51
|
Baghdadi M, Badwey ME, Khalil M, Dawoud RM. Brain magnetic resonance imaging surface-based analysis and cortical thickness measurement in relapsing remission multiple sclerosis. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2022. [DOI: 10.1186/s43055-021-00686-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Damage occurs in the brain tissue in MS which appears normal on standard conventional imaging (normal appearing brain tissue). This slow, evolving damage can be monitored by nonconventional advanced MR imaging techniques. New techniques for the measurement of cortical thickness have been validated against histological analysis and manual measurements. The aim of our study was to study the role of MRI surface-based analysis and cortical thickness measurement in the evaluation of patients with Relapsing Remitting Multiple Sclerosis and to detect if there is localized rather than generalized cortical atrophy in Multiple Sclerosis patients and correlating these findings with clinical data.
Results
30 patients and 30 healthy control were included in this study and they were subjected to cortical thickness analysis using MRI. The patients in our study showed decreased thickness of the precentral, paracentral, postcentral, posterior cingulate cortices and mean cortical thickness in both hemispheres when compared with the normal control group. Statistical analysis was significant (P value < 0.05) for the precentral, paracentral, postcentral, posterior cingulate cortices and mean cortical thickness in both hemispheres. On the other hand, statistical analysis was not significant (P value > 0.05) for other cortices. There was a significant negative correlation between the precentral, paracentral, postcentral, posterior cingulate cortices and mean cortical thickness in both hemispheres and EDSS scores with correlation coefficients ranging from − 0.9878 to − 0.7977.
Conclusions
MRI and post-processing segmentation analysis for cortical thickness is non-invasive imaging techniques that can increase the level of diagnostic confidence in diagnosis of MS patients and should be included as routine modality when evaluating patients with MS.
Collapse
|
52
|
Tranfa M, Pontillo G, Petracca M, Brunetti A, Tedeschi E, Palma G, Cocozza S. Quantitative MRI in Multiple Sclerosis: From Theory to Application. AJNR Am J Neuroradiol 2022; 43:1688-1695. [PMID: 35680161 DOI: 10.3174/ajnr.a7536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023]
Abstract
Quantitative MR imaging techniques allow evaluating different aspects of brain microstructure, providing meaningful information about the pathophysiology of damage in CNS disorders. In the study of patients with MS, quantitative MR imaging techniques represent an invaluable tool for studying changes in myelin and iron content occurring in the context of inflammatory and neurodegenerative processes. In the first section of this review, we summarize the physics behind quantitative MR imaging, here defined as relaxometry and quantitative susceptibility mapping, and describe the neurobiological correlates of quantitative MR imaging findings. In the second section, we focus on quantitative MR imaging application in MS, reporting the main findings in both the gray and white matter compartments, separately addressing macroscopically damaged and normal-appearing parenchyma.
Collapse
Affiliation(s)
- M Tranfa
- From the Departments of Advanced Biomedical Sciences (M.T., G. Pontillo, A.B., E.T., S.C.)
| | - G Pontillo
- From the Departments of Advanced Biomedical Sciences (M.T., G. Pontillo, A.B., E.T., S.C.) .,Electrical Engineering and Information Technology (G. Pontillo), University of Naples "Federico II," Naples, Italy
| | - M Petracca
- Department of Human Neurosciences (M.P.), Sapienza University of Rome, Rome, Italy
| | - A Brunetti
- From the Departments of Advanced Biomedical Sciences (M.T., G. Pontillo, A.B., E.T., S.C.)
| | - E Tedeschi
- From the Departments of Advanced Biomedical Sciences (M.T., G. Pontillo, A.B., E.T., S.C.)
| | - G Palma
- Institute of Nanotechnology (G. Palma), National Research Council, Lecce, Italy
| | - S Cocozza
- From the Departments of Advanced Biomedical Sciences (M.T., G. Pontillo, A.B., E.T., S.C.)
| |
Collapse
|
53
|
Senol AD, Pinto G, Beau M, Guillemot V, Dupree JL, Stadelmann C, Ranft J, Lubetzki C, Davenne M. Alterations of the axon initial segment in multiple sclerosis grey matter. Brain Commun 2022; 4:fcac284. [PMID: 36451656 PMCID: PMC9700164 DOI: 10.1093/braincomms/fcac284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/14/2022] [Accepted: 11/02/2022] [Indexed: 07/22/2023] Open
Abstract
Grey matter damage has been established as a key contributor to disability progression in multiple sclerosis. Aside from neuronal loss and axonal transections, which predominate in cortical demyelinated lesions, synaptic alterations have been detected in both demyelinated plaques and normal-appearing grey matter, resulting in functional neuronal damage. The axon initial segment is a key element of neuronal function, responsible for action potential initiation and maintenance of neuronal polarity. Despite several reports of profound axon initial segment alterations in different pathological models, among which experimental auto-immune encephalomyelitis, whether the axon initial segment is affected in multiple sclerosis is still unknown. Using immunohistochemistry, we analysed axon initial segments from control and multiple sclerosis tissue, focusing on layer 5/6 pyramidal neurons in the neocortex and Purkinje cells in the cerebellum and performed analysis on the parameters known to control neuronal excitability, i.e. axon initial segment length and position. We found that the axon initial segment length was increased only in pyramidal neurons of inactive demyelinated lesions, compared with normal appearing grey matter tissue. In contrast, in both cell types, the axon initial segment position was altered, with an increased soma-axon initial segment gap, in both active and inactive demyelinated lesions. In addition, using a computational model, we show that this increased gap between soma and axon initial segment might increase neuronal excitability. Taken together, these results show, for the first time, changes of axon initial segments in multiple sclerosis, in active as well as inactive grey matter lesions in both neocortex and cerebellum, which might alter neuronal function.
Collapse
Affiliation(s)
- Aysegul Dilsizoglu Senol
- Sorbonne University, Paris Brain Institute—ICM, Inserm, CNRS, Pitié-Salpêtrière Hospital, Paris, France
| | - Giulia Pinto
- Sorbonne University, Paris Brain Institute—ICM, Inserm, CNRS, Pitié-Salpêtrière Hospital, Paris, France
| | - Maxime Beau
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, Inserm, PSL Research University, Paris, France
| | - Vincent Guillemot
- Sorbonne University, Paris Brain Institute—ICM, Inserm, CNRS, Pitié-Salpêtrière Hospital, Paris, France
- Institut Pasteur, Université de Paris, Bioinformatics and Biostatistics Hub, Paris F-75015, France
| | - Jeffrey L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
- Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Jonas Ranft
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, Inserm, PSL Research University, Paris, France
| | - Catherine Lubetzki
- Sorbonne University, Paris Brain Institute—ICM, Inserm, CNRS, Pitié-Salpêtrière Hospital, Paris, France
- Assistance Publique des Hôpitaux de Paris (APHP), Pitié-Salpêtrière Hospital, DMU Neurosciences, Paris, France
| | - Marc Davenne
- Correspondence to: Dr Marc Davenne Paris Brain Institute, Pitié-Salpêtrière Hospital 47, bd de l’hôpital, F-75013 Paris, France E-mail:
| |
Collapse
|
54
|
Gharagozloo M, Mace JW, Calabresi PA. Animal models to investigate the effects of inflammation on remyelination in multiple sclerosis. Front Mol Neurosci 2022; 15:995477. [PMID: 36407761 PMCID: PMC9669474 DOI: 10.3389/fnmol.2022.995477] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/22/2022] [Indexed: 09/19/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, demyelinating, and neurodegenerative disease of the central nervous system (CNS). In people with MS, impaired remyelination and axonal loss lead to debilitating long-term neurologic deficits. Current MS disease-modifying drugs mainly target peripheral immune cells and have demonstrated little efficacy for neuroprotection or promoting repair. To elucidate the pathological mechanisms and test therapeutic interventions, multiple animal models have been developed to recapitulate specific aspects of MS pathology, particularly the acute inflammatory stage. However, there are few animal models that facilitate the study of remyelination in the presence of inflammation, and none fully replicate the biology of chronic demyelination in MS. In this review, we describe the animal models that have provided insight into the mechanisms underlying demyelination, myelin repair, and potential therapeutic targets for remyelination. We highlight the limitations of studying remyelination in toxin-based demyelination models and discuss the combinatorial models that recapitulate the inflammatory microenvironment, which is now recognized to be a major inhibitor of remyelination mechanisms. These models may be useful in identifying novel therapeutics that promote CNS remyelination in inflammatory diseases such as MS.
Collapse
Affiliation(s)
- Marjan Gharagozloo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jackson W. Mace
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
55
|
Pitt D, Lo CH, Gauthier SA, Hickman RA, Longbrake E, Airas LM, Mao-Draayer Y, Riley C, De Jager PL, Wesley S, Boster A, Topalli I, Bagnato F, Mansoor M, Stuve O, Kister I, Pelletier D, Stathopoulos P, Dutta R, Lincoln MR. Toward Precision Phenotyping of Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/6/e200025. [PMID: 36041861 PMCID: PMC9427000 DOI: 10.1212/nxi.0000000000200025] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 02/07/2022] [Indexed: 11/15/2022]
Abstract
The classification of multiple sclerosis (MS) has been established by Lublin in 1996 and revised in 2013. The revision includes clinically isolated syndrome, relapsing-remitting, primary progressive and secondary progressive MS, and has added activity (i.e., formation of white matter lesions or clinical relapses) as a qualifier. This allows for the distinction between active and nonactive progression, which has been shown to be of clinical importance. We propose that a logical extension of this classification is the incorporation of additional key pathological processes, such as chronic perilesional inflammation, neuroaxonal degeneration, and remyelination. This will distinguish MS phenotypes that may present as clinically identical but are driven by different combinations of pathological processes. A more precise description of MS phenotypes will improve prognostication and personalized care as well as clinical trial design. Thus, our proposal provides an expanded framework for conceptualizing MS and for guiding development of biomarkers for monitoring activity along the main pathological axes in MS.
Collapse
Affiliation(s)
- David Pitt
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada.
| | - Chih Hung Lo
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Susan A Gauthier
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Richard A Hickman
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Erin Longbrake
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Laura M Airas
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Yang Mao-Draayer
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Claire Riley
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Philip Lawrence De Jager
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Sarah Wesley
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Aaron Boster
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Ilir Topalli
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Francesca Bagnato
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Mohammad Mansoor
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Olaf Stuve
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Ilya Kister
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Daniel Pelletier
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Panos Stathopoulos
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Ranjan Dutta
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| | - Matthew R Lincoln
- From the Yale University (David Pitt, C.H.L., E.L., M.M., M.R.L.), New Haven; Nanyang Technological University (C.H.L.), Singapore; Weill Cornell Medicine (S.A.G.), New York; Memorial Sloan Kettering Cancer Center (R.A.H.), New York; University of Turku (L.M.A.), Finland; University of Michigan Medical School (Y.M.-D.), Ann Arbor; Columbia University Medical Center (C.R., P.L.D.J., S.W.), New York; The Boster Center for Multiple Sclerosis (A.B.), Columbus, OH; Cerneris Inc (I.T.), Wilmington, DE; Vanderbilt University Medical Center (F.B.), Nashville, TN; University of Texas Southwestern Medical Center (O.S.), Dallas; NYU Langone Medical Center (I.K.), New York; University of Southern California (Daniel Pelletier), Los Angeles; National and Kapodistrian University of Athens Medical School (P.S.), Greece; Cleveland Clinic Lerner College of Medicine (R.D.), Case Western Reserve University, OH; and University of Toronto and St. Michael's Hospital (M.L.), ON, Canada
| |
Collapse
|
56
|
Voskuhl RR, MacKenzie-Graham A. Chronic experimental autoimmune encephalomyelitis is an excellent model to study neuroaxonal degeneration in multiple sclerosis. Front Mol Neurosci 2022; 15:1024058. [PMID: 36340686 PMCID: PMC9629273 DOI: 10.3389/fnmol.2022.1024058] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 09/30/2022] [Indexed: 08/19/2023] Open
Abstract
Animal models of multiple sclerosis (MS), specifically experimental autoimmune encephalomyelitis (EAE), have been used extensively to develop anti-inflammatory treatments. However, the similarity between MS and one particular EAE model does not end at inflammation. MS and chronic EAE induced in C57BL/6 mice using myelin oligodendrocyte glycoprotein (MOG) peptide 35-55 share many neuropathologies. Beyond both having white matter lesions in spinal cord, both also have widespread neuropathology in the cerebral cortex, hippocampus, thalamus, striatum, cerebellum, and retina/optic nerve. In this review, we compare neuropathologies in each of these structures in MS with chronic EAE in C57BL/6 mice, and find evidence that this EAE model is well suited to study neuroaxonal degeneration in MS.
Collapse
Affiliation(s)
- Rhonda R. Voskuhl
- UCLA MS Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | | |
Collapse
|
57
|
Saez-Calveras N, Brewster AL, Stuve O. The validity of animal models to explore the pathogenic role of the complement system in multiple sclerosis: A review. Front Mol Neurosci 2022; 15:1017484. [PMID: 36311030 PMCID: PMC9606595 DOI: 10.3389/fnmol.2022.1017484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Animal models of multiple sclerosis (MS) have been extensively used to characterize the disease mechanisms in MS, as well as to identify potential pharmacologic targets for this condition. In recent years, the immune complement system has gained increased attention as an important effector in the pathogenesis of MS. Evidence from histological, serum, and CSF studies of patients supports an involvement of complement in both relapsing-remitting and progressive MS. In this review, we discuss the history and advances made on the use of MS animal models to profile the effects of the complement system in this condition. The first studies that explored the complement system in the context of MS used cobra venom factor (CVF) as a complement depleting agent in experimental autoimmune encephalomyelitis (EAE) Lewis rats. Since then, multiple mice and rat models of MS have revealed a role of C3 and the alternative complement cascade in the opsonization and phagocytosis of myelin by microglia and myeloid cells. Studies using viral vectors, genetic knockouts and pharmacologic complement inhibitors have also shown an effect of complement in synaptic loss. Antibody-mediated EAE models have revealed an involvement of the C1 complex and the classical complement as an effector of the humoral response in this disease. C1q itself may also be involved in modulating microglia activation and oligodendrocyte differentiation in these animals. In addition, animal and in vitro models have revealed that multiple complement factors may act as modulators of both the innate and adaptive immune responses. Finally, evidence gathered from mice models suggests that the membrane attack complex (MAC) may even exert protective roles in the chronic stages of EAE. Overall, this review summarizes the importance of MS animal models to better characterize the role of the complement system and guide future therapeutic approaches in this condition.
Collapse
Affiliation(s)
- Nil Saez-Calveras
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, Parkland Hospital, Dallas, TX, United States
| | - Amy L. Brewster
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, VA North Texas Health Care System, Dallas, TX, United States
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
- *Correspondence: Olaf Stuve,
| |
Collapse
|
58
|
Cordano C, Nourbakhsh B, Yiu HH, Papinutto N, Caverzasi E, Abdelhak A, Oertel FC, Beaudry-Richard A, Santaniello A, Sacco S, Bennett DJ, Gomez A, Sigurdson CJ, Hauser SL, Magliozzi R, Cree BA, Henry RG, Green AJ. Differences in Age-related Retinal and Cortical Atrophy Rates in Multiple Sclerosis. Neurology 2022; 99:e1685-e1693. [PMID: 36038272 PMCID: PMC9559941 DOI: 10.1212/wnl.0000000000200977] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/01/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The timing of neurodegeneration in multiple sclerosis (MS) remains unclear. It is critical to understand the dynamics of neuroaxonal loss if we hope to prevent or forestall permanent disability in MS. We therefore used a deeply phenotyped longitudinal cohort to assess and compare rates of neurodegeneration in retina and brain throughout the MS disease course. METHODS We analyzed 597 patients with MS who underwent longitudinal optical coherence tomography imaging annually for 4.5 ± 2.4 years and 432 patients who underwent longitudinal MRI scans for 10 ± 3.4 years, quantifying macular ganglion cell-inner plexiform layer (GCIPL) volume and cortical gray matter (CGM) volume. The association between the slope of decline in the anatomical structure and the age of entry in the cohort (categorized by the MRI cohort's age quartiles) was assessed by hierarchical linear models. RESULTS The rate of CGM volume loss declined with increasing age of study entry (1.3% per year atrophy for the age of entry in the cohort younger than 35 years; 1.1% for older than 35 years and younger than 41; 0.97% for older than 41 years and younger than 49; 0.9% for older than 49 years) while the rate of GCIPL thinning was highest in patients in the youngest quartile, fell by more than 50% in the following age quartile, and then stabilized (0.7% per year thinning for the age of entry in the cohort younger than 35 years; 0.29% for age older than 35 and younger than 41 years; 0.34% for older than 41 and younger than 49 years; 0.33% for age older than 49 years). DISCUSSION An age-dependent reduction in retinal and cortical volume loss rates during relapsing-remitting MS suggests deceleration in neurodegeneration in the earlier period of disease and further indicates that the period of greatest adaptive immune-mediated inflammatory activity is also the period with the greatest neuroaxonal loss.
Collapse
Affiliation(s)
- Christian Cordano
- From the Department of Neurology (C.C., N.P., E.C., A.A., F.C.O., A.B.-R., A.S., S.S., D.J.B., A.G., S.L.H., B.A.C.C., R.G.H., A.J.G.), UCSF Weill Institute for Neurosciences, University of California, San Francisco; Department of Neurology (B.N.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biology (H.H.Y.), University of Maryland, College Park; Department of Pathology (C.J.S.), University of California, San Diego, La Jolla; and Department of Neurosciences (R.M.), Biomedicine and Movement Sciences, University of Verona, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Magliozzi R, Fadda G, Brown RA, Bar‐Or A, Howell OW, Hametner S, Marastoni D, Poli A, Nicholas R, Calabrese M, Monaco S, Reynolds R. "Ependymal-in" Gradient of Thalamic Damage in Progressive Multiple Sclerosis. Ann Neurol 2022; 92:670-685. [PMID: 35748636 PMCID: PMC9796378 DOI: 10.1002/ana.26448] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 01/01/2023]
Abstract
Leptomeningeal and perivenular infiltrates are important contributors to cortical grey matter damage and disease progression in multiple sclerosis (MS). Whereas perivenular inflammation induces vasculocentric lesions, leptomeningeal involvement follows a subpial "surface-in" gradient. To determine whether similar gradient of damage occurs in deep grey matter nuclei, we examined the dorsomedial thalamic nuclei and cerebrospinal fluid (CSF) samples from 41 postmortem secondary progressive MS cases compared with 5 non-neurological controls and 12 controls with other neurological diseases. CSF/ependyma-oriented gradient of reduction in NeuN+ neuron density was present in MS thalamic lesions compared to controls, greatest (26%) in subventricular locations at the ependyma/CSF boundary and least with increasing distance (12% at 10 mm). Concomitant graded reduction in SMI31+ axon density was observed, greatest (38%) at 2 mm from the ependyma/CSF boundary and least at 10 mm (13%). Conversely, gradient of major histocompatibility complex (MHC)-II+ microglia density increased by over 50% at 2 mm at the ependyma/CSF boundary and only by 15% at 10 mm and this gradient inversely correlated with the neuronal (R = -0.91, p < 0.0001) and axonal (R = -0.79, p < 0.0001) thalamic changes. Observed gradients were also detected in normal-appearing thalamus and were associated with rapid/severe disease progression; presence of leptomeningeal tertiary lymphoid-like structures; large subependymal infiltrates, enriched in CD20+ B cells and occasionally containing CXCL13+ CD35+ follicular dendritic cells; and high CSF protein expression of a complex pattern of soluble inflammatory/neurodegeneration factors, including chitinase-3-like-1, TNFR1, parvalbumin, neurofilament-light-chains and TNF. Substantial "ependymal-in" gradient of pathological cell alterations, accompanied by presence of intrathecal inflammation, compartmentalized either in subependymal lymphoid perivascular infiltrates or in CSF, may play a key role in MS progression. SUMMARY FOR SOCIAL MEDIA: Imaging and neuropathological evidences demonstrated the unique feature of "surface-in" gradient of damage in multiple sclerosis (MS) since early pediatric stages, often associated with more severe brain atrophy and disease progression. In particular, increased inflammation in the cerebral meninges has been shown to be strictly associated with an MS-specific gradient of neuronal, astrocyte, and oligodendrocyte loss accompanied by microglial activation in subpial cortical layers, which is not directly related to demyelination. To determine whether a similar gradient of damage occurs in deep grey matter nuclei, we examined the potential neuronal and microglia alterations in the dorsomedial thalamic nuclei from postmortem secondary progressive MS cases in combination with detailed neuropathological characterization of the inflammatory features and protein profiling of paired CSF samples. We observed a substantial "subependymal-in" gradient of neuro-axonal loss and microglia activation in active thalamic lesions of progressive MS cases, in particular in the presence of increased leptomeningeal and cerebrospinal fluid (CSF) inflammation. This altered graded pathology was found associated with more severe and rapid progressive MS and increased inflammatory degree either in large perivascular subependymal infiltrates, enriched in B cells, or within the paired CSF, in particular with elevated levels of a complex pattern of soluble inflammatory and neurodegeneration factors, including chitinase 3-like-1, TNFR1, parvalbumin, neurofilament light-chains and TNF. These data support a key role for chronic, intrathecally compartmentalized inflammation in specific disease endophenotypes. CSF biomarkers, together with advance imaging tools, may therefore help to improve not only the disease diagnosis but also the early identification of specific MS subgroups that would benefit of more personalized treatments. ANN NEUROL 2022;92:670-685.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology Section of Department of Neurological and Movement SciencesUniversity of VeronaVeronaItaly,Department of Brain Sciences, Faculty of MedicineImperial College LondonLondonUK
| | - Giulia Fadda
- Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | | | - Amit Bar‐Or
- Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Owain W. Howell
- Department of Brain Sciences, Faculty of MedicineImperial College LondonLondonUK,Institute of Life SciencesSwansea UniversitySwanseaUK
| | - Simon Hametner
- Brain Research CenterMedical University of ViennaViennaAustria
| | - Damiano Marastoni
- Neurology Section of Department of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
| | - Alberto Poli
- Neurology Section of Department of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
| | - Richard Nicholas
- Department of Brain Sciences, Faculty of MedicineImperial College LondonLondonUK
| | - Massimiliano Calabrese
- Neurology Section of Department of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
| | - Salvatore Monaco
- Neurology Section of Department of Neurological and Movement SciencesUniversity of VeronaVeronaItaly
| | - Richard Reynolds
- Department of Brain Sciences, Faculty of MedicineImperial College LondonLondonUK,Centre for Molecular Neuropathology, Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
60
|
Plastini MJ, Desu HL, Ascona MC, Lang AL, Saporta MA, Brambilla R. Transcriptional abnormalities in induced pluripotent stem cell-derived oligodendrocytes of individuals with primary progressive multiple sclerosis. Front Cell Neurosci 2022; 16:972144. [PMID: 36246526 PMCID: PMC9554611 DOI: 10.3389/fncel.2022.972144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is the most common neurological disorder in young adults and is classically defined as a chronic inflammatory demyelinating disease of the central nervous system (CNS). Although MS affects millions of people worldwide, its underlying cause remains unknown making discovery of effective treatments challenging. Whether intrinsic or extrinsic factors contribute to MS initiation and progression is still unclear. This is especially true for primary progressive MS (PPMS), the rarest form of the disease, in which progressive and irreversible loss of neurological function is often observed in the absence of an overt immune-inflammatory response. To test the hypothesis that intrinsic dysfunction in oligodendrocytes (OLs), the primary targets of damage in MS, may contribute to PPMS etiopathology, we differentiated human induced pluripotent stem cell (hiPSC) lines derived from PPMS and healthy individuals into mature OLs to compare their transcriptional profile. PPMS derived OLs displayed hundreds of differentially expressed genes compared to control OLs, many associated with cell adhesion, apoptosis and inflammation, including the inflammasome component Nlrp2, which was highly upregulated. NLRP2 immunoreactivity in OLs was confirmed in post-mortem PPMS brain tissues, with higher expression than in control tissues. Altogether, our findings suggest that mature OLs in PPMS affected individuals carry intrinsic abnormalities that could contribute, at least in part, to the pathophysiology of this form of the disease.
Collapse
Affiliation(s)
- Melanie J. Plastini
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- The Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Haritha L. Desu
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- The Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Maureen C. Ascona
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- The Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anna L. Lang
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mario A. Saporta
- The Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Roberta Brambilla
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- The Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE-Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- *Correspondence: Roberta Brambilla,
| |
Collapse
|
61
|
Kolb H, Al-Louzi O, Beck ES, Sati P, Absinta M, Reich DS. From pathology to MRI and back: Clinically relevant biomarkers of multiple sclerosis lesions. Neuroimage Clin 2022; 36:103194. [PMID: 36170753 PMCID: PMC9668624 DOI: 10.1016/j.nicl.2022.103194] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Focal lesions in both white and gray matter are characteristic of multiple sclerosis (MS). Histopathological studies have helped define the main underlying pathological processes involved in lesion formation and evolution, serving as a gold standard for many years. However, histopathology suffers from an intrinsic bias resulting from over-reliance on tissue samples from late stages of the disease or atypical cases and is inadequate for routine patient assessment. Pathological-radiological correlative studies have established advanced MRI's sensitivity to several relevant MS-pathological substrates and its practicality for assessing dynamic changes and following lesions over time. This review focuses on novel imaging techniques that serve as biomarkers of critical pathological substrates of MS lesions: the central vein, chronic inflammation, remyelination and repair, and cortical lesions. For each pathological process, we address the correlative value of MRI to MS pathology, its contribution in elucidating MS pathology in vivo, and the clinical utility of the imaging biomarker.
Collapse
Affiliation(s)
- Hadar Kolb
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA,Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv-Yaffo, Israel,Corresponding author at: Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv-Yaffo, Israel.
| | - Omar Al-Louzi
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA,Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Erin S. Beck
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pascal Sati
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA,Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Martina Absinta
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA,Institute of Experimental Neurology (INSPE), IRCSS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy,Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Daniel S. Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
62
|
Jin L, Zhang J, Hua X, Xu X, Li J, Wang J, Wang M, Liu H, Qiu H, Chen M, Zhang X, Wang Y, Huang Z. Astrocytic SARM1 promotes neuroinflammation and axonal demyelination in experimental autoimmune encephalomyelitis through inhibiting GDNF signaling. Cell Death Dis 2022; 13:759. [PMID: 36055989 PMCID: PMC9440144 DOI: 10.1038/s41419-022-05202-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 01/21/2023]
Abstract
Astrocytes are important components of the innate immune response in the central nervous system (CNS), involving in the inflammatory and neurotoxic responses that occur in CNS diseases, such as multiple sclerosis (MS). Recent studies have shown that SARM1 plays a critical role in axonal degeneration and inflammation. However, the detailed role of astrocytic SARM1 in MS remains unclear. Here, we established the MS model of mice - experimental autoimmune encephalomyelitis (EAE) and found that SARM1 was upregulated in astrocytes of the spinal cords of EAE mice. Moreover, conditional knockout of astrocytic SARM1 (SARM1GFAP-CKO mice, SARM1Aldh1L1-CKO mice) delayed EAE with later onset, alleviated the inflammatory infiltration, and inhibited the demyelination and neuronal death. Mechanically, RNA-seq revealed that the expression of glial-derived neurotrophic factor (GDNF) was upregulated in SARM1-/- astrocytes. Western blot and immunostaining further confirmed the upregulation of GDNF in spinal cord astrocytes of SARM1GFAP-CKO EAE mice. Interestingly, the downregulation of GDNF by streptozotocin (STZ, a drug used to downregulate GDNF) treatment worsened the deficits of SARM1GFAP-CKO EAE mice. These findings identify that astrocytic SARM1 promotes neuroinflammation and axonal demyelination in EAE by inhibiting the expression of GDNF, reveal the novel role of SARM1/GDNF signaling in EAE, and provide new therapeutic ideas for the treatment of MS.
Collapse
Affiliation(s)
- Lingting Jin
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- School of Pharmacy, and Department of Neurosurgery of the Affiliated Hospital,, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Jingjing Zhang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xin Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xingxing Xu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jia Li
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiaojiao Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Mianxian Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Huitao Liu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Haoyu Qiu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Man Chen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xu Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Ying Wang
- Clinical Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China.
| | - Zhihui Huang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- School of Pharmacy, and Department of Neurosurgery of the Affiliated Hospital,, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
63
|
Balashov AV, Pankov VG, Balashov VP, Shikhanov NP, Gushchina SV. Changes in the Neuronal Population of the Spinal Cord of Mice with Experimental Autoimmune Encephalomyelitis as a Model of Multiple Sclerosis. Bull Exp Biol Med 2022; 173:590-593. [PMID: 36210424 DOI: 10.1007/s10517-022-05593-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Indexed: 06/16/2023]
Abstract
We performed a quantitative study of the neuronal population in the spinal cord of mice with acute and chronic model of experimental autoimmune encephalomyelitis. Immunohistological/immunofluorescent analysis with motor neuron marker ChAT revealed a significant decrease in the number of motor neurons in the ventral horns of the lumbar spinal cord in the acute form of autoimmune encephalomyelitis, with the further appearance of large empty (containing no motor neurons) areas in the ventral horns in the chronic form. The development of experimental autoimmune encephalomyelitis is accompanied by degradation and death of neurons, in particular ChAT+ motor neurons.
Collapse
Affiliation(s)
- A V Balashov
- Department of Cytology, Histology, and Embryology with Courses in Medical Biology and Molecular Biology of the Cell, N. P. Ogarev National Research Mordovia State University, Saransk, Republic of Mordovia, Russia.
| | - V G Pankov
- Department of Cytology, Histology, and Embryology with Courses in Medical Biology and Molecular Biology of the Cell, N. P. Ogarev National Research Mordovia State University, Saransk, Republic of Mordovia, Russia
| | - V P Balashov
- Department of Cytology, Histology, and Embryology with Courses in Medical Biology and Molecular Biology of the Cell, N. P. Ogarev National Research Mordovia State University, Saransk, Republic of Mordovia, Russia
| | - N P Shikhanov
- Department of Normal and Pathological Anatomy with a Course of Forensic Medicine, N. P. Ogarev National Research Mordovia State University, Saransk, Republic of Mordovia, Russia
| | - S V Gushchina
- Department of Cytology, Histology, and Embryology with Courses in Medical Biology and Molecular Biology of the Cell, N. P. Ogarev National Research Mordovia State University, Saransk, Republic of Mordovia, Russia
| |
Collapse
|
64
|
Müller J, La Rosa F, Beaumont J, Tsagkas C, Rahmanzadeh R, Weigel M, Bach Cuadra M, Gambarota G, Granziera C. Fluid and White Matter Suppression: New Sensitive 3 T Magnetic Resonance Imaging Contrasts for Cortical Lesion Detection in Multiple Sclerosis. Invest Radiol 2022; 57:592-600. [PMID: 35510874 PMCID: PMC10184808 DOI: 10.1097/rli.0000000000000877] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/26/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Cortical lesions are common in multiple sclerosis (MS), but their visualization is challenging on conventional magnetic resonance imaging. The uniform image derived from magnetization prepared 2 rapid acquisition gradient echoes (MP2RAGE uni ) detects cortical lesions with a similar rate as the criterion standard sequence, double inversion recovery. Fluid and white matter suppression (FLAWS) provides multiple reconstructed contrasts acquired during a single acquisition. These contrasts include FLAWS minimum image (FLAWS min ), which provides an exquisite sensitivity to the gray matter signal and therefore may facilitate cortical lesion identification, as well as high contrast FLAWS (FLAWS hco ), which gives a contrast that is similar to one of MP2RAGE uni . In this study, we compared the manual detection rate of cortical lesions on MP2RAGE uni , FLAWS min , and FLAWS hco in MS patients. Furthermore, we assessed whether the combined detection rate on FLAWS min and FLAWS hco was superior to MP2RAGE uni for cortical lesions identification. Last, we compared quantitative T1 maps (qT1) provided by both MP2RAGE and FLAWS in MS lesions. MATERIALS AND METHODS We included 30 relapsing-remitting MS patients who underwent MP2RAGE and FLAWS magnetic resonance imaging with isotropic spatial resolution of 1 mm at 3 T. Cortical lesions were manually segmented by consensus of 3 trained raters and classified as intracortical or leukocortical lesions on (1) MP2RAGE uniform/flat images, (2) FLAWS min , and (3) FLAWS hco . In addition, segmented lesions on FLAWS min and FLAWS hco were merged to produce a union lesion map (FLAWS min + hco ). Number and volume of all cortical, intracortical, and leukocortical lesions were compared among MP2RAGE uni , FLAWS min , and FLAWS hco using Friedman test and between MP2RAGE uni and FLAWS min + hco using Wilcoxon signed rank test. The FLAWS T1 maps were then compared with the reference MP2RAGE T1 maps using relative differences in percentage. In an exploratory analysis, individual cortical lesion counts of the 3 raters were compared, and interrater variability was quantified using Fleiss ϰ. RESULTS In total, 633 segmentations were made on the 3 contrasts, corresponding to 355 cortical lesions. The median number and volume of single cortical, intracortical, and leukocortical lesions were comparable among MP2RAGE uni , FLAWS min , and FLAWS hco . In patients with cortical lesions (22/30), median cumulative lesion volume was larger on FLAWS min (587 μL; IQR, 1405 μL) than on MP2RAGE uni (490 μL; IQR, 990 μL; P = 0.04), whereas there was no difference between FLAWS min and FLAWS hco , or FLAWS hco and MP2RAGE uni . FLAWS min + hco showed significantly greater numbers of cortical (median, 4.5; IQR, 15) and leukocortical (median, 3.5; IQR, 12) lesions than MP2RAGE uni (median, 3; IQR, 10; median, 2.5; IQR, 7; both P < 0.001). Interrater agreement was moderate on MP2RAGE uni (ϰ = 0.582) and FLAWS hco (ϰ = 0.584), but substantial on FLAWS min (ϰ = 0.614). qT1 in lesions was similar between MP2RAGE and FLAWS. CONCLUSIONS Cortical lesions identification in FLAWS min and FLAWS hco was comparable to MP2RAGE uni . The combination of FLAWS min and FLAWS hco allowed to identify a higher number of cortical lesions than MP2RAGE uni , whereas qT1 maps did not differ between the 2 acquisition schemes.
Collapse
Affiliation(s)
- Jannis Müller
- From the Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel
| | - Francesco La Rosa
- Signal Processing Laboratory (LTS5), École Polytechnique Fédérale de Lausanne
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
| | - Jeremy Beaumont
- Univ Rennes, Inserm, LTSI-UMR1099, Rennes, France
- The Australian e-Health Research Centre, CSIRO, Brisbane, Australia
| | - Charidimos Tsagkas
- From the Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel
| | - Reza Rahmanzadeh
- From the Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel
| | - Matthias Weigel
- From the Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel
- Division of Radiological Physics, Department of Radiology, University Hospital Basel, Basel
| | - Meritxell Bach Cuadra
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Radiology Department, Lausanne University and University Hospital, Lausanne, Switzerland
| | | | - Cristina Granziera
- From the Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel
| |
Collapse
|
65
|
Errede M, Annese T, Petrosino V, Longo G, Girolamo F, de Trizio I, d'Amati A, Uccelli A, Kerlero de Rosbo N, Virgintino D. Microglia-derived CCL2 has a prime role in neocortex neuroinflammation. Fluids Barriers CNS 2022; 19:68. [PMID: 36042496 PMCID: PMC9429625 DOI: 10.1186/s12987-022-00365-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/03/2022] [Indexed: 11/12/2022] Open
Abstract
Background In myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE), several areas of demyelination are detectable in mouse cerebral cortex, where neuroinflammation events are associated with scarce inflammatory infiltrates and blood–brain barrier (BBB) impairment. In this condition, the administration of mesenchymal stem cells (MSCs) controls neuroinflammation, attenuating astrogliosis and promoting the acquisition of stem cell traits by astrocytes. To contribute to the understanding of the mechanisms involved in the pathogenesis of EAE in gray matter and in the reverting effects of MSC treatment, the neocortex of EAE-affected mice was investigated by analyzing the cellular source(s) of chemokine CCL2, a molecule involved in immune cell recruitment and BBB-microvessel leakage. Methods The study was carried out by immunohistochemistry (IHC) and dual RNAscope IHC/in situ hybridization methods, using astrocyte, NG2-glia, macrophage/microglia, and microglia elective markers combined with CCL2. Results The results showed that in EAE-affected mice, hypertrophic microglia are the primary source of CCL2, surround the cortex neurons and the damaged BBB microvessels. In EAE-affected mice treated with MSCs, microgliosis appeared diminished very soon (6 h) after treatment, an observation that was long-lasting (tested after 10 days). This was associated with a reduced CCL2 expression and with apparently preserved/restored BBB features. In conclusion, the hallmark of EAE in the mouse neocortex is a condition of microgliosis characterized by high levels of CCL2 expression. Conclusions This finding supports relevant pathogenetic and clinical aspects of the human disease, while the demonstrated early control of neuroinflammation and BBB permeability exerted by treatment with MSCs may have important therapeutic implications. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00365-5.
Collapse
Affiliation(s)
- Mariella Errede
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy.,Department of Medicine and Surgery, LUM University, Casamassima Bari, Italy
| | - Valentina Petrosino
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Giovanna Longo
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Francesco Girolamo
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Ignazio de Trizio
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy.,Department of Emergency and Organ Transplantation, Pathology Section, University of Bari School of Medicine, Bari, Italy
| | - Antonio Uccelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nicole Kerlero de Rosbo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,TomaLab, Institute of Nanotechnology, Consiglio Nazionale Delle Ricerche (CNR), Rome, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy.
| |
Collapse
|
66
|
Saez-Calveras N, Stuve O. The role of the complement system in Multiple Sclerosis: A review. Front Immunol 2022; 13:970486. [PMID: 36032156 PMCID: PMC9399629 DOI: 10.3389/fimmu.2022.970486] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
The complement system has been involved in the pathogenesis of multiple neuroinflammatory and neurodegenerative conditions. In this review, we evaluated the possible role of complement activation in multiple sclerosis (MS) with a focus in progressive MS, where the disease pathogenesis remains to be fully elucidated and treatment options are limited. The evidence for the involvement of the complement system in the white matter plaques and gray matter lesions of MS stems from immunohistochemical analysis of post-mortem MS brains, in vivo serum and cerebrospinal fluid biomarker studies, and animal models of Experimental Autoimmune Encephalomyelitis (EAE). Complement knock-out studies in these animal models have revealed that this system may have a “double-edge sword” effect in MS. On the one hand, complement proteins may aid in promoting the clearance of myelin degradation products and other debris through myeloid cell-mediated phagocytosis. On the other, its aberrant activation may lead to demyelination at the rim of progressive MS white matter lesions as well as synapse loss in the gray matter. The complement system may also interact with known risk factors of MS, including as Epstein Barr Virus (EBV) infection, and perpetuate the activation of CNS self-reactive B cell populations. With the mounting evidence for the involvement of complement in MS, the development of complement modulating therapies for this condition is appealing. Herein, we also reviewed the pharmacological complement inhibitors that have been tested in MS animal models as well as in clinical trials for other neurologic diseases. The potential use of these agents, such as the C5-binding antibody eculizumab in MS will require a detailed understanding of the role of the different complement effectors in this disease and the development of better CNS delivery strategies for these compounds.
Collapse
Affiliation(s)
- Nil Saez-Calveras
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, Veterans Affairs (VA) North Texas Health Care System, Dallas, TX, United States
- *Correspondence: Olaf Stuve,
| |
Collapse
|
67
|
Abstract
PURPOSE OF REVIEW This article provides an update on progressive forms of multiple sclerosis (MS) commonly referred to as primary progressive MS and secondary progressive MS. It discusses the importance of diagnosing and detecting progression early, the similarities between progressive forms, challenges in detecting progression, factors that could augment progression, and the importance of disease-modifying therapies in patients with evidence of active progressive MS. It also discusses the overall care of progressive MS. RECENT FINDINGS The pathogenesis of primary progressive MS and secondary progressive MS is overlapping, and in both presentations, patients with relapses or focal MRI activity are classified as having active, progressive MS. All currently approved disease-modifying therapies are indicated for active secondary progressive MS. The therapeutic opportunity of anti-inflammatory drugs for the treatment of progressive MS is enhanced in those who are younger and have a shorter disease duration. Vascular comorbidities may contribute to progression in MS. SUMMARY Several challenges remain in the diagnosis, follow-up, and treatment of progressive MS. Early identification of active progressive MS is needed to maximize treatment benefit. The advantages of optimal comorbidity management (eg, hypertension, hyperlipidemia) in delaying progression are uncertain. Clinical care guidelines for advanced, severe MS are lacking.
Collapse
|
68
|
van Schaik PEM, Zuhorn IS, Baron W. Targeting Fibronectin to Overcome Remyelination Failure in Multiple Sclerosis: The Need for Brain- and Lesion-Targeted Drug Delivery. Int J Mol Sci 2022; 23:8418. [PMID: 35955549 PMCID: PMC9368816 DOI: 10.3390/ijms23158418] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is a neuroinflammatory and neurodegenerative disease with unknown etiology that can be characterized by the presence of demyelinated lesions. Prevailing treatment protocols in MS rely on the modulation of the inflammatory process but do not impact disease progression. Remyelination is an essential factor for both axonal survival and functional neurological recovery but is often insufficient. The extracellular matrix protein fibronectin contributes to the inhibitory environment created in MS lesions and likely plays a causative role in remyelination failure. The presence of the blood-brain barrier (BBB) hinders the delivery of remyelination therapeutics to lesions. Therefore, therapeutic interventions to normalize the pathogenic MS lesion environment need to be able to cross the BBB. In this review, we outline the multifaceted roles of fibronectin in MS pathogenesis and discuss promising therapeutic targets and agents to overcome fibronectin-mediated inhibition of remyelination. In addition, to pave the way for clinical use, we reflect on opportunities to deliver MS therapeutics to lesions through the utilization of nanomedicine and discuss strategies to deliver fibronectin-directed therapeutics across the BBB. The use of well-designed nanocarriers with appropriate surface functionalization to cross the BBB and target the lesion sites is recommended.
Collapse
Affiliation(s)
- Pauline E. M. van Schaik
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| | - Inge S. Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Wia Baron
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| |
Collapse
|
69
|
The mTOR Signaling Pathway in Multiple Sclerosis; from Animal Models to Human Data. Int J Mol Sci 2022; 23:ijms23158077. [PMID: 35897651 PMCID: PMC9332053 DOI: 10.3390/ijms23158077] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
This article recapitulates the evidence on the role of mammalian targets of rapamycin (mTOR) complex pathways in multiple sclerosis (MS). Key biological processes that intersect with mTOR signaling cascades include autophagy, inflammasome activation, innate (e.g., microglial) and adaptive (B and T cell) immune responses, and axonal and neuronal toxicity/degeneration. There is robust evidence that mTOR inhibitors, such as rapamycin, ameliorate the clinical course of the animal model of MS, experimental autoimmune encephalomyelitis (EAE). New, evolving data unravel mechanisms underlying the therapeutic effect on EAE, which include balance among T-effector and T-regulatory cells, and mTOR effects on myeloid cell function, polarization, and antigen presentation, with relevance to MS pathogenesis. Radiologic and preliminary clinical data from a phase 2 randomized, controlled trial of temsirolimus (a rapamycin analogue) in MS show moderate efficacy, with significant adverse effects. Large clinical trials of indirect mTOR inhibitors (metformin) in MS are lacking; however, a smaller prospective, non-randomized study shows some potentially promising radiological results in combination with ex vivo beneficial effects on immune cells that might warrant further investigation. Importantly, the study of mTOR pathway contributions to autoimmune inflammatory demyelination and multiple sclerosis illustrates the difficulties in the clinical application of animal model results. Nevertheless, it is not inconceivable that targeting metabolism in the future with cell-selective mTOR inhibitors (compared to the broad inhibitors tried to date) could be developed to improve efficacy and reduce side effects.
Collapse
|
70
|
Nwaubani P, Cercignani M, Colasanti A. In vivo quantitative imaging of hippocampal inflammation in autoimmune neuroinflammatory conditions: a systematic review. Clin Exp Immunol 2022; 210:24-38. [PMID: 35802780 PMCID: PMC9585553 DOI: 10.1093/cei/uxac058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/17/2022] [Accepted: 07/04/2022] [Indexed: 01/25/2023] Open
Abstract
The hippocampus is a morphologically complex region of the brain limbic system centrally involved in important cognitive, affective, and behavioural regulatory roles. It has exquisite vulnerability to neuroinflammatory processes, with some of its subregions found to be specific sites of neuroinflammatory pathology in ex-vivo studies. Optimizing neuroimaging correlates of hippocampal neuroinflammation would enable the direct study of functional consequences of hippocampal neuroinflammatory pathology, as well as the definition of therapeutic end-points for treatments targeting neuroinflammation, and their related affective or cognitive sequelae. However, in vivo traditional imaging of the hippocampus and its subregions is fraught with difficulties, due to methodological challenges deriving from its unique anatomical characteristics. The main objective of this review is to provide a current update on the characterization of quantitative neuroimaging correlates of hippocampal neuroinflammation by focusing on three prototypical autoimmune neuro-inflammatory conditions [multiple sclerosis (MS), systemic lupus erythematosus (SLE), and autoimmune encephalitis (AE)]. We focused on studies employing TSPO-targeting positron emission tomography (PET), quantitative magnetic resonance imaging (MRI), and spectroscopy techniques assumed to be sensitive to neuroinflammatory tissue changes. We found 18 eligible studies (14, 2, and 2 studies in MS, AE, and SLE, respectively). Across conditions, the largest effect was seen in TSPO PET and diffusion-weighted MRI studies. No study examined neuroinflammation-related changes at the hippocampal subfield level. Overall, results were largely inconsistent due to heterogeneous imaging methods, small sample sizes, and different population studies. We discuss how these data could inform future study design and conclude by suggesting further methodological directions aimed at improving the precision and sensitivity of neuroimaging techniques to characterize hippocampal neuroinflammatory pathology in the human brain.
Collapse
Affiliation(s)
- P Nwaubani
- Department of Clinical Neuroscience and Neuroimaging, Brighton and Sussex Medical School, University of Sussex, Falmer, Brighton, UK
| | - M Cercignani
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, UK
| | - A Colasanti
- Correspondence: Alessandro Colasanti, Department of Clinical Neuroscience and Neuroimaging, Brighton and Sussex Medical School, University of Sussex, Trafford Centre for Medical Research, University of Sussex, Falmer, Brighton, BN1 4RY, UK.
| |
Collapse
|
71
|
Misin O, Matilainen M, Nylund M, Honkonen E, Rissanen E, Sucksdorff M, Airas L. Innate Immune Cell–Related Pathology in the Thalamus Signals a Risk for Disability Progression in Multiple Sclerosis. NEUROLOGY - NEUROIMMUNOLOGY NEUROINFLAMMATION 2022; 9:9/4/e1182. [PMID: 35581004 PMCID: PMC9128041 DOI: 10.1212/nxi.0000000000001182] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/17/2022] [Indexed: 11/16/2022]
Abstract
Background and Objectives Our aim was to investigate whether 18-kDa translocator protein (TSPO) radioligand binding in gray matter (GM) predicts later disability progression in multiple sclerosis (MS). Methods In this prospective imaging study, innate immune cells were investigated in the MS patient brain using PET imaging. The distribution volume ratio (DVR) of the TSPO-binding radioligand [11C]PK11195 was determined in 5 GM regions: thalamus, caudate, putamen, pallidum, and cortical GM. Volumetric brain MRI parameters were obtained for comparison. The Expanded Disability Status Scale (EDSS) score was assessed at baseline and after follow-up of 3.0 ± 0.3 (mean ± SD) years. Disability progression was defined as an EDSS score increase of 1.0 point or 0.5 point if the baseline EDSS score was ≥6.0. A forward-type stepwise logistic regression model was constructed to compare multiple imaging and clinical variables in their ability to predict later disability progression. Results The cohort consisted of 66 patients with MS and 18 healthy controls. Patients with later disability progression (n = 17) had more advanced atrophy in the thalamus, caudate, and putamen at baseline compared with patients with no subsequent worsening. TSPO binding was significantly higher in the thalamus among the patients with later worsening. The thalamic DVR was the only measured imaging variable that remained a significant predictor of disability progression in the regression model. The final model predicted disability progression with 52.9% sensitivity and 93.9% specificity with an area under the curve value of 0.82 (receiver operating characteristic curve). Discussion Increased TSPO radioligand binding in the thalamus has potential in predicting short-term disability progression in MS and seems to be more sensitive for this than GM atrophy measures.
Collapse
|
72
|
Bhargava P, Hartung HP, Calabresi PA. Contribution of B cells to cortical damage in multiple sclerosis. Brain 2022; 145:3363-3373. [PMID: 35775595 DOI: 10.1093/brain/awac233] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/06/2022] [Accepted: 06/17/2022] [Indexed: 11/14/2022] Open
Abstract
Multiple sclerosis is associated with lesions not just in the white matter, but also involving the cortex. Cortical involvement has been linked to greater disease severity and hence understanding the factor underlying cortical pathology could help identify new therapeutic strategies for multiple sclerosis. The critical role of B cells in multiple sclerosis has been clarified by multiple pivotal trials of B cell depletion in people with multiple sclerosis. The presence of B cell rich areas of meningeal inflammation in multiple sclerosis has been identified at all stages of multiple sclerosis. Leptomeningeal inflammation is associated with greater extent of cortical demyelination and neuronal loss and with greater disease severity. Recent studies have identified several potential mechanisms by which B cells may mediate cortical injury including antibody production, extracellular vesicles containing neurotoxic substances and production of pro-inflammatory cytokines. Additionally, B cells may indirectly mediate cortical damage through effects on T cells, macrophages or microglia. Several animal models replicate the meningeal inflammation and cortical injury noted in people with multiple sclerosis. Studies in these models have identified BTK inhibition and type II anti-CD20 antibodies as potential agents that can impact meningeal inflammation. Trials of anti-CD20 monoclonal antibodies in people with multiple sclerosis have unsuccessfully attempted to eliminate B cells in the leptomeninges. New strategies to target B cells in multiple sclerosis include BTK inhibition and cell-based therapies aimed at B cells infected with Epstein Barr virus. Future studies will clarify the mechanisms by which B cells mediate cortical injury and treatment strategies that can target B cells in the leptomeninges and CNS parenchyma.
Collapse
Affiliation(s)
- Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hans Peter Hartung
- Department of Neurology, Heinrich-Heine University, Dusseldorf, Germany.,Brain and Mind Center, University of Sydney, Sydney, Australia.,Department of Neurology, Medical University of Vienna, Vienna, Austria.,Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
73
|
Reciprocal Interactions between Oligodendrocyte Precursor Cells and the Neurovascular Unit in Health and Disease. Cells 2022; 11:cells11121954. [PMID: 35741083 PMCID: PMC9221698 DOI: 10.3390/cells11121954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/02/2022] [Accepted: 06/14/2022] [Indexed: 12/04/2022] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are mostly known for their capability to differentiate into oligodendrocytes and myelinate axons. However, they have been observed to frequently interact with cells of the neurovascular unit during development, homeostasis, and under pathological conditions. The functional consequences of these interactions are largely unclear, but are increasingly studied. Although OPCs appear to be a rather homogenous cell population in the central nervous system (CNS), they present with an enormous potential to adapt to their microenvironment. In this review, it is summarized what is known about the various roles of OPC-vascular interactions, and the circumstances under which they have been observed.
Collapse
|
74
|
Lazzarotto A, Tonietto M, Poirion E, Battaglini M, Palladino R, Benoit C, Ricigliano VA, Maillart E, De Stefano N, Stankoff B, Bodini B. Clinically relevant profiles of myelin content changes in patients with multiple sclerosis: A multimodal and multicompartment imaging study. Mult Scler 2022; 28:1881-1890. [PMID: 35708126 DOI: 10.1177/13524585221096975] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To investigate the clinical relevance of individual profiles of cortical and white matter lesion myelin content changes combining magnetisation transfer imaging (MTI) and 11C-PiB-positron emission tomography (PET) in patients with multiple sclerosis (MS). METHODS MTI and [11C]PiB-PET acquired in 19 patients with MS followed up over 2-4 months and in seven healthy controls (HCs), were employed to generate individual maps of cortical and white matter (WM) lesion myelin content changes, respectively. These maps were used to calculate individual indices of demyelination and remyelination, and to investigate their association with clinical scores. RESULTS Cortical remyelination ranged between 1% and 5% of the total cortical volume (17%-45% of the cortical volume demyelinated at baseline). WM lesion remyelination ranged between 8% and 22% of the lesional volume. An extensive cortical remyelination was associated with a shorter disease duration (rho = -0.63, p = 0.01) and, in combination with WM lesion remyelination, explained 68%-70% of the variance of clinical scores (p < 0.01). CONCLUSION Our multimodal and multicompartment approach allows us to explore single-patient cortical and WM lesion demyelination and remyelination, and to generate clinically relevant indices of myelin repair. These indices may be used as outcome measures in clinical trials, thus increasing the chance to identify successful promyelinating treatments in patients with MS.
Collapse
Affiliation(s)
- Andrea Lazzarotto
- Sorbonne Université, Paris Brain Institute, CNRS, Inserm, Paris, France/Padova Neuroscience Center, University of Padua, Padua, Italy; AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Matteo Tonietto
- Sorbonne Université, Paris Brain Institute, CNRS, Inserm, Paris, France/CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Emilie Poirion
- Sorbonne Université, Paris Brain Institute, CNRS, Inserm, Paris, France; Hospital Foundation A. de Rothschild, Imaging department, Paris, France
| | - Marco Battaglini
- Department of Neurological and Behavioural Sciences, University of Siena, Siena, Italy
| | - Raffaele Palladino
- Department of Primary Care and Public Health, School of Public Health, Imperial College of London, London, UK/Department of Public Health, University 'Federico II' of Naples, Naples, Italy
| | - Charline Benoit
- Sorbonne Université, Paris Brain Institute, CNRS, Inserm, Paris, France
| | - Vito Ag Ricigliano
- Sorbonne Université, Paris Brain Institute, CNRS, Inserm, Paris, France/AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Elisabeth Maillart
- Sorbonne Université, Paris Brain Institute, CNRS, Inserm, Paris, France/AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Nicola De Stefano
- Department of Neurological and Behavioural Sciences, University of Siena, Siena, Italy
| | - Bruno Stankoff
- Sorbonne Université, Paris Brain Institute, CNRS, Inserm, Paris, France/AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Benedetta Bodini
- Sorbonne Université, Paris Brain Institute, CNRS, Inserm, Paris, France/AP-HP, Hôpital Saint-Antoine, Paris, France/Institut du Cerveau et de la moelle épinière, ICM, UPMC Univ Paris 06, UMR S 1127, and CNRS UMR 7225, Hopital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
75
|
Cao X, Lai SWT, Chen S, Wang S, Feng M. Targeting tumor-associated macrophages for cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:61-108. [PMID: 35636930 DOI: 10.1016/bs.ircmb.2022.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Tumor-associated macrophages (TAMs) are one of the most abundant immune components in the tumor microenvironment and play a plethora of roles in regulating tumorigenesis. Therefore, the therapeutic targeting of TAMs has emerged as a new paradigm for immunotherapy of cancer. Herein, the review summarizes the origin, polarization, and function of TAMs in the progression of malignant diseases. The understanding of such knowledge leads to several distinct therapeutic strategies to manipulate TAMs to battle cancer, which include those to reduce TAM abundance, such as depleting TAMs or inhibiting their recruitment and differentiation, and those to harness or boost the anti-tumor activities of TAMs such as blocking phagocytosis checkpoints, inducing antibody-dependent cellular phagocytosis, and reprogramming TAM polarization. In addition, modulation of TAMs may reshape the tumor microenvironment and therefore synergize with other cancer therapeutics. Therefore, the rational combination of TAM-targeting therapeutics with conventional therapies including radiotherapy, chemotherapy, and other immunotherapies is also reviewed. Overall, targeting TAMs presents itself as a promising strategy to add to the growing repertoire of treatment approaches in the fight against cancer, and it is hopeful that these approaches currently being pioneered will serve to vastly improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| | - Seigmund W T Lai
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Siqi Chen
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Sadira Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| |
Collapse
|
76
|
Madsen MAJ, Wiggermann V, Marques MFM, Lundell H, Cerri S, Puonti O, Blinkenberg M, Christensen JR, Sellebjerg F, Siebner HR. Linking lesions in sensorimotor cortex to contralateral hand function in multiple sclerosis: a 7 T MRI study. Brain 2022; 145:3522-3535. [PMID: 35653498 PMCID: PMC9586550 DOI: 10.1093/brain/awac203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Cortical lesions constitute a key manifestation of multiple sclerosis and contribute to clinical disability and cognitive impairment. Yet it is unknown whether local cortical lesions and cortical lesion subtypes contribute to domain-specific impairments attributable to the function of the lesioned cortex.
In this cross-sectional study, we assessed how cortical lesions in the primary sensorimotor hand area (SM1-HAND) relate to corticomotor physiology and sensorimotor function of the contralateral hand. 50 relapse-free patients with relapsing-remitting or secondary-progressive multiple sclerosis and 28 healthy age- and sex-matched participants underwent whole-brain 7 T MRI to map cortical lesions. Brain scans were also used to estimate normalized brain volume, pericentral cortical thickness, white matter lesion fraction of the corticospinal tract, infratentorial lesion volume and the cross-sectional area of the upper cervical spinal cord. We tested sensorimotor hand function and calculated a motor and sensory composite score for each hand. In 37 patients and 20 healthy controls, we measured maximal motor evoked potential (MEP) amplitude, resting motor threshold and corticomotor conduction time with transcranial magnetic stimulation (TMS) and the N20 latency from somatosensory evoked potentials (SSEPs).
Patients showed at least one cortical lesion in the SM1-HAND in 47 of 100 hemispheres. The presence of a lesion was associated with worse contralateral sensory (P = 0.014) and motor (P = 0.009) composite scores. TMS of a lesion-positive SM1-HAND revealed a decreased maximal MEP amplitude (P < 0.001) and delayed corticomotor conduction (P = 0.002) relative to a lesion-negative SM1-HAND. Stepwise mixed linear regressions showed that the presence of an SM1-HAND lesion, higher white-matter lesion fraction of the corticospinal tract, reduced spinal cord cross-sectional area and higher infratentorial lesion volume were associated with reduced contralateral motor hand function. Cortical lesions in SM1-HAND, spinal cord cross-sectional area and normalized brain volume were also associated with smaller maximal MEP amplitude and longer corticomotor conduction times. The effect of cortical lesions on sensory function was no longer significant when controlling for MRI-based covariates. Lastly, we found that intracortical and subpial lesions had the largest effect on reduced motor hand function, intracortical lesions on reduced MEP amplitude and leukocortical lesions on delayed corticomotor conduction.
Together, this comprehensive multi-level assessment of sensorimotor brain damage shows that the presence of a cortical lesion in SM1-HAND is associated with impaired corticomotor function of the hand, after accounting for damage at the subcortical level. The results also provide preliminary evidence that cortical lesion types may affect the various facets of corticomotor function differentially.
Collapse
Affiliation(s)
- Mads A. J. Madsen
- Copenhagen University Hospital - Amager & Hvidovre Danish Research Centre for Magnetic Resonance, , 2650 Hvidovre, Denmark
| | - Vanessa Wiggermann
- Copenhagen University Hospital - Amager & Hvidovre Danish Research Centre for Magnetic Resonance, , 2650 Hvidovre, Denmark
| | - Marta F. M. Marques
- Copenhagen University Hospital - Amager & Hvidovre Danish Research Centre for Magnetic Resonance, , 2650 Hvidovre, Denmark
| | - Henrik Lundell
- Copenhagen University Hospital - Amager & Hvidovre Danish Research Centre for Magnetic Resonance, , 2650 Hvidovre, Denmark
| | - Stefano Cerri
- Copenhagen University Hospital - Amager & Hvidovre Danish Research Centre for Magnetic Resonance, , 2650 Hvidovre, Denmark
- Technical University of Denmark Department of Health Technology, , 2800 Kgs. Lyngby, Denmark
| | - Oula Puonti
- Copenhagen University Hospital - Amager & Hvidovre Danish Research Centre for Magnetic Resonance, , 2650 Hvidovre, Denmark
| | - Morten Blinkenberg
- Copenhagen University Hospital – Rigshospitalet Danish Multiple Sclerosis Center, Department of Neurology, , 2600 Glostrup, Denmark
| | - Jeppe Romme Christensen
- Copenhagen University Hospital – Rigshospitalet Danish Multiple Sclerosis Center, Department of Neurology, , 2600 Glostrup, Denmark
| | - Finn Sellebjerg
- Copenhagen University Hospital – Rigshospitalet Danish Multiple Sclerosis Center, Department of Neurology, , 2600 Glostrup, Denmark
- University of Copenhagen Department of Clinical Medicine, , 2200 Copenhagen, Denmark
| | - Hartwig R. Siebner
- Copenhagen University Hospital - Amager & Hvidovre Danish Research Centre for Magnetic Resonance, , 2650 Hvidovre, Denmark
- Copenhagen University Hospital - Bispebjerg & Frederiksberg Department of Neurology, , 2400 Copenhagen, Denmark
- University of Copenhagen Department of Clinical Medicine, , 2200 Copenhagen, Denmark
| |
Collapse
|
77
|
Preziosa P, Pagani E, Bonacchi R, Cacciaguerra L, Falini A, Rocca MA, Filippi M. In vivo detection of damage in multiple sclerosis cortex and cortical lesions using NODDI. J Neurol Neurosurg Psychiatry 2022; 93:628-636. [PMID: 34799405 DOI: 10.1136/jnnp-2021-327803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/28/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To characterise in vivo the microstructural abnormalities of multiple sclerosis (MS) normal-appearing (NA) cortex and cortical lesions (CLs) and their relations with clinical phenotypes and disability using neurite orientation dispersion and density imaging (NODDI). METHODS One hundred and seventy-two patients with MS (101 relapsing-remitting multiple sclerosis (RRMS), 71 progressive multiple sclerosis (PMS)) and 62 healthy controls (HCs) underwent a brain 3T MRI. Brain cortex and CLs were segmented from three-dimensional T1-weighted and double inversion recovery sequences. Using NODDI on diffusion-weighted sequence, intracellular volume fraction (ICV_f) and Orientation Dispersion Index (ODI) were assessed in NA cortex and CLs with default or optimised parallel diffusivity for the cortex (D//=1.7 or 1.2 µm2/ms, respectively). RESULTS The NA cortex of patients with MS had significantly lower ICV_f versus HCs' cortex with both D// values (false discovery rate (FDR)-p <0.001). CLs showed significantly decreased ICV_f and ODI versus NA cortex of both HCs and patients with MS with both D// values (FDR-p ≤0.008). Patients with PMS versus RRMS had significantly decreased NA cortex ICV_f and ODI (FDR-p=0.050 and FDR-p=0.032) with only D//=1.7 µm2/ms. No CL microstructural differences were found between MS clinical phenotypes. MS NA cortex ICV_f and ODI were significantly correlated with disease duration, clinical disability, lesion burden and global and regional brain atrophy (r from -0.51 to 0.71, FDR-p from <0.001 to 0.045). CONCLUSIONS A significant neurite loss occurs in MS NA cortex. CLs show a further neurite density reduction and a reduced ODI suggesting a simplification of neurite complexity. NODDI is relevant to investigate in vivo the heterogeneous pathology affecting the MS cortex.
Collapse
Affiliation(s)
- Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Elisabetta Pagani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Raffaello Bonacchi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Laura Cacciaguerra
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Vita-Salute San Raffaele University, Milano, Italy
| | - Andrea Falini
- Vita-Salute San Raffaele University, Milano, Italy.,Department of Neuroradiology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Vita-Salute San Raffaele University, Milano, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy .,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Vita-Salute San Raffaele University, Milano, Italy.,Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
78
|
Collongues N, Becker G, Jolivel V, Ayme-Dietrich E, de Seze J, Binamé F, Patte-Mensah C, Monassier L, Mensah-Nyagan AG. A Narrative Review on Axonal Neuroprotection in Multiple Sclerosis. Neurol Ther 2022; 11:981-1042. [PMID: 35610531 PMCID: PMC9338208 DOI: 10.1007/s40120-022-00363-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/03/2022] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) resulting in demyelination and neurodegeneration. The therapeutic strategy is now largely based on reducing inflammation with immunosuppressive drugs. Unfortunately, when disease progression is observed, no drug offers neuroprotection apart from its anti-inflammatory effect. In this review, we explore current knowledge on the assessment of neurodegeneration in MS and look at putative targets that might prove useful in protecting the axon from degeneration. Among them, Bruton's tyrosine kinase inhibitors, anti-apoptotic and antioxidant agents, sex hormones, statins, channel blockers, growth factors, and molecules preventing glutamate excitotoxicity have already been studied. Some of them have reached phase III clinical trials and carry a great message of hope for our patients with MS.
Collapse
Affiliation(s)
- Nicolas Collongues
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France. .,Center for Clinical Investigation, INSERM U1434, Strasbourg, France. .,Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France. .,University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.
| | - Guillaume Becker
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Valérie Jolivel
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Estelle Ayme-Dietrich
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Jérôme de Seze
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France.,Center for Clinical Investigation, INSERM U1434, Strasbourg, France.,Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Fabien Binamé
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Christine Patte-Mensah
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Laurent Monassier
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Ayikoé Guy Mensah-Nyagan
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| |
Collapse
|
79
|
Bells S, Longoni G, Berenbaum T, de Medeiros CB, Narayanan S, Banwell BL, Arnold DL, Mabbott DJ, Ann Yeh E. Patterns of white and gray structural abnormality associated with paediatric demyelinating disorders. Neuroimage Clin 2022; 34:103001. [PMID: 35381508 PMCID: PMC8980471 DOI: 10.1016/j.nicl.2022.103001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 11/26/2022]
Abstract
A multi-modal approach was used to evaluate the visual pathway from anterior (retina) to posterior (visual cortex) in both paediatric MOGAD and MS patients. MS patients exhibited more widespread white matter abnormalities; MOGAD patients exhibited white matter changes primarily within the optic radiation. The pattern of cortical thinning differed in MS and MOGAD patients. Reduced RNFLT was associated with lower axonal density in MOGAD and tortuosity in MS.
The impact of multiple sclerosis (MS) and myelin oligodendrocyte glycoprotein (MOG) - associated disorders (MOGAD) on brain structure in youth remains poorly understood. Reductions in cortical mantle thickness on structural MRI and abnormal diffusion-based white matter metrics (e.g., diffusion tensor parameters) have been well documented in MS but not in MOGAD. Characterizing structural abnormalities found in children with these disorders can help clarify the differences and similarities in their impact on neuroanatomy. Importantly, while MS and MOGAD affect the entire CNS, the visual pathway is of particular interest in both groups, as most patients have evidence for clinical or subclinical involvement of the anterior visual pathway. Thus, the visual pathway is of key interest in analyses of structural abnormalities in these disorders and may distinguish MOGAD from MS patients. In this study we collected MRI data on 18 MS patients, 14 MOGAD patients and 26 age- and sex-matched typically developing children (TDC). Full-brain group differences in fixel diffusion measures (fibre-bundle populations) and cortical thickness measures were tested using age and sex as covariates. Visual pathway analysis was performed by extracting mean diffusion measures within lesion free optic radiations, cortical thickness within the visual cortex, and retinal nerve fibre layer (RNFL) and ganglion cell layer thickness measures from optical coherence tomography (OCT). Fixel based analysis (FBA) revealed MS patients have widespread abnormal white matter within the corticospinal tract, inferior longitudinal fasciculus, and optic radiations, while within MOGAD patients, non-lesional impact on white matter was found primarily in the right optic radiation. Cortical thickness measures were reduced predominately in the temporal and parietal lobes in MS patients and in frontal, cingulate and visual cortices in MOGAD patients. Additionally, our findings of associations between reduced RNFLT and axonal density in MOGAD and TORT in MS patients in the optic radiations imply widespread axonal and myelin damage in the visual pathway, respectively. Overall, our approach of combining FBA, cortical thickness and OCT measures has helped evaluate similarities and differences in brain structure in MS and MOGAD patients in comparison to TDC.
Collapse
Affiliation(s)
- Sonya Bells
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada; Pediatric Neurology, Spectrum Health Helen Devos Children's Hospital, Grand Rapids, USA; Department of Pediatrics and Human Development, Michigan State University, East Lansing, USA
| | - Giulia Longoni
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada; Department of Neurology, Hospital for Sick Children, Toronto, Canada; Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Tara Berenbaum
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Cynthia B de Medeiros
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Sridar Narayanan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Brenda L Banwell
- Division of Child Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, USA
| | - Douglas L Arnold
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Donald J Mabbott
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada; Department of Psychology, University of Toronto, Toronto, Canada
| | - E Ann Yeh
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Canada; Department of Neurology, Hospital for Sick Children, Toronto, Canada; Department of Paediatrics, University of Toronto, Toronto, Canada.
| |
Collapse
|
80
|
Wang F, Cai YJ, Ma X, Wang N, Wu ZB, Sun Y, Xu YX, Yang H, Liu TT, Xia Q, Yu Z, Zhu DF. Synaptic loss in a mouse model of euthyroid Hashimoto's thyroiditis: possible involvement of the microglia. BMC Neurosci 2022; 23:25. [PMID: 35468730 PMCID: PMC9036731 DOI: 10.1186/s12868-022-00710-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 04/05/2022] [Indexed: 11/24/2022] Open
Abstract
Background Hashimoto’s thyroiditis (HT) is an autoimmune illness that renders individuals vulnerable to neuropsychopathology even in the euthyroid state, the mechanisms involved remain unclear. We hypothesized that activated microglia might disrupt synapses, resulting in cognitive disturbance in the context of euthyroid HT, and designed the present study to test this hypothesis. Methods Experimental HT model was induced by immunizing NOD mice with thyroglobulin and adjuvant twice. Morris Water Maze was measured to determine mice spatial learning and memory. The synaptic parameters such as the synaptic density, synaptic ultrastructure and synaptic-markers (SYN and PSD95) as well as the interactions of microglia with synapses were also determined. Results HT mice had poorer performance in Morris Water Maze than controls. Concurrently, HT resulted in a significant reduction in synapse density and ultrastructure damage, along with decreased synaptic puncta visualized by immunostaining with synaptophysin and PSD-95. In parallel, frontal activated microglia in euthyroid HT mice showed increased engulfment of PSD95 and EM revealed that the synaptic structures were visible within the microglia. These functional alterations in microglia corresponded to structural increases in their attachment to neuronal perikarya and a reduction in presynaptic terminals covering the neurons. Conclusion Our results provide initial evidence that HT can induce synaptic loss in the euthyroid state with deficits might be attributable to activated microglia, which may underlie the deleterious effects of HT on spatial learning and memory. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-022-00710-2.
Collapse
Affiliation(s)
- Fen Wang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yao-Jun Cai
- Department of Endocrinology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiao Ma
- Department of Respiratoration, Wuhu Hospital of Traditional Chinese Medicine, Wuhu, 241000, China
| | - Nan Wang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zhang-Bi Wu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yan Sun
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yong-Xia Xu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Hao Yang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Tian-Tian Liu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Qin Xia
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zhen Yu
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - De-Fa Zhu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
81
|
Andica C, Hagiwara A, Yokoyama K, Kato S, Uchida W, Nishimura Y, Fujita S, Kamagata K, Hori M, Tomizawa Y, Hattori N, Aoki S. Multimodal magnetic resonance imaging quantification of gray matter alterations in relapsing-remitting multiple sclerosis and neuromyelitis optica spectrum disorder. J Neurosci Res 2022; 100:1395-1412. [PMID: 35316545 DOI: 10.1002/jnr.25035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 02/07/2022] [Accepted: 02/13/2022] [Indexed: 11/08/2022]
Abstract
Herein, we combined neurite orientation dispersion and density imaging (NODDI) and synthetic magnetic resonance imaging (SyMRI) to evaluate the spatial distribution and extent of gray matter (GM) microstructural alterations in patients with relapsing-remitting multiple sclerosis (RRMS) and neuromyelitis optica spectrum disorder (NMOSD). The NODDI (neurite density index [NDI], orientation dispersion index [ODI], and isotropic volume fraction [ISOVF]) and SyMRI (myelin volume fraction [MVF]) measures were compared between age- and sex-matched groups of 30 patients with RRMS (6 males and 24 females; mean age, 51.43 ± 8.02 years), 18 patients with anti-aquaporin-4 antibody-positive NMOSD (2 males and 16 females; mean age, 52.67 ± 16.07 years), and 19 healthy controls (6 males and 13 females; mean age, 51.47 ± 9.25 years) using GM-based spatial statistical analysis. Patients with RRMS showed reduced NDI and MVF and increased ODI and ISOVF, predominantly in the limbic and paralimbic regions, when compared with healthy controls, while only increases in ODI and ISOVF were observed when compared with NMOSD. Compared to NDI and MVF, the changes in ODI and ISOVF were observed more widely, including in the cerebellar cortex. These abnormalities were associated with disease progression and disability. In contrast, patients with NMOSD only showed reduced NDI mainly in the cerebellar, limbic, and paralimbic cortices when compared with healthy controls and patients with RRMS. Taken together, our study supports the notion that GM pathologies in RRMS are distinct from those of NMOSD. However, owing to the limitations of the study, the results should be cautiously interpreted.
Collapse
Affiliation(s)
- Christina Andica
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akifumi Hagiwara
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kazumasa Yokoyama
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shimpei Kato
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Wataru Uchida
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuma Nishimura
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Radiological Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Shohei Fujita
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaaki Hori
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Radiology, Toho University Omori Medical Center, Tokyo, Japan
| | - Yuji Tomizawa
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigeki Aoki
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
82
|
Weerasinghe-Mudiyanselage PDE, Ang MJ, Kang S, Kim JS, Moon C. Structural Plasticity of the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:3349. [PMID: 35328770 PMCID: PMC8955928 DOI: 10.3390/ijms23063349] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/10/2022] Open
Abstract
Neuroplasticity is the capacity of neural networks in the brain to alter through development and rearrangement. It can be classified as structural and functional plasticity. The hippocampus is more susceptible to neuroplasticity as compared to other brain regions. Structural modifications in the hippocampus underpin several neurodegenerative diseases that exhibit cognitive and emotional dysregulation. This article reviews the findings of several preclinical and clinical studies about the role of structural plasticity in the hippocampus in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. In this study, literature was surveyed using Google Scholar, PubMed, Web of Science, and Scopus, to review the mechanisms that underlie the alterations in the structural plasticity of the hippocampus in neurodegenerative diseases. This review summarizes the role of structural plasticity in the hippocampus for the etiopathogenesis of neurodegenerative diseases and identifies the current focus and gaps in knowledge about hippocampal dysfunctions. Ultimately, this information will be useful to propel future mechanistic and therapeutic research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Poornima D. E. Weerasinghe-Mudiyanselage
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Mary Jasmin Ang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
- College of Veterinary Medicine, University of the Philippines Los Baños, Los Baños 4031, Philippines
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| |
Collapse
|
83
|
Ahmed SM, Fransen NL, Touil H, Michailidou I, Huitinga I, Gommerman JL, Bar-Or A, Ramaglia V. Accumulation of meningeal lymphocytes correlates with white matter lesion activity in progressive multiple sclerosis. JCI Insight 2022; 7:151683. [PMID: 35104246 PMCID: PMC8983127 DOI: 10.1172/jci.insight.151683] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Subpial cortical demyelination is an important component of multiple sclerosis (MS) pathology contributing to disease progression, yet mechanism(s) underlying its development remain unclear. Compartmentalized inflammation involving the meninges may drive this type of injury. Given recent findings identifying substantial white matter (WM) lesion activity in patients with progressive MS, elucidating whether and how WM lesional activity relates to meningeal inflammation and subpial cortical injury is of interest. Using postmortem FFPE tissue blocks (range, 5-72 blocks; median, 30 blocks) for each of 27 patients with progressive MS, we assessed the relationship between meningeal inflammation, the extent of subpial cortical demyelination, and the state of subcortical WM lesional activity. Meningeal accumulations of T cells and B cells, but not myeloid cells, were spatially adjacent to subpial cortical lesions, and greater immune cell accumulation was associated with larger subpial lesion areas. Patients with a higher extent of meningeal inflammation harbored a greater proportion of active and mixed active/inactive WM lesions and an overall lower proportion of inactive and remyelinated WM lesions. Our findings support the involvement of meningeal lymphocytes in subpial cortical injury and point to a potential link between inflammatory subpial cortical demyelination and pathological mechanisms occurring in the subcortical WM.
Collapse
Affiliation(s)
- Shanzeh M. Ahmed
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Nina L. Fransen
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Meibergdreef, Amsterdam, Netherlands.,Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Hanane Touil
- Department of Neurology and Center for Neuroinflammation and Neurotherapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Iliana Michailidou
- Department of Clinical Genetics, Leiden University Medical Center, Einthovenweg, Leiden, Netherlands
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Meibergdreef, Amsterdam, Netherlands.,Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | | | - Amit Bar-Or
- Department of Neurology and Center for Neuroinflammation and Neurotherapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
84
|
Wu H, Sun C, Huang X, Wei R, Li Z, Ke D, Bai R, Liang H. Short-Range Structural Connections Are More Severely Damaged in Early-Stage MS. AJNR Am J Neuroradiol 2022; 43:361-367. [PMID: 35177546 PMCID: PMC8910797 DOI: 10.3174/ajnr.a7425] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 12/11/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Long-range connections are more severely damaged and relevant for cognition in long-standing MS. However, the evolution of such coordinated network damage in patients with MS is unclear. We investigated whether short- and long-range structural connections sustained equal damage in early-stage MS. MATERIALS AND METHODS Sixteen patients with early-stage MS and 17 healthy controls were scanned by high-resolution, multishell diffusion imaging on 7T MR imaging and assessed cognitively. We investigated macrostructural properties in short- and long-range fibers and of microstructural metrics derived from 2 quantitative diffusion MR imaging models: DTI and neurite orientation dispersion and density imaging. RESULTS Patients had significant WM integrity damage-that is, higher radial diffusivity and a lower intracellular volume fraction in the focal WM lesions. Compared with the healthy controls, the patients had noticeable microstructure changes in both short- and long-range fibers, including increased radial diffusivity, mean diffusivity, and axial diffusivity. Z scores further indicated greater damage in the short-range fibers than in the long-range fibers. CONCLUSIONS Our findings demonstrate that more severe demyelination preceding axonal degeneration occurs in short-range connections but not in long-range connections in early-stage MS, suggesting the possibility that there are cortical lesions that are undetectable by current MR imaging.
Collapse
Affiliation(s)
- H. Wu
- Frpm the Department of Neurology (H.W., X.H., R.W., D.K., H.L.), First Affiliated Hospital
| | - C. Sun
- Key Laboratory of Biomedical Engineering of Ministry of Education (C.S., Z.L.), College of Biomedical Engineering and Instrument Science
| | - X. Huang
- Frpm the Department of Neurology (H.W., X.H., R.W., D.K., H.L.), First Affiliated Hospital
| | - R. Wei
- Frpm the Department of Neurology (H.W., X.H., R.W., D.K., H.L.), First Affiliated Hospital
| | - Z. Li
- Key Laboratory of Biomedical Engineering of Ministry of Education (C.S., Z.L.), College of Biomedical Engineering and Instrument Science
| | - D. Ke
- Frpm the Department of Neurology (H.W., X.H., R.W., D.K., H.L.), First Affiliated Hospital
| | - R. Bai
- Department of Physical Medicine and Rehabilitation of the Affiliated Sir Run Run Shaw Hospital and Interdisciplinary Institute of Neuroscience and Technology (R.B.), School of Medicine, Zhejiang University, Hangzhou, China
| | - H. Liang
- Frpm the Department of Neurology (H.W., X.H., R.W., D.K., H.L.), First Affiliated Hospital
| |
Collapse
|
85
|
The role of glial cells in multiple sclerosis disease progression. Nat Rev Neurol 2022; 18:237-248. [PMID: 35190704 DOI: 10.1038/s41582-022-00624-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2022] [Indexed: 12/13/2022]
Abstract
Despite the development of highly effective treatments for relapsing-remitting multiple sclerosis (MS), limited progress has been made in addressing primary progressive or secondary progressive MS, both of which lead to loss of oligodendrocytes and neurons and axons, and to irreversible accumulation of disability. Neuroinflammation is central to all forms of MS. The current effective therapies for relapsing-remitting MS target the peripheral immune system; these treatments, however, have repeatedly failed in progressive MS. Greater understanding of inflammation driven by CNS-resident cells - including astrocytes and microglia - is, therefore, required to identify novel potential therapeutic opportunities. Advances in imaging, biomarker analysis and genomics suggest that microglia and astrocytes have central roles in the progressive disease process. In this Review, we provide an overview of the involvement of astrocytes and microglia at major sites of pathology in progressive MS. We discuss current and future therapeutic approaches to directly target glial cells, either to inhibit pathogenic functions or to restore homeostatic functions lost during the course of the disease. We also discuss how bidirectional communication between astrocytes and microglia needs to be considered, as therapeutic targeting of one is likely to alter the functions of the other.
Collapse
|
86
|
Beck ES, Maranzano J, Luciano NJ, Parvathaneni P, Filippini S, Morrison M, Suto DJ, Wu T, van Gelderen P, de Zwart JA, Antel S, Fetco D, Ohayon J, Andrada F, Mina Y, Thomas C, Jacobson S, Duyn J, Cortese I, Narayanan S, Nair G, Sati P, Reich DS. Cortical lesion hotspots and association of subpial lesions with disability in multiple sclerosis. Mult Scler 2022; 28:1351-1363. [PMID: 35142571 DOI: 10.1177/13524585211069167] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Dramatic improvements in visualization of cortical (especially subpial) multiple sclerosis (MS) lesions allow assessment of impact on clinical course. OBJECTIVE Characterize cortical lesions by 7 tesla (T) T2*-/T1-weighted magnetic resonance imaging (MRI); determine relationship with other MS pathology and contribution to disability. METHODS Sixty-four adults with MS (45 relapsing-remitting/19 progressive) underwent 3 T brain/spine MRI, 7 T brain MRI, and clinical testing. RESULTS Cortical lesions were found in 94% (progressive: median 56/range 2-203; relapsing-remitting: 15/0-168; p = 0.004). Lesion distribution across 50 cortical regions was nonuniform (p = 0.006), with highest lesion burden in supplementary motor cortex and highest prevalence in superior frontal gyrus. Leukocortical and white matter lesion volumes were strongly correlated (r = 0.58, p < 0.0001), while subpial and white matter lesion volumes were moderately correlated (r = 0.30, p = 0.002). Leukocortical (p = 0.02) but not subpial lesions (p = 0.40) were correlated with paramagnetic rim lesions; both were correlated with spinal cord lesions (p = 0.01). Cortical lesion volumes (total and subtypes) were correlated with expanded disability status scale, 25-foot timed walk, nine-hole peg test, and symbol digit modality test scores. CONCLUSION Cortical lesions are highly prevalent and are associated with disability and progressive disease. Subpial lesion burden is not strongly correlated with white matter lesions, suggesting differences in inflammation and repair mechanisms.
Collapse
Affiliation(s)
- Erin S Beck
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josefina Maranzano
- McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada; Department of Anatomy, University of Quebec in Trois-Rivières, Trois-Rivières, QC, Canada
| | - Nicholas J Luciano
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Prasanna Parvathaneni
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Stefano Filippini
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.,Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
| | - Mark Morrison
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Daniel J Suto
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Tianxia Wu
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Peter van Gelderen
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jacco A de Zwart
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Samson Antel
- McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Dumitru Fetco
- McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Joan Ohayon
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Frances Andrada
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Yair Mina
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chevaz Thomas
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Steve Jacobson
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jeff Duyn
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Irene Cortese
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Sridar Narayanan
- McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Govind Nair
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Pascal Sati
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.,Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel S Reich
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
87
|
Kamma E, Lasisi W, Libner C, Ng HS, Plemel JR. Central nervous system macrophages in progressive multiple sclerosis: relationship to neurodegeneration and therapeutics. J Neuroinflammation 2022; 19:45. [PMID: 35144628 PMCID: PMC8830034 DOI: 10.1186/s12974-022-02408-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/31/2022] [Indexed: 02/08/2023] Open
Abstract
There are over 15 disease-modifying drugs that have been approved over the last 20 years for the treatment of relapsing–remitting multiple sclerosis (MS), but there are limited treatment options available for progressive MS. The development of new drugs for the treatment of progressive MS remains challenging as the pathophysiology of progressive MS is poorly understood. The progressive phase of MS is dominated by neurodegeneration and a heightened innate immune response with trapped immune cells behind a closed blood–brain barrier in the central nervous system. Here we review microglia and border-associated macrophages, which include perivascular, meningeal, and choroid plexus macrophages, during the progressive phase of MS. These cells are vital and are largely the basis to define lesion types in MS. We will review the evidence that reactive microglia and macrophages upregulate pro-inflammatory genes and downregulate homeostatic genes, that may promote neurodegeneration in progressive MS. We will also review the factors that regulate microglia and macrophage function during progressive MS, as well as potential toxic functions of these cells. Disease-modifying drugs that solely target microglia and macrophage in progressive MS are lacking. The recent treatment successes for progressive MS include include B-cell depletion therapies and sphingosine-1-phosphate receptor modulators. We will describe several therapies being evaluated as a potential treatment option for progressive MS, such as immunomodulatory therapies that can target myeloid cells or as a potential neuroprotective agent.
Collapse
Affiliation(s)
- Emily Kamma
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Wendy Lasisi
- Recovery and Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, NL, Canada
| | - Cole Libner
- Department of Health Sciences and the Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Huah Shin Ng
- Division of Neurology and the Djavad Mowafaghian Centre for Brain Health, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jason R Plemel
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, Canada. .,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada. .,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada. .,University of Alberta, 5-64 Heritage Medical Research Centre, Edmonton, AB, T6G2S2, Canada.
| |
Collapse
|
88
|
White matter microglia heterogeneity in the CNS. Acta Neuropathol 2022; 143:125-141. [PMID: 34878590 DOI: 10.1007/s00401-021-02389-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/17/2021] [Accepted: 11/28/2021] [Indexed: 02/07/2023]
Abstract
Microglia, the resident myeloid cells in the central nervous system (CNS) play critical roles in shaping the brain during development, responding to invading pathogens, and clearing tissue debris or aberrant protein aggregations during ageing and neurodegeneration. The original concept that like macrophages, microglia are either damaging (pro-inflammatory) or regenerative (anti-inflammatory) has been updated to a kaleidoscope view of microglia phenotypes reflecting their wide-ranging roles in maintaining homeostasis in the CNS and, their contribution to CNS diseases, as well as aiding repair. The use of new technologies including single cell/nucleus RNA sequencing has led to the identification of many novel microglia states, allowing for a better understanding of their complexity and distinguishing regional variations in the CNS. This has also revealed differences between species and diseases, and between microglia and other myeloid cells in the CNS. However, most of the data on microglia heterogeneity have been generated on cells isolated from the cortex or whole brain, whereas white matter changes and differences between white and grey matter have been relatively understudied. Considering the importance of microglia in regulating white matter health, we provide a brief update on the current knowledge of microglia heterogeneity in the white matter, how microglia are important for the development of the CNS, and how microglial ageing affects CNS white matter homeostasis. We discuss how microglia are intricately linked to the classical white matter diseases such as multiple sclerosis and genetic white matter diseases, and their putative roles in neurodegenerative diseases in which white matter is also affected. Understanding the wide variety of microglial functions in the white matter may provide the basis for microglial targeted therapies for CNS diseases.
Collapse
|
89
|
van Wageningen T, Gerrits E, Brouwer N, Breve J, Geurts J, Eggen B, Boddeke H, van Dam AM. Distinct gene expression in demyelinated white and grey matter areas of patients with multiple sclerosis. Brain Commun 2022; 4:fcac005. [PMID: 35282162 PMCID: PMC8914505 DOI: 10.1093/braincomms/fcac005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/04/2021] [Accepted: 01/12/2022] [Indexed: 11/14/2022] Open
Abstract
Demyelination of the central nervous system is a prominent pathological hallmark of multiple sclerosis and affects both white and grey matter. However, demyelinated white and grey matter exhibit clear pathological differences, most notably the presence or absence of inflammation and activated glial cells in white and grey matter, respectively. In order to gain more insight into the differential pathology of demyelinated white and grey matter areas, we micro-dissected neighbouring white and grey matter demyelinated areas as well as normal-appearing matter from leucocortical lesions of human post-mortem material and used these samples for RNA sequencing. Our data show that even neighbouring demyelinated white and grey matter of the same leucocortical have a distinct gene expression profile and cellular composition. We propose that, based on their distinct expression profile, pathological processes in neighbouring white and grey matter are likely different which could have implications for the efficacy of treating grey matter lesions with current anti-inflammatory-based multiple sclerosis drugs.
Collapse
Affiliation(s)
- T.A. van Wageningen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Dept. Anatomy & Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - E. Gerrits
- Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, University Medical Center Groningen, Groningen, The Netherlands
| | - N. Brouwer
- Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, University Medical Center Groningen, Groningen, The Netherlands
| | - J. Breve
- Center for Healthy Ageing, Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| | - J.J.G. Geurts
- Amsterdam UMC, Vrije Universiteit Amsterdam, Dept. Anatomy & Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - B.J.L. Eggen
- Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, University Medical Center Groningen, Groningen, The Netherlands
| | - H.W.G.M. Boddeke
- Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, University Medical Center Groningen, Groningen, The Netherlands
- Center for Healthy Ageing, Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| | - A-M. van Dam
- Amsterdam UMC, Vrije Universiteit Amsterdam, Dept. Anatomy & Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
90
|
Rai NK, Singh V, Li L, Willard B, Tripathi A, Dutta R. Comparative Proteomic Profiling Identifies Reciprocal Expression of Mitochondrial Proteins Between White and Gray Matter Lesions From Multiple Sclerosis Brains. Front Neurol 2022; 12:779003. [PMID: 35002930 PMCID: PMC8740228 DOI: 10.3389/fneur.2021.779003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/29/2021] [Indexed: 12/27/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and demyelinating disease of the central nervous system, where ongoing demyelination and remyelination failure are the major factors for progressive neurological disability. In this report, we employed a comprehensive proteomic approach and immunohistochemical validation to gain insight into the pathobiological mechanisms that may be associated with the progressive phase of MS. Isolated proteins from myelinated regions, demyelinated white-matter lesions (WMLs), and gray-matter lesions (GMLs) from well-characterized progressive MS brain tissues were subjected to label-free quantitative mass spectrometry. Using a system-biology approach, we detected increased expression of proteins belonging to mitochondrial electron transport complexes and oxidative phosphorylation pathway in WMLs. Intriguingly, many of these proteins and pathways had opposite expression patterns and were downregulated in GMLs of progressive MS brains. A comparison to the human MitoCarta database mapped the mitochondrial proteins to mitochondrial subunits in both WMLs and GMLs. Taken together, we provide evidence of opposite expression of mitochondrial proteins in response to demyelination of white- and gray-matter regions in progressive MS brain.
Collapse
Affiliation(s)
- Nagendra Kumar Rai
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, United States
| | - Vaibhav Singh
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, United States
| | - Ling Li
- Proteomic Core Facility, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Belinda Willard
- Proteomic Core Facility, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.,Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, United States
| | - Ajai Tripathi
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, United States
| | - Ranjan Dutta
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, United States.,Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
91
|
Different Susceptibility of T and B Cells to Cladribine Depends On Their Levels of Deoxycytidine Kinase Activity Linked to Activation Status. J Neuroimmune Pharmacol 2022; 17:195-205. [PMID: 33851318 PMCID: PMC9726780 DOI: 10.1007/s11481-021-09994-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 03/31/2021] [Indexed: 02/07/2023]
Abstract
Deoxycytidine kinase (dCK) and 5' deoxynucleotidase (NT5C2) are involved in metabolism of cladribine (2CdA), the immunomodulatory drug for multiple sclerosis; by mediating phosphorylation (activation) or phosphorolysis (deactivation) of 2CdA, respectively, these enzymes promote or prevent its accumulation in the cell, which leads to cell death. In particular, lymphocytes which present with a high intracellular dCK/NT5C2 ratio are more sensitive to 2CdA than other immune cells. We aim at determining if the expression of these enzymes and/or their activity differ in specific progenitor and mature immune cells and are influenced by cellular activation and/or exposure to 2CdA. Flow cytometry analysis showed no difference in dCK/NT5C2 ratio in progenitor and mature immune cells. 2CdA induced apoptosis in stimulated T and B cells and unstimulated B cells. dCK expression was enhanced by 2CdA at mRNA and protein levels in activated T cells and mRNA level in activated B cells. dCK activity, measured through an in-house luminescence release enzyme assay was higher in activated T and B cells, and such an increase was abrogated in activated B cells, but not T cells, upon exposure to 2CdA. These results reveal an important relationship between dCK activity and the effect of 2CdA on B and T cells, according to their activation status. Further study is warranted to evaluate whether dCK activity could, in the future, be a suitable predictive biomarker of lymphocyte response to 2CdA.
Collapse
|
92
|
B cells in central nervous system disease: diversity, locations and pathophysiology. Nat Rev Immunol 2022; 22:513-524. [PMID: 34903877 PMCID: PMC8667979 DOI: 10.1038/s41577-021-00652-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 12/13/2022]
Abstract
B cells represent a relatively minor cell population within both the healthy and diseased central nervous system (CNS), yet they can have profound effects. This is emphasized in multiple sclerosis, in which B cell-depleting therapies are arguably the most efficacious treatment for the condition. In this Review, we discuss how B cells enter and persist in the CNS and how, in many neurological conditions, B cells concentrate within CNS barriers but are rarely found in the parenchyma. We highlight how B cells can contribute to CNS pathology through antibody secretion, antigen presentation and secretion of neurotoxic molecules, using examples from CNS tumours, CNS infections and autoimmune conditions such as neuromyelitis optica and, in particular, multiple sclerosis. Overall, understanding common and divergent principles of B cell accumulation and their effects within the CNS could offer new insights into treating these devastating neurological conditions.
Collapse
|
93
|
Age-related changes in multiple sclerosis and experimental autoimmune encephalomyelitis. Semin Immunol 2022; 59:101631. [PMID: 35752572 DOI: 10.1016/j.smim.2022.101631] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
A better understanding of the pathological mechanisms that drive neurodegeneration in people living with multiple sclerosis (MS) is needed to design effective therapies to treat and/or prevent disease progression. We propose that CNS-intrinsic inflammation and re-modelling of the sub-arachnoid space of the leptomeninges sets the stage for neurodegeneration from the earliest stages of MS. While neurodegenerative processes are clinically silent early in disease, ageing results in neurodegenerative changes that become clinically manifest as progressive disability. Here we review pathological correlates of MS disease progression, highlight emerging mouse models that mimic key progressive changes in MS, and provide new perspectives on therapeutic approaches to protect against MS-associated neurodegeneration.
Collapse
|
94
|
Wang Z, Ji L, Ren Y, Liu M, Ai X, Yang C. Preparation and Anti-tumor Study of Dextran 70,000-Selenium Nanoparticles and Poloxamer 188-Selenium Nanoparticles. AAPS PharmSciTech 2021; 23:29. [PMID: 34931279 DOI: 10.1208/s12249-021-02141-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022] Open
Abstract
The anti-tumor effect of selenium nanoparticles (SeNPs) has received more and more attention. However, the clinical application of SeNPs is not optimistic due to the poor stability. To improve the stability of SeNPs, many polymers are used to modify the SeNPs. However, most of the polymers are not approved by FDA. It is significant to develop a SeNPs product with good stability for clinic application. Dextran 70,000 (T70) and poloxamer 188 (P188) are FDA-approved pharmaceutical injection excipients. In this study, we decorate SeNPs with T70 and P188 and assess the physicochemical characterization, storage stability, and anti-tumor activities of T70-SeNPs and P188-SeNPs. Transmission electron microscopy (TEM) shows that T70-SeNPs and P188-SeNPs are spherical particles with particle sizes of 110 nm and 60 nm respectively. Fourier-Transform Infrared Spectra (FT-IR) show that T70 or P188 can interact with SeNPs through hydrogen bonding. Stability study shows that P188-SeNPs freeze-dried powder and T70-SeNPs freeze-dried powder remain stable at 4℃ for 6 months. T70-SeNPs and P188-SeNPs can aggregate in cell matrix and play an anti-tumor role to HepG2 by promoting apoptosis, increasing reactive oxygen species (ROS) content and reducing mitochondrial membrane potential (MMP). This study can provide reference for industrial production of SeNPs products.
Collapse
|
95
|
Möck EEA, Honkonen E, Airas L. Synaptic Loss in Multiple Sclerosis: A Systematic Review of Human Post-mortem Studies. Front Neurol 2021; 12:782599. [PMID: 34912290 PMCID: PMC8666414 DOI: 10.3389/fneur.2021.782599] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/26/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Gray matter pathology plays a central role in the progression of multiple sclerosis (MS). The occurrence of synaptic loss appears to be important but, to date, still poorly investigated aspect of MS pathology. In this systematic review, we drew from the recent knowledge about synaptic loss in human post-mortem studies. Methods: We conducted a systematic search with PubMed to identify relevant publications. Publications available from15 June 2021 were taken into account. We selected human post-mortem studies that quantitatively assessed the synapse number in MS tissue. Results: We identified 14 relevant publications out of which 9 reported synaptic loss in at least one investigated subregion. The most commonly used synaptic marker was synaptophysin; non-etheless, we found substantial differences in the methodology and the selection of reference tissue. Investigated regions included the cortex, the hippocampus, the cerebellum, the thalamus, and the spinal cord. Conclusion: Synaptic loss seems to take place throughout the entire central nervous system. However, the results are inconsistent, probably due to differences in the methodology. Moreover, synaptic loss appears to be a dynamic process, and thus the nature of this pathology might be captured using in vivo synaptic density measurements.
Collapse
Affiliation(s)
- E E Amelie Möck
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Eveliina Honkonen
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Laura Airas
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
96
|
Neuroprotective Effect of Glatiramer Acetate on Neurofilament Light Chain Leakage and Glutamate Excess in an Animal Model of Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms222413419. [PMID: 34948217 PMCID: PMC8707261 DOI: 10.3390/ijms222413419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 11/29/2022] Open
Abstract
Axonal and neuronal pathologies are a central constituent of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), induced by the myelin oligodendrocyte glycoprotein (MOG) 35–55 peptide. In this study, we investigated neurodegenerative manifestations in chronic MOG 35–55 induced EAE and the effect of glatiramer acetate (GA) treatment on these manifestations. We report that the neuronal loss seen in this model is not attributed to apoptotic neuronal cell death. In EAE-affected mice, axonal damage prevails from the early disease phase, as revealed by analysis of neurofilament light (NFL) leakage into the sera along the disease duration, as well as by immunohistological examination. Elevation of interstitial glutamate concentrations measured in the cerebrospinal fluid (CSF) implies that glutamate excess plays a role in the damage processes inflicted by this disease. GA applied as a therapeutic regimen to mice with apparent clinical symptoms significantly reduces the pathological manifestations, namely apoptotic cell death, NFL leakage, histological tissue damage, and glutamate excess, thus corroborating the neuroprotective consequences of this treatment.
Collapse
|
97
|
Yates RL, Pansieri J, Li Q, Bell JS, Yee SA, Palace J, Esiri MM, DeLuca GC. The influence of HLA-DRB1*15 on the relationship between microglia and neurons in multiple sclerosis normal appearing cortical grey matter. Brain Pathol 2021; 32:e13041. [PMID: 34904300 PMCID: PMC9245937 DOI: 10.1111/bpa.13041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/06/2022] Open
Abstract
Cortical tissue injury is common in multiple sclerosis (MS) and associates with disability progression. We have previously shown that HLA‐DRB1*15 genotype status associates with the extent of cortical inflammatory pathology. In the current study, we sought to examine the influence of HLA‐DRB1*15 on relationships between inflammation and neurodegeneration in MS. Human post‐mortem MS cases (n = 47) and controls (n = 10) were used. Adjacent sections of motor cortex were stained for microglia (Iba1+, CD68+, TMEM119+), lymphocytes (CD3+, CD8+), GFAP+ astrocytes, and neurons (NeuN+). A subset of MS cases (n = 20) and controls (n = 7) were double‐labeled for neurofilament and glutamic acid decarboxylase 65/67 (GAD+) to assess the extent of the inhibitory synaptic loss. In MS cases, microglial protein expression positively correlated with neuron density (Iba1+: r = 0.548, p < 0.001, CD68+: r = 0.498, p = 0.001, TMEM119+ r = 0.437, p = 0.003). This finding was restricted to MS cases not carrying HLA‐DRB1*15. Evidence of a 14% reduction in inhibitory synapses in MS was detected (MS: 0.299 ± 0.006 synapses/μm2 neuronal membrane versus control: 0.348 ± 0.009 synapses/μm2 neuronal membrane, p = 0.005). Neurons expressing inhibitory synapses were 24% smaller in MS cases compared to the control (MS: 403 ± 15 μm2 versus control: 531 ± 29 μm2, p = 0.001), a finding driven by HLA‐DRB1*15+ cases (15+: 376 ± 21 μm2 vs. 15−: 432 ± 22 μm2, p = 0.018). Taken together, our results demonstrate that HLA‐DRB1*15 modulates the relationship between microglial inflammation, inhibitory synapses, and neuronal density in the MS cortex.
Collapse
Affiliation(s)
- Richard L Yates
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jonathan Pansieri
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Qizhu Li
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Jack S Bell
- Salford Royal NHS Foundation Trust, Salford, UK
| | - Sydney A Yee
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Margaret M Esiri
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
98
|
Lema Dopico A, Choi S, Hua J, Li X, Harrison DM. Multi-layer analysis of quantitative 7 T magnetic resonance imaging in the cortex of multiple sclerosis patients reveals pathology associated with disability. Mult Scler 2021; 27:2040-2051. [PMID: 33596719 PMCID: PMC8371108 DOI: 10.1177/1352458521994556] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Cortical demyelination is a relevant aspect of tissue damage in multiple sclerosis (MS). Microstructural changes may affect each layer in the cortex differently. OBJECTIVES To evaluate the sensitivity of quantitative magnetic resonance imaging (qMRI) measurements on cortical layers as clinically accessible biomarkers of grey matter (GM) pathology. METHODS Forty-five participants with MS underwent 7 T magnetic resonance imaging (MRI) of the brain. Magnetization prepared two rapid acquisition gradient echoes (MP2RAGE) was processed for T1-weighted images and a T1 map. Multi-echo gradient echo images were processed for quantitative susceptibility and R2* maps. Cortical GM volumes were segmented into four cortical layers, and relaxometry metrics were calculated within and between these layers. RESULTS Significant correlations were found for disability scales and multi-layer metrics, for example, Expanded Disability Status Scale (EDSS) and peak height (PH) in the subpial (T1: ρ = -0.372, p < 0.050) and inner (R2*: ρ = -0.359, p < 0.050) cortical layers. Multivariate regression showed interdependency between atrophy and cortical metrics in some instances, but an independent relationship between cortical metrics and disability in others. CONCLUSION Cortical layer 7 T qMRI analyses reveal layer-specific relationships with disability in MS and allow emergence of clinically relevant associations that are hidden when analysing the full cortex.
Collapse
Affiliation(s)
- Alfonso Lema Dopico
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Seongjin Choi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jun Hua
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA/Neurosection, Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xu Li
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA/Neurosection, Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel M Harrison
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA/F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| |
Collapse
|
99
|
Fazia T, Marzanati D, Carotenuto AL, Beecham A, Hadjixenofontos A, McCauley JL, Saddi V, Piras M, Bernardinelli L, Gentilini D. Homozygosity Haplotype and Whole-Exome Sequencing Analysis to Identify Potentially Functional Rare Variants Involved in Multiple Sclerosis among Sardinian Families. Curr Issues Mol Biol 2021; 43:1778-1793. [PMID: 34889895 PMCID: PMC8929092 DOI: 10.3390/cimb43030125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/13/2021] [Accepted: 10/23/2021] [Indexed: 12/24/2022] Open
Abstract
Multiple Sclerosis (MS) is a complex multifactorial autoimmune disease, whose sex- and age-adjusted prevalence in Sardinia (Italy) is among the highest worldwide. To date, 233 loci were associated with MS and almost 20% of risk heritability is attributable to common genetic variants, but many low-frequency and rare variants remain to be discovered. Here, we aimed to contribute to the understanding of the genetic basis of MS by investigating potentially functional rare variants. To this end, we analyzed thirteen multiplex Sardinian families with Immunochip genotyping data. For five families, Whole Exome Sequencing (WES) data were also available. Firstly, we performed a non-parametric Homozygosity Haplotype analysis for identifying the Region from Common Ancestor (RCA). Then, on these potential disease-linked RCA, we searched for the presence of rare variants shared by the affected individuals by analyzing WES data. We found: (i) a variant (43181034 T > G) in the splicing region on exon 27 of CUL9; (ii) a variant (50245517 A > C) in the splicing region on exon 16 of ATP9A; (iii) a non-synonymous variant (43223539 A > C), on exon 9 of TTBK1; (iv) a non-synonymous variant (42976917 A > C) on exon 9 of PPP2R5D; and v) a variant (109859349-109859354) in 3'UTR of MYO16.
Collapse
Affiliation(s)
- Teresa Fazia
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (D.M.); (A.L.C.); (L.B.); (D.G.)
| | - Daria Marzanati
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (D.M.); (A.L.C.); (L.B.); (D.G.)
| | - Anna Laura Carotenuto
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (D.M.); (A.L.C.); (L.B.); (D.G.)
| | - Ashley Beecham
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.B.); (A.H.); (J.L.M.)
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL 33136, USA
| | - Athena Hadjixenofontos
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.B.); (A.H.); (J.L.M.)
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL 33136, USA
| | - Jacob L. McCauley
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.B.); (A.H.); (J.L.M.)
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL 33136, USA
| | - Valeria Saddi
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, 08100 Nuoro, Italy; (V.S.); (M.P.)
| | - Marialuisa Piras
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, 08100 Nuoro, Italy; (V.S.); (M.P.)
| | - Luisa Bernardinelli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (D.M.); (A.L.C.); (L.B.); (D.G.)
| | - Davide Gentilini
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (D.M.); (A.L.C.); (L.B.); (D.G.)
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, 20095 Cusano Milanino, Italy
| |
Collapse
|
100
|
Sherafat A, Pfeiffer F, Nishiyama A. Shaping of Regional Differences in Oligodendrocyte Dynamics by Regional Heterogeneity of the Pericellular Microenvironment. Front Cell Neurosci 2021; 15:721376. [PMID: 34690700 PMCID: PMC8531270 DOI: 10.3389/fncel.2021.721376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are glial cells that differentiate into mature oligodendrocytes (OLs) to generate new myelin sheaths. While OPCs are distributed uniformly throughout the gray and white matter in the developing and adult brain, those in white matter proliferate and differentiate into oligodendrocytes at a greater rate than those in gray matter. There is currently lack of evidence to suggest that OPCs comprise genetically and transcriptionally distinct subtypes. Rather, the emerging view is that they exist in different cell and functional states, depending on their location and age. Contrary to the normal brain, demyelinated lesions in the gray matter of multiple sclerosis brains contain more OPCs and OLs and are remyelinated more robustly than those in white matter. The differences in the dynamic behavior of OL lineage cells are likely to be influenced by their microenvironment. There are regional differences in astrocytes, microglia, the vasculature, and the composition of the extracellular matrix (ECM). We will discuss how the regional differences in these elements surrounding OPCs might shape their phenotypic variability in normal and demyelinated states.
Collapse
Affiliation(s)
- Amin Sherafat
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Friederike Pfeiffer
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States.,Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States.,Institute of Systems Genomics, University of Connecticut, Storrs, CT, United States.,The Institute of Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, United States
| |
Collapse
|