1
|
Gao M, Wang X, Su S, Feng W, Lai Y, Huang K, Cao D, Wang Q. Meningeal lymphatic vessel crosstalk with central nervous system immune cells in aging and neurodegenerative diseases. Neural Regen Res 2025; 20:763-778. [PMID: 38886941 PMCID: PMC11433890 DOI: 10.4103/nrr.nrr-d-23-01595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/22/2023] [Indexed: 06/20/2024] Open
Abstract
Meningeal lymphatic vessels form a relationship between the nervous system and periphery, which is relevant in both health and disease. Meningeal lymphatic vessels not only play a key role in the drainage of brain metabolites but also contribute to antigen delivery and immune cell activation. The advent of novel genomic technologies has enabled rapid progress in the characterization of myeloid and lymphoid cells and their interactions with meningeal lymphatic vessels within the central nervous system. In this review, we provide an overview of the multifaceted roles of meningeal lymphatic vessels within the context of the central nervous system immune network, highlighting recent discoveries on the immunological niche provided by meningeal lymphatic vessels. Furthermore, we delve into the mechanisms of crosstalk between meningeal lymphatic vessels and immune cells in the central nervous system under both homeostatic conditions and neurodegenerative diseases, discussing how these interactions shape the pathological outcomes. Regulation of meningeal lymphatic vessel function and structure can influence lymphatic drainage, cerebrospinal fluid-borne immune modulators, and immune cell populations in aging and neurodegenerative disorders, thereby playing a key role in shaping meningeal and brain parenchyma immunity.
Collapse
Affiliation(s)
- Minghuang Gao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Xinyue Wang
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shijie Su
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Weicheng Feng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Yaona Lai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Kongli Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Dandan Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Yazdani M. Cellular and Molecular Responses to Mitochondrial DNA Deletions in Kearns-Sayre Syndrome: Some Underlying Mechanisms. Mol Neurobiol 2024; 61:5665-5679. [PMID: 38224444 DOI: 10.1007/s12035-024-03938-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Kearns-Sayre syndrome (KSS) is a rare multisystem mitochondrial disorder. It is caused by mitochondrial DNA (mtDNA) rearrangements, mostly large-scale deletions of 1.1-10 kb. These deletions primarily affect energy supply through impaired oxidative phosphorylation and reduced ATP production. This impairment gives rise to dysfunction of several tissues, in particular those with high energy demand like brain and muscles. Over the past decades, changes in respiratory chain complexes and energy metabolism have been emphasized, whereas little attention has been paid to other reports on ROS overproduction, protein synthesis inhibition, myelin vacuolation, demyelination, autophagy, apoptosis, and involvement of lipid raft and oligodendrocytes in KSS. Therefore, this paper draws attention towards these relatively underemphasized findings that might further clarify the pathologic cascades following deletions in the mtDNA.
Collapse
Affiliation(s)
- Mazyar Yazdani
- Department of Medical Biochemistry, Oslo University Hospital, Rikshospitalet, Oslo, 0027, Norway.
| |
Collapse
|
3
|
Alomar HA, Nadeem A, Ansari MA, Attia SM, Bakheet SA, Al-Mazroua HA, Alhazzani K, Assiri MA, Alqinyah M, Almudimeegh S, Ahmad SF. Mitogen-activated protein kinase inhibitor PD98059 improves neuroimmune dysfunction in experimental autoimmune encephalomyelitis in SJL/J mice through the inhibition of nuclear factor-kappa B signaling in B cells. Brain Res Bull 2023; 194:45-53. [PMID: 36646144 DOI: 10.1016/j.brainresbull.2023.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Multiple sclerosis (MS) is a severe autoimmune disease leading to demyelination, followed by consequent axonal degeneration, causing sensory, motor, cognitive, and visual symptoms. Experimental autoimmune encephalomyelitis (EAE) is the most well-studied animal model of MS. Most current MS treatments are not completely effective, and severe side effects remain a great challenge. In this study, we report the therapeutic efficacy of PD98059, a potent mitogen-activated protein kinase inhibitor, on proteolipid protein (PLP)139-151-induced EAE in SJL/J mice. Following the induction of EAE, mice were intraperitoneally treated with PD98059 (5 mg/kg for 14 days) daily from day 14 to day 28. This study investigated the effects of PD98059 on C-C motif chemokine receptor 6 (CCR6), CD14, NF-κB p65, IκBα, GM-CSF, iNOS, IL-6, TNF-α in CD45R+ B lymphocytes using flow cytometry. Furthermore, we analyzed the effect of PD98059 on CCR6, CD14, NF-κB p65, GM-CSF, iNOS, IL-6, and TNF-α mRNA and protein expression levels using qRT-PCR analysis in brain tissues. Mechanistic investigations revealed that PD98059-treated in mice with EAE had reduced CD45R+CCR6+, CD45R+CD14+, CD45R+NF-κB p65+, CD45R+GM-CSF+, CD45R+iNOS+, CD45R+IL-6+, and CD45R+TNF-α+ cells and increased CD45R+IκBα+ cells compared with vehicle-treated control mice in the spleen. Moreover, downregulation of CCR6, CD14, NF-κB p65, GM-CSF, iNOS, IL-6, and TNF-α mRNA expression level was observed in PD98059-treated mice with EAE compared with vehicle-treated control mice in the brain tissue. The results of this study demonstrate that PD98059 modulates inflammatory mediators through multiple cellular mechanisms. The results of this study suggest that PD98059 may be pursued as a therapeutic agent for the treatment of MS.
Collapse
Affiliation(s)
- Hatun A Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sultan Almudimeegh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
4
|
Becktel DA, Zbesko JC, Frye JB, Chung AG, Hayes M, Calderon K, Grover JW, Li A, Garcia FG, Tavera-Garcia MA, Schnellmann RG, Wu HJJ, Nguyen TVV, Doyle KP. Repeated Administration of 2-Hydroxypropyl-β-Cyclodextrin (HPβCD) Attenuates the Chronic Inflammatory Response to Experimental Stroke. J Neurosci 2022; 42:325-348. [PMID: 34819339 PMCID: PMC8802936 DOI: 10.1523/jneurosci.0933-21.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/24/2021] [Accepted: 11/16/2021] [Indexed: 11/30/2022] Open
Abstract
Globally, more than 67 million people are living with the effects of ischemic stroke. Importantly, many stroke survivors develop a chronic inflammatory response that may contribute to cognitive impairment, a common and debilitating sequela of stroke that is insufficiently studied and currently untreatable. 2-Hydroxypropyl-β-cyclodextrin (HPβCD) is an FDA-approved cyclic oligosaccharide that can solubilize and entrap lipophilic substances. The goal of the present study was to determine whether the repeated administration of HPβCD curtails the chronic inflammatory response to stroke by reducing lipid accumulation within stroke infarcts in a distal middle cerebral artery occlusion mouse model of stroke. To achieve this goal, we subcutaneously injected young adult and aged male mice with vehicle or HPβCD 3 times per week, with treatment beginning 1 week after stroke. We evaluated mice at 7 weeks following stroke using immunostaining, RNA sequencing, lipidomic, and behavioral analyses. Chronic stroke infarct and peri-infarct regions of HPβCD-treated mice were characterized by an upregulation of genes involved in lipid metabolism and a downregulation of genes involved in innate and adaptive immunity, reactive astrogliosis, and chemotaxis. Correspondingly, HPβCD reduced the accumulation of lipid droplets, T lymphocytes, B lymphocytes, and plasma cells in stroke infarcts. Repeated administration of HPβCD also preserved NeuN immunoreactivity in the striatum and thalamus and c-Fos immunoreactivity in hippocampal regions. Additionally, HPβCD improved recovery through the protection of hippocampal-dependent spatial working memory and reduction of impulsivity. These results indicate that systemic HPβCD treatment following stroke attenuates chronic inflammation and secondary neurodegeneration and prevents poststroke cognitive decline.SIGNIFICANCE STATEMENT Dementia is a common and debilitating sequela of stroke. Currently, there are no available treatments for poststroke dementia. Our study shows that lipid metabolism is disrupted in chronic stroke infarcts, which causes an accumulation of uncleared lipid debris and correlates with a chronic inflammatory response. To our knowledge, these substantial changes in lipid homeostasis have not been previously recognized or investigated in the context of ischemic stroke. We also provide a proof of principle that solubilizing and entrapping lipophilic substances using HPβCD could be an effective strategy for treating chronic inflammation after stroke and other CNS injuries. We propose that using HPβCD for the prevention of poststroke dementia could improve recovery and increase long-term quality of life in stroke sufferers.
Collapse
Affiliation(s)
- Danielle A Becktel
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Jacob C Zbesko
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Jennifer B Frye
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Amanda G Chung
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Megan Hayes
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Kylie Calderon
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Jeffrey W Grover
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona 85719
| | - Anna Li
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Arizona Arthritis Center, University of Arizona, Tucson, Arizona 85719
| | - Frankie G Garcia
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | | | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85719
| | - Hsin-Jung Joyce Wu
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Arizona Arthritis Center, University of Arizona, Tucson, Arizona 85719
| | - Thuy-Vi V Nguyen
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Department of Neurology, University of Arizona, Tucson, Arizona 85719
| | - Kristian P Doyle
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Department of Neurology, University of Arizona, Tucson, Arizona 85719
- BIO5 Institute, University of Arizona, Tucson, Arizona 85719
- Arizona Center on Aging, University of Arizona, Tucson, Arizona 85719
- Department of Psychology, University of Arizona, Tucson, Arizona 85719
- Department of Neurosurgery, University of Arizona, Tucson, Arizona 85719
| |
Collapse
|
5
|
Prineas JW, Parratt JDE. Multiple Sclerosis: Microglia, Monocytes, and Macrophage-Mediated Demyelination. J Neuropathol Exp Neurol 2021; 80:975-996. [PMID: 34553215 PMCID: PMC8557350 DOI: 10.1093/jnen/nlab083] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This study examined the roles of microglia and monocytes in myelin destruction in patients with early multiple sclerosis (MS). Twenty-two cases were studied; the clinical duration was <9 weeks in 10 cases. Twenty myeloid cell subtypes or categories were identified including 2 cell types not known previously to occur in demyelinating diseases. Commencing myelin breakdown in plaques and in perivascular and subpial tissues occurred in the immediate presence of infiltrating monocytes and was effected by a homogeneous population of IgG-positive Fc receptor-bearing early phagocytes interacting with abnormal myelin. Oligodendrocyte apoptosis was observed in intact myelinated tissue bordering areas of active demyelination. Capillaries in the cerebral cortex plugged by large numbers of monocytes were common in acute cases of MS and in a patient with a neuromyelitis optica variant and extreme systemic recruitment of monocytes. In an MS patient with progressive disease, microglial nodules centered on MHC-II-positive capillaries plugged by monocytes were present in the cerebral cortex. This constitutes a new gray matter lesion in MS.
Collapse
Affiliation(s)
- John W Prineas
- From the Department of Medicine, University of Sydney, Camperdown, NSW, Australia
| | - John D E Parratt
- Department of Neurology, Royal North Shore Hospital, St. Leonards, NSW, Australia
| |
Collapse
|
6
|
Yu X, Zizzo Z, Kennedy PG. An appraisal of antigen identification and IgG effector functions driving host immune responses in multiple sclerosis. Mult Scler Relat Disord 2021; 56:103328. [PMID: 34666240 DOI: 10.1016/j.msard.2021.103328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/05/2021] [Accepted: 10/10/2021] [Indexed: 12/16/2022]
Abstract
Increased immunoglobulin G (IgG) antibodies and oligoclonal bands (OCB) are the most characteristic features of multiple sclerosis (MS), a neuroinflammatory demyelinating disease with neurodegeneration at chronic stages. OCB are shown to be associated with disease activity and brain atrophy. Despite intensive research over the last several decades, the antigen specificities of the IgG in MS have remained elusive. We present evidence which supports that intrathecal IgG is not driven by antigen-stimulation, therefore provide reasoning for failed MS antigen identification. Further, the presence of co-deposition of IgG and activated complement products in MS lesions suggest that the IgG effector functions may play a critical role in disease pathogenesis.
Collapse
Affiliation(s)
- Xiaoli Yu
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America.
| | - Zoe Zizzo
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Peter Ge Kennedy
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| |
Collapse
|
7
|
Macrophages and Autoantibodies in Demyelinating Diseases. Cells 2021; 10:cells10040844. [PMID: 33917929 PMCID: PMC8068327 DOI: 10.3390/cells10040844] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 12/15/2022] Open
Abstract
Myelin phagocytosis by macrophages has been an essential feature of demyelinating diseases in the central and peripheral nervous systems, including Guillain–Barré syndrome (GBS), chronic inflammatory demyelinating polyneuropathy (CIDP), and multiple sclerosis (MS). The discovery of autoantibodies, including anti-ganglioside GM1 antibodies in the axonal form of GBS, anti-neurofascin 155 and anti-contactin 1 antibodies in typical and distal forms of CIDP, and anti-aquaporin 4 antibodies in neuromyelitis optica, contributed to the understanding of the disease process in a subpopulation of patients conventionally diagnosed with demyelinating diseases. However, patients with these antibodies are now considered to have independent disease entities, including acute motor axonal neuropathy, nodopathy or paranodopathy, and neuromyelitis optica spectrum disorder, because primary lesions in these diseases are distinct from those in conventional demyelinating diseases. Therefore, the mechanisms underlying demyelination caused by macrophages remain unclear. Electron microscopy studies revealed that macrophages destroy myelin as if they are the principal players in the demyelination process. Recent studies suggest that macrophages seem to select specific sites of myelinated fibers, including the nodes of Ranvier, paranodes, and internodes, for the initiation of demyelination in individual cases, indicating that specific components localized to these sites play an important role in the behavior of macrophages that initiate myelin phagocytosis. Along with the search for autoantibodies, the ultrastructural characterization of myelin phagocytosis by macrophages is a crucial step in understanding the pathophysiology of demyelinating diseases and for the future development of targeted therapies.
Collapse
|
8
|
Chase Huizar C, Raphael I, Forsthuber TG. Genomic, proteomic, and systems biology approaches in biomarker discovery for multiple sclerosis. Cell Immunol 2020; 358:104219. [PMID: 33039896 PMCID: PMC7927152 DOI: 10.1016/j.cellimm.2020.104219] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is a neuroinflammatory disorder characterized by autoimmune-mediated inflammatory lesions in CNS leading to myelin damage and axonal loss. MS is a heterogenous disease with variable and unpredictable disease course. Due to its complex nature, MS is difficult to diagnose and responses to specific treatments may vary between individuals. Therefore, there is an indisputable need for biomarkers for early diagnosis, prediction of disease exacerbations, monitoring the progression of disease, and for measuring responses to therapy. Genomic and proteomic studies have sought to understand the molecular basis of MS and find biomarker candidates. Advances in next-generation sequencing and mass-spectrometry techniques have yielded an unprecedented amount of genomic and proteomic data; yet, translation of the results into the clinic has been underwhelming. This has prompted the development of novel data science techniques for exploring these large datasets to identify biologically relevant relationships and ultimately point towards useful biomarkers. Herein we discuss optimization of omics study designs, advances in the generation of omics data, and systems biology approaches aimed at improving biomarker discovery and translation to the clinic for MS.
Collapse
Affiliation(s)
- Carol Chase Huizar
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh, UPMC Children's Hospital, Pittsburgh, PA, USA.
| | - Thomas G Forsthuber
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
9
|
Mayne K, White JA, McMurran CE, Rivera FJ, de la Fuente AG. Aging and Neurodegenerative Disease: Is the Adaptive Immune System a Friend or Foe? Front Aging Neurosci 2020; 12:572090. [PMID: 33173502 PMCID: PMC7538701 DOI: 10.3389/fnagi.2020.572090] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases of the central nervous system (CNS) are characterized by progressive neuronal death and neurological dysfunction, leading to increased disability and a loss of cognitive or motor functions. Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis have neurodegeneration as a primary feature. However, in other CNS diseases such as multiple sclerosis, stroke, traumatic brain injury, and spinal cord injury, neurodegeneration follows another insult, such as demyelination or ischaemia. Although there are different primary causes to these diseases, they all share a hallmark of neuroinflammation. Neuroinflammation can occur through the activation of resident immune cells such as microglia, cells of the innate and adaptive peripheral immune system, meningeal inflammation and autoantibodies directed toward components of the CNS. Despite chronic inflammation being pathogenic in these diseases, local inflammation after insult can also promote endogenous regenerative processes in the CNS, which are key to slowing disease progression. The normal aging process in the healthy brain is associated with a decline in physiological function, a steady increase in levels of neuroinflammation, brain shrinkage, and memory deficits. Likewise, aging is also a key contributor to the progression and exacerbation of neurodegenerative diseases. As there are associated co-morbidities within an aging population, pinpointing the precise relationship between aging and neurodegenerative disease progression can be a challenge. The CNS has historically been considered an isolated, "immune privileged" site, however, there is mounting evidence that adaptive immune cells are present in the CNS of both healthy individuals and diseased patients. Adaptive immune cells have also been implicated in both the degeneration and regeneration of the CNS. In this review, we will discuss the key role of the adaptive immune system in CNS degeneration and regeneration, with a focus on how aging influences this crosstalk.
Collapse
Affiliation(s)
- Katie Mayne
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, Belfast, United Kingdom
| | - Jessica A. White
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, Belfast, United Kingdom
| | | | - Francisco J. Rivera
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Alerie G. de la Fuente
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, Belfast, United Kingdom
- *Correspondence: Alerie G. de la Fuente,
| |
Collapse
|
10
|
Ancau M, Berthele A, Hemmer B. CD20 monoclonal antibodies for the treatment of multiple sclerosis: up-to-date. Expert Opin Biol Ther 2019; 19:829-843. [DOI: 10.1080/14712598.2019.1611778] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Mihai Ancau
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, München,
Germany
| | - Achim Berthele
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, München,
Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, München,
Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich,
Germany
| |
Collapse
|
11
|
D'Amico E, Zanghì A, Gastaldi M, Patti F, Zappia M, Franciotta D. Placing CD20-targeted B cell depletion in multiple sclerosis therapeutic scenario: Present and future perspectives. Autoimmun Rev 2019; 18:665-672. [PMID: 31059839 DOI: 10.1016/j.autrev.2019.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 01/18/2019] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis (MS) is an acquired demyelinating disease of the central nervous system (CNS) that traditionally has been considered to be mediated primarily by T cells. Increasing evidence, however, suggests the fundamental role of B cells in the pathogenesis and development of the disease. Recently, anti-CD20 B cell-based therapies have demonstrated impressive and somewhat surprising results in MS, showing profound anti-inflammatory effects with a favorable risk-benefit ratio. Moreover, for the first time in the MS therapeutic scenario, the anti-CD20 monoclonal antibody ocrelizumab has been granted for the treatment of the primary progressive form of the disease. In this review, we provide a brief overview about anti-CD20 B cell-based therapies in MS, in the perspective of their influence on the future management of the disease, and of their possible positioning in a new wider therapeutic scenario.
Collapse
Affiliation(s)
| | - Aurora Zanghì
- Department G.F.Ingrassia, University of Catania, Italy
| | - Matteo Gastaldi
- Neuroimmunology Laboratory, IRCCS Mondino Foundation, Pavia, Italy
| | | | - Mario Zappia
- Department G.F.Ingrassia, University of Catania, Italy
| | - Diego Franciotta
- Neuroimmunology Laboratory, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
12
|
Busto R, Serna J, Perianes-Cachero A, Quintana-Portillo R, García-Seisdedos D, Canfrán-Duque A, Paino CL, Lerma M, Casado ME, Martín-Hidalgo A, Arilla-Ferreiro E, Lasunción MA, Pastor Ó. Ellagic acid protects from myelin-associated sphingolipid loss in experimental autoimmune encephalomyelitis. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:958-967. [DOI: 10.1016/j.bbalip.2018.05.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 05/10/2018] [Accepted: 05/19/2018] [Indexed: 11/29/2022]
|
13
|
Xie YJ, Zhou L, Wang Y, Jiang NW, Cao S, Shao CY, Wang XT, Li XY, Shen Y, Zhou L. Leucine-Rich Glioma Inactivated 1 Promotes Oligodendrocyte Differentiation and Myelination via TSC-mTOR Signaling. Front Mol Neurosci 2018; 11:231. [PMID: 30034322 PMCID: PMC6043672 DOI: 10.3389/fnmol.2018.00231] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/12/2018] [Indexed: 12/26/2022] Open
Abstract
Leucine-rich glioma inactivated 1 (Lgi1), a putative tumor suppressor, is tightly associated with autosomal dominant lateral temporal lobe epilepsy (ADLTE). It has been shown that Lgi1 regulates the myelination of Schwann cells in the peripheral nervous system (PNS). However, the function and underlying mechanisms for Lgi1 regulation of oligodendrocyte differentiation and myelination in the central nervous system (CNS) remain elusive. In addition, whether Lgi1 is required for myelin maintenance is unknown. Here, we show that Lgi1 is necessary and sufficient for the differentiation of oligodendrocyte precursor cells and is also required for the maintenance of myelinated fibers. The hypomyelination in Lgi1-/- mice attributes to the inhibition of the biosynthesis of lipids and proteins in oligodendrocytes (OLs). Moreover, we found that Lgi1 deficiency leads to a decrease in expression of tuberous sclerosis complex 1 (TSC1) and activates mammalian target of rapamycin signaling. Together, the present work establishes that Lgi1 is a regulator of oligodendrocyte development and myelination in CNS.
Collapse
Affiliation(s)
- Ya-Jun Xie
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Lin Zhou
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Yin Wang
- Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical UniversityYinchuan, China
| | - Nan-Wei Jiang
- Ningbo Key Laboratory of Behavioral Neuroscience, Department of Physiology and Pharmacology, Ningbo University School of MedicineNingbo, China
| | - Shenglong Cao
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of MedicineHangzhou, China
| | - Chong-Yu Shao
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Xin-Tai Wang
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Xiang-Yao Li
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Ying Shen
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Liang Zhou
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| |
Collapse
|
14
|
Seil FJ. Myelin Antigens and Antimyelin Antibodies. Antibodies (Basel) 2018; 7:E2. [PMID: 31544855 PMCID: PMC6698820 DOI: 10.3390/antib7010002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 11/16/2022] Open
Abstract
The purpose of this review is to provide an historical perspective on studies of serum derived antimyelin antibodies. Antimyelin antibodies can be defined by their action on myelinating organotypic nervous system tissue cultures and include demyelinating antibodies, which have destructive effects on myelin when applied to already myelinated cultures, and myelination inhibiting antibodies, which prevent myelin formation when applied to cultures prior to myelination. Myelin antigens were evaluated in animal studies for their ability to induce experimental allergic encephalomyelitis, an inflammatory demyelinating disease, and correlated with the induction of antimyelin antibodies. As tissue culture demyelinating activity was also found in sera from some patients with multiple sclerosis, a human inflammatory demyelinating disease, studies were undertaken to characterize the nature of the demyelinating factors.
Collapse
Affiliation(s)
- Fredrick J Seil
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
15
|
Khare P, Challa DK, Devanaboyina SC, Velmurugan R, Hughes S, Greenberg BM, Ober RJ, Ward ES. Myelin oligodendrocyte glycoprotein-specific antibodies from multiple sclerosis patients exacerbate disease in a humanized mouse model. J Autoimmun 2018; 86:104-115. [DOI: 10.1016/j.jaut.2017.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 09/09/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023]
|
16
|
Stimmer L, Fovet CM, Serguera C. Experimental Models of Autoimmune Demyelinating Diseases in Nonhuman Primates. Vet Pathol 2017; 55:27-41. [DOI: 10.1177/0300985817712794] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human idiopathic inflammatory demyelinating diseases (IIDD) are a heterogeneous group of autoimmune inflammatory and demyelinating disorders of the central nervous system (CNS). These include multiple sclerosis (MS), the most common chronic IIDD, but also rarer disorders such as acute disseminated encephalomyelitis (ADEM) and neuromyelitis optica (NMO). Great efforts have been made to understand the pathophysiology of MS, leading to the development of a few effective treatments. Nonetheless, IIDD still require a better understanding of the causes and underlying mechanisms to implement more effective therapies and diagnostic methods. Experimental autoimmune encephalomyelitis (EAE) is a commonly used animal model to study the pathophysiology of IIDD. EAE is principally induced through immunization with myelin antigens combined with immune-activating adjuvants. Nonhuman primates (NHP), the phylogenetically closest relatives of humans, challenged by similar microorganisms as other primates may recapitulate comparable immune responses to that of humans. In this review, the authors describe EAE models in 3 NHP species: rhesus macaques ( Macaca mulatta), cynomolgus macaques ( Macaca fascicularis), and common marmosets ( Callithrix jacchus), evaluating their respective contribution to the understanding of human IIDD. EAE in NHP is a heterogeneous disease, including acute monophasic and chronic polyphasic forms. This diversity makes it a versatile model to use in translational research. This clinical variability also creates an opportunity to explore multiple facets of immune-mediated mechanisms of neuro-inflammation and demyelination as well as intrinsic protective mechanisms. Here, the authors review current insights into the pathogenesis and immunopathological mechanisms implicated in the development of EAE in NHP.
Collapse
Affiliation(s)
- Lev Stimmer
- U1169/US27 Platform for experimental pathology, Molecular Imaging Research Center, INSERM-CEA, Fontenay-aux-Roses, France
| | - Claire-Maëlle Fovet
- U1169/US27 Platform for general surgery, Molecular Imaging Research Center, INSERM-CEA, Fontenay-aux-Roses, France
| | - Ché Serguera
- US27, Molecular Imaging Research Center, INSERM-CEA, Fontenay-aux-Roses, France
| |
Collapse
|
17
|
Tolpeeva OA, Zakharova MN. The diagnostic significance of antibodies to myelin proteins in demyelinating diseases of the central nervous system. NEUROCHEM J+ 2017. [DOI: 10.1134/s1819712417010135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Abstract
BACKGROUND It is uncertain whether there are autoantibodies detectable by indirect immunofluorescence in the serum of patients with multiple sclerosis (MS). OBJECTIVE To determine whether there are anti-central nervous system (CNS) autoantibodies detectable by indirect immunofluorescence in the serum of MS patients. METHODS Sera and in some cases cerebrospinal fluid from 106 patients with multiple sclerosis, 156 patients with other neurological diseases, and 70 healthy control subjects were examined by indirect immunofluorescence using cryostat sections of rat cerebrum fixed by perfusion with paraformaldehyde. RESULTS Autoantibodies were detected that recognized more than 30 neuronal, glial, and mesodermal structures in 28 of 106 MS cases. Most were also detected in patients with other related and unrelated neurological diseases and several were also found in healthy controls. Novel anti-CNS autoantibodies recognizing particular sets of interneurons were detected in both normal controls and in subjects with CNS diseases. INTERPRETATION Serum anti-CNS autoantibodies of diverse specificities are common in MS patients. The same anti-CNS autoantibodies are not uncommon in patients with other neurological diseases. The findings provide no support for the proposition that myelin breakdown in MS is caused by exposure of intact myelin sheaths or oligodendrocytes to a pathogenic serum anti-myelin or anti-oligodendrocyte autoantibody.
Collapse
Affiliation(s)
- John W Prineas
- Brain and Mind Centre, Department of Medicine, The University of Sydney, Camperdown, NSW, Australia
| | - John D E Parratt
- Parratt Brain and Mind Centre, Department of Medicine, The University of Sydney, Camperdown, NSW, Australia and Department of Neurology, Royal North Shore Hospital, St Leonards, NSW, Australia
| |
Collapse
|
19
|
Peschl P, Bradl M, Höftberger R, Berger T, Reindl M. Myelin Oligodendrocyte Glycoprotein: Deciphering a Target in Inflammatory Demyelinating Diseases. Front Immunol 2017; 8:529. [PMID: 28533781 PMCID: PMC5420591 DOI: 10.3389/fimmu.2017.00529] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/19/2017] [Indexed: 12/23/2022] Open
Abstract
Myelin oligodendrocyte glycoprotein (MOG), a member of the immunoglobulin (Ig) superfamily, is a myelin protein solely expressed at the outermost surface of myelin sheaths and oligodendrocyte membranes. This makes MOG a potential target of cellular and humoral immune responses in inflammatory demyelinating diseases. Due to its late postnatal developmental expression, MOG is an important marker for oligodendrocyte maturation. Discovered about 30 years ago, it is one of the best-studied autoantigens for experimental autoimmune models for multiple sclerosis (MS). Human studies, however, have yielded controversial results on the role of MOG, especially MOG antibodies (Abs), as a biomarker in MS. But with improved detection methods using different expression systems to detect Abs in patients' samples, this is meanwhile no longer the case. Using cell-based assays with recombinant full-length, conformationally intact MOG, several recent studies have revealed that MOG Abs can be found in a subset of predominantly pediatric patients with acute disseminated encephalomyelitis (ADEM), aquaporin-4 (AQP4) seronegative neuromyelitis optica spectrum disorders (NMOSD), monophasic or recurrent isolated optic neuritis (ON), or transverse myelitis, in atypical MS and in N-methyl-d-aspartate receptor-encephalitis with overlapping demyelinating syndromes. Whereas MOG Abs are only transiently observed in monophasic diseases such as ADEM and their decline is associated with a favorable outcome, they are persistent in multiphasic ADEM, NMOSD, recurrent ON, or myelitis. Due to distinct clinical features within these diseases it is controversially disputed to classify MOG Ab-positive cases as a new disease entity. Neuropathologically, the presence of MOG Abs is characterized by MS-typical demyelination and oligodendrocyte pathology associated with Abs and complement. However, it remains unclear whether MOG Abs are a mere inflammatory bystander effect or truly pathogenetic. This article provides deeper insight into recent developments, the clinical relevance of MOG Abs and their role in the immunpathogenesis of inflammatory demyelinating disorders.
Collapse
Affiliation(s)
- Patrick Peschl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Romana Höftberger
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
20
|
Agahozo MC, Peferoen L, Baker D, Amor S. CD20 therapies in multiple sclerosis and experimental autoimmune encephalomyelitis - Targeting T or B cells? Mult Scler Relat Disord 2016; 9:110-7. [PMID: 27645355 DOI: 10.1016/j.msard.2016.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/06/2016] [Accepted: 07/19/2016] [Indexed: 01/03/2023]
Abstract
MS is widely considered to be a T cell-mediated disease although T cell immunotherapy has consistently failed, demonstrating distinct differences with experimental autoimmune encephalomyelitis (EAE), an animal model of MS in which T cell therapies are effective. Accumulating evidence has highlighted that B cells also play key role in MS pathogenesis. The high frequency of oligoclonal antibodies in the CSF, the localization of immunoglobulin in brain lesions and pathogenicity of antibodies originally pointed to the pathogenic role of B cells as autoantibody producing plasma cells. However, emerging evidence reveal that B cells also act as antigen presenting cells, T cell activators and cytokine producers suggesting that the strong efficacy of anti-CD20 antibody therapy observed in people with MS may reduce disease progression by several different mechanisms. Here we review the evidence and mechanisms by which B cells contribute to disease in MS compared to findings in the EAE model.
Collapse
Affiliation(s)
- Marie Colombe Agahozo
- Pathology Department, VU Medical Centre, VU University of Amsterdam, The Netherlands
| | - Laura Peferoen
- Pathology Department, VU Medical Centre, VU University of Amsterdam, The Netherlands
| | - David Baker
- Neuroimmunolgy Unit, Blizard Institute, Barts and the London School of Medicine & Dentistry Queen Mary University of London, United Kingdom
| | - Sandra Amor
- Pathology Department, VU Medical Centre, VU University of Amsterdam, The Netherlands; Neuroimmunolgy Unit, Blizard Institute, Barts and the London School of Medicine & Dentistry Queen Mary University of London, United Kingdom.
| |
Collapse
|
21
|
Weil MT, Möbius W, Winkler A, Ruhwedel T, Wrzos C, Romanelli E, Bennett JL, Enz L, Goebels N, Nave KA, Kerschensteiner M, Schaeren-Wiemers N, Stadelmann C, Simons M. Loss of Myelin Basic Protein Function Triggers Myelin Breakdown in Models of Demyelinating Diseases. Cell Rep 2016. [PMID: 27346352 DOI: 10.1016/j.celrep.2016.06.008;] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Breakdown of myelin sheaths is a pathological hallmark of several autoimmune diseases of the nervous system. We employed autoantibody-mediated animal models of demyelinating diseases, including a rat model of neuromyelitis optica (NMO), to target myelin and found that myelin lamellae are broken down into vesicular structures at the innermost region of the myelin sheath. We demonstrated that myelin basic proteins (MBP), which form a polymer in between the myelin membrane layers, are targeted in these models. Elevation of intracellular Ca(2+) levels resulted in MBP network disassembly and myelin vesiculation. We propose that the aberrant phase transition of MBP molecules from their cohesive to soluble and non-adhesive state is a mechanism triggering myelin breakdown in NMO and possibly in other demyelinating diseases.
Collapse
Affiliation(s)
- Marie-Theres Weil
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Göttingen, Germany
| | - Anne Winkler
- Department of Neuropathology, University of Göttingen Medical Center, 37075 Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Göttingen, Germany
| | - Claudia Wrzos
- Department of Neuropathology, University of Göttingen Medical Center, 37075 Göttingen, Germany
| | - Elisa Romanelli
- Institute of Clinical Neuroimmunology and Biomedical Center, Ludwig-Maximillians University, 80539 Munich, Germany
| | - Jeffrey L Bennett
- Departments of Neurology, University of Denver, Denver, CO 80045, USA
| | - Lukas Enz
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Norbert Goebels
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Göttingen, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology and Biomedical Center, Ludwig-Maximillians University, 80539 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Nicole Schaeren-Wiemers
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Christine Stadelmann
- Department of Neuropathology, University of Göttingen Medical Center, 37075 Göttingen, Germany
| | - Mikael Simons
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; Institute of Neuronal Cell Biology, Technical University Munich, 80805 Munich, Germany; German Center for Neurodegenerative Disease (DZNE), 6250 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany.
| |
Collapse
|
22
|
Loss of Myelin Basic Protein Function Triggers Myelin Breakdown in Models of Demyelinating Diseases. Cell Rep 2016; 16:314-322. [PMID: 27346352 PMCID: PMC4949381 DOI: 10.1016/j.celrep.2016.06.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/22/2016] [Accepted: 05/26/2016] [Indexed: 11/23/2022] Open
Abstract
Breakdown of myelin sheaths is a pathological hallmark of several autoimmune diseases of the nervous system. We employed autoantibody-mediated animal models of demyelinating diseases, including a rat model of neuromyelitis optica (NMO), to target myelin and found that myelin lamellae are broken down into vesicular structures at the innermost region of the myelin sheath. We demonstrated that myelin basic proteins (MBP), which form a polymer in between the myelin membrane layers, are targeted in these models. Elevation of intracellular Ca(2+) levels resulted in MBP network disassembly and myelin vesiculation. We propose that the aberrant phase transition of MBP molecules from their cohesive to soluble and non-adhesive state is a mechanism triggering myelin breakdown in NMO and possibly in other demyelinating diseases.
Collapse
|
23
|
Khalaj AJ, Hasselmann J, Augello C, Moore S, Tiwari-Woodruff SK. Nudging oligodendrocyte intrinsic signaling to remyelinate and repair: Estrogen receptor ligand effects. J Steroid Biochem Mol Biol 2016; 160:43-52. [PMID: 26776441 PMCID: PMC5233753 DOI: 10.1016/j.jsbmb.2016.01.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 01/08/2016] [Accepted: 01/11/2016] [Indexed: 01/06/2023]
Abstract
Demyelination in multiple sclerosis (MS) leads to significant, progressive axonal and neuronal degeneration. Currently existing immunosuppressive and immunomodulatory therapies alleviate MS symptoms and slow, but fail to prevent or reverse, disease progression. Restoration of damaged myelin sheath by replenishment of mature oligodendrocytes (OLs) should not only restore saltatory axon conduction, but also provide a major boost to axon survival. Our previous work has shown that therapeutic treatment with the modestly selective generic estrogen receptor (ER) β agonist diarylpropionitrile (DPN) confers functional neuroprotection in a chronic experimental autoimmune encephalomyelitis (EAE) mouse model of MS by stimulating endogenous remyelination. Recently, we found that the more potent, selective ERβ agonist indazole-chloride (Ind-Cl) improves clinical disease and motor performance. Importantly, electrophysiological measures revealed improved corpus callosal conduction and reduced axon refractoriness. This Ind-Cl treatment-induced functional remyelination was attributable to increased OL progenitor cell (OPC) and mature OL numbers. At the intracellular signaling level, transition of early to late OPCs requires ERK1/2 signaling, and transition of immature to mature OLs requires mTOR signaling; thus, the PI3K/Akt/mTOR pathway plays a major role in the late stages of OL differentiation and myelination. Indeed, therapeutic treatment of EAE mice with various ERβ agonists results in increased brain-derived neurotrophic factor (BDNF) and phosphorylated (p) Akt and p-mTOR levels. It is notable that while DPN's neuroprotective effects occur in the presence of peripheral and central inflammation, Ind-Cl is directly neuroprotective, as demonstrated by remyelination effects in the cuprizone-induced demyelination model, as well as immunomodulatory. Elucidating the mechanisms by which ER agonists and other directly remyelinating agents modulate endogenous OPC and OL regulatory signaling is critical to the development of effective remyelinating drugs. The discovery of signaling targets to induce functional remyelination will valuably contribute to the treatment of demyelinating neurological diseases, including MS, stroke, and traumatic brain and spinal cord injury.
Collapse
Affiliation(s)
- Anna J Khalaj
- Division of Biomedical Sciences, School of Medicine at the University of California, Riverside, United States
| | - Jonathan Hasselmann
- Division of Biomedical Sciences, School of Medicine at the University of California, Riverside, United States
| | - Catherine Augello
- Division of Biomedical Sciences, School of Medicine at the University of California, Riverside, United States
| | - Spencer Moore
- Division of Biomedical Sciences, School of Medicine at the University of California, Riverside, United States
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine at the University of California, Riverside, United States; Neuroscience Graduate Program, University of California, Riverside, United States.
| |
Collapse
|
24
|
Duncan ID, Radcliff AB. Inherited and acquired disorders of myelin: The underlying myelin pathology. Exp Neurol 2016; 283:452-75. [PMID: 27068622 PMCID: PMC5010953 DOI: 10.1016/j.expneurol.2016.04.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 01/26/2023]
Abstract
Remyelination is a major therapeutic goal in human myelin disorders, serving to restore function to demyelinated axons and providing neuroprotection. The target disorders that might be amenable to the promotion of this repair process are diverse and increasing in number. They range primarily from those of genetic, inflammatory to toxic origin. In order to apply remyelinating strategies to these disorders, it is essential to know whether the myelin damage results from a primary attack on myelin or the oligodendrocyte or both, and whether indeed these lead to myelin breakdown and demyelination. In some disorders, myelin sheath abnormalities are prominent but demyelination does not occur. This review explores the range of human and animal disorders where myelin pathology exists and focusses on defining the myelin changes in each and their cause, to help define whether they are targets for myelin repair therapy. We reviewed myelin disorders of the CNS in humans and animals. Myelin damage results from primary attack on the oligodendrocyte or myelin sheath. All major categories of disease can affect CNS myelin. Myelin vacuolation is common, yet does not always result in demyelination.
Collapse
Affiliation(s)
- Ian D Duncan
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States.
| | - Abigail B Radcliff
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
25
|
Li R, Rezk A, Healy LM, Muirhead G, Prat A, Gommerman JL, Bar-Or A. Cytokine-Defined B Cell Responses as Therapeutic Targets in Multiple Sclerosis. Front Immunol 2016; 6:626. [PMID: 26779181 PMCID: PMC4705194 DOI: 10.3389/fimmu.2015.00626] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/30/2015] [Indexed: 02/04/2023] Open
Abstract
Important antibody-independent pathogenic roles of B cells are emerging in autoimmune diseases, including multiple sclerosis (MS). The contrasting results of different treatments targeting B cells in patients (in spite of predictions of therapeutic benefits from animal models) call for a better understanding of the multiple roles that distinct human B cell responses likely play in MS. In recent years, both murine and human B cells have been identified with distinct functional properties related to their expression of particular cytokines. These have included regulatory (Breg) B cells (secreting interleukin (IL)-10 or IL-35) and pro-inflammatory B cells (secreting tumor necrosis factor α, LTα, IL-6, and granulocyte macrophage colony-stimulating factor). Better understanding of human cytokine-defined B cell responses is necessary in both health and diseases, such as MS. Investigation of their surface phenotype, distinct functions, and the mechanisms of regulation (both cell intrinsic and cell extrinsic) may help develop effective treatments that are more selective and safe. In this review, we focus on mechanisms by which cytokine-defined B cells contribute to the peripheral immune cascades that are thought to underlie MS relapses, and the impact of B cell-directed therapies on these mechanisms.
Collapse
Affiliation(s)
- Rui Li
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University , Montreal, QC , Canada
| | - Ayman Rezk
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University , Montreal, QC , Canada
| | - Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University , Montreal, QC , Canada
| | - Gillian Muirhead
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University , Montreal, QC , Canada
| | - Alexandre Prat
- Neuroimmunology Unit, Department of Neuroscience, Centre de Recherche du CHUM (CRCHUM), Université de Montreal , Montreal, QC , Canada
| | | | - Amit Bar-Or
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada; Experimental Therapeutics Program, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | | |
Collapse
|
26
|
't Hart BA, van Kooyk Y, Geurts JJG, Gran B. The primate autoimmune encephalomyelitis model; a bridge between mouse and man. Ann Clin Transl Neurol 2015; 2:581-93. [PMID: 26000330 PMCID: PMC4435712 DOI: 10.1002/acn3.194] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/19/2015] [Accepted: 02/19/2015] [Indexed: 12/13/2022] Open
Abstract
Introduction Multiple sclerosis (MS) is an enigmatic autoimmune-driven inflammatory/demyelinating disease of the human central nervous system (CNS), affecting brain, spinal cord, and optic nerves. The cause of the disease is not known and the number of effective treatments is limited. Despite some clear successes, translation of immunological discoveries in the mouse experimental autoimmune encephalomyelitis (EAE) model into effective therapies for MS patients has been difficult. This translation gap between MS and its elected EAE animal model reflects the phylogenetic distance between humans and their experimental counterpart, the inbred/specific pathogen free (SPF) laboratory mouse. Objective Here, we discuss that important new insights can be obtained into the mechanistic basis of the therapy paradox from the study of nonhuman primate EAE (NHP-EAE) models, the well-validated EAE model in common marmosets (Callithrix jacchus) in particular. Interpretation Data presented in this review demonstrate that due to a considerable immunological and pathological overlap with mouse EAE on one side and MS on the other, the NHP EAE model can help us bridge the translation gap.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre Rijswijk, The Netherlands ; Department Neuroscience, University Medical Center, University of Groningen Groningen, The Netherlands
| | - Yvette van Kooyk
- Department of Cell Biology and Immunology, Free University Medical Center Amsterdam, The Netherlands
| | - Jeroen J G Geurts
- Department of Anatomy and Neuroscience, Free University Medical Center Amsterdam, The Netherlands
| | - Bruno Gran
- Division of Clinical Neuroscience, University of Nottingham School of Medicine Nottingham, United Kingdom
| |
Collapse
|
27
|
Abstract
Autoimmune B cells play a major role in mediating tissue damage in multiple sclerosis (MS). In MS, B cells are believed to cross the blood-brain barrier and undergo stimulation, antigen-driven affinity maturation and clonal expansion within the supportive CNS environment. These highly restricted populations of clonally expanded B cells and plasma cells can be detected in MS lesions, in cerebrospinal fluid, and also in peripheral blood. In phase II trials in relapsing MS, monoclonal antibodies that target circulating CD20-positive B lymphocytes dramatically reduced disease activity. These beneficial effects occurred within weeks of treatment, indicating that a direct effect on B cells--and likely not on putative autoantibodies--was responsible. The discovery that depletion of B cells has an impact on MS biology enabled a paradigm shift in understanding how the inflammatory phase of MS develops, and will hopefully lead to development of increasingly selective therapies against culprit B cells and related humoral immune system pathways. More broadly, these studies illustrate how lessons learned from the bedside have unique power to inform translational research. They highlight the essential role of clinician scientists, currently endangered, who navigate the rocky and often unpredictable terrain between the worlds of clinical medicine and biomedical research.
Collapse
Affiliation(s)
- Stephen L Hauser
- Department of Neurology, University of California, San Francisco, USA
| |
Collapse
|
28
|
Carbohydrate Microarrays. POLYSACCHARIDES 2015. [PMCID: PMC7123348 DOI: 10.1007/978-3-319-16298-0_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Carbohydrates, like nucleic acids and proteins, are essential biological molecules. Owing to their intrinsic physicochemical properties, carbohydrates are capable of generating structural diversity in a multitude of ways and are prominently displayed on the surfaces of cell membranes or on the exposed regions of macromolecules. Recent studies highlight that carbohydrate moieties are critical for molecular recognition, cell-cell interactions, and cell signaling in many physiological and pathological processes, and for biocommunication between microbes and host species. Modern carbohydrate microarrays emerged in 2002 and brought in new high-throughput tools for “glyco code” exploration. In this section, some basic concepts of sugar chain diversity, glyco-epitope recognition, and the evolving area of glyco-epitomics and biomarker discovery are discussed. Two complementary technologies, carbohydrate antigen arrays and photogenerated glyco-chips, serve as models to illustrate how to apply carbohydrate microarrays to address biomedical questions.
Collapse
|
29
|
Retinoic acid enhances the levels of IL-10 in TLR-stimulated B cells from patients with relapsing-remitting multiple sclerosis. J Neuroimmunol 2014; 278:11-8. [PMID: 25595247 DOI: 10.1016/j.jneuroim.2014.11.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/07/2014] [Accepted: 11/20/2014] [Indexed: 01/31/2023]
Abstract
We have explored the beneficial effects of retinoic acid (RA) on B cells from multiple sclerosis (MS) patients. When co-stimulated via the toll-like receptors (TLRs) TLR9 and RP105, MS B cells secreted less of the anti-inflammatory cytokine interleukin 10 (IL-10) compared to B cells from healthy controls. Importantly, RA enhanced the secretion of IL-10 by MS-derived B cells without affecting the levels of the pro-inflammatory cytokine TNF-α. RA revealed the same ability to induce IL-10 as did interferon-β-1b (IFN-β-1b), and B-cells from patients treated with glatiramer acetate or IFN-β-1b still displayed the beneficial effects of RA on the IL-10/TNF-α ratio.
Collapse
|
30
|
The co-occurrence of multiple sclerosis and type 1 diabetes: shared aetiologic features and clinical implication for MS aetiology. J Neurol Sci 2014; 348:126-31. [PMID: 25480016 DOI: 10.1016/j.jns.2014.11.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 10/21/2014] [Accepted: 11/14/2014] [Indexed: 12/16/2022]
Abstract
We reviewed the evidence for the co-occurrence of type 1 diabetes mellitus (T1D) and multiple sclerosis (MS), and assessed the clinical significance of this association and the shared aetiological features of the two diseases. T1D and MS contribute considerably to the burden of autoimmune diseases in young adults. The co-occurrence of MS and T1D has been reported by a number of studies, suggesting that the two conditions share one or more aetiological components. Both conditions have been associated with distinct human leukocyte antigen (HLA) haplotypes but share a number of similarities in clinical, epidemiological and immunological features, leading to suggestions of possible common mechanisms of development. While underlying genetic factors may be important for the co-occurrence of both conditions, some evidence suggests that environmental factors such as vitamin D deficiency may also modulate an individual's risk for the development of both conditions. Evidence on whether the co-occurrence of the two autoimmune conditions will affect the disease course and severity of MS is merely absent. Further studies need to be conducted to ascertain whether the neuropathology associated with T1D might influence the disease course and contribute to the severity of MS.
Collapse
|
31
|
Cantor JM. CD98 is a potential target for ablating B cell clonal expansion and autoantibody in multiple sclerosis. J Neuroimmunol 2014; 274:230-3. [PMID: 25002078 DOI: 10.1016/j.jneuroim.2014.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/01/2014] [Accepted: 06/17/2014] [Indexed: 01/15/2023]
Abstract
Current B cell-directed therapies for multiple sclerosis impact multiple B cell functions. CD98hc enables B cell clonal expansion and antibody production. I probed the relative importance of autoantibody secretion vs. other B cell functions in MS and targeted CD98hc as a possible therapeutic strategy. I report that the loss of CD98hc function in B cells largely prevents autoantibody production while preserving antigen-presenting and T cell-directing capacities. Mice lacking CD98hc in B cells are protected from EAE; importantly this is overcome with autoantibody-containing plasma. Thus CD98hc blockade is a possible avenue to treat MS by inhibiting clonal expansion and autoantibody.
Collapse
Affiliation(s)
- Joseph M Cantor
- Department of Medicine, University of California San Diego, MC 0726, 9500 Gilman Drive, La Jolla, CA 92093-0726, United States.
| |
Collapse
|
32
|
Bakhti M, Aggarwal S, Simons M. Myelin architecture: zippering membranes tightly together. Cell Mol Life Sci 2014; 71:1265-77. [PMID: 24165921 PMCID: PMC11113231 DOI: 10.1007/s00018-013-1492-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 09/11/2013] [Accepted: 10/07/2013] [Indexed: 12/12/2022]
Abstract
Rapid nerve conduction requires the coating of axons by a tightly packed multilayered myelin membrane. In the central nervous system, myelin is formed from cellular processes that extend from oligodendrocytes and wrap in a spiral fashion around an axon, resulting in the close apposition of adjacent myelin membrane bilayers. In this review, we discuss the physical principles underlying the zippering of the plasma membrane of oligodendrocytes at the cytoplasmic and extracellular leaflet. We propose that the interaction of the myelin basic protein with the cytoplasmic leaflet of the myelin bilayer triggers its polymerization into a fibrous network that drives membrane zippering and protein extrusion. In contrast, the adhesion of the extracellular surfaces of myelin requires the down-regulation of repulsive components of the glycocalyx, in order to uncover weak and unspecific attractive forces that bring the extracellular surfaces into close contact. Unveiling the mechanisms of myelin membrane assembly at the cytoplasmic and extracelluar sites may help to understand how the myelin bilayers are disrupted and destabilized in the different demyelinating diseases.
Collapse
Affiliation(s)
- Mostafa Bakhti
- Max-Planck-Institute of Experimental Medicine, Hermann-Rein-Str. 3, Göttingen, Germany
- Department of Neurology, University of Göttingen, Robert-Koch-Str. 40, Göttingen, Germany
- Present Address: Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Shweta Aggarwal
- Max-Planck-Institute of Experimental Medicine, Hermann-Rein-Str. 3, Göttingen, Germany
- Department of Neurology, University of Göttingen, Robert-Koch-Str. 40, Göttingen, Germany
| | - Mikael Simons
- Max-Planck-Institute of Experimental Medicine, Hermann-Rein-Str. 3, Göttingen, Germany
- Department of Neurology, University of Göttingen, Robert-Koch-Str. 40, Göttingen, Germany
| |
Collapse
|
33
|
Wang D, Bhat R, Sobel RA, Huang W, Wang LX, Olsson T, Steinman L. Uncovering cryptic glycan markers in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). Drug Dev Res 2014; 75:172-88. [PMID: 24648292 DOI: 10.1002/ddr.21169] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 02/10/2014] [Indexed: 01/18/2023]
Abstract
Using an integrated antigen microarray approach, we observed epitope-spreading of autoantibody responses to a variety of antigenic structures in the cerebrospinal fluid (CSF) of patients with multiple sclerosis (MS) and in the serum of mice with experimental autoimmune encephalomyelitis (EAE). These included previously described protein- and lipid-based antigenic targets and newly discovered autoimmunogenic sugar moieties, notably, autoantibodies specific for the oligomannoses in both MS patient CSF and the sera of mice with EAE. These glycans are often masked by other sugar moieties and belong to a class of cryptic autoantigens. We further determined that these targets are highly expressed on multiple cell types in MS and EAE lesions. Co-immunization of SJL/J mice with a Man9-KLH conjugate at the time of EAE induction elicited highly significant levels of anti-Man9-cluster autoantibodies. Nevertheless, this anti-glycan autoantibody response was associated with a significantly reduced clinical severity of EAE. The potential of these cryptic glycan markers and targeting antibodies for diagnostic and therapeutic interventions of neurological disorders has yet to be explored.
Collapse
Affiliation(s)
- Denong Wang
- Tumor Glycomics Laboratory, SRI International Biosciences Division, Menlo Park, CA, 94025, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Fillatreau S, Anderton SM. B-cell function in CNS inflammatory demyelinating disease: a complexity of roles and a wealth of possibilities. Expert Rev Clin Immunol 2014; 3:565-78. [DOI: 10.1586/1744666x.3.4.565] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
35
|
Lolli F, Rovero P, Chelli M, Papini AM. Toward biomarkers in multiple sclerosis: new advances. Expert Rev Neurother 2014; 6:781-94. [PMID: 16734525 DOI: 10.1586/14737175.6.5.781] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Multiple sclerosis is an autoimmune disease that commonly affects young adults. If initially characterized by acute relapses, it is later followed by only incomplete remission. Over years, progressive disability and irreversible deficit lead to chronic neurological deficits in the majority of patients. The clinical course is protracted and unpredictable, and no biological marker is useful in predicting the evolution of autoaggression and disability. It is difficult to diagnose and to monitor disease progression after the initial symptoms or even during the major clinical manifestations, and it is difficult to treat. In this review, the authors report recent advances in the field, focusing on the search of new antigens as a marker of the disease, in their relevance to the pathophysiology and diagnosis of the disease.
Collapse
Affiliation(s)
- Francesco Lolli
- Laboratorio Interdipartimentale di Chimica & Biologia dei Peptidi & Proteine, Polo Scientifico e Tecnologico, Università degli Studi di Firenze, via Ugo Schiff 6, I-50019 Sesto Fiorentino, Italy.
| | | | | | | |
Collapse
|
36
|
Lim ET, Giovannoni G. Immunopathogenesis and immunotherapeutic approaches in multiple sclerosis. Expert Rev Neurother 2014; 5:379-90. [PMID: 15938671 DOI: 10.1586/14737175.5.3.379] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple sclerosis is an organ-specific autoimmune disease, characterized pathologically by cell-mediated inflammation, demyelination and variable degrees of axonal loss. Although inflammation is considered central to the pathogenesis of multiple sclerosis, to date, the only licensed and hence widely used multiple sclerosis immunotherapies are interferon-beta, glatiramer acetate and mitoxantrone. This review discusses the immunopathogenesis of multiple sclerosis, focusing on a number of emerging immunotherapies. A number of new approaches likely to manipulate the immunopathogenesis of multiple sclerosis and which may ultimately allow for the development of more effective immunotherapy are also highlighted.
Collapse
Affiliation(s)
- Ee Tuan Lim
- University College London, Department of Neuroinflammation, Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | | |
Collapse
|
37
|
Wang D, Tang J, Wolfinger RD, Carroll GT. Carbohydrate Microarrays. POLYSACCHARIDES 2014. [DOI: 10.1007/978-3-319-03751-6_35-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
38
|
Criste G, Trapp B, Dutta R. Axonal loss in multiple sclerosis: causes and mechanisms. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:101-13. [PMID: 24507515 DOI: 10.1016/b978-0-444-52001-2.00005-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system and the leading cause of non-traumatic neurologic disability in young adults in the United States and Europe. The disease course is variable and starts with reversible episodes of neurologic disability which transforms into continuous and irreversible neurologic decline. It is well established that loss of axons and neurons is the major cause of the progressive neurologic decline that most MS patients endure. Current hypotheses support primary inflammatory demyelination as the underlying cause of axonal loss during earlier stages in MS. The transition to progressive disease course is thought to occur when a threshold of neuronal and axonal loss is reached and the compensatory capacity of the central nervous system is surpassed. Available immunomodulatory therapies are of little benefit to MS after entering this irreversible phase of the disease. Elucidation of mechanisms that are responsible for axonal loss is therefore essential for the development of therapies directed to stop neurologic decline in MS patients. The current chapter reviews existing data on mechanisms of axonal pathology in MS.
Collapse
Affiliation(s)
- Gerson Criste
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bruce Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
39
|
Hartung HP, Aktas O, Menge T, Kieseier BC. Immune regulation of multiple sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:3-14. [PMID: 24507511 DOI: 10.1016/b978-0-444-52001-2.00001-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Multiple sclerosis (MS) is considered a prototype inflammatory autoimmune disorder of the central nervous system (CNS). The etiology of this disease remains unknown, but an interplay between as yet unidentified environmental factors and susceptibility genes appears most likely. In consequence, these factors trigger a cascade, involving an inflammatory response within the CNS that results in demyelination, oligodendrocyte death, axonal damage, gliosis, and neurodegeneration. How these complex traits translate into the clinical presentation of the disease is a focus of ongoing research. The central hypothesis is that T lymphocytes with receptors for CNS myelin components are driving the disease. The initial activation of autoreactive lymphocytes is thought to take place in the systemic lymphoid organs, most likely through molecular mimickry or nonspecifically through bystander activation. These autoreactive lymphocytes can migrate to the CNS where they become reactivated upon encountering their target antigen, initiating an autoimmune inflammatory attack. This ultimately leads to demyelination and axonal damage. This chapter focuses on the role of T and B lymphocytes in the immunopathogenesis of MS.
Collapse
Affiliation(s)
- Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Til Menge
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Bernd C Kieseier
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
40
|
Schulze-Topphoff U, Casazza S, Varrin-Doyer M, Pekarek K, Sobel RA, Hauser SL, Oksenberg JR, Zamvil SS, Baranzini SE. Tob1 plays a critical role in the activation of encephalitogenic T cells in CNS autoimmunity. ACTA ACUST UNITED AC 2013; 210:1301-9. [PMID: 23797093 PMCID: PMC3698524 DOI: 10.1084/jem.20121611] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Loss of antiproliferative gene TOB1 results in more severe EAE driven by augmented pathogenic T cell responses. Reliable biomarkers corresponding to disease progression or therapeutic responsiveness in multiple sclerosis (MS) have not been yet identified. We previously reported that low expression of the antiproliferative gene TOB1 in CD4+ T cells of individuals presenting with an initial central nervous system (CNS) demyelinating event (a clinically isolated syndrome), correlated with high risk for progression to MS. We report that experimental autoimmune encephalomyelitis (EAE) in Tob1−/− mice was associated with augmented CNS inflammation, increased infiltrating CD4+ and CD8+ T cell counts, and increased myelin-reactive Th1 and Th17 cells, with reduced numbers of regulatory T cells. Reconstitution of Rag1−/− mice with Tob1−/− CD4+ T cells recapitulated the aggressive EAE phenotype observed in Tob1−/− mice. Furthermore, severe spontaneous EAE was observed when Tob1−/− mice were crossed to myelin oligodendrocyte glycoprotein–specific T cell receptor transgenic (2D2) mice. Collectively, our results reveal a critical role for Tob1 in adaptive T cell immune responses that drive development of EAE, thus providing support for the development of Tob1 as a biomarker for demyelinating disease activity.
Collapse
|
41
|
Abstract
OBJECTIVE Multiple sclerosis (MS) is a chronic, progressive central nervous system (CNS) disease with unknown cause. Considerable evidence supports an autoimmune origin with an important role for cellular immune responses in its pathogenesis. METHODS We have reviewed the current literature dealing with lymphocyte responses and their interactions as it relates to MS and present supporting evidence from animal models. RESULTS Issues regarding CD4+ T-cell subpopulations, their functional differentiation and regulatory interactions as they relate to their presumed role in MS-related pathology have been updated with references to the current literature. DISCUSSION The evidence reviewed supports an important role of CD4+ T-cells in the immunopathogenesis of MS. The successful outcome of blocking CD4 cells entry into the CNS of animals with experimental demyelinating disease and humans with MS is a strong support for other evidence of an important role of these cell populations in the pathogenesis of MS. The understanding of the specific roles of CD4+ T-cells in the development of MS is crucial for better disease management and the prevention of neurological disability.
Collapse
Affiliation(s)
- Sylvia Delgado
- Multiple Sclerosis Center, University of Miami Miller School of Medicine, FL 33136, USA
| | | |
Collapse
|
42
|
Bansal P, Khan T, Bussmeyer U, Challa DK, Swiercz R, Velmurugan R, Ober RJ, Ward ES. The Encephalitogenic, Human Myelin Oligodendrocyte Glycoprotein–Induced Antibody Repertoire Is Directed toward Multiple Epitopes in C57BL/6-Immunized Mice. THE JOURNAL OF IMMUNOLOGY 2013; 191:1091-101. [DOI: 10.4049/jimmunol.1300019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
43
|
Pedotti R, Musio S, Scabeni S, Farina C, Poliani PL, Colombo E, Costanza M, Berzi A, Castellucci F, Ciusani E, Confalonieri P, Hemmer B, Mantegazza R, Antozzi C. Exacerbation of experimental autoimmune encephalomyelitis by passive transfer of IgG antibodies from a multiple sclerosis patient responsive to immunoadsorption. J Neuroimmunol 2013; 262:19-26. [PMID: 23768729 DOI: 10.1016/j.jneuroim.2013.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 05/10/2013] [Accepted: 05/17/2013] [Indexed: 10/26/2022]
Abstract
The pathogenic role of antibodies in multiple sclerosis (MS) is still controversial. We transferred to mice with experimental autoimmune encephalomyelitis (EAE), animal model of MS, IgG antibodies purified from a MS patient presenting a dramatic clinical improvement during relapse after selective IgG removal with immunoadsorption. Passive transfer of patient's IgG exacerbated motor paralysis and increased mouse central nervous system (CNS) inflammation and demyelination. Binding of patient's IgG was demonstrated in mouse CNS, with a diffuse staining of white matter oligodendrocytes. These data support a growing body of evidence that antibodies can play an important role in the pathobiology of MS.
Collapse
Affiliation(s)
- Rosetta Pedotti
- Neuroimmunology and Neuromuscular Disorders Unit, Foundation IRCCS Neurological Institute C. Besta, Via Celoria 11, 20133 Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sulkowski G, Dąbrowska-Bouta B, Chalimoniuk M, Strużyńska L. Effects of antagonists of glutamate receptors on pro-inflammatory cytokines in the brain cortex of rats subjected to experimental autoimmune encephalomyelitis. J Neuroimmunol 2013; 261:67-76. [PMID: 23746391 DOI: 10.1016/j.jneuroim.2013.05.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/18/2013] [Accepted: 05/10/2013] [Indexed: 10/26/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). Inflammatory cytokines and glutamate neurotoxicity have been proposed as major determinants accompanying the demyelination and axonal degeneration observed during the course of MS. The present study using the animal model of MS known as experimental autoimmune encephalomyelitis (EAE) demonstrates that pharmacological inhibition of ionotropic NMDA glutamate receptors by their antagonists (amantadine and memantine) suppresses neurological symptoms of disease in EAE rats and reduces expression of pro-inflammatory cytokines in the brain. Conversely, antagonists of group I metabotropic glutamate receptors, mGluRs (LY 367385 and MPEP), do not affect the inflammatory process and the neurological condition of EAE rats.
Collapse
Affiliation(s)
- Grzegorz Sulkowski
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawińskiego str., 02-106 Warsaw, Poland.
| | | | | | | |
Collapse
|
45
|
Expansion of CD27high plasmablasts in transverse myelitis patients that utilize VH4 and JH6 genes and undergo extensive somatic hypermutation. Genes Immun 2013; 14:291-301. [PMID: 23594958 DOI: 10.1038/gene.2013.18] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/08/2013] [Accepted: 02/12/2013] [Indexed: 12/18/2022]
Abstract
Patients with the autoimmune disease multiple sclerosis (MS) typically present with the clinically isolated syndromes (CIS) transverse myelitis (TM) or optic neuritis (ON). B-cell disturbances have been well documented in patients with MS and CIS patients with ON, but not in CIS patients with TM, despite the fact that these patients have the worst clinical outcome of all CIS types. The goal of this study was to characterize the B-cell populations and immunoglobulin genetics in TM patients. We found a unique expansion of CD27(high) plasmablasts in both the cerebrospinal fluid and periphery of TM patients that is not present in ON patients. Additionally, plasmablasts from TM patients show evidence for positive selection with increased somatic hypermutation accumulation in VH4(+) B cells and receptor editing that is not observed in ON patients. These characteristics unique to TM patients may impact disease severity and progression.
Collapse
|
46
|
Benkhoucha M, Molnarfi N, Santiago-Raber ML, Weber MS, Merkler D, Collin M, Lalive PH. IgG glycan hydrolysis by EndoS inhibits experimental autoimmune encephalomyelitis. J Neuroinflammation 2012; 9:209. [PMID: 22943418 PMCID: PMC3458989 DOI: 10.1186/1742-2094-9-209] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 08/14/2012] [Indexed: 01/03/2023] Open
Abstract
Studies in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis, have shown that B cells markedly influence the course of the disease, although whether their effects are protective or pathological is a matter of debate. EndoS hydrolysis of the IgG glycan has profound effects on IgG effector functions, such as complement activation and Fc receptor binding, suggesting that the enzyme could be used as an immunomodulatory therapeutic agent against IgG-mediated diseases. We demonstrate here that EndoS has a protective effect in myelin oligodendrocyte glycoprotein peptide amino acid 35–55 (MOG35-55)-induced EAE, a chronic neuroinflammatory demyelinating disorder of the central nervous system (CNS) in which humoral immune responses are thought to play only a minor role. EndoS treatment in chronic MOG35-55-EAE did not impair encephalitogenic T cell priming and recruitment into the CNS of mice, consistent with a primary role of EndoS in controlling IgG effector functions. In contrast, reduced EAE severity coincided with poor serum complement activation and deposition within the spinal cord, suggesting that EndoS treatment impairs B cell effector function. These results identify EndoS as a potential therapeutic agent against antibody-mediated CNS autoimmune disorders.
Collapse
Affiliation(s)
- Mahdia Benkhoucha
- Department of Pathology and Immunology, University of Geneva, 1211, Geneva, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
47
|
Chen LP, Li ZF, Ping M, Li R, Liu J, Xie XH, Song XJ, Guo L. Regulation of Olig2 during astroglial differentiation in the subventricular zone of a cuprizone-induced demyelination mouse model. Neuroscience 2012; 221:96-107. [PMID: 22771621 DOI: 10.1016/j.neuroscience.2012.06.063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 05/18/2012] [Accepted: 06/23/2012] [Indexed: 10/28/2022]
Abstract
The mammalian subventricular zone (SVZ) is the largest germinative zone of the adult brain. Progenitor cells generated from the SVZ play important roles during the remyelination process. To determine the functional role of Olig2 in regulating astroglial differentiation in the mouse SVZ, we used the cuprizone mouse model to investigate demyelination. We found that cuprizone administration significantly enhanced the expression of Olig2 and increased astroglial differentiation in the SVZ, as compared with control. Moreover, cytoplasmic translocation of Olig2 occurred after demyelination. In vitro studies further revealed that supplementation of culture media with growth factors enhanced the oligodendroglial differentiation of oligodendrocyte progenitor cells (OPCs), whereas serum alone promoted astroglial differentiation and cytoplasmic translocation of Olig2. Additionally, the expression levels of bone morphogenetic proteins 2 and 4 (BMP2 and BMP4) and inhibitor of DNA binding 2 and 4 (Id2 and Id4) were greatly elevated during astroglial differentiation. BMP inhibition by noggin suppressed the astroglial differentiation of OPCs. Our results indicate that Olig2 may serve as a key regulator during the directional differentiation of progenitor cells after demyelination. The BMP signaling pathway may contribute to the cytoplasmic translocation and altered expression of Olig2 during the remyelination process. These findings provide a better understanding of the mechanisms involved in remyelination.
Collapse
Affiliation(s)
- L P Chen
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Zhao J, Gao Y, Cheng C, Yan M, Wang J. Upregulation of β-1,4-galactosyltransferase I in rat spinal cord with experimental autoimmune encephalomyelitis. J Mol Neurosci 2012; 49:437-45. [PMID: 22706684 DOI: 10.1007/s12031-012-9824-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 05/28/2012] [Indexed: 12/23/2022]
Abstract
Inflammatory infiltration has been recently emphasized in the demyelinating diseases of the central nervous system including multiple sclerosis. β-1,4-Galactosyltransferase I (β-1,4-GalT-I) is a major galactosyltransferase responsible for selectin-ligand biosynthesis, mediating rolling of the inflammatory lymphocytes. In the present study, Western blot showed that expression of β-1,4-GalT-I was low in normal or complete Freund's adjuvant (CFA) control rats' spinal cords, and it began to increase since early stage and peaked at E4 stage of experimental autoimmune encephalomyelitis (EAE) and restored approximately at normal level in the recovery stage. Immunohistochemisty revealed that upregulation of β-1,4-GalT-I was predominantly distributed in the white matter of spinal cord , while there was also some increased staining of β-1,4-GalT-I in the grey matter. Meanwhile, the expression of E-selectin, the substrate of β-1,4-GalT-I, was significantly increased, with a peak at E4 stage of EAE, and gradually decreased thereafter. Lectin blot showed that the protein bands with molecular weights of 65-25 kDa reacted a remarkable increase at the peak stage of EAE when compared with the normal and CFA control. Ricinus Communis Agglutinin-I (RCA-I) histochemistry revealed that RCA-Ι-positive signals were most intense in white matter of lumbosacral spinal cord at the peak stage of EAE (E4). Immunohistochemistry showed that β-1,4-GalT-I and CD62E, a marker for E-selectin stainings located in a considerable number of ED1 (+) macrophages in perivascular or in the white matter in EAE lesions, and a good co-localization of ED1 (+) cells with CD62E was observed. All these results suggest that β-1,4-GalT-I might serve as an inflammatory mediator regulating adhesion and migration of inflammatory cells in EAE, possibly through influencing the modification of galactosylated carbohydrate chains to modulate selectin-ligand biosynthesis and interaction with E-selectin.
Collapse
Affiliation(s)
- Jianmei Zhao
- Affiliated Children's Hospital of Soochow University, Suzhou, Jiangsu Province, 225121, People's Republic of China
| | | | | | | | | |
Collapse
|
49
|
Elliott C, Lindner M, Arthur A, Brennan K, Jarius S, Hussey J, Chan A, Stroet A, Olsson T, Willison H, Barnett SC, Meinl E, Linington C. Functional identification of pathogenic autoantibody responses in patients with multiple sclerosis. ACTA ACUST UNITED AC 2012; 135:1819-33. [PMID: 22561643 PMCID: PMC3359756 DOI: 10.1093/brain/aws105] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pathological and clinical studies implicate antibody-dependent mechanisms in the immunopathogenesis of multiple sclerosis. We tested this hypothesis directly by investigating the ability of patient-derived immunoglobulins to mediate demyelination and axonal injury in vitro. Using a myelinating culture system, we developed a sensitive and reproducible bioassay to detect and quantify these effects and applied this to investigate the pathogenic potential of immunoglobulin G preparations obtained from patients with multiple sclerosis (n = 37), other neurological diseases (n = 10) and healthy control donors (n = 13). This identified complement-dependent demyelinating immunoglobulin G responses in approximately 30% of patients with multiple sclerosis, which in two cases was accompanied by significant complement-dependent antibody mediated axonal loss. No pathogenic immunoglobulin G responses were detected in patients with other neurological disease or healthy controls, indicating that the presence of these demyelinating/axopathic autoantibodies is specific for a subset of patients with multiple sclerosis. Immunofluorescence microscopy revealed immunoglobulin G preparations with demyelinating activity contained antibodies that specifically decorated the surface of myelinating oligodendrocytes and their contiguous myelin sheaths. No other binding was observed indicating that the response is restricted to autoantigens expressed by terminally differentiated myelinating oligodendrocytes. In conclusion, our study identifies axopathic and/or demyelinating autoantibody responses in a subset of patients with multiple sclerosis. This observation underlines the mechanistic heterogeneity of multiple sclerosis and provides a rational explanation why some patients benefit from antibody depleting treatments.
Collapse
Affiliation(s)
- Christina Elliott
- Institute of Immunology, Immunity and Infection, University of Glasgow, Glasgow, G12 8TA, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lee DH, Linker RA. The role of myelin oligodendrocyte glycoprotein in autoimmune demyelination: a target for multiple sclerosis therapy? Expert Opin Ther Targets 2012; 16:451-62. [DOI: 10.1517/14728222.2012.677438] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|