1
|
Moregola A, Bonacina F, Vingiani GB, Frapolli R, Turrini R, Norata GD. Profiling the impact of anti-human CD20 monoclonal antibodies on lymphocyte B cell subsets and their precursors in the bone marrow and in lymphoid tissues in an immunocompromised mouse engrafted with human cells. Pharmacol Res 2024; 209:107442. [PMID: 39374887 DOI: 10.1016/j.phrs.2024.107442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024]
Abstract
Ofatumumab (OFA) and ocrelizumab (OCRE) are two anti-CD20 monoclonal antibodies approved for the treatment of relapsing forms of multiple sclerosis due to their ability to deplete B lymphocytes. The aim of this study was to investigate the impact of these anti-hCD20 antibodies on B lymphocyte subsets in the circulation and in primary and secondary lymphoid organs in an immune system humanized mouse model (immunocompromised Rag2-/-Il2rg-/-CD47-/-) engrafted with human CD34+ hematopoietic stem cells. Three months after humanization, mice, which present adaptive immune cells only of human origin, were treated with OFA (0.3 mg/Kg; day 1, 3 and 5), or OCRE (10 mg/kg; day 1) or saline. Seven days after the last injection a robust (>90 %) decrease of circulating human CD20+ B lymphocytes was observed in both OFA- and OCRE-treated mice. A partial replenishment of B lymphocytes was detectable in blood 36 days from the last injection in OFA-treated mice, while no B lymphocytes could be detected in OCRE-treated mice up to 65 days post injection. Bone marrow profiling showed that during hCD20+ B cell depletion and replenishment, OCRE-treated mice preserved only preB-I cells in the bone marrow, while the bone marrow of OFA-treated mice presented both preB-I as well as preB-II cells, with the latter subset being the one closest to differentiate into immature B cells. These data together with changes in B cell distribution in other tissues suggest that ofatumumab preserve BM niches, critical for B lymphocyte replenishment, limiting potential side effects of the treatment associated with the increased risk of infection.
Collapse
Affiliation(s)
- Annalisa Moregola
- Department of Excellence of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Fabrizia Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Giovanni Battista Vingiani
- Department of Excellence of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Roberta Frapolli
- Laboratory of Cancer Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Giuseppe Danilo Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
2
|
Okutan B, Frederiksen JL, Houen G, Sellebjerg F, Kyllesbech C, Magyari M, Paunovic M, Sørensen PS, Jacobsen C, Lassmann H, Bramow S. Subcortical plaques and inflammation reflect cortical and meningeal pathologies in progressive multiple sclerosis. Brain Pathol 2024:e13314. [PMID: 39460678 DOI: 10.1111/bpa.13314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
It remains elusive whether lesions and inflammation in the sub/juxtacortical white matter reflect cortical and/or meningeal pathologies. Elucidating this could have implications for MRI monitoring as sub/juxtacortical lesions are detectable by routine MRI, while cortical lesions and meningeal inflammation are not. By large-area microscopy, we quantified total and mixed active plaque loads along with densities and sizes of perivascular mononuclear infiltrates (infiltrates) in the sub/juxtacortical white matter ≤2 mm from the cortex, intra-cortically and in the meninges. Data were related to ante-mortem clinical parameters in a false discovery rate-corrected analysis. We compared 12 patients with primary progressive multiple sclerosis (PPMS) and 15 with secondary progressive MS to 22 controls. Fifteen patients and 11 controls contributed with hemispheric sections. Sections were stained with haematoxylin-eosin, for myelin and for microglia/macrophages. B cells and T cells were confirmed in a subset. Immunoglobulin G depositions in selected cortical plaques resembled depositions described before in "slowly expanding" plaques in the white matter. We quantified plaque activity by measuring microglia-dominated and macrophage-dominated areas. Sub/juxtacortical plaques (load and activity) reflected plaque activity in the cerebral cortex. Plaque activity and infiltrates were more pronounced in the sub/juxtacortical white matter than in the cerebral cortex while conversely, the total plaque load was highest in the cortex. Infiltrates correlated trans-cortically and sub/juxtacortical plaque activity reflected cortical and meningeal infiltrates. Sub/juxtacortical infiltrate sizes correlated with shorter survival after progression onset. Two patients with PPMS and putatively fatal brain stem lesions argue against incidental findings. Trans-cortical inflammatory flares and plaque activity may be pathogenic in progressive MS. We suggest emphasis on sub/juxtacortical MRI lesions as plausible surrogates for cortical and meningeal pathologies and, when present, as indicators for cognitive testing.
Collapse
Affiliation(s)
- Betül Okutan
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Jette L Frederiksen
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gunnar Houen
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Finn Sellebjerg
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Kyllesbech
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Melinda Magyari
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Danish Multiple Sclerosis Registry, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Manuela Paunovic
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Per S Sørensen
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina Jacobsen
- Section of Forensic Pathology, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Stephan Bramow
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Pathology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
3
|
Schild H, Bopp T. [Immunological foundations of neurological diseases]. DER NERVENARZT 2024; 95:894-908. [PMID: 38953921 DOI: 10.1007/s00115-024-01696-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/07/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Neurodegenerative diseases represent an increasing challenge in ageing societies, as only limited treatment options are currently available. OBJECTIVE New research methods and interdisciplinary interaction of different disciplines have changed the way neurological disorders are viewed and paved the way for the comparatively new field of neuroimmunology, which was established in the early 1980s. Starting from neurological autoimmune diseases, such as multiple sclerosis, knowledge about the involvement of immunological processes in other contexts, such as stroke or traumatic brain injury, has been significantly expanded in recent years. MATERIAL AND METHODS This review article provides an overview of the role of the immune system and the resulting potential for novel treatment approaches. RESULTS The immune system plays a central role in fighting infections but is also able to react to the body's own signals under sterile conditions and cause inflammation and subsequent adaptive immune responses through the release of immune mediators and the recruitment and differentiation of certain immune cell types. This can be beneficial in initiating healing processes; however, chronic inflammatory conditions usually have destructive consequences for the tissue and the organism and must be interrupted. CONCLUSION It is now known that different cells of the immune system play an important role in neurological diseases. Regulatory mechanisms, which are mediated by regulatory T cells or Th2 cells, are usually associated with a good prognosis, whereas inflammatory processes and polarization towards Th1 or Th17 have a destructive character. Novel immunomodulators, which are also increasingly being used in cancer treatment, can now be used in a tissue-specific manner and therefore offer great potential for use in neurological diseases.
Collapse
Affiliation(s)
- Hansjörg Schild
- Institut für Immunologie, Universitätsmedizin Mainz, Langenbeckstraße 1, 55131, Mainz, Deutschland
| | - Tobias Bopp
- Institut für Immunologie, Universitätsmedizin Mainz, Langenbeckstraße 1, 55131, Mainz, Deutschland.
| |
Collapse
|
4
|
Stys PK, Tsutsui S, Gafson AR, ‘t Hart BA, Belachew S, Geurts JJG. New views on the complex interplay between degeneration and autoimmunity in multiple sclerosis. Front Cell Neurosci 2024; 18:1426231. [PMID: 39161786 PMCID: PMC11330826 DOI: 10.3389/fncel.2024.1426231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/14/2024] [Indexed: 08/21/2024] Open
Abstract
Multiple sclerosis (MS) is a frequently disabling neurological disorder characterized by symptoms, clinical signs and imaging abnormalities that typically fluctuate over time, affecting any level of the CNS. Prominent lymphocytic inflammation, many genetic susceptibility variants involving immune pathways, as well as potent responses of the neuroinflammatory component to immunomodulating drugs, have led to the natural conclusion that this disease is driven by a primary autoimmune process. In this Hypothesis and Theory article, we discuss emerging data that cast doubt on this assumption. After three decades of therapeutic experience, what has become clear is that potent immune modulators are highly effective at suppressing inflammatory relapses, yet exhibit very limited effects on the later progressive phase of MS. Moreover, neuropathological examination of MS tissue indicates that degeneration, CNS atrophy, and myelin loss are most prominent in the progressive stage, when lymphocytic inflammation paradoxically wanes. Finally, emerging clinical observations such as "progression independent of relapse activity" and "silent progression," now thought to take hold very early in the course, together argue that an underlying "cytodegenerative" process, likely targeting the myelinating unit, may in fact represent the most proximal step in a complex pathophysiological cascade exacerbated by an autoimmune inflammatory overlay. Parallels are drawn with more traditional neurodegenerative disorders, where a progressive proteopathy with prion-like propagation of toxic misfolded species is now known to play a key role. A potentially pivotal contribution of the Epstein-Barr virus and B cells in this process is also discussed.
Collapse
Affiliation(s)
- Peter K. Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shigeki Tsutsui
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Arie R. Gafson
- Biogen Digital Health, Biogen, Cambridge, MA, United States
| | - Bert A. ‘t Hart
- Department of Anatomy and Neurosciences, Amsterdam University Medical Centers (location VUmc), Amsterdam, Netherlands
| | - Shibeshih Belachew
- TheraPanacea, Paris, France
- Indivi (DBA of Healios AG), Basel, Switzerland
| | - Jeroen J. G. Geurts
- Department of Anatomy and Neurosciences, Amsterdam University Medical Centers (location VUmc), Amsterdam, Netherlands
| |
Collapse
|
5
|
Hemond CC, Gaitán MI, Absinta M, Reich DS. New Imaging Markers in Multiple Sclerosis and Related Disorders: Smoldering Inflammation and the Central Vein Sign. Neuroimaging Clin N Am 2024; 34:359-373. [PMID: 38942521 PMCID: PMC11213979 DOI: 10.1016/j.nic.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Concepts of multiple sclerosis (MS) biology continue to evolve, with observations such as "progression independent of disease activity" challenging traditional phenotypic categorization. Iron-sensitive, susceptibility-based imaging techniques are emerging as highly translatable MR imaging sequences that allow for visualization of at least 2 clinically useful biomarkers: the central vein sign and the paramagnetic rim lesion (PRL). Both biomarkers demonstrate high specificity in the discrimination of MS from other mimics and can be seen at 1.5 T and 3 T field strengths. Additionally, PRLs represent a subset of chronic active lesions engaged in "smoldering" compartmentalized inflammation behind an intact blood-brain barrier.
Collapse
Affiliation(s)
- Christopher C Hemond
- Department of Neurology, University of Massachusetts Memorial Medical Center and University of Massachusetts Chan Medical School, Worcester, MA, USA; National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - María I Gaitán
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Martina Absinta
- Translational Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
6
|
Calabrese M, Preziosa P, Scalfari A, Colato E, Marastoni D, Absinta M, Battaglini M, De Stefano N, Di Filippo M, Hametner S, Howell OW, Inglese M, Lassmann H, Martin R, Nicholas R, Reynolds R, Rocca MA, Tamanti A, Vercellino M, Villar LM, Filippi M, Magliozzi R. Determinants and Biomarkers of Progression Independent of Relapses in Multiple Sclerosis. Ann Neurol 2024; 96:1-20. [PMID: 38568026 DOI: 10.1002/ana.26913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/04/2024] [Accepted: 02/15/2024] [Indexed: 06/20/2024]
Abstract
Clinical, pathological, and imaging evidence in multiple sclerosis (MS) suggests that a smoldering inflammatory activity is present from the earliest stages of the disease and underlies the progression of disability, which proceeds relentlessly and independently of clinical and radiological relapses (PIRA). The complex system of pathological events driving "chronic" worsening is likely linked with the early accumulation of compartmentalized inflammation within the central nervous system as well as insufficient repair phenomena and mitochondrial failure. These mechanisms are partially lesion-independent and differ from those causing clinical relapses and the formation of new focal demyelinating lesions; they lead to neuroaxonal dysfunction and death, myelin loss, glia alterations, and finally, a neuronal network dysfunction outweighing central nervous system (CNS) compensatory mechanisms. This review aims to provide an overview of the state of the art of neuropathological, immunological, and imaging knowledge about the mechanisms underlying the smoldering disease activity, focusing on possible early biomarkers and their translation into clinical practice. ANN NEUROL 2024;96:1-20.
Collapse
Affiliation(s)
- Massimiliano Calabrese
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Scalfari
- Centre of Neuroscience, Department of Medicine, Imperial College, London, UK
| | - Elisa Colato
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Damiano Marastoni
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Martina Absinta
- Translational Neuropathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Battaglini
- Siena Imaging S.r.l., Siena, Italy
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Owain W Howell
- Institute of Life Sciences, Swansea University Medical School, Swansea, UK
| | - Matilde Inglese
- Dipartimento di neuroscienze, riabilitazione, oftalmologia, genetica e scienze materno-infantili - DINOGMI, University of Genova, Genoa, Italy
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Therapeutic Design Unit, Center for Molecular Medicine, Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
- Cellerys AG, Schlieren, Switzerland
| | - Richard Nicholas
- Department of Brain Sciences, Faculty of Medicine, Burlington Danes, Imperial College London, London, UK
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Agnese Tamanti
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Marco Vercellino
- Multiple Sclerosis Center & Neurologia I U, Department of Neuroscience, University Hospital AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Luisa Maria Villar
- Department of Immunology, Ramon y Cajal University Hospital. IRYCIS. REI, Madrid, Spain
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Magliozzi
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| |
Collapse
|
7
|
Marchi L, Mariottini A, Viti V, Bianchi A, Nozzoli C, Repice AM, Boncompagni R, Ginestroni A, Damato V, Barilaro A, Chiti S, Saccardi R, Fainardi E, Massacesi L. Leptomeningeal enhancement in multiple sclerosis: a focus on patients treated with hematopoietic stem cell transplantation. Front Neurol 2024; 15:1373385. [PMID: 38899059 PMCID: PMC11186296 DOI: 10.3389/fneur.2024.1373385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Background Leptomeningeal enhancement (LME) is considered an MRI marker of leptomeningeal inflammation in inflammatory neurological disorders, including multiple sclerosis (MS). To our knowledge, no disease-modifying therapies (DMTs) have been demonstrated to affect LME number or morphology so far. Methods Monocentric study investigating the frequency and number of LME in a cohort of people with (pw)MS who performed a 3 T brain MRI with a standardized protocol (including a post-contrast FLAIR sequence), and exploring the impact of autologous hematopoietic stem cell transplantation (AHSCT) on this marker. In a longitudinal pilot study, consecutive MRIs were also analyzed in a subgroup of pwMS, including patients evaluated both pre- and post-AHSCT. Results Fifty-five pwMS were included: 24/55 (44%) had received AHSCT (AHSCT group) and 31 other treatments (CTRL group). At least one LME was identified in 19/55 (35%) cases (42 and 29% in the AHSCT and CTRL groups, respectively; p = 0.405). In the AHSCT group, LME number correlated with age at AHSCT (R = 0.50; p = 0.014), but not with age at post-treatment MRI. In the longitudinal pilot study (n = 8), one LME disappeared following AHSCT in 1/4 patients, whereas LME number was unchanged in the remaining four pwMS from the CTRL group. Discussion These results suggest that AHSCT may affect development and persistence of LME, strengthening the indication for early use of effective therapies bioavailable within the central nervous system (CNS), and therefore potentially targeting compartmentalized inflammation.
Collapse
Affiliation(s)
- Leonardo Marchi
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
| | - Alice Mariottini
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
- Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Center, Careggi University Hospital, Florence, Italy
| | - Vittorio Viti
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
| | - Andrea Bianchi
- Neuroradiology Unit, Careggi University Hospital, Florence, Italy
| | - Chiara Nozzoli
- Cell Therapy and Transfusion Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Anna Maria Repice
- Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Center, Careggi University Hospital, Florence, Italy
| | - Riccardo Boncompagni
- Cell Therapy and Transfusion Medicine Unit, Careggi University Hospital, Florence, Italy
| | | | - Valentina Damato
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
- Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Center, Careggi University Hospital, Florence, Italy
| | - Alessandro Barilaro
- Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Center, Careggi University Hospital, Florence, Italy
| | - Stefano Chiti
- Health Physics Unit, Careggi University Hospital, Florence, Italy
| | - Riccardo Saccardi
- Cell Therapy and Transfusion Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Enrico Fainardi
- Neuroradiology Unit, Careggi University Hospital, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Luca Massacesi
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
- Department of Neurology 2 and Tuscan Region Multiple Sclerosis Referral Center, Careggi University Hospital, Florence, Italy
| |
Collapse
|
8
|
Bonadio JD, Bashiri G, Halligan P, Kegel M, Ahmed F, Wang K. Delivery technologies for therapeutic targeting of fibronectin in autoimmunity and fibrosis applications. Adv Drug Deliv Rev 2024; 209:115303. [PMID: 38588958 PMCID: PMC11111362 DOI: 10.1016/j.addr.2024.115303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/29/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Fibronectin (FN) is a critical component of the extracellular matrix (ECM) contributing to various physiological processes, including tissue repair and immune response regulation. FN regulates various cellular functions such as adhesion, proliferation, migration, differentiation, and cytokine release. Alterations in FN expression, deposition, and molecular structure can profoundly impact its interaction with other ECM proteins, growth factors, cells, and associated signaling pathways, thus influencing the progress of diseases such as fibrosis and autoimmune disorders. Therefore, developing therapeutics that directly target FN or its interaction with cells and other ECM components can be an intriguing approach to address autoimmune and fibrosis pathogenesis.
Collapse
Affiliation(s)
- Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Fatima Ahmed
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, PA, United States.
| |
Collapse
|
9
|
Nicholas R, Magliozzi R, Marastoni D, Howell O, Roncaroli F, Muraro P, Reynolds R, Friede T. High Levels of Perivascular Inflammation and Active Demyelinating Lesions at Time of Death Associated with Rapidly Progressive Multiple Sclerosis Disease Course: A Retrospective Postmortem Cohort Study. Ann Neurol 2024; 95:706-719. [PMID: 38149648 DOI: 10.1002/ana.26870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 12/23/2023] [Accepted: 12/24/2023] [Indexed: 12/28/2023]
Abstract
OBJECTIVE Analysis of postmortem multiple sclerosis (MS) tissues combined with in vivo disease milestones suggests that whereas perivascular white matter infiltrates are associated with demyelinating activity in the initial stages, leptomeningeal immune cell infiltration, enriched in B cells, and associated cortical lesions contribute to disease progression. We systematically examine the association of inflammatory features and white matter demyelination at postmortem with clinical milestones. METHODS In 269 MS brains, 20 sites were examined using immunohistochemistry for active lesions (ALs) and perivenular inflammation (PVI). In a subset of 22, a detailed count of CD20+ B cells and CD3+ T cells in PVIs was performed. RESULTS ALs were detected in 22%, whereas high levels of PVI were detected in 52% of cases. ALs were present in 35% of cases with high levels of PVI. Shorter time from onset of progression to death was associated with increased prevalence and higher levels of PVI (both p < 0.0001). Shorter time from onset of progression to wheelchair use was associated with higher prevalence of ALs (odds ratio [OR] = 0.921, 95% confidence interval [CI] = 0.858-0.989, p = 0.0230) and higher level of PVI (OR = 0.932, 95% CI = 0.886-0.981, p = 0.0071). High levels of PVI were associated with meningeal inflammation and increased cortical demyelination and significantly higher levels of B lymphocytes within the PVI. INTERPRETATION ALs, a feature of early disease stage, persist up to death in a subgroup with high levels of PVI. These features link to a rapid progressive phase and higher levels of meningeal inflammation and B-cell infiltrates, supporting the hypothesis that chronic inflammation drives progression in MS. ANN NEUROL 2024;95:706-719.
Collapse
Affiliation(s)
- Richard Nicholas
- Imperial College Healthcare NHS Trust, London, UK
- Department of Brain Sciences, UK Multiple Sclerosis Society Tissue Bank, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Roberta Magliozzi
- Department of Brain Sciences, UK Multiple Sclerosis Society Tissue Bank, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Damiano Marastoni
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Owain Howell
- Department of Brain Sciences, UK Multiple Sclerosis Society Tissue Bank, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
- Institute for Life Sciences, Swansea University, Swansea, UK
| | - Federico Roncaroli
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Paolo Muraro
- Department of Brain Sciences, UK Multiple Sclerosis Society Tissue Bank, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Richard Reynolds
- Department of Brain Sciences, UK Multiple Sclerosis Society Tissue Bank, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Tim Friede
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
10
|
Wang PF, Jiang F, Zeng QM, Yin WF, Hu YZ, Li Q, Hu ZL. Mitochondrial and metabolic dysfunction of peripheral immune cells in multiple sclerosis. J Neuroinflammation 2024; 21:28. [PMID: 38243312 PMCID: PMC10799425 DOI: 10.1186/s12974-024-03016-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder characterized by the infiltration of inflammatory cells and demyelination of nerves. Mitochondrial dysfunction has been implicated in the pathogenesis of MS, as studies have shown abnormalities in mitochondrial activities, metabolism, mitochondrial DNA (mtDNA) levels, and mitochondrial morphology in immune cells of individuals with MS. The presence of mitochondrial dysfunctions in immune cells contributes to immunological dysregulation and neurodegeneration in MS. This review provided a comprehensive overview of mitochondrial dysfunction in immune cells associated with MS, focusing on the potential consequences of mitochondrial metabolic reprogramming on immune function. Current challenges and future directions in the field of immune-metabolic MS and its potential as a therapeutic target were also discussed.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, 139 Ren-Min Central Road, Changsha City, 410011, Hunan, China
| | - Fei Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha City, 410011, Hunan, China
| | - Qiu-Ming Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha City, 410011, Hunan, China
| | - Wei-Fan Yin
- Department of Neurology, The Second Xiangya Hospital, Central South University, 139 Ren-Min Central Road, Changsha City, 410011, Hunan, China
| | - Yue-Zi Hu
- Clinical Laboratory, The Second Hospital of Hunan University of Chinese Medicine, 233 Cai' e North Road, Changsha City, 410005, Hunan, China
| | - Qiao Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, 139 Ren-Min Central Road, Changsha City, 410011, Hunan, China
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, 139 Ren-Min Central Road, Changsha City, 410011, Hunan, China.
| |
Collapse
|
11
|
Ahn JJ, Islam Y, Clarkson-Paredes C, Karl MT, Miller RH. B cell depletion modulates glial responses and enhances blood vessel integrity in a model of multiple sclerosis. Neurobiol Dis 2023; 187:106290. [PMID: 37709209 DOI: 10.1016/j.nbd.2023.106290] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023] Open
Abstract
Multiple sclerosis (MS) is characterized by a compromised blood-brain barrier (BBB) resulting in central nervous system (CNS) entry of peripheral lymphocytes, including T cells and B cells. While T cells have largely been considered the main contributors to neuroinflammation in MS, the success of B cell depletion therapies suggests an important role for B cells in MS pathology. Glial cells in the CNS are essential components in both disease progression and recovery, raising the possibility that they represent targets for B cell functions. Here, we examine astrocyte and microglia responses to B cell depleting treatments in an animal model of MS, experimental autoimmune encephalomyelitis (EAE). B cell depleted EAE animals had markedly reduced disease severity and myelin damage accompanied by reduced microglia and astrocyte reactivity 20 days after symptom onset. To identify potential initial mechanisms mediating functional changes following B cell depletion, astrocyte and microglia transcriptomes were analyzed 3 days following B cell depletion. In control EAE animals, transcriptomic analysis revealed astrocytic inflammatory pathways were activated and microglial influence on neuronal function were inhibited. Following B cell depletion, initial functional recovery was associated with an activation of astrocytic pathways linked with restoration of neurovascular integrity and of microglial pathways associated with neuronal function. These studies reveal an important role for B cell depletion in influencing glial function and CNS vasculature in an animal model of MS.
Collapse
Affiliation(s)
- Julie J Ahn
- The George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Ross Hall, 2300 I St NW, Washington, DC 20037, United States of America
| | - Yusra Islam
- The George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Ross Hall, 2300 I St NW, Washington, DC 20037, United States of America
| | - Cheryl Clarkson-Paredes
- The George Washington University School of Medicine and Health Sciences, Nanofabrication and Imaging Center, Science and Engineering Hall, 800 22(nd) St NW, Washington, DC 20037, United States of America
| | - Molly T Karl
- The George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Ross Hall, 2300 I St NW, Washington, DC 20037, United States of America
| | - Robert H Miller
- The George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Ross Hall, 2300 I St NW, Washington, DC 20037, United States of America.
| |
Collapse
|
12
|
Touil H, Li R, Zuroff L, Moore CS, Healy L, Cignarella F, Piccio L, Ludwin S, Prat A, Gommerman J, Bennett FC, Jacobs D, Benjamins JA, Lisak RP, Antel JP, Bar-Or A. Cross-talk between B cells, microglia and macrophages, and implications to central nervous system compartmentalized inflammation and progressive multiple sclerosis. EBioMedicine 2023; 96:104789. [PMID: 37703640 PMCID: PMC10505984 DOI: 10.1016/j.ebiom.2023.104789] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/07/2023] [Accepted: 08/22/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND B cells can be enriched within meningeal immune-cell aggregates of multiple sclerosis (MS) patients, adjacent to subpial cortical demyelinating lesions now recognized as important contributors to progressive disease. This subpial demyelination is notable for a 'surface-in' gradient of neuronal loss and microglial activation, potentially reflecting the effects of soluble factors secreted into the CSF. We previously demonstrated that MS B-cell secreted products are toxic to oligodendrocytes and neurons. The potential for B-cell-myeloid cell interactions to propagate progressive MS is of considerable interest. METHODS Secreted products of MS-implicated pro-inflammatory effector B cells or IL-10-expressing B cells with regulatory potential were applied to human brain-derived microglia or monocyte-derived macrophages, with subsequent assessment of myeloid phenotype and function through measurement of their expression of pro-inflammatory, anti-inflammatory and homeostatic/quiescent molecules, and phagocytosis (using flow cytometry, ELISA and fluorescently-labeled myelin). Effects of secreted products of differentially activated microglia on B-cell survival and activation were further studied. FINDINGS Secreted products of MS-implicated pro-inflammatory B cells (but not IL-10 expressing B cells) substantially induce pro-inflammatory cytokine (IL-12, IL-6, TNFα) expression by both human microglia and macrophage (in a GM-CSF dependent manner), while down-regulating their expression of IL-10 and of quiescence-associated molecules, and suppressing their myelin phagocytosis. In contrast, secreted products of IL-10 expressing B cells upregulate both human microglia and macrophage expression of quiescence-associated molecules and enhance their myelin phagocytosis. Secreted factors from pro-inflammatory microglia enhance B-cell activation. INTERPRETATION Potential cross-talk between disease-relevant human B-cell subsets and both resident CNS microglia and infiltrating macrophages may propagate CNS-compartmentalized inflammation and injury associated with MS disease progression. These interaction represents an attractive therapeutic target for agents such as Bruton's tyrosine kinase inhibitors (BTKi) that modulate responses of both B cells and myeloid cells. FUNDING Stated in Acknowledgments section of manuscript.
Collapse
Affiliation(s)
- Hanane Touil
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Li
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leah Zuroff
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Luke Healy
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University, Canada
| | - Francesca Cignarella
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, USA
| | - Laura Piccio
- Charles Perkins Centre and School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - Samuel Ludwin
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Alexandre Prat
- Université de Montréal Centre de Recherche du CHUM (CRCHUM) and Department of Neuroscience, Université de Montréal, 900 Saint Denis Street, Montréal, QC, H2X 0A9, Canada
| | - Jennifer Gommerman
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Frederick C Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dina Jacobs
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joyce A Benjamins
- Departments of Neurology and Biochemistry, Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert P Lisak
- Departments of Neurology and Biochemistry, Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jack P Antel
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University, Canada
| | - Amit Bar-Or
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Pukoli D, Vécsei L. Smouldering Lesion in MS: Microglia, Lymphocytes and Pathobiochemical Mechanisms. Int J Mol Sci 2023; 24:12631. [PMID: 37628811 PMCID: PMC10454160 DOI: 10.3390/ijms241612631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated, chronic inflammatory, demyelinating, and neurodegenerative disease of the central nervous system (CNS). Immune cell infiltration can lead to permanent activation of macrophages and microglia in the parenchyma, resulting in demyelination and neurodegeneration. Thus, neurodegeneration that begins with acute lymphocytic inflammation may progress to chronic inflammation. This chronic inflammation is thought to underlie the development of so-called smouldering lesions. These lesions evolve from acute inflammatory lesions and are associated with continuous low-grade demyelination and neurodegeneration over many years. Their presence is associated with poor disease prognosis and promotes the transition to progressive MS, which may later manifest clinically as progressive MS when neurodegeneration exceeds the upper limit of functional compensation. In smouldering lesions, in the presence of only moderate inflammatory activity, a toxic environment is clearly identifiable and contributes to the progressive degeneration of neurons, axons, and oligodendrocytes and, thus, to clinical disease progression. In addition to the cells of the immune system, the development of oxidative stress in MS lesions, mitochondrial damage, and hypoxia caused by the resulting energy deficit and iron accumulation are thought to play a role in this process. In addition to classical immune mediators, this chronic toxic environment contains high concentrations of oxidants and iron ions, as well as the excitatory neurotransmitter glutamate. In this review, we will discuss how these pathobiochemical markers and mechanisms, alone or in combination, lead to neuronal, axonal, and glial cell death and ultimately to the process of neuroinflammation and neurodegeneration, and then discuss the concepts and conclusions that emerge from these findings. Understanding the role of these pathobiochemical markers would be important to gain a better insight into the relationship between the clinical classification and the pathomechanism of MS.
Collapse
Affiliation(s)
- Dániel Pukoli
- Department of Neurology, Esztergomi Vaszary Kolos Hospital, 2500 Esztergom, Hungary;
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, University of Szeged, Semmelweis u. 6., H-6725 Szeged, Hungary
- Danube Neuroscience Research Laboratory, ELKH-SZTE Neuroscience Research Group, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| |
Collapse
|
14
|
Magliozzi R, Howell OW, Calabrese M, Reynolds R. Meningeal inflammation as a driver of cortical grey matter pathology and clinical progression in multiple sclerosis. Nat Rev Neurol 2023:10.1038/s41582-023-00838-7. [PMID: 37400550 DOI: 10.1038/s41582-023-00838-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 07/05/2023]
Abstract
Growing evidence from cerebrospinal fluid samples and post-mortem brain tissue from individuals with multiple sclerosis (MS) and rodent models indicates that the meninges have a key role in the inflammatory and neurodegenerative mechanisms underlying progressive MS pathology. The subarachnoid space and associated perivascular spaces between the membranes of the meninges are the access points for entry of lymphocytes, monocytes and macrophages into the brain parenchyma, and the main route for diffusion of inflammatory and cytotoxic molecules from the cerebrospinal fluid into the brain tissue. In addition, the meningeal spaces act as an exit route for CNS-derived antigens, immune cells and metabolites. A number of studies have demonstrated an association between chronic meningeal inflammation and a more severe clinical course of MS, suggesting that the build-up of immune cell aggregates in the meninges represents a rational target for therapeutic intervention. Therefore, understanding the precise cell and molecular mechanisms, timing and anatomical features involved in the compartmentalization of inflammation within the meningeal spaces in MS is vital. Here, we present a detailed review and discussion of the cellular, molecular and radiological evidence for a role of meningeal inflammation in MS, alongside the clinical and therapeutic implications.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy.
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Owain W Howell
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
- Institute of Life Sciences, Swansea University, Swansea, UK
| | - Massimiliano Calabrese
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Richard Reynolds
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
15
|
Li Y, Noto D, Hoshino Y, Mizuno M, Yoshikawa S, Miyake S. Immunoglobulin directly enhances differentiation of oligodendrocyte-precursor cells and remyelination. Sci Rep 2023; 13:9394. [PMID: 37296298 PMCID: PMC10256778 DOI: 10.1038/s41598-023-36532-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease characterized by multiple lesions in the central nervous system. Although the role of B cells in MS pathogenesis has attracted much attention, but the detailed mechanisms remain unclear. To investigate the effects of B cells on demyelination, we analyzed a cuprizone-induced demyelination model, and found that demyelination was significantly exacerbated in B cell-deficient mice. We next investigated whether immunoglobulin affected the myelin formation process using organotypic brain slice cultures and revealed that remyelination was improved in immunoglobulin-treated groups compared with the control group. Analysis of oligodendrocyte-precursor cell (OPC) monocultures showed that immunoglobulins directly affected on OPCs and promoted their differentiation and myelination. Furthermore, OPCs expressed FcγRI and FcγRIII, two receptors that were revealed to mediate the effects of IgG. To the best of our knowledge, this is the first study to demonstrate that B cells act in an inhibitory manner against cuprizone-induced demyelination, while immunoglobulins enhance remyelination following demyelination. Analysis of the culture system revealed that immunoglobulins directly act on OPCs to promote their differentiation and myelination. Future studies to elucidate the effects of immunoglobulins on OPCs in vivo and the detailed mechanisms of these effects may lead to new treatments for demyelinating diseases.
Collapse
Affiliation(s)
- Yaguang Li
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Daisuke Noto
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yasunobu Hoshino
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Miho Mizuno
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Soichiro Yoshikawa
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
16
|
Wu GF. The cerebrospinal fluid immune cell landscape in animal models of multiple sclerosis. Front Mol Neurosci 2023; 16:1143498. [PMID: 37122618 PMCID: PMC10130411 DOI: 10.3389/fnmol.2023.1143498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/15/2023] [Indexed: 05/02/2023] Open
Abstract
The fluid compartment surrounding the central nervous system (CNS) is a unique source of immune cells capable of reflecting the pathophysiology of neurologic diseases. While human clinical and experimental studies often employ cerebrospinal fluid (CSF) analysis, assessment of CSF in animal models of disease are wholly uncommon, particularly in examining the cellular component. Barriers to routine assessment of CSF in animal models of multiple sclerosis (MS) include limited sample volume, blood contamination, and lack of feasible longitudinal approaches. The few studies characterizing CSF immune cells in animal models of MS are largely outdated, but recent work employing transcriptomics have been used to explore new concepts in CNS inflammation and MS. Absence of extensive CSF data from rodent and other systems has curbed the overall impact of experimental models of MS. Future approaches, including examination of CSF myeloid subsets, single cell transcriptomics incorporating antigen receptor sequencing, and use of diverse animal models, may serve to overcome current limitations and provide critical insights into the pathogenesis of, and therapeutic developments for, MS.
Collapse
Affiliation(s)
- Gregory F. Wu
- Departments of Neurology and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, United States
- Neurology Service, VA St. Louis Health Care System, St. Louis, MO, United States
| |
Collapse
|
17
|
Chen HY, Zhao Y, Xie YZ. Immunosenescence of brain accelerates Alzheimer's disease progression. Rev Neurosci 2023; 34:85-101. [PMID: 35791032 DOI: 10.1515/revneuro-2022-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/04/2022] [Indexed: 01/07/2023]
Abstract
Most of Alzheimer's disease (AD) cases are sporadic and occur after age 65. With prolonged life expectancy and general population aging, AD is becoming a significant public health concern. The immune system supports brain development, plasticity, and homeostasis, yet it is particularly vulnerable to aging-related changes. Aging of the immune system, called immunosenescence, is the multifaceted remodeling of the immune system during aging. Immunosenescence is a contributing factor to various age-related diseases, including AD. Age-related changes in brain immune cell phenotype and function, crosstalk between immune cells and neural cells, and neuroinflammation work together to promote neurodegeneration and age-related cognitive impairment. Although numerous studies have confirmed the correlation between systemic immune changes and AD, few studies focus on the immune state of brain microenvironment in aging and AD. This review mainly addresses the changes of brain immune microenvironment in aging and AD. Specifically, we delineate how various aspects of the brain immune microenvironment, including immune gateways, immune cells, and molecules, and the interplay between immune cells and neural cells, accelerate AD pathogenesis during aging. We also propose a theoretical framework of therapeutic strategies selectively targeting the different mechanisms to restore brain immune homeostasis.
Collapse
Affiliation(s)
- Hou-Yu Chen
- Department of Abdominal Surgery, Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangdong 510095, China
| | - Yan Zhao
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| | - Yong-Zhi Xie
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
18
|
Curran C, Vaitaitis G, Waid D, Volmer T, Alverez E, Wagner DH. Ocrevus reduces TH40 cells, a biomarker of systemic inflammation, in relapsing multiple sclerosis (RMS) and in progressive multiple sclerosis (PMS). J Neuroimmunol 2023; 374:578008. [PMID: 36535240 PMCID: PMC9868100 DOI: 10.1016/j.jneuroim.2022.578008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/16/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Treating MS has been difficult. One successful drug is Ocrelizumab (anti-CD20), used for the chronic relapsing MS (RMS) and the progressive MS (PMS) forms. TH40 cells are pathogenic effector T cells that increase in percentage and numbers during chronic inflammation. Here we show that in the earliest MS course, clinically isolated syndrome (CIS), TH40 cells expand in number. In PMS TH40 cell numbers remain expanded demonstrating sustained chronic inflammation. In RMS TH40 cells were found in CSF and express CD20. Ocrelizumab reduced TH40 cells to healthy control levels in patients. During treatment inflammatory cytokine producing TH40 cells were decreased.
Collapse
Affiliation(s)
- Christian Curran
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Gisela Vaitaitis
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Dan Waid
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Timothy Volmer
- The Department of Neurology, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - Enrique Alverez
- The Department of Neurology, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America
| | - David H Wagner
- The Webb Waring Center and Department of Medicine, The University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd, Aurora, CO 80045, United States of America.
| |
Collapse
|
19
|
Thomas OG, Rickinson A, Palendira U. Epstein-Barr virus and multiple sclerosis: moving from questions of association to questions of mechanism. Clin Transl Immunology 2023; 12:e1451. [PMID: 37206956 PMCID: PMC10191779 DOI: 10.1002/cti2.1451] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/21/2023] Open
Abstract
The link between Epstein-Barr virus (EBV) and multiple sclerosis (MS) has puzzled researchers since it was first discovered over 40 years ago. Until that point, EBV was primarily viewed as a cancer-causing agent, but the culmination of evidence now shows that EBV has a pivotal role in development of MS. Early MS disease is characterised by episodic neuroinflammation and focal lesions in the central nervous system (CNS) that over time develop into progressive neurodegeneration and disability. Risk of MS is vanishingly low in EBV seronegative individuals, history of infectious mononucleosis (acute symptomatic primary infection with EBV) significantly increases risk and elevated antibody titres directed against EBV antigens are well-characterised in patients. However, the underlying mechanism - or mechanisms - responsible for this interplay remains to be fully elucidated; how does EBV-induced immune dysregulation either trigger or drive MS in susceptible individuals? Furthermore, deep understanding of virological and immunological events during primary infection and long-term persistence in B cells will help to answer the many questions that remain regarding MS pathogenesis. This review discusses the current evidence and mechanisms surrounding EBV and MS, which have important implications for the future of MS therapies and prevention.
Collapse
Affiliation(s)
- Olivia G Thomas
- Department of Clinical Neuroscience, Therapeutic Immune Design, Centre for Molecular MedicineKarolinska InstituteStockholmSweden
| | - Alan Rickinson
- Institute of Cancer and Genomic Sciences, College of Medical and Dental SciencesUniversity of Birmingham, EdgbastonBirminghamUK
| | - Umaimainthan Palendira
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneyCamperdownNSWAustralia
- Charles Perkins CentreThe University of SydneyCamperdownNSWAustralia
| |
Collapse
|
20
|
Attfield KE, Jensen LT, Kaufmann M, Friese MA, Fugger L. The immunology of multiple sclerosis. Nat Rev Immunol 2022; 22:734-750. [PMID: 35508809 DOI: 10.1038/s41577-022-00718-z] [Citation(s) in RCA: 128] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Our incomplete understanding of the causes and pathways involved in the onset and progression of multiple sclerosis (MS) limits our ability to effectively treat this complex neurological disease. Recent studies explore the role of immune cells at different stages of MS and how they interact with cells of the central nervous system (CNS). The findings presented here begin to question the exclusivity of an antigen-specific cause and highlight how seemingly distinct immune cell types can share common functions that drive disease. Innovative techniques further expose new disease-associated immune cell populations and reinforce how environmental context is critical to their phenotype and subsequent role in disease. Importantly, the differentiation of immune cells into a pathogenic state is potentially reversible through therapeutic manipulation. As such, understanding the mechanisms that provide plasticity to causal cell types is likely key to uncoupling these disease processes and may identify novel therapeutic targets that replace the need for cell ablation.
Collapse
Affiliation(s)
- Kathrine E Attfield
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, UK
| | - Lise Torp Jensen
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Max Kaufmann
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Fugger
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, UK.
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
21
|
Yuan Y, Sun J, You T, Shen W, Xu W, Dong Q, Cui M. Extracellular Vesicle-Based Therapeutics in Neurological Disorders. Pharmaceutics 2022; 14:pharmaceutics14122652. [PMID: 36559145 PMCID: PMC9783774 DOI: 10.3390/pharmaceutics14122652] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/26/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Neurological diseases remain some of the major causes of death and disability in the world. Few types of drugs and insufficient delivery across the blood-brain barrier limit the treatment of neurological disorders. The past two decades have seen the rapid development of extracellular vesicle-based therapeutics in many fields. As the physiological and pathophysiological roles of extracellular vesicles are recognized in neurological diseases, they have become promising therapeutics and targets for therapeutic interventions. Moreover, advanced nanomedicine technologies have explored the potential of extracellular vesicles as drug delivery systems in neurological diseases. In this review, we discussed the preclinical strategies for extracellular vesicle-based therapeutics in neurological disorders and the struggles involved in their clinical application.
Collapse
Affiliation(s)
- Yiwen Yuan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Jian Sun
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Tongyao You
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Weiwei Shen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Wenqing Xu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- Correspondence: (Q.D.); (M.C.)
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- Correspondence: (Q.D.); (M.C.)
| |
Collapse
|
22
|
Höftberger R, Lassmann H, Berger T, Reindl M. Pathogenic autoantibodies in multiple sclerosis - from a simple idea to a complex concept. Nat Rev Neurol 2022; 18:681-688. [PMID: 35970870 DOI: 10.1038/s41582-022-00700-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2022] [Indexed: 11/08/2022]
Abstract
The role of autoantibodies in multiple sclerosis (MS) has been enigmatic since the first description, many decades ago, of intrathecal immunoglobulin production in people with this condition. Some studies have indicated that MS pathology is heterogeneous, with an antibody-associated subtype - characterized by B cells (in varying quantities), antibodies and complement - existing alongside other subtypes with different pathologies. However, subsequent evidence suggested that some cases originally diagnosed as MS with autoantibody-mediated demyelination were more likely to be neuromyelitis optica spectrum disorder or myelin oligodendrocyte glycoprotein antibody-associated disease. These findings raise the important question of whether an autoantibody-mediated MS subtype exists and whether pathogenic MS-associated autoantibodies remain to be identified. Potential roles of autoantibodies in MS could range from specific antibodies defining the disease to a non-disease-specific amplification of cellular immune responses and other pathophysiological processes. In this Perspective, we review studies that have attempted to identify MS-associated autoantibodies and provide our opinions on their possible roles in the pathophysiology of MS.
Collapse
Affiliation(s)
- Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
23
|
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
|
24
|
Abstract
PURPOSE OF REVIEW This article provides an overview of genetic, environmental, and lifestyle risk factors affecting the disease course of multiple sclerosis (MS) and reviews the pathophysiologic characteristics of both relapsing and progressive MS. RECENT FINDINGS The prevalence of MS has increased in recent decades, and costs of care for patients with MS have risen dramatically. Black, Asian, and Hispanic individuals may be at risk for more severe MS-related disability. Multiple genetic MS risk factors have been identified. Factors such as low vitamin D levels and a history of Epstein-Barr virus, smoking, and obesity, especially during childhood, also influence MS risk. Traditionally thought to be a T-cell-mediated disease, recent research has highlighted the additional roles of B cells and microglia in both relapsing and progressive MS. SUMMARY Complex interactions between genetic, environmental, and lifestyle factors affect the risk for MS as well as the disease course. People of color have historically been underrepresented in both MS clinical trials and literature, but current research is attempting to better clarify unique considerations in these groups. MS pathology consists of the focal inflammatory lesions that have been well characterized in relapsing MS, as well as a more widespread neurodegenerative component that is posited to drive progressive disease. Recent advances in characterization of both the inflammatory and neurodegenerative aspects of MS pathophysiology have yielded potential targets for future therapeutic options.
Collapse
|
25
|
Kölliker Frers RA, Otero-Losada M, Kobiec T, Udovin LD, Aon Bertolino ML, Herrera MI, Capani F. Multidimensional overview of neurofilament light chain contribution to comprehensively understanding multiple sclerosis. Front Immunol 2022; 13:912005. [PMID: 35967312 PMCID: PMC9368191 DOI: 10.3389/fimmu.2022.912005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory neurodegenerative disease characterized by demyelination, progressive axonal loss, and varying clinical presentations. Axonal damage associated with the inflammatory process causes neurofilaments, the major neuron structural proteins, to be released into the extracellular space, reaching the cerebrospinal fluid (CSF) and the peripheral blood. Methodological advances in neurofilaments’ serological detection and imaging technology, along with many clinical and therapeutic studies in the last years, have deepened our understanding of MS immunopathogenesis. This review examines the use of light chain neurofilaments (NFLs) as peripheral MS biomarkers in light of the current clinical and therapeutic evidence, MS immunopathology, and technological advances in diagnostic tools. It aims to highlight NFL multidimensional value as a reliable MS biomarker with a diagnostic-prognostic profile while improving our comprehension of inflammatory neurodegenerative processes, mainly RRMS, the most frequent clinical presentation of MS.
Collapse
Affiliation(s)
- Rodolfo A. Kölliker Frers
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
- Unidad de Parasitología, Hospital J. M. Ramos Mejía, Buenos Aires, Argentina
| | - Matilde Otero-Losada
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
- *Correspondence: Matilde Otero-Losada,
| | - Tamara Kobiec
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Lucas D. Udovin
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
| | - María Laura Aon Bertolino
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
| | - María I. Herrera
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Francisco Capani
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
- Departamento de Biología, Universidad Argentina John Kennedy (UAJK), Buenos Aires, Argentina
| |
Collapse
|
26
|
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
|
27
|
Oh J, Bar-Or A. Emerging therapies to target CNS pathophysiology in multiple sclerosis. Nat Rev Neurol 2022; 18:466-475. [PMID: 35697862 DOI: 10.1038/s41582-022-00675-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2022] [Indexed: 12/13/2022]
Abstract
The rapidly evolving therapeutic landscape of multiple sclerosis (MS) has contributed to paradigm shifts in our understanding of the biological mechanisms that contribute to CNS injury and in treatment philosophies. Opportunities remain to further improve treatment of relapsing-remitting MS, but two major therapeutic gaps are the limiting of progressive disease mechanisms and the repair of CNS injury. In this Review, we provide an overview of selected emerging therapies that predominantly target processes within the CNS that are thought to be involved in limiting non-relapsing, progressive disease injury or promoting tissue repair. Among these, we consider agents that modulate adaptive and innate CNS-compartmentalized inflammation, which can be mediated by infiltrating immune cells and/or resident CNS cells, including microglia and astrocytes. We also discuss agents that target degenerative disease mechanisms, agents that might confer neuroprotection, and agents that create a more favourable environment for or actively contribute to oligodendrocyte precursor cell differentiation, remyelination and axonal regeneration. We focus on agents that are novel for MS, that are known to or are presumed to penetrate the CNS, and that have already entered early stages of development in MS clinical trials.
Collapse
Affiliation(s)
- Jiwon Oh
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Ontario, Canada.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics, and Multiple Sclerosis Division, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Tout I, Miossec P. The role of B cells and their interactions with stromal cells in the context of inflammatory autoimmune diseases. Clin Exp Rheumatol 2022; 21:103098. [PMID: 35417796 DOI: 10.1016/j.autrev.2022.103098] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/08/2022] [Indexed: 02/07/2023]
Abstract
Interactions between B cells and stromal cells have essential functions in immune cell development and responses. During chronic inflammation, the pro-inflammatory microenvironment leads to changes in stromal cells, which acquire a pathogenic phenotype specific to each organ and disease. B cells are recruited to the site of inflammation and interact with these pathogenic stromal cells contributing to the disease's severity. In addition to producing autoantibodies, B cells contribute to the pathogenesis of autoimmune inflammatory diseases by serving as professional antigen-presenting cells, producing cytokines, and through additional mechanisms. This review describes the role of B cells and their interactions with stromal cells in chronic inflammation, with a focus on human disease, using three selected autoimmune inflammatory diseases: rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis. Understanding B cells roles and their interaction with stromal cells will help develop new therapeutic options for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Issam Tout
- Department of Clinical Immunology and Rheumatology, Immunogenomics and Inflammation Research Unit, University of Lyon, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437 Lyon, France
| | - Pierre Miossec
- Department of Clinical Immunology and Rheumatology, Immunogenomics and Inflammation Research Unit, University of Lyon, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437 Lyon, France.
| |
Collapse
|
29
|
Effects of a Fully Humanized Type II Anti-CD20 Monoclonal Antibody on Peripheral and CNS B Cells in a Transgenic Mouse Model of Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23063172. [PMID: 35328594 PMCID: PMC8949956 DOI: 10.3390/ijms23063172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/09/2022] [Indexed: 11/17/2022] Open
Abstract
Successful therapy with anti-CD20 monoclonal antibodies (mAbs) has reinforced the key role of B cells in the immunopathology of multiple sclerosis (MS). This study aimed to determine the effects of a novel class of anti-CD20 mAbs on vascular and extravascular central nervous system (CNS)-infiltrating B cells in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Male hCD20xhIgR3 mice and wild-type C57BL/6 (B6) mice were immunized with human myelin oligodendrocyte glycoprotein (MOG)1–125 to induce EAE. While hCD20xhIgR3 mice were injected intravenously with an anti-human CD20 mAb (5 mg/kg) (rituximab (a type I anti-CD20 mAb) or obinutuzumab (a type II anti-CD20 mAb), B6 mice received the anti-mouse CD20 antibody 18B12. Neither mAb affected clinical disease or serum antibody levels. Obinutuzumab and rituximab had an impact on splenic and CNS-infiltrated B cells with slightly differential depletion efficacy. Additionally, obinutuzumab had beneficial effects on spinal cord myelination. B cell depletion rates in the 18B12/B6 model were comparable with those observed in obinutuzumab-treated hCD20xhIgR3 mice. Our results demonstrate the usefulness of anti-CD20 mAbs for the modulation of B cell-driven peripheral immune response and CNS pathology, with type II antibodies potentially being superior to type I in the depletion of tissue-infiltrating B cells.
Collapse
|
30
|
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
|
31
|
Pachner AR. The Neuroimmunology of Multiple Sclerosis: Fictions and Facts. Front Neurol 2022; 12:796378. [PMID: 35197914 PMCID: PMC8858985 DOI: 10.3389/fneur.2021.796378] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
There have been tremendous advances in the neuroimmunology of multiple sclerosis over the past five decades, which have led to improved diagnosis and therapy in the clinic. However, further advances must take into account an understanding of some of the complex issues in the field, particularly an appreciation of "facts" and "fiction." Not surprisingly given the incredible complexity of both the nervous and immune systems, our understanding of the basic biology of the disease is very incomplete. This lack of understanding has led to many controversies in the field. This review identifies some of these controversies and facts/fictions with relation to the basic neuroimmunology of the disease (cells and molecules), and important clinical issues. Fortunately, the field is in a healthy transition from excessive reliance on animal models to a broader understanding of the disease in humans, which will likely lead to many improved treatments especially of the neurodegeneration in multiple sclerosis (MS).
Collapse
Affiliation(s)
- Andrew R. Pachner
- Dartmouth–Hitchcock Medical Center, Lebanon, NH, United States
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
32
|
Boldrini VO, Marques AM, Quintiliano RPS, Moraes AS, Stella CRAV, Longhini ALF, Santos I, Andrade M, Ferrari B, Damasceno A, Carneiro RPD, Brandão CO, Farias AS, Santos LMB. Cytotoxic B Cells in Relapsing-Remitting Multiple Sclerosis Patients. Front Immunol 2022; 13:750660. [PMID: 35197967 PMCID: PMC8859463 DOI: 10.3389/fimmu.2022.750660] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/13/2022] [Indexed: 12/02/2022] Open
Abstract
Background Emerging evidence of antibody-independent functions, as well as the clinical efficacy of anti-CD20 depleting therapies, helped to reassess the contribution of B cells during multiple sclerosis (MS) pathogenesis. Objective To investigate whether CD19+ B cells may share expression of the serine-protease granzyme-B (GzmB), resembling classical cytotoxic CD8+ T lymphocytes, in the peripheral blood from relapsing-remitting MS (RRMS) patients. Methods In this study, 104 RRMS patients during different treatments and 58 healthy donors were included. CD8, CD19, Runx3, and GzmB expression was assessed by flow cytometry analyses. Results RRMS patients during fingolimod (FTY) and natalizumab (NTZ) treatment showed increased percentage of circulating CD8+GzmB+ T lymphocytes when compared to healthy volunteers. An increase in circulating CD19+GzmB+ B cells was observed in RRMS patients during FTY and NTZ therapies when compared to glatiramer (GA), untreated RRMS patients, and healthy donors but not when compared to interferon-β (IFN). Moreover, regarding Runx3, the transcriptional factor classically associated with cytotoxicity in CD8+ T lymphocytes, the expression of GzmB was significantly higher in CD19+Runx3+-expressing B cells when compared to CD19+Runx3- counterparts in RRMS patients. Conclusions CD19+ B cells may exhibit cytotoxic behavior resembling CD8+ T lymphocytes in MS patients during different treatments. In the future, monitoring “cytotoxic” subsets might become an accessible marker for investigating MS pathophysiology and even for the development of new therapeutic interventions.
Collapse
Affiliation(s)
- Vinícius O. Boldrini
- Autoimmune Research Laboratory, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- *Correspondence: Vinícius O. Boldrini, ; Alessandro S. Farias, ; Leonilda M. B. Santos,
| | - Ana M. Marques
- Autoimmune Research Laboratory, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Raphael P. S. Quintiliano
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Adriel S. Moraes
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Carla R. A. V. Stella
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Department of Neurology, University of Campinas, Campinas, Brazil
| | - Ana Leda F. Longhini
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Department of Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Irene Santos
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Marília Andrade
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Breno Ferrari
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | | | - Rafael P. D. Carneiro
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- MS Clinic of Santa Casa de São Paulo (CATEM), Irmandade da Santa Casa de Misericordia de São Paulo, São Paulo, Brazil
| | - Carlos Otávio Brandão
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Department of Neurology, University of Campinas, Campinas, Brazil
| | - Alessandro S. Farias
- Autoimmune Research Laboratory, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
- Experimental Medicine Research Cluster (EMRC), São Paulo, Brazil
- *Correspondence: Vinícius O. Boldrini, ; Alessandro S. Farias, ; Leonilda M. B. Santos,
| | - Leonilda M. B. Santos
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
- *Correspondence: Vinícius O. Boldrini, ; Alessandro S. Farias, ; Leonilda M. B. Santos,
| |
Collapse
|
33
|
Li H, Chen Y, Niu J, Yi C. New insights into the immunologic role of oligodendrocyte lineage cells in demyelination diseases. J Biomed Res 2022; 36:343-352. [PMID: 35578762 PMCID: PMC9548433 DOI: 10.7555/jbr.36.20220016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Oligodendrocyte lineage cells (OL-lineage cells) are a cell population that are crucial for mammalian central nervous system (CNS) myelination. OL-lineage cells go through developmental stages, initially differentiating into oligodendrocyte precursor cells (OPCs), before becoming immature oligodendrocytes, then mature oligodendrocytes (OLs). While the main function of cell lineage is in myelin formation, and increasing number of studies have turned to explore the immunological characteristics of these cells. Initially, these studies focused on discovering how OPCs and OLs are affected by the immune system, and then, how these immunological changes influence the myelination process. However, recent studies have uncovered another feature of OL-lineage cells in our immune systems. It would appear that OL-lineage cells also express immunological factors such as cytokines and chemokines in response to immune activation, and the expression of these factors changes under various pathologic conditions. Evidence suggests that OL-lineage cells actually modulate immune functions. Indeed, OL-lineage cells appear to play both "victim" and "agent" in the CNS which raises a number of questions. Here, we summarize immunologic changes in OL-lineage cells and their effects, as well as consider OL-lineage cell changes which influence immune cells under pathological conditions. We also describe some of the underlying mechanisms of these changes and their effects. Finally, we describe several studies which use OL-lineage cells as immunotherapeutic targets for demyelination diseases.
Collapse
Affiliation(s)
- Hui Li
- Research Centre, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Yang Chen
- Research Centre, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Jianqin Niu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
- Jianqin Niu, Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Gaotanyan Main street, Chongqing 400038, China. Tel: +86-13668016001, E-mail:
| | - Chenju Yi
- Research Centre, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
- Chenju Yi, Research Centre, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Guangming (New) District, Shenzhen 518107, China. Tel: +86-13419189905, E-mail:
| |
Collapse
|
34
|
Chisari CG, Sgarlata E, Arena S, Toscano S, Luca M, Patti F. Rituximab for the treatment of multiple sclerosis: a review. J Neurol 2022; 269:159-183. [PMID: 33416999 PMCID: PMC7790722 DOI: 10.1007/s00415-020-10362-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 01/07/2023]
Abstract
In the last decades, evidence suggesting the direct or indirect involvement of B cells on multiple sclerosis (MS) pathogenesis has accumulated. The increased amount of data on the efficacy and safety of B-cell-depleting therapies from several studies has suggested the addition of these drugs as treatment options to the current armamentarium of disease modifying therapies (DMTs) for MS. Particularly, rituximab (RTX), a chimeric monoclonal antibody directed at CD20 positive B lymphocytes resulting in cell-mediated apoptosis, has been demonstrated to reduce inflammatory activity, incidence of relapses and new brain lesions on magnetic resonance imaging (MRI) in patients with relapsing-remitting MS (RRMS). Additional evidence also demonstrated that patients with progressive MS (PMS) may benefit from RTX, which also showed to be well tolerated, with acceptable safety risks and favorable cost-effectiveness profile.Despite these encouraging results, RTX is currently approved for non-Hodgkin's lymphoma, chronic lymphocytic leukemia, several forms of vasculitis and rheumatoid arthritis, while it can only be administered off-label for MS treatment. Between Northern European countries exist different rules for using not licensed drug for treating MS. The Sweden MS register reports a high rate (53.5%) of off-label RTX prescriptions in relation to other annually started DMTs to treat MS patients, while Danish and Norwegian neurologists have to use other anti-CD20 drugs, as ocrelizumab, in most of the cases.In this paper, we review the pharmacokinetics, pharmacodynamics, clinical efficacy, safety profile and cost effectiveness aspects of RTX for the treatment of MS. Particularly, with the approval of new anti-CD20 DMTs, the recent worldwide COVID-19 emergency and the possible increased risk of infection with this class of drugs, this review sheds light on the use of RTX as an alternative treatment option for MS management, while commenting the gaps of knowledge regarding this drug.
Collapse
Affiliation(s)
- Clara Grazia Chisari
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Eleonora Sgarlata
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
- Stroke Unit, Department of Medicine, Umberto I Hospital, Siracusa, Italy
| | - Sebastiano Arena
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Simona Toscano
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Maria Luca
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Francesco Patti
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy.
| |
Collapse
|
35
|
Eliseeva D, Zakharova M. Mechanisms of Neurodegeneration in Multiple Sclerosis. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:5-13. [DOI: 10.17116/jnevro20221220725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
36
|
Walo-Delgado PE, Monreal E, Medina S, Quintana E, Sainz de la Maza S, Fernández-Velasco JI, Lapuente P, Comabella M, Ramió-Torrentà L, Montalban X, Midaglia L, Villarrubia N, Carrasco-Sayalero A, Rodríguez-Martín E, Roldán E, Meca-Lallana J, Alvarez-Lafuente R, Masjuan J, Costa-Frossard L, Villar LM. Role of B Cell Profile for Predicting Secondary Autoimmunity in Patients Treated With Alemtuzumab. Front Immunol 2021; 12:760546. [PMID: 34691084 PMCID: PMC8531491 DOI: 10.3389/fimmu.2021.760546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/21/2021] [Indexed: 11/27/2022] Open
Abstract
Objective To explore if baseline blood lymphocyte profile could identify relapsing remitting multiple sclerosis (RRMS) patients at higher risk of developing secondary autoimmune adverse events (AIAEs) after alemtuzumab treatment. Methods Multicenter prospective study including 57 RRMS patients treated with alemtuzumab followed for 3.25 [3.5-4.21] years, (median [interquartile range]). Blood samples were collected at baseline, and leukocyte subsets determined by flow cytometry. We had additional samples one year after the first cycle of alemtuzumab treatment in 39 cases. Results Twenty-two patients (38.6%) developed AIAEs during follow-up. They had higher B-cell percentages at baseline (p=0.0014), being differences mainly due to plasmablasts/plasma cells (PB/PC, p=0.0011). Those with no AIAEs had higher percentages of CD4+ T cells (p=0.013), mainly due to terminally differentiated (TD) (p=0.034) and effector memory (EM) (p=0.031) phenotypes. AIAEs- patients also showed higher values of TNF-alpha-producing CD8+ T cells (p=0.029). The percentage of PB/PC was the best variable to differentiate both groups of patients. Baseline values >0.10% closely associated with higher AIAE risk (Odds ratio [OR]: 5.91, 95% CI: 1.83-19.10, p=0.004). When excluding the 12 patients with natalizumab, which decreases blood PB/PC percentages, being the last treatment before alemtuzumab, baseline PB/PC >0.1% even predicted more accurately the risk of AIAEs (OR: 11.67, 95% CI: 2.62-51.89, p=0.0007). The AIAEs+ group continued having high percentages of PB/PC after a year of alemtuzumab treatment (p=0.0058). Conclusions A PB/PC percentage <0.1% at baseline identifies MS patients at low risk of secondary autoimmunity during alemtuzumab treatment.
Collapse
Affiliation(s)
- Paulette Esperanza Walo-Delgado
- Department of Immunology, Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Enric Monreal
- Department of Neurology, Ramón y Cajal University Hospital, IRYCIS, Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Silvia Medina
- Department of Immunology, Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Ester Quintana
- Neuroimmunology and Multiple Sclerosis Unit, Neurology Department, Neurodegeneration and Neuroinflammation Research Group, Biomedical Research Institute (IDIBGI), Red Española de Esclerosis Múltiple (REEM), Girona, Spain
| | - Susana Sainz de la Maza
- Department of Neurology, Ramón y Cajal University Hospital, IRYCIS, Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - José Ignacio Fernández-Velasco
- Department of Immunology, Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Paloma Lapuente
- Department of Immunology, Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain
| | - Lluis Ramió-Torrentà
- Neuroimmunology and Multiple Sclerosis Unit, Neurology Department, Neurodegeneration and Neuroinflammation Research Group, Biomedical Research Institute (IDIBGI), Red Española de Esclerosis Múltiple (REEM), Girona, Spain
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain
| | - Luciana Midaglia
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain
| | - Noelia Villarrubia
- Department of Immunology, Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Angela Carrasco-Sayalero
- Department of Immunology, Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Eulalia Rodríguez-Martín
- Department of Immunology, Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Ernesto Roldán
- Department of Immunology, Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - José Meca-Lallana
- Department of Neurology, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Roberto Alvarez-Lafuente
- Grupo de Investigación de Factores Ambientales en Enfermedades Degenerativas, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Hospital Clínico San Carlos, Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Jaime Masjuan
- Department of Neurology, Ramón y Cajal University Hospital, IRYCIS, Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Lucienne Costa-Frossard
- Department of Neurology, Ramón y Cajal University Hospital, IRYCIS, Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| | - Luisa Maria Villar
- Department of Immunology, Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Madrid, Spain
| |
Collapse
|
37
|
Couloume L, Michel L. New concepts on immunology of Multiple Sclerosis. Presse Med 2021; 50:104072. [PMID: 34547375 DOI: 10.1016/j.lpm.2021.104072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/15/2021] [Accepted: 09/14/2021] [Indexed: 12/27/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and immune-driven demyelinating disease of the central nervous system (CNS). During the past decade, major advances have been made to understand the development of MS as well as its progressive stage. Here, we discuss some emerging concepts on immunology of MS, including the growing interest in the involvement of gut microbiota and the recent pathological concepts on the progression phase. Finally, we present some immuno-tools recently available that contribute to better understand diversity and function of the immune system.
Collapse
Affiliation(s)
| | - Laure Michel
- Univ Rennes, CHU Rennes, Neurology, Inserm, CIC 1414 (Centre d'Investigation Clinique de Rennes), F-35000 Rennes, France; Unité Mixte de Recherche (UMR) S1236, INSERM, University of Rennes, Etablissement Français du Sang, Rennes, France; Suivi Immunologique des Thérapeutiques Innovantes, Centre Hospitalier Universitaire de Rennes, Etablissement Français du Sang, Rennes, France.
| |
Collapse
|
38
|
Immune cell compartmentalization for brain surveillance and protection. Nat Immunol 2021; 22:1083-1092. [PMID: 34429552 DOI: 10.1038/s41590-021-00994-2] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023]
Abstract
For decades, it was commonly accepted that the brain is secluded from peripheral immune activity and is self-sufficient for its maintenance and repair. This simplistic perception was based on the presence of resident immune cells, the microglia, and barrier systems within the brain, and the assumption that the central nervous system (CNS) lacks lymphatic drainage. This view was revised with the discoveries that higher functions of the CNS, homeostasis and repair are supported by peripheral innate and adaptive immune cells. The findings of bone marrow-derived immune cells in specialized niches, and the renewed observation that a lymphatic drainage system exists within the brain, further contributed to this revised model. In this Review, we describe the immune niches within the brain, the contribution of professional immune cells to brain functions, the bidirectional relationships between the CNS and the immune system and the relevance of immune components to brain aging and neurodegenerative diseases.
Collapse
|
39
|
Boyko AN, Smirnova NF, Shchukin IA, Guseva ME, Volkov AI. [Ofatumumab - a new drug for the treatment of multiple sclerosis]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:37-43. [PMID: 34387444 DOI: 10.17116/jnevro202112107237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Recently anti-B-cell therapy has been increasingly integrated into the treatment of multiple sclerosis (MS). This review is devoted to ofatumumab, a new drug of this line. Ofatumumab, an all-human monoclonal antibody used to treat chronic leukemia, binds to a different region than the binding site of other CD20 antibodies, including both a small and large loop in the CD20 receptor structure. This monoclonal antibody provides favorable results for MS by reducing the frequency of exacerbations and the risk of disability progression, significantly more pronounced when compared with teriflunomide. The drug can be used in patients with active relapsing MS and SPMS with exacerbations, with the ineffectiveness of first-line drugs as one of the options for second-line therapy, in patients with highly active MS, especially with a high risk of PML (transfer from natalizumab), as well as if there are difficulties in organizing intravenous courses in day hospitals (produced as outpatient injections).
Collapse
Affiliation(s)
- A N Boyko
- Pirogov Russian National Research Medical University, Moscow, Russia.,Federal Center for Brain and Neurotechnology of the FMBA, Moscow, Russia
| | - N F Smirnova
- Pirogov Russian National Research Medical University, Moscow, Russia.,Federal Center for Brain and Neurotechnology of the FMBA, Moscow, Russia
| | - I A Shchukin
- Pirogov Russian National Research Medical University, Moscow, Russia.,Federal Center for Brain and Neurotechnology of the FMBA, Moscow, Russia
| | - M E Guseva
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - A I Volkov
- Federal Center for Brain and Neurotechnology of the FMBA, Moscow, Russia
| |
Collapse
|
40
|
Tadayon Zadeh F, Amini H, Habibi S, Shahedi V, Isanejad A, Akbarpour M. The Effects of 8-Week Combined Exercise Training on Inflammatory Markers in Women with Multiple Sclerosis. NEURODEGENER DIS 2021; 20:212-216. [PMID: 34348347 DOI: 10.1159/000518580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/19/2021] [Indexed: 11/19/2022] Open
Abstract
PURPOSE The present study was designed to investigate the effects of 8-week combined endurance, resistance, and balance exercise training on IL-6, CRP, and IL-10 concentrations in women with multiple sclerosis. METHODS Thirty participants with multiple sclerosis (Expanded Disability Status Scale ≤6) were randomized into either an exercise and control groups. The exercise group performed 8-weeks of endurance, resistance, and balance exercise training. Serum concentrations of IL-6, CRP, and IL-10 were measured before and after the 8-week intervention. Moreover, anthropometric measures were determined at the onset of and after the intervention. For within- and between groups comparisons of all variables, t test (independent and dependent) was used (p < 0.05). RESULTS The results revealed that IL-6 and CRP levels significantly decreased after exercise training (from 6.8 ± 1.52 to 3.2 ± 0.96, p < 0.001 and from 2.76 ± 0.98 to 1.55 ± 0.44, p = <0.001; respectively). Also, exercise training significantly increased IL-10 in the exercise group (from 16.4 ± 2.74 to 23.2 ± 2.11, p < 0.001). There was a significant difference between the 2 groups in all markers in the after 8-week exercise (p < 0.05). CONCLUSIONS One of the characteristics of MS disease is inflammation. Exercise training through physiological mechanisms and without aggravating the inflammatory pathology can be effective in functional and symptom reduction of patients with MS. In confirmation of this, the present study showed that 8 weeks of combined exercise training decreased pro-inflammatory markers (IL-6 and CRP) and increased anti-inflammatory cytokine (IL-10). Our findings suggested that an exercise training program can be an effective strategy for managing the immune system of women with MS at least by its significant effect on inflammatory markers.
Collapse
Affiliation(s)
- Fahime Tadayon Zadeh
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Hamid Amini
- Department of Physical Education & Sport Sciences, Tolou-e-Mehr Non-profit Institute of Higher Education, Qom, Iran
| | - Saeed Habibi
- Physical Education Faculty, Khorasgan Branch, Islamic Azad University, Isfahan, Iran
| | - Valiallah Shahedi
- Department of Physical Education and Sport Sciences, Parand Branch, Islamic Azad University, Parand, Iran
| | - Amin Isanejad
- Department of Physical Education & Sport Sciences, Faculty of Humanities, Shahed University, Tehran, Iran
| | - Mohsen Akbarpour
- Department of Physical Education, Faculty of Literature and Humanities, University of Qom, Qom, Iran
| |
Collapse
|
41
|
B cell depletion changes the immune cell profile in multiple sclerosis patients: One-year report. J Neuroimmunol 2021; 359:577676. [PMID: 34364105 DOI: 10.1016/j.jneuroim.2021.577676] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 01/02/2023]
Abstract
B cell depletion therapy has been shown to be beneficial in multiple sclerosis (MS). However, the mechanism by which B cell depletion mediates its beneficial effects in MS is still unclear. To better understand how B cell depletion may benefit patients with a disease previously thought to be primarily mediated by CD4 T cells, immune profiles were monitored in 48 patients in a phase II trial of ublituximab, a glycoengineered CD20 monoclonal antibody, at 18 time points over a year. As we previously described there was a significant shift in the percentages of T cells, NK cells, and myeloid cells following the initial dose of ublituximab, but this shift normalized within a week and these populations remained stable for the duration of the study. However, T cell subsets changed with an increase in the percentage of naïve CD4 and CD8 T cells and a decline in memory T cells. Importantly, the percentage of Th1 and CD4+GM-CSF+ T cells decreased, while the percentage of Tregs continued to increase over the year. Ublituximab not only depleted CD20+ B cells, but also CD20+ T cells. The favorable changes in the T cell subsets may contribute to the beneficial effects of B cell depletion therapy.
Collapse
|
42
|
Abstract
Since the initial observation of increased immunoglobulin concentrations in the cerebrospinal fluid of multiple sclerosis (MS) patients in the 1940s, B cells have been considered to participate in the pathology of MS through the production of autoantibodies reactive against central nervous system antigens. However, it is now recognized that B cells contribute to MS relapses via antibody-independent activities, including the presentation of antigens to T cells and the release of pro-inflammatory cytokines. In addition, the recent identification of B cell-rich follicle-like structures in the meninges of progressive MS patients suggests that the pathogenic roles of B cells also exist at the progressive phase of this disease. Recently, large-scale clinical trials have demonstrated the efficacy of B-cell depletion therapy using anti-CD20 antibodies in relapsing as well as primary progressive MS. B-cell depletion therapy has become an essential treatment option for MS based on its unique benefit to risk balance in relapsing MS, and because it is the only drug that has been shown to be effective in primary progressive MS to date.
Collapse
Affiliation(s)
- Yusei Miyazaki
- Department of Neurology, National Hospital Organization Hokkaido Medical Center, Sapporo, Japan
| | - Masaaki Niino
- Department of Clinical Research, National Hospital Organization Hokkaido Medical Center, Sapporo, Japan
| |
Collapse
|
43
|
Podbielska M, O’Keeffe J, Pokryszko-Dragan A. New Insights into Multiple Sclerosis Mechanisms: Lipids on the Track to Control Inflammation and Neurodegeneration. Int J Mol Sci 2021; 22:ijms22147319. [PMID: 34298940 PMCID: PMC8303889 DOI: 10.3390/ijms22147319] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) is a central nervous system disease with complex pathogenesis, including two main processes: immune-mediated inflammatory demyelination and progressive degeneration with axonal loss. Despite recent progress in our understanding and management of MS, availability of sensitive and specific biomarkers for these both processes, as well as neuroprotective therapeutic options targeted at progressive phase of disease, are still being sought. Given their abundance in the myelin sheath, lipids are believed to play a central role in underlying immunopathogenesis in MS and seem to be a promising subject of investigation in this field. On the basis of our previous research and a review of the literature, we discuss the current understanding of lipid-related mechanisms involved in active relapse, remission, and progression of MS. These insights highlight potential usefulness of lipid markers in prediction or monitoring the course of MS, particularly in its progressive stage, still insufficiently addressed. Furthermore, they raise hope for new, effective, and stage-specific treatment options, involving lipids as targets or carriers of therapeutic agents.
Collapse
Affiliation(s)
- Maria Podbielska
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Laboratory of Microbiome Immunobiology, Ludwik Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
- Correspondence: ; Tel.: +48-71-370-9912
| | - Joan O’Keeffe
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science & Computing, Galway-Mayo Institute of Technology, Galway, Ireland;
| | | |
Collapse
|
44
|
Cencioni MT, Mattoscio M, Magliozzi R, Bar-Or A, Muraro PA. B cells in multiple sclerosis - from targeted depletion to immune reconstitution therapies. Nat Rev Neurol 2021; 17:399-414. [PMID: 34075251 DOI: 10.1038/s41582-021-00498-5] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2021] [Indexed: 02/04/2023]
Abstract
Increasing evidence indicates the involvement of B cells in the pathogenesis of multiple sclerosis (MS), but their precise roles are unclear. In this Review, we provide an overview of the development and physiological functions of B cells and the main mechanisms through which B cells are thought to contribute to CNS autoimmunity. In MS, abnormalities of B cell function include pro-inflammatory cytokine production, defective B cell regulatory function and the formation of tertiary lymphoid-like structures in the CNS, which are the likely source of abnormal immunoglobulin production detectable in the cerebrospinal fluid. We also consider the hypothesis that Epstein-Barr virus (EBV) is involved in the B cell overactivation that leads to inflammatory injury to the CNS in MS. We also review the immunological effects - with a focus on the effects on B cell subsets - of several successful therapeutic approaches in MS, including agents that selectively deplete B cells (rituximab, ocrelizumab and ofatumumab), agents that less specifically deplete lymphocytes (alemtuzumab and cladribine) and autologous haematopoietic stem cell transplantation, in which the immune system is unselectively ablated and reconstituted. We consider the insights that these effects on B cell populations provide and their potential to further our understanding and targeting of B cells in MS.
Collapse
Affiliation(s)
- Maria T Cencioni
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Miriam Mattoscio
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Roberta Magliozzi
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.,Department of Neurology, University of Verona, Verona, Italy
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paolo A Muraro
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
45
|
Matejuk A, Vandenbark AA, Offner H. Cross-Talk of the CNS With Immune Cells and Functions in Health and Disease. Front Neurol 2021; 12:672455. [PMID: 34135852 PMCID: PMC8200536 DOI: 10.3389/fneur.2021.672455] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
The immune system's role is much more than merely recognizing self vs. non-self and involves maintaining homeostasis and integrity of the organism starting from early development to ensure proper organ function later in life. Unlike other systems, the central nervous system (CNS) is separated from the peripheral immune machinery that, for decades, has been envisioned almost entirely as detrimental to the nervous system. New research changes this view and shows that blood-borne immune cells (both adaptive and innate) can provide homeostatic support to the CNS via neuroimmune communication. Neurodegeneration is mostly viewed through the lens of the resident brain immune populations with little attention to peripheral circulation. For example, cognition declines with impairment of peripheral adaptive immunity but not with the removal of microglia. Therapeutic failures of agents targeting the neuroinflammation framework (inhibiting immune response), especially in neurodegenerative disorders, call for a reconsideration of immune response contributions. It is crucial to understand cross-talk between the CNS and the immune system in health and disease to decipher neurodestructive and neuroprotective immune mechanisms for more efficient therapeutic strategies.
Collapse
Affiliation(s)
- Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, Zielona Góra, Poland
| | - Arthur A Vandenbark
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, United States.,Department of Neurology, Oregon Health and Science University, Portland, OR, United States.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States
| | - Halina Offner
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, United States.,Department of Neurology, Oregon Health and Science University, Portland, OR, United States.,Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
46
|
De Meo E, Storelli L, Moiola L, Ghezzi A, Veggiotti P, Filippi M, Rocca MA. In vivo gradients of thalamic damage in paediatric multiple sclerosis: a window into pathology. Brain 2021; 144:186-197. [PMID: 33221873 DOI: 10.1093/brain/awaa379] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/12/2020] [Accepted: 08/21/2020] [Indexed: 01/01/2023] Open
Abstract
The thalamus represents one of the first structures affected by neurodegenerative processes in multiple sclerosis. A greater thalamic volume reduction over time, on its CSF side, has been described in paediatric multiple sclerosis patients. However, its determinants and the underlying pathological changes, likely occurring before this phenomenon becomes measurable, have never been explored. Using a multiparametric magnetic resonance approach, we quantified, in vivo, the different processes that can involve the thalamus in terms of focal lesions, microstructural damage and atrophy in paediatric multiple sclerosis patients and their distribution according to the distance from CSF/thalamus interface and thalamus/white matter interface. In 70 paediatric multiple sclerosis patients and 26 age- and sex-matched healthy controls, we tested for differences in thalamic volume and quantitative MRI metrics-including fractional anisotropy, mean diffusivity and T1/T2-weighted ratio-in the whole thalamus and in thalamic white matter, globally and within concentric bands originating from CSF/thalamus interface. In paediatric multiple sclerosis patients, the relationship of thalamic abnormalities with cortical thickness and white matter lesions was also investigated. Compared to healthy controls, patients had significantly increased fractional anisotropy in whole thalamus (f2 = 0.145; P = 0.03), reduced fractional anisotropy (f2 = 0.219; P = 0.006) and increased mean diffusivity (f2 = 0.178; P = 0.009) in thalamic white matter and a trend towards a reduced thalamic volume (f2 = 0.027; P = 0.058). By segmenting the whole thalamus and thalamic white matter into concentric bands, in paediatric multiple sclerosis we detected significant fractional anisotropy abnormalities in bands nearest to CSF (f2 = 0.208; P = 0.002) and in those closest to white matter (f2 range = 0.183-0.369; P range = 0.010-0.046), while we found significant mean diffusivity (f2 range = 0.101-0.369; P range = 0.018-0.042) and T1/T2-weighted ratio (f2 = 0.773; P = 0.001) abnormalities in thalamic bands closest to CSF. The increase in fractional anisotropy and decrease in mean diffusivity detected at the CSF/thalamus interface correlated with cortical thickness reduction (r range = -0.27-0.34; P range = 0.004-0.028), whereas the increase in fractional anisotropy detected at the thalamus/white matter interface correlated with white matter lesion volumes (r range = 0.24-0.27; P range = 0.006-0.050). Globally, our results support the hypothesis of heterogeneous pathological processes, including retrograde degeneration from white matter lesions and CSF-mediated damage, leading to thalamic microstructural abnormalities, likely preceding macroscopic tissue loss. Assessing thalamic microstructural changes using a multiparametric magnetic resonance approach may represent a target to monitor the efficacy of neuroprotective strategies early in the disease course.
Collapse
Affiliation(s)
- Ermelinda De Meo
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Loredana Storelli
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Moiola
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Ghezzi
- Multiple Sclerosis Center, Ospedale di Gallarate, Gallarate, Italy
| | - Pierangelo Veggiotti
- Paediatric Neurology Unit, V. Buzzi Children's Hospital, Milan, Italy.,Biomedical and Clinical Science Department, University of Milan, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurophysiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
47
|
Haase S, Linker RA. Inflammation in multiple sclerosis. Ther Adv Neurol Disord 2021; 14:17562864211007687. [PMID: 33948118 PMCID: PMC8053832 DOI: 10.1177/17562864211007687] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/15/2021] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) that is characterised pathologically by demyelination, gliosis, neuro-axonal damage and inflammation. Despite intense research, the underlying pathomechanisms driving inflammatory demyelination in MS still remain incompletely understood. It is thought to be caused by an autoimmune response towards CNS self-antigens in genetically susceptible individuals, assuming autoreactive T cells as disease-initiating immune cells. Yet, B cells were recognized as crucial immune cells in disease pathology, including antibody-dependent and independent effects. Moreover, myeloid cells are important contributors to MS pathology, and it is becoming increasingly evident that different cell types act in concert during MS immunopathology. This is supported by the finding that the beneficial effects of actual existing disease-modifying therapies cannot be attributed to one single immune cell-type, but rather involve immunological cooperation. The current strategy of MS therapies thus aims to shift the immune cell repertoire from a pro-inflammatory towards an anti-inflammatory phenotype, involving regulatory T and B cells and anti-inflammatory macrophages. Although no existing therapy actually exists that directly induces an enhanced regulatory immune cell pool, numerous studies identified potential net effects on these cell types. This review gives a conceptual overview on T cells, B cells and myeloid cells in the immunopathology of relapsing-remitting MS and discusses potential contributions of actual disease-modifying therapies on these immune cell phenotypes.
Collapse
Affiliation(s)
- Stefanie Haase
- Neuroimmunologie, Klinik und Poliklinik für Neurologie, Universitätsklinik Regensburg, Franz-Josef-Strauss Allee, Regensburg, 93053, Germany
| | - Ralf A Linker
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
48
|
Silva BA, Miglietta E, Ferrari CC. Insights into the role of B cells in the cortical pathology of Multiple sclerosis: evidence from animal models and patients. Mult Scler Relat Disord 2021; 50:102845. [PMID: 33636613 DOI: 10.1016/j.msard.2021.102845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 02/03/2021] [Accepted: 02/13/2021] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated disease of the central nervous system (CNS) that affects both white and gray matter. Although it has been traditionally considered as a T cell mediated disease, the role of B cell in MS pathology has become a topic of great research interest. Cortical lesions, key feature of the progressive forms of MS, are involved in cognitive impairment and worsening of the patients' outcome. These lesions present pathognomonic hallmarks, such as: absence of blood-brain barrier (BBB) disruption, limited inflammatory events, reactive microglia, neurodegeneration, demyelination and meningeal inflammation. B cells located in the meninges, either as part of diffuse inflammation or as part of follicle-like structures, are strongly associated with cortical damage. The function of CD20-expressing B cells in MS is further highlighted by the success of specific therapies using anti-CD20 antibodies. The possible roles of B cells in pathology go beyond their ability to produce antibodies, as they also present antigens to T cells, secrete cytokines (both pathogenic and protective) within the CNS to modulate T and myeloid cell functions, and are involved in meningeal inflammation. Here, we will review the contributions of B cells to the pathogenesis of meningeal inflammation and cortical lesions in MS patients as well as in preclinical animal models.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA, CONICET, Buenos Aires, Argentina; Centro Universitario de Esclerosis Múltiple, División Neurología, Hospital JM Ramos Mejía, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Esteban Miglietta
- Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA, CONICET, Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
49
|
Bakhuraysah MM, Theotokis P, Lee JY, Alrehaili AA, Aui PM, Figgett WA, Azari MF, Abou-Afech JP, Mackay F, Siatskas C, Alderuccio F, Strittmatter SM, Grigoriadis N, Petratos S. B-cells expressing NgR1 and NgR3 are localized to EAE-induced inflammatory infiltrates and are stimulated by BAFF. Sci Rep 2021; 11:2890. [PMID: 33536561 PMCID: PMC7858582 DOI: 10.1038/s41598-021-82346-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 01/14/2021] [Indexed: 02/01/2023] Open
Abstract
We have previously reported evidence that Nogo-A activation of Nogo-receptor 1 (NgR1) can drive axonal dystrophy during the neurological progression of experimental autoimmune encephalomyelitis (EAE). However, the B-cell activating factor (BAFF/BlyS) may also be an important ligand of NgR during neuroinflammation. In the current study we define that NgR1 and its homologs may contribute to immune cell signaling during EAE. Meningeal B-cells expressing NgR1 and NgR3 were identified within the lumbosacral spinal cords of ngr1+/+ EAE-induced mice at clinical score 1. Furthermore, increased secretion of immunoglobulins that bound to central nervous system myelin were shown to be generated from isolated NgR1- and NgR3-expressing B-cells of ngr1+/+ EAE-induced mice. In vitro BAFF stimulation of NgR1- and NgR3-expressing B cells, directed them into the cell cycle DNA synthesis phase. However, when we antagonized BAFF signaling by co-incubation with recombinant BAFF-R, NgR1-Fc, or NgR3 peptides, the B cells remained in the G0/G1 phase. The data suggest that B cells express NgR1 and NgR3 during EAE, being localized to infiltrates of the meninges and that their regulation is governed by BAFF signaling.
Collapse
Affiliation(s)
- Maha M Bakhuraysah
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
- Faculty of Applied Medical Sciences, Taif University, Taif, 26521, Kingdom of Saudi Arabia
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, 54636, Thessaloniki, Macedonia, Greece
| | - Jae Young Lee
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
- Toolgen Inc., Gasan Digital-Ro, 08594, Geumcheon, Seoul, Korea
| | - Amani A Alrehaili
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
- Faculty of Applied Medical Sciences, Taif University, Taif, 26521, Kingdom of Saudi Arabia
| | - Pei-Mun Aui
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - William A Figgett
- Department of Microbiology and Immunology, School of Biomedical Science, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3000, Australia
| | - Michael F Azari
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - John-Paul Abou-Afech
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - Fabienne Mackay
- Department of Microbiology and Immunology, School of Biomedical Science, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3000, Australia
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | | | - Frank Alderuccio
- Department of Immunology and Pathology, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, 54636, Thessaloniki, Macedonia, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia.
| |
Collapse
|
50
|
Ramaglia V, Rojas O, Naouar I, Gommerman JL. The Ins and Outs of Central Nervous System Inflammation-Lessons Learned from Multiple Sclerosis. Annu Rev Immunol 2021; 39:199-226. [PMID: 33524273 DOI: 10.1146/annurev-immunol-093019-124155] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by the inappropriate invasion of lymphocytes and monocytes into the central nervous system (CNS), where they orchestrate the demyelination of axons, leading to physical and cognitive disability. There are many reasons immunologists should be interested in MS. Aside from the fact that there is still significant unmet need for patients living with the progressive form of the disease, MS is a case study for how immune cells cross CNS barriers and subsequently interact with specialized tissue parenchymal cells. In this review, we describe the types of immune cells that infiltrate the CNS and then describe interactions between immune cells and glial cells in different types of lesions. Lastly, we provide evidence for CNS-compartmentalized immune cells and speculate on how this impacts disease progression for MS patients.
Collapse
Affiliation(s)
- Valeria Ramaglia
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Olga Rojas
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Ikbel Naouar
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | | |
Collapse
|