201
|
Straudi S, Manfredini F, Lamberti N, Zamboni P, Bernardi F, Marchetti G, Pinton P, Bonora M, Secchiero P, Tisato V, Volpato S, Basaglia N. The effectiveness of Robot-Assisted Gait Training versus conventional therapy on mobility in severely disabled progressIve MultiplE sclerosis patients (RAGTIME): study protocol for a randomized controlled trial. Trials 2017; 18:88. [PMID: 28241776 PMCID: PMC5330064 DOI: 10.1186/s13063-017-1838-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 02/12/2017] [Indexed: 11/22/2022] Open
Abstract
Background Gait and mobility impairments affect the quality of life (QoL) of patients with progressive multiple sclerosis (MS). Robot-assisted gait training (RAGT) is an effective rehabilitative treatment but evidence of its superiority compared to other options is lacking. Furthermore, the response to rehabilitation is multidimensional, person-specific and possibly involves functional reorganization processes. The aims of this study are: (1) to test the effectiveness on gait speed, mobility, balance, fatigue and QoL of RAGT compared to conventional therapy (CT) in progressive MS and (2) to explore changes of clinical and circulating biomarkers of neural plasticity. Methods This will be a parallel-group, randomized controlled trial design with the assessor blinded to the group allocation of participants. Ninety-eight (49 per arm) progressive MS patients (EDSS scale 6–7) will be randomly assigned to receive twelve 2-h training sessions over a 4-week period (three sessions/week) of either: (1) RAGT intervention on a robotic-driven gait orthosis (Lokomat, Hocoma, Switzerland). The training parameters (torque of the knee and hip drives, treadmill speed, body weight support) are set during the first session and progressively adjusted during training progression or (2) individual conventional physiotherapy focusing on over-ground walking training performed with the habitual walking device. The same assessors will perform outcome measurements at four time points: baseline (before the first intervention session); intermediate (after six training sessions); end of treatment (after the completion of 12 sessions); and follow-up (after 3 months from the end of the training program). The primary outcome is gait speed, assessed by the Timed 25-Foot Walk Test. We will also assess walking endurance, balance, depression, fatigue and QoL as well as instrumental laboratory markers (muscle metabolism, cerebral venous hemodynamics, cortical activation) and circulating laboratory markers (rare circulating cell populations pro and anti-inflammatory cytokines/chemokines, growth factors, neurotrophic factors, coagulation factors, other plasma proteins suggested by transcriptomic analysis and metabolic parameters). Discussion The RAGT training is expected to improve mobility compared to the active control intervention in progressive MS. Unique to this study is the analysis of various potential markers of plasticity in relation with clinical outcomes. Trial registration ClinicalTrials.gov, identifier: NCT02421731. Registered on 19 January 2015 (retrospectively registered). Electronic supplementary material The online version of this article (doi:10.1186/s13063-017-1838-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sofia Straudi
- Neuroscience and Rehabilitation Department, Ferrara University Hospital, Via Aldo Moro 8, 44124, Ferrara, Italy
| | - Fabio Manfredini
- Neuroscience and Rehabilitation Department, Ferrara University Hospital, Via Aldo Moro 8, 44124, Ferrara, Italy. .,Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy.
| | - Nicola Lamberti
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Paolo Zamboni
- Unit of Translational Surgery and Vascular Diseases Center, Ferrara University Hospital, Ferrara, Italy
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giovanna Marchetti
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Massimo Bonora
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine, Section of Anatomy and Histology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Veronica Tisato
- Department of Morphology, Surgery and Experimental Medicine, Section of Anatomy and Histology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Stefano Volpato
- Center for Clinical Epidemiology, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Nino Basaglia
- Neuroscience and Rehabilitation Department, Ferrara University Hospital, Via Aldo Moro 8, 44124, Ferrara, Italy
| |
Collapse
|
202
|
Lassmann H, Bradl M. Multiple sclerosis: experimental models and reality. Acta Neuropathol 2017; 133:223-244. [PMID: 27766432 PMCID: PMC5250666 DOI: 10.1007/s00401-016-1631-4] [Citation(s) in RCA: 378] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 01/01/2023]
Abstract
One of the most frequent statements, provided in different variations in the introduction of experimental studies on multiple sclerosis (MS), is that "Multiple sclerosis is a demyelinating autoimmune disease and experimental autoimmune encephalomyelitis (EAE) is a suitable model to study its pathogenesis". However, so far, no single experimental model covers the entire spectrum of the clinical, pathological, or immunological features of the disease. Many different models are available, which proved to be highly useful for studying different aspects of inflammation, demyelination, remyelination, and neurodegeneration in the central nervous system. However, the relevance of results from such models for MS pathogenesis has to be critically validated. Current EAE models are mainly based on inflammation, induced by auto-reactive CD4+ T-cells, and these models reflect important aspects of MS. However, pathological data and results from clinical trials in MS indicate that CD8+ T-cells and B-lymphocytes may play an important role in propagating inflammation and tissue damage in established MS. Viral models may reflect key features of MS-like inflammatory demyelination, but are difficult to use due to their very complex pathogenesis, involving direct virus-induced and immune-mediated mechanisms. Furthermore, evidence for a role of viruses in MS pathogenesis is indirect and limited, and an MS-specific virus infection has not been identified so far. Toxic models are highly useful to unravel mechanisms of de- and remyelination, but do not reflect other important aspects of MS pathology and pathogenesis. For all these reasons, it is important to select the right experimental model to answer specific questions in MS research.
Collapse
Affiliation(s)
- Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| | - Monika Bradl
- Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| |
Collapse
|
203
|
Smorodchenko A, Schneider S, Rupprecht A, Hilse K, Sasgary S, Zeitz U, Erben RG, Pohl EE. UCP2 up-regulation within the course of autoimmune encephalomyelitis correlates with T-lymphocyte activation. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1002-1012. [PMID: 28130201 DOI: 10.1016/j.bbadis.2017.01.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/07/2017] [Accepted: 01/23/2017] [Indexed: 01/20/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating autoimmune disorder of the central nervous system (CNS) associated with severe neurological disability. Reactive oxygen species (ROS) and mitochondrial dysfunction play a pivotal role in the pathogenesis of this disease. Several members of the mitochondrial uncoupling protein subfamily (UCP2-UCP5) were suggested to regulate ROS by diminishing the mitochondrial membrane potential and constitute therefore a promising pharmacological target for MS. To evaluate the role of different uncoupling proteins in neuroinflammation, we have investigated their expression patterns in murine brain and spinal cord (SC) during different stages of experimental autoimmune encephalomyelitis (EAE), an animal model for MS. At mRNA and protein levels we found that only UCP2 is up-regulated in the SC, but not in brain. The increase in UCP2 expression was antigen-independent, reached its maximum between 14 and 21days in both OVA and MOG immunized animals and correlated with an augmented number of CD3+ T-lymphocytes in SC parenchyma. The decrease in abundance of UCP4 was due to neuronal injury and was only detected in CNS of MOG-induced EAE animals. The results provide evidence that the involvement of mitochondrial UCP2 in CNS inflammation during EAE may be mainly explained by the invasion of activated T-lymphocytes. This conclusion coincides with our previous observation that UCP2 is up-regulated in activated and rapidly proliferating T-cells and participates in fast metabolic re-programming of cells during proliferation.
Collapse
Affiliation(s)
- Alina Smorodchenko
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria; Institute of Vegetative Anatomy, Charité - Universitätsmedizin Berlin, Germany.
| | - Stephanie Schneider
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Anne Rupprecht
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Karoline Hilse
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Soleman Sasgary
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Ute Zeitz
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Reinhold G Erben
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Elena E Pohl
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria.
| |
Collapse
|
204
|
Giovannoni G, Cutter G, Sormani MP, Belachew S, Hyde R, Koendgen H, Knappertz V, Tomic D, Leppert D, Herndon R, Wheeler-Kingshott CAM, Ciccarelli O, Selwood D, di Cantogno EV, Ben-Amor AF, Matthews P, Carassiti D, Baker D, Schmierer K. Is multiple sclerosis a length-dependent central axonopathy? The case for therapeutic lag and the asynchronous progressive MS hypotheses. Mult Scler Relat Disord 2017; 12:70-78. [PMID: 28283111 DOI: 10.1016/j.msard.2017.01.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 01/06/2017] [Accepted: 01/12/2017] [Indexed: 10/20/2022]
Abstract
Trials of anti-inflammatory therapies in non-relapsing progressive multiple sclerosis (MS) have been stubbornly negative except recently for an anti-CD20 therapy in primary progressive MS and a S1P modulator siponimod in secondary progressive MS. We argue that this might be because trials have been too short and have focused on assessing neuronal pathways, with insufficient reserve capacity, as the core component of the primary outcome. Delayed neuroaxonal degeneration primed by prior inflammation is not expected to respond to disease-modifying therapies targeting MS-specific mechanisms. However, anti-inflammatory therapies may modify these damaged pathways, but with a therapeutic lag that may take years to manifest. Based on these observations we propose that clinically apparent neurodegenerative components of progressive MS may occur in a length-dependent manner and asynchronously. If this hypothesis is confirmed it may have major implications for the future design of progressive MS trials.
Collapse
Affiliation(s)
- Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK.
| | - Gary Cutter
- Department of Biostatistics, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA.
| | - Maria Pia Sormani
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy.
| | | | | | | | | | | | | | - Robert Herndon
- University of Mississippi Medical Center, Jackson, MS, USA.
| | - Claudia A M Wheeler-Kingshott
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Queen Square, London, UK.
| | - Olga Ciccarelli
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, Queen Square, London, UK; NIHR University College Hospitals Biomedical Research Centre (BRC), UK.
| | - David Selwood
- Wolfson Institute for Biomedical Research, UCL, London, UK.
| | | | | | - Paul Matthews
- Department of Medicine, Imperial College, London, UK.
| | - Daniele Carassiti
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK.
| | - David Baker
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK.
| | - Klaus Schmierer
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK.
| |
Collapse
|
205
|
Kipp M, Nyamoya S, Hochstrasser T, Amor S. Multiple sclerosis animal models: a clinical and histopathological perspective. Brain Pathol 2017; 27:123-137. [PMID: 27792289 DOI: 10.1111/bpa.12454] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 10/26/2016] [Indexed: 12/11/2022] Open
Abstract
There is a broad consensus that multiple sclerosis (MS) represents more than an inflammatory disease: it harbors several characteristic aspects of a classical neurodegenerative disorder, that is, damage to axons, synapses and nerve cell bodies. While we are equipped with appropriate therapeutic options to prevent immune-cell driven relapses, effective therapeutic options to prevent the progressing neurodegeneration are still missing. In this review article, we will discuss to what extent pathology of the progressive disease stage can be modeled in MS animal models. While acute and relapsing-remitting forms of experimental autoimmune encephalomyelitis (EAE), which are T cell dependent, are aptly suited to model relapsing-remitting phases of MS, other EAE models, especially the secondary progressive EAE stage in Biozzi ABH mice is better representing the secondary progressive phase of MS, which is refractory to many immune therapies. Besides EAE, the cuprizone model is rapidly gaining popularity to study the formation and progression of demyelinating CNS lesions without T cell involvement. Here, we discuss these two non-popular MS models. It is our aim to point out the pathological hallmarks of MS, and discuss which pathological aspects of the disease can be best studied in the various animal models available.
Collapse
Affiliation(s)
- Markus Kipp
- Department of Neuroanatomy, Faculty of Medicine, LMU München University, München, 80336, Germany
| | - Stella Nyamoya
- Department of Neuroanatomy, Faculty of Medicine, LMU München University, München, 80336, Germany.,Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, D-52074, Germany
| | - Tanja Hochstrasser
- Department of Neuroanatomy, Faculty of Medicine, LMU München University, München, 80336, Germany
| | - Sandra Amor
- Department of Pathology, VU University Medical Centre, Amsterdam, The Netherlands.,Barts and The London School of Medicine and Dentistry, Neuroimmunology Unit, , Queen Mary University of London, Neuroscience Centre, Blizard Institute of Cell and Molecular Science, London, UK
| |
Collapse
|
206
|
Buzzard K, Chan WH, Kilpatrick T, Murray S. Multiple Sclerosis: Basic and Clinical. ADVANCES IN NEUROBIOLOGY 2017; 15:211-252. [DOI: 10.1007/978-3-319-57193-5_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
207
|
Kunert JM, Maia PD, Kutz JN. Functionality and Robustness of Injured Connectomic Dynamics in C. elegans: Linking Behavioral Deficits to Neural Circuit Damage. PLoS Comput Biol 2017; 13:e1005261. [PMID: 28056097 PMCID: PMC5215891 DOI: 10.1371/journal.pcbi.1005261] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 11/22/2016] [Indexed: 01/12/2023] Open
Abstract
Using a model for the dynamics of the full somatic nervous system of the nematode C. elegans, we address how biological network architectures and their functionality are degraded in the presence of focal axonal swellings (FAS) arising from neurodegenerative disease and/or traumatic brain injury. Using biophysically measured FAS distributions and swelling sizes, we are able to simulate the effects of injuries on the neural dynamics of C. elegans, showing how damaging the network degrades its low-dimensional dynamical responses. We visualize these injured neural dynamics by mapping them onto the worm's low-dimensional postures, i.e. eigenworm modes. We show that a diversity of functional deficits arise from the same level of injury on a connectomic network. Functional deficits are quantified using a statistical shape analysis, a procrustes analysis, for deformations of the limit cycles that characterize key behaviors such as forward crawling. This procrustes metric carries information on the functional outcome of injuries in the model. Furthermore, we apply classification trees to relate injury structure to the behavioral outcome. This makes testable predictions for the structure of an injury given a defined functional deficit. More critically, this study demonstrates the potential role of computational simulation studies in understanding how neuronal networks process biological signals, and how this processing is impacted by network injury.
Collapse
Affiliation(s)
- James M. Kunert
- Department of Physics, University of Washington, Seattle, Washington, United States of America
| | - Pedro D. Maia
- Department of Applied Mathematics, University of Washington, Seattle, Washington, United States of America
| | - J. Nathan Kutz
- Department of Applied Mathematics, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
208
|
Nyamoya S, Schweiger F, Kipp M, Hochstrasser T. Cuprizone as a model of myelin and axonal damage. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.ddmod.2018.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
209
|
Chamberlain KA, Nanescu SE, Psachoulia K, Huang JK. Oligodendrocyte regeneration: Its significance in myelin replacement and neuroprotection in multiple sclerosis. Neuropharmacology 2016; 110:633-643. [PMID: 26474658 PMCID: PMC4841742 DOI: 10.1016/j.neuropharm.2015.10.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 09/22/2015] [Accepted: 10/05/2015] [Indexed: 12/12/2022]
Abstract
Oligodendrocytes readily regenerate and replace myelin membranes around axons in the adult mammalian central nervous system (CNS) following injury. The ability to regenerate oligodendrocytes depends on the availability of neural progenitors called oligodendrocyte precursor cells (OPCs) in the adult CNS that respond to injury-associated signals to induce OPC expansion followed by oligodendrocyte differentiation, axonal contact and myelin regeneration (remyelination). Remyelination ensures the maintenance of axonal conduction, and the oligodendrocytes themselves provide metabolic factors that are necessary to maintain neuronal integrity. Recent advances in oligodendrocyte regeneration research are beginning to shed light on critical intrinsic signals, as well as extrinsic, environmental factors that regulate the distinct steps of oligodendrocyte lineage progression and myelin replacement under CNS injury. These studies may offer novel pharmacological targets for regenerative medicine in inflammatory demyelinating disorders in the CNS such as multiple sclerosis. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
Affiliation(s)
- Kelly A Chamberlain
- Department of Biology, Georgetown University, Washington, D.C., USA; Interdisciplinary Program in Neuroscience, Georgetown University, Washington, D.C., USA
| | - Sonia E Nanescu
- Department of Biology, Georgetown University, Washington, D.C., USA
| | | | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, D.C., USA; Interdisciplinary Program in Neuroscience, Georgetown University, Washington, D.C., USA.
| |
Collapse
|
210
|
Michailidou I, Naessens DMP, Hametner S, Guldenaar W, Kooi EJ, Geurts JJG, Baas F, Lassmann H, Ramaglia V. Complement C3 on microglial clusters in multiple sclerosis occur in chronic but not acute disease: Implication for disease pathogenesis. Glia 2016; 65:264-277. [PMID: 27778395 PMCID: PMC5215693 DOI: 10.1002/glia.23090] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/04/2016] [Indexed: 12/11/2022]
Abstract
Microglial clusters with C3d deposits are observed in the periplaque of multiple sclerosis (MS) brains and were proposed as early stage of lesion formation. As such they should appear in the brain of MS donors with acute disease but thus far this has not been shown. Using postmortem brain tissue from acute (n = 10) and chronic (n = 15) MS cases we investigated whether C3d+ microglial clusters are part of an acute attack against myelinated axons, which could have implications for disease pathogenesis. The specificity of our findings to MS was tested in ischemic stroke cases (n = 8) with initial or advanced lesions and further analyzed in experimental traumatic brain injury (TBI, n = 26), as both conditions are primarily nondemyelinating but share essential features of neurodegeneration with MS lesions. C3d+ microglial clusters were found in chronic but not acute MS. They were not associated with antibody deposits or terminal complement activation. They were linked to slowly expanding lesions, localized on axons with impaired transport and associated with neuronal C3 production. C3d+ microglial clusters were not specific to MS as they were also found in stroke and experimental TBI. We conclude that C3d+ microglial clusters in MS are not part of an acute attack against myelinated axons. As such it is unlikely that they drive formation of new lesions but could represent a physiological mechanism to remove irreversibly damaged axons in chronic disease. GLIA 2017;65:264–277
Collapse
Affiliation(s)
- Iliana Michailidou
- Department of Genome Analysis, Academic Medical Center, Meibergdreef 9, Amsterdam, 1105, The Netherlands
| | - Daphne M P Naessens
- Department of Genome Analysis, Academic Medical Center, Meibergdreef 9, Amsterdam, 1105, The Netherlands
| | - Simon Hametner
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna, 1090, Austria
| | - Willemijn Guldenaar
- Department of Anatomy and Neurosciences, VU University Medical Center, De Boelelaan 1118, Amsterdam, 1081, The Netherlands
| | - Evert-Jan Kooi
- Department of Anatomy and Neurosciences, VU University Medical Center, De Boelelaan 1118, Amsterdam, 1081, The Netherlands
| | - Jeroen J G Geurts
- Department of Anatomy and Neurosciences, VU University Medical Center, De Boelelaan 1118, Amsterdam, 1081, The Netherlands
| | - Frank Baas
- Department of Genome Analysis, Academic Medical Center, Meibergdreef 9, Amsterdam, 1105, The Netherlands
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna, 1090, Austria
| | - Valeria Ramaglia
- Department of Genome Analysis, Academic Medical Center, Meibergdreef 9, Amsterdam, 1105, The Netherlands
| |
Collapse
|
211
|
Pietroboni AM, Schiano di Cola F, Scarioni M, Fenoglio C, Spanò B, Arighi A, Cioffi SM, Oldoni E, De Riz MA, Basilico P, Calvi A, Fumagalli GG, Triulzi F, Galimberti D, Bozzali M, Scarpini E. CSF β-amyloid as a putative biomarker of disease progression in multiple sclerosis. Mult Scler 2016; 23:1085-1091. [PMID: 27754941 DOI: 10.1177/1352458516674566] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Neurodegeneration plays a major role in determining disability in multiple sclerosis (MS) patients. Hence, there is increasing need to identify reliable biomarkers, which could serve as prognostic measure of disease progression. OBJECTIVES To assess whether cerebrospinal fluid (CSF) tau and β-amyloid (Aβ) levels were altered in newly diagnosed MS patients and correlated with disability. Moreover, we investigated whether these CSF biomarkers associate with macroscopic brain tissue damage measures. METHODS CSF Aβ and tau levels were determined by enzyme-linked immunosorbent assay in CSF samples from 48 newly diagnosed MS patients, followed-up clinically for 3 years by recording their Expanded Disability Status Scale score at 6-month intervals, and 45 controls. All patients underwent magnetic resonance imaging at baseline and at the end of follow-up to quantify their lesion load (LL). RESULTS CSF Aβ levels were significantly reduced in patients compared to controls ( p < 0.001). Lower CSF Aβ levels at baseline were a disability predictor at 3-year follow-up ( p = 0.009). CSF tau levels correlated with T2- and T1-LL ( p < 0.001). CONCLUSION CSF Aβ reduction is a promising biomarker of neurodegeneration and may predict patients' clinical outcome. Therefore, CSF Aβ should be considered as a potential biomarker of prognostic value.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Schiano di Cola
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Scarioni
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Barbara Spanò
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Andrea Arighi
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Mg Cioffi
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Emanuela Oldoni
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Milena A De Riz
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Basilico
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Calvi
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio G Fumagalli
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Triulzi
- Neuroradiology Unit, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Galimberti
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Bozzali
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Elio Scarpini
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
212
|
Hannoun S, Durand-Dubief F, Roch JA, Sappey-Marinier D, Cotton F. Tracking successive Wallerian degenerations in a relapsing-remitting multiple sclerosis patient. J Neuroradiol 2016; 43:359-61. [DOI: 10.1016/j.neurad.2016.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 05/25/2016] [Accepted: 05/30/2016] [Indexed: 11/27/2022]
|
213
|
Mañé-Martínez M, Olsson B, Bau L, Matas E, Cobo-Calvo Á, Andreasson U, Blennow K, Romero-Pinel L, Martínez-Yélamos S, Zetterberg H. Glial and neuronal markers in cerebrospinal fluid in different types of multiple sclerosis. J Neuroimmunol 2016; 299:112-117. [DOI: 10.1016/j.jneuroim.2016.08.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/17/2016] [Accepted: 08/04/2016] [Indexed: 11/29/2022]
|
214
|
Wootla B, Denic A, Watzlawik JO, Warrington AE, Rodriguez M. Antibody-Mediated Oligodendrocyte Remyelination Promotes Axon Health in Progressive Demyelinating Disease. Mol Neurobiol 2016; 53:5217-28. [PMID: 26409478 PMCID: PMC5012151 DOI: 10.1007/s12035-015-9436-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/10/2015] [Indexed: 02/03/2023]
Abstract
Demyelination underlies early neurological symptoms in multiple sclerosis (MS); however, axonal damage is considered critical for permanent chronic deficits. The precise mechanisms by which axonal injury occurs in MS are unclear; one hypothesis is the absence or failure of remyelination, suggesting that promoting remyelination may protect axons from death. This report provides direct evidence that promoting oligodendrocyte remyelination protects axons and maintains transport function. Persistent Theiler's virus infection of Swiss Jim Lambert (SJL)/J mice was used as a model of MS to assess the effects of remyelination on axonal injury following demyelination in the spinal cord. Remyelination was induced using an oligodendrocyte/myelin-specific recombinant human monoclonal IgM, rHIgM22. The antibody is endowed with strong anti-apoptotic and pro-proliferative effects on oligodendrocyte progenitor cells. We used (1)H-magnetic resonance spectroscopy (MRS) at the brainstem to measure N-acetyl-aspartate (NAA) as a surrogate of neuronal health and spinal cord integrity. We found increased brainstem NAA concentrations at 5 weeks post-treatment with rHIgM22, which remained stable out to 10 weeks. Detailed spinal cord morphology studies revealed enhanced remyelination in the rHIgM22-treated group but not in the isotype control antibody- or saline-treated groups. Importantly, we found rHIgM22-mediated remyelination protected small- and medium-caliber mid-thoracic spinal cord axons from damage despite similar demyelination and inflammation across all experimental groups. The most direct confirmation of remyelination-mediated protection of descending neurons was an improvement in retrograde transport. Treatment with rHIgM22 significantly increased the number of retrograde-labeled neurons in the brainstem, indicating that preserved axons are functionally competent. This is direct validation that remyelination preserves spinal cord axons and protects functional axon integrity.
Collapse
Affiliation(s)
- Bharath Wootla
- Departments of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Center for Regenerative Medicine, Neuroregeneration, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Aleksandar Denic
- Departments of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jens O Watzlawik
- Departments of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Center for Regenerative Medicine, Neuroregeneration, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Arthur E Warrington
- Departments of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Moses Rodriguez
- Departments of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
- Departments of Immunology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
215
|
Obert D, Helms G, Sättler MB, Jung K, Kretzschmar B, Bähr M, Dechent P, Diem R, Hein K. Brain Metabolite Changes in Patients with Relapsing-Remitting and Secondary Progressive Multiple Sclerosis: A Two-Year Follow-Up Study. PLoS One 2016; 11:e0162583. [PMID: 27636543 PMCID: PMC5026363 DOI: 10.1371/journal.pone.0162583] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 08/25/2016] [Indexed: 01/26/2023] Open
Abstract
Magnetic resonance spectroscopy (MRS) provides the unique ability to monitor several disease-related pathological processes via their characteristic metabolic markers in vivo. In the present study metabolic compositions were assessed every six months over the period of two years in 36 patients with Multiple Sclerosis (MS) including 21 relapsing-remitting (RR), 15 secondary progressive (SP) patients and 12 normal subjects. The concentrations of the main MRS-detectable metabolites N-acetylaspartate and N-acetylaspartylglutamate (tNAA), creatine and phosphocreatine (tCr), choline containing compounds (Cho), myo-Inositol (Ins), glutamine and glutamate (Glx) and their ratios were calculated in the normal appearing white matter (NAWM) and in selected non-enhancing white matter (WM) lesions. Association between metabolic concentrations in the NAWM and disability were investigated. Concentration of tNAA, a marker for neuroaxonal integrity, did not show any difference between the investigated groups. However, the patients with SPMS showed significant reduction of tNAA in the NAWM over the investigation period of two years indicating diffuse neuroaxonal loss during the disease course. Furthermore, we found a significant increase of Ins, Ins/tCr and Ins/tNAA in WM lesions independently from the course of the disease suggesting ongoing astrogliosis in silent-appearing WM lesions. Analyzing correlations between MRS metabolites in the NAWM and patients clinical status we found the positive correlation of Ins/tNAA with disability in patients with RRMS. In SPMS positive correlation of Cho with disability was found.
Collapse
Affiliation(s)
- Dorothea Obert
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Gunther Helms
- Department of Cognitive Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Muriel B. Sättler
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Klaus Jung
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Benedikt Kretzschmar
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Peter Dechent
- Department of Cognitive Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Ricarda Diem
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany
| | - Katharina Hein
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- * E-mail:
| |
Collapse
|
216
|
Seehusen F, Kiel K, Jottini S, Wohlsein P, Habierski A, Seibel K, Vogel T, Urlaub H, Kollmar M, Baumgärtner W, Teichmann U. Axonopathy in the Central Nervous System Is the Hallmark of Mice with a Novel Intragenic Null Mutation of Dystonin. Genetics 2016; 204:191-203. [PMID: 27401753 PMCID: PMC5012385 DOI: 10.1534/genetics.116.186932] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 07/03/2016] [Indexed: 01/13/2023] Open
Abstract
Dystonia musculorum is a neurodegenerative disorder caused by a mutation in the dystonin gene. It has been described in mice and humans where it is called hereditary sensory autonomic neuropathy. Mutated mice show severe movement disorders and die at the age of 3-4 weeks. This study describes the discovery and molecular, clinical, as well as pathological characterization of a new spontaneously occurring mutation in the dystonin gene in C57BL/6N mice. The mutation represents a 40-kb intragenic deletion allele of the dystonin gene on chromosome 1 with exactly defined deletion borders. It was demonstrated by Western blot, mass spectrometry, and immunohistology that mice with a homozygous mutation were entirely devoid of the dystonin protein. Pathomorphological lesions were restricted to the brain stem and spinal cord and consisted of swollen, argyrophilic axons and dilated myelin sheaths in the white matter and, less frequently, total chromatolysis of neurons in the gray matter. Axonal damage was detected by amyloid precursor protein and nonphosphorylated neurofilament immunohistology. Axonopathy in the central nervous system (CNS) represents the hallmark of this disease. Mice with the dystonin mutation also showed suppurative inflammation in the respiratory tract, presumably due to brain stem lesion-associated food aspiration, whereas skeletal muscles showed no pathomorphological changes. This study describes a novel mutation in the dystonin gene in mice leading to axonopathy in the CNS. In further studies, this model may provide new insights into the pathogenesis of neurodegenerative diseases and may elucidate the complex interactions of dystonin with various other cellular proteins especially in the CNS.
Collapse
Affiliation(s)
- Frauke Seehusen
- Department of Pathology, University of Veterinary Medicine, D-30559 Hannover, Germany
| | - Kirsten Kiel
- Animal Facility, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
| | - Stefano Jottini
- Department of Pathology, University of Veterinary Medicine, D-30559 Hannover, Germany Department of Animal Health, Pathology Unit, Faculty of Veterinary Medicine, University of Parma, I-43100, Italy
| | - Peter Wohlsein
- Department of Pathology, University of Veterinary Medicine, D-30559 Hannover, Germany
| | - Andre Habierski
- Department of Pathology, University of Veterinary Medicine, D-30559 Hannover, Germany
| | - Katharina Seibel
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
| | - Tanja Vogel
- Institute of Anatomy and Cell Biology, University of Freiburg, D-79104, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany Bioanalytics, Department of Clinical Chemistry, University Medical Center Göttingen, D-37075, Germany
| | - Martin Kollmar
- Department of NMR-Based Structural Biology, Research Group Systems Biology of Motor Proteins, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine, D-30559 Hannover, Germany Center for Systems Neuroscience, University of Veterinary Medicine, D-30559 Hannover, Germany
| | - Ulrike Teichmann
- Animal Facility, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
| |
Collapse
|
217
|
Goodman AD, Gyang T, Smith AD. Ibudilast for the treatment of multiple sclerosis. Expert Opin Investig Drugs 2016; 25:1231-7. [DOI: 10.1080/13543784.2016.1221924] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
218
|
Abstract
Discussions of multiple sclerosis (MS) pathophysiology tend to focus on T cells and B cells of the adaptive immune response. The innate immune system is less commonly considered in this context, although dendritic cells, monocytes, macrophages and microglia - collectively referred to as myeloid cells - have prominent roles in MS pathogenesis. These populations of myeloid cells function as antigen-presenting cells and effector cells in neuroinflammation. Furthermore, a vicious cycle of interactions between T cells and myeloid cells exacerbates pathology. Several disease-modifying therapies are now available to treat MS, and insights into their mechanisms of action have largely focused on the adaptive immune system, but these therapies also have important effects on myeloid cells. In this Review, we discuss the evidence for the roles of myeloid cells in MS and the experimental autoimmune encephalomyelitis model of MS, and consider how interactions between myeloid cells and T cells and/or B cells promote MS pathology. Finally, we discuss the direct and indirect effects of existing MS medications on myeloid cells.
Collapse
Affiliation(s)
- Manoj K Mishra
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| |
Collapse
|
219
|
Manocha GD, Floden AM, Rausch K, Kulas JA, McGregor BA, Rojanathammanee L, Puig KR, Puig KL, Karki S, Nichols MR, Darland DC, Porter JE, Combs CK. APP Regulates Microglial Phenotype in a Mouse Model of Alzheimer's Disease. J Neurosci 2016; 36:8471-86. [PMID: 27511018 PMCID: PMC4978805 DOI: 10.1523/jneurosci.4654-15.2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 05/20/2016] [Accepted: 06/24/2016] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED Prior work suggests that amyloid precursor protein (APP) can function as a proinflammatory receptor on immune cells, such as monocytes and microglia. Therefore, we hypothesized that APP serves this function in microglia during Alzheimer's disease. Although fibrillar amyloid β (Aβ)-stimulated cytokine secretion from both wild-type and APP knock-out (mAPP(-/-)) microglial cultures, oligomeric Aβ was unable to stimulate increased secretion from mAPP(-/-) cells. This was consistent with an ability of oligomeric Aβ to bind APP. Similarly, intracerebroventricular infusions of oligomeric Aβ produced less microgliosis in mAPP(-/-) mice compared with wild-type mice. The mAPP(-/-) mice crossed to an APP/PS1 transgenic mouse line demonstrated reduced microgliosis and cytokine levels and improved memory compared with wild-type mice despite robust fibrillar Aβ plaque deposition. These data define a novel function for microglial APP in regulating their ability to acquire a proinflammatory phenotype during disease. SIGNIFICANCE STATEMENT A hallmark of Alzheimer's disease (AD) brains is the accumulation of amyloid β (Aβ) peptide within plaques robustly invested with reactive microglia. This supports the notion that Aβ stimulation of microglial activation is one source of brain inflammatory changes during disease. Aβ is a cleavage product of the ubiquitously expressed amyloid precursor protein (APP) and is able to self-associate into a wide variety of differently sized and structurally distinct multimers. In this study, we demonstrate both in vitro and in vivo that nonfibrillar, oligomeric forms of Aβ are able to interact with the parent APP protein to stimulate microglial activation. This provides a mechanism by which metabolism of APP results in possible autocrine or paracrine Aβ production to drive the microgliosis associated with AD brains.
Collapse
Affiliation(s)
- Gunjan D Manocha
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Angela M Floden
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Keiko Rausch
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Joshua A Kulas
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Brett A McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Lalida Rojanathammanee
- Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000 Thailand
| | - Kelley R Puig
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Kendra L Puig
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Sanjib Karki
- Department of Chemistry and Biochemistry, University of Missouri-St. Louis, St. Louis, Missouri 63121-4400, and
| | - Michael R Nichols
- Department of Chemistry and Biochemistry, University of Missouri-St. Louis, St. Louis, Missouri 63121-4400, and
| | - Diane C Darland
- Department of Biology, University of North Dakota, Grand Forks, North Dakota 58202
| | - James E Porter
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Colin K Combs
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203,
| |
Collapse
|
220
|
Lovelace MD, Varney B, Sundaram G, Franco NF, Ng ML, Pai S, Lim CK, Guillemin GJ, Brew BJ. Current Evidence for a Role of the Kynurenine Pathway of Tryptophan Metabolism in Multiple Sclerosis. Front Immunol 2016; 7:246. [PMID: 27540379 PMCID: PMC4972824 DOI: 10.3389/fimmu.2016.00246] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022] Open
Abstract
The kynurenine pathway (KP) is the major metabolic pathway of the essential amino acid tryptophan (TRP). Stimulation by inflammatory molecules, such as interferon-γ (IFN-γ), is the trigger for induction of the KP, driving a complex cascade of production of both neuroprotective and neurotoxic metabolites, and in turn, regulation of the immune response and responses of brain cells to the KP metabolites. Consequently, substantial evidence has accumulated over the past couple of decades that dysregulation of the KP and the production of neurotoxic metabolites are associated with many neuroinflammatory and neurodegenerative diseases, including Parkinson’s disease, AIDS-related dementia, motor neurone disease, schizophrenia, Huntington’s disease, and brain cancers. In the past decade, evidence of the link between the KP and multiple sclerosis (MS) has rapidly grown and has implicated the KP in MS pathogenesis. KP enzymes, indoleamine 2,3-dioxygenase (IDO-1) and tryptophan dioxygenase (highest expression in hepatic cells), are the principal enzymes triggering activation of the KP to produce kynurenine from TRP. This is in preference to other routes such as serotonin and melatonin production. In neurological disease, degradation of the blood–brain barrier, even if transient, allows the entry of blood monocytes into the brain parenchyma. Similar to microglia and macrophages, these cells are highly responsive to IFN-γ, which upregulates the expression of enzymes, including IDO-1, producing neurotoxic KP metabolites such as quinolinic acid. These metabolites circulate systemically or are released locally in the brain and can contribute to the excitotoxic death of oligodendrocytes and neurons in neurological disease principally by virtue of their agonist activity at N-methyl-d-aspartic acid receptors. The latest evidence is presented and discussed. The enzymes that control the checkpoints in the KP represent an attractive therapeutic target, and consequently several KP inhibitors are currently in clinical trials for other neurological diseases, and hence may make suitable candidates for MS patients. Underpinning these drug discovery endeavors, in recent years, several advances have been made in how KP metabolites are assayed in various biological fluids, and tremendous advancements have been made in how specimens are imaged to determine disease progression and involvement of various cell types and molecules in MS.
Collapse
Affiliation(s)
- Michael D Lovelace
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Bianca Varney
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Gayathri Sundaram
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Nunzio F Franco
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Mei Li Ng
- Faculty of Medicine, Sydney Medical School, University of Sydney , Sydney, NSW , Australia
| | - Saparna Pai
- Sydney Medical School, University of Sydney , Sydney, NSW , Australia
| | - Chai K Lim
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University , Sydney, NSW , Australia
| | - Gilles J Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University , Sydney, NSW , Australia
| | - Bruce J Brew
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia; Department of Neurology, St Vincent's Hospital, Sydney, NSW, Australia
| |
Collapse
|
221
|
Levin MC, Lee S, Gardner LA, Shin Y, Douglas JN, Salapa H. Autoantibodies to heterogeneous nuclear ribonuclear protein A1 (hnRNPA1) cause altered 'ribostasis' and neurodegeneration; the legacy of HAM/TSP as a model of progressive multiple sclerosis. J Neuroimmunol 2016; 304:56-62. [PMID: 27449854 DOI: 10.1016/j.jneuroim.2016.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/06/2016] [Indexed: 12/23/2022]
Abstract
Several years following its discovery in 1980, infection with human T-lymphotropic virus type 1 (HTLV-1) was shown to cause HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP), a disease biologically similar to progressive forms of multiple sclerosis (MS). In this manuscript, we review some of the clinical, pathological, and immunological similarities between HAM/TSP and MS with an emphasis on how autoantibodies to an RNA binding protein, heterogeneous nuclear ribonuclear protein A1 (hnRNP A1), might contribute to neurodegeneration in immune mediated diseases of the central nervous system.
Collapse
Affiliation(s)
- Michael C Levin
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Anatomy/Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Sangmin Lee
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lidia A Gardner
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yoojin Shin
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joshua N Douglas
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Anatomy/Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hannah Salapa
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Anatomy/Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
222
|
Lee H, Narayanan S, Brown RA, Chen JT, Atkins HL, Freedman MS, Arnold DL. Brain atrophy after bone marrow transplantation for treatment of multiple sclerosis. Mult Scler 2016; 23:420-431. [DOI: 10.1177/1352458516650992] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: A cohort of patients with poor-prognosis multiple sclerosis (MS) underwent chemotherapy-based immune ablation followed by immune reconstitution with an autologous hematopoietic stem cell transplant (IA/aHSCT). This eliminated new focal inflammatory activity, but resulted in early acceleration of brain atrophy. Objective: We modeled the time course of whole-brain volume in 19 patients to identify the baseline predictors of atrophy and to estimate the average rate of atrophy after IA/aHSCT. Methods: Percentage whole-brain volume changes were calculated between the baseline and follow-up magnetic resonance imaging (MRI; mean duration: 5 years). A mixed-effects model was applied using two predictors: total busulfan dose and baseline volume of T1-weighted white-matter lesions. Results: Treatment was followed by accelerated whole-brain volume loss averaging 3.3%. Both the busulfan dose and the baseline lesion volume were significant predictors. The atrophy slowed progressively over approximately 2.5 years. There was no evidence that resolution of edema contributed to volume loss. The mean rate of long-term atrophy was −0.23% per year, consistent with the rate expected from normal aging. Conclusion: Following IA/aHSCT, MS patients showed accelerated whole-brain atrophy that was likely associated with treatment-related toxicity and degeneration of “committed” tissues. Atrophy eventually slowed to that expected from normal aging, suggesting that stopping inflammatory activity in MS can reduce secondary degeneration and atrophy.
Collapse
Affiliation(s)
- Hyunwoo Lee
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Sridar Narayanan
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Robert A Brown
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Jacqueline T Chen
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Harold L Atkins
- Ottawa Hospital Blood and Marrow Transplant Program, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Mark S Freedman
- University of Ottawa and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Douglas L Arnold
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
223
|
Douglas JN, Gardner LA, Salapa HE, Lalor SJ, Lee S, Segal BM, Sawchenko PE, Levin MC. Antibodies to the RNA-binding protein hnRNP A1 contribute to neurodegeneration in a model of central nervous system autoimmune inflammatory disease. J Neuroinflammation 2016; 13:178. [PMID: 27391474 PMCID: PMC4938923 DOI: 10.1186/s12974-016-0647-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/29/2016] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Neurodegeneration is believed to be the primary cause of permanent, long-term disability in patients with multiple sclerosis. The cause of neurodegeneration in multiple sclerosis appears to be multifactorial. One mechanism that has been implicated in the pathogenesis of neurodegeneration in multiple sclerosis is the targeting of neuronal and axonal antigens by autoantibodies. Multiple sclerosis patients develop antibodies to the RNA-binding protein, heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1), which is enriched in neurons. We hypothesized that anti-hnRNP A1 antibodies would contribute to neurodegeneration in an animal model of multiple sclerosis. METHODS Following induction of experimental autoimmune encephalomyelitis (EAE) by direct immunization with myelin oligodendrocyte glycoprotein, mice were injected with anti-hnRNP A1 or control antibodies. Animals were examined clinically, and the central nervous system (CNS) tissues were tested for neurodegeneration with Fluoro-Jade C, a marker of degenerating neural elements. RESULTS Injection of anti-hnRNP A1 antibodies in mice with EAE worsened clinical disease, altered the clinical disease phenotype, and caused neurodegeneration preferentially in the ventral spinocerebellar tract and deep white matter of the cerebellum in the CNS. Neurodegeneration in mice injected with hnRNP A1-M9 antibodies compared to control groups was consistent with "dying back" axonal degeneration. CONCLUSIONS These data suggest that antibodies to the RNA-binding protein hnRNP A1 contribute to neurodegeneration in immune-mediated disease of the CNS.
Collapse
Affiliation(s)
- Joshua N. Douglas
- />Department of Neurology, University of Tennessee Health Science Center, 855 Monroe Avenue, Room 415, Memphis, TN 38163 USA
- />The Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN USA
| | - Lidia A. Gardner
- />Research Service, VA Medical Center, Memphis, TN USA
- />Department of Neurology, University of Tennessee Health Science Center, 855 Monroe Avenue, Room 415, Memphis, TN 38163 USA
- />The Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN USA
| | - Hannah E. Salapa
- />Department of Neurology, University of Tennessee Health Science Center, 855 Monroe Avenue, Room 415, Memphis, TN 38163 USA
- />The Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN USA
| | - Stephen J. Lalor
- />Department of Neurology, University of Michigan Medical School, Ann Arbor, MI USA
| | - Sangmin Lee
- />Research Service, VA Medical Center, Memphis, TN USA
- />Department of Neurology, University of Tennessee Health Science Center, 855 Monroe Avenue, Room 415, Memphis, TN 38163 USA
- />The Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN USA
| | - Benjamin M. Segal
- />Department of Neurology, University of Michigan Medical School, Ann Arbor, MI USA
- />Neurology Service, VA Ann Arbor Health Care System, Ann Arbor, MI USA
| | - Paul E. Sawchenko
- />Laboratory of Neuronal Structure & Function, The Salk Institute, La Jolla, CA USA
| | - Michael C. Levin
- />Research Service, VA Medical Center, Memphis, TN USA
- />Department of Neurology, University of Tennessee Health Science Center, 855 Monroe Avenue, Room 415, Memphis, TN 38163 USA
- />The Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN USA
| |
Collapse
|
224
|
Colucci M, Roccatagliata L, Capello E, Narciso E, Latronico N, Tabaton M, Mancardi GL. The 14-3-3 protein in multiple sclerosis: a marker of disease severity. Mult Scler 2016; 10:477-81. [PMID: 15471360 DOI: 10.1191/1352458504ms1089oa] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Context: In multiple sclerosis (MS) axonal damage is an early event and is probably to be considered the most relevant cause of permanent and progressive disability. Objectives: To investigate the value of the increase of 14-3-3 and tau proteins in the cerebrospinal fluid (CSF) as peripheral markers of axonal pathology and predictors of disease evolution. Patients and methods: In the CSF samples obtained from 63 patients with demyelinating diseases (DD), including 20 clinically isolated syndromes (CIS) and 43 clinically defined MS, as well as from 56 controls, we analysed the presence of 14-3-3 reactivity by immunoblot analysis along with the concentration of tau protein by sandwich ELISA. Results: The percentage of DD subjects showing a positive 14-3-3 protein CSF reactivity (38%) was significantly higher than the one previously detected, and was correlated in the MS patients with a more severe clinical phenotype in terms of degree of disability and rate of disease progression, during a 10-month mean clinical follow-up. On the contrary, the levels of the CSF-tau protein were highly variable in DD and control subjects, and the mean CSF-tau concentration was similar in both groups. Conclusions: The immunoblot analysis of 14-3-3 protein in the CSF could be a useful marker to identify a subgroup of DD patients with high risk of developing severe disability.
Collapse
Affiliation(s)
- M Colucci
- Department of Neuroscience, Ophthalmology and Genetic and Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | | | | | | | | | | | | |
Collapse
|
225
|
Arnold DL. The Axonal Hypothesis and Magnetic Resonance Spectroscopy of MS. Mult Scler 2016. [DOI: 10.1177/135245859800400607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
226
|
Lim ET, Grant D, Pashenkov M, Keir G, Thompson EJ, Söderström M, Giovannoni G. Cerebrospinal fluid levels of brain specific proteins in optic neuritis. Mult Scler 2016; 10:261-5. [PMID: 15222688 DOI: 10.1191/1352458504ms1020oa] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This study evaluates levels of cerebrospinal fluid (C SF) brain-specific proteins (BSP) in subjects with optic neuritis (O N) who are at high risk of progression to multiple sclerosis (MS). Forty-one subjects had acute O N and 17 subjects with other neurological diseases (OND) served as controls. Twenty-o ne subjects with O N had white matter lesions on magnetic resonance imaging (MRI) and intrathecal synthesis of oligoclonal IgG bands (OB) consistent with being at high risk of progression to MS; eight of whom later were diagnosed with clinically definite MS (C DMS). Levels of S100B, ferritin and two neurofilament heavy chain phosphoforms (NfHSMI34 and NfHSMI35) were analysed using ELISA technique. A putative index of ‘axonal health’ was expressed as a ratio of NfHSMI34 to NfHSMI35. NfHSMI34 and the NfHSMI34:SMI35 were significantly elevated in subjects with O N compared to controls. No significant differences in levels of C SF BSP were seen between O N subjects with C DMS plus those at high risk of progression to MS and O N subjects with normal MRI and negative C SF analysis. In conclusion, there is evidence of axonal damage in subjects who present with O N, which is independent of the diagnosis of C DMS.
Collapse
Affiliation(s)
- E T Lim
- Department of Neuroinflammation, Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | | | | | | | | | | | | |
Collapse
|
227
|
Davies CL, Miron VE. Distinct origins, gene expression and function of microglia and monocyte-derived macrophages in CNS myelin injury and regeneration. Clin Immunol 2016; 189:57-62. [PMID: 27377535 DOI: 10.1016/j.clim.2016.06.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/27/2016] [Accepted: 06/30/2016] [Indexed: 01/07/2023]
Abstract
Central nervous system (CNS) injury incurs a rapid innate immune response, including that from macrophages derived from endogenous microglia and circulating monocytes infiltrating the lesion site. One example of such injury is the demyelination observed in the autoimmune disease multiple sclerosis (MS), where macrophages are implicated in both myelin injury and regeneration. Although initially microglia and monocyte-derived macrophages were considered to have identical origins, gene expression, and function, recent advances have revealed important distinctions in all three categories and have caused a paradigm shift in view of their unique identity and roles. This has important consequences for understanding their individual contribution to neurological function and therapeutic targeting of these populations in diseases like MS. Here, we address the differences between CNS endogenous and exogenously-derived macrophages with a particular focus on myelin damage and regeneration.
Collapse
Affiliation(s)
- Claire L Davies
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | - Veronique E Miron
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| |
Collapse
|
228
|
Prognostic factors associated with long-term disability and secondary progression in patients with Multiple Sclerosis. Mult Scler Relat Disord 2016; 8:27-34. [DOI: 10.1016/j.msard.2016.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/18/2016] [Accepted: 03/22/2016] [Indexed: 11/20/2022]
|
229
|
Trapp BD, Ransohoff RM, Fisher E, Rudick RA. Neurodegeneration in Multiple Sclerosis: Relationship to Neurological Disability. Neuroscientist 2016. [DOI: 10.1177/107385849900500107] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the CNS. Most MS patients follow a relapsing-remitting course (RR-MS) for 8 to 15 years that transforms into a secondary progressive disease course (SP-MS). In this review, we discuss current data that describe MS as a neurodegenerative disease in which axonal loss is the major cause of irreversible neurological disability in MS patients. Neurological deficits in MS patients have two pathogeneses: acute inflammatory demyelination and axonal degeneration. Disability caused by inflammatory demyelination clinically dominates the early stages of RR-MS and is reversible. Axonal transection occurs at sites of inflammation and begins at disease onset but is clinically silent in RR-MS because the CNS compensates for neuronal loss. Once a threshold of axon loss is ex ceeded, MS patients enter an irreversible secondary progressive stage. In SP-MS, axonal degeneration is caused by chronic demyelination and may be irreversibly progressive. This view of MS provides a concep tional framework that explains conversion of RR-MS to SP-MS and provides a rationale for early aggressive anti-inflammatory and neuroprotective therapies. NEUROSCIENTIST 5:48-57, 1999
Collapse
Affiliation(s)
- Bruce D. Trapp
- Departments of Neurosciences, The Cleveland Clinic Foundation
Cleveland, Ohio
| | - Richard M. Ransohoff
- Departments of Neurosciences, Lerner Research Institute
and the Mellen Center for Multiple Sclerosis Treatment and Research (RMR,
RAR) The Cleveland Clinic Foundation Cleveland, Ohio
| | - Elizabeth Fisher
- Biomedical Engineering, The Cleveland Clinic Foundation
Cleveland, Ohio
| | - Richard A. Rudick
- Departments of Neurosciences, Lerner Research Institute
and the Mellen Center for Multiple Sclerosis Treatment and Research (RMR,
RAR) The Cleveland Clinic Foundation Cleveland, Ohio
| |
Collapse
|
230
|
Olsen JA, Kenna LA, Tipon RC, Spelios MG, Stecker MM, Akirav EM. A Minimally-invasive Blood-derived Biomarker of Oligodendrocyte Cell-loss in Multiple Sclerosis. EBioMedicine 2016; 10:227-35. [PMID: 27381476 PMCID: PMC5006601 DOI: 10.1016/j.ebiom.2016.06.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/15/2016] [Accepted: 06/23/2016] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system (CNS). Minimally invasive biomarkers of MS are required for disease diagnosis and treatment. Differentially methylated circulating-free DNA (cfDNA) is a useful biomarker for disease diagnosis and prognosis, and may offer to be a viable approach for understanding MS. Here, methylation-specific primers and quantitative real-time PCR were used to study methylation patterns of the myelin oligodendrocyte glycoprotein (MOG) gene, which is expressed primarily in myelin-producing oligodendrocytes (ODCs). MOG-DNA was demethylated in O4+ ODCs in mice and in DNA from human oligodendrocyte precursor cells (OPCs) when compared with other cell types. In the cuprizone-fed mouse model of demyelination, ODC derived demethylated MOG cfDNA was increased in serum and was associated with tissue-wide demyelination, demonstrating the utility of demethylated MOG cfDNA as a biomarker of ODC death. Collected sera from patients with active (symptomatic) relapsing-remitting MS (RRMS) demonstrated a higher signature of demethylated MOG cfDNA when compared with patients with inactive disease and healthy controls. Taken together, these results offer a minimally invasive approach to measuring ODC death in the blood of MS patients that may be used to monitor disease progression. Currently, there are no molecular biomarkers of multiple sclerosis (MS). A minimally invasive assay for measuring oligodendrocyte (ODC) cell loss in relapsing-remitting MS is described. DNA methylation of the myelin oligodendrocyte glycoprotein gene is used as a measure of ODC loss in the blood.
Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system. Currently, there are no molecular biomarkers of MS, thereby limiting disease diagnosis, prognosis, and assessment of new clinical interventions. Myelin oligodendrocyte glycoprotein (MOG) is expressed solely by oligodendrocytes (ODCs) as an integral part of the myelin sheath. This report describes a minimally invasive biomarker assay for measuring ODC-derived DNA in the blood of MS patients. It describes the presence of unique DNA methylation patterns in the MOG gene in ODCs, which is used to design methylation-specific primers. Analysis of sera from patients with active relapsing-remitting MS shows an increase in levels of ODC-derived circulating free DNA when compared with inactive disease and healthy controls.
Collapse
Affiliation(s)
- John A Olsen
- Research Institute, Islet Biology, Winthrop-University Hospital, Mineola, NY, USA
| | - Lauren A Kenna
- Research Institute, Islet Biology, Winthrop-University Hospital, Mineola, NY, USA
| | - Regine C Tipon
- Research Institute, Islet Biology, Winthrop-University Hospital, Mineola, NY, USA
| | - Michael G Spelios
- Research Institute, Islet Biology, Winthrop-University Hospital, Mineola, NY, USA
| | - Mark M Stecker
- Department of Neuroscience, Winthrop-University Hospital Mineola, NY, USA
| | - Eitan M Akirav
- Research Institute, Islet Biology, Winthrop-University Hospital, Mineola, NY, USA; Stony Brook University School of Medicine, Stony Brook, NY, USA.
| |
Collapse
|
231
|
Bhise V, Dhib-Jalbut S. Further understanding of the immunopathology of multiple sclerosis: impact on future treatments. Expert Rev Clin Immunol 2016; 12:1069-89. [PMID: 27191526 DOI: 10.1080/1744666x.2016.1191351] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The understanding of the immunopathogenesis of multiple sclerosis (MS) has expanded with more research into T-cell subtypes, cytokine contributors, B-cell participation, mitochondrial dysfunction, and more. Treatment options have rapidly expanded with three relatively recent oral therapy alternatives entering the arena. AREAS COVERED In the following review, we discuss current mechanisms of immune dysregulation in MS, how they relate to current treatments, and the impact these findings will have on the future of therapy. Expert commentary: The efficacy of these medications and understanding their mechanisms of actions validates the immunopathogenic mechanisms thought to underlie MS. Further research has exposed new targets, while new promising therapies have shed light on new aspects into the pathophysiology of MS.
Collapse
Affiliation(s)
- Vikram Bhise
- a Rutgers Biomedical and Health Sciences - Departments of Pediatrics , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| | - Suhayl Dhib-Jalbut
- b Rutgers Biomedical and Health Sciences - Departments of Neurology , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| |
Collapse
|
232
|
van Luijn MM, van Meurs M, Stoop MP, Verbraak E, Wierenga-Wolf AF, Melief MJ, Kreft KL, Verdijk RM, 't Hart BA, Luider TM, Laman JD, Hintzen RQ. Elevated Expression of the Cerebrospinal Fluid Disease Markers Chromogranin A and Clusterin in Astrocytes of Multiple Sclerosis White Matter Lesions. J Neuropathol Exp Neurol 2016; 75:86-98. [PMID: 26683597 DOI: 10.1093/jnen/nlv004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Using proteomics, we previously identified chromogranin A (CgA) and clusterin (CLU) as disease-related proteins in the cerebrospinal fluid (CSF) of patients with multiple sclerosis (MS). CgA and CLU are involved in cell survival and are implicated in neurodegenerative disorders and may also have roles in MS pathophysiology. We investigated CgA and CLU expression in lesions and nonlesional regions in postmortem brains of MS patients and controls and in the brains of marmosets with experimental autoimmune encephalomyelitis. By quantitative PCR, mRNA levels of CgA and CLU were elevated in white matter but not in grey matter of MS patients. In situ analyses showed greater expression of CgA and CLU in white matter lesions than in normal-appearing regions in MS patients and in the marmosets, primarily in or adjacent to perivascular spaces and inflammatory infiltrates. Both proteins were expressed by glial fibrillary acidic protein-positive astrocytes. CgA was more localized in astrocytic processes and endfeet surrounding blood vessels and was abundant in the superficial glia limitans and ependyma, 2 CSF-brain borders. Increased expression of CgA and CLU in reactive astrocytes in MS white matter lesions supports a role for these molecules as neuro-inflammatory mediators and their potential as CSF markers of active pathological processes in MS patients.
Collapse
|
233
|
Matías-Guíu J, Oreja-Guevara C, Matias-Guiu JA, Gomez-Pinedo U. Vitamin D and remyelination in multiple sclerosis. Neurologia 2016; 33:177-186. [PMID: 27321170 DOI: 10.1016/j.nrl.2016.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 05/12/2016] [Indexed: 12/01/2022] Open
Abstract
INTRODUCTION Several studies have found an association between multiple sclerosis and vitamin D (VD) deficiency, which suggests that VD may play a role in the immune response. However, few studies have addressed its role in remyelination. DEVELOPMENT The VD receptor and the enzymes transforming VD into metabolites which activate the VD receptor are expressed in central nervous system (CNS) cells, which suggests a potential effect of VD on the CNS. Both in vitro and animal model studies have shown that VD may play a role in myelination by acting on factors that influence the microenvironment which promotes both proliferation and differentiation of neural stem cells into oligodendrocyte progenitor cells and oligodendrocytes. It remains unknown whether the mechanisms of internalisation of VD in the CNS are synergistic with or antagonistic to the mechanisms that facilitate the entry of VD metabolites into immune cells. CONCLUSIONS VD seems to play a role in the CNS and our hypothesis is that VD is involved in remyelination. Understanding the basic mechanisms of VD in myelination is necessary to manage multiple sclerosis patients with VD deficiency.
Collapse
Affiliation(s)
- J Matías-Guíu
- Servicio de Neurología, Hospital Clínico San Carlos, Facultad de Medicina, Universidad Complutense, IdiSSC, Madrid, España.
| | - C Oreja-Guevara
- Servicio de Neurología, Hospital Clínico San Carlos, Facultad de Medicina, Universidad Complutense, IdiSSC, Madrid, España
| | - J A Matias-Guiu
- Servicio de Neurología, Hospital Clínico San Carlos, Facultad de Medicina, Universidad Complutense, IdiSSC, Madrid, España
| | - U Gomez-Pinedo
- Servicio de Neurología, Hospital Clínico San Carlos, Facultad de Medicina, Universidad Complutense, IdiSSC, Madrid, España
| |
Collapse
|
234
|
Rawji KS, Mishra MK, Yong VW. Regenerative Capacity of Macrophages for Remyelination. Front Cell Dev Biol 2016; 4:47. [PMID: 27243011 PMCID: PMC4873494 DOI: 10.3389/fcell.2016.00047] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 05/06/2016] [Indexed: 11/13/2022] Open
Abstract
White matter injury, consisting of loss of axons, myelin, and oligodendrocytes, is common in many neurological disorders and is believed to underlie several motor and sensory deficits. Remyelination is the process in which the insulative myelin sheath is restored to axons, thereby facilitating recovery from functional loss. Remyelination proceeds with oligodendrocyte precursor cells (OPCs) that differentiate into oligodendrocytes to synthesize the new myelin sheath after demyelination. This process is influenced by several factors, including trophic factors, inhibitory molecules in the lesion microenvironment, age of the subject, as well as the inflammatory response. Currently studied strategies that enhance remyelination consist of pharmacological approaches that directly induce OPC differentiation or using agents to neutralize the inhibitory microenvironment. Another strategy is to harness a reparative inflammatory response. This response, coordinated by central nervous system resident microglia and peripherally-derived infiltrating macrophages, has been shown to be important in the remyelination process. These innate immune cells perform important functions in remyelination, including the proteolysis and phagocytosis of inhibitory molecules present in the lesion microenvironment, the provision of trophic and metabolic factors to OPCs, in addition to iron handling capacity. Additionally, an initial pro-inflammatory phase followed by a regulatory/anti-inflammatory phase has been shown to be important for OPC proliferation and differentiation, respectively. This review will discuss the beneficial roles of macrophages/microglia in remyelination and discuss therapeutic strategies to obtain the optimal regenerative macrophage phenotype for enhanced remyelination.
Collapse
Affiliation(s)
- Khalil S Rawji
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, AB, Canada
| | - Manoj K Mishra
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, AB, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, AB, Canada
| |
Collapse
|
235
|
Yandamuri SS, Lane TE. Imaging Axonal Degeneration and Repair in Preclinical Animal Models of Multiple Sclerosis. Front Immunol 2016; 7:189. [PMID: 27242796 PMCID: PMC4871863 DOI: 10.3389/fimmu.2016.00189] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/02/2016] [Indexed: 12/25/2022] Open
Abstract
Multiple sclerosis (MS) is a central nervous system (CNS) disease characterized by chronic neuroinflammation, demyelination, and axonal damage. Infiltration of activated lymphocytes and myeloid cells are thought to be primarily responsible for white matter damage and axonopathy. Over time, this neurologic damage manifests clinically as debilitating motor and cognitive symptoms. Existing MS therapies focus on symptom relief and delay of disease progression through reduction of neuroinflammation. However, long-term strategies to remyelinate, protect, or regenerate axons have remained elusive, posing a challenge to treating progressive forms of MS. Preclinical mouse models and techniques, such as immunohistochemistry, flow cytometry, and genomic and proteomic analysis have provided advances in our understanding of discrete time-points of pathology following disease induction. More recently, in vivo and in situ two-photon (2P) microscopy has made it possible to visualize continuous real-time cellular behavior and structural changes occurring within the CNS during neuropathology. Research utilizing 2P imaging to study axonopathy in neuroinflammatory demyelinating disease has focused on five areas: (1) axonal morphologic changes, (2) organelle transport and health, (3) relationship to inflammation, (4) neuronal excitotoxicity, and (5) regenerative therapies. 2P imaging may also be used to identify novel therapeutic targets via identification and clarification of dynamic cellular and molecular mechanisms of axonal regeneration and remyelination. Here, we review tools that have made 2P accessible for imaging neuropathologies and advances in our understanding of axonal degeneration and repair in preclinical models of demyelinating diseases.
Collapse
Affiliation(s)
| | - Thomas E. Lane
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
- Department of Pathology, School of Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
236
|
Sheth V, Shao H, Chen J, Vandenberg S, Corey-Bloom J, Bydder GM, Du J. Magnetic resonance imaging of myelin using ultrashort Echo time (UTE) pulse sequences: Phantom, specimen, volunteer and multiple sclerosis patient studies. Neuroimage 2016; 136:37-44. [PMID: 27155128 DOI: 10.1016/j.neuroimage.2016.05.012] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 04/25/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022] Open
Abstract
Clinical magnetic resonance imaging of multiple sclerosis (MS) has focused on indirect imaging of myelin in white matter by detecting signal from protons in the water associated with myelin. Here we show that protons in myelin can be directly imaged using ultrashort echo time (UTE) free induction decay (FID) and imaging sequences on a clinical 3T MR scanner. An adiabatic inversion recovery UTE (IR-UTE) sequence was used to detect signal from myelin and simultaneously suppress signal from water protons. Validation studies were performed on myelin lipid and myelin basic protein (MBP) phantoms in the forms of lyophilized powders as well as suspensions in D2O and H2O. IR-UTE sequences were then used to image MS brain specimens, healthy volunteers, and patients. The T2* of myelin was measured using a UTE FID sequence, as well as UTE and IR-UTE sequences at different TEs. T2* values of ~110-330μs were measured with UTE FID, as well as with UTE and IR-UTE sequences for myelin powders, myelin-D2O and myelin-H2O phantoms, consistent with selective imaging of myelin protons with IR-UTE sequences. Our studies showed myelin selective imaging of white matter in the brains in vitro and in vivo. Complete or partial signal loss was observed in specimens in areas of the brain with histopathologic evidence of myelin loss, and in the brain of patients with MS.
Collapse
Affiliation(s)
- Vipul Sheth
- Department of Radiology, University of California, San Diego, United States
| | - Hongda Shao
- Department of Radiology, University of California, San Diego, United States
| | - Jun Chen
- Department of Radiology, University of California, San Diego, United States
| | - Scott Vandenberg
- Department of Pathology, University of California, San Francisco, United States
| | - Jody Corey-Bloom
- Department of Neurosciences, University of California, San Diego, United States
| | - Graeme M Bydder
- Department of Radiology, University of California, San Diego, United States
| | - Jiang Du
- Department of Radiology, University of California, San Diego, United States.
| |
Collapse
|
237
|
Graham SL, Klistorner A. Afferent visual pathways in multiple sclerosis: a review. Clin Exp Ophthalmol 2016; 45:62-72. [DOI: 10.1111/ceo.12751] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 02/04/2023]
Affiliation(s)
- Stuart L Graham
- Faculty of Medicine and Human Science; Macquarie University; Sydney New South Wales Australia
- Save Sight Institute; Sydney University; Sydney New South Wales Australia
| | - Alexander Klistorner
- Faculty of Medicine and Human Science; Macquarie University; Sydney New South Wales Australia
- Save Sight Institute; Sydney University; Sydney New South Wales Australia
| |
Collapse
|
238
|
Faizi M, Salimi A, Seydi E, Naserzadeh P, Kouhnavard M, Rahimi A, Pourahmad J. Toxicity of cuprizone a Cu(2+) chelating agent on isolated mouse brain mitochondria: a justification for demyelination and subsequent behavioral dysfunction. Toxicol Mech Methods 2016; 26:276-83. [PMID: 27088566 DOI: 10.3109/15376516.2016.1172284] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multiple Sclerosis (MS) is a complex disease with an unknown etiology and no effective cure, despite decades of extensive research that led to the development of several partially effective treatments. In this study we aimed to investigate brain mitochondrial dysfunction in demyelination induced by cuprizone in mice. Cuprizone was used for induction of demyelination in mice through a diet containing 0.2% w/w cuprizone for 5 weeks. Behavioral tests for proving of MS was performed and then mitochondria from brain of animals were isolated and afterwards parameters of mitochondrial dysfunction examined. Results of mitochondrial dysfunction parameters such as mitochondrial swelling, production ROS, collapse of the membrane potential showed that isolated mitochondria from cuprizone treated mice have been damaged compared to those of untreated control mice. It is likely that demyelination induced mitochondrial damage led to increased mitochondrial ROS formation and progression of oxidative damages in neurons. It is suggested that cuprizone which is a Cu(2+) chelating agent causes impairment of electron transport chain (complex IV) and antioxidant system (SOD) in mitochondria leading to decreased ATP production and increased ROS formation.
Collapse
Affiliation(s)
- Mehrdad Faizi
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Ahmad Salimi
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran ;,b Department of Pharmacology and Toxicology , School of Pharmacy, Ardabil University of Medical Science , Ardabil , Iran ;,c Students Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Enayatolla Seydi
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran ;,c Students Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Parvaneh Naserzadeh
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran ;,c Students Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Mehdi Kouhnavard
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Atena Rahimi
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Jalal Pourahmad
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| |
Collapse
|
239
|
Hares K, Redondo J, Kemp K, Rice C, Scolding N, Wilkins A. Axonal motor protein KIF5A and associated cargo deficits in multiple sclerosis lesional and normal-appearing white matter. Neuropathol Appl Neurobiol 2016; 43:227-241. [DOI: 10.1111/nan.12305] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/05/2016] [Accepted: 01/20/2016] [Indexed: 11/28/2022]
Affiliation(s)
- K. Hares
- MS and Stem Cell Group; School of Clinical Sciences; University of Bristol; Bristol UK
| | - J. Redondo
- MS and Stem Cell Group; School of Clinical Sciences; University of Bristol; Bristol UK
| | - K. Kemp
- MS and Stem Cell Group; School of Clinical Sciences; University of Bristol; Bristol UK
| | - C. Rice
- MS and Stem Cell Group; School of Clinical Sciences; University of Bristol; Bristol UK
| | - N. Scolding
- MS and Stem Cell Group; School of Clinical Sciences; University of Bristol; Bristol UK
| | - A. Wilkins
- MS and Stem Cell Group; School of Clinical Sciences; University of Bristol; Bristol UK
| |
Collapse
|
240
|
Matías-Guiu JA, Oreja-Guevara C, Cabrera-Martín MN, Moreno-Ramos T, Carreras JL, Matías-Guiu J. Amyloid Proteins and Their Role in Multiple Sclerosis. Considerations in the Use of Amyloid-PET Imaging. Front Neurol 2016; 7:53. [PMID: 27065425 PMCID: PMC4814935 DOI: 10.3389/fneur.2016.00053] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 03/22/2016] [Indexed: 02/06/2023] Open
Abstract
Thioflavin T derivatives are used in positron-emission tomography (PET) studies to detect amyloid protein deposits in patients with Alzheimer disease. These tracers bind extensively to white matter, which suggests that they may be useful in studies of multiple sclerosis (MS), and that proteins resulting from proteolytic processing of the amyloid precursor protein (APP) may contribute to MS. This article reviews data from both clinical and preclinical studies addressing the role of these proteins, whether they are detected in CSF studies or using PET imaging. APP is widely expressed in demyelinated axons and may have a protective effect in MS and in experimental allergic encephalomyelitis in animals. Several mechanisms associated with this increased expression may affect the degree of remyelination in MS. Amyloid-PET imaging may help determine the degree of demyelination and provide information on the molecular changes linked to APP proteolytic processing experienced by patients with MS.
Collapse
Affiliation(s)
- Jordi A Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Celia Oreja-Guevara
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - María Nieves Cabrera-Martín
- Department of Nuclear Medicine, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Teresa Moreno-Ramos
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - José Luis Carreras
- Department of Nuclear Medicine, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Jorge Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| |
Collapse
|
241
|
Patejdl R, Penner IK, Noack TK, Zettl UK. Multiple sclerosis and fatigue: A review on the contribution of inflammation and immune-mediated neurodegeneration. Autoimmun Rev 2016; 15:210-20. [DOI: 10.1016/j.autrev.2015.11.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 11/13/2015] [Indexed: 02/02/2023]
|
242
|
Ossola B, Zhao C, Compston A, Pluchino S, Franklin RJM, Spillantini MG. Neuronal expression of pathological tau accelerates oligodendrocyte progenitor cell differentiation. Glia 2016; 64:457-71. [PMID: 26576485 PMCID: PMC5132073 DOI: 10.1002/glia.22940] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/19/2015] [Indexed: 12/03/2022]
Abstract
Oligodendrocyte progenitor cell (OPC) differentiation is an important therapeutic target to promote remyelination in multiple sclerosis (MS). We previously reported hyperphosphorylated and aggregated microtubule-associated protein tau in MS lesions, suggesting its involvement in axonal degeneration. However, the influence of pathological tau-induced axonal damage on the potential for remyelination is unknown. Therefore, we investigated OPC differentiation in human P301S tau (P301S-htau) transgenic mice, both in vitro and in vivo following focal demyelination. In 2-month-old P301S-htau mice, which show hyperphosphorylated tau in neurons, we found atrophic axons in the spinal cord in the absence of prominent axonal degeneration. These signs of early axonal damage were associated with microgliosis and an upregulation of IL-1β and TNFα. Following in vivo focal white matter demyelination we found that OPCs differentiated more efficiently in P301S-htau mice than wild type (Wt) mice. We also found an increased level of myelin basic protein within the lesions, which however did not translate into increased remyelination due to higher susceptibility of P301S-htau axons to demyelination-induced degeneration compared to Wt axons. In vitro experiments confirmed higher differentiation capacity of OPCs from P301S-htau mice compared with Wt mice-derived OPCs. Because the OPCs from P301S-htau mice do not ectopically express the transgene, and when isolated from newborn mice behave like Wt mice-derived OPCs, we infer that their enhanced differentiation capacity must have been acquired through microenvironmental priming. Our data suggest the intriguing concept that damaged axons may signal to OPCs and promote their differentiation in the attempt at rescue by remyelination.
Collapse
Affiliation(s)
- Bernardino Ossola
- Department of Clinical Neurosciences, Clifford Allbutt BuildingUniversity of CambridgeCambridge CB2 0AHUnited Kingdom
| | - Chao Zhao
- Department of Clinical Neurosciences, Clifford Allbutt BuildingUniversity of CambridgeCambridge CB2 0AHUnited Kingdom
- Wellcome Trust‐Medical Research Council Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AHUnited Kingdom
| | - Alastair Compston
- Department of Clinical Neurosciences, Clifford Allbutt BuildingUniversity of CambridgeCambridge CB2 0AHUnited Kingdom
| | - Stefano Pluchino
- Department of Clinical Neurosciences, Clifford Allbutt BuildingUniversity of CambridgeCambridge CB2 0AHUnited Kingdom
- Wellcome Trust‐Medical Research Council Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AHUnited Kingdom
- NIHR Biomedical Research CentreCambridgeCB2 0AHUnited Kingdom
| | - Robin J. M. Franklin
- Department of Clinical Neurosciences, Clifford Allbutt BuildingUniversity of CambridgeCambridge CB2 0AHUnited Kingdom
- Wellcome Trust‐Medical Research Council Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AHUnited Kingdom
| | - Maria Grazia Spillantini
- Department of Clinical Neurosciences, Clifford Allbutt BuildingUniversity of CambridgeCambridge CB2 0AHUnited Kingdom
| |
Collapse
|
243
|
Nantes JC, Zhong J, Holmes SA, Whatley B, Narayanan S, Lapierre Y, Arnold DL, Koski L. Intracortical inhibition abnormality during the remission phase of multiple sclerosis is related to upper limb dexterity and lesions. Clin Neurophysiol 2016; 127:1503-1511. [DOI: 10.1016/j.clinph.2015.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 07/24/2015] [Accepted: 08/24/2015] [Indexed: 11/24/2022]
|
244
|
Brown JWL, Chard DT. The role of MRI in the evaluation of secondary progressive multiple sclerosis. Expert Rev Neurother 2016; 16:157-71. [PMID: 26692498 DOI: 10.1586/14737175.2016.1134323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Magnetic resonance imaging already has an established role in the diagnosis of multiple sclerosis, but it also has the potential to provide prognostic information, and to monitor [corrected] disease progression in clinical trials and practice. Magnetic resonance imaging measures are increasingly being used as the primary outcome in early phase clinical trials of immunomodulatory therapies (for example brain white matter lesion counts or volumes, and gadolinium contrast enhancing lesions) and putatively neuroprotective agents (for example measures of whole brain atrophy), and trials of agents that promote remyelination are also likely to follow suit. In this review we consider the use of magnetic resonance imaging measures as predictors and markers of disease progression in multiple sclerosis, and explore possible future directions in this rapidly developing field.
Collapse
Affiliation(s)
- J William L Brown
- a Department of Clinical Neurosciences , University of Cambridge , Cambridge , UK.,b NMR Research Unit, Queen Square Multiple Sclerosis Centre, Institute of Neurology , University College London (UCL) , London , UK
| | - Declan T Chard
- b NMR Research Unit, Queen Square Multiple Sclerosis Centre, Institute of Neurology , University College London (UCL) , London , UK.,c Biomedical Research Centre, National Institute for Health Research (NIHR) , University College London Hospitals (UCLH) , London , UK
| |
Collapse
|
245
|
Ratai EM, Gilberto González R. Clinical magnetic resonance spectroscopy of the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2016; 135:93-116. [PMID: 27432661 DOI: 10.1016/b978-0-444-53485-9.00005-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Proton magnetic resonance spectroscopy (1H MRS) is a noninvasive imaging technique that can easily be added to the conventional magnetic resonance (MR) imaging sequences. Using MRS one can directly compare spectra from pathologic or abnormal tissue and normal tissue. Metabolic changes arising from pathology that can be visualized by MRS may not be apparent from anatomy that can be visualized by conventional MR imaging. In addition, metabolic changes may precede anatomic changes. Thus, MRS is used for diagnostics, to observe disease progression, monitor therapeutic treatments, and to understand the pathogenesis of diseases. MRS may have an important impact on patient management. The purpose of this chapter is to provide practical guidance in the clinical application of MRS of the brain. This chapter provides an overview of MRS-detectable metabolites and their significance. In addition some specific current clinical applications of MRS will be discussed, including brain tumors, inborn errors of metabolism, leukodystrophies, ischemia, epilepsy, and neurodegenerative diseases. The chapter concludes with technical considerations and challenges of clinical MRS.
Collapse
Affiliation(s)
- Eva-Maria Ratai
- Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, and Athinoula A. Martinos Center for Biomedical Imaging, Boston, MA, USA.
| | - R Gilberto González
- Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, and Athinoula A. Martinos Center for Biomedical Imaging, Boston, MA, USA
| |
Collapse
|
246
|
Goodin DS. The epidemiology of multiple sclerosis: insights to a causal cascade. HANDBOOK OF CLINICAL NEUROLOGY 2016; 138:173-206. [PMID: 27637959 DOI: 10.1016/b978-0-12-802973-2.00011-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
MS-pathogenesis involves both genetic-susceptibility and environmental determinants. Three (or more) sequential environmental-factors are implicated. The first acts near birth, the second acts during childhood/adolescence, and the third acts subsequently. Two candidate factors (vitamin D deficiency and Epstein-Barr viral infection) seem particularly well-suited to the first two environmental-events but other factors (e.g., obesity and smoking behavior) seem also to be involved in the causal scheme. MS-pathogenesis can be modeled by incorporating both the environmental and genetic-factors into a causal scheme, which can then help to explain some of the changes in MS-epidemiology (e.g., increasing disease-prevalence, changing sex-ratio, and regional-variations in monozygotic-twin-concordance-rates), which have been taking place recently. This model suggests that genetic-susceptibility is overwhelmingly the most important determinant of MS and that, at least, 92.5% of individuals (and likely much more) are, essentially, incapable of developing MS, regardless of their specific environmental-exposures. Nevertheless, the genetics is complex and the contribution of any specific gene to MS-susceptibility seems to be quite modest. Thus, even for the DRB1*1501 allele (the strongest known MS-susceptibility marker), most carriers are not in the genetically-susceptible group. Moreover, 45-50% of individuals with MS lack this allele entirely and some of the haplotypes that carry this allele don't also confer any disease-risk. Finally, because the prevalence of genetic-susceptibility seems to be so similar throughout North America and Europe, and despite the crucial importance of a person's genetic make-up to disease pathogenesis, it is the environmental-factors, which largely responsible for the observed regional variations in disease-characteristics. Thus, despite MS being more common in women, men are more likely to be genetically-susceptible. This apparent paradox seems to relate to the fact that women are much more responsive than men to the recent changes in environmental-exposure (whatever these have been). These gender-differences may help to explain changes in the sex-ratio and the increasing disease-prevalence, which have both been observed recently. The potential importance of these conclusions regarding the role of environment in MS-pathogenesis is that they open the door to the possibility of pursuing strategies for primary primary disease prevention in the future.
Collapse
Affiliation(s)
- D S Goodin
- Multiple Sclerosis Center at the University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
247
|
Höftberger R, Leisser M, Bauer J, Lassmann H. Autoimmune encephalitis in humans: how closely does it reflect multiple sclerosis ? Acta Neuropathol Commun 2015; 3:80. [PMID: 26637427 PMCID: PMC4670499 DOI: 10.1186/s40478-015-0260-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 11/20/2015] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system. Immunological studies suggest that it is a T-cell mediated autoimmune disease, although an MS-specific target antigen for autoimmunity has so far not been identified. Models of experimental autoimmune encephalomyelitis in part reproduce features of MS, but none of the models so far covers the entire spectrum of pathology and immunology. Autoimmune disease of the nervous system has occasionally been observed in humans after active sensitization with brain tissue or brain cells, giving rise to acute demyelinating polyradiculoneuritis, acute disseminated encephalomyelitis and in rare cases reflecting an inflammatory demyelinating condition similar to acute multiple sclerosis. In this study we analyzed in detail the immunopathology in archival autopsy tissue of a patient who died with an MS like disease after repeated exposure to subcutaneous injections of lyophilized brain cells. RESULTS The pathology of this patient fulfilled all pathological diagnostic criteria of MS. Demyelination and tissue injury was associated with antibody (IgM) deposition at active lesion sites and complement activation. Major differences to classical EAE models were seen in the composition of inflammatory infiltrates, being dominated by B-cells, infiltration of IgM positive plasma cells, profound infiltration of the tissue by CD8(+) T-lymphocytes and a nearly complete absence of CD4(+) T-cells. CONCLUSIONS Our study shows that auto-sensitization of humans with brain tissue can induce a disease, which closely reflects the pathology of MS, but that the mechanisms leading to demyelination and tissue injury differ from those, generally implicated in the pathophysiology of MS through studies in experimental autoimmune encephalomyelitis.
Collapse
Affiliation(s)
- Romana Höftberger
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Marianne Leisser
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Jan Bauer
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
248
|
Veber D, Scalabrino G. Are PrPCs involved in some human myelin diseases? Relating experimental studies to human pathology. J Neurol Sci 2015; 359:396-403. [DOI: 10.1016/j.jns.2015.09.365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/04/2015] [Accepted: 09/23/2015] [Indexed: 11/29/2022]
|
249
|
Lee JY, Biemond M, Petratos S. Axonal degeneration in multiple sclerosis: defining therapeutic targets by identifying the causes of pathology. Neurodegener Dis Manag 2015; 5:527-48. [DOI: 10.2217/nmt.15.50] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Current therapeutics in multiple sclerosis (MS) target the putative inflammation and immune attack on CNS myelin. Despite their effectiveness in blunting the relapse rate in MS patients, such therapeutics do not prevent MS disease progression. Importantly, specific clinical dilemma arises through inability to predict MS progression and thereby therapeutically target axonal injury during MS, limiting permanent disability. The current review identifies immune and neurobiological principles that govern the sequelae of axonal degeneration during MS disease progression. Defining the specific disease arbiters, inflammatory and autoimmune, oligodendrocyte dystrophy and degenerative myelin, we discuss a basis for a molecular mechanism in axons that may be targeted therapeutically, in spatial and temporal manner to limit axonal degeneration and thereby halt progression of MS.
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| | - Melissa Biemond
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| |
Collapse
|
250
|
Lisak RP, Nedelkoska L, Bealmear B, Benjamins JA. Melanocortin receptor agonist ACTH 1-39 protects rat forebrain neurons from apoptotic, excitotoxic and inflammation-related damage. Exp Neurol 2015; 273:161-167. [PMID: 26300474 DOI: 10.1016/j.expneurol.2015.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/12/2015] [Accepted: 08/14/2015] [Indexed: 12/22/2022]
Abstract
Patients with relapsing-remitting multiple sclerosis (RRMS) are commonly treated with high doses of intravenous corticosteroids (CS). ACTH 1-39, a member of the melanocortin family, stimulates production of CS by the adrenals, but melanocortin receptors are also found in the central nervous system (CNS) and on immune cells. ACTH is produced within the CNS and may have direct protective effects on glia and neurons independent of CS. We previously reported that ACTH 1-39 protected oligodendroglia (OL) and their progenitors (OPC) from a panel of excitotoxic and inflammation-related agents. Neurons are the most vulnerable cells in the CNS. They are terminally differentiated, and sensitive to inflammatory and excitotoxic insults. For potential therapeutic protection of gray matter, it is important to investigate the direct effects of ACTH on neurons. Cultures highly enriched in neurons were isolated from 2-3 day old rat brain. After 4-7 days in culture, the neurons were treated for 24h with selected toxic agents with or without ACTH 1-39. ACTH 1-39 protected neurons from death induced by staurosporine, glutamate, NMDA, AMPA, kainate, quinolinic acid, reactive oxygen species and, to a modest extent, from rapidly released NO, but did not protect against kynurenic acid or slowly released nitric oxide. Our results show that ACTH 1-39 protects neurons in vitro from several apoptotic, excitotoxic and inflammation-related insults.
Collapse
Affiliation(s)
- Robert P Lisak
- Department of Neurology, Wayne State University School of Medicine, 8D University Health Center, 4201 St. Antoine St., Detroit, MI 48201, USA.
| | - Liljana Nedelkoska
- Department of Neurology, Wayne State University School of Medicine, 8D University Health Center, 4201 St. Antoine St., Detroit, MI 48201, USA
| | - Beverly Bealmear
- Department of Neurology, Wayne State University School of Medicine, 8D University Health Center, 4201 St. Antoine St., Detroit, MI 48201, USA
| | - Joyce A Benjamins
- Department of Neurology, Wayne State University School of Medicine, 8D University Health Center, 4201 St. Antoine St., Detroit, MI 48201, USA
| |
Collapse
|