1
|
Stevenson M, Algarzae NK, Moussa C. Tyrosine kinases: multifaceted receptors at the intersection of several neurodegenerative disease-associated processes. FRONTIERS IN DEMENTIA 2024; 3:1458038. [PMID: 39221072 PMCID: PMC11361951 DOI: 10.3389/frdem.2024.1458038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
Tyrosine kinases (TKs) are catalytic enzymes activated by auto-phosphorylation that function by phosphorylating tyrosine residues on downstream substrates. Tyrosine kinase inhibitors (TKIs) have been heavily exploited as cancer therapeutics, primarily due to their role in autophagy, blood vessel remodeling and inflammation. This suggests tyrosine kinase inhibition as an appealing therapeutic target for exploiting convergent mechanisms across several neurodegenerative disease (NDD) pathologies. The overlapping mechanisms of action between neurodegeneration and cancer suggest that TKIs may play a pivotal role in attenuating neurodegenerative processes, including degradation of misfolded or toxic proteins, reduction of inflammation and prevention of fibrotic events of blood vessels in the brain. In this review, we will discuss the distinct roles that select TKs have been shown to play in various disease-associated processes, as well as identify TKs that have been explored as targets for therapeutic intervention and associated pharmacological agents being investigated as treatments for NDDs.
Collapse
Affiliation(s)
- Max Stevenson
- The Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC, United States
| | - Norah K. Algarzae
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Charbel Moussa
- The Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
2
|
Wu Y, Zhou H, Ci X, Lu J. Low T3 syndrome is associated with the severity of myelin oligodendrocyte glycoprotein antibody-associated disease exacerbation. Front Neurosci 2024; 18:1357633. [PMID: 38835837 PMCID: PMC11148359 DOI: 10.3389/fnins.2024.1357633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
Background Myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) is a rare autoimmune inflammatory disease of the central nervous system, (CNS) different from multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). While numerous studies have delved into the involvement of thyroid antibodies (ATAbs) and thyroid function in NMOSD and MS. The objective of this study is to explore the clinical significance of thyroid dysfunction and ATAbs abnormalities in adult patients with MOGAD. Methods 36 adult inpatients diagnosed with MOGAD and 47 sex- and age-matched healthy controls were enrolled. Patients were divided into two groups based on the presence or absence of low T3 syndrome. Demographics, clinical characteristics, and results of auxiliary examinations were compared across the subgroups. Moreover, an analysis was conducted to explore the correlations between thyroid hormone levels and Expanded Disability Status Scale (EDSS) scores. Results Thyroid dysfunction was notably more frequent in MOGAD patients than healthy controls (p < 0.0001), particularly low T3 syndrome (p=0.03). Furthermore, subgroup analyses revealed that the low T3 syndrome group exhibited higher EDSS scores and a higher proportion of individuals with EDSS scores > 3, in comparison to the non-low T3 syndrome group (p = 0.014, p = 0.046). However, no significant differences were observed in demographic characteristics, annual relapse rates, clinical phenotypes, laboratory and MRI results, and EEG abnormalities between the two groups. Additional Spearman's analysis showed significantly negative correlations between the TT3 and FT3 levels with EDSS scores (r = -0.367, p = 0.028; r = -0.377, p = 0.024). Typical brain lesions and paralateral ventricle lesions were significantly rare in patients with positive ATAbs compared to those with negative ATAbs (p = 0.0001, p = 0.03), although the incidence of ATAbs abnormalities did not differ significantly between MOGAD patients and healthy controls. Conclusions Overall, this study confirmed thyroid dysfunction, especially low T3 syndrome, is frequent in adult MOGAD patients. Patients with low T3 syndrome exhibited elevated EDSS scores and a significantly higher incidence of unfavorable condition. additionally, the correlation analysis model manifests that FT3 and TT3 levels were negatively correlated with EDSS scores. These evidences indicate that low T3 syndrome is associated with the severity of MOGAD exacerbation.
Collapse
Affiliation(s)
- Yuqing Wu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Zhou
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojiao Ci
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Lu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Parrilla GE, Gupta V, Wall RV, Salkar A, Basavarajappa D, Mirzaei M, Chitranshi N, Graham SL, You Y. The role of myelin in neurodegeneration: implications for drug targets and neuroprotection strategies. Rev Neurosci 2024; 35:271-292. [PMID: 37983528 DOI: 10.1515/revneuro-2023-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023]
Abstract
Myelination of axons in the central nervous system offers numerous advantages, including decreased energy expenditure for signal transmission and enhanced signal speed. The myelin sheaths surrounding an axon consist of a multi-layered membrane that is formed by oligodendrocytes, while specific glycoproteins and lipids play various roles in this formation process. As beneficial as myelin can be, its dysregulation and degeneration can prove detrimental. Inflammation, oxidative stress, and changes in cellular metabolism and the extracellular matrix can lead to demyelination of these axons. These factors are hallmark characteristics of certain demyelinating diseases including multiple sclerosis. The effects of demyelination are also implicated in primary degeneration in diseases such as glaucoma and Alzheimer's disease, as well as in processes of secondary degeneration. This reveals a relationship between myelin and secondary processes of neurodegeneration, including resultant degeneration following traumatic injury and transsynaptic degeneration. The role of myelin in primary and secondary degeneration is also of interest in the exploration of strategies and targets for remyelination, including the use of anti-inflammatory molecules or nanoparticles to deliver drugs. Although the use of these methods in animal models of diseases have shown to be effective in promoting remyelination, very few clinical trials in patients have met primary end points. This may be due to shortcomings or considerations that are not met while designing a clinical trial that targets remyelination. Potential solutions include diversifying disease targets and requiring concomitant interventions to promote rehabilitation.
Collapse
Affiliation(s)
- Gabriella E Parrilla
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Vivek Gupta
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Roshana Vander Wall
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Akanksha Salkar
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Devaraj Basavarajappa
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Mehdi Mirzaei
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Nitin Chitranshi
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Stuart L Graham
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
- Save Sight Institute, University of Sydney, 8 Macquarie St, Sydney, NSW 2000, Australia
| | - Yuyi You
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
- Save Sight Institute, University of Sydney, 8 Macquarie St, Sydney, NSW 2000, Australia
| |
Collapse
|
4
|
Oertel FC, Hastermann M, Paul F. Delimiting MOGAD as a disease entity using translational imaging. Front Neurol 2023; 14:1216477. [PMID: 38333186 PMCID: PMC10851159 DOI: 10.3389/fneur.2023.1216477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/23/2023] [Indexed: 02/10/2024] Open
Abstract
The first formal consensus diagnostic criteria for myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) were recently proposed. Yet, the distinction of MOGAD-defining characteristics from characteristics of its important differential diagnoses such as multiple sclerosis (MS) and aquaporin-4 antibody seropositive neuromyelitis optica spectrum disorder (NMOSD) is still obstructed. In preclinical research, MOG antibody-based animal models were used for decades to derive knowledge about MS. In clinical research, people with MOGAD have been combined into cohorts with other diagnoses. Thus, it remains unclear to which extent the generated knowledge is specifically applicable to MOGAD. Translational research can contribute to identifying MOGAD characteristic features by establishing imaging methods and outcome parameters on proven pathophysiological grounds. This article reviews suitable animal models for translational MOGAD research and the current state and prospect of translational imaging in MOGAD.
Collapse
Affiliation(s)
- Frederike Cosima Oertel
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Hastermann
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
5
|
Sharifian A, Varshosaz J, Aliomrani M, Kazemi M. Nose to brain delivery of ibudilast micelles for treatment of multiple sclerosis in an experimental autoimmune encephalomyelitis animal model. Int J Pharm 2023; 638:122936. [PMID: 37030640 DOI: 10.1016/j.ijpharm.2023.122936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Multiple sclerosis is a chronic inflammatory disease of the central nervous system ultimate to neurodegeneration and demyelination. Ibudilast is a phosphodiesterase inhibitor, effective on the function of glial cells and lymphocytes, and inhibits the release of TNF-α by inflammatory cells. Dysregulation of glia is one of the most important pathological causes of MS. Therefore, ibudilast as a glial attenuator can be a useful treatment. The objective of the present study was to investigate the effect of nasal spray of polydopamine coated micelles of surfactin, a biosurfactant, loaded with ibudilast on its brain targeted delivery and effectiveness in remylination and neuroprotection in animal model of MS. In animal studies the micelles were administrated intranasally in different doses of 10, 25 and 50 mg/kg/day in C57/BL6 mice immunized by experimental autoimmune encephalomyelitis (EAE) model. The results of Luxol fast blue staining indicated increment in myelin fiber percent more significantly (p<0.05) in the groups treated with the polydopamine coated micelles (PDAM) compared to nasal spray of free drug or oral administration. These formulations also increased expression of Mbp, Olig2 and Mog genes in the corpus callosum. These results suggest a positive outcome of polydopamine coated micelles loaded with ibudilast in active MS as an anti-inflammatory and neuroprotective agent.
Collapse
Affiliation(s)
- Akram Sharifian
- Department of Pharmaceutics, Novel Drug Delivery Systems Research Centre, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Jaleh Varshosaz
- Department of Pharmaceutics, Novel Drug Delivery Systems Research Centre, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mehdi Aliomrani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammad Kazemi
- Department of Genetics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
6
|
Amatruda M, Harris K, Matis A, Davies AL, McElroy D, Clark M, Linington C, Desai R, Smith KJ. Oxygen treatment reduces neurological deficits and demyelination in two animal models of multiple sclerosis. Neuropathol Appl Neurobiol 2023; 49:e12868. [PMID: 36520661 PMCID: PMC10107096 DOI: 10.1111/nan.12868] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 11/07/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
AIMS The objective of the study is to explore the importance of tissue hypoxia in causing neurological deficits and demyelination in the inflamed CNS, and the value of inspiratory oxygen treatment, using both active and passive experimental autoimmune encephalomyelitis (EAE). METHODS Normobaric oxygen treatment was administered to Dark Agouti rats with either active or passive EAE, compared with room air-treated, and naïve, controls. RESULTS Severe neurological deficits in active EAE were significantly improved after just 1 h of breathing approximately 95% oxygen. The improvement was greater and more persistent when oxygen was applied either prophylactically (from immunisation for 23 days), or therapeutically from the onset of neurological deficits for 24, 48, or 72 h. Therapeutic oxygen for 72 h significantly reduced demyelination and the integrated stress response in oligodendrocytes at the peak of disease, and protected from oligodendrocyte loss, without evidence of increased oxidative damage. T-cell infiltration and cytokine expression in the spinal cord remained similar to that in untreated animals. The severe neurological deficit of animals with passive EAE occurred in conjunction with spinal hypoxia and was significantly reduced by oxygen treatment initiated before their onset. CONCLUSIONS Severe neurological deficits in both active and passive EAE can be caused by hypoxia and reduced by oxygen treatment. Oxygen treatment also reduces demyelination in active EAE, despite the autoimmune origin of the disease.
Collapse
Affiliation(s)
- Mario Amatruda
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kate Harris
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Alina Matis
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Andrew L Davies
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Daniel McElroy
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, Glasgow Biomedical Research Centre, Glasgow, UK
| | - Michael Clark
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Christopher Linington
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, Glasgow Biomedical Research Centre, Glasgow, UK
| | - Roshni Desai
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Kenneth J Smith
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
7
|
Zeng XD, Chen H, Hu WG. Myelin Oligodendrocyte Glycoprotein (MOG) antibody-associated meningoencephalitis due to Mycoplasma pneumoniae infection. Neurol Res 2023; 45:124-126. [PMID: 36111693 DOI: 10.1080/01616412.2022.2124794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Although myelin oligodendrocyte glycoprotein (MOG) antibody-associated disorders include a wide spectrum of syndromes, manifestations with meningoencephalitis symptoms due to M. pneumoniae infection were quite infrequent. We admitted an 8-years-old girl who presented with recurrent fever accompanied by headache and mild cough, her Cerebral spinal fluid polynucleated cells was elevated and progressively higher, her cranial MRI showed meningeal enhancement initially and multiple intracranial lesions later, serum M. pneumoniae-IgM and MOG-IgG were positive, she was diagnosed with MOG-IgG associated meningoencephalitis due to M. pneumoniae infection, the treatment consisted of intravenous immunoglobulin, glucocorticoid, and erythromycin, then she was completely recovered.
Collapse
Affiliation(s)
- Xiang-Dong Zeng
- Department of Pediatric Neurology Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hui Chen
- Department of Pediatric Neurology Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wen-Guang Hu
- Department of Pediatric Neurology Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
8
|
Munera M, Buendía E, Sanchez A, Viasus D, Sanchez J. AQP4 as a vintage autoantigen: what do we know till now? Heliyon 2022; 8:e12132. [PMID: 36506380 PMCID: PMC9730132 DOI: 10.1016/j.heliyon.2022.e12132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/09/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- M. Munera
- Medical Research Group (GINUMED), Universitary Corporation Rafael Nuñez, Colombia,Corresponding author.
| | - E. Buendía
- Faculty of Medicine, University of Cartagena, Cartagena, Colombia,Department of Internal Medicine, Centro Hospitalario Serena del Mar, Cartagena, Colombia,Clinical and Biomedical Research Group, Faculty of Medicine, University of Cartagena, Colombia
| | - A. Sanchez
- Faculty of Medicine, University of Cartagena, Cartagena, Colombia,Clinical and Biomedical Research Group, Faculty of Medicine, University of Cartagena, Colombia
| | - D. Viasus
- Division of Health Sciences, Universidad del Norte, Barranquilla, Colombia
| | - J. Sanchez
- Group of Clinical and Experimental Allergy (GACE), IPS Universitaria, University of Antioquia, Medellín, Colombia
| |
Collapse
|
9
|
Vaishnav J, Khan F, Yadav M, Parmar N, Buch H, Jadeja SD, Dwivedi M, Begum R. V-set domain containing T-cell activation inhibitor-1 (VTCN1): A potential target for the treatment of autoimmune diseases. Immunobiology 2022; 227:152274. [PMID: 36095871 DOI: 10.1016/j.imbio.2022.152274] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/13/2022] [Accepted: 09/01/2022] [Indexed: 11/23/2022]
Abstract
Autoimmunity eventuates when the immune system attacks self-molecules as a result of the breakdown in immune tolerance. Targeting autoimmune diseases via immunomodulation has become an essential strategy in today's era. A B7 superfamily member immune checkpoint, the V-set domain containing T-cell activation inhibitor-1 (VTCN1), also known as B7-H4, B7S1, and B7x, is involved in negatively regulating T-cell activation. VTCN1 transcript has been reported in various lymphoid and non-lymphoid tissues, but its protein expression is restricted, indicating its translational regulation. Dysregulation of VTCN1 has resulted in the exacerbation of various autoimmune diseases. Moreover, increased soluble form of VTCN1 in the patient's sera positively correlates with the disease progression and severity. The current review summarizes all the reports till date, unfolding the role of VTCN1 in various autoimmune diseases and its therapeutic potential.
Collapse
Affiliation(s)
- Jayvadan Vaishnav
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Farheen Khan
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Madhu Yadav
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Nishant Parmar
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Hiteshree Buch
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Shahnawaz D Jadeja
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Mitesh Dwivedi
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India; C. G. Bhakta Institute of Biotechnology, Uka Tarsadia University, Tarsadi, Surat 394350, Gujarat, India
| | - Rasheedunnisa Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India.
| |
Collapse
|
10
|
Coexistence of Myelin Oligodendrocyte Glycoprotein Immunoglobulin G and Neuronal or Glial Antibodies in the Central Nervous System: A Systematic Review. Brain Sci 2022; 12:brainsci12080995. [PMID: 36009058 PMCID: PMC9405704 DOI: 10.3390/brainsci12080995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/23/2022] [Accepted: 07/24/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Myelin oligodendrocyte glycoprotein immunoglobulin G (MOG-IgG) has been considered a diagnostic marker for patients with demyelinating disease, termed “MOG-IgG associated disorder” (MOGAD). Recently, the coexistence of MOG-IgG and other neuronal or glial antibodies has attracted extensive attention from clinicians. In this article, we systematically review the characteristics of MOG-IgG-related antibody coexistence syndrome. Methods: Two authors independently searched PubMed for relevant studies published before October 2021. We also manually searched the references of each related article. The appropriateness of the included studies was assessed by reading the titles, abstracts, and full texts if necessary. Results: Thirty-five relevant publications that met our inclusion criteria were finally included, of which fourteen were retrospective studies and twenty-one were case reports. A total of 113 patients were reported to show the coexistence of MOG-IgG and neuronal or glial antibodies. Additionally, 68.14% of patients were double positive for MOG-IgG and N-Methyl-D-Aspartate Receptor-IgG (NMDAR-IgG), followed by 23.01% of patients who were double positive for MOG-IgG and aquaporin4-IgG (AQP4-IgG). Encephalitis was the predominant phenotype when MOG-IgG coexisted with NMDAR-IgG, probably accompanied by imaging features of demyelination. Patients with dual positivity for MOG-IgG and AQP4-IgG experienced more severe disease and more frequent relapses. The coexistence of MOG-IgG and antibodies other than NMDAR-IgG and AQP4-IgG was extremely rare, and the clinical presentations were diverse and atypical. Except for patients who were double positive for MOG-IgG and AQP4-IgG, most patients with multiple antibodies had a good prognosis. Conclusions: MOG-IgG may coexist with neuronal or glial antibodies. Expanded screening for neuronal or glial antibodies should be performed in patients with atypical clinical and radiological features.
Collapse
|
11
|
Nefodov OO, Belenichev IF, Fedchenko MP, Popazova OO, Ryzhenko VP, Morozova OV. Evaluation of methods of modeling and formation of experimental allergic encephalomyelitis. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.77361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: Experimental autoimmune (allergic) encephalomyelitis (EAE) induced by intradermal injection of homogenate of the brain, spinal cord and peripheral nerve with Freund’s stimulator, refers to a true autoimmune disease of the nervous system.
Materials and methods: Experimental studies were conducted on white nonlinear rats. To induce experimental allergic encephalomyelitis (EAE), homologous brain homogenates was used, which leads among other drugs (homologous, heterogeneous brain and spinal cord homogenates) by encephalitogenity. The connective tissue of the animal’s tail base was injected with a mixture of encephalitogenic suspension of 0.1 ml per 100 g of the body weight.
Results and discussion: According to the results, in the rats, there was weight loss, and the abnormal neurological symptoms were found on an average of 10–12th days. Our experimental studies on the formation of EAE were confirmed morphologically by electron microscopy.
Conclusion: Thus, the use of this technique allowed us to obtain a simulated pathologic condition of multiple sclerosis in the form of experimental allergic encephalomyelitis and can be used in future studies to identify appropriate laws, the extent and nature of changes in the immune and nervous systems of the body when inducing experimental pathological conditions.
Collapse
|
12
|
Chen X, Ren Y, Zhang Y, Lu W. Comparative study of AQP4 antibody-related diseases and MOG antibody-related diseases among the population in Hunan, China. Acta Neurol Belg 2021; 121:1649-1659. [PMID: 32737792 DOI: 10.1007/s13760-020-01455-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/23/2020] [Indexed: 01/31/2023]
Abstract
To compare the clinical, imaging, and prognostic characteristics of AQP4 antibody-related diseases and MOG antibody-related diseases. The clinical data of 56 AQP4 antibody-positive patients and 14 MOG antibody-positive patients in the Second Xiangya Hospital of Central South University from June 2016 to June 2019 were collected. 92.9% of the patients with positive AQP4 antibody were females and 64.3% of patients with positive MOG antibody were females (P = 0.004). Patients with positive AQP4 antibody were more likely to have limb movement (P < 0.001) or limb sensory dysfunction (P < 0.001), and were more likely to have limb twitching (P = 0.036). In addition, AQP4 antibody-positive patients were more likely to have positive ANA (P = 0.013) and SSA antibody (P = 0.029), Ro-52 antibody (P = 0.047), immunoglobulin (P = 0.007), thyroid antibody (P = 0.007), abnormal C3 (P = 0.011), abnormal C4 (P = 0.014) than MOG antibody-positive patients. The involvement rate of head in MOG antibody-positive patients was higher than AQP4 antibody-positive patients (P = 0.029). The severity of clinical symptoms in AQP4-positive patients was usually more serious than that in MOG-positive patients (P < 0.001). The residual neurological deficit after treatment in AQP4-positive group was usually more serious than that in MOG-positive group (P < 0.001). AQP4 antibody-positive patients had a higher prevalence in women than MOG antibody-positive patients, and AQP4 antibody-positive patients were more likely to have spinal cord involvement symptoms and connective tissue antibody abnormalities. The EDSS score of them were higher than that of MOG antibody-positive patients after treatment, and the prognosis was worse.
Collapse
Affiliation(s)
- Xiqian Chen
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yijun Ren
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yiliu Zhang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Lu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
13
|
Lai QL, Zhang YX, Cai MT, Zheng Y, Qiao S, Fang GL, Shen CH. Efficacy and safety of immunosuppressive therapy in myelin oligodendrocyte glycoprotein antibody-associated disease: a systematic review and meta-analysis. Ther Adv Neurol Disord 2021; 14:17562864211054157. [PMID: 34790259 PMCID: PMC8591780 DOI: 10.1177/17562864211054157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 09/30/2021] [Indexed: 12/29/2022] Open
Abstract
Background: A considerable number of patients with myelin oligodendrocyte glycoprotein antibody–associated disease (MOGAD) will experience a relapse, but the effect of maintenance therapies on re-attack rates is currently unknown. Objective: To investigate the efficacy and safety of immunosuppressive therapy for preventing disease relapses in patients with MOGAD, including rituximab (RTX), mycophenolate mofetil (MMF), and azathioprine (AZA). Methods: English-language studies published prior to August 31, 2020, were searched in the NCBI (PubMed), ISI Web of Science, and the Cochrane Library databases. Patient characteristics, treatment regimens, outcome measures, and adverse effects were retrieved. Results: We enrolled 11 studies in the final meta-analysis, including 346 patients with MOGAD. RTX therapy was demonstrated to result in reduced mean annualized relapse rate (ARR) by 1.35 (95% confidence interval (CI): 0.85–1.85) and reduced mean Expanded Disability Status Scale score by 0.80 (95% CI: 0.53–1.08) in patients with MOGAD. MMF therapy was associated with the mean ARR decreasing by 0.83 (95% CI: 0.31–1.35), and AZA was related to the mean ARR decreasing by 1.71 (95% CI: 0.83–2.58). The reported discontinuation rates of RTX, MMF, and AZA therapy due to adverse effects were 3/197 (1.52%), 3/39 (7.69%), and 4/37 (10.81%), respectively. Conclusion: The study provided evidence to support the efficacy of RTX, MMF, and AZA on the preventive treatment in patients with MOGAD. However, large randomized controlled trials are still needed in the future.
Collapse
Affiliation(s)
- Qi-Lun Lai
- Department of Neurology, Zhejiang Hospital, Hangzhou, China
| | - Yin-Xi Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Meng-Ting Cai
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Zheng
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Song Qiao
- Department of Neurology, Zhejiang Hospital, Hangzhou, China
| | - Gao-Li Fang
- Department of Neurology, Zhejiang Chinese Medicine and Western Medicine Integrated Hospital, Hangzhou, China
| | - Chun-Hong Shen
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| |
Collapse
|
14
|
Marignier R, Hacohen Y, Cobo-Calvo A, Pröbstel AK, Aktas O, Alexopoulos H, Amato MP, Asgari N, Banwell B, Bennett J, Brilot F, Capobianco M, Chitnis T, Ciccarelli O, Deiva K, De Sèze J, Fujihara K, Jacob A, Kim HJ, Kleiter I, Lassmann H, Leite MI, Linington C, Meinl E, Palace J, Paul F, Petzold A, Pittock S, Reindl M, Sato DK, Selmaj K, Siva A, Stankoff B, Tintore M, Traboulsee A, Waters P, Waubant E, Weinshenker B, Derfuss T, Vukusic S, Hemmer B. Myelin-oligodendrocyte glycoprotein antibody-associated disease. Lancet Neurol 2021; 20:762-772. [PMID: 34418402 DOI: 10.1016/s1474-4422(21)00218-0] [Citation(s) in RCA: 285] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 06/07/2021] [Accepted: 07/01/2021] [Indexed: 10/20/2022]
Abstract
Myelin-oligodendrocyte glycoprotein antibody-associated disease (MOGAD) is a recently identified autoimmune disorder that presents in both adults and children as CNS demyelination. Although there are clinical phenotypic overlaps between MOGAD, multiple sclerosis, and aquaporin-4 antibody-associated neuromyelitis optica spectrum disorder (NMOSD) cumulative biological, clinical, and pathological evidence discriminates between these conditions. Patients should not be diagnosed with multiple sclerosis or NMOSD if they have anti-MOG antibodies in their serum. However, many questions related to the clinical characterisation of MOGAD and pathogenetic role of MOG antibodies are still unanswered. Furthermore, therapy is mainly based on standard protocols for aquaporin-4 antibody-associated NMOSD and multiple sclerosis, and more evidence is needed regarding how and when to treat patients with MOGAD.
Collapse
Affiliation(s)
- Romain Marignier
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-Inflammation, and Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle, Hôpital Neurologique Pierre Wertheimer, Bron, France; Centre des Neurosciences de Lyon, INSERM 1028 et CNRS UMR5292, Lyon, France; Université Claude Bernard Lyon 1, Lyon, France.
| | - Yael Hacohen
- Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Alvaro Cobo-Calvo
- Centre d'Esclerosi Múltiple de Catalunya, Department of Neurology/Neuroimmunology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anne-Katrin Pröbstel
- Neurologic Clinic and Policlinic and Research Center for Clinical Neuroimmunology and Neuroscience, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Orhan Aktas
- Medical Faculty, Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Harry Alexopoulos
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Pia Amato
- IRCCS Fondazione Don Carlo Gnocchi, University of Florence, Florence, Italy
| | - Nasrin Asgari
- Institute of Regional Health Research and Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Brenda Banwell
- Division of Child Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neurology and Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey Bennett
- Department of Neurology and Department of Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Fabienne Brilot
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Marco Capobianco
- Regional Multiple Sclerosis Centre, Department of Neurology, University Hospital San Luigi, Orbassano, Italy
| | - Tanuja Chitnis
- Department of Pediatric Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Olga Ciccarelli
- Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Kumaran Deiva
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital Bicêtre, Pediatric Neurology Department, National Referral Center for Rare Inflammatory Brain and Spinal Diseases, Université Paris-Sud, and UMR 1184-CEA-IDMIT, Center for Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin Bicêtre, France
| | - Jérôme De Sèze
- Department of Neurology, Strasbourg University Hospital and Clinical Investigation Center, INSERM 1434, Strasbourg, France
| | - Kazuo Fujihara
- Department of Multiple Sclerosis Therapeutics, Fukushima Medical University School of Medicine Koriyama, Japan; Multiple Sclerosis and Neuromyelitis Optica Center, Southern TOHOKU Research Institute for Neuroscience, Koriyama, Japan
| | - Anu Jacob
- Division of Multiple Sclerosis and Autoimmune Neurology, Neurological Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates; Walton Centre National Health Service Trust, Liverpool, UK
| | - Ho Jin Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Ingo Kleiter
- Marianne-Strauß-Klinik, Behandlungszentrum Kempfenhausen für Multiple Sklerose Kranke, Berg, Germany; Department of Neurology, Ruhr-University Bochum, Bochum, Germany
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Maria-Isabel Leite
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Christopher Linington
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospitals, Ludwig Maximilian University Munich, Germany
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Friedemann Paul
- NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, and Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Axel Petzold
- Moorfields Eye Hospital and National Hospital for Neurology and Neurosurgery, London, UK; University College London Queen Square Institute of Neurology, London, UK; National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, UK
| | - Sean Pittock
- Department of Neurology and Laboratory Medicine and Pathology, and Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Douglas Kazutoshi Sato
- Brain Institute of Rio Grande do Sul and School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Krzysztof Selmaj
- University of Warmia and Mazury, Olsztyn, Poland; Center of Neurology, Łódź, Poland
| | - Aksel Siva
- Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Department of Neurology, Istanbul, Turkey
| | - Bruno Stankoff
- Sorbonne Université, Paris Brain Institute, ICM, CNRS, Inserm, and Saint Antoine Hospital, APHP, Paris, France
| | - Mar Tintore
- Centre d'Esclerosi Múltiple de Catalunya, Department of Neurology/Neuroimmunology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anthony Traboulsee
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Patrick Waters
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Emmanuelle Waubant
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Brian Weinshenker
- Department of Neurology and Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Tobias Derfuss
- Neurologic Clinic and Policlinic and Research Center for Clinical Neuroimmunology and Neuroscience, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sandra Vukusic
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-Inflammation, and Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle, Hôpital Neurologique Pierre Wertheimer, Bron, France; Centre des Neurosciences de Lyon, INSERM 1028 et CNRS UMR5292, Lyon, France; Université Claude Bernard Lyon 1, Lyon, France
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
15
|
Comparative analysis of clinical and imaging data between patients with myelin oligodendrocyte glycoprotein antibody disease and patients with aquaporin 4 antibody-positive neuromyelitis optica spectrum disorder. J Neurol 2021; 269:1641-1650. [PMID: 34383114 DOI: 10.1007/s00415-021-10749-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND We aimed to compare the clinical data, laboratory findings, and imaging characteristics of myelin oligodendrocyte glycoprotein antibody disease (MOGAD) and aquaporin 4 antibody (AQP4)-positive neuromyelitis optica spectrum disorder (NMOSD), as detailed comparative analyses of laboratory data for both diseases are rare. METHODS Our retrospective study compared the clinical data, laboratory findings, and imaging characteristics of 118 AQP4-positive patients with first-episode NMOSD and 25 patients with first-episode MOGAD. Logistic regression was used to determine the factors that differentiated MOGAD and AQP4-positive NMOSD. RESULTS There were significant differences in age, symptoms, recurrence rate, laboratory indicators, and imaging examinations between patients with MOGAD and patients with AQP4-positive NMOSD. Patients with MOGAD were younger and had higher levels of uric acid than those with AQP4-positive NMOSD. The proportion of cortical gray matter/juxtacortical white matter lesions was significantly higher in the MOGAD group than in the NMOSD group. Logistic regression revealed that young age [odds ratio (OR) = 0.947, 95% confidence interval (CI) = 0.905-0.99], high uric acid level (OR = 1.016, 95% CI = 1.006-1.027), and cortical gray matter/juxtacortical white matter involvement (OR = 3.889, 95% CI = 1.048-14.442) were significantly related to MOGAD. CONCLUSION The multivariate analysis of the present study demonstrated that age, uric acid level, and the presence of lesions in the cortical gray matter/juxtacortical white matter can aid in distinguishing patients with AQP4-positive NMOSD from those with MOGAD. These factors may also aid in determining which patients should be tested for antibodies.
Collapse
|
16
|
Greiner T, Kipp M. What Guides Peripheral Immune Cells into the Central Nervous System? Cells 2021; 10:cells10082041. [PMID: 34440810 PMCID: PMC8392645 DOI: 10.3390/cells10082041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS), an immune-mediated demyelinating disease of the central nervous system (CNS), initially presents with a relapsing-remitting disease course. During this early stage of the disease, leukocytes cross the blood–brain barrier to drive the formation of focal demyelinating plaques. Disease-modifying agents that modulate or suppress the peripheral immune system provide a therapeutic benefit during relapsing-remitting MS (RRMS). The majority of individuals with RRMS ultimately enter a secondary progressive disease stage with a progressive accumulation of neurologic deficits. The cellular and molecular basis for this transition is unclear and the role of inflammation during the secondary progressive disease stage is a subject of intense and controversial debate. In this review article, we discuss the following main hypothesis: during both disease stages, peripheral immune cells are triggered by CNS-intrinsic stimuli to invade the brain parenchyma. Furthermore, we outline the different neuroanatomical routes by which peripheral immune cells might migrate from the periphery into the CNS.
Collapse
|
17
|
Chen X, Yang X, Yuan P, Jin R, Bao L, Qiu X, Liu S, Liu T, Gooding JJ, Chen W, Liu G, Bai Y, Liu S, Jin Y. Modular immune-homeostatic microparticles promote immune tolerance in mouse autoimmune models. Sci Transl Med 2021; 13:13/584/eaaw9668. [PMID: 33692135 DOI: 10.1126/scitranslmed.aaw9668] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/14/2020] [Accepted: 01/26/2021] [Indexed: 12/29/2022]
Abstract
The therapeutic goal for autoimmune diseases is disease antigen-specific immune tolerance without nonspecific immune suppression. However, it is a challenge to induce antigen-specific immune tolerance in a dysregulated immune system. In this study, we developed immune-homeostatic microparticles (IHMs) that treat multiple mouse models of autoimmunity via induction of apoptosis in activated T cells and reestablishment of regulatory T cells. Specifically, in an experimental model of colitis, IHMs rapidly released monocyte chemotactic protein-1 after intravenous administration, which recruited activated T cells and then induced their apoptosis by conjugated Fas ligand on the IHM surface. This triggered professional macrophages to ingest apoptotic T cells and produce high quantities of transforming growth factor-β, which drove regulatory T cell differentiation. Furthermore, the modular design of IHMs allowed IHMs to be engineered with the autoantigen peptides that can reduce disease in an experimental autoimmune encephalomyelitis mouse model and a nonobese diabetic mouse model. This was accomplished by sustained release of the autoantigens after induction of T cell apoptosis and transforming growth factor-β production by macrophages, which promoted to establish an immune tolerant environment. Thus, IHMs may be an efficient therapeutic strategy for autoimmune diseases through induction of apoptosis and reestablishment of tolerant immune responses.
Collapse
Affiliation(s)
- Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China.
| | - Xiaoshan Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Pingyun Yuan
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Ronghua Jin
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Lili Bao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xinyu Qiu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Siying Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Tao Liu
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - John Justin Gooding
- School of Chemistry, Australian Centre for NanoMedicine and ARC Australian Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney 2052, Australia
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Guozhen Liu
- Graduate School of Biomedical Engineering, ARC Centre of Excellence for Nanoscale BioPhotonics and Australian Centre for NanoMedicine, Faculty of Engineering, University of New South Wales, Sydney 2052, Australia
| | - Yongkang Bai
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
18
|
Abstract
Animal models with high translational validity are essential tools in understanding disease pathogenesis and in the development of therapeutic strategies. Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system characterized by progressive neurological deficits and socioeconomic burden. Experimental autoimmune encephalomyelitis (EAE) is the most extensively utilized animal model of MS, with well-characterized rodent and non-human primate variants. The EAE model is typically induced by either active immunization with myelin-derived proteins or peptides in adjuvant or by passive transfer of activated myelin-specific CD4+ T lymphocytes. To date, the EAE model has been an essential tool in the development of at least seven U.S. Food and Drug Administration (FDA)-approved immunomodulatory drugs for the treatment of MS, including glatiramer acetate, fingolimod, and natalizumab. However, the translational validity of the EAE model is frequently compromised due to poor study design, inconsistent clinical scoring endpoints, and inappropriate statistical calculations. No single animal model accurately reflects the complexity of human MS pathogenesis. Beyond EAE, multiple additional animal models are described, including Theiler's murine encephalomyelitis virus and cuprizone-induced demyelination, which facilitate the study of pathogen-induced CNS autoimmunity and remyelination, respectively. This overview summarizes several of the most frequently used animal models of MS and highlights key factors that significantly influence the experimental outcome and affect translational validity. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Paul Smith
- Incyte Research Institute, Wilmington, Delaware
| |
Collapse
|
19
|
Horellou P, de Chalus A, Giorgi L, Leroy C, Chrétien P, Hacein-Bey-Abina S, Bourgeois C, Mariette X, Serguera C, Le Grand R, Deiva K. Regulatory T Cells Increase After rh-MOG Stimulation in Non-Relapsing but Decrease in Relapsing MOG Antibody-Associated Disease at Onset in Children. Front Immunol 2021; 12:679770. [PMID: 34220827 PMCID: PMC8243969 DOI: 10.3389/fimmu.2021.679770] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Background Myelin oligodendrocytes glycoprotein (MOG) antibody-associated disease (MOGAD) represent 25% of pediatric acquired demyelinating syndrome (ADS); 40% of them may relapse, mimicking multiple sclerosis (MS), a recurrent and neurodegenerative ADS, which is MOG-Abs negative. Aims To identify MOG antigenic immunological response differences between MOGAD, MS and control patients, and between relapsing versus non-relapsing subgroups of MOGAD. Methods Three groups of patients were selected: MOGAD (n=12 among which 5 relapsing (MOGR) and 7 non-relapsing (MOGNR)), MS (n=10) and control patients (n=7). Peripheral blood mononuclear cells (PBMC) collected at the time of the first demyelinating event were cultured for 48 h with recombinant human (rh)-MOG protein (10 μg/ml) for a specific stimulation or without stimulation as a negative control. The T cells immunophenotypes were analyzed by flow cytometry. CD4+ T cells, T helper (Th) cells including Th1, Th2, and Th17 were analyzed by intracellular staining of cytokines. Regulatory T cells (Tregs, Foxp3+), CD45RA-Foxp3+ Tregs and subpopulation naive Tregs (CD45RA+Foxp3int), effector Tregs (CD45RA-Foxp3high) and non-suppressive Tregs (CD45RA-Foxp3int) proportions were determined. Results The mean onset age of each group, ranging from 9.9 to 13.8, and sex ratio, were similar between MOGR, MOGNR, MS and control patients as analyzed by one-way ANOVA and Chi-square test. When comparing unstimulated to rh-MOG stimulated T cells, a significant increase in the proportion of Th2 and Th17 cells was observed in MOGAD. Increase of Th17 cells was significant in MOGNR (means: 0.63 ± 0.15 vs. 1.36 ± 0.43; Wilcoxon-test p = 0.03) but not in MOGR. CD4+ Tregs were significantly increased in MOGNR (means: 3.51 ± 0.7 vs. 4.59 ± 1.33; Wilcoxon-test p = 0.046) while they decreased in MOGR. CD45RA-Foxp3+ Tregs were significantly decreased in MOGR (means: 2.37 ± 0.23 vs. 1.99 ± 0.17; paired t-test p = 0.021), but not in MOGNR. MOGR showed the highest ratio of effector Tregs/non suppressive-Tregs, which was significantly higher than in MOGNR. Conclusions Our findings suggest that CD4+ Th2 and Th17 cells are involved in the pathophysiology of MOGAD in children. The opposite response of Tregs to rh-MOG in MOGNR, where CD4+ Tregs increased, and in MOGR, where CD45RA-Foxp3+ Tregs decreased, suggests a probable loss of tolerance toward MOG autoantigen in MOGR which may explain relapses in this recurrent pediatric autoimmune disease.
Collapse
Affiliation(s)
- Philippe Horellou
- Université Paris-Saclay, CEA, INSERM UMR 1184, Le Kremlin Bicêtre, France
| | - Aliénor de Chalus
- Université Paris-Saclay, CEA, INSERM UMR 1184, Le Kremlin Bicêtre, France.,Assistance Publique-Hôpitaux de Paris, Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, Le Kremlin Bicêtre, France
| | - Laetitia Giorgi
- Université Paris-Saclay, CEA, INSERM UMR 1184, Le Kremlin Bicêtre, France.,Assistance Publique-Hôpitaux de Paris, Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, Le Kremlin Bicêtre, France
| | - Carole Leroy
- Université Paris-Saclay, CEA, INSERM UMR 1184, Le Kremlin Bicêtre, France.,Assistance Publique-Hôpitaux de Paris, Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, Le Kremlin Bicêtre, France
| | - Pascale Chrétien
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France.,Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Salima Hacein-Bey-Abina
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France.,Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
| | | | - Xavier Mariette
- Université Paris-Saclay, CEA, INSERM UMR 1184, Le Kremlin Bicêtre, France.,Department of Rheumatology, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin-Bicêtre, France
| | - Ché Serguera
- Institut du Cerveau (ICM), Hôpital Pitié-Salpêtrière, Paris, France
| | - Roger Le Grand
- Université Paris-Saclay, CEA, INSERM UMR 1184, Le Kremlin Bicêtre, France
| | - Kumaran Deiva
- Université Paris-Saclay, CEA, INSERM UMR 1184, Le Kremlin Bicêtre, France.,Assistance Publique-Hôpitaux de Paris, Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, Le Kremlin Bicêtre, France.,National Referral Center for Rare Inflammatory and Auto-Immune Brain and Spinal Diseases (MIRCEM), Pediatric Neurology Department, Hôpital Bicêtre, AP-HP, Le Kremlin Bicêtre, France
| |
Collapse
|
20
|
Nitsch L, Petzinna S, Zimmermann J, Schneider L, Krauthausen M, Heneka MT, Getts DR, Becker A, Müller M. Astrocyte-specific expression of interleukin 23 leads to an aggravated phenotype and enhanced inflammatory response with B cell accumulation in the EAE model. J Neuroinflammation 2021; 18:101. [PMID: 33906683 PMCID: PMC8080359 DOI: 10.1186/s12974-021-02140-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/26/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Interleukin 23 is a critical cytokine in the pathogenesis of multiple sclerosis. But the local impact of interleukin 23 on the course of neuroinflammation is still not well defined. To further characterize the effect of interleukin 23 on CNS inflammation, we recently described a transgenic mouse model with astrocyte-specific expression of interleukin 23 (GF-IL23 mice). The GF-IL23 mice spontaneously develop a progressive ataxic phenotype with cerebellar tissue destruction and inflammatory infiltrates with high amounts of B cells most prominent in the subarachnoid and perivascular space. METHODS To further elucidate the local impact of the CNS-specific interleukin 23 synthesis in autoimmune neuroinflammation, we induced a MOG35-55 experimental autoimmune encephalomyelitis (EAE) in GF-IL23 mice and WT mice and analyzed the mice by histology, flow cytometry, and transcriptome analysis. RESULTS We were able to demonstrate that local interleukin 23 production in the CNS leads to aggravation and chronification of the EAE course with a severe paraparesis and an ataxic phenotype. Moreover, enhanced multilocular neuroinflammation was present not only in the spinal cord, but also in the forebrain, brainstem, and predominantly in the cerebellum accompanied by persisting demyelination. Thereby, interleukin 23 creates a pronounced proinflammatory response with accumulation of leukocytes, in particular B cells, CD4+ cells, but also γδ T cells and activated microglia/macrophages. Furthermore, transcriptome analysis revealed an enhanced proinflammatory cytokine milieu with upregulation of lymphocyte activation markers, co-stimulatory markers, chemokines, and components of the complement system. CONCLUSION Taken together, the GF-IL23 model allowed a further breakdown of the different mechanisms how IL-23 drives neuroinflammation in the EAE model and proved to be a useful tool to further dissect the impact of interleukin 23 on neuroinflammatory models.
Collapse
Affiliation(s)
- Louisa Nitsch
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany.
| | - Simon Petzinna
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Julian Zimmermann
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Linda Schneider
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany.,Department of Surgery, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Marius Krauthausen
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Daniel R Getts
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Albert Becker
- Department of Neuropathology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Marcus Müller
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany.,School of Molecular Bioscience, University of Sydney, Sydney, Australia
| |
Collapse
|
21
|
Brill L, Ganelin-Cohen E, Dabby R, Rabinowicz S, Zohar-Dayan E, Rein N, Aloni E, Karmon Y, Vaknin-Dembinsky A. Age-Related Clinical Presentation of MOG-IgG Seropositivity in Israel. Front Neurol 2021; 11:612304. [PMID: 33584514 PMCID: PMC7874097 DOI: 10.3389/fneur.2020.612304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Introduction: Myelin oligodendrocyte glycoprotein (MOG) antibody associated disorders (MOGAD) have been recognized over the past 10 years as distinct inflammatory, demyelinating diseases of the central nervous system (CNS). Antibodies against MOG are found mostly in patients with optic neuritis (ON), acute disseminated encephalomyelitis (ADEM), and aquaporin-4 antibody (AQP4-abs)-seronegative neuromyelitis optica spectrum disorders (NMOSD). However, data on the disease course and disability outcomes of these patients are scarce. Aim: To describe clinical and paraclinical features associated with MOG antibodies (abs) in a cohort of patients in Israel, and to assess baseline prognostic features of MOG-ab-associated diseases after a first acute demyelinating event. Methods: MOG-abs were identified in serum using a cell-based assay, and clinical data were collected from the patients' medical records. Results: Of 683 patients with demyelinating diseases tested for MOG-abs, 53 were positive (7.7%), with ON the most common presenting phenotype (68%). The age range of MOG-abs seropositive patients was 1-66 years, with increased prevalence in children (19% compared to 6.7% in adults) (p < 0.01). The highest prevalence of seropositivity was observed in children aged younger than 10 years (25.5%), followed by those aged 31-40 years (16.6%). Conclusions: MOGAD are distinct autoimmune diseases that occurs at all stages of life with a significantly higher prevalence in children; the main clinical presenting phenotype in the entire cohort is ON and young children most often presented with ON or ADEM. Our data highlight the need for repeated evaluation of MOG-abs in patients with acquired CNS demyelinating disorders, especially in children under 10 and adults between 31 and 40 years of age.
Collapse
Affiliation(s)
- Livnat Brill
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes Ginges Center for Neurogenetics, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Esther Ganelin-Cohen
- Schneider Children's Medical Center, Institute of Pediatric Neurology, Affiliated With Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ron Dabby
- Department of Neurology, Edith Wolfson Medical Center, Holon, Affiliated With Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Shira Rabinowicz
- Pediatric Neurology Unit, Sheba Medical Center, Ramat Gan, Israel. Sackler School of Medicine, The Edmond and Lilly Safra Children's Hospital, Tel-Aviv University, Tel-Aviv, Israel
| | - Efrat Zohar-Dayan
- Pediatric Neurology Unit, Sheba Medical Center, Ramat Gan, Israel. Sackler School of Medicine, The Edmond and Lilly Safra Children's Hospital, Tel-Aviv University, Tel-Aviv, Israel
| | - Netaniel Rein
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes Ginges Center for Neurogenetics, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eyal Aloni
- Department of Ophthalmology, Barzilai University Medical Center, Ashkelon, Israel
| | - Yuval Karmon
- Department of Neurology, Meir General Hospital, Kfar Saba, Israel
| | - Adi Vaknin-Dembinsky
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes Ginges Center for Neurogenetics, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
22
|
Armangue T, Capobianco M, de Chalus A, Laetitia G, Deiva K. E.U. paediatric MOG consortium consensus: Part 3 - Biomarkers of paediatric myelin oligodendrocyte glycoprotein antibody-associated disorders. Eur J Paediatr Neurol 2020; 29:22-31. [PMID: 33191096 DOI: 10.1016/j.ejpn.2020.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022]
Abstract
A first episode of acquired demyelinating disorder (ADS) in children is a diagnostic challenge as different diseases can express similar clinical features. Recently, antibodies against myelin oligodendrocyte glycoprotein (MOG) have emerged as a new ADS biomarker, which clearly allow the identification of monophasic and relapsing ADS forms different from MS predominantly in children. Due to the novelty of this antibody there are still challenges and controversies about its pathogenicity and best technique to detect it. In this manuscript we will discuss the recommendations and caveats on MOG antibody assays, role in the pathogenesis, and additionally discuss the usefulness of other potential new biomarkers in MOG-antibody associated disorders (MOGAD).
Collapse
Affiliation(s)
- Thaís Armangue
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; Pediatric Neuroimmunology Unit, Neurology Department, Sant Joan de Déu (SJD) Children's Hospital, University of Barcelona, Barcelona, Spain.
| | - Marco Capobianco
- Department of Neurology and Regional Multiple Sclerosis Centre, University Hospital San Luigi Gonzaga, Orbassano, Italy
| | - Aliénor de Chalus
- Assistance Publique-Hôpitaux de Paris, Pediatric Neurology Department, University Hospitals Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
| | - Giorgi Laetitia
- Assistance Publique-Hôpitaux de Paris, Pediatric Neurology Department, University Hospitals Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
| | - Kumaran Deiva
- Assistance Publique-Hôpitaux de Paris, Pediatric Neurology Department, University Hospitals Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France; French Reference Network of Rare Inflammatory Brain and Spinal Diseases, Le Kremlin Bicêtre, France and European Reference Network-RITA, France
| | | |
Collapse
|
23
|
Hegen H, Reindl M. Recent developments in MOG-IgG associated neurological disorders. Ther Adv Neurol Disord 2020; 13:1756286420945135. [PMID: 33029200 PMCID: PMC7521831 DOI: 10.1177/1756286420945135] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
In the past few years, acquired demyelinating syndromes of the central nervous system associated with antibodies against myelin oligodendrocyte glycoprotein (MOG) have evolved into a new inflammatory disease entity distinct from neuromyelitis optica spectrum disorders or multiple sclerosis. The meticulous clinical description of patients with MOG IgG antibodies (MOG-IgG) has been achieved by development and use of highly specific cell-based assays. MOG-IgG associated disorders comprise a wide spectrum of syndromes ranging from acute disseminated encephalomyelitis predominantly in children to optic neuritis or myelitis mostly in adults. In recent studies, phenotype of MOG-IgG associated disorders has further broadened with the description of cases of brainstem encephalitis, encephalitis with seizures and overlap syndromes with other types of autoimmune encephalitis. In this review, we provide an overview of current knowledge of MOG-IgG associated disorders, describe the clinical presentations identified, highlight differences from neuromyelitis optica spectrum disorders and multiple sclerosis, summarize clinical outcome and concepts of immune treatment, depict the underlying mechanisms of antibody pathogenicity and provide the methodological essentials of MOG-IgG assays.
Collapse
Affiliation(s)
- Harald Hegen
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, A-6020, Austria
| |
Collapse
|
24
|
Hofer LS, Ramberger M, Gredler V, Pescoller AS, Rostásy K, Sospedra M, Hegen H, Berger T, Lutterotti A, Reindl M. Comparative Analysis of T-Cell Responses to Aquaporin-4 and Myelin Oligodendrocyte Glycoprotein in Inflammatory Demyelinating Central Nervous System Diseases. Front Immunol 2020; 11:1188. [PMID: 32625206 PMCID: PMC7311656 DOI: 10.3389/fimmu.2020.01188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/13/2020] [Indexed: 12/30/2022] Open
Abstract
Autoantibodies against aquaporin-4 (AQP4-Ab) and myelin oligodendrocyte glycoprotein (MOG-Ab) are associated with rare central nervous system inflammatory demyelinating diseases like neuromyelitis optica spectrum disorders (NMOSD). Previous studies have shown that not only antibodies, but also autoreactive T-cell responses against AQP4 are present in NMOSD. However, no study has yet analyzed the presence of MOG reactive T-cells in patients with MOG antibodies. Therefore, we compared AQP4 and MOG specific peripheral T-cell response in individuals with AQP4-Ab (n = 8), MOG-Ab (n = 10), multiple sclerosis (MS, n = 8), and healthy controls (HC, n = 14). Peripheral blood mononuclear cell cultures were stimulated with eight AQP4 and nine MOG peptides selected from previous studies and a tetanus toxoid peptide mix as a positive control. Antigen-specific T-cell responses were assessed using the carboxyfluorescein diacetate succinimidyl ester proliferation assay and the detection of granulocyte macrophage colony-stimulating factor (GM-CSF), interferon (IFN)-ɤ and interleukin (IL)-4, IL-6, and IL-17A in cell culture supernatants. Additionally, human leukocyte antigen (HLA)-DQ and HLA-DR genotyping of all participants was performed. We classified a T-cell response as positive if proliferation (measured by a cell division index ≥3) was confirmed by the secretion of at least one cytokine. Reactivity against AQP4 peptides was observed in many groups, but the T-cell response against AQP4 p156-170 was present only in patients with AQP4-Ab (4/8, 50%) and absent in patients with MOG-Ab, MS and HC (corrected p = 0.02). This AQP4 p156-170 peptide specific T-cell response was significantly increased in participants with AQP4-Ab compared to those without [Odds ratio (OR) = 59.00, 95% confidence interval-CI 2.70–1,290.86]. Moreover, T-cell responses against at least one AQP4 peptide were also more frequent in participants with AQP4-Ab (OR = 11.45, 95% CI 1.24–106.05). We did not observe any significant differences for the other AQP4 peptides or any MOG peptide. AQP4-Ab were associated with HLA DQB1*02 (OR = 5.71, 95% CI 1.09–30.07), DRB1*01 (OR = 9.33, 95% CI 1.50–58.02) and DRB1*03 (OR = 6.75, 95% CI = 1.19–38.41). Furthermore, HLA DRB1*01 was also associated with the presence of AQP4 p156-170 reactive T-cells (OR = 31.67, 95% CI 1.30–772.98). To summarize, our findings suggest a role of AQP4-specific, but not MOG-specific T-cells, in NMOSD.
Collapse
Affiliation(s)
- Livia Sophie Hofer
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Melanie Ramberger
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria.,Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Viktoria Gredler
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Sophie Pescoller
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kevin Rostásy
- Paediatric Neurology, Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany
| | - Mireia Sospedra
- Department of Neuroimmunology, University of Zurich, Zurich, Switzerland
| | - Harald Hegen
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Andreas Lutterotti
- Department of Neuroimmunology, University of Zurich, Zurich, Switzerland
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
25
|
Foiadelli T, Gastaldi M, Scaranzin S, Franciotta D, Savasta S. Seizures and myelin oligodendrocyte glycoprotein (MOG) antibodies: Two paradigmatic cases and a review of the literature. Mult Scler Relat Disord 2020; 41:102011. [DOI: 10.1016/j.msard.2020.102011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/10/2020] [Accepted: 02/14/2020] [Indexed: 12/13/2022]
|
26
|
James J, Jose J, Gafoor VA, Smita B, Balaram N, Ramachandran A. Clinical Course, Imaging Characteristics, and Therapeutic Response in Myelin Oligodendrocyte Glycoprotein Antibody Disease: A Case Series. J Neurosci Rural Pract 2020; 11:205-210. [PMID: 32140030 PMCID: PMC7055612 DOI: 10.1055/s-0040-1701371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Myelin oligodendrocyte glycoprotein (MOG) antibody disease is a novel central nervous system autoimmune disorder which forms part of aquaporin 4 (AQP-4) negative, neuromyelitis optica (NMO) spectrum disorder. It has a distinct clinical profile, neuroimaging features and courses from AQP-4 positive NMO and multiple sclerosis. This article is a case series of six patients with MOG antibody disease with longitudinal follow-up for up to 8 months.
Collapse
Affiliation(s)
- Joe James
- Department of Neurology, Government Medical College Kozhikode, Kerala, India
| | - James Jose
- Department of Neurology, Government Medical College Kozhikode, Kerala, India
| | - V Abdul Gafoor
- Department of Neurology, Government Medical College Kozhikode, Kerala, India
| | - B Smita
- Department of Neurology, Government Medical College Kozhikode, Kerala, India
| | - Neetha Balaram
- Department of Neurology, Government Medical College Kozhikode, Kerala, India
| | - Aparna Ramachandran
- Department of Neurology, Government Medical College Kozhikode, Kerala, India
| |
Collapse
|
27
|
Abstract
Anti-myelin oligodendrocyte glycoprotein (MOG) antibodies (MOG-Abs) were first detected by immunoblot and enzyme-linked immunosorbent assay nearly 30 years ago, but their association with multiple sclerosis (MS) was not specific. Use of cell-based assays with native MOG as the substrate enabled identification of a group of MOG-Ab-positive patients with demyelinating phenotypes. Initially, MOG-Abs were reported in children with acute disseminated encephalomyelitis (ADEM). Further studies identified MOG-Abs in adults and children with ADEM, seizures, encephalitis, anti-aquaporin-4-antibody (AQP4-Ab)-seronegative neuromyelitis optica spectrum disorder (NMOSD) and related syndromes (optic neuritis, myelitis and brainstem encephalitis), but rarely in MS. This shift in our understanding of the diagnostic assays has re-invigorated the examination of MOG-Abs and their role in autoimmune and demyelinating disorders of the CNS. The clinical phenotypes, disease courses and responses to treatment that are associated with MOG-Abs are currently being defined. MOG-Ab-associated disease is different to AQP4-Ab-positive NMOSD and MS. This Review provides an overview of the current knowledge of MOG, the metrics of MOG-Ab assays and the clinical associations identified. We collate the data on antibody pathogenicity and the mechanisms that are thought to underlie this. We also highlight differences between MOG-Ab-associated disease, NMOSD and MS, and describe our current understanding on how best to treat MOG-Ab-associated disease.
Collapse
|
28
|
Fransen NL, Crusius JBA, Smolders J, Mizee MR, van Eden CG, Luchetti S, Remmerswaal EBM, Hamann J, Mason MRJ, Huitinga I. Post-mortem multiple sclerosis lesion pathology is influenced by single nucleotide polymorphisms. Brain Pathol 2020; 30:106-119. [PMID: 31228212 PMCID: PMC6916567 DOI: 10.1111/bpa.12760] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/10/2019] [Indexed: 12/25/2022] Open
Abstract
Over the last few decades, several common single nucleotide polymorphisms (SNPs) have been identified that correlate with clinical outcome in multiple sclerosis (MS), but the pathogenic mechanisms underlying the clinical effects of these SNPs are unknown. This is in part because of the difficulty in the functional translation of genotype into disease-relevant mechanisms. Building on our recent work showing the association of clinical disease course with post-mortem MS lesion characteristics, we hypothesized that SNPs that correlate with clinical disease course would also correlate with specific MS lesion characteristics in autopsy tissue. To test this hypothesis, 179 MS brain donors from the Netherlands Brain Bank MS autopsy cohort were genotyped for 102 SNPs, selected based on their reported associations with clinical outcome or their associations with genes that show differential gene expression in MS lesions. Three SNPs linked to MS clinical severity showed a significant association between the genotype and either the proportion of active lesions (rs2234978/FAS and rs11957313/KCNIP1) or the proportion of mixed active/inactive lesions (rs8056098/CLEC16A). Three SNPs linked to MS pathology-associated genes showed a significant association with either proportion of active lesions (rs3130253/MOG), incidence of cortical gray matter lesions (rs1064395/NCAN) or the proportion of remyelinated lesions (rs5742909/CTLA4). In addition, rs2234978/FAS T-allele carriers showed increased FAS gene expression levels in perivascular T cells and perilesional oligodendrocytes, cell types that have been implicated in MS lesion formation. Thus, by combining pathological characterization of MS brain autopsy tissue with genetics, we now start to translate genotypes linked to clinical outcomes in MS into mechanisms involved in MS lesion pathogenesis.
Collapse
Affiliation(s)
- Nina L. Fransen
- Department of NeuroimmunologyThe Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
| | - Jakob B. A. Crusius
- Laboratory for Immunogenetics, Department of Medical Microbiology and Infection ControlAmsterdam UMC, VU UniversityAmsterdamThe Netherlands
| | - Joost Smolders
- Department of NeuroimmunologyThe Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
- MS Center CWZ, Department of NeurologyCanisius Wilhelmina HospitalNijmegenThe Netherlands
| | - Mark R. Mizee
- Department of NeuroimmunologyThe Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
| | - Corbert G. van Eden
- Department of NeuroimmunologyThe Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
| | - Sabina Luchetti
- Department of NeuroimmunologyThe Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
| | - Ester B. M. Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection & Immunity InstituteAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Renal Transplant Unit, Department of Internal Medicine, Amsterdam Infection & Immunity InstituteAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Jörg Hamann
- Department of NeuroimmunologyThe Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity InstituteAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Matthew R. J. Mason
- Department of NeuroimmunologyThe Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
| | - Inge Huitinga
- Department of NeuroimmunologyThe Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
| |
Collapse
|
29
|
Bando Y, Geisler JG. Disease modifying mitochondrial uncouplers, MP101, and a slow release ProDrug, MP201, in models of Multiple Sclerosis. Neurochem Int 2019; 131:104561. [DOI: 10.1016/j.neuint.2019.104561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 09/23/2019] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
|
30
|
Banerjee PP, Pang L, Soldan SS, Miah SM, Eisenberg A, Maru S, Waldman A, Smith EA, Rosenberg-Hasson Y, Hirschberg D, Smith A, Ablashi DV, Campbell KS, Orange JS. KIR2DL4-HLAG interaction at human NK cell-oligodendrocyte interfaces regulates IFN-γ-mediated effects. Mol Immunol 2019; 115:39-55. [PMID: 30482463 PMCID: PMC6543535 DOI: 10.1016/j.molimm.2018.09.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 09/11/2018] [Accepted: 09/30/2018] [Indexed: 12/12/2022]
Abstract
Interactions between germline-encoded natural killer (NK) cell receptors and their respective ligands on tumorigenic or virus-infected cells determine NK cell cytotoxic activity and/or cytokine secretion. NK cell cytokine responses can be augmented in and can potentially contribute to multiple sclerosis (MS), an inflammatory disease of the central nervous system focused upon the oligodendrocytes (OLs). To investigate mechanisms by which NK cells may contribute to MS pathogenesis, we developed an in vitro human model of OL-NK cell interaction. We found that activated, but not resting human NK cells form conjugates with, and mediate cytotoxicity against, human oligodendrocytes. NK cells, when in conjugate with OLs, rapidly synthesize and polarize IFN-γ toward the OLs. IFN-γ is capable of reducing myelin oligodendrocyte and myelin associated glycoproteins (MOG and MAG) content. This activity is independent of MHC class-I mediated inhibition via KIR2DL1, but dependent upon the interaction between NK cell-expressed KIR2DL4 and its oligodendrocyte-expressed ligand, HLA-G. NK cells from patients with MS express higher levels of IFN-γ following conjugation to OLs, more actively promote in vitro reduction of MOG and MAG and have higher frequencies of the KIR2DL4 positive population. These data collectively suggest a mechanism by which NK cells can promote pathogenic effects upon OLs.
Collapse
Affiliation(s)
- P P Banerjee
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA.
| | - L Pang
- Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| | - S S Soldan
- The Wistar Institute, 3601 Spruce St., Philadelphia, PA 19104, USA
| | - S M Miah
- Blood Cell Development and Function Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - A Eisenberg
- The Children's Hospital of Philadelphia Research Institute, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - S Maru
- The Children's Hospital of Philadelphia Research Institute, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - A Waldman
- The Children's Hospital of Philadelphia Research Institute, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - E A Smith
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| | - Y Rosenberg-Hasson
- Human Immune Monitoring Center, Stanford School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA
| | - D Hirschberg
- Human Immune Monitoring Center, Stanford School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA
| | - A Smith
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| | - D V Ablashi
- Human Herpes Virus 6 Foundation, 1482 East Valley Road, Suite 619 Santa Barbara, CA 93108, USA
| | - K S Campbell
- Blood Cell Development and Function Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - J S Orange
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| |
Collapse
|
31
|
DuBois JC, Ray AK, Gruber RC, Zhang Y, Aflakpui R, Macian-Juan F, Shafit-Zagardo B. Akt3-Mediated Protection Against Inflammatory Demyelinating Disease. Front Immunol 2019; 10:1738. [PMID: 31404142 PMCID: PMC6669559 DOI: 10.3389/fimmu.2019.01738] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 07/09/2019] [Indexed: 12/30/2022] Open
Abstract
Akt is a serine/threonine protein kinase that plays a major role in regulating multiple cellular processes. While the isoforms Akt1 and Akt2 are involved in apoptosis and insulin signaling, respectively, the role for Akt3 remains uncertain. Akt3 is predominantly expressed in the brain, and total deletion of Akt3 in mice results in a reduction in brain size and neurodegeneration following injury. Previously, we found that Akt3-/- mice have a significantly worse clinical course during myelin-oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE), an animal model in which autoreactive immune cells enter the CNS, resulting in inflammation, demyelination, and axonal injury. Spinal cords of Akt3-/- mice are severely demyelinated and have increased inflammation compared to WT, suggesting a neuroprotective role for Akt3 during EAE. To specifically address the role of Akt3 in neuroinflammation and maintaining neuronal integrity, we used several mouse strains with different manipulations to Akt3. During EAE, Akt3 Nmf350 mice (with enhanced Akt3 kinase activity) had lower clinical scores, a lag in disease onset, a delay in the influx of inflammatory cells into the CNS, and less axonal damage compared to WT mice. A significant increased efficiency of differentiation toward FOXP3 expressing iTregs was also observed in Akt3 Nmf350 mice relative to WT. Mice with a conditional deletion of Akt3 in CD4+ T-cells had an earlier onset of EAE symptoms, increased inflammation in the spinal cord and brain, and had fewer FOXP3+ cells and FOXP3 mRNA expression. No difference in EAE outcome was observed when Akt3 expression was deleted in neurons (Syn1-CKO). These results indicate that Akt3 signaling in T-cells and not neurons is necessary for maintaining CNS integrity during an inflammatory demyelinating disease.
Collapse
MESH Headings
- Animals
- Biomarkers
- Demyelinating Diseases/etiology
- Demyelinating Diseases/metabolism
- Demyelinating Diseases/pathology
- Disease Models, Animal
- Disease Susceptibility
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Fluorescent Antibody Technique
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Immunohistochemistry
- Immunophenotyping
- Mice
- Mice, Knockout
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Spinal Cord/immunology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Juwen C. DuBois
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Alex K. Ray
- Department of Microbiology and Immunology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ross C. Gruber
- Multiple Sclerosis and Neuroinflammation Research, Sanofi, Framingham, MA, United States
| | - Yongwei Zhang
- Department of Cell Biology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ranee Aflakpui
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Fernando Macian-Juan
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Bridget Shafit-Zagardo
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
32
|
Ramya L. Role of N-glycan in the structural changes of myelin oligodendrocyte glycoprotein and its complex with an antibody. J Biomol Struct Dyn 2019; 38:1649-1658. [PMID: 31057084 DOI: 10.1080/07391102.2019.1614999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Myelin Oligodendrocyte Glycoprotein (MOG) is found on the external surface of the myelin sheath and plays an important role in neurodegenerative diseases. It was observed that the protein MOG acts as an autoantigen and results in demyelination. The cause for the sudden change of protein to be autoantigen is still unclear. Here we present the molecular dynamics simulation studies of MOG in both unbound and bound states with an antibody. Both these systems were studied in the absence and presence of N-glycan in order to understand the effect of glycosylation in the MOG conformational changes. The results indicate that the glycosylation decreases the flexibility of protein in both free and bound states. Glycan influence the interaction of the complex with the water molecules whereas free protein MOG interaction with water molecules was not affected by the glycosylation. Glycan changes the 310 helices adjacent to the antibody interacting epitope MOG35-55 to turns.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- L Ramya
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamilnadu, India
| |
Collapse
|
33
|
Yakimov V, Schweiger F, Zhan J, Behrangi N, Horn A, Schmitz C, Hochstrasser T, Kipp M. Continuous cuprizone intoxication allows active experimental autoimmune encephalomyelitis induction in C57BL/6 mice. Histochem Cell Biol 2019; 152:119-131. [DOI: 10.1007/s00418-019-01786-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2019] [Indexed: 12/13/2022]
|
34
|
Oligodendrocyte degeneration and concomitant microglia activation directs peripheral immune cells into the forebrain. Neurochem Int 2019; 126:139-153. [PMID: 30867127 DOI: 10.1016/j.neuint.2019.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 01/21/2019] [Accepted: 03/06/2019] [Indexed: 11/22/2022]
Abstract
Brain-intrinsic degenerative cascades are a proposed factor driving inflammatory lesion formation in multiple sclerosis (MS) patients. We recently showed that encephalitogenic lymphocytes are recruited to the sites of active demyelination induced by cuprizone. Here, we investigated whether cuprizone-induced oligodendrocyte and myelin pathology is sufficient to trigger peripheral immune cell recruitment into the forebrain. We show that early cuprizone-induced white matter lesions display a striking similarity to early MS lesions, i.e., oligodendrocyte degeneration, microglia activation and absence of severe lymphocyte infiltration. Such early cuprizone lesions are sufficient to trigger peripheral immune cell recruitment secondary to subsequent EAE (experimental autoimmune encephalomyelitis) induction. The lesions are characterized by discontinuation of the perivascular glia limitans, focal axonal damage, and perivascular astrocyte pathology. Time course studies showed that the severity of cuprizone-induced lesions positively correlates with the extent of peripheral immune cell recruitment. Furthermore, results of genome-wide array analyses suggest that moesin is integral for early microglia activation in cuprizone and MS lesions. This study underpins the significance of brain-intrinsic degenerative cascades for immune cell recruitment and, in consequence, MS lesion formation.
Collapse
|
35
|
Yadav SK, Soin D, Ito K, Dhib-Jalbut S. Insight into the mechanism of action of dimethyl fumarate in multiple sclerosis. J Mol Med (Berl) 2019; 97:463-472. [PMID: 30820593 DOI: 10.1007/s00109-019-01761-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 12/26/2022]
Abstract
Dimethyl fumarate (DMF) is an oral, disease-modifying agent for the treatment of relapsing-remitting multiple sclerosis (RRMS). However, details regarding its mode of action are still emerging. It is believed that the mode of action of DMF involves both nuclear factor erythroid-derived 2-related factor (Nrf2)-dependent and independent pathways, which lead to an anti-inflammatory immune response due to type II myeloid cell and Th2 cell differentiation and neuroprotection. In this review, we will focus on the molecular and signaling effects of DMF that lead to changes in peripheral immune cell composition and function, alteration in CNS cell-specific functions, and effect on the blood-brain barrier.
Collapse
Affiliation(s)
- Sudhir Kumar Yadav
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Devika Soin
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Kouichi Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Suhayl Dhib-Jalbut
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
| |
Collapse
|
36
|
Mahmoodi M, Amiri H, Ayoobi F, Rahmani M, Taghipour Z, Ghavamabadi RT, Jafarzadeh A, Sankian M. Carvacrol ameliorates experimental autoimmune encephalomyelitis through modulating pro- and anti-inflammatory cytokines. Life Sci 2019; 219:257-263. [DOI: 10.1016/j.lfs.2018.11.051] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 11/17/2022]
|
37
|
Abstract
Purpose of review The purpose of this review is to summarize current understanding regarding the treatment of myelin oligodendrocyte glycoprotein antibody (MOG-Ab)-associated demyelination in children. Emphasis is placed on the unique obstacles we face when predicting the risk of relapse and the important implications of such challenges when planning treatment protocols. Recent findings MOG-Abs are consistently identified in a range of acquired demyelinating syndromes (ADS) in adults and children with a clinical phenotype distinct of MS and AQP4-Ab neuromyelitis optica spectrum disorder. Although initially thought to be associated with a benign disease, recent reports of children who are treatment-resistant and developed progressive disability over time raise important questions about how children with relapsing MOG-Ab disease should be managed. Summary MOG-Abs are common in children with ADS with both monophasic and relapsing disease courses. Treatment of patients with MOG-Ab-associated demyelination includes management of acute relapses and chronic immunotherapy for those with relapsing disease. Emerging consensus supports distinction of treatment strategies from those typically used for relapsing remitting MS, and several groups debate whether to follow treatment protocols akin to those for AQP4-Ab NMOSD. A key challenge remains predicting the severity of the disease at onset. Collaborative international consensus to derive shared clinical evaluative platforms standardized biological and neuroimaging protocols which can be used clinically, and partnered research programs are required to advance personalized treatment for children with MOG-Ab-associated demyelination.
Collapse
|
38
|
Glatigny S, Bettelli E. Experimental Autoimmune Encephalomyelitis (EAE) as Animal Models of Multiple Sclerosis (MS). Cold Spring Harb Perspect Med 2018; 8:cshperspect.a028977. [PMID: 29311122 DOI: 10.1101/cshperspect.a028977] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is a multifocal demyelinating disease of the central nervous system (CNS) leading to the progressive destruction of the myelin sheath surrounding axons. It can present with variable clinical and pathological manifestations, which might reflect the involvement of distinct pathogenic processes. Although the mechanisms leading to the development of the disease are not fully understood, numerous evidences indicate that MS is an autoimmune disease, the initiation and progression of which are dependent on an autoimmune response against myelin antigens. In addition, genetic susceptibility and environmental triggers likely contribute to the initiation of the disease. At this time, there is no cure for MS, but several disease-modifying therapies (DMTs) are available to control and slow down disease progression. A good number of these DMTs were identified and tested using animal models of MS referred to as experimental autoimmune encephalomyelitis (EAE). In this review, we will recapitulate the characteristics of EAE models and discuss how they help shed light on MS pathogenesis and help test new treatments for MS patients.
Collapse
Affiliation(s)
- Simon Glatigny
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98101.,Department of Immunology, University of Washington, Seattle, Washington 98109
| | - Estelle Bettelli
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98101.,Department of Immunology, University of Washington, Seattle, Washington 98109
| |
Collapse
|
39
|
Neuroimmune disorders of the central nervous system in children in the molecular era. Nat Rev Neurol 2018; 14:433-445. [DOI: 10.1038/s41582-018-0024-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
40
|
Dos Passos GR, Oliveira LM, da Costa BK, Apostolos-Pereira SL, Callegaro D, Fujihara K, Sato DK. MOG-IgG-Associated Optic Neuritis, Encephalitis, and Myelitis: Lessons Learned From Neuromyelitis Optica Spectrum Disorder. Front Neurol 2018; 9:217. [PMID: 29670575 PMCID: PMC5893792 DOI: 10.3389/fneur.2018.00217] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/20/2018] [Indexed: 12/28/2022] Open
Abstract
Antibodies against myelin oligodendrocyte glycoprotein (MOG-IgG) have been found in some cases diagnosed as seronegative neuromyelitis optica spectrum disorder (NMOSD). MOG-IgG allowed the identification of a subgroup with a clinical course distinct from that of NMOSD patients who are seropositive for aquaporin-4-IgG antibodies. MOG-IgG is associated with a wider clinical phenotype, not limited to NMOSD, with the majority of cases presenting with optic neuritis (ON), encephalitis with brain demyelinating lesions, and/or myelitis. Therefore, we propose the term MOG-IgG-associated Optic Neuritis, Encephalitis, and Myelitis (MONEM). Depending on the clinical characteristics, these patients may currently be diagnosed with NMOSD, acute disseminated encephalomyelitis, pediatric multiple sclerosis, transverse myelitis, or ON. With specific cell-based assays, MOG-IgG is emerging as a potential biomarker of inflammatory disorders of the central nervous system. We review the growing body of evidence on MONEM, focusing on its clinical aspects.
Collapse
Affiliation(s)
- Giordani Rodrigues Dos Passos
- School of Medicine, Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Luana Michelli Oliveira
- Department of Neurology, Hospital das Clínicas from the Faculty of Medicine, University of São Paulo (HC-FMUSP), São Paulo, Brazil
| | - Bruna Klein da Costa
- School of Medicine, Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Samira Luisa Apostolos-Pereira
- Department of Neurology, Hospital das Clínicas from the Faculty of Medicine, University of São Paulo (HC-FMUSP), São Paulo, Brazil
| | - Dagoberto Callegaro
- Department of Neurology, Hospital das Clínicas from the Faculty of Medicine, University of São Paulo (HC-FMUSP), São Paulo, Brazil
| | - Kazuo Fujihara
- Department of Multiple Sclerosis Therapeutics, Multiple Sclerosis and Neuromyelitis Optica Center, Southern TOHOKU Research Institute for Neuroscience, Fukushima Medical University, Koriyama, Japan
| | - Douglas Kazutoshi Sato
- School of Medicine, Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Department of Neurology, Hospital das Clínicas from the Faculty of Medicine, University of São Paulo (HC-FMUSP), São Paulo, Brazil
| |
Collapse
|
41
|
Microsomal Prostaglandin E Synthase-1 Facilitates an Intercellular Interaction between CD4⁺ T Cells through IL-1β Autocrine Function in Experimental Autoimmune Encephalomyelitis. Int J Mol Sci 2017; 18:ijms18122758. [PMID: 29257087 PMCID: PMC5751357 DOI: 10.3390/ijms18122758] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 12/09/2017] [Accepted: 12/13/2017] [Indexed: 11/21/2022] Open
Abstract
Microsomal prostaglandin synthetase-1 (mPGES-1) is an inducible terminal enzyme that produces prostaglandin E2 (PGE2). In our previous study, we investigated the role of mPGES-1 in the inflammation and demyelination observed in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, using mPGES-1-deficient (mPGES-1−/−) and wild-type (wt) mice. We found that mPGES-1 facilitated inflammation, demyelination, and paralysis and was induced in vascular endothelial cells and macrophages and microglia around inflammatory foci. Here, we investigated the role of interleukin-1β (IL-1β) in the intercellular mechanism stimulated by mPGES-1 in EAE spinal cords in the presence of inflammation. We found that the area invaded by CD4-positive (CD4+) T cells was extensive, and that PGE2 receptors EP1–4 were more induced in activated CD4+ T cells of wt mice than in those of mPGES-1−/− mice. Moreover, IL-1β and IL-1 receptor 1 (IL-1r1) were produced by 65% and 48% of CD4+ T cells in wt mice and by 44% and 27% of CD4+ T cells in mPGES-1−/− mice. Furthermore, interleukin-17 (IL-17) was released from the activated CD4+ T cells. Therefore, mPGES-1 stimulates an intercellular interaction between CD4+ T cells by upregulating the autocrine function of IL-1β in activated CD4+ T cells, which release IL-17 to facilitate axonal and myelin damage in EAE mice.
Collapse
|
42
|
Kesidou E, Touloumi O, Lagoudaki R, Nousiopoulou E, Theotokis P, Poulatsidou KN, Boziki M, Kofidou E, Delivanoglou N, Minti F, Hadjigeorgiou G, Grigoriadis N, Simeonidou C. Humoral response in experimental autoimmune encephalomyelitis targets neural precursor cells in the central nervous system of naive rodents. J Neuroinflammation 2017; 14:227. [PMID: 29162133 PMCID: PMC5697419 DOI: 10.1186/s12974-017-0995-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/03/2017] [Indexed: 02/06/2023] Open
Abstract
Background Neural precursor cells (NPCs) located in the subventricular zone (SVZ), a well-defined NPC niche, play a crucial role in central nervous system (CNS) homeostasis. Moreover, NPCs are involved in the endogenous reparative process both in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). However, the possibility that NPCs may be vulnerable to immune-related components may not be ruled out. Therefore, we investigated the potential affinity of myelin oligodendrocyte glycoprotein (MOG)-induced humoral response(s) to NPCs. Methods MOG35–55-EAE was induced in C57BL/6 mice; blood-sampling was performed on days 17–21 (acute phase) along with a naive group and corresponding antisera (AS) were collected (EAE-AS, NAIVE-AS). The presence of anti-CNS autoantibodies was examined with western blotting. Furthermore, using the collected antisera and anti-MOG antibody (as positive control), immunohistochemistry and double immunofluorescence were implemented on normal neonatal, postnatal, and adult mouse brain sections. Targeted NPCs were identified with confocal microscopy. In vitro immunoreactivity assessment on NPCs challenged with autoantibodies was evaluated for apoptotic/autophagic activity. Results Western blotting verified the existence of autoantibodies in EAE mice and demonstrated bands corresponding to yet unidentified NPC surface epitopes. A dominant selective binding of EAE-AS in the subventricular zone in all age groups compared to NAIVE-AS (p < 0.001) was observed. Additionally, anti-BrdU+/EAE-AS+ colocalization was significantly higher than anti-BrdU+/anti-MOG+, a finding suggesting that the EAE humoral response colocalized with NPCs(BrdU+), cells that do not express MOG. Well-established NPC markers (Nestin, m-Musashi-1, Sox2, DCX, GFAP, NG2) were used to identify the distinct cell types which exhibited selective binding with EAE-AS. The findings verified that EAE-AS exerts cross-reactivity with NPCs which varies throughout the neonatal to adult stage, with a preference to cells of early developmental stages. Finally, increased expressions of Caspase 3 and Beclin 1 on NPCs were detected. Conclusion We provide evidence for the first time that MOG35–55 EAE induces production of antibodies with affinity to SVZ of naive mice in three different age groups. These autoantibodies target lineage-specific NPCs as brain develops and have the potential to trigger apoptotic pathways. Thus, our findings provide indication that cross-talk between immunity and NPCs may lead to functional alteration of NPCs regarding their viability and potentially oligodendrogenesis and effective remyelination. Electronic supplementary material The online version of this article (10.1186/s12974-017-0995-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Olga Touloumi
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Roza Lagoudaki
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Nousiopoulou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kyriaki-Nepheli Poulatsidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Kofidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nickoleta Delivanoglou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Fani Minti
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Constantina Simeonidou
- Laboratory of Experimental Physiology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
43
|
Bando Y, Hagiwara Y, Suzuki Y, Yoshida K, Aburakawa Y, Kimura T, Murakami C, Ono M, Tanaka T, Jiang YP, Mitrovi B, Bochimoto H, Yahara O, Yoshida S. Kallikrein 6 secreted by oligodendrocytes regulates the progression of experimental autoimmune encephalomyelitis. Glia 2017; 66:359-378. [PMID: 29086442 DOI: 10.1002/glia.23249] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 12/28/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS), and experimental autoimmune encephalomyelitis (EAE) is a well-established animal model of the disease. Here, we examined the pathophysiological role of Kallikrein 6 (Klk6), a serine protease produced by oligodendrocytes (OLs), in EAE using Klk6 knockout (Klk6-/-) mice. Compared with Klk6+/+ (wild-type) mice, Klk6-/- mice showed milder EAE symptoms, including delayed onset and milder paralysis. Loss of Klk6 suppressed matrix metalloprotease-9 expression and diminished the infiltration of peripheral inflammatory cells into the CNS by decreasing blood-brain barrier (BBB) permeability and reducing expression levels of inflammatory cytokines, chemokines and their receptors. Scanning electron microscopic analysis revealed demyelination characterized by myelin detachment from the axons in the early phase of EAE progression (days 3-7) in Klk6+/+ mice but not in Klk6-/- mice. Interestingly, anti-MOG (myelin oligodendrocyte glycoprotein) autoantibody was also detected in the cerebrospinal fluid (CSF) and spinal cord on day 3 after MOG immunization. Furthermore, treatment of primary cultured OLs with anti-MOG autoantibody induced oligodendroglial morphological changes and increases in myelin basic protein and Klk6 expression. We also developed a novel enzyme-linked immunoabsorbent assay method for detecting activated KLK6 in human CSF. In human autopsy brain samples, expression of active KLK6 was detected in OLs using an antibody that specifically recognizes the protein's activated form. Taken together, our findings demonstrate that Klk6 secreted by OLs plays a critical role in the pathogenesis of EAE/MS and that it might serve as a potential therapeutic target for MS.
Collapse
Affiliation(s)
- Yoshio Bando
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, 078-8510, Japan
| | | | - Yasuhiro Suzuki
- Department of Neurology, Asahikawa Medical Center, Asahikawa, 070-8644, Japan
| | - Kosuke Yoshida
- Department of Neurology, Asahikawa Medical Center, Asahikawa, 070-8644, Japan
| | - Yoko Aburakawa
- Department of Neurology, Asahikawa Medical Center, Asahikawa, 070-8644, Japan
| | - Takashi Kimura
- Department of Neurology, Asahikawa Medical Center, Asahikawa, 070-8644, Japan
| | - Chisato Murakami
- Department of Neurology, Asahikawa Medical Center, Asahikawa, 070-8644, Japan
| | - Miyuki Ono
- Department of Neurology, Asahikawa Medical Center, Asahikawa, 070-8644, Japan
| | - Tatsuhide Tanaka
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, 078-8510, Japan
| | - Ying-Ping Jiang
- Department of Immunology, Berlex Biosciences, Richmond, California, 94804
| | - Branka Mitrovi
- Department of Immunology, Berlex Biosciences, Richmond, California, 94804
| | - Hiroki Bochimoto
- Department of Microscopic Anatomy and Cell biology, Asahikawa Medical University, Asahikawa, 078-8510, Japan
| | - Osamu Yahara
- Department of Neurology, Asahikawa Medical Center, Asahikawa, 070-8644, Japan
| | - Shigetaka Yoshida
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, 078-8510, Japan
| |
Collapse
|
44
|
Rüther BJ, Scheld M, Dreymueller D, Clarner T, Kress E, Brandenburg LO, Swartenbroekx T, Hoornaert C, Ponsaerts P, Fallier-Becker P, Beyer C, Rohr SO, Schmitz C, Chrzanowski U, Hochstrasser T, Nyamoya S, Kipp M. Combination of cuprizone and experimental autoimmune encephalomyelitis to study inflammatory brain lesion formation and progression. Glia 2017; 65:1900-1913. [PMID: 28836302 DOI: 10.1002/glia.23202] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 12/13/2022]
Abstract
Brain-intrinsic degenerative cascades are a proposed factor driving inflammatory lesion formation in multiple sclerosis (MS) patients. We recently described a model combining noninflammatory cytodegeneration (via cuprizone) with the classic active experimental autoimmune encephalomyelitis (Cup/EAE model), which exhibits inflammatory forebrain lesions. Here, we describe the histopathological characteristics and progression of these Cup/EAE lesions. We show that inflammatory lesions develop at various topographical sites in the forebrain, including white matter tracts and cortical and subcortical grey matter areas. The lesions are characterized by focal demyelination, discontinuation of the perivascular glia limitans, focal axonal damage, and neutrophil granulocyte extravasation. Transgenic mice with enhanced green fluorescent protein-expressing microglia and red fluorescent protein-expressing monocytes reveal that both myeloid cell populations contribute to forebrain inflammatory infiltrates. EAE-triggered inflammatory cerebellar lesions were augmented in mice pre-intoxicated with cuprizone. Gene expression studies suggest roles of the chemokines Cxcl10, Ccl2, and Ccl3 in inflammatory lesion formation. Finally, follow-up experiments in Cup/EAE mice with chronic disease revealed that forebrain, but not spinal cord, lesions undergo spontaneous reorganization and repair. This study underpins the significance of brain-intrinsic degenerative cascades for immune cell recruitment and, in consequence, MS lesion formation.
Collapse
Affiliation(s)
- Bernhard Josef Rüther
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, Aachen, 52074, Germany
| | - Miriam Scheld
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, Aachen, 52074, Germany
| | - Daniela Dreymueller
- Institute of Pharmacology and Toxicology, Faculty of Medicine, RWTH Aachen University, Aachen, 52074, Germany
| | - Tim Clarner
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, Aachen, 52074, Germany
| | - Eugenia Kress
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, 52074, Germany
| | - Lars-Ove Brandenburg
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, 52074, Germany
| | - Tine Swartenbroekx
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, 2610, Belgium
| | - Chloé Hoornaert
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, 2610, Belgium
| | - Peter Ponsaerts
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, 2610, Belgium
| | - Petra Fallier-Becker
- Institute of Pathology and Neuropathology, University of Tuebingen, Tuebingen, 72076, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, Aachen, 52074, Germany
| | - Sven Olaf Rohr
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, 80336, Germany
| | - Christoph Schmitz
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, 80336, Germany
| | - Uta Chrzanowski
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, 80336, Germany
| | - Tanja Hochstrasser
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, 80336, Germany
| | - Stella Nyamoya
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, Aachen, 52074, Germany.,Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, 80336, Germany
| | - Markus Kipp
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, 80336, Germany
| |
Collapse
|
45
|
Sanna MD, Quattrone A, Galeotti N. Silencing of the RNA-binding protein HuR attenuates hyperalgesia and motor disability in experimental autoimmune encephalomyelitis. Neuropharmacology 2017; 123:116-125. [PMID: 28599923 DOI: 10.1016/j.neuropharm.2017.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/12/2017] [Accepted: 06/05/2017] [Indexed: 01/23/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system associated with progressive neuronal loss and axonal degeneration. Neuronal lesions and dysfunction lead often to neuropathic pain, the most prevalent and difficult to treat pain syndrome observed in MS patients. Despite its widespread occurrence, the underlying neural mechanisms for MS pain are not fully understood. For a better clarification of the pathophysiology of MS-associated pain, we investigated the role of HuR, an RNA-binding protein that positively regulates the stability of many target mRNAs, including several cytokines. The influence of HuR in the generation of the hypernociceptive response in a mouse model of relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE), an experimental model of MS, was investigated. HuR silencing, obtained through the repeated intrathecal administration of an antisense oligonucleotide (aODN) anti-HuR, completely attenuated hindpaw mechanical allodynia and thermal hyperalgesia developed by RR-EAE mice. Anti-HuR aODN also reduced severity of motor deficits as reflected by a reduction of clinical EAE score and improvement of rotarod performance. RR-EAE mice showed demyelination in spinal cord sections that was significantly reduced by HuR silencing. Double-staining immunofluorescence studies showed a neuronal localization of HuR within dorsal horn spinal cord, consistent with a neuronal mechanism of action. Our findings suggest the involvement of HuR in the hypernociceptive behaviour of RR-EAE mice providing the first pharmacological assessment of an antiallodynic and antihyperalgesic effect of HuR silencing. These data may provide support for HuR modulation as a therapeutic perspective for the management of MS-related neuropathic pain.
Collapse
Affiliation(s)
- Maria Domenica Sanna
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy
| | - Alessandro Quattrone
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy.
| |
Collapse
|
46
|
Cellina M, Fetoni V, Ciocca M, Pirovano M, Oliva G. Anti-myelin oligodendrocyte glycoprotein antibodies: Magnetic resonance imaging findings in a case series and a literature review. Neuroradiol J 2017; 31:69-82. [PMID: 28379073 DOI: 10.1177/1971400917698856] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Myelin oligodendrocyte glycoprotein is a protein exclusively expressed on the surface of oligodendrocytes and myelin in the central nervous system. Antibodies against myelin oligodendrocyte glycoprotein were initially detected in children with demyelinating syndromes, and more recently reported in a broad spectrum of central nervous system demyelinating diseases in adults, including neuromyelitis optica spectrum disorders and bilateral optic neuritis. Patients with myelin oligodendrocyte glycoprotein antibody-associated demyelination appear to have unique clinical and radiological features. To the best of our knowledge a series of Italian patients with optic neuritis and positivity to myelin oligodendrocyte glycoprotein antibodies has not yet been reported and the paper on myelin oligodendrocyte glycoprotein antibodies are more focused on clinical features, diagnosis and outcome than on the radiological appearance, so we want to retrospectively report magnetic resonance imaging features of a group of eight patients, who came to our Ophthalmologic Emergency Department for optic neuritis and were found seropositive for myelin oligodendrocyte glycoprotein antibodies, comparing our data with the findings described in the literature.
Collapse
Affiliation(s)
| | | | - Matteo Ciocca
- 2 Neurology Department, ASST Fatebenefratelli Sacco, Italy
| | - Marta Pirovano
- 2 Neurology Department, ASST Fatebenefratelli Sacco, Italy
| | | |
Collapse
|
47
|
Alberti TB, Barbosa WLR, Vieira JLF, Raposo NRB, Dutra RC. (-)-β-Caryophyllene, a CB2 Receptor-Selective Phytocannabinoid, Suppresses Motor Paralysis and Neuroinflammation in a Murine Model of Multiple Sclerosis. Int J Mol Sci 2017; 18:ijms18040691. [PMID: 28368293 PMCID: PMC5412277 DOI: 10.3390/ijms18040691] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/18/2017] [Accepted: 03/20/2017] [Indexed: 12/05/2022] Open
Abstract
(−)-β-caryophyllene (BCP), a cannabinoid receptor type 2 (CB2)-selective phytocannabinoid, has already been shown in precedent literature to exhibit both anti-inflammatory and analgesic effects in mouse models of inflammatory and neuropathic pain. Herein, we endeavored to investigate the therapeutic potential of BCP on experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS). Furthermore, we sought to demonstrate some of the mechanisms that underlie the modulation BCP exerts on autoimmune activated T cells, the pro-inflammatory scenery of the central nervous system (CNS), and demyelination. Our findings demonstrate that BCP significantly ameliorates both the clinical and pathological parameters of EAE. In addition, data hereby presented indicates that mechanisms underlying BCP immunomodulatory effect seems to be linked to its ability to inhibit microglial cells, CD4+ and CD8+ T lymphocytes, as well as protein expression of pro-inflammatory cytokines. Furthermore, it diminished axonal demyelination and modulated Th1/Treg immune balance through the activation of CB2 receptor. Altogether, our study represents significant implications for clinical research and strongly supports the effectiveness of BCP as a novel molecule to target in the development of effective therapeutic agents for MS.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Cannabinoid Receptor Agonists/pharmacology
- Cytokines/metabolism
- Demyelinating Diseases/prevention & control
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Female
- Humans
- Hyperalgesia/prevention & control
- Mice, Inbred C57BL
- Microglia/drug effects
- Microglia/metabolism
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/physiopathology
- Multiple Sclerosis/prevention & control
- Neurogenic Inflammation/metabolism
- Neurogenic Inflammation/physiopathology
- Neurogenic Inflammation/prevention & control
- Paralysis/metabolism
- Paralysis/physiopathology
- Paralysis/prevention & control
- Polycyclic Sesquiterpenes
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/metabolism
- Sesquiterpenes/pharmacology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/metabolism
Collapse
Affiliation(s)
- Thaís Barbosa Alberti
- Laboratory of Autoimmunity and Immunopharmacology (LAIF), Department of Health Sciences, Center of Araranguá, Federal University of Santa Catarina, Araranguá 88906-072, Brazil.
| | | | | | - Nádia Rezende Barbosa Raposo
- Research and Innovation in Health Sciences (NUPICS), Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil.
| | - Rafael Cypriano Dutra
- Laboratory of Autoimmunity and Immunopharmacology (LAIF), Department of Health Sciences, Center of Araranguá, Federal University of Santa Catarina, Araranguá 88906-072, Brazil.
| |
Collapse
|
48
|
Zhou L, Huang Y, Li H, Fan J, Zhangbao J, Yu H, Li Y, Lu J, Zhao C, Lu C, Wang M, Quan C. MOG-antibody associated demyelinating disease of the CNS: A clinical and pathological study in Chinese Han patients. J Neuroimmunol 2017; 305:19-28. [PMID: 28284341 DOI: 10.1016/j.jneuroim.2017.01.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/08/2017] [Accepted: 01/11/2017] [Indexed: 01/26/2023]
Abstract
We aim to evaluate the clinical relevance of MOG-ab in a cohort of Chinese Han adults with CNS inflammatory demyelinating diseases (IDDs). MOG-ab and AQP4-ab were examined through a fixed cell based indirect immune-fluorescence assay in 86 patients with CNS-IDDs. MOG-ab was positive in 12 patients, while AQP4-ab was positive in 31 patients; none double positives. Optic neuritis (ON) was the most frequent symptom at onset (75.0%) or during the whole disease course (83.3%) of MOG-ab associated IDDs (MOG-IDDs); 79.5% of the episodes involved only the optic nerve in MOG-IDDs. MOG-ab related ON (MOG-ON) usually caused severe visual impairment, longitudinally extensive optic nerve lesion with anterior enhancement and perineural soft tissue enhancement, responded well to steroid, but still could leave remarkable thinning of retinal nerve fiber layer (RNFL) and ganglion cell complex (GCC). MOG-IDDs had less spinal cord involvement compared to AQP4-ab mediated NMO/SD. Heterogeneous brain lesions existed in 66.7% of the patients with MOG-IDDs. Large, edematous white matter lesions were observed with the pathological feature of obvious demyelination yet preservation of astrocyte and axon, fundamentally different from the astrocytopathy typically seen in NMO/SD. Our investigations suggest that MOG-ab mediates a distinct disease entity separate from NMO/SD.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Yongheng Huang
- Department of Ophthalmology, Eye and ENT Hospital, Shanghai Medical College, Fudan University, China
| | - Haiqing Li
- Department of Radiology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Jie Fan
- Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Jingzi Zhangbao
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Hai Yu
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Yuxin Li
- Department of Radiology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Jiahong Lu
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China; Department of Neurology, Jing'an District Centre Hospital of Shanghai, China
| | - Chuanzhen Lu
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Min Wang
- Department of Ophthalmology, Eye and ENT Hospital, Shanghai Medical College, Fudan University, China.
| | - Chao Quan
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China.
| |
Collapse
|
49
|
Valverde P, Kawai T, Taubman MA. Potassium Channel-blockers as Therapeutic Agents to Interfere with Bone Resorption of Periodontal Disease. J Dent Res 2016; 84:488-99. [PMID: 15914584 DOI: 10.1177/154405910508400603] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Inflammatory lesions of periodontal disease contain all the cellular components, including abundant activated/memory T- and B-cells, necessary to control immunological interactive networks and to accelerate bone resorption by RANKL-dependent and -independent mechanisms. Blockade of RANKL function has been shown to ameliorate periodontal bone resorption and other osteopenic disorders without affecting inflammation. Development of therapies aimed at decreasing the expression of RANKL and pro-inflammatory cytokines by T-cells constitutes a promising strategy to ameliorate not only bone resorption, but also inflammation. Several reports have demonstrated that the potassium channels Kv1.3 and IKCa1, through the use of selective blockers, play important roles in T-cell-mediated events, including T-cell proliferation and the production of pro-inflammatory cytokines. More recently, a potassium channel-blocker for Kv1.3 has been shown to down-regulate bone resorption by decreasing the ratio of RANKL-to-OPG expression by memory-activated T-cells. In this article, we first summarize the mechanisms by which chronically activated/memory T-cells, in concert with B-cells and macrophages, trigger inflammatory bone resorption. Then, we describe the main structural and functional characteristics of potassium channels Kv1.3 and IKCa1 in some of the cells implicated in periodontal disease progression. Finally, this review elucidates some recent advances in the use of potassium channel-blockers of Kv1.3 and IKCa1 to ameliorate the clinical signs or side-effects of several immunological disorders and to decrease inflammatory bone resorption in periodontal disease. ABBREVIATIONS: AICD, activation-induced cell death; APC, antigen-presenting cells; B(K), large conductance; CRAC, calcium release-activated calcium channels; DC, dendritic cell; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; IFN-γ, interferon-γ; IP3, inositol (1,4,5)-triphosphate; (K)ir, inward rectifier; JNK, c-Jun N-terminal kinase; I(K), intermediate conductance; LPS, lipopolysaccharide; L, ligand; MCSF, macrophage colony-stimulating factor; MHC, major histocompatibility complex; NFAT, nuclear factor of activated T-cells; RANK, receptor activator of nuclear factor-κB; TCM, central memory T-cells; TEM, effector memory T-cells; TNF, tumor necrosis factor; TRAIL, TNF-related apoptosis-inducing ligand; OPG, osteoprotegerin; Omp29, 29-kDa outer membrane protein; PKC, protein kinase C; PLC, phospholipase C; RT-PCR, reverse-transcriptase polymerase chain-reaction; S(K), small conductance; TCR, T-cell receptor; and (K)v, voltage-gated.
Collapse
Affiliation(s)
- P Valverde
- Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA.
| | | | | |
Collapse
|
50
|
Castillo EF, Zheng H, Van Cabanlong C, Dong F, Luo Y, Yang Y, Liu M, Kao WWY, Yang XO. Lumican negatively controls the pathogenicity of murine encephalitic TH17 cells. Eur J Immunol 2016; 46:2852-2861. [PMID: 27682997 DOI: 10.1002/eji.201646507] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/02/2016] [Accepted: 09/27/2016] [Indexed: 11/11/2022]
Abstract
TH17 cells play an essential role in the development of both human multiple sclerosis and animal experimental autoimmune encephalomyelitis (EAE). Nevertheless, it is not well understood how the pathogenicity of TH17 cells is controlled in the autoimmune neuroinflammation. In vitro, we found Lumican (Lum), an extracellular matrix (ECM) protein, is selectively expressed by TH17 cells among tested murine TH subsets. Lum deficiency leads to earlier onset and enhanced severity of EAE. This enhanced disease in Lum-deficient mice is associated with increased production of IL-17 and IL-21 and decreased TH17 cell apoptosis. Dysregulation in cytokine production appears to be specific to TH17 cells as TH1 and TH2 cell polarization and/or cytokine production were unaltered. Furthermore, adoptive transfer of myelin oligodendrocyte glycoprotein specific TH17 cells derived from Lum-deficient mice led to earlier onset and increased severity of disease compared to controls highlighting a TH17-cell-intrinsic effect of Lum. Taken together, our results suggest that Lum negatively regulates encephalitic TH17 cells, implicating a potential therapeutic pathway in TH17 cell mediated autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Eliseo F Castillo
- Department of Molecular Genetics and Microbiology and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Handong Zheng
- Department of Molecular Genetics and Microbiology and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Christian Van Cabanlong
- Department of Molecular Genetics and Microbiology and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Fei Dong
- Deparment of Ophthalmology, University of Cincinnati, Cincinnati, OH, USA
| | - Yan Luo
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Winston W-Y Kao
- Deparment of Ophthalmology, University of Cincinnati, Cincinnati, OH, USA
| | - Xuexian O Yang
- Department of Molecular Genetics and Microbiology and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|