1
|
Wills O, Wright B, Greenwood LM, Solowij N, Schira M, Maller JJ, Gupta A, Magnussen J, Probst Y. Lifestyle management and brain MRI metrics in female Australian adults living with multiple sclerosis: a feasibility and acceptability study. Pilot Feasibility Stud 2024; 10:71. [PMID: 38698454 PMCID: PMC11064336 DOI: 10.1186/s40814-024-01495-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 04/14/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Limited studies of multiple sclerosis (MS) exist whereby magnetic resonance imaging (MRI) of the brain with consistent imaging protocols occurs at the same time points as collection of healthy lifestyle measures. The aim of this study was to test the feasibility, acceptability and preliminary efficacy of acquiring MRI data as an objective, diagnostic and prognostic marker of MS, at the same time point as brain-healthy lifestyle measures including diet. METHODS Participants living with relapsing remitting MS partook in one structural MRI scanning session of the brain, completed two online 24-hour dietary recalls and demographic and self-reported lifestyle questionnaires (e.g. self-reported disability, comorbidities, physical activity, smoking status, body mass index (BMI), stress). Measures of central tenancy and level of dispersion were calculated for feasibility and acceptability of the research protocols. Lesion count was determined by one radiologist and volumetric analyses by a data analysis pipeline based on FreeSurfer software suite. Correlations between white matter lesion count, whole brain volume analyses and lifestyle measures were assessed using Spearman's rank-order correlation coefficient. RESULTS Thirteen female participants were included in the study: eligibility rate 90.6% (29/32), recruitment rate 46.9% (15/32) and compliance rate 87% (13/15). The mean time to complete all required tasks, including MRI acquisition was 115.86 minutes ( ± 23.04), over 4 days. Conversion to usual dietary intake was limited by the small sample. There was one strong, negative correlation between BMI and brain volume (rs = -0.643, p = 0.018) and one strong, positive correlation between physical activity and brain volume (rs = 0.670, p = 0.012) that were both statistically significant. CONCLUSIONS Acquiring MRI brain scans at the same time point as lifestyle profiles in adults with MS is both feasible and accepted among adult females living with MS. Quantification of volumetric MRI data support further investigations using semi-automated pipelines among people living with MS, with pre-processing steps identified to increase automated feasibility. This protocol may be used to determine relationships between elements of a brain-healthy lifestyle, including dietary intake, and measures of disease burden and brain health, as assessed by T1-weighted and T2-weighted lesion count and whole brain volume, in an adequately powered sample. TRIAL REGISTRATION The study protocol was retrospectively registered in the Australia New Zealand Clinical Trials Registry (ACTRN12624000296538).
Collapse
Affiliation(s)
- Olivia Wills
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, NSW, 2522, Australia.
| | - Brooklyn Wright
- School of Psychology, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Lisa-Marie Greenwood
- Illawara Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
- School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia
| | - Nadia Solowij
- School of Psychology, University of Wollongong, Wollongong, NSW, 2522, Australia
- Illawara Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Mark Schira
- School of Psychology, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Jerome J Maller
- General Electric Healthcare, Richmond, Melbourne, Australia
- Monash Alfred Psychiatry Research Centre, Melbourne, VIC, Australia
| | - Alok Gupta
- Illawara Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
- Wollongong Diagnostic Imaging Group, Wollongong, NSW, Australia
| | - John Magnussen
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie Park, NSW, 2109, Australia
| | - Yasmine Probst
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, NSW, 2522, Australia
- Illawara Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| |
Collapse
|
2
|
Zhang W, Joshi C, Smith C, Ujas TA, Rivas JR, Cowell L, Christley S, Stowe AM, Monson NL. Neuronal binding by antibodies can be influenced by low pH stress during the isolation procedure. J Immunol Methods 2023; 521:113535. [PMID: 37558123 PMCID: PMC11249026 DOI: 10.1016/j.jim.2023.113535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 07/18/2023] [Accepted: 08/05/2023] [Indexed: 08/11/2023]
Abstract
Low pH stress and its influence on antibody binding is a common consideration among chemists, but is only recently emerging as a consideration in Immunological studies. Antibody characterizations in Multiple Sclerosis (MS), an autoimmune disease of the Central Nervous System (CNS) has revealed that antibodies in the cerebrospinal fluid (CSF) of patients with Multiple Sclerosis bind to myelin-related and non-myelin antigen targets. Many laboratories have used molecular biology techniques to generate recombinant human antibodies (rhAbs) expressed by individual B cells from healthy donors and patients with systemic autoimmune disease to identify antigen targets. This approach has been adapted within the Neuroimmunology research community to investigate antigen targets of individual B cells in the CSF of MS patients. Our laboratory determines which antibodies to clone based on their immunogenetics and this method enriches for cloning of rhAbs that bind to neurons. However, newer technologies to assist in purification of these rhAbs from culture supernatants use an acidic elution buffer which may enhance low pH stress on the antibody structure. Our laboratory routinely uses a basic elution buffer to purify rhAbs from culture supernatants to avoid low pH stress to the antibody structure. Our goal was to investigate whether acidic elution of our rhAbs using Next Generation Chromatography would impact the rhAbs' ability to bind neurons. The limited data presented here for two neuron-binding rhAbs tested indicated that acidic elution buffers used during rhAb purification impacted the ability of rhAbs with low CDR3 charge to maintain binding to neuronal targets. Reproducibility in a larger panel of rhAbs and factors underlying these observations remain untested.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Chaitanya Joshi
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Chad Smith
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Thomas A Ujas
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Jacqueline R Rivas
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Lindsay Cowell
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Scott Christley
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Ann M Stowe
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America
| | - Nancy L Monson
- Department of Neurology, Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, United States of America.
| |
Collapse
|
3
|
Mohamadi Y, Borhani-Haghighi M. TGN020 application against aquaporin 4 improved multiple sclerosis by inhibiting astrocytes, microglia, and NLRP3 inflammasome in a cuprizone mouse model. J Chem Neuroanat 2023; 132:102306. [PMID: 37394105 DOI: 10.1016/j.jchemneu.2023.102306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/20/2023] [Accepted: 06/30/2023] [Indexed: 07/04/2023]
Abstract
In multiple sclerosis (MS), activation of the astrocytes and microglia induces a cascading inflammatory response. Overexpression of the aquaporin 4 (AQP4) in the glia is a trigger for this reaction. This study aimed to block AQP4 by injecting TGN020 to alleviate the symptoms of MS. Total of 30 male mice were randomly divided into control (intact), cuprizone model of MS (fed with 0.2% cuprizone for 35 days), and TGN020-treated (received daily intraperitoneal injections of 200 mg/kg TGN020 with cuprizone intake) groups. Astrogliosis, M1-M2 microglia polarization, NLRP3 inflammasome activation, and demyelination were investigated in the corpus callosum by immunohistochemistry, real-time PCR, western blot, and luxol fast blue staining. The Rotarod test was performed for a behavior assessment. AQP4 inhibition caused a significant decrease in the expression of the astrocyte-specific marker, GFAP. It also changed the microglia polarization from M1 to M2 indicated by a significant downregulation of iNOS, CD86, MHC-ІІ, and upregulation of arginase1, CD206, and TREM-2. In addition, western blot data showed a significant decrease in the NLRP3, caspase1, and IL-1b proteins in the treatment group, which indicated inflammasome inactivation. The molecular changes following the TGN020 injection resulted in remyelination and motor recovery enhancement in the treatment group. In conclusion, the results draw the attention to the role of AQP4 in the cuprizone model of MS.
Collapse
Affiliation(s)
- Yousef Mohamadi
- Department of Anatomy, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Gkika A, Androutsou ME, Aletras AJ, Tselios T. Competitive ELISA for the identification of 35-55 myelin oligodendrocyte glycoprotein immunodominant epitope conjugated with mannan. J Pept Sci 2023; 29:e3493. [PMID: 37041122 DOI: 10.1002/psc.3493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/02/2023] [Accepted: 04/05/2023] [Indexed: 04/13/2023]
Abstract
Analogs of immunodominant myelin peptides involved in multiple sclerosis (MS: the most common autoimmune disease) have been extensively used to modify the immune response over the progression of the disease. The immunodominant 35-55 epitope of myelin oligodendrocyte glycoprotein (MOG35-55 ) is an autoantigen appearing in MS and stimulates the encephalitogenic T cells, whereas mannan polysaccharide (Saccharomyces cerevisiae) is a carrier toward the mannose receptor of dendritic cells and macrophages. The conjugate of mannan-MOG35-55 has been extensively studied for the inhibition of chronic experimental autoimmune encephalomyelitis (EAE: an animal model of MS) by inducing antigen-specific immune tolerance against the clinical symptoms of EAE in mice. Moreover, it presents a promising approach for the immunotherapy of MS under clinical investigation. In this study, a competitive enzyme-linked immunosorbent assay (ELISA) was developed to detect the MOG35-55 peptide that is conjugated to mannan. Intra- and inter-day assay experiments proved that the proposed ELISA methodology is accurate and reliable and could be used in the following applications: (i) to identify the peptide (antigen) while it is conjugated to mannan and (ii) to adequately address the alterations that the MOG35-55 peptide may undergo when it is bound to mannan during production and stability studies.
Collapse
Affiliation(s)
- Areti Gkika
- Department of Chemistry, University of Patras, Rion Patras, Greece
| | | | | | - Theodore Tselios
- Department of Chemistry, University of Patras, Rion Patras, Greece
| |
Collapse
|
5
|
Eva L, Pleș H, Covache-Busuioc RA, Glavan LA, Bratu BG, Bordeianu A, Dumitrascu DI, Corlatescu AD, Ciurea AV. A Comprehensive Review on Neuroimmunology: Insights from Multiple Sclerosis to Future Therapeutic Developments. Biomedicines 2023; 11:2489. [PMID: 37760930 PMCID: PMC10526343 DOI: 10.3390/biomedicines11092489] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/02/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
This review delves into neuroimmunology, focusing on its relevance to multiple sclerosis (MS) and potential treatment advancements. Neuroimmunology explores the intricate relationship between the immune system and the central nervous system (CNS). Understanding these mechanisms is vital for grasping the pathophysiology of diseases like MS and for devising innovative treatments. This review introduces foundational neuroimmunology concepts, emphasizing the role of immune cells, cytokines, and blood-brain barrier in CNS stability. It highlights how their dysregulation can contribute to MS and discusses genetic and environmental factors influencing MS susceptibility. Cutting-edge research methods, from omics techniques to advanced imaging, have revolutionized our understanding of MS, offering valuable diagnostic and prognostic tools. This review also touches on the intriguing gut-brain axis, examining how gut microbiota impacts neuroimmunological processes and its potential therapeutic implications. Current MS treatments, from immunomodulatory drugs to disease-modifying therapies, are discussed alongside promising experimental approaches. The potential of personalized medicine, cell-based treatments, and gene therapy in MS management is also explored. In conclusion, this review underscores neuroimmunology's significance in MS research, suggesting that a deeper understanding could pave the way for more tailored and effective treatments for MS and similar conditions. Continued research and collaboration in neuroimmunology are essential for enhancing patient outcomes.
Collapse
Affiliation(s)
- Lucian Eva
- Clinical Emergency Hospital “Prof. Dr. Nicolae Oblu”, 700309 Iasi, Romania;
| | - Horia Pleș
- Department of Neurosurgery, Centre for Cognitive Research in Neuropsychiatric Pathology (NeuroPsy-Cog), “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (L.A.G.); (B.-G.B.); (A.B.); (D.-I.D.); (A.D.C.); (A.V.C.)
| | - Luca Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (L.A.G.); (B.-G.B.); (A.B.); (D.-I.D.); (A.D.C.); (A.V.C.)
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (L.A.G.); (B.-G.B.); (A.B.); (D.-I.D.); (A.D.C.); (A.V.C.)
| | - Andrei Bordeianu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (L.A.G.); (B.-G.B.); (A.B.); (D.-I.D.); (A.D.C.); (A.V.C.)
| | - David-Ioan Dumitrascu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (L.A.G.); (B.-G.B.); (A.B.); (D.-I.D.); (A.D.C.); (A.V.C.)
| | - Antonio Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (L.A.G.); (B.-G.B.); (A.B.); (D.-I.D.); (A.D.C.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (L.A.G.); (B.-G.B.); (A.B.); (D.-I.D.); (A.D.C.); (A.V.C.)
| |
Collapse
|
6
|
Morgan A, Tallantyre E, Ontaneda D. The benefits and risks of escalation versus early highly effective treatment in patients with multiple sclerosis. Expert Rev Neurother 2023; 23:433-444. [PMID: 37129299 DOI: 10.1080/14737175.2023.2208347] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
INTRODUCTION Multiple sclerosis is a chronic, demyelinating, inflammatory, and neurodegenerative disease of the central nervous system that affects over 2 million people worldwide. Considerable advances have been made in the availability of disease modifying therapies for relapsing-remitting multiple sclerosis since their introduction in the 1990s. This has led to debate regarding the optimal first-line treatment approach: a strategy of escalation versus early highly effective treatment. AREAS COVERED This review defines the strategies of escalation and early highly effective treatment, outlines the pros and cons of each, and provides an analysis of both the current literature and expected future directions of the field. EXPERT OPINION There is growing support for using early highly effective treatment as the initial therapeutic approach in relapsing-remitting multiple sclerosis. However, much of this support stems from observational real-world studies that use historic data and lack safety outcomes or randomized control trials that compare individual high versus low-moderate efficacy therapies, instead of the approaches themselves. Randomized control trials (DELIVER-MS, TREAT-MS) are needed to systemically and prospectively compare contemporary escalation versus early highly effective treatment approaches.
Collapse
Affiliation(s)
- Annalisa Morgan
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Emma Tallantyre
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
- Department of Neurology, University Hospital of Wales, Cardiff, UK
| | - Daniel Ontaneda
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
7
|
Kumar N, Sahoo NK, Mehan S, Verma B. The importance of gut-brain axis and use of probiotics as a treatment strategy for multiple sclerosis. Mult Scler Relat Disord 2023; 71:104547. [PMID: 36805171 DOI: 10.1016/j.msard.2023.104547] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/16/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
It has been shown that the dysbiosis of the gut's microbes substantially impacts CNS illnesses, including Alzheimer's, Parkinson's, autism, and autoimmune diseases like multiple sclerosis (MS). MS is a CNS-affected autoimmune demyelination condition. Through a two-way communication pathway known as the gut-brain axis, gut microbes communicate with the CNS. When there is a disruption in the gut microbiome, cytokines and other immune cells are secreted, which affects the BBB and gastrointestinal permeability. Recent research using animal models has revealed that the gut microbiota may greatly influence the pathophysiology of EAE/MS. Any change in the gut might increase inflammatory cytokinesand affect the quantity of SCFAs, and other metabolites that cause neuroinflammation and demyelination. In- vivo and in-vitro studies have concluded that probiotics affect the immune system and can be utilized to treat gastrointestinal dysbiosis. Any alteration in the gut microbial composition caused by probiotic intake may serve as a preventive and treatment strategy for MS. The major goal of this review is to emphasize an overview of recent research on the function of gut microbiota in the onset of MS and how probiotics have a substantial impact on gastrointestinal disruption in MS and other neuro disorders. It will be easier to develop new therapeutic approaches, particularly probiotic-based supplements, for treating multiple sclerosis (MS) if we know the link between the gut and CNS.
Collapse
Affiliation(s)
- Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India.
| | - Nalini Kanta Sahoo
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, (An Autonomous College), Moga, Punjab 142001, India
| | - Bharti Verma
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| |
Collapse
|
8
|
Shiraishi W, Miyata T, Matsuyoshi A, Yamada Y, Hatano T, Hashimoto T. [A case of multiple sclerosis with a tumefactive lesion during long-term treatment with fingolimod, leading to decompressive craniotomy]. Rinsho Shinkeigaku 2023; 63:37-44. [PMID: 36567105 DOI: 10.5692/clinicalneurol.cn-001806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
We report a 57-year-old man with multiple sclerosis since his 30s who was treated with fingolimod for 9 years. He developed left hemiparesis and consciousness disturbance. Brain MRI revealed a mass lesion in the right frontal lobe with gadolinium enhancement. Cerebrospinal fluid examination showed no pleocytosis. The lesion continued to expand after admission, and on the 9th day after admission, decompressive craniectomy and brain biopsy were performed. Brain pathology revealed demyelination in the lesion, leading to the diagnosis of a tumefactive demyelinating lesion. Corticosteroid therapy ameliorated the brain lesion, and we inducted natalizumab. Tumefactive demyelinating lesions requiring decompressive craniotomy are rare, and we report this case for the further accumulation of similar cases.
Collapse
Affiliation(s)
- Wataru Shiraishi
- Department of Neurology, Kokura Memorial Hospital
- Shiraishi Internal Medicine Clinic
| | | | | | - Yui Yamada
- Department of Pathology, Kokura Memorial Hospital
| | - Taketo Hatano
- Department of Neurosurgery, Kokura Memorial Hospital
| | | |
Collapse
|
9
|
Triantafyllakou I, Clemente N, Khetavat RK, Dianzani U, Tselios T. Development of PLGA Nanoparticles with a Glycosylated Myelin Oligodendrocyte Glycoprotein Epitope (MOG 35-55) against Experimental Autoimmune Encephalomyelitis (EAE). Mol Pharm 2022; 19:3795-3805. [PMID: 36098508 DOI: 10.1021/acs.molpharmaceut.2c00277] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multiple sclerosis (MS) is one of the most common neurodegenerative diseases in young adults, with early clinical symptoms seen in the central nervous system (CNS) myelin sheaths due to an attack caused by the patient's immune system. Activation of the immune system is mediated by the induction of an antigen-specific immune response involving the interaction of multiple T-cell types with antigen-presenting cells (APCs), such as dendritic cells (DCs). Antigen-specific therapeutic approaches focus on immune cells and autoantigens involved in the onset of disease symptoms, which are the main components of myelin proteins. The ability of such therapeutics to bind strongly to DCs could lead to immune system tolerance to the disease. Many modern approaches are based on peptide-based research, as, in recent years, they have been of particular interest in the development of new pharmaceuticals. The characteristics of peptides, such as short lifespan in the body and rapid hydrolysis, can be overcome by their entrapment in nanospheres, providing better pharmacokinetics and bioavailability. The present study describes the development of polymeric nanoparticles with encapsulated myelin peptide analogues involved in the development of MS, along with their biological evaluation as inhibitors of MS development and progression. In particular, particles of poly(lactic-co-glycolic) acid (PLGA) loaded with peptides based on mouse/rat (rMOG) epitope 35-55 of myelin oligodendrocyte glycoprotein (MOG) conjugated with saccharide residues were developed. More specifically, the MOG35-55 peptide was conjugated with glucosamine to promote the interaction with mannose receptors (MRs) expressed by DCs. In addition, a study of slow release (dissolution) and quantification on both initially encapsulated peptide and daily release in saline in vitro was performed, followed by an evaluation of in vivo activity of the formulation on mouse experimental autoimmune encephalomyelitis (EAE), an animal model of MS, using both prophylactic and therapeutic protocols. Our results showed that the therapeutic protocol was effective in reducing EAE clinical scores and inflammation of the central nervous system and could be an alternative and promising approach against MS inducing tolerance against the disease.
Collapse
Affiliation(s)
- Iro Triantafyllakou
- Department of Chemistry, University of Patras, 26504 Rion Patras, Greece.,Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Nausicaa Clemente
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Ravi Kumar Khetavat
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Theodore Tselios
- Department of Chemistry, University of Patras, 26504 Rion Patras, Greece
| |
Collapse
|
10
|
Emamnejad R, Dass M, Mahlis M, Bozkurt S, Ye S, Pagnin M, Theotokis P, Grigoriadis N, Petratos S. Thyroid hormone-dependent oligodendroglial cell lineage genomic and non-genomic signaling through integrin receptors. Front Pharmacol 2022; 13:934971. [PMID: 36133808 PMCID: PMC9483185 DOI: 10.3389/fphar.2022.934971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous autoimmune disease whereby the pathological sequelae evolve from oligodendrocytes (OLs) within the central nervous system and are targeted by the immune system, which causes widespread white matter pathology and results in neuronal dysfunction and neurological impairment. The progression of this disease is facilitated by a failure in remyelination following chronic demyelination. One mediator of remyelination is thyroid hormone (TH), whose reliance on monocarboxylate transporter 8 (MCT8) was recently defined. MCT8 facilitates the entry of THs into oligodendrocyte progenitor cell (OPC) and pre-myelinating oligodendrocytes (pre-OLs). Patients with MS may exhibit downregulated MCT8 near inflammatory lesions, which emphasizes an inhibition of TH signaling and subsequent downstream targeted pathways such as phosphoinositide 3-kinase (PI3K)-Akt. However, the role of the closely related mammalian target of rapamycin (mTOR) in pre-OLs during neuroinflammation may also be central to the remyelination process and is governed by various growth promoting signals. Recent research indicates that this may be reliant on TH-dependent signaling through β1-integrins. This review identifies genomic and non-genomic signaling that is regulated through mTOR in TH-responsive pre-OLs and mature OLs in mouse models of MS. This review critiques data that implicates non-genomic Akt and mTOR signaling in response to TH-dependent integrin receptor activation in pre-OLs. We have also examined whether this can drive remyelination in the context of neuroinflammation and associated sequelae. Importantly, we outline how novel therapeutic small molecules are being designed to target integrin receptors on oligodendroglial lineage cells and whether these are viable therapeutic options for future use in clinical trials for MS.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Michael Mahlis
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Salome Bozkurt
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Paschalis Theotokis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
- *Correspondence: Steven Petratos,
| |
Collapse
|
11
|
Židó M, Kačer D, Valeš K, Svobodová Z, Zimová D, Štětkárová I. Metabolomics of Cerebrospinal Fluid in Multiple Sclerosis Compared With Healthy Controls: A Pilot Study. Front Neurol 2022; 13:874121. [PMID: 35693010 PMCID: PMC9178205 DOI: 10.3389/fneur.2022.874121] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) leading to the loss of myelin and axons. Diagnosis is based on clinical findings, MRI, and analysis of cerebrospinal fluid (CSF). CSF is an ultrafiltrate of plasma and reflects inflammatory processes in the CNS. The aim of this study was to perform metabolomics analysis of CSF in patients after the first attack of MS and healthy controls and try to find new specific analytes for MS including those potentially predicting disease activities at the onset. Methods We collected CSF from 19 patients (16 females, aged 19–55 years) after the first attack of clinical symptoms who fulfilled revised McDonald criteria of MS and CSF of 19 controls (16 females, aged 19–50 years). Analyses of CSF samples were provided using the high-performance liquid chromatography system coupled with a mass spectrometer with a high-resolution detector (TripleTOF 5600, AB Sciex, Canada). Results Approximately 130 selected analytes were identified, and 30 of them were verified. During the targeted analysis, a significant decrease in arginine and histidine and a less significant decrease in the levels of asparagine, leucine/isoleucine, and tryptophan, together with a significant increase of palmitic acid in the patient group, were found. Conclusion We observed significant differences in amino and fatty acids in the CSF of newly diagnosed patients with MS in comparison with controls. The most significant changes were observed in levels of arginine, histidine, and palmitic acid that may predict inflammatory disease activity. Further studies are necessary to support these findings as potential biomarkers of MS.
Collapse
Affiliation(s)
- Michal Židó
- Department of Neurology, Third Faculty of Medicine, Charles University, Prague, Czechia
- Department of Neurology, Faculty Hospital Královské Vinohrady, Prague, Czechia
| | - David Kačer
- National Institute of Mental Health, Klecany, Czechia
| | - Karel Valeš
- National Institute of Mental Health, Klecany, Czechia
- Institute of Physiology, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia
| | - Zuzana Svobodová
- Department of Neurology, Faculty Hospital Královské Vinohrady, Prague, Czechia
| | - Denisa Zimová
- Department of Neurology, Faculty Hospital Královské Vinohrady, Prague, Czechia
| | - Ivana Štětkárová
- Department of Neurology, Third Faculty of Medicine, Charles University, Prague, Czechia
- Department of Neurology, Faculty Hospital Královské Vinohrady, Prague, Czechia
- *Correspondence: Ivana Štětkárová
| |
Collapse
|
12
|
Liu H, Chen B, Zhu Q. Potential application of hydrogel to the diagnosis and treatment of multiple sclerosis. J Biol Eng 2022; 16:10. [PMID: 35395765 PMCID: PMC8991948 DOI: 10.1186/s13036-022-00288-7] [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: 12/31/2021] [Accepted: 03/12/2022] [Indexed: 11/18/2022] Open
Abstract
Abstract Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system. This disorder may cause progressive and permanent impairment, placing significant physical and psychological strain on sufferers. Each progress in MS therapy marks a significant advancement in neurological research. Hydrogels can serve as a scaffold with high water content, high expansibility, and biocompatibility to improve MS cell proliferation in vitro and therapeutic drug delivery to cells in vivo. Hydrogels may also be utilized as biosensors to detect MS-related proteins. Recent research has employed hydrogels as an adjuvant imaging agent in immunohistochemistry assays. Following an overview of the development and use of hydrogels in MS diagnostic and therapy, this review discussed hydrogel’s advantages and future opportunities in the diagnosis and treatment of MS. Graphical abstract ![]()
Collapse
Affiliation(s)
- Haochuan Liu
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Xiantai Street No. 126, Changchun, TX, 130031, PR China
| | - Bing Chen
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Xiantai Street No. 126, Changchun, TX, 130031, PR China.
| | - Qingsan Zhu
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Xiantai Street No. 126, Changchun, TX, 130031, PR China.
| |
Collapse
|
13
|
McGill MM, Richman AR, Boyd JR, Sabikunnahar B, Lahue KG, Montgomery TL, Caldwell S, Varnum S, Frietze S, Krementsov DN. p38 MAP Kinase Signaling in Microglia Plays a Sex-Specific Protective Role in CNS Autoimmunity and Regulates Microglial Transcriptional States. Front Immunol 2021; 12:715311. [PMID: 34707603 PMCID: PMC8542909 DOI: 10.3389/fimmu.2021.715311] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/17/2021] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system, representing the leading cause of non-traumatic neurologic disease in young adults. This disease is three times more common in women, yet more severe in men, but the mechanisms underlying these sex differences remain largely unknown. MS is initiated by autoreactive T helper cells, but CNS-resident and CNS-infiltrating myeloid cells are the key proximal effector cells regulating disease pathology. We have previously shown that genetic ablation of p38α MAP kinase broadly in the myeloid lineage is protective in the autoimmune model of MS, experimental autoimmune encephalomyelitis (EAE), but only in females, and not males. To precisely define the mechanisms responsible, we used multiple genetic approaches and bone marrow chimeras to ablate p38α in microglial cells, peripheral myeloid cells, or both. Deletion of p38α in both cell types recapitulated the previous sex difference, with reduced EAE severity in females. Unexpectedly, deletion of p38α in the periphery was protective in both sexes. In contrast, deletion of p38α in microglia exacerbated EAE in males only, revealing opposing roles of p38α in microglia vs. periphery. Bulk transcriptional profiling revealed that p38α regulated the expression of distinct gene modules in male vs. female microglia. Single-cell transcriptional analysis of WT and p38α-deficient microglia isolated from the inflamed CNS revealed a diversity of complex microglial states, connected by distinct convergent transcriptional trajectories. In males, microglial p38α deficiency resulted in enhanced transition from homeostatic to disease-associated microglial states, with the downregulation of regulatory genes such as Atf3, Rgs1, Socs3, and Btg2, and increased expression of inflammatory genes such as Cd74, Trem2, and MHC class I and II genes. In females, the effect of p38α deficiency was divergent, exhibiting a unique transcriptional profile that included an upregulation of tissue protective genes, and a small subset of inflammatory genes that were also upregulated in males. Taken together, these results reveal a p38α-dependent sex-specific molecular pathway in microglia that is protective in CNS autoimmunity in males, suggesting that autoimmunity in males and females is driven by distinct cellular and molecular pathways, thus suggesting design of future sex-specific therapeutic approaches.
Collapse
Affiliation(s)
- Mahalia M McGill
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Alyssa R Richman
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Joseph R Boyd
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Bristy Sabikunnahar
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Karolyn G Lahue
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Theresa L Montgomery
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Sydney Caldwell
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Stella Varnum
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Seth Frietze
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Dimitry N Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| |
Collapse
|
14
|
Avşar T, Çelikyapi Erdem G, Terzioğlu G, Tahir Turanli E. Investigation of neuro-inflammatory parameters in a cuprizone induced mouse model of multiple sclerosis. Turk J Biol 2021; 45:644-655. [PMID: 34803461 PMCID: PMC8574193 DOI: 10.3906/biy-2104-88] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/27/2021] [Indexed: 01/02/2023] Open
Abstract
Cuprizone, copper chelator, treatment of mouse is a toxic model of multiple sclerosis (MS) in which oligodendrocyte death, demyelination and remyelination can be observed. Understanding T and B cell subset as well as their cytokines involved in MS pathogenesis still requires further scrutiny to better understand immune component of MS. The study presented here, aimed to evaluate relevant cytokines, lymphocytes, and gene expressions profiles during demyelination and remyelination in the cuprizone mouse model of MS. Eighty male C57BL/6J mice fed with 0.2% cuprizone for eight weeks. Cuprizone has been removed from the diet in the following eight weeks. Cuprizone treated and control mice sacrificed biweekly, and corpus callosum of the brain was investigated by staining. Lymphocyte cells of mice analyzed by flow cytometry with CD3e, CD11b, CD19, CD80, CD86, CD4, CD25 and FOXP3 antibodies. IFN-gamma, IL-1alpha, IL-2, IL-5, IL-6, IL-10, IL-17, TNF-alpha cytokines were analyzed in plasma samples. Neuregulin 1 (Nrg1), ciliary neurotrophic factor (Cntf) and C-X-C chemokine receptor type 4 (Cxcr4) gene expressions in corpus callosum sections of the mice brain were quantified. Histochemistry analysis showed that demyelination began at the fourth week of cuprizone administration and total demyelination occurred at the twelfth week in chronic model. Remyelination occurred at the fourth week of following withdrawal of cuprizone from diet. The level of mature and activated T cells, regulatory T cells, T helper cells and mature B cells increased during demyelination and decreased when cuprizone removed from diet. Further, both type 1 and type 2 cytokines together with the proinflammatory cytokines increased. The level of oligodendrocyte maturation and survival genes showed differential gene expression in parallel to that of demyelination and remyelination. In conclusion, for the first-time, involvement of both cellular immune response and antibody response as well as oligodendrocyte maturation and survival factors having role in demyelination and remyelination of cuprizone mouse model of MS have been shown.
Collapse
Affiliation(s)
- Timucin Avşar
- Medical Biology Department, School of Medicine, Bahçeşehir University, İstanbul Turkey
| | - Gökçe Çelikyapi Erdem
- Dr. Orhan Ocalgiray Molecular Biology and Genetics Research Center, İstanbul Technical University, İstanbul Turkey
| | - Gökhan Terzioğlu
- Department of Biotechnology, Institute of Science, Yeditepe University, İstanbul Turkey
| | - Eda Tahir Turanli
- Dr. Orhan Ocalgiray Molecular Biology and Genetics Research Center, İstanbul Technical University, İstanbul Turkey
- Molecular Biology and Genetics Department, Acıbadem University, İstanbul Turkey
| |
Collapse
|
15
|
Yu X, Zizzo Z, Kennedy PG. An appraisal of antigen identification and IgG effector functions driving host immune responses in multiple sclerosis. Mult Scler Relat Disord 2021; 56:103328. [PMID: 34666240 DOI: 10.1016/j.msard.2021.103328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/05/2021] [Accepted: 10/10/2021] [Indexed: 12/16/2022]
Abstract
Increased immunoglobulin G (IgG) antibodies and oligoclonal bands (OCB) are the most characteristic features of multiple sclerosis (MS), a neuroinflammatory demyelinating disease with neurodegeneration at chronic stages. OCB are shown to be associated with disease activity and brain atrophy. Despite intensive research over the last several decades, the antigen specificities of the IgG in MS have remained elusive. We present evidence which supports that intrathecal IgG is not driven by antigen-stimulation, therefore provide reasoning for failed MS antigen identification. Further, the presence of co-deposition of IgG and activated complement products in MS lesions suggest that the IgG effector functions may play a critical role in disease pathogenesis.
Collapse
Affiliation(s)
- Xiaoli Yu
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America.
| | - Zoe Zizzo
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Peter Ge Kennedy
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| |
Collapse
|
16
|
Tagge IJ, Anderson VC, Springer CS, Sammi MK, Bourdette DN, Spain RI, Rooney WD. Gray matter blood-brain barrier water exchange dynamics are reduced in progressive multiple sclerosis. J Neuroimaging 2021; 31:1111-1118. [PMID: 34355458 PMCID: PMC9291753 DOI: 10.1111/jon.12912] [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: 05/26/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose To compare transcapillary wall water exchange, a putative marker of cerebral metabolic health, in brain T2 white matter (WM) lesions and normal appearing white and gray matter (NAWM and NAGM, respectively) in individuals with progressive multiple sclerosis (PMS) and healthy controls (HC). Methods Dynamic‐contrast‐enhanced 7T MRI data were obtained from 19 HC and 23 PMS participants. High‐resolution pharmacokinetic parametric maps representing tissue microvascular and microstructural properties were created by shutter‐speed (SS) paradigm modeling to obtain estimates of blood volume fraction (vb), water molecule capillary efflux rate constant (kpo), and the water capillary wall permeability surface area product (PwS ≡ vb*kpo). Linear regression models were used to investigate differences in (i) kpo and PwS between groups in NAWM and NAGM, and (ii) between WM lesions and NAWM in PMS. Results High‐resolution parametric maps were produced to visualize tissue classes and resolve individual WM lesions. Normal‐appearing gray matter kpo and PwS were significantly decreased in PMS compared to HC (p ≤ .01). Twenty‐one T2 WM lesions were analyzed in 10 participants with PMS. kpo was significantly decreased in WM lesions compared to PMS NAWM (p < .0001). Conclusions Transcapillary water exchange is reduced in PMS NAGM compared to HC and is further reduced in PMS WM lesions, suggesting pathologically impaired brain metabolism. kpo provides a sensitive measure of cerebral metabolic activity and/or coupling, and can be mapped at higher spatial resolution than conventional imaging techniques assessing metabolic activity.
Collapse
Affiliation(s)
- Ian J Tagge
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA.,Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Valerie C Anderson
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Charles S Springer
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Manoj K Sammi
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Dennis N Bourdette
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Rebecca I Spain
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA.,Neurology Division, Veterans Affairs Portland Health Care System, Portland, Oregon, USA
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA.,Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
17
|
Liu H, Yang X, Yang J, Yuan Y, Wang Y, Zhang R, Xiong H, Xu Y. IL-17 Inhibits Oligodendrocyte Progenitor Cell Proliferation and Differentiation by Increasing K + Channel Kv1.3. Front Cell Neurosci 2021; 15:679413. [PMID: 34239419 PMCID: PMC8258110 DOI: 10.3389/fncel.2021.679413] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/20/2021] [Indexed: 12/02/2022] Open
Abstract
Interleukin 17 (IL-17) is a signature cytokine of Th17 cells. IL-17 level is significantly increased in inflammatory conditions of the CNS, including but not limited to post-stroke and multiple sclerosis. IL-17 has been detected direct toxicity on oligodendrocyte (Ol) lineage cells and inhibition on oligodendrocyte progenitor cell (OPC) differentiation, and thus promotes myelin damage. The cellular mechanism of IL-17 in CNS inflammatory diseases remains obscure. Voltage-gated K+ (Kv) channel 1.3 is the predominant Kv channel in Ol and potentially involved in Ol function and cell cycle regulation. Kv1.3 of T cells involves in immunomodulation of inflammatory progression, but the role of Ol Kv1.3 in inflammation-related pathogenesis has not been fully investigated. We hypothesized that IL-17 induces myelin injury through Kv1.3 activation. To test the hypothesis, we studied the involvement of OPC/Ol Kv1.3 in IL-17-induced Ol/myelin injury in vitro and in vivo. Kv1.3 currents and channel expression gradually decreased during the OPC development. Application of IL-17 to OPC culture increased Kv1.3 expression, leading to a decrease of AKT activation, inhibition of proliferation and myelin basic protein reduction, which were prevented by a specific Kv1.3 blocker 5-(4-phenoxybutoxy) psoralen. IL-17-caused myelin injury was validated in LPC-induced demyelination mouse model, particularly in corpus callosum, which was also mitigated by aforementioned Kv1.3 antagonist. IL-17 altered Kv1.3 expression and resultant inhibitory effects on OPC proliferation and differentiation may by interrupting AKT phosphorylating activation. Taken together, our results suggested that IL-17 impairs remyelination and promotes myelin damage by Kv1.3-mediated Ol/myelin injury. Thus, blockade of Kv1.3 as a potential therapeutic strategy for inflammatory CNS disease may partially attribute to the direct protection on OPC proliferation and differentiation other than immunomodulation.
Collapse
Affiliation(s)
- Han Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueke Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanpeng Yuan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanlin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huangui Xiong
- Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
18
|
Schättin A, Häfliger S, Meyer A, Früh B, Böckler S, Hungerbühler Y, de Bruin ED, Frese S, Steinlin Egli R, Götz U, Bauer R, Martin-Niedecken AL. Design and Evaluation of User-Centered Exergames for Patients With Multiple Sclerosis: Multilevel Usability and Feasibility Studies. JMIR Serious Games 2021; 9:e22826. [PMID: 33960956 PMCID: PMC8140386 DOI: 10.2196/22826] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/03/2021] [Accepted: 02/12/2021] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system. Patients with MS experience a wide range of physical and cognitive dysfunctions that affect their quality of life. A promising training approach that concurrently trains physical and cognitive functions is video game-based physical exercising (ie, exergaming). Previous studies have indicated that exergames have positive effects on balance and cognitive functions in patients with MS. However, there is still a need for specific, user-centered exergames that function as a motivating and effective therapy tool for patients with MS and studies investigating their usability and feasibility. OBJECTIVE The aim of this interdisciplinary research project is to develop usable and feasible user-centered exergames for the pressure-sensitive plate Dividat Senso by incorporating theoretical backgrounds from movement sciences, neuropsychology, and game research as well as participatory design processes. METHODS Focus groups (patients and therapists) were set up to define the user-centered design process. This was followed by the field testing of newly developed exergame concepts. Two sequential usability and feasibility studies were conducted on patients with MS. The first study included a single exergaming session followed by measurements. Between the first and second studies, prototypes were iterated based on the findings. The second study ran for 4 weeks (1-2 trainings per week), and measurements were taken before and after the intervention. For each study, participants answered the System Usability Scale (SUS; 10 items; 5-point Likert Scale; score range 0-100) and interview questions. In the second study, participants answered game experience-related questionnaires (Flow Short Scale [FSS]: 13 items; 7-point Likert Scale; score range 1-7; Game Flow questionnaire: 17 items; 6-point Likert Scale; score range 1-6). Mixed methods were used to analyze the quantitative and qualitative data. RESULTS In the first study (N=16), usability was acceptable, with a median SUS score of 71.3 (IQR 58.8-80.0). In the second study (N=25), the median SUS scores were 89.7 (IQR 78.8-95.0; before) and 82.5 (IQR 77.5-90.0; after), and thus, a significant decrease was observed after training (z=-2.077; P=.04; r=0.42). Moreover, high values were observed for the overall FSS (pre: median 5.9, IQR 4.6-6.4; post: median 5.8, IQR 5.4-6.2) and overall Game Flow Questionnaire (pre: median 5.0, IQR 4.7-5.3; post: median 5.1, IQR 4.9-5.3). A significant decrease was observed in the item perceived importance (FSS: z=-2.118; P=.03; r=0.42). Interviews revealed that user-centered exergames were usable, well accepted, and enjoyable. Points of reference were identified for future research and development. CONCLUSIONS The project revealed that the newly developed, user-centered exergames were usable and feasible for patients with MS. Furthermore, exergame elements should be considered in the development phase of user-centered exergames (for patients with MS). Future studies are needed to provide indications about the efficacy of user-centered exergames for patients with MS.
Collapse
Affiliation(s)
- Alexandra Schättin
- Department of Health Sciences and Technology, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Stephan Häfliger
- Department of Health Sciences and Technology, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Alain Meyer
- Department of Health Sciences and Technology, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Barbara Früh
- Department of Health Sciences and Technology, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Sonja Böckler
- Department of Design, Subject Area in Game Design, Zurich University of the Arts, Zurich, Switzerland
| | - Yannic Hungerbühler
- Department of Design, Subject Area in Game Design, Zurich University of the Arts, Zurich, Switzerland
| | - Eling D de Bruin
- Department of Health Sciences and Technology, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
- Division of Physiotherapy, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Sebastian Frese
- Technology and Innovation Unit and Department of Research, ZURZACH Care, Bad Zurzach, Switzerland
| | | | - Ulrich Götz
- Department of Design, Subject Area in Game Design, Zurich University of the Arts, Zurich, Switzerland
| | - René Bauer
- Department of Design, Subject Area in Game Design, Zurich University of the Arts, Zurich, Switzerland
| | | |
Collapse
|
19
|
Pérez Sánchez S, Eichau Madueño S, Rus Hidalgo M, Domínguez Mayoral A, Vilches-Arenas A, Navarro Mascarell G, Izquierdo G. Usefulness of optic nerve ultrasound to predict clinical progression in multiple sclerosis. NEUROLOGÍA (ENGLISH EDITION) 2021. [DOI: 10.1016/j.nrleng.2017.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
20
|
Multiple sclerosis risk gene Mertk is required for microglial activation and subsequent remyelination. Cell Rep 2021; 34:108835. [PMID: 33691116 DOI: 10.1016/j.celrep.2021.108835] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/23/2020] [Accepted: 02/17/2021] [Indexed: 12/26/2022] Open
Abstract
In multiple sclerosis (MS) and other neurological diseases, the failure to repair demyelinated lesions contributes to axonal damage and clinical disability. Here, we provide evidence that Mertk, a gene highly expressed by microglia that alters MS risk, is required for efficient remyelination. Compared to wild-type (WT) mice, Mertk-knockout (KO) mice show impaired clearance of myelin debris and remyelination following demyelination. Using single-cell RNA sequencing, we characterize Mertk-influenced responses to cuprizone-mediated demyelination and remyelination across different cell types. Mertk-KO brains show an attenuated microglial response to demyelination but an elevated proportion of interferon (IFN)-responsive microglia. In addition, we identify a transcriptionally distinct subtype of surviving oligodendrocytes specific to demyelinated lesions. The inhibitory effect of myelin debris on remyelination is mediated in part by IFNγ, which further impedes microglial clearance of myelin debris and inhibits oligodendrocyte differentiation. Together, our work establishes a role for Mertk in microglia activation, phagocytosis, and migration during remyelination.
Collapse
|
21
|
Sarkar A, Saha S, Paul A, Maji A, Roy P, Maity TK. Understanding stem cells and its pivotal role in regenerative medicine. Life Sci 2021; 273:119270. [PMID: 33640402 DOI: 10.1016/j.lfs.2021.119270] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/06/2021] [Accepted: 02/14/2021] [Indexed: 02/07/2023]
Abstract
Stem cells (SCs) are clonogenic cells that develop into the specialized cells which later responsible for making up various types of tissue in the human body. SCs are not only the appropriate source of information for cell division, molecular and cellular processes, and tissue homeostasis but also one of the major putative biological aids to diagnose and cure various degenerative diseases. This study emphasises on various research outputs that occurred in the past two decades. This will give brief information on classification, differentiation, detection, and various isolation techniques of SCs. Here, the various signalling pathways which includes WNT, Sonic hedgehog, Notch, BMI1 and C-met pathways and how does it effect on the regeneration of various classes of SCs and factors that regulates the potency of the SCs are also been discussed. We also focused on the application of SCs in the area of regenerative medicine along with the cellular markers that are useful as salient diagnostic or curative tools or in both, by the process of reprogramming, which includes diabetes, cancer, cardiovascular disorders and neurological disorders. The biomarkers that are mentioned in various literatures and experiments include PDX1, FOXA2, HNF6, and NKX6-1 (for diabetes); CD33, CD24, CD133 (for cancer); c-Kit, SCA-1, Wilm's tumor 1 (for cardiovascular disorders); and OCT4, SOX2, c-MYC, EN1, DAT and VMAT2 (for neurological disorders). In this review, we come to know the advancements and scopes of potential SC-based therapies, its diverse applications in clinical fields that can be helpful in the near future.
Collapse
Affiliation(s)
- Arnab Sarkar
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Sanjukta Saha
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Abhik Paul
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Avik Maji
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Puspita Roy
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Tapan Kumar Maity
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India.
| |
Collapse
|
22
|
Block VJ, Gopal A, Rowles W, -Yueh C, Gelfand JM, Bove R. CoachMS, an innovative closed-loop, interdisciplinary platform to monitor and proactively treat MS symptoms: A pilot study. Mult Scler J Exp Transl Clin 2021; 7:2055217321988937. [PMID: 33796329 PMCID: PMC7970691 DOI: 10.1177/2055217321988937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/01/2021] [Indexed: 12/02/2022] Open
Abstract
Background There are numerous challenges to treating co-occurring symptoms in multiple sclerosis (MS). Objective To pilot the feasibility of a novel symptom management platform, CoachMS, to monitor MS symptoms (bladder function, ambulation, and mood: BAM) and respond to changes in real-time. Methods In this 12-week randomized controlled pilot trial, participants’ symptoms were monitored using weekly questionnaires and remote ambulatory monitoring (Fitbit Flex2®). Behavioral change principles used included shared goal setting at 2 weeks. Between weeks 2-12, the CoachMS group received targeted contact and interventions if symptoms worsened; the control group were treated through usual clinic practice. Our outcomes were feasibility (retention, adherence and acceptability; primary) and proportion of recommended treatments pursued (secondary); efficacy was explored. Results Of 21 participants enrolled, 13 (62%) completed the study; protocol adherence was excellent. CoachMS participants demonstrated greater follow-through with clinical recommendations than controls (OR 9.3, 95% CI (0.9, 97.6)). As a cohort, each BAM symptom tended to improve. Suicidality was detected in one control participant, resulting in urgent evaluation and hospitalization. Conclusions The innovative CoachMS platform was feasible and acceptable in this cohort with baseline BAM symptoms. It could represent an accessible, cost-effective tool to monitor MS symptoms in real-time; a larger trial is planned.
Collapse
Affiliation(s)
- Valerie J Block
- UCSF Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Arpita Gopal
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, CA, USA
| | - William Rowles
- UCSF Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Chu -Yueh
- UCSF Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey M Gelfand
- UCSF Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Riley Bove
- UCSF Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW To outline recent applications of e-health data and digital tools for improving the care and management of healthcare for people with multiple sclerosis. RECENT FINDINGS The digitization of most clinical data, along with developments in communication technologies, miniaturization of sensors and computational advances are enabling aggregation and clinically meaningful analyses of real-world data from patient registries, digital patient-reported outcomes and electronic health records (EHR). These data are allowing more confident descriptions of prognoses for multiple sclerosis patients and the long-term relative benefits and safety of disease-modifying treatments (DMT). Registries allow detailed, multiple sclerosis-specific data to be shared between clinicians more easily, provide data needed to improve the impact of DMT and, with EHR, characterize clinically relevant interactions between multiple sclerosis and other diseases. Wearable sensors provide continuous, long-term measures of performance dynamics in relevant ecological settings. In conjunction with telemedicine and online apps, they promise a major expansion of the scope for patients to manage aspects of their own care. Advances in disease understanding, decision support and self-management using these Big Data are being accelerated by machine learning and artificial intelligence. SUMMARY Both health professionals and patients can employ e-health approaches and tools for development of a more patient-centred learning health system.
Collapse
|
24
|
Johnson DK, Reynolds KM, Poole BD, Montierth MD, Todd VM, Barnado A, Davis MF. Contribution of viral infection to risk for cancer in systemic lupus erythematosus and multiple sclerosis. PLoS One 2021; 16:e0243150. [PMID: 33481783 PMCID: PMC7822541 DOI: 10.1371/journal.pone.0243150] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/17/2020] [Indexed: 11/19/2022] Open
Abstract
Patients with autoimmune disorders (AD) have altered cancer risks compared to the general population. Systemic lupus erythematosus and multiple sclerosis lead to a heightened risk for hematological malignancies and decreased risk for breast, ovarian, and prostate malignancies. Often patients with autoimmune disease have dysregulated antiviral immune responses, including against oncogenic viruses. To uncover the relationship between viral incidence and cancer risk in the context of autoimmune disease, we extracted electronic health records (EHR) from Vanderbilt University. ICD-9/10 codes and laboratory values were collected for hematological, lung, anal-vaginal, thyroid, hepatobiliary, bladder, prostate, and breast cancers; and viruses including Epstein Barr virus (EBV), Human papilloma virus (HPV), and Hepatitis A/B/C (Hep). Only viral infections that led to a physician visit or laboratory test were entered into the EMR; therefore, only clinically relevant cases were noted and considered positive in this study. The relationship between virus infection and cancer in an SLE cohort (SLE-cases n = 2,313, and SLE-controls n = 5,702) and an MS cohort (MS-case n = 7,277, MS-control n = 7,277) was examined by multilinear logistic regression. Viral infection was strongly associated with increased risk for cancer overall. SLE and MS patients were more susceptible to all viral infections. MS patients trended toward increased risk for cancers overall, while decreased risk for hormone-based cancers in SLE patients non-significantly reduced their risk for overall cancer. Both SLE and MS patients had increased clinically relevant EBV infection, which was associated with risk for hematological cancers. Preventing viral infections by vaccination may be especially helpful in controlling risk for cancer in SLE and MS patients.
Collapse
Affiliation(s)
- Deborah K. Johnson
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States of America
| | - Kaylia M. Reynolds
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States of America
| | - Brian D. Poole
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States of America
| | - Matthew D. Montierth
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States of America
| | - Vera M. Todd
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States of America
| | - April Barnado
- Division of Rheumatology & Immunology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Mary F. Davis
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States of America
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States of America
- * E-mail:
| |
Collapse
|
25
|
Levy O, Rothhammer V, Mascanfroni I, Tong Z, Kuai R, De Biasio M, Wang Q, Majid T, Perrault C, Yeste A, Kenison JE, Safaee H, Musabeyezu J, Heinelt M, Milton Y, Kuang H, Lan H, Siders W, Multon MC, Rothblatt J, Massadeh S, Alaamery M, Alhasan AH, Quintana FJ, Karp JM. A cell-based drug delivery platform for treating central nervous system inflammation. J Mol Med (Berl) 2021; 99:663-671. [PMID: 33398468 DOI: 10.1007/s00109-020-02003-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/22/2020] [Accepted: 10/29/2020] [Indexed: 01/18/2023]
Abstract
Mesenchymal stem cells (MSCs) are promising candidates for the development of cell-based drug delivery systems for autoimmune inflammatory diseases, such as multiple sclerosis (MS). Here, we investigated the effect of Ro-31-8425, an ATP-competitive kinase inhibitor, on the therapeutic properties of MSCs. Upon a simple pretreatment procedure, MSCs spontaneously took up and then gradually released significant amounts of Ro-31-8425. Ro-31-8425 (free or released by MSCs) suppressed the proliferation of CD4+ T cells in vitro following polyclonal and antigen-specific stimulation. Systemic administration of Ro-31-8425-loaded MSCs ameliorated the clinical course of experimental autoimmune encephalomyelitis (EAE), a murine model of MS, displaying a stronger suppressive effect on EAE than control MSCs or free Ro-31-8425. Ro-31-8425-MSC administration resulted in sustained levels of Ro-31-8425 in the serum of EAE mice, modulating immune cell trafficking and the autoimmune response during EAE. Collectively, these results identify MSC-based drug delivery as a potential therapeutic strategy for the treatment of autoimmune diseases. KEY MESSAGES: MSCs can spontaneously take up the ATP-competitive kinase inhibitor Ro-31-8425. Ro-31-8425-loaded MSCs gradually release Ro-31-8425 and exhibit sustained suppression of T cells. Ro-31-8425-loaded MSCs have more sustained serum levels of Ro-31-8425 than free Ro-31-8425. Ro-31-8425-loaded MSCs are more effective than MSCs and free Ro-31-8425 for EAE therapy.
Collapse
Affiliation(s)
- Oren Levy
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Veit Rothhammer
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ivan Mascanfroni
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhixiang Tong
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Rui Kuai
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael De Biasio
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Qingping Wang
- Department of Drug Metabolism and Pharmacokinetics, Sanofi R&D, Waltham, MA, USA
| | - Tahir Majid
- Global Research Program and Portfolio Management, Sanofi-Genzyme, Cambridge, MA, USA
| | - Christelle Perrault
- Sanofi R&D, In Vitro Pharmacology, Integrated Drug Discovery, Centre de Recherche Vitry-Alfortville, Vitry-Sur-Seine, France
| | - Ada Yeste
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica E Kenison
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Helia Safaee
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Juliet Musabeyezu
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Martina Heinelt
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Yuka Milton
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Heidi Kuang
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Haoyue Lan
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - William Siders
- Genzyme R&D, Neuroimmunology Research, Framingham, MA, USA
| | - Marie-Christine Multon
- Sanofi R&D, Translational Sciences, Centre de Recherche Vitry-Alfortville, Vitry-Sur-Seine, France
| | | | - Salam Massadeh
- Developmental Medicine Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Manal Alaamery
- Developmental Medicine Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Ali H Alhasan
- Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
- National Center of Pharmaceutical Technology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Francisco J Quintana
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Jeffrey M Karp
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA.
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
26
|
Block VJ, Mestas O, Anderson A, Singh J, Wylie L, Guo CY, Green AJ, Gelfand JM, Bove R. Underutilization of physical therapy for symptomatic women with MS during and following pregnancy. Mult Scler Relat Disord 2020; 48:102703. [PMID: 33387862 DOI: 10.1016/j.msard.2020.102703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/17/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND Many patients with MS continue to have symptoms of their disease even when inflammatory activity is reduced by DMTs. Although disease activity tends to be reduced during pregnancy - especially in the third trimester - women with MS can experience ongoing symptoms during pregnancy, or new ones in the immediate postpartum period, that degrade quality of life. While many MS-related and postpartum symptoms can be improved with physical therapy (PT), there are currently no guidelines on pregnancy-related rehabilitation in MS. In this analysis, we evaluated the prevalence of PT-amenable symptoms and patterns of PT referrals in a cohort of UCSF MS Clinic patients who became pregnant. METHODS We extracted electronic medical records (EMR) data for the year before conception, during pregnancy, and year postpartum for women with MS cared for at UCSF between 09-2005 and 08-2019. This included clinical visits, MS therapies and symptoms (as defined by the National MS Society). PT and pelvic floor PT orders and notes were also extracted. RESULTS We included 142 live birth pregnancies from 118 women. During the course of their pregnancy and within the year postpartum, 107 women (75.4%) reported at least one PT-amenable symptom. A total of 30 (28.0%) referrals were made to PT, with attendance confirmed for 10 (33.3% of referrals). Symptoms most commonly triggering a referral for PT evaluation were numbness and urinary incontinence. Falls were reported after 10 of the pregnancies; 4 resulted in a referral to PT. Forty-one women reported urinary incontinence: 11 (26.8%) were referred to PT, and 2 to pelvic floor PT. Nineteen women experienced a documented relapse during pregnancy and/or postpartum: 11 received a PT referral, and 4 attended PT. CONCLUSIONS While women with MS recorded at least 1 PT-amenable symptom during or following 75.4% of their pregnancies, only 28% of these were referred to PT - and only a third attended PT. Of significance was the 4.9% referral rate for pelvic floor PT in postpartum women with a record of urinary incontinence. Pelvic floor PT is a mainstay of general postpartum care in many European countries. These data illustrate critical gaps in rehabilitation referral, access and use at the intersection of neurological conditions and pregnancy in a large US-based MS clinic. They lend support for quality improvement efforts to improve care pathways and for telerehabilitation innovations to reduce barriers to access and improve synergistic care between PT, MD and urologic care.
Collapse
Affiliation(s)
- Valerie J Block
- Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, University of California San Francisco, Department of Neurology.
| | - Olivia Mestas
- Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, University of California San Francisco, Department of Neurology.
| | - Annika Anderson
- Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, University of California San Francisco, Department of Neurology.
| | - Jessica Singh
- Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, University of California San Francisco, Department of Neurology.
| | - Leah Wylie
- Rehabilitation Service Mount Zion Campus, University of California San Francisco, Department of Neurology, Department of Physical Therapy and Rehabilitation Science.
| | - Chu-Yueh Guo
- Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, University of California San Francisco, Department of Neurology.
| | - Ari J Green
- Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, University of California San Francisco, Department of Neurology.
| | - Jeffrey M Gelfand
- Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, University of California San Francisco, Department of Neurology.
| | - Riley Bove
- Weill Institute for Neurosciences, MS and Neuroinflammation Clinic, University of California San Francisco, Department of Neurology.
| |
Collapse
|
27
|
Chauhan P, Kakkar AK, Singh H, Gautam CS. Minocycline for the management of multiple sclerosis: repositioning potential, opportunities, and challenges. Expert Rev Neurother 2020; 21:35-43. [PMID: 33059513 DOI: 10.1080/14737175.2020.1838276] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic demyelinating inflammatory disorder with variable clinical and pathologic characteristics reflecting multiple underlying pathophysiologic mechanisms. Repositioning of existing drugs for the new indications offers several advantages including significant reduction in the cost and time of drug development and exemption from early phase clinical trials. Minocycline has been reported to exhibit immunomodulation in several pre-clinical and clinical studies through suppression of migratory inflammatory cells, modulation of peripheral immune response, and inhibition of microglial activation within the CNS. AREAS COVERED Here, the authors review the repositioning potential of minocycline for the treatment of MS along with appraisal of the evidence obtained from preclinical and clinical research. The authors also discuss the advantages and potential safety concerns related to the use of minocycline for the management of MS. EXPERT OPINION Minocycline offers several distinct advantages in terms of well-known safety profile, lower cost of therapy, widespread availability, and being available as an oral formulation. The authors call upon the public and private funders to facilitate well designed and adequately powered randomized clinical trials that can provide conclusive evidence regarding the safety and efficacy of minocycline in patients with MS.
Collapse
Affiliation(s)
- Prerna Chauhan
- Department of Pharmacology, All India Institute of Medical Sciences , New Delhi, India
| | - Ashish Kumar Kakkar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research , Chandigarh, India
| | - Harmanjit Singh
- Department of Pharmacology, Government Medical College and Hospital , Chandigarh, India
| | - C S Gautam
- Department of Pharmacology, Government Medical College and Hospital , Chandigarh, India
| |
Collapse
|
28
|
Inhibition of Human Neutrophil Functions In Vitro by Multiple Sclerosis Disease-Modifying Therapies. J Clin Med 2020; 9:jcm9113542. [PMID: 33147889 PMCID: PMC7692529 DOI: 10.3390/jcm9113542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/21/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
There is a growing optimism about the potential of new disease-modifying therapies (DMTs) in the management of relapsing-remitting multiple sclerosis (RRMS) patients. However, this initial enthusiasm has been tempered by evidence indicating that multiple sclerosis (MS) patients undergoing DMT may be at higher risk of developing infections through incompletely understood mechanisms. As neutrophils provide the first line of defense against pathogens, here we have compared the effects of some of the commonly used MS DMTs (i.e., moderate-efficacy injective, first-line: interferonβ-1b (IFNβ-1b), glatiramer acetate (GA); and high-efficacy, second-line: fingolimod (FTY) and natalizumab (NAT)) on the in vitro viability and functions of neutrophils isolated from healthy subjects. All the DMTs tested impaired the ability of neutrophils to kill Klebsiella pneumoniae, whereas none of them affected the rate of neutrophil apoptosis or CD11b and CD62L cell surface expression. Intriguingly, only FTY exposure negatively affected K. pneumoniae-induced production of reactive oxygen species (ROS) in polymorphonuclear leukocytes (PMNs). Furthermore, neutrophils exposed to K. pneumoniae secreted enhanced amounts of CXCL8, IL-1β and TNF-α, which were differentially regulated following DMT pretreatment. Altogether, these findings suggest that DMTs may increase the susceptibility of MS patients to microbial infections, in part, through inhibition of neutrophil functions. In light of these data, we recommend that the design of personalized therapies for RRMS patients should take into account not just the mechanism of action of the chosen DMT but also the potential risk of infection associated with the administration of such therapeutic compounds to this highly vulnerable population.
Collapse
|
29
|
Bacmeister CM, Barr HJ, McClain CR, Thornton MA, Nettles D, Welle CG, Hughes EG. Motor learning promotes remyelination via new and surviving oligodendrocytes. Nat Neurosci 2020; 23:819-831. [PMID: 32424285 PMCID: PMC7329620 DOI: 10.1038/s41593-020-0637-3] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 04/07/2020] [Indexed: 11/24/2022]
Abstract
Oligodendrocyte loss in neurological disease leaves axons vulnerable to damage and degeneration, and activity-dependent myelination may represent an endogenous mechanism to improve remyelination following injury. Here, we report that while learning a forelimb reach task transiently suppresses oligodendrogenesis, it subsequently increases OPC differentiation, oligodendrocyte generation, and myelin sheath remodeling in the forelimb motor cortex. Immediately followingdemyelination, neurons exhibit hyperexcitability, learning is impaired, and behavioral intervention provides no benefit to remyelination. However, partial remyelination restores neuronal and behavioral function allowing learning to enhance oligodendrogenesis, remyelination of denuded axons, and the ability of surviving oligodendrocytes to generate new myelinsheaths. Previously considered controversial, we show that sheath generation by mature oligodendrocytes is not only possible but also increases myelin pattern preservation following demyelination, presenting a new target for therapeutic interventions. Together, our findings demonstrate that precisely-timed motor learning improves recovery from demyelinating injury via enhanced remyelination from new and surviving oligodendrocytes.
Collapse
Affiliation(s)
- Clara M Bacmeister
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Helena J Barr
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Crystal R McClain
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael A Thornton
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Dailey Nettles
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Neurosurgery, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Cristin G Welle
- Department of Neurosurgery, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ethan G Hughes
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
30
|
McGill MM, Sabikunnahar B, Fang Q, Teuscher C, Krementsov DN. The sex-specific role of p38 MAP kinase in CNS autoimmunity is regulated by estrogen receptor alpha. J Neuroimmunol 2020; 342:577209. [PMID: 32200131 PMCID: PMC8978838 DOI: 10.1016/j.jneuroim.2020.577209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/30/2022]
Abstract
Biological sex is a critical factor in regulating immune function. A striking example of this is the higher prevalence of autoimmune diseases such as multiple sclerosis (MS) and lupus in females compared to males. While many studies have implicated the role of sex hormones such as estrogens and androgens in these sex differences, surprisingly little is known about other molecular pathways that underlie sex differences or interact with sex hormones. We have previously shown that conditional ablation of p38α MAP kinase signaling in myeloid cells (p38αCKO) was protective in a mouse model of MS, experimental autoimmune encephalomyelitis (EAE), in female but not male mice. This sex difference was dependent on the presence of sex hormones, leading us to hypothesize that the pathogenic function of p38α in EAE depends on estrogen signaling via one of the two nuclear estrogen receptors, encoded by Esr1 and Esr2 . To test this hypothesis, we performed experiments with p38αCKO macrophages, which demonstrated that the effects of estradiol and p38α were independent of one another in vitro . Since many sex hormone effects are lost in vitro, we generated p38αCKO mice lacking either Esr1 or Esr2 , and evaluated their EAE susceptibility in vivo . Myeloid-specific deletion of Esr1 abrogated protection in p38αCKO females, although global deletion of Esr1 and Esr2 did not. Moreover, global or myeloid-specific disruption of Esr1 unexpectedly promoted protection from EAE in p38αCKO males. Mechanistically, Esr1 deletion resulted in partial reprogramming of p38α-dependent transcriptional modules in male macrophages, in particular those regulated by TGFβ, BRD4, and SMARCA4. These results demonstrate that estrogen signaling in myeloid cells plays an important sex-specific role in programming their dependence on specific intracellular signaling pathways in the context of autoimmune disease pathogenesis, suggesting potential avenues for sex-specific therapeutics or combinatorial approaches for the treatment of such diseases.
Collapse
Affiliation(s)
- Mahalia M McGill
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Bristy Sabikunnahar
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Qian Fang
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Cory Teuscher
- Department of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Dimitry N Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA.
| |
Collapse
|
31
|
Bar-Or A, Pender MP, Khanna R, Steinman L, Hartung HP, Maniar T, Croze E, Aftab BT, Giovannoni G, Joshi MA. Epstein-Barr Virus in Multiple Sclerosis: Theory and Emerging Immunotherapies. Trends Mol Med 2019; 26:296-310. [PMID: 31862243 PMCID: PMC7106557 DOI: 10.1016/j.molmed.2019.11.003] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/31/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022]
Abstract
New treatments for multiple sclerosis (MS) focused on B cells have created an atmosphere of excitement in the MS community. B cells are now known to play a major role in disease, demonstrated by the highly impactful effect of a B cell-depleting antibody on controlling MS. The idea that a virus may play a role in the development of MS has a long history and is supported mostly by studies demonstrating a link between B cell-tropic Epstein–Barr virus (EBV) and disease onset. Efforts to develop antiviral strategies for treating MS are underway. Although gaps remain in our understanding of the etiology of MS, the role, if any, of viruses in propagating pathogenic immune responses deserves attention.
Collapse
Affiliation(s)
- Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P Pender
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Rajiv Khanna
- Centre for Immunotherapy and Vaccine Development, Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Beckman Center for Molecular Medicine, Stanford University Medical Center, Stanford, CA, USA
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Tap Maniar
- Clinical Development, Torque Therapeutics, Boston, MA, USA
| | - Ed Croze
- IRIS-Bay, San Francisco, CA, USA.
| | - Blake T Aftab
- Preclinical Science and Translational Medicine, Atara Biotherapeutics, South San Francisco, CA, USA
| | - Gavin Giovannoni
- Blizard Institute, Queen Mary University London, Barts and the London School of Medicine, London, UK
| | - Manher A Joshi
- Medical Affairs, Atara Biotherapeutics, South San Francisco, CA, USA
| |
Collapse
|
32
|
Combinatory Multifactor Treatment Effects on Primary Nanofiber Oligodendrocyte Cultures. Cells 2019; 8:cells8111422. [PMID: 31726669 PMCID: PMC6912369 DOI: 10.3390/cells8111422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system. Neurological deficits are attributed to inflammatory demyelination, which compromises axonal function and survival. These are mitigated in experimental models by rapid and often complete remyelination of affected axons, but in MS this endogenous repair mechanism frequently fails, leaving axons increasingly vulnerable to the detrimental effects of inflammatory and metabolic stress. Understanding the molecular basis of remyelination and remyelination failure is essential to develop improved therapies for this devastating disease. However, recent studies suggest that this is not due to a single dominant mechanism, but rather represents the biological outcome of multiple changes in the lesion microenvironment that combine to disrupt oligodendrocyte differentiation. This identifies a pressing need to develop technical platforms to investigate combinatory and/or synergistic effects of factors differentially expressed in MS lesions on oligodendrocyte proliferation and differentiation. Here we describe protocols using primary oligodendrocyte cultures from Bl6 mice on 384-well nanofiber plates to model changes affecting oligodendrogenesis and differentiation in the complex signaling environment associated with multiple sclerosis lesions. Using platelet-derived growth factor (PDGF–AA), fibroblast growth factor 2 (FGF2), bone morphogenetic protein 2 (BMP2) and bone morphogenetic protein 4 (BMP4) as representative targets, we demonstrate that we can assess their combinatory effects across a wide range of concentrations in a single experiment. This in vitro model is ideal for assessing the combinatory effects of changes in availability of multiple factors, thus more closely modelling the situation in vivo and furthering high-throughput screening possibilities.
Collapse
|
33
|
Kim D, Park Y. Molecular mechanism for the multiple sclerosis risk variant rs17594362. Hum Mol Genet 2019; 28:3600-3609. [PMID: 31509193 PMCID: PMC6927461 DOI: 10.1093/hmg/ddz216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/31/2019] [Accepted: 09/02/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is known as an autoimmune demyelinating disease of the central nervous system. However, its cause remains elusive. Given previous studies suggesting that dysfunctional oligodendrocytes (OLs) may trigger MS, we tested whether single nucleotide polymorphisms (SNPs) associated with MS affect OL enhancers, potentially increasing MS risk by dysregulating gene expression of OL lineage cells. We found that two closely spaced OL enhancers, which are 3 Kb apart on chromosome 13, overlap two MS SNPs in linkage disequilibrium-rs17594362 and rs12429256. Our data revealed that the two MS SNPs significantly up-regulate the associated OL enhancers, which we have named as Rgcc-E1 and Rgcc-E2. Analysis of Hi-C data and epigenome editing experiments shows that Rgcc is the primary target of Rgcc-E1 and Rgcc-E2. Collectively, these data indicate that the molecular mechanism of rs17594362 and rs12429256 is to induce Rgcc overexpression by potentiating the enhancer activity of Rgcc-E1 and Rgcc-E2. Importantly, the dosage of the rs17594362/rs12429256 risk allele is positively correlated with the expression level of Rgcc in the human population, confirming our molecular mechanism. Our study also suggests that Rgcc overexpression in OL lineage cells may be a key cellular mechanism of rs17594362 and rs12429256 for MS.
Collapse
Affiliation(s)
- Dongkyeong Kim
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Yungki Park
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
34
|
Correa-Díaz EP, Ortiz MA, Toral AM, Guillen F, Terán E, Ontaneda D, García-Castillo M, Jácome-Sánchez C, Torres-Herrán G, Ortega-Heredia A, Buestán ME, Murillo-Calle J, Raza P, Baño G. Prevalence of multiple sclerosis in Cuenca, Ecuador. Mult Scler J Exp Transl Clin 2019; 5:2055217319884952. [PMID: 31695924 PMCID: PMC6822194 DOI: 10.1177/2055217319884952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/28/2019] [Accepted: 10/04/2019] [Indexed: 11/16/2022] Open
Abstract
Background Methods Results Conclusions
Collapse
Affiliation(s)
| | | | | | | | - Enrique Terán
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito
| | - Daniel Ontaneda
- Meller Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic
| | | | | | | | | | | | | | - Praneeta Raza
- Meller Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic
| | - Guillermo Baño
- Department of Neurology, Carlos Andrade Marín Hospital, Ecuador
| |
Collapse
|
35
|
Afrang N, Tavakoli R, Tasharrofi N, Alian A, Naderi Sohi A, Kabiri M, Fathi-Roudsari M, Soufizomorrod M, Rajaei F, Soleimani M, Kouhkan F. A critical role for miR-184 in the fate determination of oligodendrocytes. Stem Cell Res Ther 2019; 10:112. [PMID: 30922384 PMCID: PMC6440085 DOI: 10.1186/s13287-019-1208-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/14/2019] [Accepted: 03/06/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND New insights on cellular and molecular aspects of both oligodendrocyte (OL) differentiation and myelin synthesis pathways are potential avenues for developing a cell-based therapy for demyelinating disorders comprising multiple sclerosis. MicroRNAs (miRNA) have broad implications in all aspects of cell biology including OL differentiation. MiR-184 has been identified as one of the most highly enriched miRNAs in oligodendrocyte progenitor cells (OPCs). However, the exact molecular mechanism of miR-184 in OL differentiation is yet to be elucidated. METHODS AND RESULTS Based on immunochemistry assays, qRT-PCR, and western blotting findings, we hypothesized that overexpression of miR-184 in either neural progenitor cells (NPCs) or embryonic mouse cortex stimulated the differentiation of OL lineage efficiently through regulating crucial developmental genes. Luciferase assays demonstrated that miR-184 directly represses positive regulators of neural and astrocyte differentiation, i.e., SOX1 and BCL2L1, respectively, including the negative regulator of myelination, LINGO1. Moreover, blocking the function of miR-184 reduced the number of committed cells to an OL lineage. CONCLUSIONS Our data highlighted that miR-184 could promote OL differentiation even in the absence of exogenous growth factors and propose a novel strategy to improve the efficacy of OL differentiation, with potential applications in cell therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Negin Afrang
- Stem Cell Technology Research Center, P.O. Box: 15856-36473, Tehran, Iran
- School of Paramedical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Rezvan Tavakoli
- Stem Cell Technology Research Center, P.O. Box: 15856-36473, Tehran, Iran
| | - Nooshin Tasharrofi
- Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Amir Alian
- Stem Cell Technology Research Center, P.O. Box: 15856-36473, Tehran, Iran
- Department of Chemistry, Rice University, Houston, TX 77054 USA
| | | | - Mahboubeh Kabiri
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | | | - Mina Soufizomorrod
- Tissue Engineering and Applied Cell Sciences Division, Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farzad Rajaei
- School of Paramedical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Masoud Soleimani
- Stem Cell Technology Research Center, P.O. Box: 15856-36473, Tehran, Iran
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box: 14115-331, Tehran, Iran
| | - Fatemeh Kouhkan
- Stem Cell Technology Research Center, P.O. Box: 15856-36473, Tehran, Iran
| |
Collapse
|
36
|
Yu WB, Wang Q, Chen S, Cao L, Tang J, Ma CG, Xiao W, Xiao BG. The therapeutic potential of ginkgolide K in experimental autoimmune encephalomyelitis via peripheral immunomodulation. Int Immunopharmacol 2019; 70:284-294. [PMID: 30851709 DOI: 10.1016/j.intimp.2019.02.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/14/2019] [Accepted: 02/20/2019] [Indexed: 11/29/2022]
Abstract
Multiple sclerosis is a T cell-mediated inflammatory, demyelinating disease of the central nervous system, accompanied by neuronal degeneration. Based on the anti-inflammatory effects of Ginkgolide K (GK), a platelet activating factor antagonist, we explored the possible application of GK in the treatment of MS. The results showed that GK effectively ameliorated the severity of experimental autoimmune encephalomyelitis. The intervention of GK inhibited the infiltration of inflammatory cells and demyelination in the spinal cord. At the same time, the expression of the inflammation-related molecules TLR4, NF-κB, and COX2 in the spinal cord was significantly lower in the GK-treated mice, indicating that GK intervention can inhibit the inflammatory microenvironment of the spinal cord in EAE mice. In mouse spleen lymphocytes, GK increased the proportion of regulatory T cells (Treg) and reduced the proportion of T helper 17 cells (Th17), modifying the imbalance between Th17/Treg cells. Additionally, GK shifted macrophage/microglia polarization from M1 to M2 cell type. Importantly, GK inhibited the expression of chemotactic molecules CCL-2, CCL-3 and CCL-5, thereby limiting the migration of inflammatory cells to the spinal cord. Our results provide the possibility that GK may be a promising naturally small molecule compound for the future treatment of MS.
Collapse
Affiliation(s)
- Wen-Bo Yu
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Qing Wang
- 2011 Collaborative Innovation Center/Research Center of Neurobiology, University of Shanxi Traditional Chinese Medicine, Taiyuan, Shanxi, China
| | - Sheng Chen
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Liang Cao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, Jiangsu, China
| | - Jie Tang
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Cun-Gen Ma
- 2011 Collaborative Innovation Center/Research Center of Neurobiology, University of Shanxi Traditional Chinese Medicine, Taiyuan, Shanxi, China
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, Jiangsu, China
| | - Bao-Guo Xiao
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| |
Collapse
|
37
|
Sapkota A, Park SJ, Choi JW. Neuroprotective Effects of 6-Shogaol and Its Metabolite, 6-Paradol, in a Mouse Model of Multiple Sclerosis. Biomol Ther (Seoul) 2019; 27:152-159. [PMID: 30001610 PMCID: PMC6430232 DOI: 10.4062/biomolther.2018.089] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 05/23/2018] [Accepted: 05/29/2018] [Indexed: 12/21/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by progressive neuronal loss, neuroinflammation, axonal degeneration, and demyelination. Previous studies have reported that 6-shogaol, a major constituent of ginger (Zingiber officinale rhizome), and its biological metabolite, 6-paradol, have anti-inflammatory and anti-oxidative properties in the central nervous system (CNS). In the present study, we investigated whether 6-shogaol and 6-paradol could ameliorate against experimental autoimmune encephalomyelitis (EAE), a mouse model of MS elicited by myelin oligodendrocyte glycoprotein (MOG35-55) peptide immunization with injection of pertussis toxin. Once-daily administration of 6-shogaol and 6-paradol (5 mg/kg/day, p.o.) to symptomatic EAE mice significantly alleviated clinical signs of the disease along with remyelination and reduced cell accumulation in the white matter of spinal cord. Administration of 6-shogaol and 6-paradol into EAE mice markedly reduced astrogliosis and microglial activation as key features of immune responses inside the CNS. Furthermore, administration of these two molecules significantly suppressed expression level of tumor necrosis factor-α, a major proinflammatory cytokine, in EAE spinal cord. Collectively, these results demonstrate therapeutic efficacy of 6-shogaol or 6-paradol for EAE by reducing neuroinflammatory responses, further indicating the therapeutic potential of these two active ingredients of ginger for MS.
Collapse
Affiliation(s)
- Arjun Sapkota
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Republic of Korea
| | - Se Jin Park
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ji Woong Choi
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Republic of Korea
| |
Collapse
|
38
|
Selective Estrogen Receptor Modulators Enhance CNS Remyelination Independent of Estrogen Receptors. J Neurosci 2019; 39:2184-2194. [PMID: 30696729 DOI: 10.1523/jneurosci.1530-18.2019] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 01/17/2019] [Accepted: 01/20/2019] [Indexed: 11/21/2022] Open
Abstract
A significant unmet need for patients with multiple sclerosis (MS) is the lack of U.S. Food and Drug Administration (FDA)-approved remyelinating therapies. We have identified a compelling remyelinating agent, bazedoxifene (BZA), a European Medicines Agency (EMA)-approved (and FDA-approved in combination with conjugated estrogens) selective estrogen receptor (ER) modulator (SERM) that could move quickly from bench to bedside. This therapy stands out as a tolerable alternative to previously identified remyelinating agents and other candidates within this family. Using an unbiased high-throughput screen, with subsequent validation in both murine and human oligodendrocyte precursor cells (OPCs) and coculture systems, we find that BZA enhances differentiation of OPCs into functional oligodendrocytes. Using an in vivo murine model of focal demyelination, we find that BZA enhances OPC differentiation and remyelination. Of critical importance, we find that BZA acts independently of its presumed target, the ER, in both in vitro and in vivo systems. Using a massive computational data integration approach, we independently identify six possible candidate targets through which SERMs may mediate their effect on remyelination. Of particular interest, we identify EBP (encoding 3β-hydroxysteroid-Δ8,Δ7-isomerase), a key enzyme in the cholesterol biosynthesis pathway, which was previously implicated as a target for remyelination. These findings provide valuable insights into the implications for SERMs in remyelination for MS and hormonal research at large.SIGNIFICANCE STATEMENT Therapeutics targeted at remyelination failure, which results in axonal degeneration and ultimately disease progression, represent a large unmet need in the multiple sclerosis (MS) population. Here, we have validated a tolerable European Medicines Agency-approved (U.S. Food and Drug Administration-approved in combination with conjugated estrogens) selective estrogen receptor (ER) modulator (SERM), bazedoxifene (BZA), as a potent agent of oligodendrocyte precursor cell (OPC) differentiation and remyelination. SERMs, which were developed as nuclear ER-α and ER-β agonists/antagonists, have previously been implicated in remyelination and neuroprotection, following a heavy focus on estrogens with underwhelming and conflicting results. We show that nuclear ERs are not required for SERMs to mediate their potent effects on OPC differentiation and remyelination in vivo and highlight EBP, an enzyme in the cholesterol biosynthesis pathway that could potentially act as a target for SERMs.
Collapse
|
39
|
Jahanbazi Jahan-Abad A, Karima S, Sahab Negah S, Noorbakhsh F, Borhani-Haghighi M, Gorji A. Therapeutic potential of conditioned medium derived from oligodendrocytes cultured in a self-assembling peptide nanoscaffold in experimental autoimmune encephalomyelitis. Brain Res 2019; 1711:226-235. [PMID: 30703369 DOI: 10.1016/j.brainres.2019.01.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/26/2019] [Indexed: 01/12/2023]
Abstract
The use of neurotrophic factors is considered to be a novel therapeutic approach for restoring and/or maintaining neurological function in neurodegenerative disorders, such as multiple sclerosis (MS). Various studies have shown that conditioned medium produced by oligodendrocyte (OL-CM) contain a variety of neurotrophic factors. Here, we investigated the restorative effects of OL-CM, collected from oligodendrocytes cultured in a self-assembling peptide hydrogels scaffold (PuraMatrix), in experimental autoimmune encephalomyelitis (EAE) mouse model. Neural stem/progenitor cells, isolated from the embryonic mouse brain, were cultured and differentiated into oligodendrocyte. Cell viability and proliferation of oligodendrocytes were assessed by live/dead and MTT assays. Motor functions, myelination, cell infiltration, gliosis, and inflammatory process were assessed in EAE mice after intracranial injection of OL-CM at different concentrations. Application of OL-CM improved clinical score and neurological function in EAE mice and reduced the inflammatory cell infiltration and demyelination. Furthermore, administration of OL-CM reduced the expression of pro-inflammatory cytokines and suppressed the activation of NLRP3-inflammasome complex in EAE mice. These data suggest the potential therapeutic effect of OL-CM for MS treatment.
Collapse
Affiliation(s)
- Ali Jahanbazi Jahan-Abad
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Sajad Sahab Negah
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, Building No. 7, School of Medicine, Tehran University of Medical Sciences, Poursina Avenue, Tehran, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Ali Gorji
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran; Department of Neurosurgery, Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany; Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany.
| |
Collapse
|
40
|
Morillo Verdugo R, Ramírez Herráiz E, Fernández-Del Olmo R, Roig Bonet M, Valdivia García M. Adherence to disease-modifying treatments in patients with multiple sclerosis in Spain. Patient Prefer Adherence 2019; 13:261-272. [PMID: 30863016 PMCID: PMC6388740 DOI: 10.2147/ppa.s187983] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PURPOSE Adherence to disease-modifying treatments is essential in order to maximize the beneficial effects of treatment for multiple sclerosis (MS). There are numerous treatments that have been approved. Treatment selection is essential in patient adherence. In addition, patient preference plays an increasingly significant role in treatment decision-making. This study aims to evaluate the degree of adherence, along with other variables that may influence this adherence, in Spain. METHODS A cross-sectional study was conducted with 157 MS patients with disease-modifying treatments. Adherence was assessed using the Morisky Green scale, and other related factors were measured using a questionnaire that addressed demographics, disease characteristics, global perception of pathology, impact of medication on patient's life, and treatment decision-making. RESULTS The adherence rate was 71% and was associated with the following variables: older age, more treatments received, time to diagnosis 5-10 years, absence of exacerbations, better cognitive status, being married/in a union, clear information about the disease, and higher treatment satisfaction. The main cause for non-compliance was forgetfulness (27%). CONCLUSION The adherence rate is acceptable. It is widely known that treatment satisfaction is related to adherence. In our study, patients' level of satisfaction was higher with oral treatments. However, oral administration showed a greater lack of adherence. The main cause of lack of adherence was forgetfulness. In relation to other variables, cognitive status and family support showed a correlation with treatment adherence.
Collapse
Affiliation(s)
| | | | | | - Montserrat Roig Bonet
- Project Department, Technical Advisory of Projects and Innovation "Esclerosis Múltiple España", Madrid, Spain
| | | |
Collapse
|
41
|
Zheng W, Li Q, Zhao C, Da Y, Zhang HL, Chen Z. Differentiation of Glial Cells From hiPSCs: Potential Applications in Neurological Diseases and Cell Replacement Therapy. Front Cell Neurosci 2018; 12:239. [PMID: 30140204 PMCID: PMC6094089 DOI: 10.3389/fncel.2018.00239] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/17/2018] [Indexed: 12/20/2022] Open
Abstract
Glial cells are the most abundant cell type in the central nervous system (CNS) and play essential roles in maintaining brain homeostasis, forming myelin, and providing support and protection for neurons, etc. Over the past decade, significant progress has been made in the reprogramming field. Given the limited accessibility of human glial cells, in vitro differentiation of human induced pluripotent stem cells (hiPSCs) into glia may provide not only a valuable research tool for a better understanding of the functions of glia in the CNS but also a potential cellular source for clinical therapeutic purposes. In this review, we will summarize up-to-date novel strategies for the committed differentiation into the three major glial cell types, i.e., astrocyte, oligodendrocyte, and microglia, from hiPSCs, focusing on the non-neuronal cell effects on the pathology of some representative neurological diseases. Furthermore, the application of hiPSC-derived glial cells in neurological disease modeling will be discussed, so as to gain further insights into the development of new therapeutic targets for treatment of neurological disorders.
Collapse
Affiliation(s)
- Wei Zheng
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, China
| | - Qian Li
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, China
| | - Chao Zhao
- Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Yuwei Da
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, Beijing, China
| | - Zhiguo Chen
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
42
|
De Angelis F, Plantone D, Chataway J. Pharmacotherapy in Secondary Progressive Multiple Sclerosis: An Overview. CNS Drugs 2018; 32:499-526. [PMID: 29968175 DOI: 10.1007/s40263-018-0538-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis is an immune-mediated inflammatory disease of the central nervous system characterised by demyelination, neuroaxonal loss and a heterogeneous clinical course. Multiple sclerosis presents with different phenotypes, most commonly a relapsing-remitting course and, less frequently, a progressive accumulation of disability from disease onset (primary progressive multiple sclerosis). The majority of people with relapsing-remitting multiple sclerosis, after a variable time, switch to a stage characterised by gradual neurological worsening known as secondary progressive multiple sclerosis. We have a limited understanding of the mechanisms underlying multiple sclerosis, and it is believed that multiple genetic, environmental and endogenous factors are elements driving inflammation and ultimately neurodegeneration. Axonal loss and grey matter damage have been regarded as amongst the leading causes of irreversible neurological disability in the progressive stages. There are over a dozen disease-modifying therapies currently licenced for relapsing-remitting multiple sclerosis, but none of these has provided evidence of effectiveness in secondary progressive multiple sclerosis. Recently, there has been some early modest success with siponimod in secondary progressive multiple sclerosis and ocrelizumab in primary progressive multiple sclerosis. Finding treatments to delay or prevent the courses of secondary progressive multiple sclerosis is an unmet and essential goal of the research in multiple sclerosis. In this review, we discuss new findings regarding drugs with immunomodulatory, neuroprotective or regenerative properties and possible treatment strategies for secondary progressive multiple sclerosis. We examine the field broadly to include trials where participants have progressive or relapsing phenotypes. We summarise the most relevant results from newer investigations from phase II and III randomised controlled trials over the past decade, with particular attention to the last 5 years.
Collapse
Affiliation(s)
- Floriana De Angelis
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK.
| | - Domenico Plantone
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
| | - Jeremy Chataway
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
| |
Collapse
|
43
|
Pérez Sánchez S, Eichau Madueño S, Rus Hidalgo M, Domínguez Mayoral AM, Vilches-Arenas A, Navarro Mascarell G, Izquierdo G. Usefulness of optic nerve ultrasound to predict clinical progression in multiple sclerosis. Neurologia 2018; 36:209-214. [PMID: 29573900 DOI: 10.1016/j.nrl.2017.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 12/17/2017] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Progressive neuronal and axonal loss are considered the main causes of disability in patients with multiple sclerosis (MS). The disease frequently involves the visual system; the accessibility of the system for several functional and structural tests has made it a model for the in vivo study of MS pathogenesis. Orbital ultrasound is a non-invasive technique that enables various structures of the orbit, including the optic nerve, to be evaluated in real time. MATERIAL AND METHODS We conducted an observational, ambispective study of MS patients. Disease progression data were collected. Orbital ultrasound was performed on all patients, with power set according to the 'as low as reasonably achievable' (ALARA) principle. Optical coherence tomography (OCT) data were also collected for those patients who underwent the procedure. Statistical analysis was conducted using SPSS version 22.0. RESULTS Disease progression was significantly correlated with ultrasound findings (P=.041 for the right eye and P=.037 for the left eye) and with Expanded Disability Status Scale (EDSS) score at the end of the follow-up period (P=.07 for the right eye and P=.043 for the left eye). No statistically significant differences were found with relation to relapses or other clinical variables. DISCUSSION Ultrasound measurement of optic nerve diameter constitutes a useful, predictive factor for the evaluation of patients with MS. Smaller diameters are associated with poor clinical progression and greater disability (measured by EDSS).
Collapse
Affiliation(s)
- S Pérez Sánchez
- Servicio de Neurología, Hospital Universitario Virgen Macarena, Sevilla, España.
| | - S Eichau Madueño
- Servicio de Neurología, Hospital Universitario Virgen Macarena, Sevilla, España
| | - M Rus Hidalgo
- Servicio de Neurología, Hospital Universitario Virgen Macarena, Sevilla, España
| | | | - A Vilches-Arenas
- Servicio de Medicina Preventiva, Hospital Universitario Virgen Macarena, Sevilla, España
| | - G Navarro Mascarell
- Servicio de Neurología, Hospital Universitario Virgen Macarena, Sevilla, España
| | - G Izquierdo
- Servicio de Neurología, Hospital Universitario Virgen Macarena, Sevilla, España
| |
Collapse
|
44
|
Yukitake M. Drug-induced progressive multifocal leukoencephalopathy in multiple sclerosis: A comprehensive review. ACTA ACUST UNITED AC 2018. [DOI: 10.1111/cen3.12440] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Motohiro Yukitake
- Division of Neurology; Japan Community Health Care Organization; Saga Central Hospital; Saga Japan
| |
Collapse
|
45
|
JC Virus-DNA Detection Is Associated with CD8 Effector Accumulation in Peripheral Blood of Patients with Multiple Sclerosis under Natalizumab Treatment, Independently from JC Virus Serostatus. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5297980. [PMID: 29682547 PMCID: PMC5848061 DOI: 10.1155/2018/5297980] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 01/29/2018] [Indexed: 11/18/2022]
Abstract
Although natalizumab (anti-α4 integrin) represents an effective therapy for relapsing remitting multiple sclerosis (RRMS), it is associated with an increased risk of developing progressive multifocal leukoencephalopathy (PML), caused by the polyomavirus JC (JCV). The aim of this study was to explore natalizumab-induced phenotypic changes in peripheral blood T-lymphocytes and their relationship with JCV reactivation. Forty-four patients affected by RRMS were enrolled. Blood and urine samples were classified according to natalizumab infusion number: 0 (N0), 1–12 (N12), 13–24 (N24), 25–36 (N36), and over 36 (N > 36) infusions. JCV-DNA was detected in plasma and urine. T-lymphocyte phenotype was evaluated with flow cytometry. JCV serostatus was assessed. Ten healthy donors (HD), whose ages and sexes matched with the RRMS patients of the N0 group, were enrolled. CD8 effector (CD8 E) percentages were increased in natalizumab treated patients with detectable JCV-DNA in plasma or urine compared to JCV-DNA negative patients (JCV−) (p < 0.01 and p < 0.001, resp.). Patients with CD8 E percentages above 10.4% tended to show detectable JCV-DNA in plasma and/or urine (ROC curve p = 0.001). The CD8 E was increased when JCV-DNA was detectable in plasma or urine, independently from JCV serology, for N12 and N24 groups (p < 0.01). As long as PML can affect RRMS patients under natalizumab treatment with a negative JCV serology, the assessment of CD8 E could help in the evaluation of JCV reactivation.
Collapse
|
46
|
Affiliation(s)
- Yanan Chen
- Department of Neurology, Center for Peripheral Neuropathy, University of Chicago, Chicago, IL 60637, USA
| | - Brian Popko
- Department of Neurology, Center for Peripheral Neuropathy, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
47
|
Baecher-Allan C, Kaskow BJ, Weiner HL. Multiple Sclerosis: Mechanisms and Immunotherapy. Neuron 2018; 97:742-768. [DOI: 10.1016/j.neuron.2018.01.021] [Citation(s) in RCA: 432] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/25/2017] [Accepted: 01/09/2018] [Indexed: 12/17/2022]
|
48
|
Laquinimod ameliorates excitotoxic damage by regulating glutamate re-uptake. J Neuroinflammation 2018; 15:5. [PMID: 29304807 PMCID: PMC5756343 DOI: 10.1186/s12974-017-1048-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/21/2017] [Indexed: 11/10/2022] Open
Abstract
Background Laquinimod is an immunomodulatory drug under clinical investigation for the treatment of the progressive form of multiple sclerosis (MS) with both anti-inflammatory and neuroprotective effects. Excitotoxicity, a prominent pathophysiological feature of MS and of its animal model, experimental autoimmune encephalomyelitis (EAE), involves glutamate transporter (GluT) dysfunction in glial cells. The aim of this study was to assess whether laquinimod might exert direct neuroprotective effects by interfering with the mechanisms of excitotoxicity linked to GluT function impairments in EAE. Methods Osmotic minipumps allowing continuous intracerebroventricular (icv) infusion of laquinimod for 4 weeks were implanted into C57BL/6 mice before EAE induction. EAE cerebella were taken to perform western blot and qPCR experiments. For ex vivo experiments, EAE cerebellar slices were incubated with laquinimod before performing electrophysiology, western blot, and qPCR. Results In vivo treatment with laquinimod attenuated EAE clinical score at the peak of the disease, without remarkable effects on inflammatory markers. In vitro application of laquinimod to EAE cerebellar slices prevented EAE-linked glutamatergic alterations without mitigating astrogliosis and inflammation. Moreover, such treatment induced an increase of Slcla3 mRNA coding for the glial glutamate–aspartate transporter (GLAST) without affecting the protein content. Concomitantly, laquinimod significantly increased the levels of the glial glutamate transporter 1 (GLT-1) protein and pharmacological blockade of GLT-1 function fully abolished laquinimod anti-excitotoxic effect. Conclusions Overall, our results suggest that laquinimod protects against glutamate excitotoxicity of the cerebellum of EAE mice by bursting the expression of glial glutamate transporters, independently of its anti-inflammatory effects.
Collapse
|
49
|
Gaissmaier W, Giese H, Galesic M, Garcia-Retamero R, Kasper J, Kleiter I, Meuth SG, Köpke S, Heesen C. Numeracy of multiple sclerosis patients: A comparison of patients from the PERCEPT study to a German probabilistic sample. PATIENT EDUCATION AND COUNSELING 2018; 101:74-78. [PMID: 28764895 DOI: 10.1016/j.pec.2017.07.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/05/2017] [Accepted: 07/14/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE A shared decision-making approach is suggested for multiple sclerosis (MS) patients. To properly evaluate benefits and risks of different treatment options accordingly, MS patients require sufficient numeracy - the ability to understand quantitative information. It is unknown whether MS affects numeracy. Therefore, we investigated whether patients' numeracy was impaired compared to a probabilistic national sample. METHODS As part of the larger prospective, observational, multicenter study PERCEPT, we assessed numeracy for a clinical study sample of German MS patients (N=725) with a standard test and compared them to a German probabilistic sample (N=1001), controlling for age, sex, and education. Within patients, we assessed whether disease variables (disease duration, disability, annual relapse rate, cognitive impairment) predicted numeracy beyond these demographics. RESULTS MS patients showed a comparable level of numeracy as the probabilistic national sample (68.9% vs. 68.5% correct answers, P=0.831). In both samples, numeracy was higher for men and the highly educated. Disease variables did not predict numeracy beyond demographics within patients, and predictability was generally low. CONCLUSION This sample of MS patients understood quantitative information on the same level as the general population. PRACTICE IMPLICATIONS There is no reason to withhold quantitative information from MS patients.
Collapse
Affiliation(s)
- Wolfgang Gaissmaier
- Department of Psychology, University of Konstanz, Konstanz, Germany; Center for Adaptive Behavior and Cognition, Max Planck Institute for Human Development, Berlin, Germany.
| | - Helge Giese
- Department of Psychology, University of Konstanz, Konstanz, Germany
| | | | | | - Juergen Kasper
- Department of Health and Care Sciences, University of Tromsø, Tromsø, Norway
| | - Ingo Kleiter
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Sven G Meuth
- Department of Neurology, University of Münster, Münster, Germany
| | - Sascha Köpke
- Institute for Social Medicine and Epidemiology, University of Lübeck, Lübeck, Germany
| | - Christoph Heesen
- Institute for Neuroimmunology and Clinical MS Research and Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
50
|
Green AJ, Gelfand JM, Cree BA, Bevan C, Boscardin WJ, Mei F, Inman J, Arnow S, Devereux M, Abounasr A, Nobuta H, Zhu A, Friessen M, Gerona R, von Büdingen HC, Henry RG, Hauser SL, Chan JR. Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): a randomised, controlled, double-blind, crossover trial. Lancet 2017; 390:2481-2489. [PMID: 29029896 DOI: 10.1016/s0140-6736(17)32346-2] [Citation(s) in RCA: 394] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 07/10/2017] [Accepted: 08/06/2017] [Indexed: 01/24/2023]
Abstract
BACKGROUND Multiple sclerosis is a degenerative inflammatory disease of the CNS characterised by immune-mediated destruction of myelin and progressive neuroaxonal loss. Myelin in the CNS is a specialised extension of the oligodendrocyte plasma membrane and clemastine fumarate can stimulate differentiation of oligodendrocyte precursor cells in vitro, in animal models, and in human cells. We aimed to analyse the efficacy and safety of clemastine fumarate as a treatment for patients with multiple sclerosis. METHODS We did this single-centre, 150-day, double-blind, randomised, placebo-controlled, crossover trial (ReBUILD) in patients with relapsing multiple sclerosis with chronic demyelinating optic neuropathy on stable immunomodulatory therapy. Patients who fulfilled international panel criteria for diagnosis with disease duration of less than 15 years were eligible. Patients were randomly assigned (1:1) via block randomisation using a random number generator to receive either clemastine fumarate (5·36 mg orally twice daily) for 90 days followed by placebo for 60 days (group 1), or placebo for 90 days followed by clemastine fumarate (5·36 mg orally twice daily) for 60 days (group 2). The primary outcome was shortening of P100 latency delay on full-field, pattern-reversal, visual-evoked potentials. We analysed by intention to treat. The trial is registered with ClinicalTrials.gov, number NCT02040298. FINDINGS Between Jan 1, 2014, and April 11, 2015, we randomly assigned 50 patients to group 1 (n=25) or group 2 (n=25). All patients completed the study. The primary efficacy endpoint was met with clemastine fumarate treatment, which reduced the latency delay by 1·7 ms/eye (95% CI 0·5-2·9; p=0·0048) when analysing the trial as a crossover. Clemastine fumarate treatment was associated with fatigue, but no serious adverse events were reported. INTERPRETATION To our knowledge, this is the first randomised controlled trial to document efficacy of a remyelinating drug for the treatment of chronic demyelinating injury in multiple sclerosis. Our findings suggest that myelin repair can be achieved even following prolonged damage. FUNDING University of California, San Francisco and the Rachleff Family.
Collapse
Affiliation(s)
- Ari J Green
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA.
| | - Jeffrey M Gelfand
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce A Cree
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Carolyn Bevan
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - W John Boscardin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Feng Mei
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Program in Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Justin Inman
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Sam Arnow
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Michael Devereux
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Aya Abounasr
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Hiroko Nobuta
- Department of Pediatrics and Neurosurgery and Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Alyssa Zhu
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Matt Friessen
- Department of Obstetrics and Gynecology, University of California, San Francisco, San Francisco, CA, USA
| | - Roy Gerona
- Department of Obstetrics and Gynecology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Roland G Henry
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA; Bioengineering Graduate Group, University of California, Berkeley, and San Francisco, San Francisco, CA, USA
| | - Stephen L Hauser
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Program in Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Jonah R Chan
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Program in Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|