1
|
He J, Meng Q, Miao C, Hao J, Dai M. Unravelling the neuroimmune nexus: insights into epilepsy pathology and the role of S100b protein in brain-gut axis modulation: a literature review. Postgrad Med J 2025; 101:181-188. [PMID: 39400536 DOI: 10.1093/postmj/qgae125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/30/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024]
Abstract
Epilepsy, a chronic neurological condition marked by recurrent, unprovoked seizures, involves complex pathophysiological mechanisms. Recent advancements have expanded our understanding from traditional neuronal dysfunction to include neuroimmune interactions and the influence of the brain-gut-bio-axis. This review explores the role of the S100b protein within these contexts, noted for its involvement in neuroinflammatory processes and as a potential biomarker. Furthermore, it discusses the emerging significance of the gut microbiome in modulating neuroimmune responses and seizure activity. The review integrates findings from recent studies, emphasizing the critical role of the S100b signalling pathway and the gut-brain axis in epilepsy pathology. The interplay between neuroimmune mechanisms and gut microbiota offers novel insights and potential therapeutic targets, underlining the need for further research to exploit these connections for clinical benefit.
Collapse
Affiliation(s)
- Jianxun He
- Department of Neurosurgery, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, Gansu 730050, China
| | - Qianling Meng
- Department of Rehabilitation Medicine, Shanghai Xuhui Central Hospital, Shanghai 200030, China
| | - Chuhan Miao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam 999077, Hong Kong SAR of China
| | - Jing Hao
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Woodhouse, Leeds LS2 9JT, United Kingdom
| | - Mengliang Dai
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| |
Collapse
|
2
|
Qiao Y, Wang X, Ma Y, Hu J. Variations in vitamin D status among Chinese children aged 1-6 years during the COVID-19 pandemic. Front Public Health 2025; 13:1514355. [PMID: 39916704 PMCID: PMC11799289 DOI: 10.3389/fpubh.2025.1514355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
Background Vitamin D deficiency has been a critical global health issue within the pediatric population. Closed-off management brought about by the COVID-19 pandemic has drastically impacted outdoor activities and sunlight exposure, however, whether it indirectly further exacerbated the vitamin D deficiency has not been largely investigated, especially among children in China. The purpose of this study was to evaluate 25(OH)D concentrations in children before and during the COVID-19 lockdown and to analyze the factors influencing their vitamin D status. Methods A cross-sectional survey included children aged 1-6 years from Han Zhong Central Hospital in the southern Shanxi Province of China. This study examined healthy children from a pediatric health care department over two periods: before COVID-19 (March 2019-February 2020), and during COVID-19 (March 2020-February 2021). Total 25(OH)D concentrations were compared between the two observation periods. Vitamin D status was determined by 25(OH)D concentrations: deficient (<20 ng/ml), insufficient (20-29 ng/ml), and sufficient (30-100 ng/ml). Results The study involved 6,780 children, with 52.8% being 1-year-olds, 23.1% being 2-year-olds, and 24.1% being 3 to 6-year-olds. Boys and girls were 52.8 and 47.2%, respectively. The actual prevalence of deficiency in vitamin D nutritional status among children was 2.8%, with 87.1% of cases in those aged 3 to 6 years. Vitamin D insufficiency was 18.3%, affecting 54.8% of the same demographic. The average of 25(OH)D concentration were 38.2 ± 9.8 ng/ml, significantly varying by age and season. 25(OH)D concentrations decreased with age, from 42.3 ± 8.8 ng/ml at 1-year-olds to 37.4 ± 8.2 ng/ml at 2-year-olds, and further to 30.2 ± 8.1 ng/ml at 3 to 6-year-olds. Seasonal variations showed that 25(OH)D concentrations were higher in spring (38.7 ± 10.1 ng/ml), summer (38.7 ± 10.0 ng/ml), and fall (38.6 ± 9.2 ng/ml) in comparison to winter (36.0 ± 9.8 ng/ml). Additionally, the concentrations of 25(OH)D in spring exhibited a decrease during the COVID-19 pandemic (37.9 ± 10.3 ng/ml) in comparison to the pre-pandemic measurements (39.3 ± 9.9 ng/ml) (p = 0.008), while winter concentrations increased from (35.1 ± 10.4 ng/ml) to (37.9 ± 10.3 ng/ml) during the pandemic (p = 0.002). Conclusion The research indicated that vitamin D deficiency is uncommon among Chinese children, with 25(OH)D concentrations experiencing a notable decline in those aged 3-6 years. The findings suggested a potential need for tailored supplementation strategies and possibly higher doses for this age group, along with monitoring 25(OH)D concentrations to evaluate supplementation effectiveness. COVID-19-related restrictions minimally affected children's 25(OH)D concentrations, revealing the nutritional implications of the pandemic.
Collapse
Affiliation(s)
- Yongfeng Qiao
- Department of Clinical Laboratory, Hanzhong Central Hospital, Hanzhong, China
| | - Xiaoqin Wang
- Department of Clinical Laboratory, The First affiliated hospital of Xi’an Jiaotong University, Xi'an, China
| | - Yanfen Ma
- Department of Clinical Laboratory, The First affiliated hospital of Xi’an Jiaotong University, Xi'an, China
| | - Jian Hu
- Department of Clinical Laboratory, The First affiliated hospital of Xi’an Jiaotong University, Xi'an, China
| |
Collapse
|
3
|
Yüksel D, Yardimci F. Development and psychometric evaluation of the treatment management adherence scale for children with multiple sclerosis. Mult Scler Relat Disord 2024; 92:106162. [PMID: 39541822 DOI: 10.1016/j.msard.2024.106162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/15/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Pediatric multiple sclerosis (pMS) is a chronic inflammatory, demyelinating, and neurodegenerative disease affecting the central nervous system in children and adolescents The aim of this correlational, comparative study was to develop an assessment scale for adherence to treatment management in pMS. METHODS Two measurement tools were used to develop a scientifically sound tool to assess adherence in pediatric patients (12-18 years) diagnosed with multiple sclerosis (MS). Cases of pMS (n = 120) in 7 hospitals in Turkey were included between August 2021-February 2022. The tools were a "Sociodemographic and Disease-Related Information" and a newly developed "Treatment Management Adherence Scale for Children with Multiple Sclerosis". The form and questionnaire were completed by the children through online using the Zoom platform in approximately 10 min. The questionnaire on adherence contains 16 items related to the disease and treatment, scored in a 5-point Likert type. Face validity was established by pretesting with 20 children, and construct validity was established using the statistical methods of exploratory factor analysis and confirmatory factor analysis. For the reliability of the scale, Cronbach's Alpha and omega coefficients, item test correlation values, split-half, test-retest techniques were used. RESULTS There were 120 eligible patients, 71.2 % girls, with mean age (±SD) 13,6 ± 2,2 years at disease onset and 15,7 ± 1,5 at the time of the study, all under disease-modifying therapy. The sample size and items were sufficient to conduct a factor analysis. The Cronbach's Alpha and Omega value was 0.75, indicating participants' opinions were consistent across items. The mean content validity index was 0.93, showing the scale represented the measured data, and the exploratory factor analysis showed the scale measures adherence in 55 % of patients (desired figures: >0.80 and 40-60 % respectively). The 16 items of the questionnaire were grouped into 4 dimensions. These dimensions were termed 'physiological', 'self-concept', 'role function' and 'interdependence', in line with different styles of adaptation. The total score can be between 16 and 80, with higher scores indicating strong adherence to treatment. The mean total score of 54,3 ± 9,53 (min=31, max= 75) in this study was in the "moderate adherence" range. CONCLUSIONS This new scale is the first to assess adherence in pMS. The study supports its validity, reliability, and likelihood to address adjustment issues in children and adolescents with MS accurately and can be recommended for clinical use.
Collapse
Affiliation(s)
- Didem Yüksel
- Assistant Professor, Atılım University, Faculty of Health Sciences, Department of Nursing, Child Health and Diseases Nursing, Ankara, Turkey.
| | - Figen Yardimci
- Associate Professor, Ege University, Faculty of Nursing, Department of Child Health and Diseases Nursing, Izmir, Turkey
| |
Collapse
|
4
|
Yeh WZ, Gresle M, Lea R, Taylor B, Lucas RM, Ponsonby AL, Mason D, Andrew J, Campbell H, Morahan J, Sampangi S, Campagna MP, Stankovich J, Van der Walt A, Jokubaitis V, Butzkueven H. The immune cell transcriptome is modulated by vitamin D 3 supplementation in people with a first demyelinating event participating in a randomized placebo-controlled trial. Clin Immunol 2024; 262:110183. [PMID: 38479439 DOI: 10.1016/j.clim.2024.110183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/18/2024]
Abstract
Vitamin D deficiency is a risk factor for developing multiple sclerosis. The PrevANZ trial was conducted to determine if vitamin D3 supplementation can prevent recurrent disease activity in people with a first demyelinating event. As a sub-study of this trial, we investigated the effect of supplementation on peripheral immune cell gene expression. Participants were randomized to 1000, 5000 or 10,000 international units daily of vitamin D3 or placebo. Peripheral blood was collected at baseline and 12 weeks and sent for ribonucleic acid sequencing. Datasets from 55 participants were included. Gene expression was modulated by high dose supplementation. Antigen presentation and viral response pathways were upregulated. Oxidative phosphorylation and immune signaling pathways, including tumor necrosis factor-alpha and interleukin-17 signaling, were downregulated. Overall, vitamin D3 supplementation for 12 weeks modulated the peripheral immune cell transcriptome with induction of anti-inflammatory gene expression profiles. Our results support a dose-dependent effect of vitamin D3 supplementation on immune gene expression.
Collapse
Affiliation(s)
- Wei Zhen Yeh
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| | - Melissa Gresle
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Rodney Lea
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia; Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Bruce Taylor
- Royal Hobart Hospital, Department of Neurology, Hobart, Australia; University of Tasmania, Menzies Institute for Medical Research, Hobart, Australia
| | - Robyn M Lucas
- Australian National University, National Centre for Epidemiology and Population Health, Canberra, Australia
| | - Anne-Louise Ponsonby
- The Florey Institute of Neuroscience and Mental Health, Early Brain Division, Parkville, Australia; University of Melbourne, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Deborah Mason
- Christchurch Hospital, Christchurch, New Zealand; New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Julie Andrew
- Neuroscience Trials Australia, Heidelberg, Australia
| | | | | | - Sandeep Sampangi
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Maria Pia Campagna
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Jim Stankovich
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Anneke Van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia; MSBase Foundation, Melbourne, Australia.
| |
Collapse
|
5
|
Butzkueven H, Ponsonby AL, Stein MS, Lucas RM, Mason D, Broadley S, Kilpatrick T, Lechner-Scott J, Barnett M, Carroll W, Mitchell P, Hardy TA, Macdonell R, McCombe P, Lee A, Kalincik T, van der Walt A, Lynch C, Abernethy D, Willoughby E, Barkhof F, MacManus D, Clarke M, Andrew J, Morahan J, Zhu C, Dear K, Taylor BV. Vitamin D did not reduce multiple sclerosis disease activity after a clinically isolated syndrome. Brain 2024; 147:1206-1215. [PMID: 38085047 PMCID: PMC10994527 DOI: 10.1093/brain/awad409] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/16/2023] [Accepted: 11/03/2023] [Indexed: 04/06/2024] Open
Abstract
Low serum levels of 25-hydroxyvitamin D [25(OH)D] and low sunlight exposure are known risk factors for the development of multiple sclerosis. Add-on vitamin D supplementation trials in established multiple sclerosis have been inconclusive. The effects of vitamin D supplementation to prevent multiple sclerosis is unknown. We aimed to test the hypothesis that oral vitamin D3 supplementation in high-risk clinically isolated syndrome (abnormal MRI, at least three T2 brain and/or spinal cord lesions), delays time to conversion to definite multiple sclerosis, that the therapeutic effect is dose-dependent, and that all doses are safe and well tolerated. We conducted a double-blind trial in Australia and New Zealand. Eligible participants were randomized 1:1:1:1 to placebo, 1000, 5000 or 10 000 international units (IU) of oral vitamin D3 daily within each study centre (n = 23) and followed for up to 48 weeks. Between 2013 and 2021, we enrolled 204 participants. Brain MRI scans were performed at baseline, 24 and 48 weeks. The main study outcome was conversion to clinically definite multiple sclerosis based on the 2010 McDonald criteria defined as either a clinical relapse or new brain MRI T2 lesion development. We included 199 cases in the intention-to-treat analysis based on assigned dose. Of these, 116 converted to multiple sclerosis by 48 weeks (58%). Compared to placebo, the hazard ratios (95% confidence interval) for conversion were 1000 IU 0.87 (0.50, 1.50); 5000 IU 1.37 (0.82, 2.29); and 10 000 IU 1.28 (0.76, 2.14). In an adjusted model including age, sex, latitude, study centre and baseline symptom number, clinically isolated syndrome onset site, presence of infratentorial lesions and use of steroids, the hazard ratios (versus placebo) were 1000 IU 0.80 (0.45, 1.44); 5000 IU 1.36 (0.78, 2.38); and 10 000 IU 1.07 (0.62, 1.85). Vitamin D3 supplementation was safe and well tolerated. We did not demonstrate reduction in multiple sclerosis disease activity by vitamin D3 supplementation after a high-risk clinically isolated syndrome.
Collapse
Affiliation(s)
- Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Anne-Louise Ponsonby
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mark S Stein
- Department of Diabetes and Endocrinology, The Royal Melbourne Hospital, Parkville, VIC 3010, Australia
| | - Robyn M Lucas
- National Centre for Epidemiology and Public Health, Australian National University, Canberra, ACT 0200, Australia
| | - Deborah Mason
- Department of Neurology, Christchurch Hospital, Christchurch 8011, New Zealand
| | - Simon Broadley
- Department of Neurology, School of Medicine and Dentistry, Griffith University, Southport, QLD 4222, Australia
| | - Trevor Kilpatrick
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Michael Barnett
- Brain and Mind Research Institute University of Sydney, Sydney, NSW 2050, Australia
| | - William Carroll
- Department of Neurology, Sir Charles Gairdner Hospital and Centre for Neuromuscular and Neurological Disorders and Perron Institute, University of Western Australia, WA 6009, Australia
| | - Peter Mitchell
- Department of Radiology, Royal Melbourne Hospital, Melbourne, VIC 3010, Australia
| | - Todd A Hardy
- Department of Neurology, Concord Hospital, University of Sydney, Sydney, NSW 2139, Australia
| | - Richard Macdonell
- Department of Neurology, Austin Health, Melbourne, VIC 3084, Australia
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010Australia
| | - Pamela McCombe
- University of Queensland, Centre for Clinical Research, Brisbane, QLD 4029, Australia
| | - Andrew Lee
- Department of Neurology, Flinders University College of Medicine and Public Health, Adelaide, SA 5042, Australia
| | - Tomas Kalincik
- Neuroimmunology Centre, Department of Neurology, Royal Melbourne Hospital, Melbourne, VIC 3010, Australia
- CORe, Department of Medicine, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Anneke van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Chris Lynch
- Midland Neurology, Hamilton, Waikato 3240, New Zealand
| | - David Abernethy
- Department of Neurology, Wellington Hospital, Wellington 6021, New Zealand
| | - Ernest Willoughby
- Department of Neurology, Auckland Hospital, Auckland 1023, New Zealand
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Amsterdam 1081 HV, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, WC1N 3BG, UK
| | - David MacManus
- University College London Queen Square Institute of Neurology, Queen Square MS Centre, London WC1N 3BG, UK
| | - Michael Clarke
- Metabolomics Australia (WA), School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Julie Andrew
- Neurosciences Trials Australia, North Melbourne, VIC 3051, Australia
| | - Julia Morahan
- Multiple Sclerosis Australia, North Sydney, NSW 2059, Australia
| | - Chao Zhu
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Keith Dear
- Department of Statistics, School of Public Health, University of Adelaide, SA 5005, Australia
| | - Bruce V Taylor
- MS Research Flagship, Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| |
Collapse
|
6
|
Wagner CL, Hollis BW. Vitamin D as a modifier of genomic function and phenotypic expression during pregnancy. FELDMAN AND PIKE' S VITAMIN D 2024:669-692. [DOI: 10.1016/b978-0-323-91386-7.00045-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
Balshi A, Saart E, Dempsey J, Baber U, Sloane JA. Bariatric surgery outcomes in multiple sclerosis: Interplay with vitamin D and chronic pain syndromes. Mult Scler Relat Disord 2023; 79:105006. [PMID: 37734186 DOI: 10.1016/j.msard.2023.105006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/25/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND Obesity and lower vitamin D levels are associated with adverse outcomes in multiple sclerosis (MS). Bariatric surgery is a safe intervention in patients with MS, although it lowers vitamin D levels in the general population. OBJECTIVE To investigate the effects of bariatric surgery on vitamin D levels and interrogate risk factors for unsuccessful post-operative weight loss in patients with MS. METHODS We retrospectively identified patients with MS who underwent bariatric surgery from 2001 to 2023. Wilcoxon signed rank tests for paired samples were used to compare pre- and post-operative body mass index (BMI), expanded disability status scale (EDSS), timed 25-foot walk (T25FW), and median vitamin D values. RESULTS Following bariatric surgery, patients with MS had a decrease in BMI (mean percent total weight loss of 18.4 %, range 0-38 %, p < 0.001) and an increase in vitamin D values (mean increase of 23 ng/mL, range -4-32 ng/mL, p < 0.001), while no change in EDSS or T25FW was seen. Four out of 20 patients did not lose more than 5 % of their pre-operative BMI, all of whom had chronic pain syndromes and were on gabapentin. CONCLUSION Healthy vitamin D levels are attainable following bariatric surgery in patients with MS.
Collapse
Affiliation(s)
- Alexandra Balshi
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Emma Saart
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - John Dempsey
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ursela Baber
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jacob A Sloane
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Gill AJ, Schorr EM, Gadani SP, Calabresi PA. Emerging imaging and liquid biomarkers in multiple sclerosis. Eur J Immunol 2023; 53:e2250228. [PMID: 37194443 PMCID: PMC10524168 DOI: 10.1002/eji.202250228] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/10/2023] [Accepted: 05/12/2023] [Indexed: 05/18/2023]
Abstract
The advent of highly effective disease modifying therapy has transformed the landscape of multiple sclerosis (MS) care over the last two decades. However, there remains a critical, unmet need for sensitive and specific biomarkers to aid in diagnosis, prognosis, treatment monitoring, and the development of new interventions, particularly for people with progressive disease. This review evaluates the current data for several emerging imaging and liquid biomarkers in people with MS. MRI findings such as the central vein sign and paramagnetic rim lesions may improve MS diagnostic accuracy and evaluation of therapy efficacy in progressive disease. Serum and cerebrospinal fluid levels of several neuroglial proteins, such as neurofilament light chain and glial fibrillary acidic protein, show potential to be sensitive biomarkers of pathologic processes such as neuro-axonal injury or glial-inflammation. Additional promising biomarkers, including optical coherence tomography, cytokines and chemokines, microRNAs, and extracellular vesicles/exosomes, are also reviewed, among others. Beyond their potential integration into MS clinical care and interventional trials, several of these biomarkers may be informative of MS pathogenesis and help elucidate novel targets for treatment strategies.
Collapse
Affiliation(s)
- Alexander J. Gill
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, US
| | - Emily M. Schorr
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, US
| | - Sachin P. Gadani
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, US
| | - Peter A. Calabresi
- Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, US
- Department of Neuroscience, Baltimore, MD, US
- Department of Ophthalmology, Baltimore, MD, US
| |
Collapse
|
9
|
Hedström AK. Risk factors for multiple sclerosis in the context of Epstein-Barr virus infection. Front Immunol 2023; 14:1212676. [PMID: 37554326 PMCID: PMC10406387 DOI: 10.3389/fimmu.2023.1212676] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/26/2023] [Indexed: 08/10/2023] Open
Abstract
Compelling evidence indicates that Epstein Barr virus (EBV) infection is a prerequisite for multiple sclerosis (MS). The disease may arise from a complex interplay between latent EBV infection, genetic predisposition, and various environmental and lifestyle factors that negatively affect immune control of the infection. Evidence of gene-environment interactions and epigenetic modifications triggered by environmental factors in genetically susceptible individuals supports this view. This review gives a short introduction to EBV and host immunity and discusses evidence indicating EBV as a prerequisite for MS. The role of genetic and environmental risk factors, and their interactions, in MS pathogenesis is reviewed and put in the context of EBV infection. Finally, possible preventive measures are discussed based on the findings presented.
Collapse
Affiliation(s)
- Anna Karin Hedström
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
10
|
Dinov D, Brenton JN. Environmental Influences on Risk and Disease Course in Pediatric Multiple Sclerosis. Semin Pediatr Neurol 2023; 46:101049. [PMID: 37451747 PMCID: PMC10351032 DOI: 10.1016/j.spen.2023.101049] [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: 02/15/2023] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 07/18/2023]
Abstract
Pediatric multiple sclerosis (MS) accounts for 3%-10% of all patients diagnosed with MS. Complex interplay between environmental factors impacts the risk for MS and may also affect disease course. Many of these environmental factors are shared with adult-onset MS. However, children with MS are in closer temporal proximity to the biological onset of MS and have less confounding environmental exposures than their adult counterparts. Environmental factors that contribute to MS risk include: geographical latitude, viral exposures, obesity, vitamin deficiencies, smoking, air pollution, perinatal factors, gut microbiome, and diet. More recently, research efforts have shifted to studying the impact of these risk determinants on the clinical course of MS. In this article we will examine relevant environmental risk determinants of pediatric MS and review the current knowledge on how these factors may contribute to pediatric MS disease evolution.
Collapse
Affiliation(s)
- Darina Dinov
- Department of Neurology, Virginia Commonwealth University, Richmond, VA
| | - James Nicholas Brenton
- Division of Child Neurology, Department of Neurology, University of Virginia, Charlottesville, VA.
| |
Collapse
|
11
|
Sangha A, Quon M, Pfeffer G, Orton SM. The Role of Vitamin D in Neuroprotection in Multiple Sclerosis: An Update. Nutrients 2023; 15:2978. [PMID: 37447304 DOI: 10.3390/nu15132978] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Multiple sclerosis (MS) is a complex neurological condition that involves both inflammatory demyelinating and neurodegenerative components. MS research and treatments have traditionally focused on immunomodulation, with less investigation of neuroprotection, and this holds true for the role of vitamin D in MS. Researchers have already established that vitamin D plays an anti-inflammatory role in modulating the immune system in MS. More recently, researchers have begun investigating the potential neuroprotective role of vitamin D in MS. The active form of vitamin D, 1,25(OH)2D3, has a range of neuroprotective properties, which may be important in remyelination and/or the prevention of demyelination. The most notable finding relevant to MS is that 1,25(OH)2D3 promotes stem cell proliferation and drives the differentiation of neural stem cells into oligodendrocytes, which carry out remyelination. In addition, 1,25(OH)2D3 counteracts neurodegeneration and oxidative stress by suppressing the activation of reactive astrocytes and M1 microglia. 1,25(OH)2D3 also promotes the expression of various neuroprotective factors, including neurotrophins and antioxidant enzymes. 1,25(OH)2D3 decreases blood-brain barrier permeability, reducing leukocyte recruitment into the central nervous system. These neuroprotective effects, stimulated by 1,25(OH)2D3, all enhance neuronal survival. This review summarizes and connects the current evidence supporting the vitamin D-mediated mechanisms of action for neuroprotection in MS.
Collapse
Affiliation(s)
- Amarpreet Sangha
- Faculty of Science and Technology, Mount Royal University, Calgary, AB T3E 6K6, Canada
| | - Michaela Quon
- Faculty of Science and Technology, Mount Royal University, Calgary, AB T3E 6K6, Canada
| | - Gerald Pfeffer
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Child Health Research Institute, Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Sarah-Michelle Orton
- Faculty of Science and Technology, Mount Royal University, Calgary, AB T3E 6K6, Canada
| |
Collapse
|
12
|
Ghareghani M, Zibara K, Rivest S. Melatonin and vitamin D, two sides of the same coin, better to land on its edge to improve multiple sclerosis. Proc Natl Acad Sci U S A 2023; 120:e2219334120. [PMID: 36972442 PMCID: PMC10083587 DOI: 10.1073/pnas.2219334120] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Previous studies revealed a latitudinal gradient of multiple sclerosis (MS) prevalence, increasing by moving from the equator to the poles. The duration and quality of an individual's exposure to sunlight vary with latitude. Skin exposure to sunlight activates vitamin D synthesis, while light absence, as perceived by the eyes, activates melatonin synthesis in the pineal gland. Vitamin D or melatonin deficiency/insufficiency or overdose can occur at any latitude due to specific lifestyles and diets. Moving away from the equator, especially beyond 37°, decreases vitamin D while raising melatonin. Furthermore, melatonin synthesis increases in cold habitats like northern countries. Since melatonin's beneficial role was shown in MS, it is expected that northern countries whose individuals have higher endogenous melatonin should show a lower MS prevalence; however, these are ranked with the highest scores. In addition, countries like the United States and Canada have uncontrolled over-the-counter usage. In high latitudes, vitamin D deficiency and a higher MS prevalence persist even though vitamin D is typically compensated for by supplementation and not sunlight. Recently, we found that prolonged darkness increased MS melatonin levels, mimicking the long-term increase in northern countries. This caused a reduction in cortisol and increased infiltration, inflammation, and demyelination, which were all rescued by constant light therapy. In this review, we explain melatonin and vitamin D's possible roles in MS prevalence. The possible causes in northern countries are then discussed. Finally, we suggest strategies to treat MS by manipulating vitamin D and melatonin, preferably with sunlight or darkness, not supplements.
Collapse
Affiliation(s)
- Majid Ghareghani
- Neuroscience Laboratory, Centre Hospitalier Universitaire de Québec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QCG1V 4G2, Canada
| | - Kazem Zibara
- Platform for Research and Analysis in Environmental Science (PRASE) and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut1003, Lebanon
| | - Serge Rivest
- Neuroscience Laboratory, Centre Hospitalier Universitaire de Québec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QCG1V 4G2, Canada
| |
Collapse
|
13
|
Porto CM, Leão RDCH, Sousa RAD, Diniz PRB, Silva TDPSD, Sougey EB. Brain changes in neuroimaging of adult patients with vitamin D deficiency: systematic review protocol. BMJ Open 2023; 13:e052524. [PMID: 36849215 PMCID: PMC9972426 DOI: 10.1136/bmjopen-2021-052524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
INTRODUCTION Brain abnormalities detected through neuroimaging are described in patients with vitamin D deficiency, however, it is still not clear which cerebral alterations are more frequent and characteristic in this population. Thus, this review aims to identify and classify which are the main and most frequent brain changes found by neuroimaging in patients with vitamin D deficiency. METHODS AND ANALYSIS The study protocol was constructed in accordance with Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols and the leading research question was formulated through Population, Intervention, Comparator, Outcome, Setting. The evidence will be researched at the following electronic databases: PubMed, PsycINFO, Scopus, Web of Science and EMBASE. Two researchers will work in the selection, analysis and inclusion phases of the articles. In the case of divergence, a third-party reviewer will be contacted. The following studies will be included: (1) cohort studies, case-control studies and cross-sectional studies; (2) studies carried out on patients with serum 25-hydroxyvitamin D levels below 30 ng/mL; (3) studies conducted with an adult population; (4) studies using neuroimaging methods. Articles considered eligible will be analysed by the Newcastle-Ottawa Quality Assessment Scale/cross-section studies to evaluate study quality. The survey will be conducted from June to December 2022. ETHICS AND DISSEMINATION The identification of the main and most frequent brain alterations found through neuroimaging in patients with vitamin D deficiency can guide professionals as to the identification which of the main cerebral pathologies detected through neuroimaging are related to vitamin D deficiency, in choosing more sensitive and specific neuroimaging tests to detect these brain changes, in addition to emphasising the importance of monitoring and maintaining adequate serum levels of vitamin D, in order to reduce possible cognitive sequelae. Results will be announced at national and international conferences. PROSPERO REGISTRATION NUMBER CRD42018100074.
Collapse
Affiliation(s)
| | | | - Renata Alves de Sousa
- Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, UFPE, Recife, Brazil
| | | | | | - Everton Botelho Sougey
- Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, UFPE, Recife, Brazil
| |
Collapse
|
14
|
Albiñana C, Zhu Z, Borbye-Lorenzen N, Boelt SG, Cohen AS, Skogstrand K, Wray NR, Revez JA, Privé F, Petersen LV, Bulik CM, Plana-Ripoll O, Musliner KL, Agerbo E, Børglum AD, Hougaard DM, Nordentoft M, Werge T, Mortensen PB, Vilhjálmsson BJ, McGrath JJ. Genetic correlates of vitamin D-binding protein and 25-hydroxyvitamin D in neonatal dried blood spots. Nat Commun 2023; 14:852. [PMID: 36792583 PMCID: PMC9932173 DOI: 10.1038/s41467-023-36392-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
The vitamin D binding protein (DBP), encoded by the group-specific component (GC) gene, is a component of the vitamin D system. In a genome-wide association study of DBP concentration in 65,589 neonates we identify 26 independent loci, 17 of which are in or close to the GC gene, with fine-mapping identifying 2 missense variants on chromosomes 12 and 17 (within SH2B3 and GSDMA, respectively). When adjusted for GC haplotypes, we find 15 independent loci distributed over 10 chromosomes. Mendelian randomization analyses identify a unidirectional effect of higher DBP concentration and (a) higher 25-hydroxyvitamin D concentration, and (b) a reduced risk of multiple sclerosis and rheumatoid arthritis. A phenome-wide association study confirms that higher DBP concentration is associated with a reduced risk of vitamin D deficiency. Our findings provide valuable insights into the influence of DBP on vitamin D status and a range of health outcomes.
Collapse
Affiliation(s)
- Clara Albiñana
- National Centre for Register-Based Research, Aarhus University, 8210, Aarhus V, Denmark
| | - Zhihong Zhu
- National Centre for Register-Based Research, Aarhus University, 8210, Aarhus V, Denmark
| | - Nis Borbye-Lorenzen
- Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Sanne Grundvad Boelt
- Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Clinical Mass Spectrometry, Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark
| | - Arieh S Cohen
- Testcenter Denmark, Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark
| | - Kristin Skogstrand
- Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Naomi R Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Joana A Revez
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Florian Privé
- National Centre for Register-Based Research, Aarhus University, 8210, Aarhus V, Denmark
| | - Liselotte V Petersen
- National Centre for Register-Based Research, Aarhus University, 8210, Aarhus V, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8210, Aarhus V, Denmark
| | - Cynthia M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Oleguer Plana-Ripoll
- National Centre for Register-Based Research, Aarhus University, 8210, Aarhus V, Denmark
- Department of Clinical Epidemiology, Aarhus University and Aarhus University Hospital, 8200, Aarhus N, Denmark
| | - Katherine L Musliner
- Department of Affective Disorders, Aarhus University and Aarhus University Hospital-Psychiatry, Aarhus, Denmark
| | - Esben Agerbo
- National Centre for Register-Based Research, Aarhus University, 8210, Aarhus V, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8210, Aarhus V, Denmark
- CIRRAU - Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Anders D Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8210, Aarhus V, Denmark
- Center for Genomics and Personalized Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine and the iSEQ Center, Aarhus University, Aarhus, Denmark
| | - David M Hougaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8210, Aarhus V, Denmark
- Department for Congenital Disorders, Statens Serum Institut, 2300, Copenhagen S, Denmark
| | - Merete Nordentoft
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8210, Aarhus V, Denmark
- Mental Health Services in the Capital Region of Denmark, Mental Health Center Copenhagen, University of Copenhagen, 2100, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Thomas Werge
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8210, Aarhus V, Denmark
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen N, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Lundbeck Center for Geogenetics, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Preben Bo Mortensen
- National Centre for Register-Based Research, Aarhus University, 8210, Aarhus V, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8210, Aarhus V, Denmark
- CIRRAU - Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Bjarni J Vilhjálmsson
- National Centre for Register-Based Research, Aarhus University, 8210, Aarhus V, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8210, Aarhus V, Denmark
- Bioinformatics Research Centre, Aarhus University, 8000, Aarhus C, Denmark
| | - John J McGrath
- National Centre for Register-Based Research, Aarhus University, 8210, Aarhus V, Denmark.
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia.
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Brisbane, QLD, 4076, Australia.
| |
Collapse
|
15
|
Orton SM, Sangha A, Gupta M, Martens K, Metz LM, de Koning APJ, Pfeffer G. Expression of risk genes linked to vitamin D receptor super-enhancer regions and their association with phenotype severity in multiple sclerosis. Front Neurol 2022; 13:1064008. [PMID: 36644209 PMCID: PMC9832371 DOI: 10.3389/fneur.2022.1064008] [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: 10/07/2022] [Accepted: 12/05/2022] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic debilitating neurological condition with a wide range of phenotype variability. A complex interplay of genetic and environmental factors contributes to disease onset and progression in MS patients. Vitamin D deficiency is a known susceptibility factor for MS, however the underlying mechanism of vitamin D-gene interactions in MS etiology is still poorly understood. Vitamin D receptor super-enhancers (VSEs) are enriched in MS risk variants and may modulate these environment-gene interactions. mRNA expression in total of 64 patients with contrasting MS severity was quantified in select genes. First, RNA-seq was performed on a discovery cohort (10 mild, 10 severe MS phenotype) and ten genes regulated by VSEs that have been linked to MS risk were analyzed. Four candidates showed a significant positive association (GRINA, PLEC, PARP10, and LRG1) in the discovery cohort and were then quantified using digital droplet PCR (ddPCR) in a validation cohort (33 mild, 11 severe MS phenotype). A significant differential expression persisted in the validation cohort for three of the VSE-MS genes: GRINA (p = 0.0138), LRG1 (p = 0.0157), and PLEC (p = 0.0391). In summary, genes regulated by VSE regions that contain known MS risk variants were shown to have differential expression based on disease severity (p<0.05). The findings implicate a role for vitamin D super-enhancers in modulating disease activity. In addition, expression levels may have some utility as prognostic biomarkers in the future.
Collapse
Affiliation(s)
- Sarah M. Orton
- Faculty of Science and Technology, Mount Royal University, Calgary, AB, Canada,*Correspondence: Sarah M. Orton ✉
| | - Amarpreet Sangha
- Faculty of Science and Technology, Mount Royal University, Calgary, AB, Canada
| | - Mehul Gupta
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Kristina Martens
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Luanne M. Metz
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - A. P. J. de Koning
- Department of Medical Genetics, Alberta Child Health Research Institute, Cumming of Medicine, University of Calgary, Calgary, AB, Canada
| | - Gerald Pfeffer
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,Department of Medical Genetics, Alberta Child Health Research Institute, Cumming of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
16
|
Stezin A, Pal PK. Treatable Ataxias: How to Find the Needle in the Haystack? J Mov Disord 2022; 15:206-226. [PMID: 36065614 DOI: 10.14802/jmd.22069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/05/2022] [Indexed: 11/24/2022] Open
Abstract
Treatable ataxias are a group of ataxic disorders with specific treatments. These disorders include genetic and metabolic disorders, immune-mediated ataxic disorders, and ataxic disorders associated with infectious and parainfectious etiology, vascular causes, toxins and chemicals, and endocrinopathies. This review provides a comprehensive overview of different treatable ataxias. The major metabolic and genetic treatable ataxic disorders include ataxia with vitamin E deficiency, abetalipoproteinemia, cerebrotendinous xanthomatosis, Niemann-Pick disease type C, autosomal recessive cerebellar ataxia due to coenzyme Q10 deficiency, glucose transporter type 1 deficiency, and episodic ataxia type 2. The treatment of these disorders includes the replacement of deficient cofactors and vitamins, dietary modifications, and other specific treatments. Treatable ataxias with immune-mediated etiologies include gluten ataxia, anti-glutamic acid decarboxylase antibody-associated ataxia, steroid-responsive encephalopathy associated with autoimmune thyroiditis, Miller-Fisher syndrome, multiple sclerosis, and paraneoplastic cerebellar degeneration. Although dietary modification with a gluten-free diet is adequate in gluten ataxia, other autoimmune ataxias are managed by short-course steroids, plasma exchange, or immunomodulation. For autoimmune ataxias secondary to malignancy, treatment of tumor can reduce ataxic symptoms. Chronic alcohol consumption, antiepileptics, anticancer drugs, exposure to insecticides, heavy metals, and recreational drugs are potentially avoidable and treatable causes of ataxia. Infective and parainfectious causes of cerebellar ataxias include acute cerebellitis, postinfectious ataxia, Whipple's disease, meningoencephalitis, and progressive multifocal leukoencephalopathy. These disorders are treated with steroids and antibiotics. Recognizing treatable disorders is of paramount importance when dealing with ataxias given that early treatment can prevent permanent neurological sequelae.
Collapse
Affiliation(s)
- Albert Stezin
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India.,Centre for Brain Research, Indian Institute of Science, Bengaluru, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| |
Collapse
|
17
|
Albiñana C, Boelt SG, Cohen AS, Zhu Z, Musliner KL, Vilhjálmsson BJ, McGrath JJ. Developmental exposure to vitamin D deficiency and subsequent risk of schizophrenia. Schizophr Res 2022; 247:26-32. [PMID: 34247885 DOI: 10.1016/j.schres.2021.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 01/22/2023]
Abstract
Over the last half century, a body of convergent evidence has accumulated linking disruption of early brain development with an increased risk of mental disorders, including schizophrenia. The orderly cascade of brain development may be disrupted by exposure to suboptimal concentrations of a range of biological substrates and micronutrients. We hypothesized that those exposed to vitamin D deficiency during early life, have an increased risk of neurodevelopmental disorders, including schizophrenia. The hypothesis was based on the link between an increased risk of schizophrenia in (a) those born in winter and spring, when vitamin D deficiency is more prevalent, and (b) the offspring of dark-skinned migrants living in cold climates, who have a markedly increased risk of vitamin D deficiency. In this review, we summarize evidence from analytic epidemiology related to this hypothesis. Two case-control studies based on Danish neonatal dried blood spots have found that neonatal vitamin deficiency is associated with an increased risk of schizophrenia. However, recent genetic analyses have also suggested that common variants linked to schizophrenia may lead to lower vitamin D concentrations (possibly mediated via reduced outdoor activity). We summarize limitations of the current evidence and outline suggestions that can guide future research. Based on currently available data, there is insufficient evidence to support public health recommendations related to this topic. However, we cannot reject the hypothesis that the provision of vitamin D supplementation to pregnant women and/or offspring in groups vulnerable to vitamin D deficiency may subsequently reduce the incidence of schizophrenia in the offspring.
Collapse
Affiliation(s)
- Clara Albiñana
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark; iPSYCH - the Lundbeck Foundation Initiative for Integrative Psychiatric Research, Denmark
| | | | - Arieh S Cohen
- Department of Inherited Diseases, Statens Serum Institut, Copenhagen, Denmark
| | - Zhihong Zhu
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
| | - Katherine L Musliner
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark; iPSYCH - the Lundbeck Foundation Initiative for Integrative Psychiatric Research, Denmark
| | - Bjarni J Vilhjálmsson
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark; iPSYCH - the Lundbeck Foundation Initiative for Integrative Psychiatric Research, Denmark; Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - John J McGrath
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark; Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, Australia; Queensland Brain Institute, The University of Queensland, St Lucia, Australia.
| |
Collapse
|
18
|
Zorrilla Veloz RI, McKenzie T, Palacios BE, Hu J. Nuclear hormone receptors in demyelinating diseases. J Neuroendocrinol 2022; 34:e13171. [PMID: 35734821 PMCID: PMC9339486 DOI: 10.1111/jne.13171] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/20/2022] [Accepted: 05/27/2022] [Indexed: 11/28/2022]
Abstract
Demyelination results from the pathological loss of myelin and is a hallmark of many neurodegenerative diseases. Despite the prevalence of demyelinating diseases, there are no disease modifying therapies that prevent the loss of myelin or promote remyelination. This review aims to summarize studies in the field that highlight the importance of nuclear hormone receptors in the promotion and maintenance of myelination and the relevance of nuclear hormone receptors as potential therapeutic targets for demyelinating diseases. These nuclear hormone receptors include the estrogen receptor, progesterone receptor, androgen receptor, vitamin D receptor, thyroid hormone receptor, peroxisome proliferator-activated receptor, liver X receptor, and retinoid X receptor. Pre-clinical studies in well-established animal models of demyelination have shown a prominent role of these nuclear hormone receptors in myelination through their promotion of oligodendrocyte maturation and development. The activation of the nuclear hormone receptors by their ligands also promotes the synthesis of myelin proteins and lipids in mouse models of demyelination. There are limited clinical studies that focus on how the activation of these nuclear hormone receptors could alleviate demyelination in patients with diseases such as multiple sclerosis (MS). However, the completed clinical trials have reported improved clinical outcome in MS patients treated with the ligands of some of these nuclear hormone receptors. Together, the positive results from both clinical and pre-clinical studies point to nuclear hormone receptors as promising therapeutic targets to counter demyelination.
Collapse
Affiliation(s)
- Rocío I Zorrilla Veloz
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Takese McKenzie
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Bridgitte E Palacios
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Neuroscience Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
19
|
González-Madrid E, Rangel-Ramírez MA, Mendoza-León MJ, Álvarez-Mardones O, González PA, Kalergis AM, Opazo MC, Riedel CA. Risk Factors from Pregnancy to Adulthood in Multiple Sclerosis Outcome. Int J Mol Sci 2022; 23:ijms23137080. [PMID: 35806081 PMCID: PMC9266360 DOI: 10.3390/ijms23137080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by a robust inflammatory response against myelin sheath antigens, which causes astrocyte and microglial activation and demyelination of the central nervous system (CNS). Multiple genetic predispositions and environmental factors are known to influence the immune response in autoimmune diseases, such as MS, and in the experimental autoimmune encephalomyelitis (EAE) model. Although the predisposition to suffer from MS seems to be a multifactorial process, a highly sensitive period is pregnancy due to factors that alter the development and differentiation of the CNS and the immune system, which increases the offspring’s susceptibility to develop MS. In this regard, there is evidence that thyroid hormone deficiency during gestation, such as hypothyroidism or hypothyroxinemia, may increase susceptibility to autoimmune diseases such as MS. In this review, we discuss the relevance of the gestational period for the development of MS in adulthood.
Collapse
Affiliation(s)
- Enrique González-Madrid
- Laboratorio Endocrinología-Inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8320000, Chile; (E.G.-M.); (M.A.R.-R.); (M.J.M.-L.); (O.Á.-M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8320000, Chile; (P.A.G.); (A.M.K.); (M.C.O.)
| | - Ma. Andreina Rangel-Ramírez
- Laboratorio Endocrinología-Inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8320000, Chile; (E.G.-M.); (M.A.R.-R.); (M.J.M.-L.); (O.Á.-M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8320000, Chile; (P.A.G.); (A.M.K.); (M.C.O.)
| | - María José Mendoza-León
- Laboratorio Endocrinología-Inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8320000, Chile; (E.G.-M.); (M.A.R.-R.); (M.J.M.-L.); (O.Á.-M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8320000, Chile; (P.A.G.); (A.M.K.); (M.C.O.)
| | - Oscar Álvarez-Mardones
- Laboratorio Endocrinología-Inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8320000, Chile; (E.G.-M.); (M.A.R.-R.); (M.J.M.-L.); (O.Á.-M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8320000, Chile; (P.A.G.); (A.M.K.); (M.C.O.)
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Santiago 8320000, Chile; (P.A.G.); (A.M.K.); (M.C.O.)
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago 8320000, Chile; (P.A.G.); (A.M.K.); (M.C.O.)
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
- Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Ma. Cecilia Opazo
- Millennium Institute on Immunology and Immunotherapy, Santiago 8320000, Chile; (P.A.G.); (A.M.K.); (M.C.O.)
- Instituto de Ciencias Naturales, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Manuel Montt 948, Providencia 7500000, Chile
| | - Claudia A. Riedel
- Laboratorio Endocrinología-Inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8320000, Chile; (E.G.-M.); (M.A.R.-R.); (M.J.M.-L.); (O.Á.-M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8320000, Chile; (P.A.G.); (A.M.K.); (M.C.O.)
- Correspondence:
| |
Collapse
|
20
|
Gombash SE, Lee PW, Sawdai E, Lovett-Racke AE. Vitamin D as a Risk Factor for Multiple Sclerosis: Immunoregulatory or Neuroprotective? Front Neurol 2022; 13:796933. [PMID: 35651353 PMCID: PMC9149265 DOI: 10.3389/fneur.2022.796933] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 04/13/2022] [Indexed: 12/18/2022] Open
Abstract
Vitamin D insufficiency during childhood has been linked to the development of multiple sclerosis (MS), typically an adult-onset inflammatory demyelinating disease of the central nervous system (CNS). Since vitamin D was known to have immunoregulatory properties on both innate and adaptive immunity, it was hypothesized that low vitamin D resulted in aberrant immune responses and the development of MS. However, vitamin D receptors are present on many cell types, including neurons, oligodendrocytes, astrocytes and microglia, and vitamin D has profound effects on development and function of the CNS. This leads to the possibility that low vitamin D may alter the CNS in a manner that makes it vulnerable to inflammation and the development of MS. This review analysis the role of vitamin D in the immune and nervous system, and how vitamin D insufficiency in children may contribute to the development of MS.
Collapse
Affiliation(s)
- Sara E Gombash
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Priscilla W Lee
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Elizabeth Sawdai
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Amy E Lovett-Racke
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
21
|
Boelt SG, Plana-Ripoll O, Albiñana C, Vilhjálmsson B, McGrath JJ, Cohen AS. A method to correct for the influence of bovine serum albumin-associated vitamin D metabolites in protein extracts from neonatal dried blood spots. BMC Res Notes 2022; 15:194. [PMID: 35659347 PMCID: PMC9166528 DOI: 10.1186/s13104-022-06077-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/18/2022] [Indexed: 12/04/2022] Open
Abstract
Objective We developed an assay to measure the concentration of 25 hydroxyvitamin D2 and D3 in protein extracts derived from stored neonatal dried blood spots. During this study, we postulated that these samples had been contaminated with exogenous vitamin D metabolites because of the addition of bovine serum albumin (BSA) as part of an extraction step undertaken 7 years earlier. The aim of the current study was to develop methods in order to adjust for this contamination. Results We identified between-plate variations in 25 hydroxyvitamin D2 and D3 concentrations which suggested the presence of three different BSA batches. Based on repeat extraction (without the addition of BSA) and testing of 395 samples, we developed models to correct for the exogenous 25 hydroxyvitamin D2 and D3. The regression models were Diff25OHD3 = − 8.2 + 1.8* Diff25OHD2 for low contamination, Diff25OHD3 = 23.8 + 1.7* Diff25OHD2 for middle contamination, and Diff25OHD3 = 14.3 + 3.0* Diff25OHD2 for high contamination. After these corrections, the three subsamples had comparable distributions within the expected range for both 25 hydroxyvitamin D2 and D3. Supplementary Information The online version contains supplementary material available at 10.1186/s13104-022-06077-1.
Collapse
Affiliation(s)
- Sanne Grundvad Boelt
- Center for Neonatal Screening, Department of Congenital Disorders-Clinical Mass Spectrometry Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark
| | - Oleguer Plana-Ripoll
- National Centre for Register-Based Research, Department of Economics and Business Economics, Aarhus University, Fuglesangs Allé 26, Building 2640 Aarhus V, DK-8210, Aarhus, Denmark.,Department of Clinical Epidemiology, Aarhus University and Aarhus University Hospital, Olof Palmes Allé 43-45 Aarhus N, 8200, Aarhus, Denmark
| | - Clara Albiñana
- National Centre for Register-Based Research, Department of Economics and Business Economics, Aarhus University, Fuglesangs Allé 26, Building 2640 Aarhus V, DK-8210, Aarhus, Denmark
| | - Bjarni Vilhjálmsson
- National Centre for Register-Based Research, Department of Economics and Business Economics, Aarhus University, Fuglesangs Allé 26, Building 2640 Aarhus V, DK-8210, Aarhus, Denmark.,Bioinformatics Research Centre, Aarhus University, C.F. Møllers Allé 8, Building 1110 Aarhus C, DK-8000, Aarhus, Denmark
| | - John J McGrath
- National Centre for Register-Based Research, Department of Economics and Business Economics, Aarhus University, Fuglesangs Allé 26, Building 2640 Aarhus V, DK-8210, Aarhus, Denmark. .,Queensland Centre for Mental Health Research, The Park Centre for Mental Health, University of Queensland, St. Lucia, QLD-4072, Australia. .,Queensland Brain Institute, University of Queensland, St. Lucia, QLD-4072, Australia.
| | - Arieh S Cohen
- Center for Neonatal Screening, Department of Congenital Disorders-Clinical Mass Spectrometry Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark
| |
Collapse
|
22
|
Mazrouei-Arani N, Zargar M, Nikuinejad H. Association between ApaI and TaqI polymorphisms of the vitamin D receptor gene and the multiple sclerosis. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
23
|
Makkawi S, Aljabri A, Bin Lajdam G, Albakistani A, Aljohani A, Labban S, Felemban R. Effect of Seasonal Variation on Relapse Rate in Patients With Relapsing-Remitting Multiple Sclerosis in Saudi Arabia. Front Neurol 2022; 13:862120. [PMID: 35359633 PMCID: PMC8964008 DOI: 10.3389/fneur.2022.862120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is becoming a global subject of study in which some demographic variations are thought to be correlated with its activity. Relapsing-remitting multiple sclerosis (RRMS) is the most common demyelinating disorder, characterized by periods of exacerbating attacks, followed by partial or complete remission. Several factors might play a role in disease progression and relapse frequency, such as vitamin D, ultraviolet B radiation, estrogen levels, smoking, obesity, and unhealthy lifestyles. In this study, we identified the relationship between seasonal variation and relapse rate and correlated the latter with sex, age, and vitamin D levels in patients with RRMS in Jeddah, Saudi Arabia. We retrospectively collected data from 182 RRMS patients between 2016 and 2021. A total of 219 relapses were documented in 106 patients (58.2 %). The relapse per patient ratio showed a sinusoidal pattern, peaking in January at a rate of 0.49 and troughed in June at a rate of 0.18. There was no difference in relapse rates between men and women (p =0.280). There was a significant negative correlation between vitamin D levels and relapse rate (r = −0.312, p =0.024). Therefore, the relapse rate was higher during the winter and was correlated with low vitamin D levels. However, relapses are likely multifactorial, and more population-based studies are needed to understand the role of environmental variables in MS exacerbation. A better understanding of this relationship will allow for improved treatment and possibly better prevention of relapse.
Collapse
Affiliation(s)
- Seraj Makkawi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia.,King Abdullah International Medical Research Center, Jeddah, Saudi Arabia.,Department of Medicine, Ministry of the National Guard-Health Affairs, Jeddah, Saudi Arabia
| | - Ammar Aljabri
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia.,King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Ghassan Bin Lajdam
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia.,King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Ammar Albakistani
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia.,King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Abdulrahman Aljohani
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia.,King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Suhail Labban
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia.,King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Razaz Felemban
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia.,King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| |
Collapse
|
24
|
Ingelfinger F, Gerdes LA, Kavaka V, Krishnarajah S, Friebel E, Galli E, Zwicky P, Furrer R, Peukert C, Dutertre CA, Eglseer KM, Ginhoux F, Flierl-Hecht A, Kümpfel T, De Feo D, Schreiner B, Mundt S, Kerschensteiner M, Hohlfeld R, Beltrán E, Becher B. Twin study reveals non-heritable immune perturbations in multiple sclerosis. Nature 2022; 603:152-158. [PMID: 35173329 PMCID: PMC8891021 DOI: 10.1038/s41586-022-04419-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 01/04/2022] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system underpinned by partially understood genetic risk factors and environmental triggers and their undefined interactions1,2. Here we investigated the peripheral immune signatures of 61 monozygotic twin pairs discordant for MS to dissect the influence of genetic predisposition and environmental factors. Using complementary multimodal high-throughput and high-dimensional single-cell technologies in conjunction with data-driven computational tools, we identified an inflammatory shift in a monocyte cluster of twins with MS, coupled with the emergence of a population of IL-2 hyper-responsive transitional naive helper T cells as MS-related immune alterations. By integrating data on the immune profiles of healthy monozygotic and dizygotic twin pairs, we estimated the variance in CD25 expression by helper T cells displaying a naive phenotype to be largely driven by genetic and shared early environmental influences. Nonetheless, the expanding helper T cells of twins with MS, which were also elevated in non-twin patients with MS, emerged independent of the individual genetic makeup. These cells expressed central nervous system-homing receptors, exhibited a dysregulated CD25-IL-2 axis, and their proliferative capacity positively correlated with MS severity. Together, our matched-pair analysis of the extended twin approach allowed us to discern genetically and environmentally determined features of an MS-associated immune signature.
Collapse
Affiliation(s)
- Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Lisa Ann Gerdes
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Vladyslav Kavaka
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
| | | | - Ekaterina Friebel
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Edoardo Galli
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Neurologic Clinic and Policlinic, University Hospital Basel, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Reinhard Furrer
- Department of Mathematics, University of Zurich, Zurich, Switzerland
- Department of Computational Science, University of Zurich, Zurich, Switzerland
| | - Christian Peukert
- Department of Strategy, Globalization and Society, University of Lausanne, Lausanne, Switzerland
| | - Charles-Antoine Dutertre
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Klara Magdalena Eglseer
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
| | | | - Andrea Flierl-Hecht
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Donatella De Feo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Bettina Schreiner
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Eduardo Beltrán
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
25
|
Hardy D, Chitnis T, Waubant E, Banwell B. Preventing Multiple Sclerosis: The Pediatric Perspective. Front Neurol 2022; 13:802380. [PMID: 35280298 PMCID: PMC8913516 DOI: 10.3389/fneur.2022.802380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
Pediatric-onset multiple sclerosis (MS) is a predominantly relapsing-remitting neuroinflammatory disorder characterized by frequent relapses and high magnetic resonance imaging (MRI) lesion burden early in the disease course. Current treatment for pediatric MS relies on early initiation of disease-modifying therapies designed to prevent relapses and slow progression of disability. When considering the concept of MS prevention, one can conceptualize primary prevention (population- or at-risk population interventions that prevent the earliest facet of MS pathobiology and hence reduce disease incidence), or secondary prevention (prevention of disease consequence, such as reducing relapse frequency and lesion accrual, enhancing focal lesion repair, promoting CNS resilience against the more global facets of disease injury, and ultimately, preventing progression of neurological disability). Studying the pediatric MS population provides a unique opportunity to explore early-life exposures that contribute to the development of MS including perinatal and environmental risk determinants. Research is ongoing related to targeting these risk factors for potential MS primary prevention. Here we review these key risk factors, their proposed role in the pathogenesis of MS, and their potential implications for primary MS prevention.
Collapse
Affiliation(s)
- Duriel Hardy
- Dell Children's Medical Center of Central Texas, Austin, TX, United States
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- *Correspondence: Duriel Hardy
| | - Tanuja Chitnis
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Emmanuelle Waubant
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
- San Francisco Multiple Sclerosis Center, University of California, San Francisco, San Francisco, CA, United States
| | - Brenda Banwell
- Center for Neuroinflammation and Neurotherapeutics, and Multiple Sclerosis Division, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Child Neurology, Department of Neurology, The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
26
|
Tremlett H, Munger KL, Makhani N. The Multiple Sclerosis Prodrome: Evidence to Action. Front Neurol 2022; 12:761408. [PMID: 35173664 PMCID: PMC8841819 DOI: 10.3389/fneur.2021.761408] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/14/2021] [Indexed: 01/10/2023] Open
Abstract
A growing body of work points toward the existence of a clinically symptomatic prodromal phase in multiple sclerosis (MS) that might span 5–10 years or more. A prodrome is an early set of signs or symptoms predating the onset of classical disease, which in turn predates a definitive diagnosis. Evidence for a prodromal phase in MS could have major implications for prevention, earlier recognition and treatment, as well as an improved disease course or prognosis. This Perspective provides a succinct overview of the recent advances in our understanding of the MS prodrome and current key challenges. Many of the MS prodromal features characterized thus far are non-specific and are common in the general population; no single feature alone is sufficient to identify an individual with prodromal MS. Biomarkers may increase specificity and accuracy for detecting individuals in the MS prodromal phase, but are yet to be discovered or formally validated. Progress made in the elucidation of prodromal phases in other neurological and immune-mediated diseases suggests that these barriers can be overcome. Therefore, while knowledge of a prodromal phase in MS remains nascent, how best to move from the rapidly growing evidence to research-related action is critical. Immediate implications include refining the concept of the MS continuum to include a prodromal phase. This will help inform the true “at risk” period when considering exposures that might cause MS. Major long-term implications include the earlier recognition of MS, improved prognosis, through earlier disease management, and the future possibility of MS disease prevention.
Collapse
Affiliation(s)
- Helen Tremlett
- Faculty of Medicine (Neurology), University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Helen Tremlett
| | | | - Naila Makhani
- Departments of Pediatrics and Neurology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
27
|
Serum Vitamin D as a Biomarker in Autoimmune, Psychiatric and Neurodegenerative Diseases. Diagnostics (Basel) 2022; 12:diagnostics12010130. [PMID: 35054296 PMCID: PMC8774449 DOI: 10.3390/diagnostics12010130] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 02/05/2023] Open
Abstract
Vitamin D is a steroid hormone regulating calcium-phosphorus homeostasis, immune response and brain function. In the past thirty years, an increasing number of cohort studies, meta-analyses and randomized controlled trials (RTCs) evaluated the serum levels of 25-hydroxyvitamin D [25(OH)D], which is considered the Vitamin D status biomarker, in patients affected by neurological, psychiatric and autoimmune diseases. Although an association between low 25(OH)D serum levels and the prevalence of these diseases has been found, it is still unclear whether the serum 25(OH)D measurement can be clinically useful as a biomarker for diagnosis, prognosis and predicting treatment response in neurodegeneration, mental illness and immune-mediated disorders. The lack of standardized data, as well as discrepancies among the studies (in the analytical methods, cut-offs, endpoints and study sets), weakened the findings achieved, hindered pooling data, and, consequently, hampered drawing conclusions. This narrative review summarizes the main findings from the studies performed on serum 25(OH)D in neurological, psychiatric and autoimmune diseases, and clarifies whether or not serum 25(OH)D can be used as a reliable biomarker in these diseases.
Collapse
|
28
|
Kalincik T, Tremlett H, Ascherio A. High BMI in Youths as a Modifiable Risk Factor for Multiple Sclerosis: Weighing Up the Evidence. Neurology 2021; 97:1057-1058. [PMID: 34697244 DOI: 10.1212/wnl.0000000000012966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Tomas Kalincik
- From the CORe (T.K.), Department of Medicine, University of Melbourne; MS Centre (T.K.), Department of Neurology, Royal Melbourne Hospital, Australia; Faculty of Medicine (Neurology) and the Djavad Mowafaghian Centre for Brain Health (H.T.), University of British Columbia, Vancouver, Canada; and Harvard School of Public Health (A.A.), Boston, MA.
| | - Helen Tremlett
- From the CORe (T.K.), Department of Medicine, University of Melbourne; MS Centre (T.K.), Department of Neurology, Royal Melbourne Hospital, Australia; Faculty of Medicine (Neurology) and the Djavad Mowafaghian Centre for Brain Health (H.T.), University of British Columbia, Vancouver, Canada; and Harvard School of Public Health (A.A.), Boston, MA
| | - Alberto Ascherio
- From the CORe (T.K.), Department of Medicine, University of Melbourne; MS Centre (T.K.), Department of Neurology, Royal Melbourne Hospital, Australia; Faculty of Medicine (Neurology) and the Djavad Mowafaghian Centre for Brain Health (H.T.), University of British Columbia, Vancouver, Canada; and Harvard School of Public Health (A.A.), Boston, MA
| |
Collapse
|
29
|
Mimpen M, Rolf L, Poelmans G, van den Ouweland J, Hupperts R, Damoiseaux J, Smolders J. Vitamin D related genetic polymorphisms affect serological response to high-dose vitamin D supplementation in multiple sclerosis. PLoS One 2021; 16:e0261097. [PMID: 34855907 PMCID: PMC8638856 DOI: 10.1371/journal.pone.0261097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/23/2021] [Indexed: 01/08/2023] Open
Abstract
Introduction A poor 25-hydroxyvitamin D (25(OH)D) status is a much replicated risk factor for developing multiple sclerosis (MS), and several vitamin D-associated single nucleotide polymorphisms (SNPs) have been associated with a higher risk of MS. However, studies on the benefit of vitamin D supplementation in MS show inconclusive results. Here, we explore whether vitamin D-associated SNPs and MS risk alleles confound serological response to vitamin D supplementation. Methods 34 participants from the SOLARIUM study consented to genotyping, of which 26 had vitamin D data available. The SOLARIUM study randomised relapsing-remitting MS patients to placebo or 14,000 IU vitamin D3 for 48 weeks. Participants were categorised as either ‘carriers’ or ‘non-carriers’ of the risk allele for 4 SNPs: two related to D binding protein (DBP) and associated with lower 25(OH)D levels (rs4588 and rs7041), and two related to vitamin D metabolism enzymes CYP27B1 and CYP24A1 and associated with a higher risk of MS (rs12368653; rs2248359, respectively). 25(OH)D levels were determined at baseline and after 48 weeks. Results The DBP-related SNPs showed no difference in 25(OH)D status at baseline, but carriers of the rs7041 risk allele showed lower 25(OH)D-levels compared to non-carriers after 48 weeks of supplementation (median 224.2 vs. 332.0 nmol/L, p = 0.013). For CYP related SNPs, neither showed a difference at baseline, but carriers of the rs12368653 risk allele showed higher 25(OH)D-levels compared to non-carriers after 48 weeks of supplementation (median 304.1 vs. 152.0 nmol/L, p = 0.014). Discussion Vitamin D-related SNPs affect the serological response to high-dose vitamin D supplementation. The effects on more common doses of vitamin D, as well as the clinical consequence of this altered response, need to be investigated further.
Collapse
Affiliation(s)
- Max Mimpen
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Linda Rolf
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Geert Poelmans
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jody van den Ouweland
- Department of Clinical Chemistry, Canisius-Wilhelmina Hospital, Nijmegen, Netherlands
| | - Raymond Hupperts
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Department of Neurology, Zuyderland Medical Center, Sittard-Geleen, Netherlands
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, Netherlands
| | - Joost Smolders
- MS Center ErasMS, Departments of Neurology and Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
- * E-mail:
| |
Collapse
|
30
|
Stridh P, Huang J, Hedström AK, Alfredsson L, Olsson T, Hillert J, Manouchehrinia A, Kockum I. Season of birth is associated with multiple sclerosis and disease severity. Mult Scler J Exp Transl Clin 2021; 7:20552173211065730. [PMID: 35035988 PMCID: PMC8753082 DOI: 10.1177/20552173211065730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/19/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The latitude gradient in multiple sclerosis incidence indicates that low sun exposure and therefore vitamin D deficiency is associated with multiple sclerosis risk. OBJECTIVE Investigation of the effect of month of birth, which influences postnatal vitamin D levels, on multiple sclerosis risk and severity in Sweden. METHODS Patients and population-based controls were included from three nationwide cohorts. Differences in month of birth between cases and controls were analyzed using logistic regression and examined for effect modification by calendar year and geographic region at birth. RESULTS Males had a reduced risk of multiple sclerosis if born in the winter and increased risk if born in the early fall. Individuals born before 1960 had an increased risk if born in summer or fall. Being born in late summer and early fall was associated with more severe disease. CONCLUSIONS We identified a birth cohort effect on the association between the month of birth and multiple sclerosis, with a more significant effects for births before 1960. This coincides with a period of lower breastfeeding rates, recommended intake of vitamin D, and sun exposure, resulting in a lower vitamin D exposure during the fall/winter season for infants born in the summer.
Collapse
Affiliation(s)
- P Stridh
- Pernilla Stridh, Centrum for
Molecular Medicine, Karolinska University Hospital, Solna, L8:05, SE-171 76
Stockholm, Sweden
| | - J Huang
- Center of Molecular Medicine, Karolinska University
Hospital, Solna, Sweden
- Department of Clinical Neuroscience, Karolinska
Institutet, Stockholm, Sweden
| | | | - L Alfredsson
- Department of Clinical Neuroscience, Karolinska
Institutet, Stockholm, Sweden
- Institute of Environmental Medicine, Karolinska
Institutet, Stockholm, Sweden
| | | | | | | | - I Kockum
- Center of Molecular Medicine, Karolinska University
Hospital, Solna, Sweden
- Department of Clinical Neuroscience, Karolinska
Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Greenberg BM, Casper TC, Mar SS, Ness JM, Plumb P, Liang S, Goyal M, Weinstock-Guttman B, Rodriguez M, Aaen GS, Belman A, Barcellos LF, Rose JW, Gorman MP, Benson LA, Candee M, Chitnis T, Harris YC, Kahn IL, Roalstad S, Hart J, Lotze TE, Rensel M, Rubin JP, Schreiner TL, Tillema JM, Waldman AT, Krupp L, Graves J, Drake K, Waubant E. Familial History of Autoimmune Disorders Among Patients With Pediatric Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:e1049. [PMID: 34353894 PMCID: PMC8362349 DOI: 10.1212/nxi.0000000000001049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 06/17/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND OBJECTIVE The objective of this study was to determine whether family members of patients with pediatric multiple sclerosis (MS) have an increased prevalence of autoimmune conditions compared with controls. METHODS Data collected during a pediatric MS case-control study of risk factors included information about various autoimmune diseases in family members. The frequency of these disorders was compared between cases and controls. RESULTS There was an increased rate of autoimmune diseases among family members of pediatric MS cases compared with controls with first-degree history of MS excluded (OR = 2.27, 95% CI 1.71-3.01, p < 0.001). There was an increased rate of MS among second-degree relatives of pediatric MS cases compared with controls (OR = 3.47, 95% CI 1.36-8.86, p = 0.009). The OR for MS was 2.64 when restricted to maternal relatives and 6.37 when restricted to paternal relatives. DISCUSSION The increased rates of autoimmune disorders, including thyroid disorders and MS among families of patients with pediatric MS, suggest shared genetic factors among families with children diagnosed with pediatric MS.
Collapse
Affiliation(s)
- Benjamin M. Greenberg
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Theron Charles Casper
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Soe S. Mar
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Jayne M. Ness
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Patricia Plumb
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Shannon Liang
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Manu Goyal
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Bianca Weinstock-Guttman
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Moses Rodriguez
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Gregory S. Aaen
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Anita Belman
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Lisa F. Barcellos
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - John W. Rose
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Mark P. Gorman
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Leslie A. Benson
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Meghan Candee
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Tanuja Chitnis
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Yolanda C. Harris
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Ilana L. Kahn
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Shelly Roalstad
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Janace Hart
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Timothy E. Lotze
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Mary Rensel
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Jennifer P. Rubin
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Teri L. Schreiner
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Jan-Mendelt Tillema
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Amy Tara Waldman
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Lauren Krupp
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Jennifer Graves
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Kaylea Drake
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| | - Emmanuelle Waubant
- From the University of Texas Southwestern (B.M.G.), Department of Neurology, Department of Pediatrics, Dallas; Data Coordinating and Analysis Center (T.C.C., S.S.R., K.D.), University of Utah, Salt Lake City; Washington University (S.S.M.), St. Louis, MO; University of Alabama Birmingham (J.M.N.); The University of Texas Southwestern (P.P.), Department of Neurology, Dallas; Department of Radiology (S.L., M.G.), Washington University in St. Louis, MO; Jacobs Pediatric Multiple Sclerosis Center (B.W.-G.), State University of New York at Buffalo, NY; Mayo Clinic Pediatric Multiple Sclerosis Center (M.R., J.-M.T.), Mayo Clinic, Rochester, MN; Pediatric Multiple Sclerosis Center (G.S.A.), Loma Linda University Children's Hospital, CA; Lourie Center for Pediatric Multiple Sclerosis (A.B.), Stony Brook University Hospital, NY; Epidemiology (L.F.B.), University of California, Berkeley; Department of Neurology (J.W.R.), University of Utah, Salt Lake City; Pediatric Multiple Sclerosis and Related Disorders Program (M.P.G., L.A.B.), Boston Children's Hospital, MA; Primary Children's Hospital (M.C.), University of Utah, Salt Lake City; Partners Pediatric Multiple Sclerosis Center (T.C.), Massachusetts General Hospital, Boston; Center for Pediatric-Onset Demyelinating Disease (Y.C.H.), Children's Hospital of Alabama, University of Alabama, Birmingham; Children's National Medical Center (I.L.K.), Washington, DC; Pediatric Multiple Sclerosis Center (J.H.), University of California San Francisco; The Blue Bird Circle Clinic for Multiple Sclerosis (T.E.L.), Texas Children's Hospital, Baylor College of Medicine, Houston; Mellen Center for Multiple Sclerosis (M.R.), Cleveland Clinic, OH; Lurie Children's Hospital of Chicago (J.P.R.), IL; Rocky Mountain Multiple Sclerosis Center (T.L.S.), Children's Hospital Colorado, University of Colorado at Denver, Aurora; Children's Hospital of Philadelphia (A.T.W.), PA; Pediatric Multiple Sclerosis Center (L.K.), New York University; Pediatric Multiple Sclerosis Center (J.G.), University of California San Diego; and Pediatric Multiple Sclerosis Center (E.W.), University of California San Francisco
| |
Collapse
|
32
|
Keller A, Thorsteinsdottir F, Stougaard M, Cardoso I, Frederiksen P, Cohen AS, Vaag A, Jacobsen R, Heitmann BL. Vitamin D concentrations from neonatal dried blood spots and the risk of early-onset type 2 diabetes in the Danish D-tect case-cohort study. Diabetologia 2021; 64:1572-1582. [PMID: 34028586 DOI: 10.1007/s00125-021-05450-2] [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: 09/09/2020] [Accepted: 01/04/2021] [Indexed: 11/28/2022]
Abstract
AIMS/HYPOTHESIS The aim of this study was to examine the influence of neonatal vitamin D concentration on the development of early-onset type 2 diabetes in a large population sample. METHODS We conducted a case-cohort study utilising data from the Danish biobank and registers. Neonatal vitamin D was assessed measuring 25-hydroxyvitamin D3 [25(OH)D3] concentrations on the dried blood spot samples from the Biological Specimen Bank for Neonatal Screening. Cases of type 2 diabetes (n = 731) were retrieved from the Danish National Patient Register for all individuals born in Denmark between 1 May 1981 and 31 December 1992. The sub-cohort (n = 1765) was randomly selected from all children born in the same period. We used a weighted Cox proportional hazard model assessing the hazard of first type 2 diabetes diagnoses by quintiles of 25(OH)D3 and restricted cubic spline. RESULTS The median 25(OH)D3 concentration (IQR) among cases was 21.3 nmol/l (13.3-34.1) and 23.9 nmol/l (13.7-35.7) in the sub-cohort. There was no indication of a potential lower risk of early-onset type 2 diabetes among individuals in the higher quintile of vitamin D concentration compared with the lowest (HRcrude 0.97 [95% CI 0.71, 1.33] p = 0.85; HRadjusted 1.29 [95% CI 0.92, 1.83] p = 0.14). CONCLUSIONS/INTERPRETATION The results of this study do not support the hypothesis that higher neonatal vitamin D concentrations are associated with a lower risk of early-onset type 2 diabetes in adulthood.
Collapse
Affiliation(s)
- Amélie Keller
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark.
- Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark.
| | - Fanney Thorsteinsdottir
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
| | - Maria Stougaard
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Center for Early Interventions and Family Studies, Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Isabel Cardoso
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
| | - Peder Frederiksen
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
| | - Arieh S Cohen
- Statens Serum Institute, Clinical Mass Spectrometry, Copenhagen, Denmark
| | - Allan Vaag
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Ramune Jacobsen
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Berit L Heitmann
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- The Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, University of Sydney, Sydney, Australia
- Department of Public Health, Section for General Practice, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Boltjes R, Knippenberg S, Gerlach O, Hupperts R, Damoiseaux J. Vitamin D supplementation in multiple sclerosis: an expert opinion based on the review of current evidence. Expert Rev Neurother 2021; 21:715-725. [PMID: 34058936 DOI: 10.1080/14737175.2021.1935878] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/25/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Vitamin D has long been known for its immune-modulating effects, next to its function in calcium metabolism. As a consequence, poor vitamin D status has been associated with many diseases including multiple sclerosis (MS). Epidemiological studies suggest an association between a poor vitamin D status and development of MS and a poor vitamin D status is associated with more relapses and faster progression after patients are diagnosed with MS. AREA’S COVERED The aim of the authors was to review the role of vitamin D supplementation in the treatment of MS. Pubmed was used to review literature with a focus of vitamin D supplementation trials and meta-analyses in MS. EXPERT OPINION There is no solid evidence to support the application of vitamin D therapy, based on current available supplementation trials, although there are some promising results in the clinically isolated syndrome (CIS) patients and young MS patients early after initial diagnosis. The authors recommend further larger clinical trials with selected patient groups, preferable CIS patients and young patients at the time of diagnosis, using vitamin D3 supplements to reach a 100 nmol/l level, to further investigate the effects of vitamin D supplementation in MS.
Collapse
Affiliation(s)
- Robin Boltjes
- Academic MS Center Limburg, Department of Neurology, Zuyderland Medical Center, Sittard, The Netherlands
| | - Stephanie Knippenberg
- Academic MS Center Limburg, Department of Neurology, Zuyderland Medical Center, Sittard, The Netherlands
| | - Oliver Gerlach
- Academic MS Center Limburg, Department of Neurology, Zuyderland Medical Center, Sittard, The Netherlands
- School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Raymond Hupperts
- Academic MS Center Limburg, Department of Neurology, Zuyderland Medical Center, Sittard, The Netherlands
- School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jan Damoiseaux
- School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
34
|
Badihian N, Riahi R, Goli P, Badihian S, Poursafa P, Kelishadi R. Prenatal and perinatal factors associated with developing multiple sclerosis later in life: A systematic review and meta-analysis. Autoimmun Rev 2021; 20:102823. [PMID: 33866064 DOI: 10.1016/j.autrev.2021.102823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/06/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Both genetic and environmental factors play roles in Multiple Sclerosis (MS) etiopathogenesis. The relationship between prenatal/perinatal factors/exposures and future MS occurrence in the offspring remains controversial. Here, we aimed to review the available evidence on prenatal/perinatal factors associated with later MS occurrence. METHOD We performed systematic search of PubMed, Web of Science, and Scopus from inception to October 2020. We included original observational studies conducted on human participants addressing the association between prenatal/perinatal factors and MS occurrence. Data were extracted according to the PRISMA guideline. The adjusted odds ratio (OR) with 95% confidence interval (CI) was considered as the desired effect size. The heterogeneity was evaluated by Cochran's Q and I2 and the publication bias was assessed. We excluded gestational/neonatal vitamin D level, season of birth, and latitude because of recently published systematic reviews/meta-analyses on these subjects. RESULTS Overall, 2306 records were identified in the primary search. After excluding irrelevant studies, we evaluated 34 studies with contributing data on 100 prenatal/perinatal factors associated with an increased or decreased risk of MS occurrence. In the meta-analyses, we found no statistically significant associations between later MS occurrence in offspring and prenatal smoking exposure (OR = 1.01, 95% CI = 0.77-1.34), mode of delivery (OR = 0.90, 95% CI = 0.52-1.56), birth order (OR = 0.85, 95% CI = 0.72-1.00), and maternal age (OR = 1.34, 95% CI = 0.88-2.04). Paternal age and parents' marital status at the time of childbirth, maternal preeclampsia/ toxemia, forceps use, birth weight, plurality, and preterm birth were the other most studied factors, and none reported to affect MS risk. CONCLUSION We found that prenatal smoking exposure, mode of delivery, birth order, and maternal age do not affect risk of future MS development. Moreover, most of the other investigated factors were reported not to affect MS risk in the offspring.
Collapse
Affiliation(s)
- Negin Badihian
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Riahi
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Goli
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shervin Badihian
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Parnian Poursafa
- Department of Cell and Molecular Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Roya Kelishadi
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
35
|
Lincoln MR, Schneider R, Oh J. Vitamin D as disease-modifying therapy for multiple sclerosis? Expert Rev Clin Immunol 2021; 17:691-693. [PMID: 33836645 DOI: 10.1080/1744666x.2021.1915772] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Matthew R Lincoln
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Raphael Schneider
- Division of Neurology, University of Toronto, Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, Ontario, Canada
| | - Jiwon Oh
- Division of Neurology, University of Toronto, Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, Ontario, Canada
| |
Collapse
|
36
|
Sabel CE, Pearson JF, Mason DF, Willoughby E, Abernethy DA, Taylor BV. The latitude gradient for multiple sclerosis prevalence is established in the early lifecourse. Brain 2021; 144:2038-2046. [PMID: 33704407 DOI: 10.1093/brain/awab104] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/14/2020] [Accepted: 12/30/2020] [Indexed: 11/12/2022] Open
Abstract
The strongest epidemiological clue that the environment at the population level has a significant impact on the risk of developing multiple sclerosis (MS) is the well-established, and in many instances, increasing latitudinal gradient of prevalence, incidence and mortality globally, with prevalence increasing by up to 10-fold between the equator and 60 degrees North and South. The drivers of this gradient are thought to be environmental with latitude seen as a proxy for ultraviolet radiation and thus vitamin D production, however other factors may also play a role. However several important questions remain unanswered, particularly when in the life course is the gradient established, does lifetime migration mitigate or exacerbate previously reported latitude gradients at location of diagnosis, and do factors such as sex or MS disease phenotype influence the timing or significance of the gradient? Utilising life time residence calendars collected as part of the New Zealand national MS prevalence study, we constructed lifetime latitudinal gradients for MS from birth to prevalence day 2006 taking into account migration internally and externally and then analysed by sex and MS clinical course phenotype. 2127 of 2917 people living in NZ on prevalence day 7 March 2006 with MS completed the life course questionnaire and of these 1587 were born in NZ. All cohorts and sub cohorts were representative of the overall MS population in NZ on prevalence day. We found that the prevalence gradient was present at birth and was in fact stronger than at census day, and the slope of the gradient persisted until the age of 12 before gradually declining. We found that internal and external migration into NZ had little if any effect on the gradient except to decrease the significance of the gradient somewhat. Finally, we found as we had reported previously that the lifetime prevalence gradients were largely driven by females with relapse onset MS. These findings confirm for the first time the importance of early life environmental exposures in the risk of MS indicating strongly that exposures as early as in utero and at birth drive the latitudinal gradient. Consequently, prevention studies should be focussed on high risk individuals and populations from the earliest possible time points especially, when appropriate, on females.
Collapse
Affiliation(s)
- Clive E Sabel
- BERTHA, Big Data Centre for Environment and Health, Department of Environmental Science, Aarhus University, Denmark
| | - John F Pearson
- Biostatistics and Computational Biology Unit, University of Otago, Christchurch, New Zealand
| | | | | | | | - Bruce V Taylor
- Menzies Institute for Medical Research, University of Tasmania, Australia
| |
Collapse
|
37
|
Ames BN, Grant WB, Willett WC. Does the High Prevalence of Vitamin D Deficiency in African Americans Contribute to Health Disparities? Nutrients 2021; 13:499. [PMID: 33546262 PMCID: PMC7913332 DOI: 10.3390/nu13020499] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/24/2021] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
African Americans have higher incidence of, and mortality from, many health-related problems than European Americans. They also have a 15 to 20-fold higher prevalence of severe vitamin D deficiency. Here we summarize evidence that: (i) this health disparity is partly due to insufficient vitamin D production, caused by melanin in the skin blocking the UVB solar radiation necessary for its synthesis; (ii) the vitamin D insufficiency is exacerbated at high latitudes because of the combination of dark skin color with lower UVB radiation levels; and (iii) the health of individuals with dark skin can be markedly improved by correcting deficiency and achieving an optimal vitamin D status, as could be obtained by supplementation and/or fortification. Moderate-to-strong evidence exists that high 25-hydroxyvitamin D levels and/or vitamin D supplementation reduces risk for many adverse health outcomes including all-cause mortality rate, adverse pregnancy and birth outcomes, cancer, diabetes mellitus, Alzheimer's disease and dementia, multiple sclerosis, acute respiratory tract infections, COVID-19, asthma exacerbations, rickets, and osteomalacia. We suggest that people with low vitamin D status, which would include most people with dark skin living at high latitudes, along with their health care provider, consider taking vitamin D3 supplements to raise serum 25-hydroxyvitamin D levels to 30 ng/mL (75 nmol/L) or possibly higher.
Collapse
Affiliation(s)
- Bruce N. Ames
- Molecular and Cell Biology, Emeritus, University of California, Berkeley, CA 94720, USA;
| | - William B. Grant
- Sunlight, Nutrition and Health Research Center, San Francisco, CA 94164-1603, USA
| | - Walter C. Willett
- Departments of Nutrition and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
38
|
Magyari M, Joensen H, Laursen B, Koch-Henriksen N. The Danish Multiple Sclerosis Registry. Brain Behav 2021; 11:e01921. [PMID: 33128351 PMCID: PMC7821574 DOI: 10.1002/brb3.1921] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/23/2020] [Accepted: 10/14/2020] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES The Danish Multiple Sclerosis Registry is the oldest operative and nationwide MS registry. We present The Danish Multiple Sclerosis Registry with its history, data collection, scientific contribution, and national and international research collaboration. MATERIALS AND METHODS Detailed description of data collection, completeness, quality optimizing procedures, funding, and legal, ethical and data protection issues are provided. RESULTS The total number of registered cases with clinical isolated syndrome and multiple sclerosis since 1956 was by start of May 2020 30,023 of whom 16,515 cases were alive and residing in Denmark, giving a prevalence rate of about 284 per 100,000 population. The mean annual number of new cases receiving an MS diagnosis was 649 per year in the period 2010 to 2019. In total, 7,945 patients (48.1%) are receiving disease modifying therapy at the start of May 2020. CONCLUSIONS Multiple Sclerosis registers are becoming increasingly important, not only for epidemiological research but also by quantifying the burden of the disease for the patients and society and helping health care providers and regulators in their decisions. The Danish Multiple Sclerosis Registry has served as data source for a number of scientific publications including epidemiological studies on changes in incidence and mortality, cohort studies investigating risk factors for developing MS, comorbidities and socioeconomic outcomes in the MS population, and observational studies on effectiveness of disease modifying treatments outside the narrow realms of randomized clinical trials.
Collapse
Affiliation(s)
- Melinda Magyari
- Department of Neurology, The Danish Multiple Sclerosis Registry, Rigshospitalet, Glostrup, Denmark.,Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, Glostrup, Denmark
| | - Hanna Joensen
- Department of Neurology, The Danish Multiple Sclerosis Registry, Rigshospitalet, Glostrup, Denmark
| | - Bjarne Laursen
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Nils Koch-Henriksen
- Department of Neurology, The Danish Multiple Sclerosis Registry, Rigshospitalet, Glostrup, Denmark.,Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
39
|
Vickaryous N, Jitlal M, Jacobs BM, Middleton R, Chandran S, MacDougall NJJ, Giovannoni G, Dobson R. Remote testing of vitamin D levels across the UK MS population-A case control study. PLoS One 2020; 15:e0241459. [PMID: 33378408 PMCID: PMC7773187 DOI: 10.1371/journal.pone.0241459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/16/2020] [Indexed: 11/24/2022] Open
Abstract
Objective The association between vitamin D deficiency and multiple sclerosis (MS) is well described. We set out to use remote sampling to ascertain vitamin D status and vitamin D supplementation in a cross-sectional study of people with MS across the UK. Methods People with MS and matched controls were recruited from across the UK. 1768 people with MS enrolled in the study; remote sampling kits were distributed to a subgroup. Dried blood spots (DBS) were used to assess serum 25(OH)D in people with MS and controls. Results 1768 MS participants completed the questionnaire; 388 MS participants and 309 controls provided biological samples. Serum 25(OH)D was higher in MS than controls (median 71nmol/L vs 49nmol/L). A higher proportion of MS participants than controls supplemented (72% vs 26%, p<0.001); people with MS supplemented at higher vD doses than controls (median 1600 vs 600 IU/day, p<0.001). People with MS who did not supplement had lower serum 25(OH)D levels than non-supplementing controls (median 38 nmol/L vs 44 nmol/L). Participants engaged well with remote sampling. Conclusions The UK MS population have higher serum 25(OH)D than controls, mainly as a result of vitamin D supplementation. Remote sampling is a feasible way of carrying out large studies.
Collapse
Affiliation(s)
- Nicola Vickaryous
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University London, London, United Kingdom
| | - Mark Jitlal
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University London, London, United Kingdom
| | - Benjamin Meir Jacobs
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University London, London, United Kingdom
| | - Rod Middleton
- UKMS Register, Swansea University Medical School, Swansea, United Kingdom
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, UK Dementia Research Institute at Edinburgh, University of Edinburgh, Edinburgh, United Kingdom
| | - Niall John James MacDougall
- Neurology Department, Hairmyres Hospital, East Kilbride, United Kingdom
- Neurology Department, Institute of Neurological Sciences, Glasgow, United Kingdom
| | - Gavin Giovannoni
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University London, London, United Kingdom
- Blizard Institute, Queen Mary University London, London, United Kingdom
- Department of Neurology, Royal London Hospital, BartsHealth NHS Trust, London, United Kingdom
| | - Ruth Dobson
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University London, London, United Kingdom
- Department of Neurology, Royal London Hospital, BartsHealth NHS Trust, London, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Scazzone C, Agnello L, Bivona G, Lo Sasso B, Ciaccio M. Vitamin D and Genetic Susceptibility to Multiple Sclerosis. Biochem Genet 2020; 59:1-30. [PMID: 33159645 DOI: 10.1007/s10528-020-10010-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 10/22/2020] [Indexed: 12/28/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease affecting the central nervous system (CNS), resulting from the interaction among genetic, epigenetic, and environmental factors. Vitamin D is a secosteroid, and its circulating levels are influenced by environment and genetics. In the last decades, research data on the association between MS and vitamin D status led to hypothesize a possible role for hypovitaminosis D as a risk factor for MS. Some gene variants encoding proteins involved in vitamin D metabolism, transport, and function, which are responsible for vitamin D status alterations, have been related to MS susceptibility. This review explores the current literature on the influence of vitamin D-related genes in MS susceptibility, reporting all single-nucleotide polymorphisms (SNPs) investigated to date in 12 vitamin D pathway genes. Among all, the gene codifying vitamin D receptor (VDR) is the most studied. The association between VDR SNPs and MS risk has been reported by many Authors, with a few studies producing opposite results. Other vitamin D-related genes (including DHCR7/NADSYN1, CYP2R1, CYP27A1, CYP3A4, CYP27B1, CYP24A1, Megalin-DAB2-Cubilin, FGF-23, and Klotho) have been less investigated and achieved more conflicting evidence. Taken together, findings from the studies reviewed cannot clarify whether and to what extent vitamin D-related gene variants can influence MS risk.
Collapse
Affiliation(s)
- Concetta Scazzone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, Via del Vespro, 129, CAP 90127, Palermo, Sicily, Italy
| | - Luisa Agnello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, Via del Vespro, 129, CAP 90127, Palermo, Sicily, Italy
| | - Giulia Bivona
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, Via del Vespro, 129, CAP 90127, Palermo, Sicily, Italy
| | - Bruna Lo Sasso
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, Via del Vespro, 129, CAP 90127, Palermo, Sicily, Italy
- Department of Laboratory Medicine, University-Hospital, Palermo, Italy
| | - Marcello Ciaccio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, Via del Vespro, 129, CAP 90127, Palermo, Sicily, Italy.
- Department of Laboratory Medicine, University-Hospital, Palermo, Italy.
| |
Collapse
|
41
|
Pape K, Steffen F, Zipp F, Bittner S. Supplementary medication in multiple sclerosis: Real-world experience and potential interference with neurofilament light chain measurement. Mult Scler J Exp Transl Clin 2020; 6:2055217320936318. [PMID: 32922829 PMCID: PMC7457677 DOI: 10.1177/2055217320936318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/27/2020] [Accepted: 05/30/2020] [Indexed: 01/29/2023] Open
Abstract
Background As vitamins and dietary supplements are obtainable without prescription,
treating physicians often ignore their intake by patients with multiple
sclerosis (MS) and may therefore miss potential adverse effects and
interactions. Objective We aimed to assess the spectrum and intake frequency of supplementary
medication in a cohort of MS patients and to analyse the effect of biotin
intake on measurement of serum neurofilament light chain (sNfL), an emerging
marker of disease activity. Methods MS patients visiting our neurology outpatient clinic completed a
questionnaire on their past or present use of vitamins or dietary
supplements. In addition, the impact of two different doses of biotin (10
and 300 mg/day) on sNfL was studied in healthy volunteers. Results Of 186 patients, 72.6% reported taking over-the-counter vitamins or dietary
supplements currently or previously. Most frequently used was vitamin D
(60.0%), followed by biotin. Female patients and patients with primary
progressive MS tended to use supplements more frequently. Biotin intake did
not interfere with sNfL measurement by single molecule array (Simoa). Conclusions The use of vitamins and dietary supplements is frequent among patients with
MS. Thus, treating physicians should be aware of the pitfalls of
supplementary treatment and educate their patients accordingly.
Collapse
Affiliation(s)
- Katrin Pape
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University, Germany
| | - Falk Steffen
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University, Germany
| |
Collapse
|
42
|
Kowalówka M, Główka AK, Karaźniewicz-Łada M, Kosewski G. Clinical Significance of Analysis of Vitamin D Status in Various Diseases. Nutrients 2020; 12:E2788. [PMID: 32933052 PMCID: PMC7551674 DOI: 10.3390/nu12092788] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022] Open
Abstract
Vitamin D plays a role not only in the proper functioning of the skeletal system and the calcium-phosphate equilibrium, but also in the immune system, the cardiovascular system and the growth and division of cells. Although numerous studies have reported on the analysis of vitamin D status in various groups of patients, the clinical significance of measurements of vitamin D forms and metabolites remains ambiguous. This article reviews the reports analyzing the status of vitamin D in various chronic states. Particular attention is given to factors affecting measurement of vitamin D forms and metabolites. Relevant papers published during recent years were identified by an extensive PubMed search using appropriate keywords. Measurement of vitamin D status proved to be a useful tool in diagnosis and progression of metabolic syndrome, neurological disorders and cancer. High performance liquid chromatography coupled with tandem mass spectrometry has become the preferred method for analyzing the various forms and metabolites of vitamin D in biological fluids. Factors influencing vitamin D concentration, including socio-demographic and biochemical factors as well as the genetic polymorphism of the vitamin D receptor, along with vitamin D transporters and enzymes participating in vitamin D metabolism should be considered as potential confounders of the interpretation of plasma total 25(OH)D concentrations.
Collapse
Affiliation(s)
- Magdalena Kowalówka
- Department of Bromatology, Poznan University of Medical Sciences, 42 Marcelińska Street, 60-354 Poznań, Poland; (M.K.); (A.K.G.); (G.K.)
| | - Anna K. Główka
- Department of Bromatology, Poznan University of Medical Sciences, 42 Marcelińska Street, 60-354 Poznań, Poland; (M.K.); (A.K.G.); (G.K.)
| | - Marta Karaźniewicz-Łada
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 6 Święcickiego Street, 60-781 Poznań, Poland
| | - Grzegorz Kosewski
- Department of Bromatology, Poznan University of Medical Sciences, 42 Marcelińska Street, 60-354 Poznań, Poland; (M.K.); (A.K.G.); (G.K.)
| |
Collapse
|
43
|
Yeh WZ, Gresle M, Jokubaitis V, Stankovich J, van der Walt A, Butzkueven H. Immunoregulatory effects and therapeutic potential of vitamin D in multiple sclerosis. Br J Pharmacol 2020; 177:4113-4133. [PMID: 32668009 DOI: 10.1111/bph.15201] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 12/19/2022] Open
Abstract
Initially recognised as an important factor for bone health, vitamin D is now known to have a range of effects on the immune system. Vitamin D deficiency is associated with an increased risk of multiple sclerosis (MS), a chronic immune-mediated demyelinating disease of the CNS. In this review, we explore the links between vitamin D deficiency, MS risk, and disease activity. We also discuss the known immune effects of vitamin D supplementation and the relevance of these observations to the immunopathology of MS. Finally, we review the existing evidence for vitamin D supplementation as an MS therapy, highlighting several recent clinical studies and trials.
Collapse
Affiliation(s)
- Wei Zhen Yeh
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Melissa Gresle
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Jim Stankovich
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Anneke van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
44
|
An atlas on risk factors for multiple sclerosis: a Mendelian randomization study. J Neurol 2020; 268:114-124. [PMID: 32728946 PMCID: PMC7815542 DOI: 10.1007/s00415-020-10119-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 11/08/2022]
Abstract
Objectives We conducted a systematic review and wide-angled Mendelian randomization (MR) study to examine the association between possible risk factors and multiple sclerosis (MS). Methods We used MR analysis to assess the associations between 65 possible risk factors and MS using data from a genome-wide association study including 14 498 cases and 24 091 controls of European ancestry. For 18 exposures not suitable for MR analysis, we conducted a systematic review to obtain the latest meta-analyses evidence on their associations with MS. Results Childhood and adulthood body mass index were positively associated with MS, whereas physical activity and serum 25-hydroxyvitamin D were inversely associated with MS. There was evidence of possible associations of type 2 diabetes, waist circumference, body fat percentage, age of puberty and high-density lipoprotein cholesterol. Data of systematic review showed that exposure to organic solvents, Epstein Barr virus and cytomegalovirus virus infection, and diphtheria and tetanus vaccination were associated with MS risk. Conclusions This study identified several modifiable risk factors for primary prevention of MS that should inform public health policy. Electronic supplementary material The online version of this article (10.1007/s00415-020-10119-8) contains supplementary material, which is available to authorized users.
Collapse
|
45
|
Alfredsson L, Armstrong BK, Butterfield DA, Chowdhury R, de Gruijl FR, Feelisch M, Garland CF, Hart PH, Hoel DG, Jacobsen R, Lindqvist PG, Llewellyn DJ, Tiemeier H, Weller RB, Young AR. Insufficient Sun Exposure Has Become a Real Public Health Problem. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E5014. [PMID: 32668607 PMCID: PMC7400257 DOI: 10.3390/ijerph17145014] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/01/2020] [Accepted: 07/04/2020] [Indexed: 12/13/2022]
Abstract
This article aims to alert the medical community and public health authorities to accumulating evidence on health benefits from sun exposure, which suggests that insufficient sun exposure is a significant public health problem. Studies in the past decade indicate that insufficient sun exposure may be responsible for 340,000 deaths in the United States and 480,000 deaths in Europe per year, and an increased incidence of breast cancer, colorectal cancer, hypertension, cardiovascular disease, metabolic syndrome, multiple sclerosis, Alzheimer's disease, autism, asthma, type 1 diabetes and myopia. Vitamin D has long been considered the principal mediator of beneficial effects of sun exposure. However, oral vitamin D supplementation has not been convincingly shown to prevent the above conditions; thus, serum 25(OH)D as an indicator of vitamin D status may be a proxy for and not a mediator of beneficial effects of sun exposure. New candidate mechanisms include the release of nitric oxide from the skin and direct effects of ultraviolet radiation (UVR) on peripheral blood cells. Collectively, this evidence indicates it would be wise for people living outside the tropics to ensure they expose their skin sufficiently to the sun. To minimize the harms of excessive sun exposure, great care must be taken to avoid sunburn, and sun exposure during high ambient UVR seasons should be obtained incrementally at not more than 5-30 min a day (depending on skin type and UV index), in season-appropriate clothing and with eyes closed or protected by sunglasses that filter UVR.
Collapse
Affiliation(s)
- Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institute, 171 77 Stockholm, Sweden;
| | - Bruce K. Armstrong
- School of Population and Global Health, The University of Western Australia, Perth 6009, Australia;
| | - D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA;
| | - Rajiv Chowdhury
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK;
| | - Frank R. de Gruijl
- Department of Dermatology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands;
| | - Martin Feelisch
- Clinical & Experimental Sciences, University of Southampton Medical School and University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK;
| | - Cedric F. Garland
- Division of Epidemiology, Department of Family Medicine and Public Health, University of California San Diego School of Medicine, La Jolla, CA 92093, USA;
| | - Prue H. Hart
- Telethon Kids Institute, University of Western Australia, Perth 6872, Australia;
| | - David G. Hoel
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ramune Jacobsen
- Department of Pharmacy, University of Copenhagen, DK-2100 Copenhagen, Denmark;
| | - Pelle G. Lindqvist
- Department of Clinical Science and Education, Karolinska Institute, 171 77 Stockholm, Sweden;
| | - David J. Llewellyn
- College of Medicine and Health, University of Exeter Medical School, Exeter EX1 2LU, UK;
| | - Henning Tiemeier
- Department of Social and Behavioral Science, Harvard T.H. Chan School of Public Health, Harvard University, Cambridge, MA 02115, USA;
| | - Richard B. Weller
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4SB, UK;
| | - Antony R. Young
- St John’s Institute of Dermatology, King’s College London, London SE1 9RT, UK;
| |
Collapse
|
46
|
Dietary influence on central nervous system myelin production, injury, and regeneration. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165779. [DOI: 10.1016/j.bbadis.2020.165779] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/19/2020] [Accepted: 03/22/2020] [Indexed: 02/07/2023]
|
47
|
Graves JS, Barcellos LF, Krupp L, Belman A, Shao X, Quach H, Hart J, Chitnis T, Weinstock-Guttman B, Aaen G, Benson L, Gorman M, Greenberg B, Lotze T, Soe M, Ness J, Rodriguez M, Rose J, Schreiner T, Tillema JM, Waldman A, Casper TC, Waubant E. Vitamin D genes influence MS relapses in children. Mult Scler 2020; 26:894-901. [PMID: 31081484 PMCID: PMC6851448 DOI: 10.1177/1352458519845842] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The aim of this study was to determine whether a vitamin D genetic risk score (vitDGRS) is associated with 25-hydroxyvitamin D (25(OH)D) level and multiple sclerosis (MS) relapses in children. METHODS DNA samples were typed for single nucleotide polymorphisms (SNPs) from four genes previously identified to be associated with 25(OH)D levels. SNPs with strong associations with 25(OH)D after multiple comparison correction were used to create a genetic risk score (vitDGRS). Cox regression models tested associations of vitDGRS with relapse hazard. RESULTS Two independent SNPs within or near GC and NADSYN1/DHCR7 genes were strongly associated with 25(OH)D levels in the discovery cohort (n = 182) after Bonferroni correction. The vitDGRS of these SNPs explained 4.5% of the variance of 25(OH)D level after adjustment for genetic ancestry. Having the highest versus lowest vitDGRS was associated with 11 ng/mL lower 25(OH)D level (95% confidence interval (CI) = -17.5, -4.5, p = 0.001) in the discovery cohort. Adjusting for ancestry, sex, disease-modifying therapy (DMT), and HLA-DRB1*15 carrier status, the highest versus lowest vitDGRS was associated with 2.6-fold (95% CI = 1.37, 5.03, p = 0.004) and 2.0-fold (95% CI = 0.75, 5.20, p = 0.16) higher relapse hazard in the discovery and replication cohorts, respectively. CONCLUSION The vitDGRS identifies children at greater risk of relapse. These findings support a causal role for vitamin D in MS course.
Collapse
Affiliation(s)
- Jennifer S Graves
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Lisa F Barcellos
- School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Lauren Krupp
- Pediatric Multiple Sclerosis Center, New York University Langone Medical Center, New York, NY, USA
| | | | - Xiaorong Shao
- School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Hong Quach
- School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Janace Hart
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Tanuja Chitnis
- Partners Pediatric Multiple Sclerosis Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | | | - Gregory Aaen
- Pediatric Multiple Sclerosis Center, Loma Linda University Children's Hospital, San Bernardino, CA, USA
| | - Leslie Benson
- Pediatric Multiple Sclerosis and Related Disorders Program, Boston Children's Hospital, Boston, MA, USA
| | - Mark Gorman
- Pediatric Multiple Sclerosis and Related Disorders Program, Boston Children's Hospital, Boston, MA, USA
| | | | - Timothy Lotze
- The Blue Bird Circle Clinic for Multiple Sclerosis, Texas Children's Hospital, Houston, TX, USA
| | - Mar Soe
- Pediatric MS & Demyelinating Disease Center, Washington University, St. Louis, MI, USA
| | - Jayne Ness
- Center for Pediatric-Onset Demyelinating Disease, Children's of Alabama, Birmingham, AL, USA
| | - Moses Rodriguez
- Pediatric Multiple Sclerosis Center, Mayo Clinic, Rochester, MA, USA
| | - John Rose
- Department of Neurology, The University of Utah, Salt Lake City, UT, USA
| | - Teri Schreiner
- Rocky Mountain MS Center, University of Colorado, Denver, Denver, CO, USA
| | | | - Amy Waldman
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - T Charles Casper
- Department of Pediatrics, The University of Utah, Salt Lake City, UT, USA
| | - Emmanuelle Waubant
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
48
|
Häusler D, Torke S, Peelen E, Bertsch T, Djukic M, Nau R, Larochelle C, Zamvil SS, Brück W, Weber MS. High dose vitamin D exacerbates central nervous system autoimmunity by raising T-cell excitatory calcium. Brain 2020; 142:2737-2755. [PMID: 31302671 DOI: 10.1093/brain/awz190] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 04/08/2019] [Accepted: 05/03/2019] [Indexed: 01/12/2023] Open
Abstract
Poor vitamin D status is associated with a higher relapse rate and earlier disability in multiple sclerosis. Based on these associations, patients with multiple sclerosis are frequently supplemented with the vitamin D precursor cholecalciferol, although it is unclear whether this regimen is of therapeutic benefit. To model consequences of this common practice, mice were fed for more than 3 months with a low, medium or high dose of cholecalciferol, representative of vitamin D deficiency, modest and disproportionally high supplementation, respectively, in patients with multiple sclerosis. Compared to vitamin D-deprived mice, its moderate supplementation reduced the severity of subsequent experimental autoimmune encephalomyelitis, which was associated with an expansion of regulatory T cells. Direct exposure of murine or human T cells to vitamin D metabolites inhibited their activation. In contrast, mice with 25-(OH) vitamin D levels above 200 nmol/l developed fulminant experimental autoimmune encephalomyelitis with massive CNS infiltration of activated myeloid cells, Th1 and Th17 cells. When dissecting this unexpected outcome, we observed that high, but not medium dose vitamin D had caused mild hypercalcaemia, which rendered T cells more prone to pro-inflammatory activation. Exposing murine or human T cells to equivalent calcium concentrations in vitro enhanced its influx, triggering activation, upregulation of pro-inflammatory gene products and enhanced transmigration across a blood-brain barrier model. These findings suggest that vitamin D at moderate levels may exert a direct regulatory effect, while continuous high dose vitamin D treatment could trigger multiple sclerosis disease activity by raising mean levels of T-cell excitatory calcium.
Collapse
Affiliation(s)
- Darius Häusler
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Sebastian Torke
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Evelyn Peelen
- Department of Neurosciences, Centre de recherche de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Thomas Bertsch
- Institute of Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, General Hospital Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Marija Djukic
- Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| | - Roland Nau
- Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, Göttingen, Germany
| | - Catherine Larochelle
- Department of Neurosciences, Centre de recherche de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Scott S Zamvil
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Martin S Weber
- Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Department of Neurology, University Medical Center, Göttingen, Germany
| |
Collapse
|
49
|
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. We review the two core MS features, myelin instability, fragmentation, and remyelination failure, and dominance of pathogenic CD4+ Th17 cells over protective CD4+ Treg cells. To better understand myelin pathology, we describe myelin biosynthesis, structure, and function, then highlight stearoyl-CoA desaturase (SCD) in nervonic acid biosynthesis and nervonic acid's contribution to myelin stability. Noting that vitamin D deficiency decreases SCD in the periphery, we propose it also decreases SCD in oligodendrocytes, disrupting the nervonic acid supply and causing myelin instability and fragmentation. To better understand the distorted Th17/Treg cell balance, we summarize Th17 cell contributions to MS pathogenesis, then highlight how 1,25-dihydroxyvitamin D3 signaling from microglia to CD4+ T cells restores Treg cell dominance. This signaling rapidly increases flux through the methionine cycle, removing homocysteine, replenishing S-adenosyl-methionine, and improving epigenetic marking. Noting that DNA hypomethylation and inappropriate DRB1*1501 expression were observed in MS patient CD4+ T cells, we propose that vitamin D deficiency thwarts epigenetic downregulation of DRB1*1501 and Th17 cell signature genes, and upregulation of Treg cell signature genes, causing dysregulation within the CD4+ T cell compartment. We explain how obesity reduces vitamin D status, and how estrogen and vitamin D collaborate to promote Treg cell dominance in females. Finally, we discuss the implications of this new knowledge concerning myelin and the Th17/Treg cell balance, and advocate for efforts to address the global epidemics of obesity and vitamin D deficiency in the expectation of reducing the impact of MS.
Collapse
Affiliation(s)
- Colleen E. Hayes
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - James M. Ntambi
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, 1415 Linden Drive, Madison, WI 53706, USA
| |
Collapse
|
50
|
Vitamin D's Effect on Immune Function. Nutrients 2020; 12:nu12051248. [PMID: 32353972 PMCID: PMC7281985 DOI: 10.3390/nu12051248] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/24/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023] Open
Abstract
Ever since its discovery by Windhaus, the importance of the active metabolite of vitamin D (1,25-dihydroxyvitamin D3; 1,25-(OH)2D3) has been ever expanding. In this review, the attention is shifted towards the importance of the extra-skeletal effects of vitamin D, with special emphasis on the immune system. The first hint of the significant role of vitamin D on the immune system was made by the discovery of the presence of the vitamin D receptor on almost all cells of the immune system. In vitro, the overwhelming effect of supra-physiological doses of vitamin D on the individual components of the immune system is very clear. Despite these promising pre-clinical results, the translation of the in vitro observations to solid clinical effects has mostly failed. Nevertheless, the evidence of a link between vitamin D deficiency and adverse outcomes is overwhelming and clearly points towards avoidance of vitamin D deficiency especially in early life.
Collapse
|