1
|
Yang L, Han X, Wang M, Zhang X, Wang L, Xu N, Wu H, Shi H, Pan W, Huang F, Wu X. Early Growth Response Gene 1 Benefits Autoimmune Disease by Promoting Regulatory T Cell Differentiation as a Regulator of Foxp3. RESEARCH (WASHINGTON, D.C.) 2025; 8:0662. [PMID: 40235598 PMCID: PMC11997311 DOI: 10.34133/research.0662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
Foxp3+ regulatory T (Treg) cells, as one of the subtypes of CD4+ T cells, are the crucial gatekeeper in the pathogenesis of self-antigen reactive diseases. In this context, we demonstrated that the selective ablation of early growth response gene 1 (Egr-1) in CD4+ T cells exacerbated experimental autoimmune encephalomyelitis (EAE) in murine models. The absence of Egr-1 in CD4+ T cells, obtained from EAE mice and naïve CD4+ T cells, impeded the differentiation and influence of Treg. Importantly, in CD4+ T cells of multiple sclerosis patients, both Egr-1 and Foxp3 were found to decrease. Further studies showed that distinct from the classical Smad3 route, TGF-β could activate Egr-1 through the Raf-Erk signaling route to promote Foxp3 genetic modulation, thereby promoting Treg cell differentiation and reducing EAE inflammation. A novel natural Egr-1 agonist, calycosin, was found to attenuate EAE progression by regulating the differentiation of Treg. Together, the above results indicate the value of Egr-1, as a novel Foxp3 transactivator, for the differentiation of Treg cells in the development of self-antigen reactive diseases.
Collapse
Affiliation(s)
- Liu Yang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Central Laboratory,
Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyan Han
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Neurology, Tangdu Hospital,
Air Force Medical University, Xi’an, China
| | - Mengxue Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojuan Zhang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lupeng Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nuo Xu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weidong Pan
- Department of Neurology,
Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Huang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica,
Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
2
|
Dorababu A. Experimentation of Heterocycles (2013-22) as Potent Pharmacophores in Drug Design of Multiple Sclerosis. Drug Dev Res 2025; 86:e70059. [PMID: 39907074 DOI: 10.1002/ddr.70059] [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: 09/13/2024] [Revised: 01/15/2025] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Multiple sclerosis (MS) is a demyelinating disease in which the insulating cover (myelin sheath) of the brain and spinal cord is damaged. Demyelination results in a decreased signal transmission in the nervous system. Symptoms include double vision, muscle weakness, and difficulty with coordination. Genetic and viral infections have been proposed as plausible factors responsible for MS. Although there is no cure for MS, treatment prevents future attacks. At present, chemotherapy and monoclonal antibodies are the available treatments for MS. Heterocyclic compounds are currently being tested clinically for their efficacy. Some heterocyclic scaffolds have been found to be promising for the treatment of MS. In view of this, research has been conducted towards the design and discovery of chemical agents for MS. Hence, the literature relevant to drug design for MS in the last decade has been collated and described comprehensively so that it would be helpful for efficient drug design for MS in the future. Additionally, through the structure-activity relationship, the importance of crucial structural features was emphasized. The classification was primarily based on the type of heterocycle.
Collapse
Affiliation(s)
- Atukuri Dorababu
- SKNG Government First Grade College, Gangavathi, 583227, Karnataka, India
| |
Collapse
|
3
|
Kawakami N, Wekerle H. Life history of a brain autoreactive T cell: From thymus through intestine to blood-brain barrier and brain lesion. Neurotherapeutics 2024; 21:e00442. [PMID: 39237437 PMCID: PMC11585894 DOI: 10.1016/j.neurot.2024.e00442] [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: 05/06/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024] Open
Abstract
Brain antigen-specific autoreactive T cells seem to play a key role in inducing inflammation in the central nervous system (CNS), a characteristic feature of human multiple sclerosis (MS). These T cells are generated within the thymus, where they escape negative selection and become integrated into the peripheral immune repertoire of immune cells. Typically, these autoreactive T cells rest in the periphery without attacking the CNS. When autoimmune T cells enter gut-associated lymphatic tissue (GALT), they may be stimulated by the microbiota and its metabolites. After activation, the cells migrate into the CNS through the blood‒brain barrier, become reactivated upon interacting with local antigen-presenting cells, and induce inflammatory lesions within the brain parenchyma. This review describes how microbiota influence autoreactive T cells during their life, starting in the thymus, migrating through the periphery and inducing inflammation in their target organ, the CNS.
Collapse
Affiliation(s)
- Naoto Kawakami
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich and Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Germany.
| | - Hartmut Wekerle
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich and Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Germany; Emeritus Group Neuroimmunology, Max Planck Institute of Biological Intelligence, Germany.
| |
Collapse
|
4
|
Peters A, Gerdes LA, Wekerle H. Multiple sclerosis and the intestine: Chasing the microbial offender. Immunol Rev 2024; 325:152-165. [PMID: 38809041 DOI: 10.1111/imr.13357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Multiple sclerosis (MS) affects more than 2.8 million people worldwide but the distribution is not even. Although over 200 gene variants have been associated with susceptibility, studies of genetically identical monozygotic twin pairs suggest that the genetic make-up is responsible for only about 20%-30% of the risk to develop disease, while the rest is contributed by milieu factors. Recently, a new, unexpected player has entered the ranks of MS-triggering or facilitating elements: the human gut microbiota. In this review, we summarize the present knowledge of microbial effects on formation of a pathogenic autoreactive immune response targeting the distant central nervous system and delineate the approaches, both in people with MS and in MS animal models, which have led to this concept. Finally, we propose that a tight combination of investigations of human patients with studies of suitable animal models is the best strategy to functionally characterize disease-associated microbiota and thereby contribute to deciphering pathogenesis of a complex human disease.
Collapse
Affiliation(s)
- Anneli Peters
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Lisa Ann Gerdes
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Hartmut Wekerle
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Munich, Germany
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| |
Collapse
|
5
|
Wang Z, Xu J, Mo L, Zhan R, Zhang J, Liu L, Jiang J, Zhang Y, Bai Y. The Application Potential of the Regulation of Tregs Function by Irisin in the Prevention and Treatment of Immune-Related Diseases. Drug Des Devel Ther 2024; 18:3005-3023. [PMID: 39050796 PMCID: PMC11268596 DOI: 10.2147/dddt.s465713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Irisin is a muscle factor induced by exercise, generated through the proteolytic cleavage of the membrane protein fibronectin type III domain-containing protein 5 (FNDC-5). Numerous studies have shown that irisin plays a significant role in regulating glucose and lipid metabolism, inhibiting oxidative stress, reducing systemic inflammatory responses, and providing neuroprotection. Additionally, irisin can exert immunomodulatory functions by regulating regulatory T cells (Tregs). Tregs are a highly differentiated subset of mature T cells that play a key role in maintaining self-immune homeostasis and are closely related to infections, inflammation, immune-related diseases, and tumors. Irisin exerts persistent positive effects on Treg cell functions through various mechanisms, including regulating Treg cell differentiation and proliferation, improving their function, modulating the balance of immune cells, increasing the production of anti-inflammatory cytokines, and enhancing metabolic functions, thereby helping to maintain immune homeostasis and prevent immune-related diseases. As an important myokine, irisin interacts with receptors on the cell membrane, activating multiple intracellular signaling pathways to regulate cell metabolism, proliferation, and function. Although the specific receptor for irisin has not been fully identified, integrins are considered potential receptors. Irisin activates various signaling pathways, including AMPK, MAPK, and PI3K/Akt, through integrin receptors, thereby exerting multiple biological effects. These research findings provide important clues for understanding the mechanisms of irisin's action and theoretical basis for its potential applications in metabolic diseases and immunomodulation. This article reviews the relationship between irisin and Tregs, as well as the research progress of irisin in immune-related diseases such as multiple sclerosis, myasthenia gravis, acquired immune deficiency syndrome, type 1 diabetes, sepsis, and rheumatoid arthritis. Studies have revealed that irisin plays an important role in immune regulation by improving the function of Tregs, suggesting its potential application value in the treatment of immune-related diseases.
Collapse
Affiliation(s)
- Zhengjiang Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Jiaqi Xu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Liqun Mo
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Renshu Zhan
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Yingying Zhang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Yiping Bai
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| |
Collapse
|
6
|
Jiao M, Wang C, Tang X, Dai C, Zhang N, Fan A, Qian Z, Liu S, Zhang F, Li B, Xu Y, Tan Z, Gong F, Lu Y, Zheng F. Active secretion of IL-33 from astrocytes is dependent on TMED10 and promotes central nervous system homeostasis. Brain Behav Immun 2024; 119:539-553. [PMID: 38663774 DOI: 10.1016/j.bbi.2024.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/07/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
Interleukin-33 (IL-33), secreted by astrocytes, regulates the synapse development in the spinal cord and hippocampus and suppresses autoimmune disease in the central nervous system (CNS). However, the mechanism of unconventional protein secretion of this cytokine remains unclear. In this study, we found that IFN-γ promotes the active secretion of IL-33 from astrocytes, and the active secretion of IL-33 from cytoplasm to extracellular space was dependent on interaction with transmembrane emp24 domain 10 (TMED10) via the IL-1 like cytokine domain in astrocytes. Knockout of Il-33 or its receptor St2 induced hippocampal astrocyte activation and depressive-like disorder in naive mice, as well as increased spinal cord astrocyte activation and polarization to a neurotoxic reactive subtype and aggravated passive experimental autoimmune encephalomyelitis (EAE). Our results have identified that IL-33 is actively secreted by astrocytes through the unconventional protein secretion pathway facilitated by TMED10 channels. This process helps maintain CNS homeostasis by inhibiting astrocyte activation.
Collapse
Affiliation(s)
- Mengya Jiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chenchen Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xuhuan Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chan Dai
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Na Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anqi Fan
- College of Life Science, Yangtze University, Jingzhou 434025, China
| | - Zhigang Qian
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiwang Liu
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feng Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bin Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yong Xu
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zheng Tan
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Feili Gong
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yisheng Lu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China.
| |
Collapse
|
7
|
Holt EA, Tyler A, Lakusta-Wong T, Lahue KG, Hankes KC, Teuscher C, Lynch RM, Ferris MT, Mahoney JM, Krementsov DN. Probing the basis of disease heterogeneity in multiple sclerosis using genetically diverse mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597205. [PMID: 38895248 PMCID: PMC11185616 DOI: 10.1101/2024.06.03.597205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Multiple sclerosis (MS) is a complex disease with significant heterogeneity in disease course and progression. Genetic studies have identified numerous loci associated with MS risk, but the genetic basis of disease progression remains elusive. To address this, we leveraged the Collaborative Cross (CC), a genetically diverse mouse strain panel, and experimental autoimmune encephalomyelitis (EAE). The thirty-two CC strains studied captured a wide spectrum of EAE severity, trajectory, and presentation, including severe-progressive, monophasic, relapsing remitting, and axial rotary (AR)-EAE, accompanied by distinct immunopathology. Sex differences in EAE severity were observed in six strains. Quantitative trait locus analysis revealed distinct genetic linkage patterns for different EAE phenotypes, including EAE severity and incidence of AR-EAE. Machine learning-based approaches prioritized candidate genes for loci underlying EAE severity ( Abcc4 and Gpc6 ) and AR-EAE ( Yap1 and Dync2h1 ). This work expands the EAE phenotypic repertoire and identifies novel loci controlling unique EAE phenotypes, supporting the hypothesis that heterogeneity in MS disease course is driven by genetic variation. Summary The genetic basis of disease heterogeneity in multiple sclerosis (MS) remains elusive. We leveraged the Collaborative Cross to expand the phenotypic repertoire of the experimental autoimmune encephalomyelitis (EAE) model of MS and identify loci controlling EAE severity, trajectory, and presentation.
Collapse
|
8
|
Stegnjaić G, Jevtić B, Lazarević M, Ignjatović Đ, Tomić M, Nikolovski N, Bjelobaba I, Momčilović M, Dimitrijević M, Miljković Đ, Stanisavljević S. Brain inflammation in experimental autoimmune encephalomyelitis induced in Dark Agouti rats with spinal cord homogenate. Immunol Lett 2024; 267:106852. [PMID: 38508497 DOI: 10.1016/j.imlet.2024.106852] [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: 12/14/2023] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 03/22/2024]
Abstract
We have recently characterized experimental autoimmune encephalomyelitis (EAE) induced in DA rats with spinal cord homogenate without complete Freund's adjuvant (CFA). The main advantage of this multiple sclerosis model is the lack of CFA-related confounding effects which represent the major obstacles in translating findings from EAE to multiple sclerosis. Here, antigen specificity of the cellular and humoral immune response directed against the central nervous system was explored. The reactivity of T and B cells to myelin basic protein, myelin oligodendrocyte glycoprotein, and β-synuclein was detected. Having in mind that reactivity against β-synuclein was previously associated with autoimmunity against the brain, the infiltration of immune cells into different brain compartments, i.e. pons, cerebellum, hippocampus, and cortex was determined. T cell infiltration was observed in all structures examined. This finding stimulated investigation of the effects of immunization on DA rat behavior using the elevated plus maze and the open field test. Rats recovered from EAE displayed increased anxiety-like behavior. These data support CFA-free EAE in DA rats as a useful model for multiple sclerosis research.
Collapse
Affiliation(s)
- Goran Stegnjaić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia
| | - Milica Lazarević
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia
| | - Đurđica Ignjatović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia
| | - Mirko Tomić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia
| | - Neda Nikolovski
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia
| | - Ivana Bjelobaba
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia
| | - Miljana Momčilović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia
| | - Mirjana Dimitrijević
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia.
| | - Suzana Stanisavljević
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia
| |
Collapse
|
9
|
Funaki M, Nio-Kobayashi J, Suzuki R, Bando Y. Galectin-3 Plays a Role in Neuroinflammation in the Visual Pathway in Experimental Optic Neuritis. Cells 2024; 13:612. [PMID: 38607051 PMCID: PMC11011492 DOI: 10.3390/cells13070612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/18/2024] [Accepted: 03/30/2024] [Indexed: 04/13/2024] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) featuring numerous neuropathologies, including optic neuritis (ON) in some patients. However, the molecular mechanisms of ON remain unknown. Galectins, β-galactoside-binding lectins, are involved in various pathophysiological processes. We previously showed that galectin-3 (gal-3) is associated with the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of MS. In the current study, we investigated the expression of gal-3 in the visual pathway in EAE mice to clarify its role in the pathogenesis of ON. Immunohistochemical analysis revealed upregulation of gal-3 in the visual pathway of the EAE mice during the peak stage of the disease, compared with naïve and EAE mice during the chronic stage. Gal-3 was detected mainly in microglia/macrophages and astrocytes in the visual pathway in EAE mice. In addition, gal-3+/Iba-1+ cells, identified as phagocytic by immunostaining for cathepsin D, accumulated in demyelinating lesions in the visual pathway during the peak disease stage of EAE. Moreover, NLRP3 expression was detected in most gal-3+/Iba-1+ cells. These results strongly suggest that gal-3 regulates NLRP3 signaling in microglia/macrophages and neuroinflammatory demyelination in ON. In astrocytes, gal-3 was expressed from the peak to the chronic disease stages. Taken together, our findings suggest a critical role of gal-3 in the pathogenesis of ON. Thus, gal-3 in glial cells may serve as a potential therapeutic target for ON.
Collapse
Affiliation(s)
- Masako Funaki
- Department of Anatomy, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Junko Nio-Kobayashi
- Department of Functional Glycobiology in Infectious Diseases, National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki 852-8523, Japan
| | - Ryoji Suzuki
- Department of Anatomy, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Yoshio Bando
- Department of Anatomy, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| |
Collapse
|
10
|
Jahromi AS, Erfanian S, Roustazadeh A. Association of OX40L gene polymorphism with multiple sclerosis in Iranians. Heliyon 2024; 10:e27304. [PMID: 38496859 PMCID: PMC10944201 DOI: 10.1016/j.heliyon.2024.e27304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction The exact etiology of multiple sclerosis is unknown but recent studies indicated a link between tumor necrosis factor superfamily member 4 and the disease. Polymorphisms located in the regulatory region of the gene may affect its phenotype. Hence, we aimed to investigate the association of promoter polymorphisms of the gene with multiple sclerosis and also to estimate the frequency of haplotypes in the patients and healthy subjects. Methods Two hundred age- and sex-matched subjects including 100 patients and 100 healthy subjects were investigated in the study. Genotype and allele distributions of rs3850641, rs1234313, and rs10912580 polymorphisms in the promoter region of the gene were investigated by polymerase chain reaction-restriction fragment length polymorphism. In addition, haplotype frequencies estimation and linkage disequilibrium analysis were performed by SNPStats web tool. Results The distribution of AA, AG and GG genotypes of rs3850641 was significantly different between the patient and healthy groups (P = 0.009). In addition, frequencies of A and G alleles of rs3850641 were different between the groups (P < 0.001). Also the distribution of rs3850641 genotypes was different between the women of the both groups (P = 0.007). Our analysis revealed that rs3850641 AG (Odds ratio = 0.393, 95 % confidence interval = 0.170-0.907, P = 0.029) and GG (Odds ratio = 0.373, 95 % confidence interval = 0.168-0.830, P = 0.016) genotypes were associated with decreased risk of the disease. However, rs1234313 genotype and allele distributions were not different between the groups. The distribution of rs10912580polymorphism. AA, AG, and GG genotypes was significantly different between the groups (P = 0.007). rs10912580 AG genotype was associated with low risk of the disease (Odds ratio = 0.252, 95 % confidence interval = 0.102-0.623, P = 0.003). The distribution of haplotypes was statistically different between the patient and healthy groups (P < 0.001). A-G-A was the most frequent haplotype among the patients and the estimated frequency was higher than that of the control group (0.5527 versus 0.3739). Conclusion The distribution of rs3850641 and rs10912580 genotypes was different between the patients and healthy subjects. Moreover, rs3850641 AG and GG genotypes and also rs10912580 AG genotype were associated with low risk of the disease in Iranians. Further studies with large groups are recommended to show whether genotype variation in the patients could alter the response to treatment or not.
Collapse
Affiliation(s)
- Abdolreza Sotoodeh Jahromi
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
- Medical Immunology Department, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Saiedeh Erfanian
- Department of Biochemistry, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- Department of Advanced Medical Sciences and Technologies, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Abazar Roustazadeh
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
- Department of Biochemistry, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- Department of Advanced Medical Sciences and Technologies, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| |
Collapse
|
11
|
Yazdanpanah E, Dadfar S, Shadab A, Orooji N, Nemati M, Pazoki A, Esmaeili S, Baharlou R, Haghmorad D. Berberine: A natural modulator of immune cells in multiple sclerosis. Immun Inflamm Dis 2024; 12:e1213. [PMID: 38477663 PMCID: PMC10936236 DOI: 10.1002/iid3.1213] [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: 08/25/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Berberine is a benzylisoquinoline alkaloid found in such plants as Berberis vulgaris, Berberis aristata, and others, revealing a variety of pharmacological properties as a result of interacting with different cellular and molecular targets. Recent studies have shown the immunomodulatory effects of Berberine which result from its impacts on immune cells and immune response mediators such as diverse T lymphocyte subsets, dendritic cells (DCs), and different inflammatory cytokines. Multiple sclerosis (MS) is a chronic disabling and neurodegenerative disease of the central nervous system (CNS) characterized by the recruitment of autoreactive T cells into the CNS causing demyelination, axonal damage, and oligodendrocyte loss. There have been considerable changes discovered in MS regards to the function and frequency of T cell subsets such as Th1 cells, Th17 cells, Th2 cells, Treg cells, and DCs. In the current research, we reviewed the outcomes of in vitro, experimental, and clinical investigations concerning the modulatory effects that Berberine provides on the function and numbers of T cell subsets and DCs, as well as important cytokines that are involved in MS.
Collapse
Affiliation(s)
| | - Sepehr Dadfar
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Alireza Shadab
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Niloufar Orooji
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - MohammadHossein Nemati
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Alireza Pazoki
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | | | - Rasoul Baharlou
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
- Cancer Research CenterSemnan University of Medical SciencesSemnanIran
| | - Dariush Haghmorad
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
- Cancer Research CenterSemnan University of Medical SciencesSemnanIran
| |
Collapse
|
12
|
Jakimovski D, Bittner S, Zivadinov R, Morrow SA, Benedict RH, Zipp F, Weinstock-Guttman B. Multiple sclerosis. Lancet 2024; 403:183-202. [PMID: 37949093 DOI: 10.1016/s0140-6736(23)01473-3] [Citation(s) in RCA: 164] [Impact Index Per Article: 164.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 06/08/2023] [Accepted: 07/12/2023] [Indexed: 11/12/2023]
Abstract
Multiple sclerosis remains one of the most common causes of neurological disability in the young adult population (aged 18-40 years). Novel pathophysiological findings underline the importance of the interaction between genetics and environment. Improvements in diagnostic criteria, harmonised guidelines for MRI, and globalised treatment recommendations have led to more accurate diagnosis and an earlier start of effective immunomodulatory treatment than previously. Understanding and capturing the long prodromal multiple sclerosis period would further improve diagnostic abilities and thus treatment initiation, eventually improving long-term disease outcomes. The large portfolio of currently available medications paved the way for personalised therapeutic strategies that will balance safety and effectiveness. Incorporation of cognitive interventions, lifestyle recommendations, and management of non-neurological comorbidities could further improve quality of life and outcomes. Future challenges include the development of medications that successfully target the neurodegenerative aspect of the disease and creation of sensitive imaging and fluid biomarkers that can effectively predict and monitor disease changes.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA; Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA; Center for Biomedical Imaging at the Clinical Translational Science Institute, State University of New York at Buffalo, Buffalo, NY, USA
| | - Sarah A Morrow
- Department of Clinical Neurological Sciences, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Ralph Hb Benedict
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Bianca Weinstock-Guttman
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
13
|
Gadhave DG, Sugandhi VV, Kokare CR. Potential biomaterials and experimental animal models for inventing new drug delivery approaches in the neurodegenerative disorder: Multiple sclerosis. Brain Res 2024; 1822:148674. [PMID: 37952871 DOI: 10.1016/j.brainres.2023.148674] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
The tight junction of endothelial cells in the central nervous system (CNS) has an ideal characteristic, acting as a biological barrier that can securely regulate the movement of molecules in the brain. Tightly closed astrocyte cell junctions on blood capillaries are the blood-brain barrier (BBB). This biological barrier prohibits the entry of polar drugs, cells, and ions, which protect the brain from harmful toxins. However, delivering any therapeutic agent to the brain in neurodegenerative disorders (i.e., schizophrenia, multiple sclerosis, etc.) is extremely difficult. Active immune responses such as microglia, astrocytes, and lymphocytes cross the BBB and attack the nerve cells, which causes the demyelination of neurons. Therefore, there is a hindrance in transmitting electrical signals properly, resulting in blindness, paralysis, and neuropsychiatric problems. The main objective of this article is to shed light on the performance of biomaterials, which will help researchers to create nanocarriers that can cross the blood-brain barrier and achieve a therapeutic concentration of drugs in the CNS of patients with multiple sclerosis (MS). The present review focuses on the importance of biomaterials with diagnostic and therapeutic efficacy that can help enhance multiple sclerosis therapeutic potential. Currently, the development of MS in animal models is limited by immune responses, which prevent MS induction in healthy animals. Therefore, this article also showcases animal models currently used for treating MS. A future advance in developing a novel effective strategy for treating MS is now a potential area of research.
Collapse
Affiliation(s)
- Dnyandev G Gadhave
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA; Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune 413130, Maharashtra, India.
| | - Vrashabh V Sugandhi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Chandrakant R Kokare
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India
| |
Collapse
|
14
|
Liblau RS, Latorre D, Kornum BR, Dauvilliers Y, Mignot EJ. The immunopathogenesis of narcolepsy type 1. Nat Rev Immunol 2024; 24:33-48. [PMID: 37400646 DOI: 10.1038/s41577-023-00902-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2023] [Indexed: 07/05/2023]
Abstract
Narcolepsy type 1 (NT1) is a chronic sleep disorder resulting from the loss of a small population of hypothalamic neurons that produce wake-promoting hypocretin (HCRT; also known as orexin) peptides. An immune-mediated pathology for NT1 has long been suspected given its exceptionally tight association with the MHC class II allele HLA-DQB1*06:02, as well as recent genetic evidence showing associations with polymorphisms of T cell receptor genes and other immune-relevant loci and the increased incidence of NT1 that has been observed after vaccination with the influenza vaccine Pandemrix. The search for both self-antigens and foreign antigens recognized by the pathogenic T cell response in NT1 is ongoing. Increased T cell reactivity against HCRT has been consistently reported in patients with NT1, but data demonstrating a primary role for T cells in neuronal destruction are currently lacking. Animal models are providing clues regarding the roles of autoreactive CD4+ and CD8+ T cells in the disease. Elucidation of the pathogenesis of NT1 will allow for the development of targeted immunotherapies at disease onset and could serve as a model for other immune-mediated neurological diseases.
Collapse
Affiliation(s)
- Roland S Liblau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, Toulouse, France.
- Department of Immunology, Toulouse University Hospitals, Toulouse, France.
| | | | - Birgitte R Kornum
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yves Dauvilliers
- National Reference Center for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia and Kleine-Levin Syndrome, Department of Neurology, Gui-de-Chauliac Hospital, CHU de Montpellier, Montpellier, France
- INSERM Institute for Neurosciences of Montpellier, Montpellier, France
| | - Emmanuel J Mignot
- Stanford University, Center for Narcolepsy, Department of Psychiatry and Behavioral Sciences, Palo Alto, CA, USA.
| |
Collapse
|
15
|
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
|
16
|
Jiao M, Sun Y, Shi J, Zhang N, Tang X, Fan A, Liu S, Dai C, Qian Z, Zhang F, Wang C, Chen H, Zheng F. IL-33 and HMGB1 modulate the progression of EAE via oppositely regulating each other. Int Immunopharmacol 2023; 122:110653. [PMID: 37467690 DOI: 10.1016/j.intimp.2023.110653] [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: 12/27/2022] [Revised: 05/19/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
Interleukin-33 (IL-33) and high mobility group box 1 (HMGB1) have been reported to play crucial and distinct roles in experimental autoimmune encephalomyelitis (EAE). However, little is known about their interaction in the progression of EAE. In this study, the dynamic expression and release of IL-33 and HMGB1 in different stages of EAE in vivo, and their interaction in vitro were explored. We found that HMGB1 was dominant in pre-onset stage of EAE, while IL-33 was dominant in peak stage. Moreover, both blockade of extracellular HMGB1 in the central nervous system (CNS) and conditional knockout of HMGB1 in astrocytes decreased IL-33 release. HMGB1 promoted the release of IL-33, while IL-33 reduced the release of HMGB1 from primary astrocytes in vitro. Taken together, IL-33 and HMGB1 in the CNS jointly participate in the EAE progression and the inhibitory effect of IL-33 on HMGB1 may be involved in the self-limiting of EAE.
Collapse
Affiliation(s)
- Mengya Jiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yan Sun
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central Minzu University, Wuhan 430074, China; College of Life Sciences, South-Central Minzu University, Wuhan 430074, China
| | - Junyu Shi
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Na Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xuhuan Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Anqi Fan
- College of Life Science, Yangtze University, Jingzhou 434025, China
| | - Shiwang Liu
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chan Dai
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhigang Qian
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Feng Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chenchen Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Huoying Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin 541199, China; Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin 541199, China.
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China.
| |
Collapse
|
17
|
Zhao M, Zhang Y, Wu J, Li X, Gao Y. Early urinary candidate biomarkers and clinical outcomes of intervention in a rat model of experimental autoimmune encephalomyelitis. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230118. [PMID: 37621667 PMCID: PMC10445012 DOI: 10.1098/rsos.230118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023]
Abstract
Multiple sclerosis is a chronic autoimmune demyelinating disease of the central nervous system and is difficult to diagnose in early stages. Without homeostatic control, urine was reported to have the ability to accumulate early changes in the body. We expect that urinary proteome can reflect early changes in the nervous system. The early urinary proteome changes in a most employed multiple sclerosis rat model (experimental autoimmune encephalomyelitis) were analysed to explore early urinary candidate biomarkers, and early treatment of methylprednisolone was used to evaluate the therapeutic effect. Twenty-five urinary proteins were altered at day 7 when there were no clinical symptoms and obvious histological changes. Fourteen were reported to be differently expressed in the serum/cerebrospinal fluid/brain tissues of multiple sclerosis patients or animals such as angiotensinogen and matrix metallopeptidase 8. Functional analysis showed that the dysregulated proteins were associated with asparagine degradation, neuroinflammation and lipid metabolism. After the early treatment of methylprednisolone, the incidence of encephalomyelitis in the intervention group was only 1/13. This study demonstrates that urine may be a good source of biomarkers for the early detection of multiple sclerosis. These findings may provide important information for early diagnosis and intervention of multiple sclerosis in the future.
Collapse
Affiliation(s)
- Mindi Zhao
- Department of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yameng Zhang
- Gene Engineering Drug and Biotechnology Beijing Key Laboratory, College of Life Sciences, Beijing Normal University, Beijing 100875, People's Republic of China
- Department of Pathology, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jianqiang Wu
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Xundou Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences/School of Basic Medicine, Peking Union Medical College, Beijing, People's Republic of China
| | - Youhe Gao
- Gene Engineering Drug and Biotechnology Beijing Key Laboratory, College of Life Sciences, Beijing Normal University, Beijing 100875, People's Republic of China
| |
Collapse
|
18
|
Dedoni S, Scherma M, Camoglio C, Siddi C, Dazzi L, Puliga R, Frau J, Cocco E, Fadda P. An overall view of the most common experimental models for multiple sclerosis. Neurobiol Dis 2023:106230. [PMID: 37453561 DOI: 10.1016/j.nbd.2023.106230] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis (MS) is a complex chronic disease with an unknown etiology. It is considered an inflammatory demyelinating and neurodegenerative disorder of the central nervous system (CNS) characterized, in most cases, by an unpredictable onset of relapse and remission phases. The disease generally starts in subjects under 40; it has a higher incidence in women and is described as a multifactorial disorder due to the interaction between genetic and environmental risk factors. Unfortunately, there is currently no definitive cure for MS. Still, therapies can modify the disease's natural history, reducing the relapse rate and slowing the progression of the disease or managing symptoms. The limited access to human CNS tissue slows down. It limits the progression of research on MS. This limit has been partially overcome over the years by developing various experimental models to study this disease. Animal models of autoimmune demyelination, such as experimental autoimmune encephalomyelitis (EAE) and viral and toxin or transgenic MS models, represent the most significant part of MS research approaches. These models have now been complemented by ex vivo studies, using organotypic brain slice cultures and in vitro, through induced Pluripotent Stem cells (iPSCs). We will discuss which clinical features of the disorders might be reproduced and investigated in vivo, ex vivo, and in vitro in models commonly used in MS research to understand the processes behind the neuropathological events occurring in the CNS of MS patients. The primary purpose of this review is to give the reader a global view of the main paradigms used in MS research, spacing from the classical animal models to transgenic mice and 2D and 3D cultures.
Collapse
Affiliation(s)
- S Dedoni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - M Scherma
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Camoglio
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Siddi
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - L Dazzi
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - R Puliga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - J Frau
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy
| | - E Cocco
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy; Department Medical Science and Public Health, University of Cagliari, Italy.
| | - P Fadda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
19
|
Ackun-Farmmer MA, Jewell CM. Delivery route considerations for designing antigen-specific biomaterial strategies to combat autoimmunity. ADVANCED NANOBIOMED RESEARCH 2023; 3:2200135. [PMID: 36938103 PMCID: PMC10019031 DOI: 10.1002/anbr.202200135] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Disease modifying drugs and biologics used to treat autoimmune diseases, although promising, are non-curative. As the field moves towards development of new approaches to treat autoimmune disease, antigen-specific therapies immunotherapies (ASITs) have emerged. Despite clinical approval of ASITs for allergies, clinical trials using soluble ASITs for autoimmunity have been largely unsuccessful. A major effort to address this shortcoming is the use of biomaterials to harness the features unique to specific delivery routes. This review focuses on biomaterials being developed for delivery route-specific strategies to induce antigen-specific responses in autoimmune diseases such as multiple sclerosis, type 1 diabetes, rheumatoid arthritis, and celiac disease. We first discuss the delivery strategies used in ongoing and completed clinical trials in autoimmune ASITs. Next, we highlight pre-clinical biomaterial approaches from the most recent 3 years in the context of these same delivery route considerations. Lastly, we provide discussion on the gaps remaining in biomaterials development and comment on the need to consider delivery routes in the process of designing biomaterials for ASITs.
Collapse
Affiliation(s)
- Marian A Ackun-Farmmer
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- US Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, 21201, USA
- Robert E. Fischell Institute for Biomedical Devices, College Park, MD, 20742, USA
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, 21201, USA
| |
Collapse
|
20
|
Offner H, Lockwood D, Meza-Romero R, Vandenbark AA. PD-L1 is required for estrogen-induced protection against severe EAE in IL-10 deficient mice 1. Metab Brain Dis 2023; 38:589-599. [PMID: 36454506 PMCID: PMC9976593 DOI: 10.1007/s11011-022-01129-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND IL-10 knockout (KO) mice can be protected against experimental autoimmune encephalomyelitis (EAE) with low-dose estrogen (E2) treatment similar to wild type (WT) mice, indicating that IL-10 is not required for E2-induced EAE protection. Our previous study demonstrated that E2 treatment induced an increase in programmed death ligands 1 (PD-L1) and 2 (PD-L2) on monocytes and macrophages in the periphery and within the CNS. In this study, we selectively inhibited the function of PD-L1 and PD-L2 to evaluate their critical role in maintaining E2-induced protection against EAE in IL-10-KO mice. METHODS This study used female IL-10 KO mice pre-treated with either E2 or sham pellets seven days prior to induction of EAE and subsequently treated with Vehicle or antibodies to PD-L1, PD-L2 or respective isotype controls. Mice were scored daily for EAE severity over 21 days post-EAE induction. Cells from the spleen and brain were evaluated by flow cytometry. RESULTS Differences in EAE severity were assessed in E2 and sham pre-treated IL-10-KO mice treated with α-PD-L1 or α-PD-L2 antibodies over the course of disease compared to treatment with Vehicle or isotype control antibodies. The results revealed real-time development of severe EAE in E2-pre-treated IL-10-KO mice treated with α-PD-L1 but not α-PD-L2 antibodies, mediated in part by increased percentages of activated CD74+CD11b+ myeloid cells in spleen and brain as well as splenic B-cells, T-cells and CD73+ cells. CONCLUSION These results demonstrate unequivocally that PD-L1 but not PD-L2 was required to retain the inhibitory effects of E2 on clinical EAE scores in female IL-10-KO mice and further implicate the emergence of the MIF/CD74 axis as a contributing pathogenic mechanism.
Collapse
Affiliation(s)
- Halina Offner
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA.
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.
- Veterans Affairs Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., 97239, Portland, OR, USA.
| | - Denesa Lockwood
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
| | - Roberto Meza-Romero
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
21
|
Phenethyl Ester of Gallic Acid Ameliorates Experimental Autoimmune Encephalomyelitis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248770. [PMID: 36557903 PMCID: PMC9782083 DOI: 10.3390/molecules27248770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/18/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Gallic acid is a phenolic acid present in various plants, nuts, and fruits. It is well known for its anti-oxidative and anti-inflammatory properties. The phenethyl ester of gallic acid (PEGA) was synthesized with the aim of increasing the bioavailability of gallic acid, and thus its pharmacological potential. Here, the effects of PEGA on encephalitogenic cells were examined, and PEGA was found to modulate the inflammatory activities of T cells and macrophages/microglia. Specifically, PEGA reduced the release of interleukin (IL)-17 and interferon (IFN)-γ from T cells, as well as NO, and IL-6 from macrophages/microglia. Importantly, PEGA ameliorated experimental autoimmune encephalomyelitis, an animal model of chronic inflammatory disease of the central nervous system (CNS)-multiple sclerosis. Thus, PEGA is a potent anti-inflammatory compound with a perspective to be further explored in the context of CNS autoimmunity and other chronic inflammatory disorders.
Collapse
|
22
|
Humanized zebrafish as a tractable tool for in vivo evaluation of pro-myelinating drugs. Cell Chem Biol 2022; 29:1541-1555.e7. [PMID: 36126653 DOI: 10.1016/j.chembiol.2022.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/25/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
Therapies that promote neuroprotection and axonal survival by enhancing myelin regeneration are an unmet need to prevent disability progression in multiple sclerosis. Numerous potentially beneficial compounds have originated from phenotypic screenings but failed in clinical trials. It is apparent that current cell- and animal-based disease models are poor predictors of positive treatment options, arguing for novel experimental approaches. Here we explore the experimental power of humanized zebrafish to foster the identification of pro-remyelination compounds via specific inhibition of GPR17. Using biochemical and imaging techniques, we visualize the expression of zebrafish (zf)-gpr17 during the distinct stages of oligodendrocyte development, thereby demonstrating species-conserved expression between zebrafish and mammals. We also demonstrate species-conserved function of zf-Gpr17 using genetic loss-of-function and rescue techniques. Finally, using GPR17-humanized zebrafish, we provide proof of principle for in vivo analysis of compounds acting via targeted inhibition of human GPR17. We anticipate that GPR17-humanized zebrafish will markedly improve the search for effective pro-myelinating pharmacotherapies.
Collapse
|
23
|
Stegnjaić G, Lazarević M, Diamantis D, Djedović N, Jevtić B, Stanisavljević S, Dimitrijević M, Momčilović M, Tzakos AG, Miljković Đ. Phenethyl ester of rosmarinic acid ameliorates experimental autoimmune encephalomyelitis. Immunol Lett 2022; 251-252:9-19. [DOI: 10.1016/j.imlet.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/13/2022] [Accepted: 09/25/2022] [Indexed: 11/24/2022]
|
24
|
Osorio-Querejeta I, Alberro A, Suárez J, Sáenz-Cuesta M, Oregi A, Moles L, Muñoz-Culla M, Otaegui D. The innovative animal monitoring device for experimental autoimmune encephalomyelitis (“I AM D EAE”): A more detailed evaluation for improved results. Mult Scler Relat Disord 2022; 63:103836. [DOI: 10.1016/j.msard.2022.103836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
|
25
|
Hohlfeld R, Liblau RS. Toward identification of personalized immunological profiles in multiple sclerosis. SCIENCE ADVANCES 2022; 8:eabq4849. [PMID: 35476442 PMCID: PMC11324011 DOI: 10.1126/sciadv.abq4849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 06/14/2023]
Abstract
The diversity of four previously unidentified autoantigens found in multiple sclerosis mirrors its notorious clinical variability.
Collapse
Affiliation(s)
- Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-University of Munich, and Munich Cluster of Systems Neurology, Munich, Germany
| | - Roland S. Liblau
- Department of Immunology, Toulouse University Hospital, 31300 Toulouse, France
- Toulouse Institute for Infectious and Inflammatory Diseases, University of Toulouse, CNRS, INSERM, Toulouse, France
| |
Collapse
|
26
|
Remlinger J, Madarasz A, Guse K, Hoepner R, Bagnoud M, Meli I, Feil M, Abegg M, Linington C, Shock A, Boroojerdi B, Kiessling P, Smith B, Enzmann V, Chan A, Salmen A. Antineonatal Fc Receptor Antibody Treatment Ameliorates MOG-IgG-Associated Experimental Autoimmune Encephalomyelitis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/2/e1134. [PMID: 35027475 PMCID: PMC8759074 DOI: 10.1212/nxi.0000000000001134] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/03/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND OBJECTIVES Myelin oligodendrocyte glycoprotein antibody-associated disorder (MOGAD) is a rare, autoimmune demyelinating CNS disorder, distinct from multiple sclerosis and neuromyelitis optica spectrum disorder. Characterized by pathogenic immunoglobulin G (IgG) antibodies against MOG, a potential treatment strategy for MOGAD is to reduce circulating IgG levels, e.g., by interference with the IgG recycling pathway mediated by the neonatal Fc receptor (FcRn). Although the optic nerve is often detrimentally involved in MOGAD, the effect of FcRn blockade on the visual pathway has not been assessed. Our objective was to investigate effects of a monoclonal anti-FcRn antibody in murine MOG-IgG-associated experimental autoimmune encephalomyelitis (EAE). METHODS We induced active MOG35-55 EAE in C57Bl/6 mice followed by the application of a monoclonal MOG-IgG (8-18C5) 10 days postimmunization (dpi). Animals were treated with either a specific monoclonal antibody against FcRn (α-FcRn, 4470) or an isotype-matched control IgG on 7, 10, and 13 dpi. Neurologic disability was scored daily on a 10-point scale. Visual acuity was assessed by optomotor reflex. Histopathologic hallmarks of disease were assessed in the spinal cord, optic nerve, and retina. Immune cell infiltration was visualized by immunohistochemistry, demyelination by Luxol fast blue staining and complement deposition and number of retinal ganglion cells by immunofluorescence. RESULTS In MOG-IgG-augmented MOG35-55 EAE, anti-FcRn treatment significantly attenuated neurologic disability over the course of disease (mean area under the curve and 95% confidence intervals (CIs): α-FcRn [n = 27], 46.02 [37.89-54.15]; isotype IgG [n = 24], 66.75 [59.54-73.96], 3 independent experiments), correlating with reduced amounts of demyelination and macrophage infiltration into the spinal cord. T- and B-cell infiltration and complement deposition remained unchanged. Compared with isotype, anti-FcRn treatment prevented reduction of visual acuity over the course of disease (median cycles/degree and interquartile range: α-FcRn [n = 16], 0.50 [0.48-0.55] to 0.50 [0.48-0.58]; isotype IgG [n = 17], 0.50 [0.49-0.54] to 0.45 [0.39-0.51]). DISCUSSION We show preserved optomotor response and ameliorated course of disease after anti-FcRn treatment in an experimental model using a monoclonal MOG-IgG to mimic MOGAD. Selectively targeting FcRn might represent a promising therapeutic approach in MOGAD.
Collapse
Affiliation(s)
- Jana Remlinger
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Adrian Madarasz
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Kirsten Guse
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Robert Hoepner
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Maud Bagnoud
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Ivo Meli
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Moritz Feil
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Mathias Abegg
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Christopher Linington
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Anthony Shock
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Babak Boroojerdi
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Peter Kiessling
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Bryan Smith
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Volker Enzmann
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Andrew Chan
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Anke Salmen
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany.
| |
Collapse
|
27
|
Jiang R, Qin Y, Wang Y, Xu X, Chen H, Xu K, Zhang M. Dynamic Number and Function of IL-10-Producing Regulatory B Cells in the Immune Microenvironment at Distinct Stages of Type 1 Diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1034-1041. [PMID: 35140133 DOI: 10.4049/jimmunol.2100357] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 12/17/2021] [Indexed: 01/12/2023]
Abstract
The critical role of IL-10-producing B cells (B10 cells) with a unique CD1dhiCD5+ phenotype in suppressing autoimmune responses and relieving inflammation has been demonstrated in several models of autoimmune diseases. However, the regulatory role of B10 cells in T cell-mediated autoimmune responses during the natural history of type 1 diabetes is unclear. In this study, we used the NOD mouse model of autoimmune diabetes to clarify the changes and potential mechanisms of B10 cells for disease. Compared with B10 cells present in the 4-wk-old normoglycemic NOD mice, the frequency of B10 cells was increased in the insulitis and diabetic NOD mice, with the highest proportion in the insulitis NOD mice. The changes in the relative number of B10 cells were most pronounced in the pancreas-draining lymph nodes. The pathogenic T cells, including Th1 and Th17 cells, remarkably increased. The assays in vitro showed that B10 cells in the NOD mice did not inhibit the proliferation of CD4+CD25- T cells. They also had no regulatory effect on IFN-γ and IL-4 secretion or on Foxp3 expression of T cells. B10 cells suppressed T cell-mediated autoimmune responses via an IL-10-dependent pathway. In contrast, B10 cells in the NOD mice exhibited a significant reduction in IL-10 production. In summary, a defect in the number and function of B10 cells may participate in the development and progression of type 1 diabetes.
Collapse
Affiliation(s)
- Ruimei Jiang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Endocrinology, Fuyang People's Hospital, Fuyang, China; and
| | - Yao Qin
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yueshu Wang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyu Xu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Heng Chen
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kuanfeng Xu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mei Zhang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China;
| |
Collapse
|
28
|
Song YC, Liu CT, Lee HJ, Yen HR. Cordycepin prevents and ameliorates experimental autoimmune encephalomyelitis by inhibiting leukocyte infiltration and reducing neuroinflammation. Biochem Pharmacol 2022; 197:114918. [DOI: 10.1016/j.bcp.2022.114918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/30/2022]
|
29
|
Weissert R, Hugosson T, Petzold A. Upregulated Retinal Neurofilament Expression in Experimental Optic Neuritis. Neuroophthalmology 2022; 46:215-219. [PMID: 35859627 PMCID: PMC9291678 DOI: 10.1080/01658107.2022.2025852] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In optic neuritis (ON), transient thickening of the macular retinal nerve fibre layer (RNFL) can be observed. This optical coherence tomography-based observation is not understood. The axonal diameter correlates with the neurofilament (Nf) protein content, but there are no data on the retinal tissue concentration of Nfs. The myelin-oligodendrocyte-glycoprotein (MOG) induced experimental autoimmune encephalomyelitis (EAE) model was used to investigate the retinas of Brown Norway rats with (i) visual evoked potentials (VEP) confirmed ON, (ii) VEP confirmed absence of ON and (iii) control animals. Twenty retinas were collected from MOG-EAE and control rats 27 days after immunisation. Retinal tissue Nf concentrations per total protein (μg/mg) were significantly higher in MOG-EAE rats with ON (median 4.29, interquartile range [IQR] 3.41-5.97) compared with MOG-EAE rats without ON (1.14, IQR 1.10-1.67) or control rats (0.93, IQR 0.45-4.00). The data suggest that up-regulation of Nf expression in the retinal ganglion cells precedes development of RNFL atrophy and plausibly explains the transient increase of axonal diameter and RNFL thickening.
Collapse
Affiliation(s)
- Robert Weissert
- Department of Neurology, University Hospital of Regensburg, Regensburg, Germany
| | - Therése Hugosson
- Department of Ophthalmology, St Erik Eye Hospital, Stockholm, Sweden,Department of Clinical Sciences, Ophthalmology, Lund University, Lund, Sweden
| | - Axel Petzold
- Queen Square Institute of Neurology, UCL, Department of Neuroimmunology & The National Hospital for Neurology and Neurosurgery, Moorfields Eye Hospital, London, UK,Amsterdam UMC, Locatie VUmc, Departments of Neurology and Ophthalmology, NL, Moorfields Eye Hospital, London, UK,CONTACT Axel PetzoldQueen Square Institute of Neurology, 9th floor, Room 914, Queen Square, London, WC1N 3BG, UK
| |
Collapse
|
30
|
Mirabelli E, Elkabes S. Neuropathic Pain in Multiple Sclerosis and Its Animal Models: Focus on Mechanisms, Knowledge Gaps and Future Directions. Front Neurol 2022; 12:793745. [PMID: 34975739 PMCID: PMC8716468 DOI: 10.3389/fneur.2021.793745] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/17/2021] [Indexed: 12/22/2022] Open
Abstract
Multiple sclerosis (MS) is a multifaceted, complex and chronic neurological disease that leads to motor, sensory and cognitive deficits. MS symptoms are unpredictable and exceedingly variable. Pain is a frequent symptom of MS and manifests as nociceptive or neuropathic pain, even at early disease stages. Neuropathic pain is one of the most debilitating symptoms that reduces quality of life and interferes with daily activities, particularly because conventional pharmacotherapies do not adequately alleviate neuropathic pain. Despite advances, the mechanisms underlying neuropathic pain in MS remain elusive. The majority of the studies investigating the pathophysiology of MS-associated neuropathic pain have been performed in animal models that replicate some of the clinical and neuropathological features of MS. Experimental autoimmune encephalomyelitis (EAE) is one of the best-characterized and most commonly used animal models of MS. As in the case of individuals with MS, rodents affected by EAE manifest increased sensitivity to pain which can be assessed by well-established assays. Investigations on EAE provided valuable insights into the pathophysiology of neuropathic pain. Nevertheless, additional investigations are warranted to better understand the events that lead to the onset and maintenance of neuropathic pain in order to identify targets that can facilitate the development of more effective therapeutic interventions. The goal of the present review is to provide an overview of several mechanisms implicated in neuropathic pain in EAE by summarizing published reports. We discuss current knowledge gaps and future research directions, especially based on information obtained by use of other animal models of neuropathic pain such as nerve injury.
Collapse
Affiliation(s)
- Ersilia Mirabelli
- Reynolds Family Spine Laboratory, Department of Neurosurgery, New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, United States.,Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA, United States
| | - Stella Elkabes
- Reynolds Family Spine Laboratory, Department of Neurosurgery, New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
31
|
Kesharwani A, Schwarz K, Dembla E, Dembla M, Schmitz F. Early Changes in Exo- and Endocytosis in the EAE Mouse Model of Multiple Sclerosis Correlate with Decreased Synaptic Ribbon Size and Reduced Ribbon-Associated Vesicle Pools in Rod Photoreceptor Synapses. Int J Mol Sci 2021; 22:ijms221910789. [PMID: 34639129 PMCID: PMC8509850 DOI: 10.3390/ijms221910789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/17/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system that finally leads to demyelination. Demyelinating optic neuritis is a frequent symptom in MS. Recent studies also revealed synapse dysfunctions in MS patients and MS mouse models. We previously reported alterations of photoreceptor ribbon synapses in the experimental auto-immune encephalomyelitis (EAE) mouse model of MS. In the present study, we found that the previously observed decreased imunosignals of photoreceptor ribbons in early EAE resulted from a decrease in synaptic ribbon size, whereas the number/density of ribbons in photoreceptor synapses remained unchanged. Smaller photoreceptor ribbons are associated with fewer docked and ribbon-associated vesicles. At a functional level, depolarization-evoked exocytosis as monitored by optical recording was diminished even as early as on day 7 after EAE induction. Moreover compensatory, post-depolarization endocytosis was decreased. Decreased post-depolarization endocytosis in early EAE correlated with diminished synaptic enrichment of dynamin3. In contrast, basal endocytosis in photoreceptor synapses of resting non-depolarized retinal slices was increased in early EAE. Increased basal endocytosis correlated with increased de-phosphorylation of dynamin1. Thus, multiple endocytic pathways in photoreceptor synapse are differentially affected in early EAE and likely contribute to the observed synapse pathology in early EAE.
Collapse
Affiliation(s)
- Ajay Kesharwani
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, 66421 Homburg, Germany; (K.S.); (E.D.); (M.D.); (F.S.)
- Correspondence:
| | - Karin Schwarz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, 66421 Homburg, Germany; (K.S.); (E.D.); (M.D.); (F.S.)
| | - Ekta Dembla
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, 66421 Homburg, Germany; (K.S.); (E.D.); (M.D.); (F.S.)
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mayur Dembla
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, 66421 Homburg, Germany; (K.S.); (E.D.); (M.D.); (F.S.)
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Frank Schmitz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, 66421 Homburg, Germany; (K.S.); (E.D.); (M.D.); (F.S.)
| |
Collapse
|
32
|
Moulson AJ, Squair JW, Franklin RJM, Tetzlaff W, Assinck P. Diversity of Reactive Astrogliosis in CNS Pathology: Heterogeneity or Plasticity? Front Cell Neurosci 2021; 15:703810. [PMID: 34381334 PMCID: PMC8349991 DOI: 10.3389/fncel.2021.703810] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/02/2021] [Indexed: 01/02/2023] Open
Abstract
Astrocytes are essential for the development and homeostatic maintenance of the central nervous system (CNS). They are also critical players in the CNS injury response during which they undergo a process referred to as "reactive astrogliosis." Diversity in astrocyte morphology and gene expression, as revealed by transcriptional analysis, is well-recognized and has been reported in several CNS pathologies, including ischemic stroke, CNS demyelination, and traumatic injury. This diversity appears unique to the specific pathology, with significant variance across temporal, topographical, age, and sex-specific variables. Despite this, there is limited functional data corroborating this diversity. Furthermore, as reactive astrocytes display significant environmental-dependent plasticity and fate-mapping data on astrocyte subsets in the adult CNS is limited, it remains unclear whether this diversity represents heterogeneity or plasticity. As astrocytes are important for neuronal survival and CNS function post-injury, establishing to what extent this diversity reflects distinct established heterogeneous astrocyte subpopulations vs. environmentally dependent plasticity within established astrocyte subsets will be critical for guiding therapeutic development. To that end, we review the current state of knowledge on astrocyte diversity in the context of three representative CNS pathologies: ischemic stroke, demyelination, and traumatic injury, with the goal of identifying key limitations in our current knowledge and suggesting future areas of research needed to address them. We suggest that the majority of identified astrocyte diversity in CNS pathologies to date represents plasticity in response to dynamically changing post-injury environments as opposed to heterogeneity, an important consideration for the understanding of disease pathogenesis and the development of therapeutic interventions.
Collapse
Affiliation(s)
- Aaron J. Moulson
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
| | - Jordan W. Squair
- Department of Clinical Neuroscience, Faculty of Life Sciences, Center for Neuroprosthetics and Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), NeuroRestore, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Robin J. M. Franklin
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Peggy Assinck
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
33
|
Wei L, Xue Z, Lan B, Yuan S, Li Y, Guo C, Zhang R, Ding R, Shen H. Arctigenin Exerts Neuroprotective Effect by Ameliorating Cortical Activities in Experimental Autoimmune Encephalomyelitis In Vivo. Front Immunol 2021; 12:691590. [PMID: 34349758 PMCID: PMC8327179 DOI: 10.3389/fimmu.2021.691590] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/01/2021] [Indexed: 11/19/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic disease in the central nervous system (CNS), characterized by inflammatory cells that invade into the brain and the spinal cord. Among a bulk of different MS models, the most widely used and best understood rodent model is experimental autoimmune encephalomyelitis (EAE). Arctigenin, a botanical extract from Arctium lappa, is reported to exhibit pharmacological properties, including anti-inflammation and neuroprotection. However, the effects of arctigenin on neural activity attacked by inflammation in MS are still unclear. Here, we use two-photon calcium imaging to observe the activity of somatosensory cortex neurons in awake EAE mice in vivo and found added hyperactive cells, calcium influx, network connectivity, and synchronization, mainly at preclinical stage of EAE model. Besides, more silent cells and decreased calcium influx and reduced network synchronization accompanied by a compensatory rise in functional connectivity are found at the remission stage. Arctigenin treatment not only restricts inordinate individually neural spiking, calcium influx, and network activity at preclinical stage but also restores neuronal activity and communication at remission stage. In addition, we confirm that the frequency of AMPA receptor-mediated spontaneous excitatory postsynaptic current (sEPSC) is also increased at preclinical stage and can be blunted by arctigenin. These findings suggest that excitotoxicity characterized by calcium influx is involved in EAE at preclinical stage. What is more, arctigenin exerts neuroprotective effect by limiting hyperactivity at preclinical stage and ameliorates EAE symptoms, indicating that arctigenin could be a potential therapeutic drug for neuroprotection in MS-related neuropsychological disorders.
Collapse
Affiliation(s)
- Liangpeng Wei
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Zhenyi Xue
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin, China
| | - Baihui Lan
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Shiyang Yuan
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Yuanyuan Li
- Innovation Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cunle Guo
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Rongxin Zhang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ran Ding
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Hui Shen
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
- Research Institute of Neurology, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
34
|
Dash PK, Gorantla S, Poluektova L, Hasan M, Waight E, Zhang C, Markovic M, Edagwa B, Machhi J, Olson KE, Wang X, Mosley RL, Kevadiya B, Gendelman HE. Humanized Mice for Infectious and Neurodegenerative disorders. Retrovirology 2021; 18:13. [PMID: 34090462 PMCID: PMC8179712 DOI: 10.1186/s12977-021-00557-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022] Open
Abstract
Humanized mice model human disease and as such are used commonly for research studies of infectious, degenerative and cancer disorders. Recent models also reflect hematopoiesis, natural immunity, neurobiology, and molecular pathways that influence disease pathobiology. A spectrum of immunodeficient mouse strains permit long-lived human progenitor cell engraftments. The presence of both innate and adaptive immunity enables high levels of human hematolymphoid reconstitution with cell susceptibility to a broad range of microbial infections. These mice also facilitate investigations of human pathobiology, natural disease processes and therapeutic efficacy in a broad spectrum of human disorders. However, a bridge between humans and mice requires a complete understanding of pathogen dose, co-morbidities, disease progression, environment, and genetics which can be mirrored in these mice. These must be considered for understanding of microbial susceptibility, prevention, and disease progression. With known common limitations for access to human tissues, evaluation of metabolic and physiological changes and limitations in large animal numbers, studies in mice prove important in planning human clinical trials. To these ends, this review serves to outline how humanized mice can be used in viral and pharmacologic research emphasizing both current and future studies of viral and neurodegenerative diseases. In all, humanized mouse provides cost-effective, high throughput studies of infection or degeneration in natural pathogen host cells, and the ability to test transmission and eradication of disease.
Collapse
Affiliation(s)
- Prasanta K Dash
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Larisa Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Emiko Waight
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chen Zhang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Milica Markovic
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Xinglong Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bhavesh Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
35
|
Abstract
Multiple sclerosis (MS) is a complex inflammatory disease of the central nervous system (CNS) with an unknown etiology. Thereby, MS is not a uniform disease but rather represents a spectrum of disorders, where each aspect needs to be modeled with specific requirements-for a systematic overview see our previous issue of this review (Kurschus, Wortge, & Waisman, 2011). However, there is broad consensus about the critical involvement of the immune system in the disease pathogenesis. To better understand how the immune system contributes to CNS autoimmunity, the model of experimental autoimmune encephalomyelitis (EAE) was developed. EAE can be induced in susceptible animals in many different ways, with the most popular protocol involving the activation of self-reactive T cells by a peptide based on the myelin oligodendrocyte glycoprotein sequence. In the last 10 years this model has led to major advances in our understanding of the immune system, especially the nature of IL-17-producing T cells (Th17 cells), host-microbiome interactions, the gut-brain axis and how the immune system can cause damage in different regions of the brain and the spinal cord. This update summarizes some of the main achievements in the field in the last 10 years.
Collapse
Affiliation(s)
- Tommy Regen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
36
|
Complete Freund's adjuvant-free experimental autoimmune encephalomyelitis in Dark Agouti rats is a valuable tool for multiple sclerosis studies. J Neuroimmunol 2021; 354:577547. [PMID: 33765502 DOI: 10.1016/j.jneuroim.2021.577547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/02/2021] [Accepted: 03/12/2021] [Indexed: 12/21/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is classically induced with complete Freund's adjuvant (CFA). The immune response against CFA has a confounding influence on the translational capacity of EAE as a multiple sclerosis model. Here, we compare clinical, cellular and molecular properties between syngeneic spinal cord homogenate (SCH)- and SCH + CFA-immunized Dark Agouti rats. EAE signs were observed earlier and the cumulative clinical score was higher without CFA. Also, a higher number of immune cells infiltrates in the spinal cords was noticed at the peak of EAE without CFA. High spinal cord abundance of CD8+CD11bc+MHC class II+ cells was detected in SCH-immunized rats. Myelin basic protein -specific response can be elicited in the cells from the lymph nodes draining the site of SCH immunization. This CFA-free EAE is a reliable multiple sclerosis model.
Collapse
|
37
|
Derdelinckx J, Cras P, Berneman ZN, Cools N. Antigen-Specific Treatment Modalities in MS: The Past, the Present, and the Future. Front Immunol 2021; 12:624685. [PMID: 33679769 PMCID: PMC7933447 DOI: 10.3389/fimmu.2021.624685] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
Antigen-specific therapy for multiple sclerosis may lead to a more effective therapy by induction of tolerance to a wide range of myelin-derived antigens without hampering the normal surveillance and effector function of the immune system. Numerous attempts to restore tolerance toward myelin-derived antigens have been made over the past decades, both in animal models of multiple sclerosis and in clinical trials for multiple sclerosis patients. In this review, we will give an overview of the current approaches for antigen-specific therapy that are in clinical development for multiple sclerosis as well provide an insight into the challenges for future antigen-specific treatment strategies for multiple sclerosis.
Collapse
Affiliation(s)
- Judith Derdelinckx
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Division of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Patrick Cras
- Division of Neurology, Antwerp University Hospital, Edegem, Belgium.,Born Bunge Institute, Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
38
|
Mukherjee A, Katiyar R, Dembla E, Dembla M, Kumar P, Belkacemi A, Jung M, Beck A, Flockerzi V, Schwarz K, Schmitz F. Disturbed Presynaptic Ca 2+ Signaling in Photoreceptors in the EAE Mouse Model of Multiple Sclerosis. iScience 2020; 23:101830. [PMID: 33305185 PMCID: PMC7711289 DOI: 10.1016/j.isci.2020.101830] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/10/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease caused by an auto-reactive immune system. Recent studies also demonstrated synapse dysfunctions in MS patients and MS mouse models. We previously observed decreased synaptic vesicle exocytosis in photoreceptor synapses in the EAE mouse model of MS at an early, preclinical stage. In the present study, we analyzed whether synaptic defects are associated with altered presynaptic Ca2+ signaling. Using high-resolution immunolabeling, we found a reduced signal intensity of Cav-channels and RIM2 at active zones in early, preclinical EAE. In line with these morphological alterations, depolarization-evoked increases of presynaptic Ca2+ were significantly smaller. In contrast, basal presynaptic Ca2+ was elevated. We observed a decreased expression of Na+/K+-ATPase and plasma membrane Ca2+ ATPase 2 (PMCA2), but not PMCA1, in photoreceptor terminals of EAE mice that could contribute to elevated basal Ca2+. Thus, complex Ca2+ signaling alterations contribute to synaptic dysfunctions in photoreceptors in early EAE. Less Cav-channels and RIM2 at the active zones of EAE photoreceptor synapses Decreased depolarization-evoked Ca2+-responses in EAE photoreceptor synapses Elevated basal, resting Ca2+ levels in preclinical EAE photoreceptor terminals Decreased expression of PMCA2 and Na+/K+-ATPase in EAE photoreceptor synapses
Collapse
Affiliation(s)
- Amrita Mukherjee
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421 Homburg, Germany
| | - Rashmi Katiyar
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421 Homburg, Germany
| | - Ekta Dembla
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421 Homburg, Germany
| | - Mayur Dembla
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421 Homburg, Germany
| | - Praveen Kumar
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421 Homburg, Germany
| | - Anouar Belkacemi
- Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, Medical School, 66421 Homburg, Germany
| | - Martin Jung
- Institute of Medical Biochemistry and Molecular Biology, Saarland University, Medical School, 66421 Homburg, Germany
| | - Andreas Beck
- Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, Medical School, 66421 Homburg, Germany
| | - Veit Flockerzi
- Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, Medical School, 66421 Homburg, Germany
| | - Karin Schwarz
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421 Homburg, Germany
| | - Frank Schmitz
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421 Homburg, Germany
| |
Collapse
|
39
|
Montgomery TL, Künstner A, Kennedy JJ, Fang Q, Asarian L, Culp-Hill R, D'Alessandro A, Teuscher C, Busch H, Krementsov DN. Interactions between host genetics and gut microbiota determine susceptibility to CNS autoimmunity. Proc Natl Acad Sci U S A 2020; 117:27516-27527. [PMID: 33077601 PMCID: PMC7959502 DOI: 10.1073/pnas.2002817117] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system. The etiology of MS is multifactorial, with disease risk determined by genetics and environmental factors. An emerging risk factor for immune-mediated diseases is an imbalance in the gut microbiome. However, the identity of gut microbes associated with disease risk, their mechanisms of action, and the interactions with host genetics remain obscure. To address these questions, we utilized the principal autoimmune model of MS, experimental autoimmune encephalomyelitis (EAE), together with a genetically diverse mouse model representing 29 unique host genotypes, interrogated by microbiome sequencing and targeted microbiome manipulation. We identified specific gut bacteria and their metabolic functions associated with EAE susceptibility, implicating short-chain fatty acid metabolism as a key element conserved across multiple host genotypes. In parallel, we used a reductionist approach focused on two of the most disparate phenotypes identified in our screen. Manipulation of the gut microbiome by transplantation and cohousing demonstrated that transfer of these microbiomes into genetically identical hosts was sufficient to modulate EAE susceptibility and systemic metabolite profiles. Parallel bioinformatic approaches identified Lactobacillus reuteri as a commensal species unexpectedly associated with exacerbation of EAE in a genetically susceptible host, which was functionally confirmed by bacterial isolation and commensal colonization studies. These results reveal complex interactions between host genetics and gut microbiota modulating susceptibility to CNS autoimmunity, providing insights into microbiome-directed strategies aimed at lowering the risk for autoimmune disease and underscoring the need to consider host genetics and baseline gut microbiome composition.
Collapse
Affiliation(s)
- Theresa L Montgomery
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05401
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Josephine J Kennedy
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05401
| | - Qian Fang
- Department of Medicine, Immunobiology Division, University of Vermont, Burlington, VT 05401
| | - Lori Asarian
- Department of Medicine, Immunobiology Division, University of Vermont, Burlington, VT 05401
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO 80045
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO 80045
| | - Cory Teuscher
- Department of Medicine, Immunobiology Division, University of Vermont, Burlington, VT 05401
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Dimitry N Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05401;
| |
Collapse
|
40
|
Faber H, Kurtoic D, Krishnamoorthy G, Weber P, Pütz B, Müller-Myhsok B, Weber F, Andlauer TFM. Gene Expression in Spontaneous Experimental Autoimmune Encephalomyelitis Is Linked to Human Multiple Sclerosis Risk Genes. Front Immunol 2020; 11:2165. [PMID: 33072080 PMCID: PMC7531036 DOI: 10.3389/fimmu.2020.02165] [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: 05/21/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Recent genome-wide association studies have identified over 230 genetic risk loci for multiple sclerosis. Current experimental autoimmune encephalomyelitis (EAE) models requiring active induction of disease may not be optimally suited for the characterization of the function of these genes. We have thus used gene expression profiling to study whether spontaneous opticospinal EAE (OSE) or MOG-induced EAE mirrors the genetic contribution to the pathogenesis of multiple sclerosis more faithfully. To this end, we compared gene expression in OSE and MOG EAE models and analyzed the relationship of both models to human multiple sclerosis risk genes and T helper cell biology. We observed stronger gene expression changes and an involvement of more pathways of the adaptive immune system in OSE than MOG EAE. Furthermore, we demonstrated a more extensive enrichment of human MS risk genes among transcripts differentially expressed in OSE than was the case for MOG EAE. Transcripts differentially expressed only in diseased OSE mice but not in MOG EAE were significantly enriched for T helper cell-specific transcripts. These transcripts are part of immune-regulatory pathways. The activation of the adaptive immune system and the enrichment of both human multiple sclerosis risk genes and T helper cell-specific transcripts were also observed in OSE mice showing only mild disease signs. These expression changes may, therefore, be indicative of processes at disease onset. In summary, more human multiple sclerosis risk genes were differentially expressed in OSE than was observed for MOG EAE, especially in TH1 cells. When studying the functional role of multiple sclerosis risk genes and pathways during disease onset and their interactions with the environment, spontaneous OSE may thus show advantages over MOG-induced EAE.
Collapse
Affiliation(s)
- Hans Faber
- Max Planck Institute of Psychiatry, Munich, Germany
| | | | | | - Peter Weber
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Benno Pütz
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Frank Weber
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Till F M Andlauer
- Max Planck Institute of Psychiatry, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
41
|
Esmaeil Amini M, Shomali N, Bakhshi A, Rezaei S, Hemmatzadeh M, Hosseinzadeh R, Eslami S, Babaie F, Aslani S, Torkamandi S, Mohammadi H. Gut microbiome and multiple sclerosis: New insights and perspective. Int Immunopharmacol 2020; 88:107024. [PMID: 33182024 DOI: 10.1016/j.intimp.2020.107024] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
The human gastrointestinal microbiota, also known as the gut microbiota living in the human gastrointestinal tract, has been shown to have a significant impact on several human disorders including rheumatoid arthritis, diabetes, obesity, and multiple sclerosis (MS). MS is an inflammatory disease characterized by the destruction of the spinal cord and nerve cells in the brain due to an attack of immune cells, causing a wide range of harmful symptoms related to inflammation in the central nervous system (CNS). Despite extensive studies on MS that have shown that many external and genetic factors are involved in its pathogenesis, the exact role of external factors in the pathophysiology of MS is still unclear. Recent studies on MS and experimental autoimmune encephalomyelitis (EAE), an animal model of encephalitis, have shown that intestinal microbiota may play a key role in the pathogenesis of MS. Therefore, modification of the intestinal microbiome could be a promising strategy for the future treatment of MS. In this study, the characteristics of intestinal microbiota, the relationship between intestine and brain despite the blood-brain barrier, various factors involved in intestinal microbiota modification, changes in intestinal microbial composition in MS, intestinal microbiome modification strategies, and possible use of intestinal microbiome and factors affecting it have been discussed.
Collapse
Affiliation(s)
- Mohammad Esmaeil Amini
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, Guilan University of Medical Sciences, Rasht, Iran
| | - Navid Shomali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arash Bakhshi
- Student Research Committee, Guilan University of Medical Sciences, Rasht, Iran
| | - Somaye Rezaei
- Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hosseinzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solat Eslami
- Dietary Supplements & Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Farhad Babaie
- Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Saeed Aslani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Torkamandi
- Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
42
|
Mortales CL, Lee SU, Manousadjian A, Hayama KL, Demetriou M. N-Glycan Branching Decouples B Cell Innate and Adaptive Immunity to Control Inflammatory Demyelination. iScience 2020; 23:101380. [PMID: 32745987 PMCID: PMC7398982 DOI: 10.1016/j.isci.2020.101380] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/10/2020] [Accepted: 07/14/2020] [Indexed: 11/21/2022] Open
Abstract
B cell depletion potently reduces episodes of inflammatory demyelination in multiple sclerosis (MS), predominantly through loss of innate rather than adaptive immunity. However, molecular mechanisms controlling innate versus adaptive B cell function are poorly understood. N-glycan branching, via interactions with galectins, controls endocytosis and signaling of cell surface receptors to control cell function. Here we report that N-glycan branching in B cells dose dependently reduces pro-inflammatory innate responses by titrating decreases in Toll-like receptor-4 (TLR4) and TLR2 surface expression via endocytosis. In contrast, a minimal level of N-glycan branching maximizes surface retention of the B cell receptor (BCR) and the CD19 co-receptor to promote adaptive immunity. Branched N-glycans inhibit antigen presentation by B cells to reduce T helper cell-17 (TH17)/TH1 differentiation and inflammatory demyelination in mice. Thus, N-glycan branching negatively regulates B cell innate function while promoting/maintaining adaptive immunity via BCR, providing an attractive therapeutic target for MS.
Collapse
Affiliation(s)
- Christie-Lynn Mortales
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92617, USA
| | - Sung-Uk Lee
- Department of Neurology, University of California, Irvine, CA 92617, USA
| | - Armen Manousadjian
- Department of Neurology, University of California, Irvine, CA 92617, USA
| | - Ken L Hayama
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92617, USA
| | - Michael Demetriou
- Department of Neurology, University of California, Irvine, CA 92617, USA; Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92617, USA.
| |
Collapse
|
43
|
Altered Regulatory B Cell Subsets in Children with Type 1 Diabetes Mellitus. J Immunol Res 2020; 2020:8935694. [PMID: 32775471 PMCID: PMC7391103 DOI: 10.1155/2020/8935694] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/24/2020] [Accepted: 07/04/2020] [Indexed: 12/12/2022] Open
Abstract
B regulatory cells (Breg) refer to characteristic subsets of B cells that generally exert anti-inflammatory functions and maintain peripheral tolerance mainly through their ability to secrete interleukin-10 (IL10). Dysregulation in the function of Breg cells was reported in several autoimmune diseases. However, the relation between Breg and children with type 1 diabetes (T1D) is poorly understood. Thus, this study is aimed at determining whether Breg cells play a role in T1D in children or not, so we hypothesized that an altered phenotype of B cell subsets is associated with T1D in children. Children with T1D (n = 29) and control children with normal blood glucose levels (n = 14) were recruited. The percentages of different circulating IL10-producing Breg subsets, including B10, immature transitional, and plasmablasts were determined using flow cytometry analysis. Furthermore, the association between different IL10-producing B cells and patient parameters was investigated. The percentage of circulating IL10+CD24hiCD27+ (B10) and IL10+CD24hiCD38hi (immature transitional) subsets of Breg cells was significantly lower in T1D patients than in healthy controls. Moreover, these cells were also negatively correlated with fasting blood glucose and HbA1c levels. Breg cells did not correlate with autoantibody levels in the serum. These findings suggest that certain Breg subsets are numerically deficient in children with T1D. This alteration in frequency is associated with deficient islet function and glycemia. These findings suggest that Breg cells may be involved in the loss of auto-tolerance and consequent destruction of pancreatic cells and could, therefore, be a potential target for immunotherapy.
Collapse
|
44
|
Rotating magnetic field ameliorates experimental autoimmune encephalomyelitis by promoting T cell peripheral accumulation and regulating the balance of Treg and Th1/Th17. Aging (Albany NY) 2020; 12:6225-6239. [PMID: 32265343 PMCID: PMC7185125 DOI: 10.18632/aging.103018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/03/2020] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by T cell infiltration and demyelination of the central nervous system (CNS). Experimental autoimmune encephalomyelitis (EAE) is a classical preclinical animal model of MS. In this study, we found that rotating magnetic field (RMF) treatment exerts potential preventive effects on the discovery of EAE, including reducing the severity of the disease and delaying the onset of the disease. The results indicated that RMF (0.2 T, 4 Hz) treatment increases the accumulation of CD4+ cells in the spleen and lymph nodes by downregulating the expression of CCL-2, CCL-3 and CCL-5, but has no significant effect on myelin oligodendrocyte glycoprotein (MOG) specific T cell responses. Simultaneously, RMF treatment adjusted the imbalance between regulatory T (Treg) cell and T helper 1 (Th1) cells or T helper 17 (Th17) cells by increasing the proportion of Treg cells and inhibiting the ratio of Th1 and Th17 cell subsets. These findings suggest that exposure to RMF may improve EAE disease by promoting CD4+ cell accumulation into peripheral lymphoid tissue, improving the imbalance between Treg and Th1/Th17 cells. Therefore, as a mild physical therapy approach, RMF, is likely to be a potential way to alter the development of EAE.
Collapse
|
45
|
Wang Y, Qin Y, Wang X, Zhang L, Wang J, Xu X, Chen H, Hsu H, Zhang M. Decrease in the proportion of CD24 hi CD38 hi B cells and impairment of their regulatory capacity in type 1 diabetes patients. Clin Exp Immunol 2020; 200:22-32. [PMID: 31849037 PMCID: PMC7066388 DOI: 10.1111/cei.13408] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2019] [Indexed: 01/11/2023] Open
Abstract
B10 cells restore immune balance by producing interleukin (IL)-10. Impaired B10 cell responses are related to numerous autoimmune diseases. However, the function of B10 cells in type 1 diabetes (T1D) patients is controversial. We hypothesized that there are numerical and functional defects of B10 cells in T1D. Sixty-two patients with T1D and 74 healthy volunteers were included in our study. We showed that B10 cells in human peripheral blood belong to a CD24hi CD38hi B cell subpopulation. CD24hi CD38hi B cells from healthy individuals possessed regulatory capacity, suppressed interferon (IFN)-γ, tumor necrosis factor (TNF)-α and IL-17A production and promoted IL-4 production and forkhead box protein 3 (FoxP3) expression in CD4+ T cells through an IL-10-dependent mechanism. Compared to healthy controls, B10 cell percentages in T1D were significantly lower (5·6 ± 3·5 versus 6·9 ± 3·3%; P < 0·05), produced less IL-10 (15·4 ± 4·3 versus 29·0 ± 4·5%; P < 0·001) and lacked regulatory capacity. In addition, Pearson's correlation analysis showed that the frequency of circulating B10 cells was negatively correlated with the frequency of CD4+ IFN-γ+ and CD4+ TNF-α+ T cells (r = -0·248 and r = -0·283, P = 0·008 and P = 0·017, respectively), positively correlating with the frequency of CD4+ CD25+ FoxP3+ T cells (r = 0·247, P = 0·001). These data offer direct proof that there is a deficiency of circulating CD24hi CD38hi B cells in peripheral blood of patients with T1D, which participate in the T1D immune imbalance involved in the development of T1D.
Collapse
Affiliation(s)
- Y. Wang
- Department of EndocrinologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Department of PediatricsThe First Affiliated Hospital with Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - Y. Qin
- Department of EndocrinologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - X. Wang
- Department of EndocrinologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - L. Zhang
- Department of EndocrinologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - J. Wang
- Department of EndocrinologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - X. Xu
- Department of EndocrinologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - H. Chen
- Department of EndocrinologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - H.‐T. Hsu
- Department of EndocrinologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - M. Zhang
- Department of EndocrinologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
46
|
Torre-Fuentes L, Moreno-Jiménez L, Pytel V, Matías-Guiu J, Gómez-Pinedo U, Matías-Guiu J. Experimental models of demyelination and remyelination. NEUROLOGÍA (ENGLISH EDITION) 2020. [PMCID: PMC7148713 DOI: 10.1016/j.nrleng.2019.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
47
|
Nataf S, Guillen M, Pays L. Common Neurodegeneration-Associated Proteins Are Physiologically Expressed by Human B Lymphocytes and Are Interconnected via the Inflammation/Autophagy-Related Proteins TRAF6 and SQSTM1. Front Immunol 2019; 10:2704. [PMID: 31824497 PMCID: PMC6886494 DOI: 10.3389/fimmu.2019.02704] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
There is circumstantial evidence that, under neurodegenerative conditions, peptides deriving from aggregated or misfolded specific proteins elicit adaptive immune responses. On another hand, several genes involved in familial forms of neurodegenerative diseases exert key innate immune functions. However, whether or not such observations are causally linked remains unknown. To start addressing this issue, we followed a systems biology strategy based on the mining of large proteomics and immunopeptidomics databases. First, we retrieved the expression patterns of common neurodegeneration-associated proteins in two professional antigen-presenting cells, namely B lymphocytes and dendritic cells. Surprisingly, we found that under physiological conditions, numerous neurodegeneration-associated proteins are abundantly expressed by human B lymphocytes. A survey of the human proteome allowed us to map a unique protein-protein interaction network linking common neurodegeneration-associated proteins and their first shell interactors in human B lymphocytes. Interestingly, network connectivity analysis identified two major hubs that both relate with inflammation and autophagy, namely TRAF6 (TNF Receptor Associated Factor 6) and SQSTM1 (Sequestosome-1). Moreover, the mapped network in B lymphocytes comprised two additional hub proteins involved in both inflammation and autoimmunity: HSPA8 (Heat Shock Protein Family A Member 8 also known as HSC70) and HSP90AA1 (Heat Shock Protein 90 Alpha Family Class A Member 1). Based on these results, we then explored the Immune Epitope Database "IEDB-AR" and actually found that a large share of neurodegeneration-associated proteins were previously reported to provide endogenous MHC class II-binding peptides in human B lymphocytes. Of note, peptides deriving from amyloid beta A4 protein, sequestosome-1 or profilin-1 were reported to bind multiple allele-specific MHC class II molecules. In contrast, peptides deriving from microtubule-associated protein tau, presenilin 2 and serine/threonine-protein kinase TBK1 were exclusively reported to bind MHC molecules encoded by the HLA-DRB1 1501 allele, a recently-identified susceptibility gene for late onset Alzheimer's disease. Finally, we observed that the whole list of proteins reported to provide endogenous MHC class II-binding peptides in human B lymphocytes is specifically enriched in neurodegeneration-associated proteins. Overall, our work indicates that immunization against neurodegeneration-associated proteins might be a physiological process which is shaped, at least in part, by B lymphocytes.
Collapse
Affiliation(s)
- Serge Nataf
- CarMeN Laboratory, INSERM U1060, INRA U1397, INSA de Lyon, Lyon-Sud Faculty of Medicine, University of Lyon, Pierre-Bénite, France
- Faculté de Médecine Lyon-Est, University of Lyon 1, Lyon, France
- Banque de Tissus et de Cellules des Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Marine Guillen
- Faculté de Médecine Lyon-Est, University of Lyon 1, Lyon, France
| | - Laurent Pays
- CarMeN Laboratory, INSERM U1060, INRA U1397, INSA de Lyon, Lyon-Sud Faculty of Medicine, University of Lyon, Pierre-Bénite, France
- Faculté de Médecine Lyon-Est, University of Lyon 1, Lyon, France
- Banque de Tissus et de Cellules des Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| |
Collapse
|
48
|
Alves da Costa T, Lang J, Torres RM, Pelanda R. The development of human immune system mice and their use to study tolerance and autoimmunity. J Transl Autoimmun 2019; 2:100021. [PMID: 32743507 PMCID: PMC7388352 DOI: 10.1016/j.jtauto.2019.100021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 11/04/2019] [Indexed: 12/26/2022] Open
Abstract
Autoimmune diseases evolve from complex interactions between the immune system and self-antigens and involve several genetic attributes, environmental triggers and diverse cell types. Research using experimental mouse models has contributed key knowledge on the mechanisms that underlie these diseases in humans, but differences between the mouse and human immune systems can and, at times, do undermine the translational significance of these findings. The use of human immune system (HIS) mice enables the utility of mouse models with greater relevance for human diseases. As the name conveys, these mice are reconstituted with mature human immune cells transferred directly from peripheral blood or via transplantation of human hematopoietic stem cells that nucleate the generation of a complex human immune system. The function of the human immune system in HIS mice has improved over the years with the stepwise development of better models. HIS mice exhibit key benefits of the murine animal model, such as small size, robust and rapid reproduction and ease of experimental manipulation. Importantly, HIS mice also provide an applicable in vivo setting that permit the investigation of the physiological and pathological functions of the human immune system and its response to novel treatments. With the gaining popularity of HIS mice in the last decade, the potential of this model has been exploited for research in basic science, infectious diseases, cancer, and autoimmunity. In this review we focus on the use of HIS mice in autoimmune studies to stimulate further development of these valuable models. Human immune system (HIS) mice bear components of the human immune system. HIS mice engraft with human blood or hematopoietic stem cells, and sometimes thymus. HIS mice are used to investigate development and function of the human immune system. Immunological tolerance and autoimmune responses can be studied in HIS mice. HIS models of autoimmunity vary in complexity and in ability to represent disease.
Collapse
Affiliation(s)
- Thiago Alves da Costa
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Julie Lang
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Raul M. Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Biomedical Research, National Jewish Health, Denver, CO, 80206, USA
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Biomedical Research, National Jewish Health, Denver, CO, 80206, USA
- Corresponding author. University of Colorado School of Medicine, 12800 East 19th Avenue Mail Stop 8333, Aurora, CO, 80045-2508, USA.
| |
Collapse
|
49
|
Boshans LL, Sherafat A, Nishiyama A. The effects of developmental and current niches on oligodendrocyte precursor dynamics and fate. Neurosci Lett 2019; 715:134593. [PMID: 31678373 DOI: 10.1016/j.neulet.2019.134593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/29/2022]
Abstract
Oligodendrocyte precursor cells (OPCs), whose primary function is to generate myelinating oligodendrocytes, are distributed widely throughout the developing and mature central nervous system. They originate from several defined subdomains in the embryonic germinal zones at different developmental stages and in the adult. While many phenotypic differences have been observed among OPCs in different anatomical regions and among those arising from different germinal zones, we know relatively little about the molecular and cellular mechanisms by which the historical and current niches shape the behavior of oligodendrocyte lineage cells. This minireview will discuss how the behavior of oligodendrocyte lineage cells is influenced by the developmental niches from which subpopulations of OPCs emerge, by the current niches surrounding OPCs in different regions, and in pathological states that cause deviations from the normal density of oligodendrocyte lineage cells and myelin.
Collapse
Affiliation(s)
- Linda L Boshans
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, 06269, USA
| | - Amin Sherafat
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, 06269, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, 06269, USA; Institute for Systems Genomics, University of Connecticut, USA; Institute for Brain and Cognitive Science, University of Connecticut, USA.
| |
Collapse
|
50
|
Seifert HA, Gerstner G, Kent G, Vandenbark AA, Offner H. Estrogen-induced compensatory mechanisms protect IL-10-deficient mice from developing EAE. J Neuroinflammation 2019; 16:195. [PMID: 31665042 PMCID: PMC6821034 DOI: 10.1186/s12974-019-1588-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/12/2019] [Indexed: 02/08/2023] Open
Abstract
Background IL-10 knockout (KO) mice are protected from experimental autoimmune encephalomyelitis (EAE) with low-dose estrogen (E2) treatment similar to wild-type (WT) mice. Previous studies have demonstrated a decrease in tumor necrosis factor in all E2-treated groups, which led to the protection of the mice. Methods This study used IL-10 KO mice and WT mice treated either with E2 or sham pellets 7 days prior to induction of EAE. Mice were observed for 21 days post-immunization. The spleen, inguinal lymph nodes, and brain were evaluated by flow cytometry. Spinal cords were evaluated using a cytokine/chemokine array, RT-PCR, and histology. Results This study demonstrates that E2 treatment induced three heightened regulatory mechanisms that potentially protect IL-10 KO mice from EAE: (1) an increase in programmed death-ligands 1 and 2 on monocytes and macrophages in the periphery and within the CNS; (2) an increase in CD73 in the inflamed CNS, which can increase the production of the anti-inflammatory molecule adenosine; and (3) a decrease in CD4+CD25+FoxP3+ regulatory T cells in the spleen. Together, these factors comprise an alternative compensatory mechanism that significantly downregulates key pro-inflammatory cytokine, chemokine, and chemokine receptor genes which are enhanced in the spinal cord of IL-10 KO mice. This group of E2-treated mice remained asymptomatic after EAE challenge similar to E2-treated WT mice, despite their having more T and B lymphocytes in the brain, and modestly increased demyelination in the spinal cord. Conclusion These results indicate that previously unrecognized compensatory mechanisms of EAE protection are stimulated by E2 in the absence of IL-10, which can provide disease protection comparable to the IL-10-dependent effects induced by E2 in WT mice.
Collapse
Affiliation(s)
- Hilary A Seifert
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Neuroimmunology Research, VA Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., Portland, OR, 97239, USA
| | - Grant Gerstner
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Neuroimmunology Research, VA Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., Portland, OR, 97239, USA
| | - Gail Kent
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Neuroimmunology Research, VA Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Neuroimmunology Research, VA Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., Portland, OR, 97239, USA.,Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Halina Offner
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA. .,Neuroimmunology Research, VA Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., Portland, OR, 97239, USA. .,Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|