) is a serious comorbidity of MS. Many dysfunctions including neuroinflammation, peripheral inflammation, gut dysbiosis, chronic oxidative and nitrosative stress, and neuroendocrine and mitochondrial abnormalities may contribute to the comorbidity between MS and MD. In addition to these actions, medical treatment and microRNA (miRNA) regulation may also be involved in the mechanisms of the comorbidity between MS and MD. In the study, I review many common miRNA biomarkers for both diseases. These common miRNA biomarkers may help further explore the association between MS and MD.
Collapse
Affiliation(s)
- Hsiuying Wang
- Institute of Statistics, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| |
Collapse
|
14
|
Kiselev IS, Kulakova OG, Boyko AN, Favorova OO. DNA Methylation As an Epigenetic Mechanism in the Development of Multiple Sclerosis. Acta Naturae 2021; 13:45-57. [PMID: 34377555 PMCID: PMC8327151 DOI: 10.32607/actanaturae.11043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
The epigenetic mechanisms of gene expression regulation are a group of the key cellular and molecular pathways that lead to inherited alterations in genes' activity without changing their coding sequence. DNA methylation at the C5 position of cytosine in CpG dinucleotides is amongst the central epigenetic mechanisms. Currently, the number of studies that are devoted to the identification of methylation patterns specific to multiple sclerosis (MS), a severe chronic autoimmune disease of the central nervous system, is on a rapid rise. However, the issue of the contribution of DNA methylation to the development of the different clinical phenotypes of this highly heterogeneous disease has only begun to attract the attention of researchers. This review summarizes the data on the molecular mechanisms underlying DNA methylation and the MS risk factors that can affect the DNA methylation profile and, thereby, modulate the expression of the genes involved in the disease's pathogenesis. The focus of our attention is centered on the analysis of the published data on the differential methylation of DNA from various biological samples of MS patients obtained using both the candidate gene approach and high-throughput methods.
Collapse
Affiliation(s)
- I. S. Kiselev
- Pirogov Russian National Research Medical University, Moscow, 117997 Russia
| | - O. G. Kulakova
- Pirogov Russian National Research Medical University, Moscow, 117997 Russia
| | - A. N. Boyko
- Pirogov Russian National Research Medical University, Moscow, 117997 Russia
| | - O. O. Favorova
- Pirogov Russian National Research Medical University, Moscow, 117997 Russia
| |
Collapse
|
15
|
Baulina N, Kiselev I, Favorova O. Imprinted Genes and Multiple Sclerosis: What Do We Know? Int J Mol Sci 2021; 22:1346. [PMID: 33572862 PMCID: PMC7866243 DOI: 10.3390/ijms22031346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune neurodegenerative disease of the central nervous system that arises from interplay between non-genetic and genetic risk factors. The epigenetics functions as a link between these factors, affecting gene expression in response to external influence, and therefore should be extensively studied to improve the knowledge of MS molecular mechanisms. Among others, the epigenetic mechanisms underlie the establishment of parent-of-origin effects that appear as phenotypic differences depending on whether the allele was inherited from the mother or father. The most well described manifestation of parent-of-origin effects is genomic imprinting that causes monoallelic gene expression. It becomes more obvious that disturbances in imprinted genes at the least affecting their expression do occur in MS and may be involved in its pathogenesis. In this review we will focus on the potential role of imprinted genes in MS pathogenesis.
Collapse
Affiliation(s)
- Natalia Baulina
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (I.K.); (O.F.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Ivan Kiselev
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (I.K.); (O.F.)
| | - Olga Favorova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (I.K.); (O.F.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
16
|
Maltby VE, Lea RA, Burnard S, Xavier A, Van Cao T, White N, Kennedy D, Groen K, Sanders KA, Seeto R, Bray S, Gresle M, Laverick L, Butzkueven H, Scott RJ, Lechner-Scott J. Epigenetic differences at the HTR2A locus in progressive multiple sclerosis patients. Sci Rep 2020; 10:22217. [PMID: 33335118 PMCID: PMC7747721 DOI: 10.1038/s41598-020-78809-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 11/22/2020] [Indexed: 11/30/2022] Open
Abstract
The pathology of progressive multiple sclerosis (MS) is poorly understood. We have previously assessed DNA methylation in the CD4+ T cells of relapsing–remitting (RR) MS patients compared to healthy controls and identified differentially methylated regions (DMRs) in HLA-DRB1 and RNF39. This study aimed to investigate the DNA methylation profiles of the CD4+ T cells of progressive MS patients. DNA methylation was measured in two separate case/control cohorts using the Illumina 450K/EPIC arrays and data was analysed with the Chip Analysis Methylation Pipeline (ChAMP). Single nucleotide polymorphisms (SNPs) were assessed using the Illumina Human OmniExpress24 arrays and analysed using PLINK. Expression was assessed using the Illumina HT12 array and analysed in R using a combination of Limma and Illuminaio. We identified three DMRs at HTR2A, SLC17A9 and HDAC4 that were consistent across both cohorts. The DMR at HTR2A is located within the bounds of a haplotype block; however, the DMR remained significant after accounting for SNPs in the region. No expression changes were detected in any DMRs. HTR2A is differentially methylated in progressive MS independent of genotype. This differential methylation is not evident in RRMS, making it a potential biomarker of progressive disease.
Collapse
Affiliation(s)
- Vicki E Maltby
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2308, Australia.,Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Rodney A Lea
- Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.,Institute of Health and Biomedical Innovations, Genomics Research Centre, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia
| | - Sean Burnard
- Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Alexandre Xavier
- Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Thao Van Cao
- Institute of Health and Biomedical Innovations, Genomics Research Centre, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia
| | - Nicole White
- Institute of Health and Biomedical Innovations, Genomics Research Centre, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia
| | - Daniel Kennedy
- Institute of Health and Biomedical Innovations, Genomics Research Centre, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia
| | - Kira Groen
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2308, Australia.,Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Katherine A Sanders
- Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.,Centre for Anatomical and Human Sciences, Hull York Medical School, Hull, UK
| | - Rebecca Seeto
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2308, Australia.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Samara Bray
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2308, Australia.,Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Melissa Gresle
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia.,Royal Melbourne Hospital, Melbourne, VIC, Australia.,MS and Neuroimmunology Unit, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Louise Laverick
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia.,Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Alfred Hospital, Melbourne, VIC, Australia.,MS and Neuroimmunology Unit, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, 2308, Australia.,Division of Molecular Genetics, Pathology North, John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia.,Centre for Cancer Research, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Jeannette Lechner-Scott
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2308, Australia. .,Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia. .,Department of Neurology, John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
17
|
Peñaloza E, Soto-Carrasco G, Krause BJ. MiR-21-5p directly contributes to regulating eNOS expression in human artery endothelial cells under normoxia and hypoxia. Biochem Pharmacol 2020; 182:114288. [PMID: 33075314 DOI: 10.1016/j.bcp.2020.114288] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022]
Abstract
Clinical conditions associated with hypoxia and oxidative stress, such as fetal growth restriction (FGR), results in endothelial dysfunction. Previous reports show that changes in eNOS expression under these conditions are tightly controlled by DNA methylation and histone posttranslational modifications. However, the contribution of an orchestrating epigenetic mechanism, such as miRNAs, on the NO-related genes expression has not been addressed. We aimed to determine the levels of miRNAs highly expressed in normal endothelial cells (EC), miR-21 and miR-126, in FGR human umbilical artery EC (HUAEC), and their effects on hypoxia-dependent regulation of both, NO-related and oxidative stress-related genes. Results were validated by transcriptome analysis of HUAEC cultured under chronic low oxygen conditions. Cultured FGR-HUAEC showed decreased hsa-miR-21, DDAH1, SOD1, and NRF2, but increased miR-126, NOX4, and eNOS levels, compared with controls. MiR-21-5p levels in FGR were associated with increased hg-miR-21 gene promoter methylation, with no changes in hg-miR-126 gene promoter methylation. HUAEC exposed to hypoxia showed a transient increase in eNOS and DDAH11, paralleled by decrease miR-21-5p levels, but no changes in miR-126-3p and the other genes under study. Transcriptome profiling showed an inverse relationship among miR-21 and several transcripts targeted by miR-21 in HUAEC exposed to hypoxia, meanwhile miR-21-5p-mimic decreased eNOS and DDAH1 transcripts stability, blocking their induction by hypoxia. Consequently, FGR programs a hypoxia-related miRNA that contributes to the regulation of the NO pathway, involving a direct effect of miR-21-5p on eNOS transcript stability, not previously reported. Moreover, hypoxia downregulates miR-21-5p, contributing to increasing the expression of NO-related genes in arterial endothelial cells.
Collapse
Affiliation(s)
- Estefania Peñaloza
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile
| | | | - Bernardo J Krause
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile.
| |
Collapse
|
18
|
Wu L, Xia J, Li D, Kang Y, Fang W, Huang P. Mechanisms of M2 Macrophage-Derived Exosomal Long Non-coding RNA PVT1 in Regulating Th17 Cell Response in Experimental Autoimmune Encephalomyelitisa. Front Immunol 2020; 11:1934. [PMID: 33013847 PMCID: PMC7500097 DOI: 10.3389/fimmu.2020.01934] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/17/2020] [Indexed: 11/13/2022] Open
Abstract
Long non-coding RNA (lncRNA) is pivotal for multiple sclerosis (MS), but the potential mechanism of lncRNA PVT1 in MS animal model, experimental autoimmune encephalomyelitis (EAE) still remains unclear. In this study, macrophages were firstly isolated and induced to polarize into M2 macrophages. M2 macrophage-derived exosomes (M2-exos) were extracted and identified, and EAE mouse model was established and treated with M2-exos. The effect of M2-exos on EAE mice was evaluated by clinical scores. The proportion of Treg and Th17 cells in spinal cord cells and splenocytes, and levels of inflammatory factors were measured. The targeting relationships among PVT1, miR-21-5p, and SOCS5 were verified. The expression of JAKs/STAT3 pathway-related proteins was measured. After M2-exo treatment, the clinical score of EAE mice decreased, and demyelination and inflammatory infiltration improved; Th17 cells decreased, Treg cells increased, and the levels of inflammatory factors decreased significantly. SOCS5 and PVT1 were downregulated and miR-21-5p was upregulated in EAE mice. PVT1 could sponge miR-21-5p to regulate SOCS5. SOCS5 alleviated EAE symptoms by repressing the JAKs/STAT3 pathway. Together, M2-exos-carried lncRNA PVT1 sponged miR-21-5p to upregulate SOCS5 and inactivate the JAKs/STAT3 pathway, thus reducing inflammation and protecting EAE mice. This study may offer novel treatments for MS.
Collapse
Affiliation(s)
- Lei Wu
- Department of Neurology, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, China
| | - Jinjin Xia
- Department of Neurology, Changxing Hospital, Second Affiliated Hospital of Medical College of Zhejiang University, Huzhou, China
| | - Donghui Li
- Department of Neurology, Changxing Hospital, Second Affiliated Hospital of Medical College of Zhejiang University, Huzhou, China
| | - Ying Kang
- Department of Pollution Source Statistics, Zhejiang Provincial Environmental Monitoring Center, Hangzhou, China
| | - Wei Fang
- School of Pharmacy, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Peng Huang
- School of Pharmacy, Anhui University of Traditional Chinese Medicine, Hefei, China
| |
Collapse
|
19
|
Abstract
Multiple sclerosis (MS) is an aggravating autoimmune disease that cripples young patients slowly with physical, sensory and cognitive deficits. The break of self-tolerance to neuronal antigens is the key to the pathogenesis of MS, with autoreactive T cells causing demyelination that subsequently leads to inflammation-mediated neurodegenerative events in the central nervous system. The exact etiology of MS remains elusive; however, the interplay of genetic and environmental factors contributes to disease development and progression. Given that genetic variation only accounts for a fraction of risk for MS, extrinsic risk factors including smoking, infection and lack of vitamin D or sunshine, which cause changes in gene expression, contribute to disease development through epigenetic regulation. To date, there is a growing body of scientific evidence to support the important roles of epigenetic processes in MS. In this chapter, the three main layers of epigenetic regulatory mechanisms, namely DNA methylation, histone modification and microRNA-mediated gene regulation, will be discussed, with a particular focus on the role of epigenetics on dysregulated immune responses and neurodegenerative events in MS. Also, the potential for epigenetic modifiers as biomarkers and therapeutics for MS will be reviewed.
Collapse
Affiliation(s)
- Vera Sau-Fong Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Queen Mary Hospital, Hong Kong SAR, China.
| |
Collapse
|
20
|
Li X, Gao Q, Yang L, Han M, Zhou C, Mu H. Matairesinol ameliorates experimental autoimmune uveitis by suppression of IRBP-specific Th17 cells. J Neuroimmunol 2020; 345:577286. [PMID: 32559555 DOI: 10.1016/j.jneuroim.2020.577286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
We investigated the effects of matairesinol (MAT) in the experimental autoimmune uveitis (EAU), a classical animal model of uveitis. We found that treatment with MAT could alleviate intraocular inflammation of EAU. Notably, Th17 cells in eyes of EAU mice could be predominantly restrained by MAT. Furthermore, MAT could inhibit Th17 differentiation in vitro. In addition, MAT inhibited the signaling of MAPK and ROR-γt, a pivotal transcription factor for Th17 cell differentiation in vitro and in vivo. Taken together, these results suggested that MAT had immune-suppressive effects on autoimmune inflammation through Th17 cells.
Collapse
Affiliation(s)
- Xin Li
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China
| | - Qiang Gao
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China
| | - Lei Yang
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China
| | - Meng Han
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China
| | - Chunlei Zhou
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China
| | - Hong Mu
- Department of Clinical Laboratory, Tianjin First Centre Hospital, Tianjin, China.
| |
Collapse
|
21
|
Jia Y, Wei Y. Modulators of MicroRNA Function in the Immune System. Int J Mol Sci 2020; 21:E2357. [PMID: 32235299 PMCID: PMC7177468 DOI: 10.3390/ijms21072357] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/23/2020] [Accepted: 03/27/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) play a key role in fine-tuning host immune homeostasis and responses through the negative regulation of mRNA stability and translation. The pathways regulated by miRNAs are well characterized, but the precise mechanisms that control the miRNA-mediated regulation of gene expression during immune cell-development and immune responses to invading pathogens are incompletely understood. Context-specific interactions of miRNAs with other RNA species or proteins may modulate the function of a given miRNA. Dysregulation of miRNA function is associated with various human diseases, such as cardiovascular diseases and cancers. Here, we review the potential modulators of miRNA function in the immune system, including the transcription regulators of miRNA genes, miRNA-processing enzymes, factors affecting miRNA targeting, and intercellular communication.
Collapse
Affiliation(s)
- Yunhui Jia
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yuanyuan Wei
- Department of Immunology, Shanghai Key laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
22
|
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: 1.3] [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
|
23
|
Celarain N, Tomas-Roig J. Aberrant DNA methylation profile exacerbates inflammation and neurodegeneration in multiple sclerosis patients. J Neuroinflammation 2020; 17:21. [PMID: 31937331 PMCID: PMC6961290 DOI: 10.1186/s12974-019-1667-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune and demyelinating disease of the central nervous system characterised by incoordination, sensory loss, weakness, changes in bladder capacity and bowel function, fatigue and cognitive impairment, creating a significant socioeconomic burden. The pathogenesis of MS involves both genetic susceptibility and exposure to distinct environmental risk factors. The gene x environment interaction is regulated by epigenetic mechanisms. Epigenetics refers to a complex system that modifies gene expression without altering the DNA sequence. The most studied epigenetic mechanism is DNA methylation. This epigenetic mark participates in distinct MS pathophysiological processes, including blood-brain barrier breakdown, inflammatory response, demyelination, remyelination failure and neurodegeneration. In this study, we also accurately summarised a list of environmental factors involved in the MS pathogenesis and its clinical course. A literature search was conducted using MEDLINE through PubMED and Scopus. In conclusion, an exhaustive study of DNA methylation might contribute towards new pharmacological interventions in MS by use of epigenetic drugs.
Collapse
Affiliation(s)
- Naiara Celarain
- Girona Neuroimmunology and Multiple Sclerosis Unit (UNIEM), Dr. Josep Trueta University Hospital and Girona Biomedical Research Institute (IDIBGI), Girona, Spain.
| | - Jordi Tomas-Roig
- Girona Neuroimmunology and Multiple Sclerosis Unit (UNIEM), Dr. Josep Trueta University Hospital and Girona Biomedical Research Institute (IDIBGI), Girona, Spain.
| |
Collapse
|
24
|
Berge T, Eriksson A, Brorson IS, Høgestøl EA, Berg-Hansen P, Døskeland A, Mjaavatten O, Bos SD, Harbo HF, Berven F. Quantitative proteomic analyses of CD4 + and CD8 + T cells reveal differentially expressed proteins in multiple sclerosis patients and healthy controls. Clin Proteomics 2019; 16:19. [PMID: 31080378 PMCID: PMC6505067 DOI: 10.1186/s12014-019-9241-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/27/2019] [Indexed: 02/07/2023] Open
Abstract
Background Multiple sclerosis (MS) is an autoimmune, neuroinflammatory disease, with an unclear etiology. However, T cells play a central role in the pathogenesis by crossing the blood–brain-barrier, leading to inflammation of the central nervous system and demyelination of the protective sheath surrounding the nerve fibers. MS has a complex inheritance pattern, and several studies indicate that gene interactions with environmental factors contribute to disease onset. Methods In the current study, we evaluated T cell dysregulation at the protein level using electrospray liquid chromatography–tandem mass spectrometry to get novel insights into immune-cell processes in MS. We have analyzed the proteomic profiles of CD4+ and CD8+ T cells purified from whole blood from 13 newly diagnosed, treatment-naive female patients with relapsing–remitting MS and 14 age- and sex-matched healthy controls. Results An overall higher protein abundance was observed in both CD4+ and CD8+ T cells from MS patients when compared to healthy controls. The differentially expressed proteins were enriched for T-cell specific activation pathways, especially CTLA4 and CD28 signaling in CD4+ T cells. When selectively analyzing proteins expressed from the genes most proximal to > 200 non-HLA MS susceptibility polymorphisms, we observed differential expression of eight proteins in T cells between MS patients and healthy controls, and there was a correlation between the genotype at three MS genetic risk loci and protein expressed from proximal genes. Conclusion Our study provides evidence for proteomic differences in T cells from relapsing–remitting MS patients compared to healthy controls and also identifies dysregulation of proteins encoded from MS susceptibility genes. Electronic supplementary material The online version of this article (10.1186/s12014-019-9241-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tone Berge
- Department of Mechanical, Electronics and Chemical Engineering, Faculty of Technology, Art and Design, Oslo Met - Oslo Metropolitan University, Postboks 4, St. Olavs Plass, 0130 Oslo, Norway.,2Neuroscience Research Unit, Oslo University Hospital, Rikshospitalet, Domus Medica 4, Nydalen, Postboks 4950, 0424 Oslo, Norway.,3Department of Research, Innovation and Education, Oslo University Hospital, Oslo, Norway
| | - Anna Eriksson
- 2Neuroscience Research Unit, Oslo University Hospital, Rikshospitalet, Domus Medica 4, Nydalen, Postboks 4950, 0424 Oslo, Norway.,4Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ina Skaara Brorson
- 2Neuroscience Research Unit, Oslo University Hospital, Rikshospitalet, Domus Medica 4, Nydalen, Postboks 4950, 0424 Oslo, Norway.,4Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,5Department of Neurology, Oslo University Hospital, Ullevål, Postboks 4950, 0424 Nydalen, Oslo, Norway
| | - Einar August Høgestøl
- 2Neuroscience Research Unit, Oslo University Hospital, Rikshospitalet, Domus Medica 4, Nydalen, Postboks 4950, 0424 Oslo, Norway.,4Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pål Berg-Hansen
- 4Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,5Department of Neurology, Oslo University Hospital, Ullevål, Postboks 4950, 0424 Nydalen, Oslo, Norway
| | - Anne Døskeland
- 6Proteomics Unit at University of Bergen (PROBE), Department of Biomedicine, University of Bergen, Postboks 7804, 5020 Bergen, Norway
| | - Olav Mjaavatten
- 6Proteomics Unit at University of Bergen (PROBE), Department of Biomedicine, University of Bergen, Postboks 7804, 5020 Bergen, Norway
| | - Steffan Daniel Bos
- 2Neuroscience Research Unit, Oslo University Hospital, Rikshospitalet, Domus Medica 4, Nydalen, Postboks 4950, 0424 Oslo, Norway.,4Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,5Department of Neurology, Oslo University Hospital, Ullevål, Postboks 4950, 0424 Nydalen, Oslo, Norway
| | - Hanne F Harbo
- 4Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,5Department of Neurology, Oslo University Hospital, Ullevål, Postboks 4950, 0424 Nydalen, Oslo, Norway
| | - Frode Berven
- 6Proteomics Unit at University of Bergen (PROBE), Department of Biomedicine, University of Bergen, Postboks 7804, 5020 Bergen, Norway
| |
Collapse
|
25
|
Ewing E, Kular L, Fernandes SJ, Karathanasis N, Lagani V, Ruhrmann S, Tsamardinos I, Tegner J, Piehl F, Gomez-Cabrero D, Jagodic M. Combining evidence from four immune cell types identifies DNA methylation patterns that implicate functionally distinct pathways during Multiple Sclerosis progression. EBioMedicine 2019; 43:411-423. [PMID: 31053557 PMCID: PMC6558224 DOI: 10.1016/j.ebiom.2019.04.042] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/15/2019] [Accepted: 04/23/2019] [Indexed: 12/22/2022] Open
Abstract
Background Multiple Sclerosis (MS) is a chronic inflammatory disease and a leading cause of progressive neurological disability among young adults. DNA methylation, which intersects genes and environment to control cellular functions on a molecular level, may provide insights into MS pathogenesis. Methods We measured DNA methylation in CD4+ T cells (n = 31), CD8+ T cells (n = 28), CD14+ monocytes (n = 35) and CD19+ B cells (n = 27) from relapsing-remitting (RRMS), secondary progressive (SPMS) patients and healthy controls (HC) using Infinium HumanMethylation450 arrays. Monocyte (n = 25) and whole blood (n = 275) cohorts were used for validations. Findings B cells from MS patients displayed most significant differentially methylated positions (DMPs), followed by monocytes, while only few DMPs were detected in T cells. We implemented a non-parametric combination framework (omicsNPC) to increase discovery power by combining evidence from all four cell types. Identified shared DMPs co-localized at MS risk loci and clustered into distinct groups. Functional exploration of changes discriminating RRMS and SPMS from HC implicated lymphocyte signaling, T cell activation and migration. SPMS-specific changes, on the other hand, implicated myeloid cell functions and metabolism. Interestingly, neuronal and neurodegenerative genes and pathways were also specifically enriched in the SPMS cluster. Interpretation We utilized a statistical framework (omicsNPC) that combines multiple layers of evidence to identify DNA methylation changes that provide new insights into MS pathogenesis in general, and disease progression, in particular. Fund This work was supported by the Swedish Research Council, Stockholm County Council, AstraZeneca, European Research Council, Karolinska Institutet and Margaretha af Ugglas Foundation.
Collapse
Affiliation(s)
- Ewoud Ewing
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | - Lara Kular
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | - Sunjay J Fernandes
- Unit of Computational Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, 17177, Sweden; Science for Life Laboratory, Solna, Sweden
| | - Nestoras Karathanasis
- Institute of Computer Science, Foundation for Research and Technology-Hellas, Heraklion, Greece; Computational Medicine Center, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Vincenzo Lagani
- Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia; Gnosis Data Analysis PC, Heraklion, Greece
| | - Sabrina Ruhrmann
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | - Ioannis Tsamardinos
- Gnosis Data Analysis PC, Heraklion, Greece; Department of Computer Science, University of Crete, Heraklion, Greece
| | - Jesper Tegner
- Unit of Computational Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, 17177, Sweden; Biological and Environmental Sciences and Engineering Division, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Saudi Arabia; Science for Life Laboratory, Solna, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17177, Sweden; Center for Neurology, Academic Specialist Clinic, Stockholm Health Services, Stockholm, Sweden
| | - David Gomez-Cabrero
- Unit of Computational Medicine, Department of Medicine, Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, 17177, Sweden; Translational Bioinformatics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; Centre for Host Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, UK
| | - Maja Jagodic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17177, Sweden.
| |
Collapse
|
26
|
Piket E, Zheleznyakova GY, Kular L, Jagodic M. Small non-coding RNAs as important players, biomarkers and therapeutic targets in multiple sclerosis: A comprehensive overview. J Autoimmun 2019; 101:17-25. [PMID: 31014917 DOI: 10.1016/j.jaut.2019.04.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 10/27/2022]
Abstract
Multiple sclerosis (MS) is a leading cause of progressive disability among young adults caused by inflammation, demyelination and axonal loss in the central nervous system. Small non-coding RNAs (sncRNAs) are important regulators of various biological processes and could therefore play important roles in MS. Over the past decade, a large number of studies investigated sncRNAs in MS patients, focusing primarily on microRNAs (miRNAs). Overwhelming 500 miRNAs have been reported as dysregulated in MS. Nevertheless, owing to a large heterogeneity between studies it is challenging to evaluate the reproducibility of findings, in turn hampering our knowledge about the functional roles of miRNAs in disease. We systematically searched main databases and evaluated results from all studies that examined sncRNAs in MS to date (n = 61) and provided a detailed overview of experimental design and findings of these studies. We focused on the mechanisms of the most dysregulated sncRNAs and used predicted targets of the most dysregulated sncRNAs as input for functional enrichment analysis to highlight affected pathways. The prime affected pathway was TGF-β signaling. This multifunctional cytokine is important in the differentiation and function of T helper type 17 (Th17) and regulatory T (Treg) cells, with opposing functions in the disease. Recent studies demonstrate the importance of miRNAs in controlling the balance between Th17/Th1 cells and Tregs and, importantly, the potential to exploit this paradigm for therapeutic purposes. Additionally, some of the discussed miRNAs could potentially serve as biomarkers of disease. In order to assist researchers in evaluating the evidence of a particular sncRNA in the pathogenesis of MS, we provide a detailed overview of experimental design and findings of these studies to date.
Collapse
Affiliation(s)
- Eliane Piket
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Lara Kular
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
27
|
Gharibi S, Moghimi B, Haghmorad D, Mahmoudi MB, Shahvazian E, Yadegari M, Yazd EF, Tahoori MT. Altered expression patterns of complement factor H and miR‐146a genes in acute‐chronic phases in experimental autoimmune encephalomyelitis mouse. J Cell Physiol 2019; 234:19842-19851. [DOI: 10.1002/jcp.28583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/10/2019] [Accepted: 03/19/2019] [Indexed: 01/10/2023]
Affiliation(s)
- Saba Gharibi
- Department of Genetics, Faculty of Medicine, International Campus Shahid Sadoughi University of Medical Sciences and Health Services Yazd Iran
| | - Bahram Moghimi
- Department of Genetics, Faculty of Medicine Shahid Sadoughi University of Medical Sciences and Health Services Yazd Iran
| | - Dariush Haghmorad
- Department of Pathology and Laboratory Medicine, School of Medicine Semnan University of Medical Sciences Semnan Iran
- Department of Immunology, School of Medicine Semnan University of Medical Sciences and Health Services Semnan Iran
| | - Mohammad Bagher Mahmoudi
- Department of Genetics, Faculty of Medicine Shahid Sadoughi University of Medical Sciences and Health Services Yazd Iran
| | - Ensieh Shahvazian
- Department of Genetics, Faculty of Medicine, International Campus Shahid Sadoughi University of Medical Sciences and Health Services Yazd Iran
| | - Maryam Yadegari
- Department of Biology & Anatomical Sciences, Shahid Sadoughi University of Medical Sciences and Health Services Faculty of Medicine Yazd Iran
| | - Ehsan Farashahi Yazd
- Department of Genetics, Faculty of Medicine Shahid Sadoughi University of Medical Sciences and Health Services Yazd Iran
- Genetic Engineering and Genome Editing Laboratory, Stem Cell Biology Research Center Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences Yazd Iran
| | - Mohammad Taher Tahoori
- Department of Immunology, Faculty of Medicine Shahid Sadoughi University of Medical Sciences and Health Services Yazd Iran
| |
Collapse
|
28
|
Ntranos A, Ntranos V, Bonnefil V, Liu J, Kim-Schulze S, He Y, Zhu Y, Brandstadter R, Watson CT, Sharp AJ, Katz Sand I, Casaccia P. Fumarates target the metabolic-epigenetic interplay of brain-homing T cells in multiple sclerosis. Brain 2019; 142:647-661. [PMID: 30698680 PMCID: PMC6821213 DOI: 10.1093/brain/awy344] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023] Open
Abstract
Cell-permeable formulations of metabolites, such as fumaric acid esters, have been used as highly effective immunomodulators in patients with multiple sclerosis and yet their mechanism of action remains elusive. Since fumaric acid esters are metabolites, and cell metabolism is highly intertwined with the epigenetic regulation of gene expression, we investigated whether this metabolic-epigenetic interplay could be leveraged for therapeutic purposes. To this end we recruited 47 treatment-naïve and 35 fumaric acid ester-treated patients with multiple sclerosis, as well as 16 glatiramer acetate-treated patients as a non-metabolite treatment control. Here we identify a significant immunomodulatory effect of fumaric acid esters on the expression of the brain-homing chemokine receptor CCR6 in CD4 and CD8 T cells of patients with multiple sclerosis, which include T helper-17 and T cytotoxic-17 cells. We report differences in DNA methylation of CD4 T cells isolated from untreated and treated patients with multiple sclerosis, using the Illumina EPIC 850K BeadChip. We first demonstrate that Krebs cycle intermediates, such as fumaric acid esters, have a significantly higher impact on epigenome-wide DNA methylation changes in CD4 T cells compared to amino-acid polymers such as glatiramer acetate. We then define a fumaric acid ester treatment-specific hypermethylation effect on microRNA MIR-21, which is critical for the differentiation of T helper-17 cells. This hypermethylation effect was attributed to the subpopulation of T helper-17 cells using a decomposition analysis and was further validated in an independent prospective cohort of seven patients before and after treatment with fumaric acid esters. In vitro treatment of CD4 and CD8 T cells with fumaric acid esters supported a direct and dose-dependent effect on DNA methylation at the MIR-21 promoter. Finally, the upregulation of miR-21 transcripts and CCR6 expression was inhibited if CD4 or CD8 T cells stimulated under T helper-17 or T cytotoxic-17 polarizing conditions were treated with fumaric acid esters in vitro. These data collectively define a direct link between fumaric acid ester treatment and hypermethylation of the MIR-21 locus in both CD4 and CD8 T cells and suggest that the immunomodulatory effect of fumaric acid esters in multiple sclerosis is at least in part due to the epigenetic regulation of the brain-homing CCR6+ CD4 and CD8 T cells.
Collapse
Affiliation(s)
- Achilles Ntranos
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Valentina Bonnefil
- Neuroscience, Advanced Science Research Center at The Graduate Center of the City University of New York, New York, NY, USA
| | - Jia Liu
- Neuroscience, Advanced Science Research Center at The Graduate Center of the City University of New York, New York, NY, USA
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ye He
- Neuroscience, Advanced Science Research Center at The Graduate Center of the City University of New York, New York, NY, USA
| | - Yunjiao Zhu
- Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Brandstadter
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Corey T Watson
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Andrew J Sharp
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ilana Katz Sand
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrizia Casaccia
- Neuroscience, Advanced Science Research Center at The Graduate Center of the City University of New York, New York, NY, USA
- Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
29
|
Ma Y, De Jager PL. Designing an epigenomic study. Mult Scler 2018; 24:604-609. [PMID: 29692225 DOI: 10.1177/1352458517750770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Yiyi Ma
- Center for Translational and Computational Neuro-immunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA/Cell Circuits Program, Broad Institute, Cambridge, MA, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuro-immunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA/Cell Circuits Program, Broad Institute, Cambridge, MA, USA
| |
Collapse
|
30
|
Zheleznyakova GY, Piket E, Marabita F, Pahlevan Kakhki M, Ewing E, Ruhrmann S, Needhamsen M, Jagodic M, Kular L. Epigenetic research in multiple sclerosis: progress, challenges, and opportunities. Physiol Genomics 2017; 49:447-461. [PMID: 28754822 DOI: 10.1152/physiolgenomics.00060.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/24/2017] [Indexed: 01/02/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and demyelinating disease of the central nervous system. MS likely results from a complex interplay between predisposing causal gene variants (the strongest influence coming from HLA class II locus) and environmental risk factors such as smoking, infectious mononucleosis, and lack of sun exposure/vitamin D. However, little is known about the mechanisms underlying MS development and progression. Moreover, the clinical heterogeneity and variable response to treatment represent additional challenges to a comprehensive understanding and efficient treatment of disease. Epigenetic processes, such as DNA methylation and histone posttranslational modifications, integrate influences from the genes and the environment to regulate gene expression accordingly. Studying epigenetic modifications, which are stable and reversible, may provide an alternative approach to better understand and manage disease. We here aim to review findings from epigenetic studies in MS and further discuss the challenges and clinical opportunities arising from epigenetic research, many of which apply to other diseases with similar complex etiology. A growing body of evidence supports a role of epigenetic processes in the mechanisms underlying immune pathogenesis and nervous system dysfunction in MS. However, disparities between studies shed light on the need to consider possible confounders and methodological limitations for a better interpretation of the data. Nevertheless, translational use of epigenetics might offer new opportunities in epigenetic-based diagnostics and therapeutic tools for a personalized care of MS patients.
Collapse
Affiliation(s)
- Galina Y Zheleznyakova
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eliane Piket
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Francesco Marabita
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Majid Pahlevan Kakhki
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ewoud Ewing
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sabrina Ruhrmann
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Needhamsen
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lara Kular
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|