201
|
Benjamins JA, Nedelkoska L, Touil H, Stemmer PM, Carruthers NJ, Jena BP, Naik AR, Bar-Or A, Lisak RP. Exosome-enriched fractions from MS B cells induce oligodendrocyte death. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:e550. [PMID: 31044144 PMCID: PMC6467686 DOI: 10.1212/nxi.0000000000000550] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/16/2019] [Indexed: 12/21/2022]
Abstract
Objective To identify whether factors toxic to oligodendrocytes (OLs), released by B cells from patients with MS, are found in extracellular microvesicles enriched in exosomes. Methods Conditioned medium (Sup) was obtained from cultures of blood B cells of patients with MS and normal controls (NCs). Exosome-enriched (Ex-En) fractions were prepared by solvent precipitation from Sup containing bovine serum and from serum-free Sup by ultracentrifugation (UC) or immunoprecipitation (IP) with antibodies to CD9. Ex-En fractions were diluted 1:4 with OL culture medium and screened for toxic effects on cultured rat OLs as measured by trypan blue uptake. Proteomic analysis was performed on Sup fractions. Results MS B cell-derived Ex-En fractions prepared from Sup by solvent extraction, UC, or IP induced OL death, whereas corresponding Ex-En fractions from NC showed little toxicity. Proteomic analysis of Sup demonstrated enrichment of proteins characteristic of exosomes from both NC and MS B-cell Sup. Ontology enrichment analysis suggested differences in the types and cargo of exosomes from MS Sup compared with NC, with proteins related to cell surface, extracellular plasma membrane, and gliogenesis enriched in MS. Conclusions Much of the in vitro toxicity of Sup from B cells of patients with relapsing-remitting MS is found in Ex-En fractions, as confirmed by 3 methods. Proteomic analysis of B-cell Sup indicates multiple differences between MS and NC.
Collapse
Affiliation(s)
- Joyce A Benjamins
- Departments of Neurology and Biochemistry, Immunology and Microbiology (J.A.B., R.P.L.), Wayne State University School of Medicine; Department of Neurology (L.N.), Wayne State University School of Medicine, Detroit, Michigan; Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (H.T., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology (H.T., A.B.-O.), McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada; Institute of Environmental Health Sciences (P.M.S., N.J.C.), Wayne State University; and Department of Physiology (B.P.J., A.R.N.), Wayne State University School of Medicine, Detroit, Michigan
| | - Liljana Nedelkoska
- Departments of Neurology and Biochemistry, Immunology and Microbiology (J.A.B., R.P.L.), Wayne State University School of Medicine; Department of Neurology (L.N.), Wayne State University School of Medicine, Detroit, Michigan; Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (H.T., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology (H.T., A.B.-O.), McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada; Institute of Environmental Health Sciences (P.M.S., N.J.C.), Wayne State University; and Department of Physiology (B.P.J., A.R.N.), Wayne State University School of Medicine, Detroit, Michigan
| | - Hanane Touil
- Departments of Neurology and Biochemistry, Immunology and Microbiology (J.A.B., R.P.L.), Wayne State University School of Medicine; Department of Neurology (L.N.), Wayne State University School of Medicine, Detroit, Michigan; Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (H.T., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology (H.T., A.B.-O.), McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada; Institute of Environmental Health Sciences (P.M.S., N.J.C.), Wayne State University; and Department of Physiology (B.P.J., A.R.N.), Wayne State University School of Medicine, Detroit, Michigan
| | - Paul M Stemmer
- Departments of Neurology and Biochemistry, Immunology and Microbiology (J.A.B., R.P.L.), Wayne State University School of Medicine; Department of Neurology (L.N.), Wayne State University School of Medicine, Detroit, Michigan; Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (H.T., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology (H.T., A.B.-O.), McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada; Institute of Environmental Health Sciences (P.M.S., N.J.C.), Wayne State University; and Department of Physiology (B.P.J., A.R.N.), Wayne State University School of Medicine, Detroit, Michigan
| | - Nicholas J Carruthers
- Departments of Neurology and Biochemistry, Immunology and Microbiology (J.A.B., R.P.L.), Wayne State University School of Medicine; Department of Neurology (L.N.), Wayne State University School of Medicine, Detroit, Michigan; Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (H.T., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology (H.T., A.B.-O.), McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada; Institute of Environmental Health Sciences (P.M.S., N.J.C.), Wayne State University; and Department of Physiology (B.P.J., A.R.N.), Wayne State University School of Medicine, Detroit, Michigan
| | - Bhanu P Jena
- Departments of Neurology and Biochemistry, Immunology and Microbiology (J.A.B., R.P.L.), Wayne State University School of Medicine; Department of Neurology (L.N.), Wayne State University School of Medicine, Detroit, Michigan; Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (H.T., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology (H.T., A.B.-O.), McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada; Institute of Environmental Health Sciences (P.M.S., N.J.C.), Wayne State University; and Department of Physiology (B.P.J., A.R.N.), Wayne State University School of Medicine, Detroit, Michigan
| | - Akshata R Naik
- Departments of Neurology and Biochemistry, Immunology and Microbiology (J.A.B., R.P.L.), Wayne State University School of Medicine; Department of Neurology (L.N.), Wayne State University School of Medicine, Detroit, Michigan; Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (H.T., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology (H.T., A.B.-O.), McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada; Institute of Environmental Health Sciences (P.M.S., N.J.C.), Wayne State University; and Department of Physiology (B.P.J., A.R.N.), Wayne State University School of Medicine, Detroit, Michigan
| | - Amit Bar-Or
- Departments of Neurology and Biochemistry, Immunology and Microbiology (J.A.B., R.P.L.), Wayne State University School of Medicine; Department of Neurology (L.N.), Wayne State University School of Medicine, Detroit, Michigan; Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (H.T., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology (H.T., A.B.-O.), McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada; Institute of Environmental Health Sciences (P.M.S., N.J.C.), Wayne State University; and Department of Physiology (B.P.J., A.R.N.), Wayne State University School of Medicine, Detroit, Michigan
| | - Robert P Lisak
- Departments of Neurology and Biochemistry, Immunology and Microbiology (J.A.B., R.P.L.), Wayne State University School of Medicine; Department of Neurology (L.N.), Wayne State University School of Medicine, Detroit, Michigan; Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (H.T., A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology (H.T., A.B.-O.), McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada; Institute of Environmental Health Sciences (P.M.S., N.J.C.), Wayne State University; and Department of Physiology (B.P.J., A.R.N.), Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
202
|
Bauckneht M, Capitanio S, Raffa S, Roccatagliata L, Pardini M, Lapucci C, Marini C, Sambuceti G, Inglese M, Gallo P, Cecchin D, Nobili F, Morbelli S. Molecular imaging of multiple sclerosis: from the clinical demand to novel radiotracers. EJNMMI Radiopharm Chem 2019; 4:6. [PMID: 31659498 PMCID: PMC6453990 DOI: 10.1186/s41181-019-0058-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
Background Brain PET imaging with different tracers is mainly clinically used in the field of neurodegenerative diseases and brain tumors. In recent years, the potential usefulness of PET has also gained attention in the field of MS. In fact, MS is a complex disease and several processes can be selected as a target for PET imaging. The use of PET with several different tracers has been mainly evaluated in the research setting to investigate disease pathophysiology (i.e. phenotypes, monitoring of progression) or to explore its use a surrogate end-point in clinical trials. Results We have reviewed PET imaging studies in MS in humans and animal models. Tracers have been grouped according to their pathophysiological targets (ie. tracers for myelin kinetic, neuroinflammation, and neurodegeneration). The emerging clinical indication for brain PET imaging in the differential diagnosis of suspected tumefactive demyelinated plaques as well as the clinical potential provided by PET images in view of the recent introduction of PET/MR technology are also addressed. Conclusion While several preclinical and fewer clinical studies have shown results, full-scale clinical development programs are needed to translate molecular imaging technologies into a clinical reality that could ideally fit into current precision medicine perspectives.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132, Genoa, Italy.
| | - Selene Capitanio
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132, Genoa, Italy
| | - Stefano Raffa
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Luca Roccatagliata
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy.,Neuroradiology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Matteo Pardini
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy.,Clinica Neurologica, IRCCS Ospedale Policlinico, San Martino, Genoa, Italy
| | - Caterina Lapucci
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
| | - Cecilia Marini
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132, Genoa, Italy.,CNR Institute of Molecular Bioimaging and Physiology, Milan, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132, Genoa, Italy.,Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Matilde Inglese
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy.,Clinica Neurologica, IRCCS Ospedale Policlinico, San Martino, Genoa, Italy
| | - Paolo Gallo
- Multiple Sclerosis Centre of the Veneto Region, Department of Neurosciences DNS, University of Padua, Padua, Italy
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine-DIMED, Padova University Hospital, Padua, Italy.,Padua Neuroscience Center, University of Padua, Padua, Italy
| | - Flavio Nobili
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy.,Clinica Neurologica, IRCCS Ospedale Policlinico, San Martino, Genoa, Italy
| | - Silvia Morbelli
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132, Genoa, Italy.,Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| |
Collapse
|
203
|
Li R, Bar-Or A. The Multiple Roles of B Cells in Multiple Sclerosis and Their Implications in Multiple Sclerosis Therapies. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a029108. [PMID: 29661809 DOI: 10.1101/cshperspect.a029108] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Increasing evidence has suggested that both antibody-dependent and antibody-independent functions of B cells are involved in multiple sclerosis (MS). The contrasting results of distinct B-cell targeting therapies in MS patients underscores the importance of elucidating these multiple B-cell functions. In this review, we discuss the generation of autoreactive B cells, migration of B cells into the central nervous system (CNS), and how different functions of B cells may contribute to MS disease activity and potentially mitigation in both the periphery and CNS compartments. In addition, we propose several future therapeutic strategies that may better target/shape B-cell responses for long-term treatment of MS.
Collapse
Affiliation(s)
- Rui Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Amit Bar-Or
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
204
|
Faizy TD, Broocks G, Thaler C, Rauch G, Gebert P, Stürner KH, Flottmann F, Leischner H, Kniep HC, Stellmann JP, Heesen C, Fiehler J, Gellißen S, Hanning U. Development of Cortical Lesion Volumes on Double Inversion Recovery MRI in Patients With Relapse-Onset Multiple Sclerosis. Front Neurol 2019; 10:133. [PMID: 30873106 PMCID: PMC6401630 DOI: 10.3389/fneur.2019.00133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/01/2019] [Indexed: 11/30/2022] Open
Abstract
Background and Objective: In multiple sclerosis (MS) patients, Double Inversion Recovery (DIR) magnetic resonance imaging (MRI) can be used to detect cortical lesions (CL). While the quantity and distribution of CLs seems to be associated with patients' disease course, literature lacks frequent assessments of CL volumes (CL-V) in this context. We investigated the reliability of DIR for the longitudinal assessment of CL-V development with frequent follow-up MRIs and examined the course of CL-V progressions in relation to white-matter lesions (WML), contrast enhancing lesions (CEL) and clinical parameters in patients with Relapsing-Remitting Multiple Sclerosis (RRMS). Methods: In this post-hoc analysis, image- and clinical data of a subset of 24 subjects that were part of a phase IIa clinical trial on the “Safety, Tolerability and Mechanisms of Action of Boswellic Acids in Multiple Sclerosis (SABA)” (ClinicalTrials.gov, NCT01450124) were included. The study was divided in three phases (screening, treatment, study-end). All patients received 12 MRI follow-up-examinations (including DIR) during a 16-months period. CL-Vs were assessed for each patient on each follow-up MRI separately by two experienced neuroradiologists. Results of neurological screening tests, as well as other MRI parameters (WML number and volume and CELs) were included from the SABA investigation data. Results: Inter-rater agreement regarding CL-V assessment over time was good-to-excellent (κ = 0.89). Mean intraobserver variability was 1.1%. In all patients, a total number of 218 CLs was found. Total CL-Vs of all patients increased during the 4 months of baseline screening followed by a continuous and significant decrease from month 5 until study-end (p < 0.001, Kendall'W = 0.413). A positive association between WML volumes and CL-Vs was observed during baseline screening. Decreased CL-V were associated with lower EDSS and also with improvements of SDMT- and SCRIPPS scores. Conclusion: DIR MRI seems to be a reliable tool for the frequent assessment of CL-Vs. Overall CL-Vs decreased during the follow-up period and were associated with improvements of cognitive and disability status scores. Our results suggest the presence of short-term CL-V dynamics in RRMS patients and we presume that the laborious evaluation of lesion volumes may be worthwhile for future investigations. Clinical Trial Numbers:www.ClinicalTrials.gov, “The SABA trial”; number: NCT01450124
Collapse
Affiliation(s)
- Tobias D Faizy
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriel Broocks
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Thaler
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Geraldine Rauch
- Institute of Medical Biometry and Epidemiology, Charité Berlin-University Medical Center, Berlin, Germany
| | - Pimrapat Gebert
- Institute of Medical Biometry and Epidemiology, Charité Berlin-University Medical Center, Berlin, Germany
| | - Klarissa H Stürner
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Neurology, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Fabian Flottmann
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hannes Leischner
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Helge C Kniep
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan-Patrick Stellmann
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Heesen
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Fiehler
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Gellißen
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uta Hanning
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
205
|
Schwenkenbecher P, Wurster U, Konen FF, Gingele S, Sühs KW, Wattjes MP, Stangel M, Skripuletz T. Impact of the McDonald Criteria 2017 on Early Diagnosis of Relapsing-Remitting Multiple Sclerosis. Front Neurol 2019; 10:188. [PMID: 30930829 PMCID: PMC6428717 DOI: 10.3389/fneur.2019.00188] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/14/2019] [Indexed: 01/21/2023] Open
Abstract
Multiple sclerosis is a chronic immune mediated demyelinating disease leading to neurological disabilities that need to be diagnosed and treated early. Guidelines on multiple sclerosis diagnosis and monitoring experienced comprehensive changes over the last decades. The first McDonald criteria published in 2001 emphasized the importance of MR imaging but also recognized the role of cerebrospinal fluid diagnostics. The demonstration of an intrathecal immunoglobulin G synthesis is a well-established additional component and has a long tradition in the diagnosis of relapsing-remitting multiple sclerosis. However, the role of cerebrospinal fluid for diagnostic purposes was rather diminished in each revision of the McDonald criteria. In the latest revision of the McDonald criteria of 2017, the detection of an intrathecal immunoglobulin G synthesis as oligoclonal bands experienced a revival. Patients with the first clinical event suggesting multiple sclerosis who fulfill the criteria for dissemination in space can be diagnosed with relapsing-remitting multiple sclerosis when oligoclonal bands in cerebrospinal fluid are detected. The diagnostic sensitivity of these novel criteria with a focus on dissemination in time and oligoclonal bands as a substitute for dissemination in time was published in different cohorts in the last year and is of special interest in this review. Recently published data show that by applying the 2017 McDonald criteria, multiple sclerosis can be diagnosed more frequently at the time of first clinical event as compared to the 2010 McDonald criteria. The main effect was due to the implementation of oligoclonal bands as a substitute for dissemination in time. However, careful differential diagnosis is essential in patients with atypical clinical manifestations to avoid misdiagnoses.
Collapse
Affiliation(s)
- Philipp Schwenkenbecher
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Ulrich Wurster
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Franz Felix Konen
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Stefan Gingele
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Kurt-Wolfram Sühs
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Mike P Wattjes
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Thomas Skripuletz
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
206
|
van der Poel M, Ulas T, Mizee MR, Hsiao CC, Miedema SSM, Adelia, Schuurman KG, Helder B, Tas SW, Schultze JL, Hamann J, Huitinga I. Transcriptional profiling of human microglia reveals grey-white matter heterogeneity and multiple sclerosis-associated changes. Nat Commun 2019; 10:1139. [PMID: 30867424 PMCID: PMC6416318 DOI: 10.1038/s41467-019-08976-7] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 02/07/2019] [Indexed: 12/22/2022] Open
Abstract
Here we report the transcriptional profile of human microglia, isolated from normal-appearing grey matter (GM) and white matter (WM) of multiple sclerosis (MS) and non-neurological control donors, to find possible early changes related to MS pathology. Microglia show a clear region-specific profile, indicated by higher expression of type-I interferon genes in GM and higher expression of NF-κB pathway genes in WM. Transcriptional changes in MS microglia also differ between GM and WM. MS WM microglia show increased lipid metabolism gene expression, which relates to MS pathology since active MS lesion-derived microglial nuclei show similar altered gene expression. Microglia from MS GM show increased expression of genes associated with glycolysis and iron homeostasis, possibly reflecting microglia reacting to iron depositions. Except for ADGRG1/GPR56, expression of homeostatic genes, such as P2RY12 and TMEM119, is unaltered in normal-appearing MS tissue, demonstrating overall preservation of microglia homeostatic functions in the initiation phase of MS.
Collapse
Affiliation(s)
- Marlijn van der Poel
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105BA, Amsterdam, The Netherlands
| | - Thomas Ulas
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
| | - Mark R Mizee
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105BA, Amsterdam, The Netherlands
| | - Cheng-Chih Hsiao
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Suzanne S M Miedema
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105BA, Amsterdam, The Netherlands
| | - Adelia
- Netherlands Brain Bank, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105BA, Amsterdam, The Netherlands
| | - Karianne G Schuurman
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105BA, Amsterdam, The Netherlands
| | - Boy Helder
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Sander W Tas
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Joachim L Schultze
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, Carl-Troll-Straße 31, 53115, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases, University of Bonn, Sigmund-Freud-Street 27, 53127, Bonn, Germany
| | - Jörg Hamann
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105BA, Amsterdam, The Netherlands.
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| | - Inge Huitinga
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105BA, Amsterdam, The Netherlands.
| |
Collapse
|
207
|
Hametner S, Dal Bianco A, Trattnig S, Lassmann H. Iron related changes in MS lesions and their validity to characterize MS lesion types and dynamics with Ultra-high field magnetic resonance imaging. Brain Pathol 2019; 28:743-749. [PMID: 30020556 PMCID: PMC8028547 DOI: 10.1111/bpa.12643] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 06/12/2018] [Indexed: 12/31/2022] Open
Abstract
Iron accumulates with age in the normal human brain. This process is altered at several levels in the brain of multiple sclerosis (MS) patients. Since iron is mainly stored in oligodendrocytes and myelin in the normal brain, its liberation in demyelinating lesions may amplify tissue damage in demyelinating lesions and its uptake in macrophages and microglia may help to more precisely define activity stages of the lesions. In addition, glia cells change their iron import, export and storage properties in MS lesions, which is reflected by alterations in the expression of iron transport molecules. Changes of iron distribution in the brain can be reliably detected by MRI, particularly upon application of Ultra‐high magnetic field (7 Tesla). Iron‐sensitive MRI allows to more accurately distinguish the lesions in MS from those in other inflammatory brain diseases, to visualize a subset of slowly expanding lesions in the progressive stage of MS and to increase the sensitivity for lesion detection in the gray matter, such as the cerebral cortex or deep gray matter nuclei.
Collapse
Affiliation(s)
- Simon Hametner
- Center for Brain Research, Medical University of Vienna, Austria.,Institute of Neuropathology, University of Göttingen, Germany
| | - Assunta Dal Bianco
- Center for Brain Research, Medical University of Vienna, Austria.,Department of Neurology, Medical University of Vienna, Austria
| | - Siegfried Trattnig
- Department of Biomedical Imaging and Image-guided Therapy, High Field Magnetic Resonance Center, Medical University of Vienna, Austria
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Austria
| |
Collapse
|
208
|
Abstract
PURPOSE OF REVIEW Clinical MRI is of paramount importance for multiple sclerosis diagnosis but lacks the specificity to investigate the pathogenic mechanisms underlying disease onset and progression. The application of advanced MR sequences allows the characterization of diverse and complex pathological mechanisms, granting insights into multiple sclerosis natural history and response to treatment. RECENT FINDINGS This review provides an update on the most recent international guidelines for optimal standard imaging of multiple sclerosis and discusses advantages and limitations of advanced imaging approaches for investigating inflammation, demyelination and neurodegeneration. An overview is provided for methods devoted to imaging leptomeningeal enhancement, microglial activation, demyelination, neuronal metabolic damage and neuronal loss. SUMMARY The application of magnetic resonance (MR) guidelines to standard-of-care MR protocols, although still limited, would substantially contribute to the optimization of multiple sclerosis management. From an academic perspective, different mechanism-specific imaging techniques are available and offer a powerful tool to elucidate multiple sclerosis pathogenesis, monitor disease progression and guide therapeutic choices.
Collapse
|
209
|
Jonkman LE, Kenkhuis B, Geurts JJG, van de Berg WDJ. Post-Mortem MRI and Histopathology in Neurologic Disease: A Translational Approach. Neurosci Bull 2019; 35:229-243. [PMID: 30790214 DOI: 10.1007/s12264-019-00342-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/29/2018] [Indexed: 01/28/2023] Open
Abstract
In this review, combined post-mortem brain magnetic resonance imaging (MRI) and histology studies are highlighted, illustrating the relevance of translational approaches to define novel MRI signatures of neuropathological lesions in neuroinflammatory and neurodegenerative disorders. Initial studies combining post-mortem MRI and histology have validated various MRI sequences, assessing their sensitivity and specificity as diagnostic biomarkers in neurologic disease. More recent studies have focused on defining new radiological (bio)markers and implementing them in the clinical (research) setting. By combining neurological and neuroanatomical expertise with radiological development and pathological validation, a cycle emerges that allows for the discovery of novel MRI biomarkers to be implemented in vivo. Examples of this cycle are presented for multiple sclerosis, Alzheimer's disease, Parkinson's disease, and traumatic brain injury. Some applications have been shown to be successful, while others require further validation. In conclusion, there is much to explore with post-mortem MRI and histology studies, which can eventually be of high relevance for clinical practice.
Collapse
Affiliation(s)
- Laura E Jonkman
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
| | - Boyd Kenkhuis
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen J G Geurts
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| |
Collapse
|
210
|
Correale J, Marrodan M, Ysrraelit MC. Mechanisms of Neurodegeneration and Axonal Dysfunction in Progressive Multiple Sclerosis. Biomedicines 2019; 7:biomedicines7010014. [PMID: 30791637 PMCID: PMC6466454 DOI: 10.3390/biomedicines7010014] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/14/2019] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
Multiple Sclerosis (MS) is a major cause of neurological disability, which increases predominantly during disease progression as a result of cortical and grey matter structures involvement. The gradual accumulation of disability characteristic of the disease seems to also result from a different set of mechanisms, including in particular immune reactions confined to the Central Nervous System such as: (a) B-cell dysregulation, (b) CD8+ T cells causing demyelination or axonal/neuronal damage, and (c) microglial cell activation associated with neuritic transection found in cortical demyelinating lesions. Other potential drivers of neurodegeneration are generation of oxygen and nitrogen reactive species, and mitochondrial damage, inducing impaired energy production, and intra-axonal accumulation of Ca2+, which in turn activates a variety of catabolic enzymes ultimately leading to progressive proteolytic degradation of cytoskeleton proteins. Loss of axon energy provided by oligodendrocytes determines further axonal degeneration and neuronal loss. Clearly, these different mechanisms are not mutually exclusive and could act in combination. Given the multifactorial pathophysiology of progressive MS, many potential therapeutic targets could be investigated in the future. This remains however, an objective that has yet to be undertaken.
Collapse
Affiliation(s)
- Jorge Correale
- Department of Neurology, FLENI, Buenos Aires 1428, Argentina.
| | | | | |
Collapse
|
211
|
Lodygin D, Hermann M, Schweingruber N, Flügel-Koch C, Watanabe T, Schlosser C, Merlini A, Körner H, Chang HF, Fischer HJ, Reichardt HM, Zagrebelsky M, Mollenhauer B, Kügler S, Fitzner D, Frahm J, Stadelmann C, Haberl M, Odoardi F, Flügel A. β-Synuclein-reactive T cells induce autoimmune CNS grey matter degeneration. Nature 2019; 566:503-508. [DOI: 10.1038/s41586-019-0964-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 01/15/2019] [Indexed: 01/17/2023]
|
212
|
Fadda G, Brown RA, Magliozzi R, Aubert-Broche B, O'Mahony J, Shinohara RT, Banwell B, Marrie RA, Yeh EA, Collins DL, Arnold DL, Bar-Or A. A surface-in gradient of thalamic damage evolves in pediatric multiple sclerosis. Ann Neurol 2019; 85:340-351. [PMID: 30719730 PMCID: PMC6593844 DOI: 10.1002/ana.25429] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 02/02/2019] [Accepted: 02/02/2019] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Central nervous system pathology in multiple sclerosis includes both focal inflammatory perivascular injury and injury to superficial structures, including the subpial region of the cortex, which reportedly exhibits a gradient of damage from the surface inward. We assessed how early in the multiple sclerosis course a "surface-in" process of injury suggesting progressive biology may begin. METHODS We focused on the thalamus, which notably has both a cerebrospinal fluid (CSF) interface and a white matter interface. Thalamic volume trajectories were assessed in a prospectively followed cohort of children from initial presentation with either multiple sclerosis or monophasic acquired demyelination, and healthy controls. Voxelwise volume changes were calculated using deformation-based morphometry, and analyzed in relation to distance from the CSF interface by mixed effects modeling and semiparametric smoothing methods. RESULTS Twenty-seven children with multiple sclerosis and 73 children with monophasic demyelination were prospectively followed with yearly brain scans (mean follow-up = 4.6 years, standard deviation = 1.9). A total of 282 healthy children with serial scans were included as controls. Relative to healthy controls, children with multiple sclerosis and children with monophasic demyelination demonstrated volume loss in thalamic regions adjacent to the white matter. However, only children with multiple sclerosis exhibited an additional surface-in gradient of thalamic injury on the ventricular side, which was already notable in the first year of clinical disease (asymptote estimate = 3.01, 95% confidence interval [CI] = 1.44-4.58, p = 0.0002) and worsened over time (asymptote:time estimate = 0.33, 95% CI = 0.12-0.54, p = 0.0021). INTERPRETATION Our results suggest that a multiple sclerosis disease-specific surface-in process of damage can manifest at the earliest stages of the disease. ANN NEUROL 2019;85:340-351.
Collapse
Affiliation(s)
- Giulia Fadda
- Department of Neurology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA.,Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Robert A Brown
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Roberta Magliozzi
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Verona, Italy.,Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | | | - Julia O'Mahony
- Institute of Health Policy, Management, and Evaluation, University of Toronto/Hospital for Sick Children, Toronto, Ontario, Canada
| | - Russell T Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Brenda Banwell
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada.,Division of Neurology, Hospital for Sick Children, Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada.,Division of Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ruth Ann Marrie
- Departments of Internal Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - E Ann Yeh
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - D Louis Collins
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Douglas L Arnold
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Amit Bar-Or
- Department of Neurology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA.,Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.,Division of Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
213
|
Kocovski P, Jiang X, D'Souza CS, Li Z, Dang PT, Wang X, Chen W, Peter K, Hale MW, Orian JM. Platelet Depletion is Effective in Ameliorating Anxiety-Like Behavior and Reducing the Pro-Inflammatory Environment in the Hippocampus in Murine Experimental Autoimmune Encephalomyelitis. J Clin Med 2019; 8:jcm8020162. [PMID: 30717130 PMCID: PMC6406682 DOI: 10.3390/jcm8020162] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/21/2019] [Accepted: 01/27/2019] [Indexed: 01/16/2023] Open
Abstract
The neuropsychiatric symptoms of multiple sclerosis (MS), such as anxiety and depression, can result from disease activity itself as well as psychological reaction to an unfavorable diagnosis. Accordingly, the literature reports evidence of increased anxiety-like behavior in experimental autoimmune encephalomyelitis (EAE), an accepted MS model. Due to the recently described critical role of platelets in inflammation and autoimmune disease, we examined the relationship between platelets, inflammation, and anxiety-like behavior in EAE. In the elevated plus maze, EAE-induced C57BL/6J mice showed decreased time spent in the open arms relative to vehicle-only controls, demonstrating an increase in anxiety-like behavior. This effect occurred in the presence of platelet–neuron association, but absence of lymphocytic infiltration, in the hippocampal parenchyma. Platelet depletion at the pre-clinical disease stage, using antibody-mediated lysis prevented the EAE-induced increase in anxiety-like behavior, while no significant difference in distance moved was recorded. Furthermore, platelet depletion was also associated with reduction of the pro-inflammatory environment to control levels in the hippocampus and prevention of EAE disease symptomology. These studies demonstrate the high efficacy of a platelet-targeting approach in preventing anxiety-like symptoms and clinical manifestations of EAE and have implications for the treatment of neuropsychiatric symptoms in MS.
Collapse
Affiliation(s)
- Pece Kocovski
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Bundoora, VIC 3086, Australia.
| | - Xiangrui Jiang
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Bundoora, VIC 3086, Australia.
| | - Claretta S D'Souza
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Bundoora, VIC 3086, Australia.
| | - Zhenjiang Li
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Bundoora, VIC 3086, Australia.
| | - Phuc T Dang
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Bundoora, VIC 3086, Australia.
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia.
- Department of Medicine, Monash University, Melbourne, VIC 3800, Australia.
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Bundoora, VIC 3086, Australia.
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia.
- Department of Medicine, Monash University, Melbourne, VIC 3800, Australia.
| | - Matthew W Hale
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Bundoora, VIC 3086, Australia.
| | - Jacqueline M Orian
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Bundoora, VIC 3086, Australia.
| |
Collapse
|
214
|
Ohno N, Ikenaka K. Axonal and neuronal degeneration in myelin diseases. Neurosci Res 2019; 139:48-57. [DOI: 10.1016/j.neures.2018.08.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/22/2018] [Accepted: 08/29/2018] [Indexed: 12/14/2022]
|
215
|
Hundehege P, Cerina M, Eichler S, Thomas C, Herrmann AM, Göbel K, Müntefering T, Fernandez-Orth J, Bock S, Narayanan V, Budde T, Speckmann EJ, Wiendl H, Schubart A, Ruck T, Meuth SG. The next-generation sphingosine-1 receptor modulator BAF312 (siponimod) improves cortical network functionality in focal autoimmune encephalomyelitis. Neural Regen Res 2019; 14:1950-1960. [PMID: 31290453 PMCID: PMC6676873 DOI: 10.4103/1673-5374.259622] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Autoimmune diseases of the central nervous system (CNS) like multiple sclerosis (MS) are characterized by inflammation and demyelinated lesions in white and grey matter regions. While inflammation is present at all stages of MS, it is more pronounced in the relapsing forms of the disease, whereas progressive MS (PMS) shows significant neuroaxonal damage and grey and white matter atrophy. Hence, disease-modifying treatments beneficial in patients with relapsing MS have limited success in PMS. BAF312 (siponimod) is a novel sphingosine-1-phosphate receptor modulator shown to delay progression in PMS. Besides reducing inflammation by sequestering lymphocytes in lymphoid tissues, BAF312 crosses the blood-brain barrier and binds its receptors on neurons, astrocytes and oligodendrocytes. To evaluate potential direct neuroprotective effects, BAF312 was systemically or locally administered in the CNS of experimental autoimmune encephalomyelitis mice with distinct grey- and white-matter lesions (focal experimental autoimmune encephalomyelitis using an osmotic mini-pump). Ex-vivo flow cytometry revealed that systemic but not local BAF312 administration lowered immune cell infiltration in animals with both grey and white matter lesions. Ex-vivo voltage-sensitive dye imaging of acute brain slices revealed an altered spatio-temporal pattern of activation in the lesioned cortex compared to controls in response to electrical stimulation of incoming white-matter fiber tracts. Here, BAF312 administration showed partial restore of cortical neuronal circuit function. The data suggest that BAF312 exerts a neuroprotective effect after crossing the blood-brain barrier independently of peripheral effects on immune cells. Experiments were carried out in accordance with German and EU animal protection law and approved by local authorities (Landesamt für Natur, Umwelt und Verbraucherschutz Nordrhein-Westfalen; 87-51.04.2010.A331) on December 28, 2010.
Collapse
Affiliation(s)
- Petra Hundehege
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Manuela Cerina
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Susann Eichler
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Christian Thomas
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Alexander M Herrmann
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Kerstin Göbel
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Thomas Müntefering
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Juncal Fernandez-Orth
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Stefanie Bock
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Venu Narayanan
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Thomas Budde
- Institute of Physiology I, Westfälische Wilhelms-Universität, Münster, Germany
| | | | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Anna Schubart
- Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Tobias Ruck
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| |
Collapse
|
216
|
Effect of glatiramer acetate on cerebral grey matter pathology in patients with relapsing-remitting multiple sclerosis. Mult Scler Relat Disord 2019; 27:305-311. [DOI: 10.1016/j.msard.2018.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/08/2018] [Accepted: 11/11/2018] [Indexed: 01/17/2023]
|
217
|
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disorder. Although all MS patients initially show a relapsing-remitting course, 20-50% subsequently enter a chronic progressive course at 10-20 years after onset that greatly influences their activities of daily living. There are 2.5 million MS patients worldwide with large regional and racial differences. In particular, there are many MS patients among Caucasians living in Europe, while the disease is relatively rare in Asians and Africans.Although MS is regarded as an autoimmune disease, many factors such as genetic background, environmental factors, and sex are involved in its pathogenesis. While the immunological mechanisms remain to be fully elucidated, invasion of autoreactive T cells into the central nervous system (CNS) tissue is considered the first step of the disease. These T cells react with myelin antigens and initiate demyelination of the CNS by activating cytotoxic T cells, macrophages, and B cells through the release of inflammatory cytokines. As a treatment option, disease-modifying therapies have recently been developed to prevent the recurrence of MS in addition to conventional treatment with corticosteroids for acute relapse. However, there are still few effective treatments for the chronic progressive phase, and it is thus imperative to decipher the mechanism for chronic progression.
Collapse
|
218
|
Petracca M, Margoni M, Bommarito G, Inglese M. Monitoring Progressive Multiple Sclerosis with Novel Imaging Techniques. Neurol Ther 2018; 7:265-285. [PMID: 29956263 PMCID: PMC6283788 DOI: 10.1007/s40120-018-0103-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Indexed: 02/04/2023] Open
Abstract
Imaging markers for monitoring disease progression in progressive multiple sclerosis (PMS) are scarce, thereby limiting the possibility to monitor disease evolution and to test effective treatments in clinical trials. Advanced imaging techniques that have the advantage of metrics with increased sensitivity to short-term tissue changes and increased specificity to the structural abnormalities characteristic of PMS have recently been applied in clinical trials of PMS. In this review, we (1) provide an overview of the pathological features of PMS, (2) summarize the findings of research and clinical trials conducted in PMS which have applied conventional and advanced magnetic resonance imaging techniques and (3) discuss recent advancements and future perspectives in monitoring PMS with imaging techniques.
Collapse
Affiliation(s)
- Maria Petracca
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Monica Margoni
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Multiple Sclerosis Centre, Department of Neurosciences DNS, University Hospital, University of Padua, Padua, Italy
| | - Giulia Bommarito
- Department of Neuroscience, Rehabilitation, Genetics and Maternal and Perinatal Sciences, University of Genoa, Genoa, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Genetics and Maternal and Perinatal Sciences, University of Genoa, Genoa, Italy.
- Departments of Neurology, Radiology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
219
|
Piaggio N, Schiavi S, Martino M, Bommarito G, Inglese M, Magioncalda P. Exploring mania-associated white matter injury by comparison with multiple sclerosis: a diffusion tensor imaging study. Psychiatry Res Neuroimaging 2018; 281:78-84. [PMID: 30268035 DOI: 10.1016/j.pscychresns.2018.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/07/2018] [Accepted: 09/21/2018] [Indexed: 12/16/2022]
Abstract
Bipolar disorder (BD), especially in its active phases, has shown some neuroimaging and immunological similarities with multiple sclerosis (MS). The objective of this study was to compare white matter (WM) alterations in BD patients in manic phase (M-BD) and MS patients at early stage of disease and with low lesion burden. We compared diffusion tensor imaging (DTI)-derived fractional anisotropy (FA), mean diffusivity (MD) and radial diffusivity (RD) in a priori selected WM regions (i.e., corpus callosum and cingulum) betwixt 23 M-BD, 23 MS patients and 46 healthy controls. Both M-BD and MS showed WM changes in the corpus callosum, which, however, showed a greater impairment in MS patients. However, considering the different sub-regions of corpus callosum separately (i.e., genu, body, splenium), M-BD and MS presented an opposite pattern in spatial distribution of WM microstructure alterations, with a greater impairment in the anterior region in M-BD and in the posterior region in MS. Common features as well as divergent patterns in DTI changes are detected in M-BD and early MS, prompting a deeper investigation of analogies and differences in WM and immunological alterations of these disorders.
Collapse
Affiliation(s)
- Niccolò Piaggio
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Neurology, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Simona Schiavi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Neurology, University of Genoa, Genoa, Italy
| | - Matteo Martino
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genova, Genoa, Italy.
| | - Giulia Bommarito
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Neurology, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Neurology, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Neurology, Radiology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Paola Magioncalda
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genova, Genoa, Italy
| |
Collapse
|
220
|
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory, demyelinating and neurodegenerative disease of the central nervous system in young adults. This disorder is a heterogeneous, multifactorial, immune-mediated disease that is influenced by both genetic and environmental factors. In most patients, reversible episodes of neurological dysfunction lasting several days or weeks characterize the initial stages of the disease (that is, clinically isolated syndrome and relapsing-remitting MS). Over time, irreversible clinical and cognitive deficits develop. A minority of patients have a progressive disease course from the onset. The pathological hallmark of MS is the formation of demyelinating lesions in the brain and spinal cord, which can be associated with neuro-axonal damage. Focal lesions are thought to be caused by the infiltration of immune cells, including T cells, B cells and myeloid cells, into the central nervous system parenchyma, with associated injury. MS is associated with a substantial burden on society owing to the high cost of the available treatments and poorer employment prospects and job retention for patients and their caregivers.
Collapse
Affiliation(s)
- 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.
| | - Amit Bar-Or
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.,Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.,Neuroimmunology Unit, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Paolo Preziosa
- 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
| | - Alessandra Solari
- Unit of Neuroepidemiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sandra Vukusic
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-inflammation, Fondation Eugène Devic EDMUS Contre la Sclérose en Plaques, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Lyon, France
| | - 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
| |
Collapse
|
221
|
Bellizzi MJ, Hammond JW, Li H, Gantz Marker MA, Marker DF, Freeman RS, Gelbard HA. The Mixed-Lineage Kinase Inhibitor URMC-099 Protects Hippocampal Synapses in Experimental Autoimmune Encephalomyelitis. eNeuro 2018; 5:ENEURO.0245-18.2018. [PMID: 30627663 PMCID: PMC6325567 DOI: 10.1523/eneuro.0245-18.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/31/2018] [Accepted: 11/07/2018] [Indexed: 12/04/2022] Open
Abstract
Treatments to stop gray matter degeneration are needed to prevent progressive disability in multiple sclerosis (MS). We tested whether inhibiting mixed-lineage kinases (MLKs), which can drive inflammatory microglial activation and neuronal degeneration, could protect hippocampal synapses in C57BL/6 mice with experimental autoimmune encephalomyelitis (EAE), a disease model that recapitulates the excitatory synaptic injury that occurs widely within the gray matter in MS. URMC-099, a broad spectrum MLK inhibitor with additional activity against leucine-rich repeat kinase 2 (LRRK2) and other kinases, prevented loss of PSD95-positive postsynaptic structures, shifted activated microglia toward a less inflammatory phenotype, and reversed deficits in hippocampal-dependent contextual fear conditioning in EAE mice when administered after the onset of motor symptoms. A narrow spectrum inhibitor designed to be highly selective for MLK3 failed to protect synapses in EAE hippocampi, and could not rescue cultured neurons from trophic deprivation in an in vitro model of MLK-driven neuronal degeneration. These results suggest that URMC-099 may have potential as a neuroprotective treatment in MS and demonstrate that a broad spectrum of inhibition against a combination of MLK and other kinases is more effective in neuroinflammatory disease than selectively targeting a single kinase.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Calcium-Binding Proteins/metabolism
- Cells, Cultured
- Conditioning, Psychological/drug effects
- Cytokines/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Enzyme Inhibitors/therapeutic use
- Fear/drug effects
- Fear/psychology
- Female
- Hippocampus/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Microfilament Proteins/metabolism
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Neurons/drug effects
- Neuroprotective Agents/therapeutic use
- Peptide Fragments/toxicity
- Pyridines/therapeutic use
- Pyrroles/therapeutic use
- Superior Cervical Ganglion/cytology
- Synapses/drug effects
Collapse
Affiliation(s)
- Matthew J. Bellizzi
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY 14642
| | - Jennetta W. Hammond
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
| | - Herman Li
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
| | - Mary A. Gantz Marker
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Daniel F. Marker
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
| | - Robert S. Freeman
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Harris A. Gelbard
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY 14642
- Departments of Pediatrics and Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
222
|
Radecki DZ, Johnson EL, Brown AK, Meshkin NT, Perrine SA, Gow A. Corticohippocampal Dysfunction In The OBiden Mouse Model Of Primary Oligodendrogliopathy. Sci Rep 2018; 8:16116. [PMID: 30382234 PMCID: PMC6208344 DOI: 10.1038/s41598-018-34414-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 09/25/2018] [Indexed: 12/18/2022] Open
Abstract
Despite concerted efforts over decades, the etiology of multiple sclerosis (MS) remains unclear. Autoimmunity, environmental-challenges, molecular mimicry and viral hypotheses have proven equivocal because early-stage disease is typically presymptomatic. Indeed, most animal models of MS also lack defined etiologies. We have developed a novel adult-onset oligodendrogliopathy using a delineated metabolic stress etiology in myelinating cells, and our central question is, “how much of the pathobiology of MS can be recapitulated in this model?” The analyses described herein demonstrate that innate immune activation, glial scarring, cortical and hippocampal damage with accompanying electrophysiological, behavioral and memory deficits naturally emerge from disease progression. Molecular analyses reveal neurofilament changes in normal-appearing gray matter that parallel those in cortical samples from MS patients with progressive disease. Finally, axon initial segments of deep layer pyramidal neurons are perturbed in entorhinal/frontal cortex and hippocampus from OBiden mice, and computational modeling provides insight into vulnerabilities of action potential generation during demyelination and early remyelination. We integrate these findings into a working model of corticohippocampal circuit dysfunction to predict how myelin damage might eventually lead to cognitive decline.
Collapse
Affiliation(s)
- Daniel Z Radecki
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, 48201, USA.,Department of Comparative Biosciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, 53706, USA
| | - Elizabeth L Johnson
- Institute of Gerontology, Wayne State University, Detroit, MI, 48202, USA.,Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA
| | - Ashley K Brown
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Nicholas T Meshkin
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, 48201, USA.,Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Alexander Gow
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, 48201, USA. .,Carman and Ann Adams Department of Pediatrics, School of Medicine, Wayne State University, Detroit, MI, 48201, USA. .,Department of Neurology, School of Medicine, Wayne State University, Detroit, MI, 48201, USA.
| |
Collapse
|
223
|
Local volume fraction distributions of axons, astrocytes, and myelin in deep subcortical white matter. Neuroimage 2018; 179:275-287. [DOI: 10.1016/j.neuroimage.2018.06.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 05/31/2018] [Accepted: 06/11/2018] [Indexed: 01/28/2023] Open
|
224
|
Kritsilis M, V Rizou S, Koutsoudaki PN, Evangelou K, Gorgoulis VG, Papadopoulos D. Ageing, Cellular Senescence and Neurodegenerative Disease. Int J Mol Sci 2018; 19:E2937. [PMID: 30261683 PMCID: PMC6213570 DOI: 10.3390/ijms19102937] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/16/2018] [Accepted: 09/19/2018] [Indexed: 01/10/2023] Open
Abstract
Ageing is a major risk factor for developing many neurodegenerative diseases. Cellular senescence is a homeostatic biological process that has a key role in driving ageing. There is evidence that senescent cells accumulate in the nervous system with ageing and neurodegenerative disease and may predispose a person to the appearance of a neurodegenerative condition or may aggravate its course. Research into senescence has long been hindered by its variable and cell-type specific features and the lack of a universal marker to unequivocally detect senescent cells. Recent advances in senescence markers and genetically modified animal models have boosted our knowledge on the role of cellular senescence in ageing and age-related disease. The aim now is to fully elucidate its role in neurodegeneration in order to efficiently and safely exploit cellular senescence as a therapeutic target. Here, we review evidence of cellular senescence in neurons and glial cells and we discuss its putative role in Alzheimer's disease, Parkinson's disease and multiple sclerosis and we provide, for the first time, evidence of senescence in neurons and glia in multiple sclerosis, using the novel GL13 lipofuscin stain as a marker of cellular senescence.
Collapse
Affiliation(s)
- Marios Kritsilis
- Laboratory of Histology & Embryology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Goudi, 115-27 Athens, Greece.
| | - Sophia V Rizou
- Laboratory of Histology & Embryology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Goudi, 115-27 Athens, Greece.
| | - Paraskevi N Koutsoudaki
- Laboratory of Histology & Embryology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Goudi, 115-27 Athens, Greece.
| | - Konstantinos Evangelou
- Laboratory of Histology & Embryology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Goudi, 115-27 Athens, Greece.
| | - Vassilis G Gorgoulis
- Laboratory of Histology & Embryology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Goudi, 115-27 Athens, Greece.
| | - Dimitrios Papadopoulos
- Laboratory of Histology & Embryology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Goudi, 115-27 Athens, Greece.
| |
Collapse
|
225
|
Ellwardt E, Pramanik G, Luchtman D, Novkovic T, Jubal ER, Vogt J, Arnoux I, Vogelaar CF, Mandal S, Schmalz M, Barger Z, Ruiz de Azua I, Kuhlmann T, Lutz B, Mittmann T, Bittner S, Zipp F, Stroh A. Maladaptive cortical hyperactivity upon recovery from experimental autoimmune encephalomyelitis. Nat Neurosci 2018; 21:1392-1403. [DOI: 10.1038/s41593-018-0193-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/17/2018] [Indexed: 12/14/2022]
|
226
|
Therapeutic Advances and Challenges in the Treatment of Progressive Multiple Sclerosis. Drugs 2018; 78:1549-1566. [DOI: 10.1007/s40265-018-0984-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
227
|
Juźwik CA, Drake S, Lécuyer MA, Johnson RM, Morquette B, Zhang Y, Charabati M, Sagan SM, Bar-Or A, Prat A, Fournier AE. Neuronal microRNA regulation in Experimental Autoimmune Encephalomyelitis. Sci Rep 2018; 8:13437. [PMID: 30194392 PMCID: PMC6128870 DOI: 10.1038/s41598-018-31542-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/21/2018] [Indexed: 01/02/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune, neurodegenerative disease but the molecular mechanisms underlying neurodegenerative aspects of the disease are poorly understood. microRNAs (miRNAs) are powerful regulators of gene expression that regulate numerous mRNAs simultaneously and can thus regulate programs of gene expression. Here, we describe miRNA expression in neurons captured from mice subjected to experimental autoimmune encephalomyelitis (EAE), a model of central nervous system (CNS) inflammation. Lumbar motor neurons and retinal neurons were laser captured from EAE mice and miRNA expression was assessed by next-generation sequencing and validated by qPCR. We describe 14 miRNAs that are differentially regulated in both neuronal subtypes and determine putative mRNA targets though in silico analysis. Several upregulated neuronal miRNAs are predicted to target pathways that could mediate repair and regeneration during EAE. This work identifies miRNAs that are affected by inflammation and suggests novel candidates that may be targeted to improve neuroprotection in the context of pathological inflammation.
Collapse
Affiliation(s)
- Camille A Juźwik
- McGill University, Montréal Neurological Institute, Montréal, QC, H3A 2B4, Canada
| | - Sienna Drake
- McGill University, Montréal Neurological Institute, Montréal, QC, H3A 2B4, Canada
| | - Marc-André Lécuyer
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Radia Marie Johnson
- McGill University, Goodman Cancer Research Centre, Montréal, H3A 1A3, Canada
| | - Barbara Morquette
- McGill University, Montréal Neurological Institute, Montréal, QC, H3A 2B4, Canada
| | - Yang Zhang
- McGill University, Montréal Neurological Institute, Montréal, QC, H3A 2B4, Canada
| | - Marc Charabati
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Selena M Sagan
- McGill University, Departments of Microbiology & Immunology and Biochemistry, Montréal, QC, H3G 0B1, Canada
| | - Amit Bar-Or
- McGill University, Montréal Neurological Institute, Montréal, QC, H3A 2B4, Canada
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alexandre Prat
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Alyson E Fournier
- McGill University, Montréal Neurological Institute, Montréal, QC, H3A 2B4, Canada.
| |
Collapse
|
228
|
Magliozzi R, Reynolds R, Calabrese M. MRI of cortical lesions and its use in studying their role in MS pathogenesis and disease course. Brain Pathol 2018; 28:735-742. [PMID: 30020563 PMCID: PMC8028295 DOI: 10.1111/bpa.12642] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/21/2018] [Indexed: 02/01/2023] Open
Abstract
Cortical grey matter (GM) demyelination is present from the earliest stages of multiple sclerosis (MS) and is associated with physical deficits and cognitive impairment. In particular, the rate of disability progression in MS, both in the relapsing and progressive phases, appears to be strictly associated with degenerative GM demyelination and diffuse cortical atrophy. In the last decade, several histopathological studies and advanced radiological methodologies have contributed to better identify the exact involvement/load of cortical pathology in MS, even if the specific inflammatory features and the precise cell and molecular mechanisms of GM demyelination and neurodegeneration in MS remain still not fully understood. It has been proposed that a combined neuropathology, imaging and molecular approach may help to define a more detailed characterization and precise assessment of the heterogeneous features of GM injury and inflammation in MS. This, in turn, will possibly identify specific imaging and biohumoral (cerebrospinal fluid/serum) correlates of cortical pathology that may have an important role in predicting and monitor the disease evolution.
Collapse
Affiliation(s)
- R. Magliozzi
- Neurology BDepartment of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
- Division of Brain SciencesDepartment of MedicineImperial College LondonLondonUnited Kingdom
| | - R. Reynolds
- Division of Brain SciencesDepartment of MedicineImperial College LondonLondonUnited Kingdom
| | - M. Calabrese
- Neurology BDepartment of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
| |
Collapse
|
229
|
7 Tesla magnetic resonance imaging of caudal anterior cingulate and posterior cingulate cortex atrophy in patients with trigeminal neuralgia. Magn Reson Imaging 2018; 51:144-150. [DOI: 10.1016/j.mri.2018.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/14/2018] [Accepted: 05/14/2018] [Indexed: 01/10/2023]
|
230
|
Luo Q, Ding L, Zhang N, Jiang Z, Gao C, Xue L, Peng B, Wang G. A stable and easily reproducible model of focal white matter demyelination. J Neurosci Methods 2018; 307:230-239. [DOI: 10.1016/j.jneumeth.2018.05.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 05/23/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022]
|
231
|
MacKenzie‐Graham A, Brook J, Kurth F, Itoh Y, Meyer C, Montag MJ, Wang H, Elashoff R, Voskuhl RR. Estriol-mediated neuroprotection in multiple sclerosis localized by voxel-based morphometry. Brain Behav 2018; 8:e01086. [PMID: 30144306 PMCID: PMC6160650 DOI: 10.1002/brb3.1086] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/05/2018] [Accepted: 07/08/2018] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Progressive gray matter (GM) atrophy is a hallmark of multiple sclerosis (MS). Cognitive impairment has been observed in 40%-70% of MS patients and has been linked to GM atrophy. In a phase 2 trial of estriol treatment in women with relapsing-remitting MS (RRMS), higher estriol levels correlated with greater improvement on the paced auditory serial addition test (PASAT) and imaging revealed sparing of localized GM in estriol-treated compared to placebo-treated patients. To better understand the significance of this GM sparing, the current study explored the relationships between the GM sparing and traditional MRI measures and clinical outcomes. METHODS Sixty-two estriol- and forty-nine placebo-treated RRMS patients underwent clinical evaluations and brain MRI. Voxel-based morphometry (VBM) was used to evaluate voxelwise GM sparing from high-resolution T1-weighted scans. RESULTS A region of treatment-induced sparing (TIS) was defined as the areas where GM was spared in estriol- as compared to placebo-treated groups, localized primarily within the frontal and parietal cortices. We observed that TIS volume was directly correlated with improvement on the PASAT. Next, a longitudinal cognitive disability-specific atlas (DSA) was defined by correlating voxelwise GM volumes with PASAT scores, that is, areas where less GM correlated with less improvement in PASAT scores. Finally, overlap between the TIS and the longitudinal cognitive DSA revealed a specific region of cortical GM that was preserved in estriol-treated subjects that was associated with better performance on the PASAT. CONCLUSIONS Discovery of this region of overlap was biology driven, not based on an a priori structure of interest. It included the medial frontal cortex, an area previously implicated in problem solving and attention. These findings indicate that localized GM sparing during estriol treatment was associated with improvement in cognitive testing, suggesting a clinically relevant, disability-specific biomarker for clinical trials of candidate neuroprotective treatments in MS.
Collapse
Affiliation(s)
- Allan MacKenzie‐Graham
- Department of NeurologyAhmanson‐Lovelace Brain Mapping CenterDavid Geffen School of Medicine at UCLALos AngelesCalifornia
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Jenny Brook
- Department of BiomathematicsDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Florian Kurth
- Department of NeurologyAhmanson‐Lovelace Brain Mapping CenterDavid Geffen School of Medicine at UCLALos AngelesCalifornia
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Yuichiro Itoh
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Cassandra Meyer
- Department of NeurologyAhmanson‐Lovelace Brain Mapping CenterDavid Geffen School of Medicine at UCLALos AngelesCalifornia
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Michael J. Montag
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - He‐Jing Wang
- Department of BiomathematicsDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Robert Elashoff
- Department of BiomathematicsDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| | - Rhonda R. Voskuhl
- UCLA Multiple Sclerosis ProgramDepartment of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCalifornia
| |
Collapse
|
232
|
Khachanova NV. [What do we know about the pathology of gray matter in multiple sclerosis]. Zh Nevrol Psikhiatr Im S S Korsakova 2018; 118:18-22. [PMID: 30160663 DOI: 10.17116/jnevro201811808218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The emergence of modern methods of immunohistochemistry and further development of MRI has led to a deeper understanding of gray matter (GM) pathology in multiple sclerosis (MS). GM involvement can be extensive including both demyelination (cortical lesions) and neuroaxonal damage. The mechanisms of GM damage in MS remain insufficiently studied. There are two concepts: the lesion of GM is primary and is paralleled by changes in white matter (WM), or secondary, i.e. it is a consequence of the pathological process in WM. More research into GM pathology using the latest MRI techniques will contribute to the understanding of pathological changes in both cortical and subcortical GM.
Collapse
Affiliation(s)
- N V Khachanova
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
233
|
Trapp BD, Vignos M, Dudman J, Chang A, Fisher E, Staugaitis SM, Battapady H, Mork S, Ontaneda D, Jones SE, Fox RJ, Chen J, Nakamura K, Rudick RA. Cortical neuronal densities and cerebral white matter demyelination in multiple sclerosis: a retrospective study. Lancet Neurol 2018; 17:870-884. [PMID: 30143361 DOI: 10.1016/s1474-4422(18)30245-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 05/18/2018] [Accepted: 06/11/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Demyelination of cerebral white matter is thought to drive neuronal degeneration and permanent neurological disability in individuals with multiple sclerosis. Findings from brain MRI studies, however, support the possibility that demyelination and neuronal degeneration can occur independently. We aimed to establish whether post-mortem brains from patients with multiple sclerosis show pathological evidence of cortical neuronal loss that is independent of cerebral white-matter demyelination. METHODS Brains and spinal cords were removed at autopsy from patients, who had died with multiple sclerosis, at the Cleveland Clinic in Cleveland, OH, USA. Visual examination of centimetre-thick slices of cerebral hemispheres was done to identify brains without areas of cerebral white-matter discoloration that were indicative of demyelinated lesions (referred to as myelocortical multiple sclerosis) and brains that had cerebral white-matter discolorations or demyelinated lesions (referred to as typical multiple sclerosis). These individuals with myelocortical multiple sclerosis were matched by age, sex, MRI protocol, multiple sclerosis disease subtype, disease duration, and Expanded Disability Status Scale, with individuals with typical multiple sclerosis. Demyelinated lesion area in tissue sections of cerebral white matter, spinal cord, and cerebral cortex from individuals classed as having myelocortical and typical multiple sclerosis were compared using myelin protein immunocytochemistry. Neuronal densities in cortical layers III, V, and VI from five cortical regions not directly connected to spinal cord (cingulate gyrus and inferior frontal cortex, superior temporal cortex, and superior insular cortex and inferior insular cortex) were also compared between the two groups and with aged-matched post-mortem brains from individuals without evidence of neurological disease. FINDINGS Brains and spinal cords were collected from 100 deceased patients between May, 1998, and November, 2012, and this retrospective study was done between Sept 6, 2011, and Feb 2, 2018. 12 individuals were identified as having myelocortical multiple sclerosis and were compared with 12 individuals identified as having typical multiple sclerosis. Demyelinated lesions were detected in spinal cord and cerebral cortex, but not in cerebral white matter, of people with myelocortical multiple sclerosis. Cortical demyelinated lesion area was similar between myelocortical and typical multiple sclerosis (median 4·45% [IQR 2·54-10·81] in myelocortical vs 9·74% [1·35-19·50] in typical multiple sclerosis; p=0·5512). Spinal cord demyelinated area was significantly greater in typical than in myelocortical multiple sclerosis (median 3·81% [IQR 1·72-7·42] in myelocortical vs 13·81% [6·51-29·01] in typical multiple sclerosis; p=0·0083). Despite the lack of cerebral white-matter demyelination in myelocortical multiple sclerosis, mean cortical neuronal densities were significantly decreased compared with control brains (349·8 neurons per mm2 [SD 51·9] in myelocortical multiple sclerosis vs 419·0 [43·6] in controls in layer III [p=0·0104]; 355·6 [46·5] vs 454·2 [48·3] in layer V [p=0·0006]; 366·6 [50·9] vs 458·3 [48·4] in layer VI [p=0·0049]). By contrast, mean cortical neuronal densities were decreased in typical multiple sclerosis brains compared with those from controls in layer V (392·5 [59·0] vs 454·2 [48·3]; p=0·0182) but not layers III and VI. INTERPRETATION We propose that myelocortical multiple sclerosis is a subtype of multiple sclerosis that is characterised by demyelination of spinal cord and cerebral cortex but not of cerebral white matter. Cortical neuronal loss is not accompanied by cerebral white-matter demyelination and can be an independent pathological event in myelocortical multiple sclerosis. Compared with control brains, cortical neuronal loss was greater in myelocortical multiple sclerosis cortex than in typical multiple sclerosis cortex. The molecular mechanisms of primary neuronal degeneration and axonal pathology in myelocortical multiple sclerosis should be investigated in future studies. FUNDING US National Institutes of Health and National Multiple Sclerosis Society.
Collapse
Affiliation(s)
- Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Megan Vignos
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Jessica Dudman
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ansi Chang
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Elizabeth Fisher
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Susan M Staugaitis
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Pathology, Pathology and Laboratory, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Sverre Mork
- Department of Pathology, Haukeland University Hospital, Bergen, Norway; Gade Lab for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Daniel Ontaneda
- Medicine Institute, Mellen Center for Treatment and Research in Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Robert J Fox
- Medicine Institute, Mellen Center for Treatment and Research in Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jacqueline Chen
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kunio Nakamura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Richard A Rudick
- Medicine Institute, Mellen Center for Treatment and Research in Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
234
|
Swanson A, Wolf T, Sitzmann A, Willette AA. Neuroinflammation in Alzheimer's disease: Pleiotropic roles for cytokines and neuronal pentraxins. Behav Brain Res 2018; 347:49-56. [PMID: 29462653 PMCID: PMC5988985 DOI: 10.1016/j.bbr.2018.02.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/30/2018] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is a potential factor speculated to underlie Alzheimer's disease (AD) etiopathogenesis and progression. The overwhelming focus in this area of research to date has been on the chronic upregulation of pro-inflammatory cytokines to understand how neuroinflammatory mechanisms contribute to neurodegeneration. Yet, it is important to understand the pleiotropic roles of these cytokines in modulating neuroinflammation in which they cannot be labeled as a strictly "good" or "bad" biomarker phenotype. As such, biomarkers with more precise functions are needed to better understand how neuroinflammation impacts the brain in AD. Neuronal pentraxins are a concentration- dependent group of pro- or anti- inflammatory cytokines. There is contradictory evidence of these pentraxins as being both neuroprotective and potentially detrimental in AD. Potential neuroprotective examples include their ability to predict AD-related outcomes such as cognition, memory function and synaptic refinement. This review will briefly outline the basis of AD and subsequently summarize findings for neuropathological mechanisms of neuroinflammation, roles for traditional pro-and anti-inflammatory cytokines, and data found thus far on the neuronal pentraxins.
Collapse
Affiliation(s)
- Ashley Swanson
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Tovah Wolf
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Alli Sitzmann
- Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States.
| | - Auriel A Willette
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States; Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States; Department of Biomedical Sciences, Iowa State University, 2008 Veterinary Medicine, Ames, IA 50011, United States; Department of Neurology, University of Iowa, 2007 Roy Carver Pavilion, 200 Hawkins Drive, Iowa City, IA 52242, United States.
| |
Collapse
|
235
|
Retinal ganglion cell function in recovered optic neuritis: Faster is not better. Clin Neurophysiol 2018; 129:1813-1818. [PMID: 29981956 DOI: 10.1016/j.clinph.2018.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/25/2018] [Accepted: 06/13/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To assess residual retinal ganglion cell (RGC) function in patients with recovered optic neuritis (ON) and multiple sclerosis (MS). METHODS Age-matched controls (C, n = 32) and MS patients (n = 17) with history of ON in one eye but normal visual acuity and color vision were tested with steady-state Pattern Electroretinogram (PERG). Light Emitting Diodes (LED)-generated bar gratings, robust signal averaging and Fourier analysis were used to assess response amplitude and latency. RESULTS PERG amplitude was similar for C, ON and fellow eyes (FE) (P = 0.4), but PERG latency was shortened in ON by 3.2 ms (P = 0.002) and in FE by 2.0 ms (P = 0.02) and was correlated (P < 0.01) with both Retinal Nerve Fiber Layer (RNFL) and Ganglion Cell Inner Plexiform Layer (GCIPL) thicknesses. PERG latency shortening could be simulated in control subjects (n = 8) by dioptrically blurring the edges of gratings (high spatial frequencies), which reduced activity of parvocellular RGCs with smaller/slower axons. The blurred PERG latency was shorter than baseline by 2.9 ms (P = 0.01). CONCLUSIONS PERG latency is shortened in both eyes of MS patients with recovered unilateral ON, suggesting relative dysfunction of RGCs with slower axons and sparing of RGCs with faster axons. SIGNIFICANCE Assessment of PERG latency in MS and ON may help identifying and monitoring RGC dysfunction. PERG latency shortening in FE suggests primary retinopathy in MS.
Collapse
|
236
|
Bajrami A, Pitteri M, Castellaro M, Pizzini F, Romualdi C, Montemezzi S, Monaco S, Calabrese M. The effect of fingolimod on focal and diffuse grey matter damage in active MS patients. J Neurol 2018; 265:2154-2161. [PMID: 29938336 DOI: 10.1007/s00415-018-8952-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 01/14/2023]
Abstract
INTRODUCTION The mechanism of action of fingolimod within the central nervous system and its efficacy in reducing/preventing both focal and diffuse grey matter (GM) damage in active multiple sclerosis (MS) are not completely understood. METHODS In this longitudinal, 2-year prospective, phase IV, single-blind study, 40 MS patients treated with fingolimod and 39 untreated age, gender, and disability-matched MS patients were enrolled. Each patient underwent a neurological examination every 6 months and a 3T MRI at the beginning of the treatment and after 24 months. The accumulation of new cortical lesions (CLs) and the progression of regional GM atrophy were compared between the two groups. RESULTS At the end of the study (T24), the percentage of patients with new CLs (13.5 vs. 89%, p < 0.001) and the percentage of GM volume change was lower in the treated group (p < 0.001). The regional analysis revealed that the treated group had also less volume loss in thalamus, caudatus, globus pallidus, cingulate cortex, and hippocampus (p < 0.001), as well as in, cerebellum, superior frontal gyrus, and insular-long gyrus (p < 0.05). Patients with no evidence of disease activity were 60% in the treated group and 10% in the untreated group (p < 0.001). CONCLUSIONS These results suggest a possible protective effect of fingolimod on focal and diffuse GM damage.
Collapse
Affiliation(s)
- Albulena Bajrami
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Policlinico "G.B. Rossi" Borgo Roma, Piazzale L.A. Scuro, 10, 37134, Verona, Italy
| | - Marco Pitteri
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Policlinico "G.B. Rossi" Borgo Roma, Piazzale L.A. Scuro, 10, 37134, Verona, Italy
| | - Marco Castellaro
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Policlinico "G.B. Rossi" Borgo Roma, Piazzale L.A. Scuro, 10, 37134, Verona, Italy
- Department of Information Engineering, University of Padova, Padua, Italy
| | - Francesca Pizzini
- Neuroradiology and Radiology Units, Department of Diagnostic and Pathology, University Hospital of Verona, Verona, Italy
| | | | - Stefania Montemezzi
- Neuroradiology and Radiology Units, Department of Diagnostic and Pathology, University Hospital of Verona, Verona, Italy
| | - Salvatore Monaco
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Policlinico "G.B. Rossi" Borgo Roma, Piazzale L.A. Scuro, 10, 37134, Verona, Italy
| | - Massimiliano Calabrese
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Policlinico "G.B. Rossi" Borgo Roma, Piazzale L.A. Scuro, 10, 37134, Verona, Italy.
| |
Collapse
|
237
|
2017 McDonald diagnostic criteria: A review of the evidence. Mult Scler Relat Disord 2018; 24:48-54. [PMID: 29936325 DOI: 10.1016/j.msard.2018.05.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/03/2018] [Accepted: 05/12/2018] [Indexed: 11/20/2022]
Abstract
The diagnosis of Multiple Sclerosis (MS) has continuously evolved, allowing for an earlier and more accurate diagnosis of MS over time. The McDonald Criteria for diagnosis of MS were originally proposed in 2001, with previous revisions in both 2005 and 2010. The International Panel on Diagnosis in MS have recently reviewed the 2010 McDonald Criteria, and made recommendations for the revised 2017 McDonald Criteria. Any revisions made relied entirely on the available evidence, and not expert opinion. In this review, we provide an overview of the recent 2017 revisions to the McDonald Criteria, focusing in particular on the motivating evidence behind the recommendations made. We also review the existing research around misdiagnosis in MS, as well as areas considered to be high priorities of research, currently lacking in sufficient evidence, which may influence future diagnostic criteria in years to come. Finally, we illustrate some clinical examples, to demonstrate the impact of new diagnostic criteria on time to MS diagnosis in a real-world setting.
Collapse
|
238
|
Ghai S, Ghai I. Effects of Rhythmic Auditory Cueing in Gait Rehabilitation for Multiple Sclerosis: A Mini Systematic Review and Meta-Analysis. Front Neurol 2018; 9:386. [PMID: 29942278 PMCID: PMC6004404 DOI: 10.3389/fneur.2018.00386] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/11/2018] [Indexed: 12/15/2022] Open
Abstract
Rhythmic auditory cueing has been shown to enhance gait performance in several movement disorders. The "entrainment effect" generated by the stimulations can enhance auditory motor coupling and instigate plasticity. However, a consensus as to its influence over gait training among patients with multiple sclerosis is still warranted. A systematic review and meta-analysis was carried out to analyze the effects of rhythmic auditory cueing in studies gait performance in patients with multiple sclerosis. This systematic identification of published literature was performed according to PRISMA guidelines, from inception until Dec 2017, on online databases: Web of science, PEDro, EBSCO, MEDLINE, Cochrane, EMBASE, and PROQUEST. Studies were critically appraised using PEDro scale. Of 602 records, five studies (PEDro score: 5.7 ± 1.3) involving 188 participants (144 females/40 males) met our inclusion criteria. The meta-analysis revealed enhancements in spatiotemporal parameters of gait i.e., velocity (Hedge's g: 0.67), stride length (0.70), and cadence (1.0), and reduction in timed 25 feet walking test (-0.17). Underlying neurophysiological mechanisms, and clinical implications are discussed. This present review bridges the gaps in literature by suggesting application of rhythmic auditory cueing in conventional rehabilitation approaches to enhance gait performance in the multiple sclerosis community.
Collapse
Affiliation(s)
- Shashank Ghai
- Institute of Sports Science, Leibniz University Hanover, Hanover, Germany
| | - Ishan Ghai
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| |
Collapse
|
239
|
Aktas O, Wattjes MP, Stangel M, Hartung HP. Diagnose der Multiplen Sklerose: Revision der McDonald-Kriterien 2017. DER NERVENARZT 2018; 89:1344-1354. [DOI: 10.1007/s00115-018-0550-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
240
|
The role of the cerebellum in multiple sclerosis—150 years after Charcot. Neurosci Biobehav Rev 2018; 89:85-98. [DOI: 10.1016/j.neubiorev.2018.02.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 01/02/2018] [Accepted: 02/18/2018] [Indexed: 12/22/2022]
|
241
|
Sati P. Diagnosis of multiple sclerosis through the lens of ultra-high-field MRI. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2018; 291:101-109. [PMID: 29705032 PMCID: PMC6022748 DOI: 10.1016/j.jmr.2018.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 06/08/2023]
Abstract
The long-standing relationship between ultra-high-field (7 T) MRI and multiple sclerosis (MS) has brought new insights to our understanding of lesion evolution and its associated pathology. With the recent FDA approval of a commercially available scanner, 7 T MRI is finally entering the clinic with great expectations about its potential added value. By looking through the prism of MS diagnosis, this perspective article discusses current limitations and prospects of 7 T MRI techniques relevant to helping clinicians diagnose patients encountered in daily practice.
Collapse
Affiliation(s)
- Pascal Sati
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive MSC 1400, Building 10 Room 5C103, Bethesda, MD 20852, USA.
| |
Collapse
|
242
|
Xing B, Brink LE, Maers K, Sullivan ML, Bodnar RJ, Stolz DB, Cambi F. Conditional depletion of GSK3b protects oligodendrocytes from apoptosis and lessens demyelination in the acute cuprizone model. Glia 2018; 66:1999-2012. [PMID: 29761559 DOI: 10.1002/glia.23453] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 02/04/2023]
Abstract
Apoptosis is recognized as the main mechanism of oligodendrocyte loss in Multiple Sclerosis caused either by immune mediated injury (Barnett & Prineas, ) or a direct degenerative process (oligodendrogliapathy; Lucchinetti et al., ). Cuprizone induced demyelination is the result of non-immune mediated apoptosis of oligodendrocytes (OL) and represents a model of oligodendrogliapathy (Simmons, Pierson, Lee, & Goverman, ). Glycogen Synthase Kinase (GSK) 3b has been shown to be pro-apoptotic for cells other than OL. Here, we sought to investigate whether GSK3b plays a role in cuprizone-induced apoptosis of OL by using a novel inducible conditional knockout (cKO) of GSK3b in mature OL. While depletion of GSK3b has no effect on survival of uninjured OL, it increases survival of mature OL exposed to cuprizone. We show that GSK3b-deficient OLs are protected against caspase-dependent, but not against caspase-independent apoptosis. Active GSK3b is present in the nuclei of OL at peak of caspase-dependent apoptosis. Significant preservation of myelinated axons is associated with GSK3b depletion and glial cell activation is markedly reduced. Collectively, the data show that GSK3b is pro-apoptotic for caspase-dependent cell death, likely through activation of nuclear GSK3b and its depletion promotes survival of oligodendrocytes and attenuates myelin loss.
Collapse
Affiliation(s)
- Bin Xing
- Veterans Administration Pittsburgh, University Drive C Bldg 30, Pittsburgh, Pennsylvania
| | - Lauren E Brink
- Veterans Administration Pittsburgh, University Drive C Bldg 30, Pittsburgh, Pennsylvania
| | - Kelly Maers
- Veterans Administration Pittsburgh, University Drive C Bldg 30, Pittsburgh, Pennsylvania
| | - Mara L Sullivan
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Richard J Bodnar
- Veterans Administration Pittsburgh, University Drive C Bldg 30, Pittsburgh, Pennsylvania
| | - Donna B Stolz
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Franca Cambi
- Veterans Administration Pittsburgh, University Drive C Bldg 30, Pittsburgh, Pennsylvania.,Department of Neurology/PIND, University of Pittsburgh, 3501 5th Avenue, Pittsburgh, Pennsylvania.,Department of Neurology, University of Kentucky, 800 Rose St, Lexington, Kentucky
| |
Collapse
|
243
|
van Wageningen TA, van Dam AM. Much, if not all, of the cortical damage in MS can be attributed to the microglial cell - Yes. Mult Scler 2018; 24:895-896. [PMID: 29754520 PMCID: PMC6029145 DOI: 10.1177/1352458517739139] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Thecla A van Wageningen
- Department of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Anne-Marie van Dam
- Department of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
244
|
Frau J, Fenu G, Signori A, Coghe G, Lorefice L, Barracciu MA, Sechi V, Cabras F, Badas M, Marrosu MG, Cocco E. A cross-sectional and longitudinal study evaluating brain volumes, RNFL, and cognitive functions in MS patients and healthy controls. BMC Neurol 2018; 18:67. [PMID: 29751782 PMCID: PMC5946463 DOI: 10.1186/s12883-018-1065-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/30/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The principal biomarker of neurodegeneration in multiple sclerosis (MS) is believed to be brain volume, which is associated with cognitive functions and retinal nerve fibre layer (RNFL). A cross-sectional and longitudinal assessment of the relationship between RNFL, cognitive functions and brain volume. METHODS At baseline, relapsing patients and healthy controls underwent 1.5 T MRI to estimate the normalized volume of brain (NBV), grey (NGV), white (NWV) and peripheral grey (pNGV) matter. Cognitive functions were evaluated by BICAMS, RNFL by Spectral-Domain OCT. Patients were re-evaluated after 12 months. RESULTS Cognitive functions, brain volume, and RNFL differed between the group of 66 patients and that of 16 healthy controls. In the MS group, at baseline, an association was found between: p-NGV and symbol-digit (SDMT) (p = 0.022); temporal-RNFL and NBV (p = 0.007), NWV (p = 0.012), NGV (p = 0.048), and p-NGV (p = 0.021); papillo-macular bundle-RNFL and NBV (p = 0.013), NWV (p = 0.02), NGV (p = 0.049), and p-NGV (p = 0.032). Over the observational period, we found a reduction of brain volume (p < 0.001), average-RNFL (p = 0.001), temporal-RNFL (p = 0.006), and papillo-macular bundle-RNFL (p = 0.009). No association was found between OCT, MRI, and cognitive changes. CONCLUSIONS Brain volume, cognitive functions, and RNFL are continuous measures of different neurodegenerative aspects. BICAMS and OCT have low costs and can be easily used in clinical practice to monitor neurodegeneration.
Collapse
Affiliation(s)
- Jessica Frau
- Multiple Sclerosis Center Binaghi Hospital, Department of Medical Sciences and Public Health, University of Cagliari, via Is Guadazzonis 2, 09126 Cagliari, Italy
| | - Giuseppe Fenu
- Multiple Sclerosis Center Binaghi Hospital, Department of Medical Sciences and Public Health, University of Cagliari, via Is Guadazzonis 2, 09126 Cagliari, Italy
| | - Alessio Signori
- Department of Health Sciences, Section of Biostatistics, University of Genova, Via Pastore, 1, 16132 Genoa, Italy
| | - Giancarlo Coghe
- Multiple Sclerosis Center Binaghi Hospital, Department of Medical Sciences and Public Health, University of Cagliari, via Is Guadazzonis 2, 09126 Cagliari, Italy
| | - Lorena Lorefice
- Multiple Sclerosis Center Binaghi Hospital, Department of Medical Sciences and Public Health, University of Cagliari, via Is Guadazzonis 2, 09126 Cagliari, Italy
| | | | - Vincenzo Sechi
- Unit of Radiology, Binaghi Hospital, ATS Sardegna, via Is Guadazzonis 2, 09126 Cagliari, Italy
| | - Federico Cabras
- Unit of Radiology, Binaghi Hospital, ATS Sardegna, via Is Guadazzonis 2, 09126 Cagliari, Italy
| | - Mauro Badas
- Multiple Sclerosis Center Binaghi Hospital, Department of Medical Sciences and Public Health, University of Cagliari, via Is Guadazzonis 2, 09126 Cagliari, Italy
| | - Maria Giovanna Marrosu
- Multiple Sclerosis Center Binaghi Hospital, Department of Medical Sciences and Public Health, University of Cagliari, via Is Guadazzonis 2, 09126 Cagliari, Italy
| | - Eleonora Cocco
- Multiple Sclerosis Center Binaghi Hospital, Department of Medical Sciences and Public Health, University of Cagliari, via Is Guadazzonis 2, 09126 Cagliari, Italy
| |
Collapse
|
245
|
Boyko AN, Boyko OV. Cladribine tablets' potential role as a key example of selective immune reconstitution therapy in multiple sclerosis. Degener Neurol Neuromuscul Dis 2018; 8:35-44. [PMID: 30050387 PMCID: PMC6053904 DOI: 10.2147/dnnd.s161450] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Multiple sclerosis (MS) is one of the most important, disabling, and prevalent neurological disorders of young adults. It is a chronic inflammatory and neurodegenerative disease when autoreactive B and T cells have downstream effects that result in demyelination and neuronal loss. Anti-inflammatory disease-modifying therapies do have proven efficacy in delaying disease and disability progression in MS. While the progress in MS treatments has already improved the prognosis and quality of patients’ lives overall, there are some clear shortcomings and unmet needs in the current MS treatment landscape. The most promising means of MS treatment is selective immune reconstitution therapy (SIRT). This therapy is given in short-duration courses of immunosuppression, producing durable effects on the immune system and preventing nervous tissue loss. This review discusses the mechanisms of action and the data of clinical trials of cladribine tablets as an example of SIRT in MS. The clinical benefits of cladribine tablets in these studies include decreased relapse rate and disability progression with large reductions in lesion activity, and protection against brain volume loss. Whether all of these neurological findings are direct results of lymphocyte depletion, or if there are downstream effects on other, unknown, neurodegenerative processes are yet to be determined, but these clearly point to an interesting area of research.
Collapse
Affiliation(s)
- Alexey N Boyko
- Pirogov's Russian National Research University, Department of Neurology, Neurosurgery and Medical Genetics, .,Neurological Department, Usupov's Hospital, Moscow, Russia,
| | - Olga V Boyko
- Pirogov's Russian National Research University, Department of Neurology, Neurosurgery and Medical Genetics, .,Neurological Department, Usupov's Hospital, Moscow, Russia,
| |
Collapse
|
246
|
Abstract
Since its technical development in the early 1980s, magnetic resonance imaging (MRI) has quickly been adopted as an essential tool in supporting the diagnosis, longitudinal monitoring, evaluation of therapeutic response, and scientific investigations in multiple sclerosis (MS). The clinical usage of MRI has increased in parallel with technical innovations in the technique itself; the widespread adoption of clinically routine MRI at 1.5T has allowed sensitive qualitative and quantitative assessments of macroscopic central nervous system (CNS) inflammatory demyelinating lesions and tissue atrophy. However, conventional MRI lesion measures lack specificity for the underlying MS pathology and only weakly correlate with clinical status. Higher field strength units and newer, advanced MRI techniques offer increased sensitivity and specificity in the detection of disease activity and disease severity. This review summarizes the current status and future prospects regarding the role of MRI in the characterization of MS-related brain and spinal cord involvement.
Collapse
Affiliation(s)
- Christopher C Hemond
- Laboratory for Neuroimaging Research, Partners Multiple Sclerosis Center, Ann Romney Center for Neurologic Diseases, Departments of Neurology and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Rohit Bakshi
- Laboratory for Neuroimaging Research, Partners Multiple Sclerosis Center, Ann Romney Center for Neurologic Diseases, Departments of Neurology and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
247
|
Amiri H, de Sitter A, Bendfeldt K, Battaglini M, Gandini Wheeler-Kingshott CAM, Calabrese M, Geurts JJG, Rocca MA, Sastre-Garriga J, Enzinger C, de Stefano N, Filippi M, Rovira Á, Barkhof F, Vrenken H. Urgent challenges in quantification and interpretation of brain grey matter atrophy in individual MS patients using MRI. Neuroimage Clin 2018; 19:466-475. [PMID: 29984155 PMCID: PMC6030805 DOI: 10.1016/j.nicl.2018.04.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 03/28/2018] [Accepted: 04/22/2018] [Indexed: 01/18/2023]
Abstract
Atrophy of the brain grey matter (GM) is an accepted and important feature of multiple sclerosis (MS). However, its accurate measurement is hampered by various technical, pathological and physiological factors. As a consequence, it is challenging to investigate the role of GM atrophy in the disease process as well as the effect of treatments that aim to reduce neurodegeneration. In this paper we discuss the most important challenges currently hampering the measurement and interpretation of GM atrophy in MS. The focus is on measurements that are obtained in individual patients rather than on group analysis methods, because of their importance in clinical trials and ultimately in clinical care. We discuss the sources and possible solutions of the current challenges, and provide recommendations to achieve reliable measurement and interpretation of brain GM atrophy in MS.
Collapse
Key Words
- BET, brain extraction tool
- Brain atrophy
- CNS, central nervous system
- CTh, cortical thickness
- DGM, deep grey matter
- DTI, diffusion tensor imaging
- FA, fractional anisotropy
- GM, grey matter
- Grey matter
- MRI, magnetic resonance imaging
- MS, multiple sclerosis
- Magnetic resonance imaging
- Multiple sclerosis
- TE, echo time
- TI, inversion time
- TR, repetition time
- VBM, voxel-based morphometry
- WM, white matter
Collapse
Affiliation(s)
- Houshang Amiri
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Alexandra de Sitter
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.
| | | | - Marco Battaglini
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | | | - Massimiliano Calabrese
- Multiple Sclerosis Centre, Neurology Section, Department of Neurosciences, Biomedicine and Movements, University of Verona, Italy
| | - Jeroen J G Geurts
- Anatomy & Neurosciences, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Maria A Rocca
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Jaume Sastre-Garriga
- Servei de Neurologia/Neuroimmunologia, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Christian Enzinger
- Department of Neurology & Division of Neuroradiology, Vascular and Interventional Radiology, Department of Radiology, Medical University of Graz, Austria
| | - Nicola de Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Álex Rovira
- Unitat de Ressonància Magnètica (Servei de Radiologia), Hospital universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; Institutes of Neurology and Healthcare Engineering, UCL, London, UK
| | - Hugo Vrenken
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
248
|
Narayan RN, Forsthuber T, Stüve O. Emerging drugs for primary progressive multiple sclerosis. Expert Opin Emerg Drugs 2018; 23:97-110. [DOI: 10.1080/14728214.2018.1463370] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Ram Narendra Narayan
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Neurology Section, VA North Texas Health Care System, Dallas VA Medical Center, Dallas, TX, USA
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
249
|
Zéphir H. Progress in understanding the pathophysiology of multiple sclerosis. Rev Neurol (Paris) 2018; 174:358-363. [PMID: 29680179 DOI: 10.1016/j.neurol.2018.03.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/08/2018] [Accepted: 03/08/2018] [Indexed: 01/17/2023]
Abstract
Multiple sclerosis (MS) arises in people who have a genetic susceptibility to environmental factors and events, which ultimately trigger the disease. It is thought that peripheral immune cells are mobilized and enter the CNS through the impaired blood-brain barrier in the subarachnoid space, as acute lesions show large numbers of macrophages and CD8+ T cells and, to a lesser extent, CD4+ T cells, B cells and plasma cells. Demyelination is mostly localized to focal lesions in early relapsing-remitting (RR) MS, whereas other areas of white matter appear normal. Over time, T-cell and B-cell infiltration becomes more diffuse and axonal injury more widespread, leading to self-perpetuating atrophy in both white and gray matter. With disease progression, inflammatory processes are predominantly driven by the action of CNS resident microglia cells. In addition, there is evidence that meningeal lymphoid-like structures can form and contribute to late-stage inflammation. In general, however, despite dynamic changes over time in MS pathology, lesions do not appear to differ significantly in the different classic forms of MS already identified. While all treatments approved for MS management target inflammatory components of RRMS, the B-cell-depleting antibody ocrelizumab is the first such treatment approved recently for primary progressive (PP) MS. However, recent pathological and imaging findings have prompted reconsideration of the clinical phenotypes of MS patients proposed by Lublin's 2013 classification, including clinical and MRI signs of activity, and new imaging biomarkers of remyelination are now being investigated for new strategies of MS management.
Collapse
Affiliation(s)
- H Zéphir
- Pôle des Neurosciences et de l'Appareil Locomoteur, CHRU de Lille, LIRIC, U995, équipe 3, Université de Lille, 59037 Lille Cedex, France.
| |
Collapse
|
250
|
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS), which gives rise to focal lesions in the gray and white matter and to diffuse neurodegeneration in the entire brain. In this review, the spectrum of MS lesions and their relation to the inflammatory process is described. Pathology suggests that inflammation drives tissue injury at all stages of the disease. Focal inflammatory infiltrates in the meninges and the perivascular spaces appear to produce soluble factors, which induce demyelination or neurodegeneration either directly or indirectly through microglia activation. The nature of these soluble factors, which are responsible for demyelinating activity in sera and cerebrospinal fluid of the patients, is currently undefined. Demyelination and neurodegeneration is finally accomplished by oxidative injury and mitochondrial damage leading to a state of "virtual hypoxia."
Collapse
Affiliation(s)
- Hans Lassmann
- Center for Brain Research, Medical University of Vienna, A-1090 Wien, Austria
| |
Collapse
|