1
|
Kurt Jellinger 90: his contribution to neuroimmunology. J Neural Transm (Vienna) 2021; 128:1545-1550. [PMID: 34110492 PMCID: PMC8528743 DOI: 10.1007/s00702-021-02358-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/27/2021] [Indexed: 11/05/2022]
Abstract
This review honors Kurt Jellinger on his 90th birthday as one of the most outstanding neuropathologists, who has contributed immensely to neuroscience due to his vast experience and collection of excellently documented autopsy cases. Two of his many insightful reports are highlighted here. One report focuses on the pathogenesis of inflammatory demyelinating diseases and investigates the neuropathology in autopsy tissue of a patient, who developed an MS-like disease after repeated treatment with lyophilized bovine brain cells in 1958. More than 60 years later, after reinvestigation of the historic samples in 2015 and subsequent mRNA isolation, next generation sequencing and reconstruction of the antibody, we succeeded in identifying myelin oligodendrocyte glycoprotein (MOG) as the target antigen and provided the missing element between the pathomechanisms in classic EAE animal models and transfer of this disease process into humans. A second significant example of Kurt Jellinger’s contribution to neuroscience was a report on the role of MS in the development of Alzheimer's disease (AD), which found that AD pathology is present to the same extent in demyelinated and non-demyelinated cortical areas in MS and the incidence for AD pathology in elderly MS patients is comparable to the normal-aging population. This indicates that chronic inflammation in the MS cortex alone does not significantly predispose to the development of cortical AD pathology. These and other findings were only possible due to the broad collection of extremely well-defined material established by Kurt Jellinger, which ultimately continues to contribute to translational neuroscience, even decades later.
Collapse
|
2
|
Mockus TE, Munie A, Atkinson JR, Segal BM. Encephalitogenic and Regulatory CD8 T Cells in Multiple Sclerosis and Its Animal Models. THE JOURNAL OF IMMUNOLOGY 2021; 206:3-10. [PMID: 33443060 DOI: 10.4049/jimmunol.2000797] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS), a neuroinflammatory disease that affects millions worldwide, is widely thought to be autoimmune in etiology. Historically, research into MS pathogenesis has focused on autoreactive CD4 T cells because of their critical role in the animal model, experimental autoimmune encephalomyelitis, and the association between MS susceptibility and single-nucleotide polymorphisms in the MHC class II region. However, recent studies have revealed prominent clonal expansions of CD8 T cells within the CNS during MS. In this paper, we review the literature on CD8 T cells in MS, with an emphasis on their potential effector and regulatory properties. We discuss the impact of disease modifying therapies, currently prescribed to reduce MS relapse rates, on CD8 T cell frequency and function. A deeper understanding of the role of CD8 T cells in MS may lead to the development of more effective and selective immunomodulatory drugs for particular subsets of patients.
Collapse
Affiliation(s)
- Taryn E Mockus
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Ashley Munie
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210.,Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Jeffrey R Atkinson
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Benjamin M Segal
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210; .,Neuroscience Research Institute, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
3
|
Roodselaar J, Zhou Y, Leppert D, Hauser AE, Urich E, Anthony DC. Anti-CD20 Disrupts Meningeal B-Cell Aggregates in a Model of Secondary Progressive Multiple Sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/3/e975. [PMID: 33653962 PMCID: PMC7931641 DOI: 10.1212/nxi.0000000000000975] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/28/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Therapies targeting B cells have been used in the clinic for multiple sclerosis (MS). In patients with relapsing MS, anti-CD20 therapy often suppresses relapse activity; yet, their effect on disease progression has been disappointing. Most anti-CD20 therapeutic antibodies are type I, but within the unique microenvironment of the brain, type II antibodies may be more beneficial, as type II antibodies exhibit reduced complement-dependent cytotoxicity and they have an increased capacity to induce direct cell death that is independent of the host immune response. METHODS We compared the effect of type I with type II anti-CD20 therapy in a new rodent model of secondary progressive MS (SPMS), which recapitulates the principal histopathologic features of MS including meningeal B-cell aggregates. Focal MS-like lesions were induced by injecting heat-killed Mycobacterium tuberculosis into the piriform cortex of MOG-immunized mice. Groups of mice were treated with anti-CD20 antibodies (type I [rituxumab, 10 mg/kg] or type II [GA101, 10 mg/kg]) 4 weeks after lesion initiation, and outcomes were evaluated by immunohistochemistry. RESULTS Anti-CD20 therapy decreased the extent of glial activation, significantly decreased the number of B and T lymphocytes in the lesion, and resulted in disruption of the meningeal aggregates. Moreover, at the given dose, the type II anti-CD20 therapy was more efficacious than the type I and also protected against neuronal death. CONCLUSIONS These results indicate that anti-CD20 may be an effective therapy for SPMS with B-cell aggregates and that the elimination of CD20+ B cells alone is sufficient to cause disruption of aggregates in the brain.
Collapse
Affiliation(s)
- Jay Roodselaar
- From the Department of Pharmacology (J.R., Y.Z.), University of Oxford; University of Basel (D.L.), Switzerland; Deutsches Rheumaforschungszentrum (DRFZ) and Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin (J.R., A.E.H.), Germany; Roche Innovation Center (E.U.), Basel, Switzerland; and Department of Pharmacology (D.C.A.), University of Oxford, UK.
| | - Yifan Zhou
- From the Department of Pharmacology (J.R., Y.Z.), University of Oxford; University of Basel (D.L.), Switzerland; Deutsches Rheumaforschungszentrum (DRFZ) and Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin (J.R., A.E.H.), Germany; Roche Innovation Center (E.U.), Basel, Switzerland; and Department of Pharmacology (D.C.A.), University of Oxford, UK
| | - David Leppert
- From the Department of Pharmacology (J.R., Y.Z.), University of Oxford; University of Basel (D.L.), Switzerland; Deutsches Rheumaforschungszentrum (DRFZ) and Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin (J.R., A.E.H.), Germany; Roche Innovation Center (E.U.), Basel, Switzerland; and Department of Pharmacology (D.C.A.), University of Oxford, UK
| | - Anja E Hauser
- From the Department of Pharmacology (J.R., Y.Z.), University of Oxford; University of Basel (D.L.), Switzerland; Deutsches Rheumaforschungszentrum (DRFZ) and Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin (J.R., A.E.H.), Germany; Roche Innovation Center (E.U.), Basel, Switzerland; and Department of Pharmacology (D.C.A.), University of Oxford, UK
| | - Eduard Urich
- From the Department of Pharmacology (J.R., Y.Z.), University of Oxford; University of Basel (D.L.), Switzerland; Deutsches Rheumaforschungszentrum (DRFZ) and Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin (J.R., A.E.H.), Germany; Roche Innovation Center (E.U.), Basel, Switzerland; and Department of Pharmacology (D.C.A.), University of Oxford, UK
| | - Daniel C Anthony
- From the Department of Pharmacology (J.R., Y.Z.), University of Oxford; University of Basel (D.L.), Switzerland; Deutsches Rheumaforschungszentrum (DRFZ) and Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin (J.R., A.E.H.), Germany; Roche Innovation Center (E.U.), Basel, Switzerland; and Department of Pharmacology (D.C.A.), University of Oxford, UK
| |
Collapse
|
4
|
Lassmann H. Neuroinflammation: 2021 update. FREE NEUROPATHOLOGY 2021; 2:1. [PMID: 37284634 PMCID: PMC10209849 DOI: 10.17879/freeneuropathology-2021-3166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 06/08/2023]
Abstract
Key requirements for the validity of a neuropathological study are the inclusion of large numbers of biopsy or autopsy cases and proper controls, the rigorous classification of the basic neuropathology and the selection of the most suitable technologies for investigation. Whether the studies are performed with the fanciest, new, and state of the art technology or with rather conventional methodology is of minor importance. Following these criteria, a spectrum of neuropathological studies has been published in 2020, which provides new insights on important questions related to neurological disease. They include the pathological substrate of brain disease in COVID-19 infected patients, the nature of the adaptive and innate inflammatory response, or the type and mechanisms of tissue injury and repair in multiple sclerosis, and diagnostically relevant or mechanistic new insights into antibody-mediated diseases of the central nervous system. Other studies describe in detail the dynamic changes of brain inflammation in patients with trisomy 21 as a disease model for Alzheimer's disease, or the presence and consequences of vascular comorbidities in a chronic inflammatory disease, such as multiple sclerosis. All these contributions have provided important, highly relevant clues for basic and translational neuroscience.
Collapse
Affiliation(s)
- Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Austria
| |
Collapse
|
5
|
Beltrán E, Paunovic M, Gebert D, Cesur E, Jeitler M, Höftberger R, Malotka J, Mader S, Kawakami N, Meinl E, Bradl M, Dornmair K, Lassmann H. Archeological neuroimmunology: resurrection of a pathogenic immune response from a historical case sheds light on human autoimmune encephalomyelitis and multiple sclerosis. Acta Neuropathol 2021; 141:67-83. [PMID: 33242149 PMCID: PMC7785560 DOI: 10.1007/s00401-020-02239-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 12/28/2022]
Abstract
Aim of our study was to identify the target auto-antigen in the central nervous system recognized by the immune system of a unique patient, who died more than 60 years ago from a disease with pathological changes closely resembling multiple sclerosis (MS), following a misguided immunization with lyophilized calf brain tissue. Total mRNA was isolated from formaldehyde fixed and paraffin embedded archival brain tissue containing chronic active inflammatory demyelinating lesions with inflammatory infiltrates rich in B-lymphocytes and plasma cells. Analysis of the transcriptome by next generation sequencing and reconstruction of the dominant antibody by bioinformatic tools revealed the presence of one strongly expanded B-cell clone, producing an autoantibody against a conformational epitope of myelin oligodendrocytes glycoprotein (MOG), similar to that recognized by the well characterized monoclonal anti-MOG antibody 8-18C5. The reconstructed antibody induced demyelination after systemic or intrathecal injection into animals with T-cell mediated encephalomyelitis. Our study suggests that immunization with bovine brain tissue in humans may-in a small subset of patients-induce a disease with an intermediate clinical and pathological presentation between MS and MOG-antibody associated inflammatory demyelinating disease (MOGAD).
Collapse
Affiliation(s)
- Eduardo Beltrán
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Manuela Paunovic
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - David Gebert
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Emine Cesur
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Markus Jeitler
- Core Facility Genomics, Medical University Vienna, Vienna, Austria
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Joachim Malotka
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Simone Mader
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Naoto Kawakami
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| |
Collapse
|
6
|
Jellinger KA. Neuropathology through the ages - My life between neurology and neuropathology. FREE NEUROPATHOLOGY 2020; 1:25. [PMID: 37283666 PMCID: PMC10240953 DOI: 10.17879/freeneuropathology-2020-2945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 06/08/2023]
|
7
|
Cheng HS, Tan WR, Low ZS, Marvalim C, Lee JYH, Tan NS. Exploration and Development of PPAR Modulators in Health and Disease: An Update of Clinical Evidence. Int J Mol Sci 2019; 20:E5055. [PMID: 31614690 PMCID: PMC6834327 DOI: 10.3390/ijms20205055] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 12/20/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that govern the expression of genes responsible for energy metabolism, cellular development, and differentiation. Their crucial biological roles dictate the significance of PPAR-targeting synthetic ligands in medical research and drug discovery. Clinical implications of PPAR agonists span across a wide range of health conditions, including metabolic diseases, chronic inflammatory diseases, infections, autoimmune diseases, neurological and psychiatric disorders, and malignancies. In this review we aim to consolidate existing clinical evidence of PPAR modulators, highlighting their clinical prospects and challenges. Findings from clinical trials revealed that different agonists of the same PPAR subtype could present different safety profiles and clinical outcomes in a disease-dependent manner. Pemafibrate, due to its high selectivity, is likely to replace other PPARα agonists for dyslipidemia and cardiovascular diseases. PPARγ agonist pioglitazone showed tremendous promises in many non-metabolic disorders like chronic kidney disease, depression, inflammation, and autoimmune diseases. The clinical niche of PPARβ/δ agonists is less well-explored. Interestingly, dual- or pan-PPAR agonists, namely chiglitazar, saroglitazar, elafibranor, and lanifibranor, are gaining momentum with their optimistic outcomes in many diseases including type 2 diabetes, dyslipidemia, non-alcoholic fatty liver disease, and primary biliary cholangitis. Notably, the preclinical and clinical development for PPAR antagonists remains unacceptably deficient. We anticipate the future design of better PPAR modulators with minimal off-target effects, high selectivity, superior bioavailability, and pharmacokinetics. This will open new possibilities for PPAR ligands in medicine.
Collapse
Affiliation(s)
- Hong Sheng Cheng
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Wei Ren Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
| | - Zun Siong Low
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
| | - Charlie Marvalim
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Justin Yin Hao Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
| | - Nguan Soon Tan
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
| |
Collapse
|
8
|
Zrzavy T, Kollaritsch H, Rommer PS, Boxberger N, Loebermann M, Wimmer I, Winkelmann A, Zettl UK. Vaccination in Multiple Sclerosis: Friend or Foe? Front Immunol 2019; 10:1883. [PMID: 31440255 PMCID: PMC6693409 DOI: 10.3389/fimmu.2019.01883] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 07/24/2019] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is a debilitating disease of the central nervous systems (CNS). Disease-modifying treatments (including immunosuppressive treatments) have shown positive effects on the disease course, but are associated with systemic consequences on the immune system and may increase the risk of infections and alter vaccine efficiency. Therefore, vaccination of MS patients is of major interest. Over the last years, vaccine hesitancy has steadily grown especially in Western countries, partly due to fear of sequelae arising from vaccination, especially neurological disorders. The interaction of vaccination and MS has been discussed for decades. In this review, we highlight the immunology of vaccination, provide a review of literature and discuss the clinical consideration of MS, vaccination and immunosuppression. In conclusion, there is consensus that MS cannot be caused by vaccines, neither by inactivated nor by live vaccines. However, particular attention should be paid to two aspects: First, in immunocompromised patients, live vaccines may lead to a stronger immune reaction with signs of the disease against which the patients have been vaccinated, albeit in weakened form. Second, protection provided by vaccination should be controlled in patients who have been vaccinated while receiving immunomodulatory or immunosuppressive treatment. In conclusion, there is evidence that systemic infections can worsen MS, thus vaccination will lower the risk of relapses by reducing the risk of infections. Therefore, vaccination should be in general recommended to MS patients.
Collapse
Affiliation(s)
- Tobias Zrzavy
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Herwig Kollaritsch
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Paulus S. Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Neuroimmunological Section, University of Rostock, Rostock, Germany
| | - Nina Boxberger
- Department of Neurology, Neuroimmunological Section, University of Rostock, Rostock, Germany
| | - Micha Loebermann
- Department of Tropical Medicine and Infectious Diseases, University of Rostock, Rostock, Germany
| | - Isabella Wimmer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | | | - Uwe K. Zettl
- Department of Neurology, Neuroimmunological Section, University of Rostock, Rostock, Germany
- Department of Neurology, University of Rostock, Rostock, Germany
| |
Collapse
|
9
|
Machado-Santos J, Saji E, Tröscher AR, Paunovic M, Liblau R, Gabriely G, Bien CG, Bauer J, Lassmann H. The compartmentalized inflammatory response in the multiple sclerosis brain is composed of tissue-resident CD8+ T lymphocytes and B cells. Brain 2018; 141:2066-2082. [PMID: 29873694 PMCID: PMC6022681 DOI: 10.1093/brain/awy151] [Citation(s) in RCA: 373] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/22/2018] [Accepted: 04/15/2018] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis is an inflammatory demyelinating disease in which active demyelination and neurodegeneration are associated with lymphocyte infiltrates in the brain. However, so far little is known regarding the phenotype and function of these infiltrating lymphocyte populations. In this study, we performed an in-depth phenotypic characterization of T and B cell infiltrates in a large set of multiple sclerosis cases with different disease and lesion stages and compared the findings with those seen in inflammatory, non-inflammatory and normal human controls. In multiple sclerosis lesions, we found a dominance of CD8+ T cells and a prominent contribution of CD20+ B cells in all disease courses and lesion stages, including acute multiple sclerosis cases with very short disease duration, while CD4+ T cells were sparse. A dominance of CD8+ T cells was also seen in other inflammatory controls, such as Rasmussen's encephalitis and viral encephalitis, but the contribution of B cells in these diseases was modest. Phenotypic analysis of the CD8+ T cells suggested that part of the infiltrating cells in active lesions proliferate, show an activated cytotoxic phenotype and are in part destroyed by apoptosis. Further characterization of the remaining cells suggest that CD8+ T cells acquire features of tissue-resident memory cells, which may be focally reactivated in active lesions of acute, relapsing and progressive multiple sclerosis, while B cells, at least in part, gradually transform into plasma cells. The loss of surface molecules involved in the egress of leucocytes from inflamed tissue, such as S1P1 or CCR7, and the upregulation of CD103 expression may be responsible for the compartmentalization of the inflammatory response in established lesions. Similar phenotypic changes of tissue-infiltrating CD8+ T cells were also seen in Rasmussen's encephalitis. Our data underline the potential importance of CD8+ T lymphocytes and B cells in the inflammatory response in established multiple sclerosis lesions. Tissue-resident T and B cells may represent guardians of previous inflammatory brain disease, which can be reactivated and sustain the inflammatory response, when they are re-exposed to their specific antigen.
Collapse
Affiliation(s)
- Joana Machado-Santos
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Etsuji Saji
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Anna R Tröscher
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Manuela Paunovic
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roland Liblau
- INSERM U1043 - CNRS UMR 5282, Centre de Physiopathologie Toulouse-Purpan, Université Toulouse III, Toulouse, F-31000, France
| | - Galina Gabriely
- Department of Neurology, Anne Romney Center for Neurologic Disease, Harvard Medical School, Boston, USA
| | | | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Höftberger R, Lassmann H. Inflammatory demyelinating diseases of the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2018; 145:263-283. [PMID: 28987175 PMCID: PMC7149979 DOI: 10.1016/b978-0-12-802395-2.00019-5] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inflammatory demyelinating diseases are a heterogeneous group of disorders, which occur against the background of an acute or chronic inflammatory process. The pathologic hallmark of multiple sclerosis (MS) is the presence of focal demyelinated lesions with partial axonal preservation and reactive astrogliosis. Demyelinated plaques are present in the white as well as gray matter, such as the cerebral or cerebellar cortex and brainstem nuclei. Activity of the disease process is reflected by the presence of lesions with ongoing myelin destruction. Axonal and neuronal destruction in the lesions is a major substrate for permanent neurologic deficit in MS patients. The MS pathology is qualitatively similar in different disease stages, such as relapsing remitting MS or secondary or primary progressive MS, but the prevalence of different lesion types differs quantitatively. Acute MS and Balo's type of concentric sclerosis appear to be variants of classic MS. In contrast, neuromyelitis optica (NMO) and spectrum disorders (NMOSD) are inflammatory diseases with primary injury of astrocytes, mediated by aquaporin-4 antibodies. Finally, we discuss the histopathology of other inflammatory demyelinating diseases such as acute disseminated encephalomyelitis and myelin oligodendrocyte glycoprotein antibody-associated demyelination. Knowledge of the heterogenous immunopathology in demyelinating diseases is important, to understand the clinical presentation and disease course and to find the optimal treatment for an individual patient.
Collapse
Affiliation(s)
- Romana Höftberger
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria,Correspondence to: Hans Lassmann, MD, Center for Brain Research, Medical University of Vienna, Spitalgasse, 1090 Vienna, Austria
| |
Collapse
|
11
|
Verrusio W, Magro VM, Summa ML, Angeloni U, Gueli N, Cacciafesta M. Acute disseminated encephalomyelitis in an elderly patient. Neurol Sci 2017; 38:2045-2047. [DOI: 10.1007/s10072-017-3044-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 06/19/2017] [Indexed: 10/19/2022]
|
12
|
Shrestha B, Jiang X, Ge S, Paul D, Chianchiano P, Pachter JS. Spatiotemporal resolution of spinal meningeal and parenchymal inflammation during experimental autoimmune encephalomyelitis. Neurobiol Dis 2017; 108:159-172. [PMID: 28844788 DOI: 10.1016/j.nbd.2017.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/10/2017] [Accepted: 08/18/2017] [Indexed: 01/14/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) induced by active immunization of C57BL/6 mice with peptide from myelin oligodendrocyte protein (MOG35-55), is a neuroinflammatory, demyelinating disease widely recognized as an animal model of multiple sclerosis (MS). Typically, EAE presents with an ascending course of paralysis, and inflammation that is predominantly localized to the spinal cord. Recent studies have further indicated that inflammation - in both MS and EAE - might initiate within the meninges and propagate from there to the underlying parenchyma. However, the patterns of inflammation within the respective meningeal and parenchymal compartments along the length of the spinal cord, and the progression with which these patterns develop during EAE, have yet to be detailed. Such analysis could hold key to identifying factors critical for spreading, as well as constraining, inflammation along the neuraxis. To address this issue, high-resolution 3-dimensional (3D) confocal microscopy was performed to visualize, in detail, the sequence of leukocyte infiltration at distinct regions of the spinal cord. High quality virtual slide scanning for imaging the entire spinal cord using epifluorescence was further conducted to highlight the directionality and relative degree of inflammation. Meningeal inflammation was found to precede parenchymal inflammation at all levels of the spinal cord, but did not develop equally or simultaneously throughout the subarachnoid space (SAS) of the meninges. Instead, meningeal inflammation was initially most obvious in the caudal SAS, from which it progressed to the immediate underlying parenchyma, paralleling the first signs of clinical disease in the tail and hind limbs. Meningeal inflammation could then be seen to extend in the caudal-to-rostral direction, followed by a similar, but delayed, trajectory of parenchymal inflammation. To additionally determine whether the course of ascending paralysis and leukocyte infiltration during EAE is reflected in differences in inflammatory gene expression by meningeal and parenchymal microvessels along the spinal cord, laser capture microdissection (LCM) coupled with gene expression profiling was performed. Expression profiles varied between these respective vessel populations at both the cervical and caudal levels of the spinal cord during disease progression, and within each vessel population at different levels of the cord at a given time during disease. These results reinforce a significant role for the meninges in the development and propagation of central nervous system inflammation associated with MS and EAE.
Collapse
Affiliation(s)
- Bandana Shrestha
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Xi Jiang
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Shujun Ge
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Debayon Paul
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Peter Chianchiano
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Joel S Pachter
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| |
Collapse
|
13
|
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: 179] [Impact Index Per Article: 22.4] [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
|
14
|
Mentis AFA, Dardiotis E, Grigoriadis N, Petinaki E, Hadjigeorgiou GM. Viruses and Multiple Sclerosis: From Mechanisms and Pathways to Translational Research Opportunities. Mol Neurobiol 2017; 54:3911-3923. [PMID: 28455696 DOI: 10.1007/s12035-017-0530-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/06/2017] [Indexed: 12/26/2022]
Abstract
Viruses are directly or indirectly implicated in multiple sclerosis (MS). Here, we review the evidence on the virus-related pathophysiology of MS, introduce common experimental models, and explore the ways in which viruses cause demyelination. By emphasizing knowledge gaps, we highlight future research directions for effective MS diagnostics and therapies: (i) identifying biomarkers for at-risk individuals, (ii) searching for direct evidence of specific causative viruses, (iii) establishing the contribution of host genetic factors and viruses, and (iv) investigating the contribution of immune regulation at extra-CNS sites. Research in these areas is likely to be facilitated by the application of high-throughput technologies, the development of systems-based bioinformatic approaches, careful selection of experimental models, and the acquisition of high-quality clinical material for tissue-based research.
Collapse
Affiliation(s)
- Alexios-Fotios A Mentis
- Department of Microbiology, University Hospital of Larissa, University of Thessaly, Larissa, Greece. .,The Johns Hopkins University, AAP, Baltimore, MD, USA.
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Nikolaos Grigoriadis
- B' Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Efthimia Petinaki
- Department of Microbiology, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Georgios M Hadjigeorgiou
- Department of Neurology, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| |
Collapse
|
15
|
Lassmann H, Bradl M. Multiple sclerosis: experimental models and reality. Acta Neuropathol 2017; 133:223-244. [PMID: 27766432 PMCID: PMC5250666 DOI: 10.1007/s00401-016-1631-4] [Citation(s) in RCA: 378] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 01/01/2023]
Abstract
One of the most frequent statements, provided in different variations in the introduction of experimental studies on multiple sclerosis (MS), is that "Multiple sclerosis is a demyelinating autoimmune disease and experimental autoimmune encephalomyelitis (EAE) is a suitable model to study its pathogenesis". However, so far, no single experimental model covers the entire spectrum of the clinical, pathological, or immunological features of the disease. Many different models are available, which proved to be highly useful for studying different aspects of inflammation, demyelination, remyelination, and neurodegeneration in the central nervous system. However, the relevance of results from such models for MS pathogenesis has to be critically validated. Current EAE models are mainly based on inflammation, induced by auto-reactive CD4+ T-cells, and these models reflect important aspects of MS. However, pathological data and results from clinical trials in MS indicate that CD8+ T-cells and B-lymphocytes may play an important role in propagating inflammation and tissue damage in established MS. Viral models may reflect key features of MS-like inflammatory demyelination, but are difficult to use due to their very complex pathogenesis, involving direct virus-induced and immune-mediated mechanisms. Furthermore, evidence for a role of viruses in MS pathogenesis is indirect and limited, and an MS-specific virus infection has not been identified so far. Toxic models are highly useful to unravel mechanisms of de- and remyelination, but do not reflect other important aspects of MS pathology and pathogenesis. For all these reasons, it is important to select the right experimental model to answer specific questions in MS research.
Collapse
Affiliation(s)
- Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| | - Monika Bradl
- Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| |
Collapse
|
16
|
Wang M, Xie Y, Zhong Y, Cen J, Wang L, Liu Y, Zhu Y, Tong L, Wei Q. Amelioration of Experimental Autoimmune Encephalomyelitis by Isogarcinol Extracted from Garcinia mangostana L. Mangosteen. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:9012-9021. [PMID: 27933873 DOI: 10.1021/acs.jafc.6b04145] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Isogarcinol is a new natural immunosuppressant that was extracted from Garcinia mangostana L. in our laboratory. Knowledge of its effects on treatable diseases and its mechanism of action is still very limited. In this study, we explored the therapeutic effect of isogarcinol in experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS). Treatment with oral 100 mg/kg isogarcinol markedly ameliorated clinical scores, alleviated inflammation and demyelination of the spinal cord, and reduced intracranial lesions in EAE mice. The percentages of Th cells and macrophages were also strongly reduced. Isogarcinol appeared to act by inhibiting T helper (Th) 1 and Th17 cell differentiation via the janus kinase/signal transducers and activators of transcription pathway and by impairing macrophage function. Our data suggest that isogarcinol has the potential to be an effective therapeutic agent of low toxicity for treating MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Mengqi Wang
- Gene Engineering and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, Beijing Normal University , Beijing 100875, People's Republic of China
| | - Yufei Xie
- Gene Engineering and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, Beijing Normal University , Beijing 100875, People's Republic of China
| | - Youxiu Zhong
- Gene Engineering and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, Beijing Normal University , Beijing 100875, People's Republic of China
- National Vaccine and Serum Institute, Beijing 100024, People's Republic of China
| | - Juren Cen
- Gene Engineering and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, Beijing Normal University , Beijing 100875, People's Republic of China
- Key Laboratory of Protection and Development Utilization of Tropical Crop Germplasm Resources, Ministry of Education, College of Landscape and Horticulture, Hainan University , Haikou 570228, People's Republic of China
| | - Lei Wang
- Gene Engineering and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, Beijing Normal University , Beijing 100875, People's Republic of China
| | - Yuanyuan Liu
- Gene Engineering and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, Beijing Normal University , Beijing 100875, People's Republic of China
| | - Ying Zhu
- Gene Engineering and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, Beijing Normal University , Beijing 100875, People's Republic of China
| | - Li Tong
- Gene Engineering and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, Beijing Normal University , Beijing 100875, People's Republic of China
| | - Qun Wei
- Gene Engineering and Biotechnology Beijing Key Laboratory, Department of Biochemistry and Molecular Biology, Beijing Normal University , Beijing 100875, People's Republic of China
| |
Collapse
|
17
|
Sack U, Boldt A, Mallouk N, Gruber R, Krenn V, Berger-Depincé AE, Conrad K, Tarnok A, Lambert C, Reinhold D, Fricke S. Cellular analyses in the monitoring of autoimmune diseases. Autoimmun Rev 2016; 15:883-9. [PMID: 27392502 DOI: 10.1016/j.autrev.2016.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/05/2016] [Indexed: 01/10/2023]
Affiliation(s)
- Ulrich Sack
- Universitätsklinikum Leipzig, Department für Diagnostik, Institut für Klinische Immunologie, Johannisallee 30, 04103, Leipzig, Germany.
| | - Andreas Boldt
- Universitätsklinikum Leipzig, Department für Diagnostik, Institut für Klinische Immunologie, Johannisallee 30, 04103, Leipzig, Germany.
| | - Nora Mallouk
- URCIP, CHU Saint-Etienne, Hôpital Nord, 42055 Saint-Etienne Cedex 02, France.
| | - Rudolf Gruber
- Institut für Labormedizin, Mikrobiologie und Krankenhaushygiene, Krankenhaus Barmherzige Brüder Regensburg, Prüfeninger Straße 86, 93049, Regensburg, Germany.
| | - Veit Krenn
- Medizinisches Versorgungszentrum für Histologie, Zytologie und Molekulare Diagnostik Trier, Max-Planck-Str. 5, 54296, Trier, Germany.
| | | | - Karsten Conrad
- Institut für Immunologie, Medizinische Fakultät "Carl Gustav Carus" der Technischen Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
| | - Attila Tarnok
- Universitätsklinikum Leipzig, Department für Diagnostik, Institut für Klinische Immunologie, Johannisallee 30, 04103, Leipzig, Germany.
| | - Claude Lambert
- Immunology laboratory, Pole de Biologie-Pathologie, University Hospital. CNRS UMR5307 Labo Georges Friedel (LGF); 42055 Saint-Etienne Cedex 02, France.
| | - Dirk Reinhold
- Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Institut für Molekulare und Klinische Immunologie, Leipziger Straße 44, 39120, Magdeburg, Germany.
| | - Stephan Fricke
- Fraunhofer Institut für Zelltherapie und Immunologie, Perlickstraße 1, 04103, Leipzig, Germany.
| |
Collapse
|