1
|
Zhang X, Song S, Chen B, Leocani L, Zhao X, Zhong Y, Pisa M, Sharma SM. The Effects of Disease-Modifying Therapies on Optic Nerve Degeneration in Multiple Sclerosis. Eur J Neurol 2025; 32:e70081. [PMID: 40047132 PMCID: PMC11883414 DOI: 10.1111/ene.70081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/30/2024] [Accepted: 01/27/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND Retinal nerve fiber layer (RNFL) and ganglion cell-inner plexiform layer (GCIPL) thinning are used as markers of subclinical retinal degeneration to evaluate the effect of disease-modifying therapies (DMTs) on disease progression in clinical trials of multiple sclerosis (MS). This study aimed to assess the available evidence regarding the effects of DMTs on retinal thinning in people with MS. METHODS Databases were searched for studies reporting longitudinal optical coherence tomography (OCT)-derived annualized RNFL and GCIPL thinning in patients receiving DMTs treatment. The standardized mean differences (Hedges g) of RNFL and GCIPL thickness between the baseline and follow-up were used as the primary effect size measure. DMTs were divided into moderate (M-DMTs) and high (H-DMTs) efficacy therapies. RESULTS Twenty-one studies including 2158 patients and 3685 eyes were included. Overall, significant annualized RNFL (g = -0.6715, p = 0.0077) and GCIPL (g = -0.31, p < 0.0001) thinning was observed at follow-up compared with baseline. Annualized RNFL thinning was only significant in the M-DMTs group (g = -0.6992, p = 0.0243). Annualized GCIPL thinning was significant in both M-DMTs (g = -0.38, p = 0.0006) and H-DMTs group (g = -0.19, p < 0.0001) but was significantly greater in the M-DMTs group compared with the H-DMTs group (g = -0.20, p = 0.0017). There was no difference in annualized GCIPL or RNFL thinning between RRMS and PMS, or between RRMS with and without ON history. CONCLUSIONS High-DMTs are more effective in reducing longitudinal thinning of RNFL and GCIPL compared with M-DMTs. GCIPL thinning could serve as a sensitive predictor for the surveillance of optic nerve degeneration and the assessment of DMT efficacy for both RRMS and PMS.
Collapse
Affiliation(s)
- Xia Zhang
- Department of OphthalmologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of OphthalmologyOxford University Hospitals NHS TrustOxfordUK
| | - Shuang Song
- Department of Biostatistics, Harvard T.H. Chan School of Public HealthHarvard UniversityBostonMassachusettsUSA
| | - Bo Chen
- Department of Neurology, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Nuffield Department of Clinical NeuroscienceUniversity of OxfordOxfordUK
| | - Letizia Leocani
- Neurological Department and Institute of Experimental Neurology (INSPE)Scientific Institute, Hospital San RaffaeleMilanItaly
| | - Xinyu Zhao
- Department of OphthalmologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yong Zhong
- Department of OphthalmologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Marco Pisa
- Nuffield Department of Clinical NeuroscienceUniversity of OxfordOxfordUK
| | | |
Collapse
|
2
|
Charmarke-Askar I, Spenlé C, Bagnard D. Complementary strategies to be used in conjunction with animal models for multiple sclerosis drug discovery: adapting preclinical validation of drug candidates to the need of remyelinating strategies. Expert Opin Drug Discov 2024; 19:1115-1124. [PMID: 39039755 DOI: 10.1080/17460441.2024.2382180] [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: 02/21/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION The quest for novel MS therapies focuses on promoting remyelination and neuroprotection, necessitating innovative drug design paradigms and robust preclinical validation methods to ensure efficient clinical translation. The complexity of new drugs action mechanisms is strengthening the need for solid biological validation attempting to address all possible pitfalls and biases precluding access to efficient and safe drugs. AREAS COVERED In this review, the authors describe the different in vitro and in vivo models that should be used to create an integrated approach for preclinical validation of novel drugs, including the evaluation of the action mechanism. This encompasses 2D, 3D in vitro models and animal models presented in such a way to define the appropriate use in a global process of drug screening and hit validation. EXPERT OPINION None of the current available tests allow the concomitant evaluation of anti-inflammatory, immune regulators or remyelinating agents with sufficient reliability. Consequently, the collaborative efforts of academia, industry, and regulatory agencies are essential for establishing standardized protocols, validating novel methodologies, and translating preclinical findings into clinically meaningful outcomes.
Collapse
|
3
|
Horng DE. Editorial for "Longitudinal Metabolite Changes in Progressive Multiple Sclerosis: A Study of 3 Potential Neuroprotective Treatments". J Magn Reson Imaging 2024; 59:2202-2203. [PMID: 37737006 DOI: 10.1002/jmri.29018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 09/23/2023] Open
Affiliation(s)
- Debra E Horng
- Deb Horng Consulting LLC, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Collongues N, Becker G, Jolivel V, Ayme-Dietrich E, de Seze J, Binamé F, Patte-Mensah C, Monassier L, Mensah-Nyagan AG. A Narrative Review on Axonal Neuroprotection in Multiple Sclerosis. Neurol Ther 2022; 11:981-1042. [PMID: 35610531 PMCID: PMC9338208 DOI: 10.1007/s40120-022-00363-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/03/2022] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) resulting in demyelination and neurodegeneration. The therapeutic strategy is now largely based on reducing inflammation with immunosuppressive drugs. Unfortunately, when disease progression is observed, no drug offers neuroprotection apart from its anti-inflammatory effect. In this review, we explore current knowledge on the assessment of neurodegeneration in MS and look at putative targets that might prove useful in protecting the axon from degeneration. Among them, Bruton's tyrosine kinase inhibitors, anti-apoptotic and antioxidant agents, sex hormones, statins, channel blockers, growth factors, and molecules preventing glutamate excitotoxicity have already been studied. Some of them have reached phase III clinical trials and carry a great message of hope for our patients with MS.
Collapse
Affiliation(s)
- Nicolas Collongues
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France. .,Center for Clinical Investigation, INSERM U1434, Strasbourg, France. .,Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France. .,University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.
| | - Guillaume Becker
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Valérie Jolivel
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Estelle Ayme-Dietrich
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Jérôme de Seze
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France.,Center for Clinical Investigation, INSERM U1434, Strasbourg, France.,Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Fabien Binamé
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Christine Patte-Mensah
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Laurent Monassier
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Ayikoé Guy Mensah-Nyagan
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| |
Collapse
|
5
|
Rupnik M, Baker D, Selwood DL. Oligodendrocytes, BK channels and remyelination. F1000Res 2021; 10:781. [PMID: 34909188 PMCID: PMC8596180 DOI: 10.12688/f1000research.53422.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 11/09/2023] Open
Abstract
Oligodendrocytes wrap multiple lamellae of their membrane, myelin, around axons of the central nervous system (CNS), to improve impulse conduction. Myelin synthesis is specialised and dynamic, responsive to local neuronal excitation. Subtle pathological insults are sufficient to cause significant neuronal metabolic impairment, so myelin preservation is necessary to safeguard neural networks. Multiple sclerosis (MS) is the most prevalent demyelinating disease of the CNS. In MS, inflammatory attacks against myelin, proposed to be autoimmune, cause myelin decay and oligodendrocyte loss, leaving neurons vulnerable. Current therapies target the prominent neuroinflammation but are mostly ineffective in protecting from neurodegeneration and the progressive neurological disability. People with MS have substantially higher levels of extracellular glutamate, the main excitatory neurotransmitter. This impairs cellular homeostasis to cause excitotoxic stress. Large conductance Ca2 +-activated K + channels (BK channels) could preserve myelin or allow its recovery by protecting cells from the resulting excessive excitability. This review evaluates the role of excitotoxic stress, myelination and BK channels in MS pathology, and explores the hypothesis that BK channel activation could be a therapeutic strategy to protect oligodendrocytes from excitotoxic stress in MS. This could reduce progression of neurological disability if used in parallel to immunomodulatory therapies.
Collapse
Affiliation(s)
- Maddalena Rupnik
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| | - David Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, WC1E 6BT, UK
| | - David L. Selwood
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| |
Collapse
|
6
|
Rupnik M, Baker D, Selwood DL. Oligodendrocytes, BK channels and the preservation of myelin. F1000Res 2021; 10:781. [PMID: 34909188 PMCID: PMC8596180 DOI: 10.12688/f1000research.53422.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2021] [Indexed: 11/20/2022] Open
Abstract
Oligodendrocytes wrap multiple lamellae of their membrane, myelin, around axons of the central nervous system (CNS), to improve impulse conduction. Myelin synthesis is specialised and dynamic, responsive to local neuronal excitation. Subtle pathological insults are sufficient to cause significant neuronal metabolic impairment, so myelin preservation is necessary to safeguard neural networks. Multiple sclerosis (MS) is the most prevalent demyelinating disease of the CNS. In MS, inflammatory attacks against myelin, proposed to be autoimmune, cause myelin decay and oligodendrocyte loss, leaving neurons vulnerable. Current therapies target the prominent neuroinflammation but are mostly ineffective in protecting from neurodegeneration and the progressive neurological disability. People with MS have substantially higher levels of extracellular glutamate, the main excitatory neurotransmitter. This impairs cellular homeostasis to cause excitotoxic stress. Large conductance Ca2 +-activated K + channels (BK channels) could preserve myelin or allow its recovery by protecting cells from the resulting excessive excitability. This review evaluates the role of excitotoxic stress, myelination and BK channels in MS pathology, and explores the hypothesis that BK channel activation could be a therapeutic strategy to protect oligodendrocytes from excitotoxic stress in MS. This could reduce progression of neurological disability if used in parallel to immunomodulatory therapies.
Collapse
Affiliation(s)
- Maddalena Rupnik
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| | - David Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, WC1E 6BT, UK
| | - David L. Selwood
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| |
Collapse
|
7
|
Failed, Interrupted, or Inconclusive Trials on Neuroprotective and Neuroregenerative Treatment Strategies in Multiple Sclerosis: Update 2015-2020. Drugs 2021; 81:1031-1063. [PMID: 34086251 PMCID: PMC8217012 DOI: 10.1007/s40265-021-01526-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
In the recent past, a plethora of drugs have been approved for the treatment of multiple sclerosis (MS). These therapeutics are mainly confined to immunomodulatory or immunosuppressive strategies but do not sufficiently address remyelination and neuroprotection. However, several neuroregenerative agents have shown potential in pre-clinical research and entered Phase I to III clinical trials. Although none of these compounds have yet proceeded to approval, understanding the causes of failure can broaden our knowledge about neuroprotection and neuroregeneration in MS. Moreover, most of the investigated approaches are characterised by consistent mechanisms of action and proved convincing efficacy in animal studies. Therefore, learning from their failure will help us to enforce the translation of findings acquired in pre-clinical studies into clinical application. Here, we summarise trials on MS treatment published since 2015 that have either failed or were interrupted due to a lack of efficacy, adverse events, or for other reasons. We further outline the rationale underlying these drugs and analyse the background of failure to gather new insights into MS pathophysiology and optimise future study designs. For conciseness, this review focuses on agents promoting remyelination and medications with primarily neuroprotective properties or unconventional approaches. Failed clinical trials that pursue immunomodulation are presented in a separate article.
Collapse
|
8
|
Lisak RP, Nedelkoska L, Benjamins JA. Sigma-1 receptor agonists as potential protective therapies in multiple sclerosis. J Neuroimmunol 2020; 342:577188. [PMID: 32179326 DOI: 10.1016/j.jneuroim.2020.577188] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/17/2020] [Accepted: 02/07/2020] [Indexed: 01/25/2023]
Abstract
The sigma-1 receptor (σ-1R) is an endoplasmic reticulum (ER) chaperone upregulated during ER stress, and regulates calcium homeostasis. Agonists of σ-1R are neuroprotective. ANAVEX2-73, a new σ-1R agonist, is undergoing several clinical trials. We show that ANAVEX2-73 protects oligodendroglia (OL) and oligodendroglial precursors (OPC) from apoptosis, excitotoxicity, reactive oxygen species (ROS) and quinolinic acid (QA), associated with inflammation. ANAVEX2-73 stimulates OPC proliferation, but does not alter early maturation to OL. We previously reported that dextromethorphan (DM), another σ-1R agonist with a different structure, had similar effects. We now show that both DM and ANAVEX2-73 protect neurons from the four cytotoxic agents.
Collapse
Affiliation(s)
- Robert P Lisak
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Liljana Nedelkoska
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Joyce A Benjamins
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
9
|
Efficacy of three neuroprotective drugs in secondary progressive multiple sclerosis (MS-SMART): a phase 2b, multiarm, double-blind, randomised placebo-controlled trial. Lancet Neurol 2020; 19:214-225. [PMID: 31981516 PMCID: PMC7029307 DOI: 10.1016/s1474-4422(19)30485-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/20/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
Background Neurodegeneration is the pathological substrate that causes major disability in secondary progressive multiple sclerosis. A synthesis of preclinical and clinical research identified three neuroprotective drugs acting on different axonal pathobiologies. We aimed to test the efficacy of these drugs in an efficient manner with respect to time, cost, and patient resource. Methods We did a phase 2b, multiarm, parallel group, double-blind, randomised placebo-controlled trial at 13 clinical neuroscience centres in the UK. We recruited patients (aged 25–65 years) with secondary progressive multiple sclerosis who were not on disease-modifying treatment and who had an Expanded Disability Status Scale (EDSS) score of 4·0–6·5. Participants were randomly assigned (1:1:1:1) at baseline, by a research nurse using a centralised web-based service, to receive twice-daily oral treatment of either amiloride 5 mg, fluoxetine 20 mg, riluzole 50 mg, or placebo for 96 weeks. The randomisation procedure included minimisation based on sex, age, EDSS score at randomisation, and trial site. Capsules were identical in appearance to achieve masking. Patients, investigators, and MRI readers were unaware of treatment allocation. The primary outcome measure was volumetric MRI percentage brain volume change (PBVC) from baseline to 96 weeks, analysed using multiple regression, adjusting for baseline normalised brain volume and minimisation criteria. The primary analysis was a complete-case analysis based on the intention-to-treat population (all patients with data at week 96). This trial is registered with ClinicalTrials.gov, NCT01910259. Findings Between Jan 29, 2015, and June 22, 2016, 445 patients were randomly allocated amiloride (n=111), fluoxetine (n=111), riluzole (n=111), or placebo (n=112). The primary analysis included 393 patients who were allocated amiloride (n=99), fluoxetine (n=96), riluzole (n=99), and placebo (n=99). No difference was noted between any active treatment and placebo in PBVC (amiloride vs placebo, 0·0% [95% CI −0·4 to 0·5; p=0·99]; fluoxetine vs placebo −0·1% [–0·5 to 0·3; p=0·86]; riluzole vs placebo −0·1% [–0·6 to 0·3; p=0·77]). No emergent safety issues were reported. The incidence of serious adverse events was low and similar across study groups (ten [9%] patients in the amiloride group, seven [6%] in the fluoxetine group, 12 [11%] in the riluzole group, and 13 [12%] in the placebo group). The most common serious adverse events were infections and infestations. Three patients died during the study, from causes judged unrelated to active treatment; one patient assigned amiloride died from metastatic lung cancer, one patient assigned riluzole died from ischaemic heart disease and coronary artery thrombosis, and one patient assigned fluoxetine had a sudden death (primary cause) with multiple sclerosis and obesity listed as secondary causes. Interpretation The absence of evidence for neuroprotection in this adequately powered trial indicates that exclusively targeting these aspects of axonal pathobiology in patients with secondary progressive multiple sclerosis is insufficient to mitigate neuroaxonal loss. These findings argue for investigation of different mechanistic targets and future consideration of combination treatment trials. This trial provides a template for future simultaneous testing of multiple disease-modifying medicines in neurological medicine. Funding Efficacy and Mechanism Evaluation (EME) Programme, an MRC and NIHR partnership, UK Multiple Sclerosis Society, and US National Multiple Sclerosis Society.
Collapse
|
10
|
Hollinger KR, Smith MD, Kirby LA, Prchalova E, Alt J, Rais R, Calabresi PA, Slusher BS. Glutamine antagonism attenuates physical and cognitive deficits in a model of MS. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:e609. [PMID: 31467038 PMCID: PMC6745721 DOI: 10.1212/nxi.0000000000000609] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/09/2019] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To measure the impact of JHU-083, a novel prodrug of the glutamine antagonist 6-diazo-5-oxo-l-norleucine, on immune cell proliferation and activation, along with physical and cognitive impairments associated with the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. METHODS Splenic-derived T cells and bone marrow-derived dendritic cells (DCs) were cultured, activated, and treated daily with vehicle or JHU-083. Proliferation and activation were measured via flow cytometry and IncuCyte live cell analysis. C57BL/6 mice were immunized for EAE. Vehicle or JHU-083 was administered orally every other day either from the time of immunization in the prevention paradigm or from the time of disease onset in the treatment paradigm. Disease scores and body weight were monitored. In the treatment paradigm, cognition was evaluated using the Barnes maze test. RESULTS JHU-083 selectively inhibits T-cell proliferation and decreases T-cell activation, with no effect on DCs. In vivo, orally administered JHU-083 significantly decreases EAE severity in both prevention and treatment paradigms and reverses EAE-induced cognitive impairment. CONCLUSIONS JHU-083, a well-tolerated, brain penetrable glutamine antagonist, is a promising novel treatment for both the physical and cognitive deficits of MS.
Collapse
Affiliation(s)
- Kristen R Hollinger
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Matthew D Smith
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Leslie A Kirby
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Eva Prchalova
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Jesse Alt
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Rana Rais
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Peter A Calabresi
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD.
| | - Barbara S Slusher
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
11
|
Lambe J, Saidha S, Bermel RA. Optical coherence tomography and multiple sclerosis: Update on clinical application and role in clinical trials. Mult Scler 2019; 26:624-639. [PMID: 32412377 DOI: 10.1177/1352458519872751] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Optical coherence tomography (OCT) has emerged as a fast, non-invasive, inexpensive, high-resolution imaging technique in multiple sclerosis (MS). Retinal layer quantification by OCT facilitates a 'window' into not only local retinal pathology but also global neurodegenerative processes, recognised to be the principal substrates of disability accumulation in MS. While OCT measures in MS have been demonstrated to reflect visual function, inflammatory activity outside of the visual pathways, disability measures including the prediction of disability progression, whole brain atrophy, and the differential neuroprotective effects of disease-modifying therapies, debate continues regarding the clinical utility of OCT in everyday practice. This review presents an overview of the evidence supporting OCT, with particular focus on its application in the MS clinic. We will also discuss the role of OCT in MS clinical trials to develop novel neuroprotective and potential remyelinating therapies.
Collapse
Affiliation(s)
- Jeffrey Lambe
- Department of Neurology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Shiv Saidha
- Department of Neurology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Robert A Bermel
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
12
|
Phillips RS, Rubin JE. Effects of persistent sodium current blockade in respiratory circuits depend on the pharmacological mechanism of action and network dynamics. PLoS Comput Biol 2019; 15:e1006938. [PMID: 31469828 PMCID: PMC6742421 DOI: 10.1371/journal.pcbi.1006938] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 09/12/2019] [Accepted: 06/15/2019] [Indexed: 02/05/2023] Open
Abstract
The mechanism(s) of action of most commonly used pharmacological blockers of voltage-gated ion channels are well understood; however, this knowledge is rarely considered when interpreting experimental data. Effects of blockade are often assumed to be equivalent, regardless of the mechanism of the blocker involved. Using computer simulations, we demonstrate that this assumption may not always be correct. We simulate the blockade of a persistent sodium current (INaP), proposed to underlie rhythm generation in pre-Bötzinger complex (pre-BötC) respiratory neurons, via two distinct pharmacological mechanisms: (1) pore obstruction mediated by tetrodotoxin and (2) altered inactivation dynamics mediated by riluzole. The reported effects of experimental application of tetrodotoxin and riluzole in respiratory circuits are diverse and seemingly contradictory and have led to considerable debate within the field as to the specific role of INaP in respiratory circuits. The results of our simulations match a wide array of experimental data spanning from the level of isolated pre-BötC neurons to the level of the intact respiratory network and also generate a series of experimentally testable predictions. Specifically, in this study we: (1) provide a mechanistic explanation for seemingly contradictory experimental results from in vitro studies of INaP block, (2) show that the effects of INaP block in in vitro preparations are not necessarily equivalent to those in more intact preparations, (3) demonstrate and explain why riluzole application may fail to effectively block INaP in the intact respiratory network, and (4) derive the prediction that effective block of INaP by low concentration tetrodotoxin will stop respiratory rhythm generation in the intact respiratory network. These simulations support a critical role for INaP in respiratory rhythmogenesis in vivo and illustrate the importance of considering mechanism when interpreting and simulating data relating to pharmacological blockade. The application of pharmacological agents that affect transmembrane ionic currents in neurons is a commonly used experimental technique. A simplistic interpretation of experiments involving these agents suggests that antagonist application removes the impacted current and that subsequently observed changes in activity are attributable to the loss of that current’s effects. The more complex reality, however, is that different drugs may have distinct mechanisms of action, some corresponding not to a removal of a current but rather to a changing of its properties. We use computational modeling to explore the implications of the distinct mechanisms associated with two drugs, riluzole and tetrodotoxin, that are often characterized as sodium channel blockers. Through this approach, we offer potential explanations for disparate findings observed in experiments on neural respiratory circuits and show that the experimental results are consistent with a key role for the persistent sodium current in respiratory rhythm generation.
Collapse
Affiliation(s)
- Ryan S. Phillips
- Department of Mathematics and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Jonathan E. Rubin
- Department of Mathematics and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
13
|
Mahmoud S, Gharagozloo M, Simard C, Gris D. Astrocytes Maintain Glutamate Homeostasis in the CNS by Controlling the Balance between Glutamate Uptake and Release. Cells 2019; 8:E184. [PMID: 30791579 PMCID: PMC6406900 DOI: 10.3390/cells8020184] [Citation(s) in RCA: 399] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 01/26/2023] Open
Abstract
Glutamate is one of the most prevalent neurotransmitters released by excitatory neurons in the central nervous system (CNS); however, residual glutamate in the extracellular space is, potentially, neurotoxic. It is now well-established that one of the fundamental functions of astrocytes is to uptake most of the synaptically-released glutamate, which optimizes neuronal functions and prevents glutamate excitotoxicity. In the CNS, glutamate clearance is mediated by glutamate uptake transporters expressed, principally, by astrocytes. Interestingly, recent studies demonstrate that extracellular glutamate stimulates Ca2+ release from the astrocytes' intracellular stores, which triggers glutamate release from astrocytes to the adjacent neurons, mostly by an exocytotic mechanism. This released glutamate is believed to coordinate neuronal firing and mediate their excitatory or inhibitory activity. Therefore, astrocytes contribute to glutamate homeostasis in the CNS, by maintaining the balance between their opposing functions of glutamate uptake and release. This dual function of astrocytes represents a potential therapeutic target for CNS diseases associated with glutamate excitotoxicity. In this regard, we summarize the molecular mechanisms of glutamate uptake and release, their regulation, and the significance of both processes in the CNS. Also, we review the main features of glutamate metabolism and glutamate excitotoxicity and its implication in CNS diseases.
Collapse
Affiliation(s)
- Shaimaa Mahmoud
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Marjan Gharagozloo
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Camille Simard
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Denis Gris
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
14
|
Rocca MA, Preziosa P, Filippi M. Application of advanced MRI techniques to monitor pharmacologic and rehabilitative treatment in multiple sclerosis: current status and future perspectives. Expert Rev Neurother 2018; 19:835-866. [PMID: 30500303 DOI: 10.1080/14737175.2019.1555038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Advances in magnetic resonance imaging (MRI) technology and analyses are improving our understanding of the pathophysiology of multiple sclerosis (MS). Due to their ability to grade the presence of irreversible tissue loss, microstructural tissue abnormalities, metabolic changes and functional plasticity, the application of these techniques is also expanding our knowledge on the efficacy and mechanisms of action of different pharmacological and rehabilitative treatments. Areas covered: This review discusses recent findings derived from the application of advanced MRI techniques to evaluate the structural and functional substrates underlying the effects of pharmacologic and rehabilitative treatments in patients with MS. Current applications as outcome in clinical trials and observational studies, their interpretation and possible pitfalls in their use are discussed. Finally, how these techniques could evolve in the future to improve monitoring of disease progression and treatment response is examined. Expert commentary: The number of treatments currently available for MS is increasing. The application of advanced MRI techniques is providing reliable and specific measures to better understand the targets of different treatments, including neuroprotection, tissue repair, and brain plasticity. This is a fundamental progress to move toward personalized medicine and individual treatment selection.
Collapse
Affiliation(s)
- Maria A Rocca
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University , Milan , Italy.,Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University , Milan , Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University , Milan , Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University , Milan , Italy.,Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University , Milan , Italy
| |
Collapse
|
15
|
LoPresti P. Silent Free Fall at Disease Onset: A Perspective on Therapeutics for Progressive Multiple Sclerosis. Front Neurol 2018; 9:973. [PMID: 30542317 PMCID: PMC6277889 DOI: 10.3389/fneur.2018.00973] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
Central nervous system (CNS) degeneration occurs during multiple sclerosis (MS) following several years of reversible autoimmune demyelination. Progressive CNS degeneration appears later during the course of relapsing-remitting MS (RRMS), although it starts insidiously at disease onset. We propose that there is an early subclinical phase also for primary-progressive (PP) MS. Consensus exists that many different cell types are involved during disease onset. Furthermore, the response to the initial damage, which is specific for each individual, would result in distinct pathological pathways that add complexity to the disease and the mechanisms underlying progressive CNS degeneration. Progressive MS is classified as either active or not active, as well as with or without progression. Different forms of progressive MS might reflect distinct or overlapping pathogenetic pathways. Disease mechanisms should be determined for each patient at diagnosis and the time of treatment. Until individualized and time-sensitive treatments that specifically target the molecular mechanisms of the progressive aspect of the disease are identified, combined therapies directed at anti-inflammation, regeneration, and neuroprotection are the most effective for preventing MS progression. This review presents selected therapeutics in support of the overall idea of a multidimensional therapy applied early in the disease. This approach could limit damage and increase CNS repair. By targeting several cellular populations (i.e., microglia, astrocytes, neurons, oligodendrocytes, and lymphocytes) and multiple pathological processes (e.g., inflammation, demyelination, synaptopathy, and excitatory/inhibitory imbalance) progressive MS could be attenuated. Early timing for such multidimensional therapy is proposed as the prerequisite for effectively halting progressive MS.
Collapse
Affiliation(s)
- Patrizia LoPresti
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
16
|
Guennoc AM, Pallix-Guyot M, Le Page E, Le Port D, Daryabin M, Hergesheimer R, Beltran S, Tourbah A, Edan G, Corcia P. Co-occurrence of MS and ALS: a clue in favor of common pathophysiological findings? Amyotroph Lateral Scler Frontotemporal Degener 2018; 19:500-505. [PMID: 30175630 DOI: 10.1080/21678421.2018.1476547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS) are two neurological disorders that seem, theoretically, completely divergent according to epidemiological, clinical, pathophysiological, and therapeutic data. However, some reports that have mentioned the occurrence of both conditions within the same patient underpin the suggestion that this co-occurrence might not be random. We report six co-occurrences of ALS and MS cases, focusing on epidemiological and clinical diseases findings. We then compare our cohort to those in the literature. Our cohort was composed of five females and one male. The age of onset for MS ranged from 27 to 54 years with either primary or secondary prominence while all being progressive. Both diseases occurred sequentially in all but one the cases. Concerning ALS, the age of onset ranged from 51 to 60 years and the site of onset was the legs in 5/6 cases. The disease lasted from four to 29 months. Although infrequent, this co-occurrence supports the hypothesis of common, pathophysiological mechanisms between ALS and MS. We discuss some arguments favoring a potential link between both conditions.
Collapse
Affiliation(s)
- Anne-Marie Guennoc
- a Neurology Department , University Hospital Center of Tours , Tours , France
| | - Maud Pallix-Guyot
- b Neurology Department , Regional Hospital Center of Orléans , Orléans , France
| | - Emmanuelle Le Page
- c Neurology Department , University Hospital Center of Rennes , Rennes , France
| | - Damien Le Port
- c Neurology Department , University Hospital Center of Rennes , Rennes , France
| | - Mathieu Daryabin
- d Neurology Department , Hospital Center of Angoulême , Angoulême , France
| | | | - Stéphane Beltran
- a Neurology Department , University Hospital Center of Tours , Tours , France.,e ALS Center, University Hospital Center of Tours , Tours , France , and
| | - Ayman Tourbah
- f Neurology Department , University Hospital Center of Reims , Reims , France
| | - Gilles Edan
- c Neurology Department , University Hospital Center of Rennes , Rennes , France
| | - Philippe Corcia
- a Neurology Department , University Hospital Center of Tours , Tours , France.,e ALS Center, University Hospital Center of Tours , Tours , France , and
| |
Collapse
|
17
|
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
|
18
|
Huang C, Wu J, Xu L, Wang J, Chen Z, Yang R. Regulation of HSF1 protein stabilization: An updated review. Eur J Pharmacol 2018; 822:69-77. [PMID: 29341886 DOI: 10.1016/j.ejphar.2018.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/11/2017] [Accepted: 01/09/2018] [Indexed: 12/12/2022]
Abstract
Heat shock factor 1 (HSF1) is a transcriptional factor that determines the efficiency of heat shock responses (HSRs) in the cell. Given its function has been extensively studied in recent years, HSF1 is considered a potential target for the treatment of disorders associated with protein aggregation. The activity of HSF1 is traditionally regulated at the transcriptional level in which the transactivation domain of HSF1 is modified by extensive array of pos-translational modifications, such as phosphorylation, sumoylation, and acetylation. Recently, HSF1 is also reported to be regulated at the monomeric level. For example, in neurodegenerative disorders such as Huntington's disease and Alzheimer's disease the expression levels of the monomeric HSF1 are found to be reduced markedly. Methylene blue (MB) and riluzole, two clinical available drugs, increase the amount of the monomeric HSF1 in both cells and animals. Since the monomeric HSF1 not only determines the efficiency of HSRs, but exerts protective effects in a trimerization-independent manner, increasing the amount of the monomeric HSF1 via stabilization of HSF1 may be an alternative strategy for the amplification of HSR. However, to date we have no outlined knowledges about HSF1 protein stabilization, though studies regarding the regulation of the monomeric HSF1 have been documented in recent years. Here, we summarize the regulation of the monomeric HSF1 by some previously reported factors, such as synuclein, Huntingtin (Htt), TDP-43, unfolded protein response (UPR), MB and doxorubicin (DOX), as well as their possible mechanisms, aiming to push the understanding about HSF1 protein stabilization.
Collapse
Affiliation(s)
- Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| | - Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China
| | - Li Xu
- Department of Ultrasound, Danyang People's Hospital, #2 Xinmin Western Road, Danyang 212300, Jiangsu, China
| | - Jili Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, # 6 North Road Hai'er Xiang, Nantong 226001, Jiangsu, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20Xisi Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
19
|
Nourbakhsh B, Revirajan N, Waubant E. Treatment of fatigue with methylphenidate, modafinil and amantadine in multiple sclerosis (TRIUMPHANT-MS): Study design for a pragmatic, randomized, double-blind, crossover clinical trial. Contemp Clin Trials 2018; 64:67-76. [DOI: 10.1016/j.cct.2017.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/01/2017] [Accepted: 11/02/2017] [Indexed: 11/17/2022]
|
20
|
Zhou T, Ahmad TK, Gozda K, Truong J, Kong J, Namaka M. Implications of white matter damage in amyotrophic lateral sclerosis (Review). Mol Med Rep 2017; 16:4379-4392. [PMID: 28791401 PMCID: PMC5646997 DOI: 10.3892/mmr.2017.7186] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 06/09/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease, which involves the progressive degeneration of motor neurons. ALS has long been considered a disease of the grey matter; however, pathological alterations of the white matter (WM), including axonal loss, axonal demyelination and oligodendrocyte death, have been reported in patients with ALS. The present review examined motor neuron death as the primary cause of ALS and evaluated the associated WM damage that is guided by neuronal‑glial interactions. Previous studies have suggested that WM damage may occur prior to the death of motor neurons, and thus may be considered an early indicator for the diagnosis and prognosis of ALS. However, the exact molecular mechanisms underlying early‑onset WM damage in ALS have yet to be elucidated. The present review explored the detailed anatomy of WM and identified several pathological mechanisms that may be implicated in WM damage in ALS. In addition, it associated the pathophysiological alterations of WM, which may contribute to motor neuron death in ALS, with similar mechanisms of WM damage that are involved in multiple sclerosis (MS). Furthermore, the early detection of WM damage in ALS, using neuroimaging techniques, may lead to earlier therapeutic intervention, using immunomodulatory treatment strategies similar to those used in relapsing‑remitting MS, aimed at delaying WM damage in ALS. Early therapeutic approaches may have the potential to delay motor neuron damage and thus prolong the survival of patients with ALS. The therapeutic interventions that are currently available for ALS are only marginally effective. However, early intervention with immunomodulatory drugs may slow the progression of WM damage in the early stages of ALS, thus delaying motor neuron death and increasing the life expectancy of patients with ALS.
Collapse
Affiliation(s)
- Ting Zhou
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Tina Khorshid Ahmad
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Kiana Gozda
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jessica Truong
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Michael Namaka
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
- Department of Medical Rehabilitation, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Department of Internal Medicine, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 1R9, Canada
| |
Collapse
|
21
|
Kuhle J, Nourbakhsh B, Grant D, Morant S, Barro C, Yaldizli Ö, Pelletier D, Giovannoni G, Waubant E, Gnanapavan S. Serum neurofilament is associated with progression of brain atrophy and disability in early MS. Neurology 2017; 88:826-831. [PMID: 28148632 DOI: 10.1212/wnl.0000000000003653] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 10/10/2016] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate a potential effect of riluzole on serum neurofilaments (Nf) compared to placebo and the relationship between longitudinal clinical and MRI outcomes and serum Nf levels. METHODS Serum samples were obtained from participants enrolled in a randomized double-blind trial of neuroprotection with riluzole vs placebo as an add-on to weekly interferon-β (IFN-β)-1a IM initiated 3 months after randomization. Nf measurements were performed by ELISA and electrochemiluminescence immunoassay. RESULTS Longitudinal serum samples were available from 22 riluzole and 20 placebo participants over 24 months. There was no observed treatment effect with riluzole. Nf light chain (NfL) levels decreased over time (p = 0.007 at 24 months), whereas the Nf heavy chain was unchanged (p = 0.997). Changes in NfL were correlated with EDSS change (p = 0.009) and neuropsychological outcomes. Brain volume decreased more rapidly in patients with high baseline NfL (p = 0.05 at 12 months and p = 0.008 at 24 months) and this relationship became stronger at 24 months (p = 0.024 for interaction). Higher and increasing NfL predicted higher number of gadolinium-enhancing lesions (p < 0.001 for both). CONCLUSIONS Our findings support the potential value of serum NfL as a marker of neuroaxonal injury in early multiple sclerosis. Its reduction over time could represent regression to the mean, or a possible treatment effect of IFN-β-1a. The association with whole brain atrophy and the formation of acute white matter lesions has relevant implications to use serum NfL as a noninvasive biomarker of the overall consequences of brain damage and ongoing disease activity. CLINICALTRIALSGOV IDENTIFIER NCT00501943.
Collapse
Affiliation(s)
- Jens Kuhle
- From Neurologic Clinic and Policlinic (J.K., C.B., O.Y.), Departments of Medicine, Clinical Research, and Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (B.N., E.W.), University of California San Francisco; Department of Neuroimmunology (D.G.), Queen Square, Institute of Neurology, London; independent statistician (S.M.), Haddenham, Bucks, UK; Department of Neurology (D.P.), University of Southern California, Los Angeles; and Department of Neuroscience and Trauma (G.G., S.G.), Queen Mary University of London, UK.
| | - Bardia Nourbakhsh
- From Neurologic Clinic and Policlinic (J.K., C.B., O.Y.), Departments of Medicine, Clinical Research, and Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (B.N., E.W.), University of California San Francisco; Department of Neuroimmunology (D.G.), Queen Square, Institute of Neurology, London; independent statistician (S.M.), Haddenham, Bucks, UK; Department of Neurology (D.P.), University of Southern California, Los Angeles; and Department of Neuroscience and Trauma (G.G., S.G.), Queen Mary University of London, UK
| | - Donna Grant
- From Neurologic Clinic and Policlinic (J.K., C.B., O.Y.), Departments of Medicine, Clinical Research, and Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (B.N., E.W.), University of California San Francisco; Department of Neuroimmunology (D.G.), Queen Square, Institute of Neurology, London; independent statistician (S.M.), Haddenham, Bucks, UK; Department of Neurology (D.P.), University of Southern California, Los Angeles; and Department of Neuroscience and Trauma (G.G., S.G.), Queen Mary University of London, UK
| | - Steve Morant
- From Neurologic Clinic and Policlinic (J.K., C.B., O.Y.), Departments of Medicine, Clinical Research, and Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (B.N., E.W.), University of California San Francisco; Department of Neuroimmunology (D.G.), Queen Square, Institute of Neurology, London; independent statistician (S.M.), Haddenham, Bucks, UK; Department of Neurology (D.P.), University of Southern California, Los Angeles; and Department of Neuroscience and Trauma (G.G., S.G.), Queen Mary University of London, UK
| | - Christian Barro
- From Neurologic Clinic and Policlinic (J.K., C.B., O.Y.), Departments of Medicine, Clinical Research, and Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (B.N., E.W.), University of California San Francisco; Department of Neuroimmunology (D.G.), Queen Square, Institute of Neurology, London; independent statistician (S.M.), Haddenham, Bucks, UK; Department of Neurology (D.P.), University of Southern California, Los Angeles; and Department of Neuroscience and Trauma (G.G., S.G.), Queen Mary University of London, UK
| | - Özgür Yaldizli
- From Neurologic Clinic and Policlinic (J.K., C.B., O.Y.), Departments of Medicine, Clinical Research, and Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (B.N., E.W.), University of California San Francisco; Department of Neuroimmunology (D.G.), Queen Square, Institute of Neurology, London; independent statistician (S.M.), Haddenham, Bucks, UK; Department of Neurology (D.P.), University of Southern California, Los Angeles; and Department of Neuroscience and Trauma (G.G., S.G.), Queen Mary University of London, UK
| | - Daniel Pelletier
- From Neurologic Clinic and Policlinic (J.K., C.B., O.Y.), Departments of Medicine, Clinical Research, and Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (B.N., E.W.), University of California San Francisco; Department of Neuroimmunology (D.G.), Queen Square, Institute of Neurology, London; independent statistician (S.M.), Haddenham, Bucks, UK; Department of Neurology (D.P.), University of Southern California, Los Angeles; and Department of Neuroscience and Trauma (G.G., S.G.), Queen Mary University of London, UK
| | - Gavin Giovannoni
- From Neurologic Clinic and Policlinic (J.K., C.B., O.Y.), Departments of Medicine, Clinical Research, and Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (B.N., E.W.), University of California San Francisco; Department of Neuroimmunology (D.G.), Queen Square, Institute of Neurology, London; independent statistician (S.M.), Haddenham, Bucks, UK; Department of Neurology (D.P.), University of Southern California, Los Angeles; and Department of Neuroscience and Trauma (G.G., S.G.), Queen Mary University of London, UK
| | - Emmanuelle Waubant
- From Neurologic Clinic and Policlinic (J.K., C.B., O.Y.), Departments of Medicine, Clinical Research, and Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (B.N., E.W.), University of California San Francisco; Department of Neuroimmunology (D.G.), Queen Square, Institute of Neurology, London; independent statistician (S.M.), Haddenham, Bucks, UK; Department of Neurology (D.P.), University of Southern California, Los Angeles; and Department of Neuroscience and Trauma (G.G., S.G.), Queen Mary University of London, UK
| | - Sharmilee Gnanapavan
- From Neurologic Clinic and Policlinic (J.K., C.B., O.Y.), Departments of Medicine, Clinical Research, and Biomedicine, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (B.N., E.W.), University of California San Francisco; Department of Neuroimmunology (D.G.), Queen Square, Institute of Neurology, London; independent statistician (S.M.), Haddenham, Bucks, UK; Department of Neurology (D.P.), University of Southern California, Los Angeles; and Department of Neuroscience and Trauma (G.G., S.G.), Queen Mary University of London, UK
| |
Collapse
|
22
|
Abstract
Brain atrophy occurs at a faster rate in patients with multiple sclerosis (MS) than in healthy individuals. In three randomized, controlled, phase III trials, fingolimod reduced the annual rate of brain volume loss (BVL) in patients with relapsing MS (RMS) by approximately one-third relative to that in individuals receiving placebo or intramuscular interferon beta-1a. Analysis of brain volume changes during study extensions has shown that this reduced rate of BVL is sustained in patients with RMS receiving fingolimod continuously. Subgroup analyses of the core phase III and extension studies have shown that reductions in the rate of BVL are observed irrespective of levels of inflammatory lesion activity seen by magnetic resonance imaging at baseline and on study; levels of disability at baseline; and treatment history. The rate of BVL in these studies was predicted independently by T2 lesion and gadolinium-enhancing lesion burdens at baseline, and correlations observed between BVL and increasing levels of disability strengthened over time. In another phase III trial in patients with primary progressive MS (PPMS), fingolimod did not reduce BVL overall relative to placebo; however, consistent with findings in RMS, there was a treatment effect on BVL in patients with PPMS with gadolinium-enhancing lesion activity at baseline. The association between treatment effects on BVL and future accumulation of disability argues in favor of measuring BVL on a more routine basis and with a more structured approach than is generally the case in clinical practice. Despite several practical obstacles, progress is being made in achieving this goal.
Collapse
|
23
|
Macrez R, Stys PK, Vivien D, Lipton SA, Docagne F. Mechanisms of glutamate toxicity in multiple sclerosis: biomarker and therapeutic opportunities. Lancet Neurol 2016; 15:1089-102. [PMID: 27571160 DOI: 10.1016/s1474-4422(16)30165-x] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/21/2016] [Accepted: 06/27/2016] [Indexed: 12/22/2022]
Abstract
Research advances support the idea that excessive activation of the glutamatergic pathway plays an important part in the pathophysiology of multiple sclerosis. Beyond the well established direct toxic effects on neurons, additional sites of glutamate-induced cell damage have been described, including effects in oligodendrocytes, astrocytes, endothelial cells, and immune cells. Such toxic effects could provide a link between various pathological aspects of multiple sclerosis, such as axonal damage, oligodendrocyte cell death, demyelination, autoimmunity, and blood-brain barrier dysfunction. Understanding of the mechanisms underlying glutamate toxicity in multiple sclerosis could help in the development of new approaches for diagnosis, treatment, and follow-up in patients with this debilitating disease. While several clinical trials of glutamatergic modulators have had disappointing results, our growing understanding suggests that there is reason to remain optimistic about the therapeutic potential of these drugs.
Collapse
Affiliation(s)
| | - Peter K Stys
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Denis Vivien
- INSERM U919, University of Caen Normandy, Caen, France
| | - Stuart A Lipton
- Scintillon Institute San Diego, CA, USA; Scripps Research Institute, La Jolla, CA, USA; School of Mecicine, University of California, San Diego, CA, USA
| | | |
Collapse
|
24
|
Nourbakhsh B, Julian L, Waubant E. Fatigue and depression predict quality of life in patients with early multiple sclerosis: a longitudinal study. Eur J Neurol 2016; 23:1482-6. [DOI: 10.1111/ene.13102] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/10/2016] [Indexed: 01/14/2023]
Affiliation(s)
- B. Nourbakhsh
- Department of Neurology; University of California San Francisco; San Francisco CA USA
| | - L. Julian
- Department of Internal Medicine; University of California San Francisco; San Francisco CA USA
| | - E. Waubant
- Department of Neurology; University of California San Francisco; San Francisco CA USA
- Department of Pediatrics; University of California San Francisco; San Francisco CA USA
| |
Collapse
|
25
|
Nourbakhsh B, Azevedo C, Maghzi AH, Spain R, Pelletier D, Waubant E. Subcortical grey matter volumes predict subsequent walking function in early multiple sclerosis. J Neurol Sci 2016; 366:229-233. [DOI: 10.1016/j.jns.2016.04.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/04/2016] [Accepted: 04/28/2016] [Indexed: 01/28/2023]
|
26
|
Pul R, Saadat M, Morbiducci F, Skripuletz T, Pul Ü, Brockmann D, Sühs KW, Schwenkenbecher P, Kahl KG, Pars K, Stangel M, Trebst C. Longitudinal time-domain optic coherence study of retinal nerve fiber layer in IFNβ-treated and untreated multiple sclerosis patients. Exp Ther Med 2016; 12:190-200. [PMID: 27347038 PMCID: PMC4906774 DOI: 10.3892/etm.2016.3300] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 02/11/2016] [Indexed: 12/14/2022] Open
Abstract
Quantification of the retinal nerve fiber layer (RNFL) by optical coherence tomography (OCT) has been proposed to provide an indirect measure for retinal axonal loss. The aim of the present study was to determine whether interferon beta (IFNβ) treatment impedes retinal axonal loss in multiple sclerosis (MS) patients. A total of 48 patients with MS (24 IFNβ-1b-treated and 24 untreated subjects) and 12 healthy controls were enrolled in a prospective longitudinal OCT study. OCT measurements were performed for both eyes of each subject at baseline, and at 3-, 6-, and 12-month follow-up examinations using a time-domain OCT. At each visit, we additionally recorded full-field visual evoked potential (VEP) responses and performed the paced auditory serial addition test (PASAT), in addition to expanded disability status scale (EDSS) scoring. Generalized estimation equation (GEE) was used to account for repeated measurements and paired-data. The model-based approach predicted a monthly reduction in the RNFL thickness by 0.19 µm in the eyes of the MS subjects. The reduction was estimated to be 0.17 µm in case of IFNβ-treatment and 0.16 µm in case of no treatment. Treatment duration and group allocation were not significantly associated with the RNFL thickness. Inclusion of further longitudinal data (EDSS, two and three second PASAT) in each of our models did not result in any significant association. In summary, over a period of one year no significant association between IFNβ-1b treatment and RNFL thinning was identified in patients with MS.
Collapse
Affiliation(s)
- Refik Pul
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Mehdi Saadat
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Franco Morbiducci
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Ünsal Pul
- Department of Thoracic and Cardiovascular Surgery, University Hospital Essen, 45147 Essen, Germany
| | - Dorothee Brockmann
- Department of Ophthalmology, Hannover Medical School, 30625 Hannover, Germany
| | - Kurt-Wolfram Sühs
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Kai Günter Kahl
- Department of Psychiatry, Hannover Medical School, 30625 Hannover, Germany
| | - Kaweh Pars
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Martin Stangel
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Corinna Trebst
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
27
|
Longitudinal associations between MRI and cognitive changes in very early MS. Mult Scler Relat Disord 2016; 5:47-52. [DOI: 10.1016/j.msard.2015.10.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 09/17/2015] [Accepted: 10/26/2015] [Indexed: 11/18/2022]
|
28
|
Longitudinal associations between brain structural changes and fatigue in early MS. Mult Scler Relat Disord 2015; 5:29-33. [PMID: 26856940 DOI: 10.1016/j.msard.2015.10.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/15/2015] [Accepted: 10/20/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Fatigue is a common and disabling symptom of multiple sclerosis (MS) patients. Structural changes in several brain areas have been reported to correlate with fatigue in MS patients but none consistently. OBJECTIVE To study the association between global and regional measures of brain atrophy and fatigue in patients with early relapsing MS. METHODS Clinically isolated syndrome and relapsing MS patients within 12 months of clinical onset were enrolled in a neuroprotection trial of riluzole versus placebo with up to 36 months of follow-up. MRI metrics included brain volumes measured by SIENAX normalized measurements [normalized brain parenchymal volume (nBPV), normalized normal-appearing white and gray matter volume (nNAWMV and nGMV)] and T2 lesion volume (T2LV). Cortical thickness, thalamic volume and cerebellar cortical volume were measured using Freesurfer's longitudinal pipeline (v5.3) and a lesion inpainting approach. Fatigue was evaluated using the Modified Fatigue Impact Scale (MFIS). Mixed model regression measured time trends and associations between imaging and fatigue severity, adjusting for age and sex. RESULTS Forty-three patients (mean age 36 years; 31 females) were enrolled within 7.5 ± 4.9 months of symptom onset. Baseline and change over baseline in lesion volumes, grey matter, white matter, basal ganglia and total parenchymal volumes were not associated with change in MFIS score over time. Lower thalamic volume at baseline predicted increasing physical subscale of MFIS score during the study (p=0.017). There was a trend toward baseline thalamic volume and cerebellar cortical volume predicting subsequent change in total MFIS score (p=0.055 and 0.082 respectively). On-study change in thalamic or cerebellar cortical volume was not associated with on-study change in MFIS score. CONCLUSION Global measures of tissue loss are not strongly associated with fatigue in patients with early MS. However, thalamic and cerebellar cortical atrophy may be predictive of subsequent changes in fatigue in these patients.
Collapse
|
29
|
Maghzi AH, Graves J, Revirajan N, Spain R, Liu S, McCulloch CE, Pelletier D, Green AJ, Waubant E. Retinal axonal loss in very early stages of multiple sclerosis. Eur J Neurol 2015; 22:1138-41. [PMID: 25929276 DOI: 10.1111/ene.12722] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/26/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE The lack of surrogates of clinical progression has limited the design of neuroprotection trials in multiple sclerosis (MS). Our aim was to study the association between time-domain optical coherence tomography measures and clinical and magnetic resonance imaging outcomes in early MS. METHODS Forty-three relapsing-remitting MS patients within 1 year of onset were followed for up to 3 years. RESULTS The peripapillary retinal nerve fiber layer (RNFL) decreased annually by 2 μm (95% confidence interval -3.89, -0.11; P = 0.038). The RNFL tended to be associated with normalized normal appearing white matter volume in cross-sectional (P = 0.08) and longitudinal analyses (P = 0.06). CONCLUSIONS There is substantial RNFL loss even in very early MS. Our data suggest that retinal axonal atrophy is associated with atrophy in global white matter volume in early MS.
Collapse
Affiliation(s)
- A-H Maghzi
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - J Graves
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - N Revirajan
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - R Spain
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - S Liu
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - C E McCulloch
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - D Pelletier
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - A J Green
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - E Waubant
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.,Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
30
|
Mallucci G, Peruzzotti-Jametti L, Bernstock JD, Pluchino S. The role of immune cells, glia and neurons in white and gray matter pathology in multiple sclerosis. Prog Neurobiol 2015; 127-128:1-22. [PMID: 25802011 PMCID: PMC4578232 DOI: 10.1016/j.pneurobio.2015.02.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/24/2014] [Accepted: 02/27/2015] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis is one of the most common causes of chronic neurological disability beginning in early to middle adult life. Multiple sclerosis is idiopathic in nature, yet increasing correlative evidence supports a strong association between one's genetic predisposition, the environment and the immune system. Symptoms of multiple sclerosis have primarily been shown to result from a disruption in the integrity of myelinated tracts within the white matter of the central nervous system. However, recent research has also highlighted the hitherto underappreciated involvement of gray matter in multiple sclerosis disease pathophysiology, which may be especially relevant when considering the accumulation of irreversible damage and progressive disability. This review aims at providing a comprehensive overview of the interplay between inflammation, glial/neuronal damage and regeneration throughout the course of multiple sclerosis via the analysis of both white and gray matter lesional pathology. Further, we describe the common pathological mechanisms underlying both relapsing and progressive forms of multiple sclerosis, and analyze how current (as well as future) treatments may interact and/or interfere with its pathology. Understanding the putative mechanisms that drive disease pathogenesis will be key in helping to develop effective therapeutic strategies to prevent, mitigate, and treat the diverse morbidities associated with multiple sclerosis.
Collapse
Affiliation(s)
- Giulia Mallucci
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
- Department of Brain and Behavioural Sciences, National Neurological Institute C. Mondino, University of Pavia, 27100 Pavia, Italy
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
| | - Joshua D. Bernstock
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bldg10/Rm5B06, MSC 1401, 10 Center Drive, Bethesda, MD 20892, USA
| | - Stefano Pluchino
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
| |
Collapse
|