1
|
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
|
2
|
Gao X, Tang Y, Kong L, Fan Y, Wang C, Wang R. Treg cell: Critical role of regulatory T-cells in depression. Pharmacol Res 2023; 195:106893. [PMID: 37611836 DOI: 10.1016/j.phrs.2023.106893] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/28/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023]
Abstract
Depression is a highly prevalent disorder of the central nervous system. The neuropsychiatric symptoms of clinical depression are persistent and include fatigue, anorexia, weight loss, altered sleep patterns, hyperalgesia, melancholia, anxiety, and impaired social behaviours. Mounting evidences suggest that neuroinflammation triggers dysregulated cellular immunity and increases susceptibility to psychiatric diseases. Neuroimmune responses have transformed the clinical approach to depression because of their roles in its pathophysiology and their therapeutic potential. In particular, activated regulatory T (Treg) cells play an increasingly evident role in the inflammatory immune response. In this review, we summarized the available data and discussed in depth the fundamental roles of Tregs in the pathogenesis of depression, as well as the clinical therapeutic potential of Tregs. We aimed to provide recent information regarding the potential of Tregs as immune-modulating biologics for the treatment and prevention of long-term neuropsychiatric symptoms of depression.
Collapse
Affiliation(s)
- Xiao Gao
- Department of Geriatrics, Qingdao Mental Health Center, 26600 Qingdao, Shandong Province, China
| | - Yuru Tang
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, 26600 Qingdao, Shandong Province, China
| | - Lingli Kong
- Department of Geriatrics, Qingdao Mental Health Center, 26600 Qingdao, Shandong Province, China
| | - Yong Fan
- Department of Geriatrics, Qingdao Mental Health Center, 26600 Qingdao, Shandong Province, China
| | - Chunxia Wang
- Department of Geriatrics, Qingdao Mental Health Center, 26600 Qingdao, Shandong Province, China.
| | - Rui Wang
- Department of Pain Management, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), 26600 Qingdao, Shandong Province, China.
| |
Collapse
|
3
|
Xiong H, Tang F, Guo Y, Xu R, Lei P. Neural Circuit Changes in Neurological Disorders: Evidence from in vivo Two-photon Imaging. Ageing Res Rev 2023; 87:101933. [PMID: 37061201 DOI: 10.1016/j.arr.2023.101933] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
Neural circuits, such as synaptic plasticity and neural activity, are critical components of healthy brain function. The consequent dynamic remodeling of neural circuits is an ongoing procedure affecting neuronal activities. Disruption of this essential process results in diseases. Advanced microscopic applications such as two-photon laser scanning microscopy have recently been applied to understand neural circuit changes during disease since it can visualize fine structural and functional cellular activation in living animals. In this review, we have summarized the latest work assessing the dynamic rewiring of postsynaptic dendritic spines and modulation of calcium transients in neurons of the intact living brain, focusing on their potential roles in neurological disorders (e.g. Alzheimer's disease, stroke, and epilepsy). Understanding the fine changes that occurred in the brain during disease is crucial for future clinical intervention developments.
Collapse
Affiliation(s)
- Huan Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China; Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Fei Tang
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Yujie Guo
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China.
| |
Collapse
|
4
|
Lee JI, Choi JH, Kwon TW, Jo HS, Kim DG, Ko SG, Song GJ, Cho IH. Neuroprotective effects of bornyl acetate on experimental autoimmune encephalomyelitis via anti-inflammatory effects and maintaining blood-brain-barrier integrity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154569. [PMID: 36842217 DOI: 10.1016/j.phymed.2022.154569] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 09/05/2022] [Accepted: 11/19/2022] [Indexed: 06/18/2023]
Abstract
BACKGROUND Bornyl acetate (BA), a chemical component of essential oil in the Pinus family, has yet to be actively studies in terms of its therapeutic effect on numerous diseases, including autoimmune diseases. PURPOSE This study aimed to investigate the pharmacological effects and molecular mechanisms of BA on myelin oligodendrocyte glycoprotein (MOG35-55)-induced experimental autoimmune encephalomyelitis (EAE) mice in an animal model of multiple sclerosis (MS), a representative autoimmune disease in central nervous system. METHODS BA (100, 200, or 400 mg/kg) was orally treated to EAE mice once daily for 30 days after immunization for the behavioral test and for the 16th-18th days for the histopathological and molecular analyses, from the onset stage (8th day) of EAE symptoms. RESULTS BA mitigated behavioral dysfunction (motor disability) and demyelination in the spinal cord that were associated with the down-regulation of representative pro-inflammatory cytokines (interleukin (IL)-1 beta, IL-6, and tumor necrosis factor-alpha), enzymes (cyclooxygenase-2 and inducible nitric oxide synthase), and chemokines (monocyte chemotactic protein-1, macrophage inflammatory protein-1 alpha, and regulated on activation), and decreased infiltration of microglia (CD11b+/CD45+(low)) and macrophages (CD11b+/CD45+(high)). The anti-inflammatory effect of BA was related to the inhibition of mitogen-activated protein kinases and nuclear factor-kappa B pathways. BA also reduced the recruitment/infiltration rates of CD4+ T, Th1, and Th17 cells into the spinal cords of EAE mice, which was related to reduced blood-spinal cord barrier (BSCB) disruption. CONCLUSION These findings strongly suggest that BA may alleviate EAE due to its anti-inflammatory and BSCB protective activities. This indicates that BA is a potential therapeutic agent for treating autoimmune demyelinating diseases including MS.
Collapse
Affiliation(s)
- Joon-Il Lee
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jong-Hee Choi
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Tae-Woo Kwon
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyo-Sung Jo
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Do-Geun Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu 41068, Republic of Korea
| | - Seong-Gyu Ko
- Korean Medicine-based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Gyun Jee Song
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Gangwon-do 25601, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Institute of Convergence Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
5
|
Gulbins A, Görtz GE, Gulbins E, Eckstein A. Sphingolipids in thyroid eye disease. Front Endocrinol (Lausanne) 2023; 14:1170884. [PMID: 37082124 PMCID: PMC10112667 DOI: 10.3389/fendo.2023.1170884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/22/2023] [Indexed: 04/22/2023] Open
Abstract
Graves' disease (GD) is caused by an autoimmune formation of autoantibodies and autoreactive T-cells against the thyroid stimulating hormone receptor (TSHR). The autoimmune reaction does not only lead to overstimulation of the thyroid gland, but very often also to an immune reaction against antigens within the orbital tissue leading to thyroid eye disease, which is characterized by activation of orbital fibroblasts, orbital generation of adipocytes and myofibroblasts and increased hyaluronan production in the orbit. Thyroid eye disease is the most common extra-thyroidal manifestation of the autoimmune Graves' disease. Several studies indicate an important role of sphingolipids, in particular the acid sphingomyelinase/ceramide system and sphingosine 1-phosphate in thyroid eye disease. Here, we discuss how the biophysical properties of sphingolipids contribute to cell signaling, in particular in the context of thyroid eye disease. We further review the role of the acid sphingomyelinase/ceramide system in autoimmune diseases and its function in T lymphocytes to provide some novel hypotheses for the pathogenesis of thyroid eye disease and potentially allowing the development of novel treatments.
Collapse
Affiliation(s)
- Anne Gulbins
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Gina-Eva Görtz
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Erich Gulbins
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- *Correspondence: Anja Eckstein, ; Erich Gulbins,
| | - Anja Eckstein
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- *Correspondence: Anja Eckstein, ; Erich Gulbins,
| |
Collapse
|
6
|
GATA1 controls numbers of hematopoietic progenitors and their response to autoimmune neuroinflammation. Blood Adv 2022; 6:5980-5994. [PMID: 36206195 PMCID: PMC9691916 DOI: 10.1182/bloodadvances.2022008234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/13/2022] [Indexed: 12/14/2022] Open
Abstract
GATA-binding factor 1 (GATA1) is a transcription factor that governs the development and function of multiple hematopoietic cell lineages. GATA1 is expressed in hematopoietic stem and progenitor cells (HSPCs) and is essential for erythroid lineage commitment; however, whether it plays a role in hematopoietic stem cell (HSC) biology and the development of myeloid cells, and what that role might be, remains unclear. We initially set out to test the role of eosinophils in experimental autoimmune encephalomyelitis (EAE), a model of central nervous system autoimmunity, using mice lacking a double GATA-site (ΔdblGATA), which lacks eosinophils due to the deletion of the dblGATA enhancer to Gata1, which alters its expression. ΔdblGATA mice were resistant to EAE, but not because of a lack of eosinophils, suggesting that these mice have an additional defect. ΔdblGATA mice with EAE had fewer inflammatory myeloid cells than the control mice, suggesting that resistance to EAE is caused by a defect in myeloid cells. Naïve ΔdblGATA mice also showed reduced frequency of CD11b+ myeloid cells in the blood, indicating a defect in myeloid cell production. Examination of HSPCs revealed fewer HSCs and myeloid cell progenitors in the ΔdblGATA bone marrow (BM), and competitive BM chimera experiments showed a reduced capacity of the ΔdblGATA BM to reconstitute immune cells, suggesting that reduced numbers of ΔdblGATA HSPCs cause a functional deficit during inflammation. Taken together, our data show that GATA1 regulates the number of HSPCs and that reduced GATA1 expression due to dblGATA deletion results in a diminished immune response following the inflammatory challenge.
Collapse
|
7
|
Artemisinin derivative TPN10466 suppresses immune cell migration and Th1/Th17 differentiation to ameliorate disease severity in experimental autoimmune encephalomyelitis. Cell Immunol 2022; 373:104500. [DOI: 10.1016/j.cellimm.2022.104500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/21/2022] [Accepted: 03/02/2022] [Indexed: 11/20/2022]
|
8
|
Ahsan N, Santoro JD. Immunopathogenesis of acute disseminated encephalomyelitis. TRANSLATIONAL AUTOIMMUNITY 2022:249-263. [DOI: 10.1016/b978-0-12-824466-1.00003-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Zeng-Brouwers J, Pandey S, Trebicka J, Wygrecka M, Schaefer L. Communications via the Small Leucine-rich Proteoglycans: Molecular Specificity in Inflammation and Autoimmune Diseases. J Histochem Cytochem 2020; 68:887-906. [PMID: 32623933 PMCID: PMC7708667 DOI: 10.1369/0022155420930303] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a highly regulated biological response of the immune system that is triggered by assaulting pathogens or endogenous alarmins. It is now well established that some soluble extracellular matrix constituents, such as small leucine-rich proteoglycans (SLRPs), can act as danger signals and trigger aseptic inflammation by interacting with innate immune receptors. SLRP inflammatory signaling cascade goes far beyond its canonical function. By choosing specific innate immune receptors, coreceptors, and adaptor molecules, SLRPs promote a switch between pro- and anti-inflammatory signaling, thereby determining disease resolution or chronification. Moreover, by orchestrating signaling through various receptors, SLRPs fine-tune inflammation and, despite their structural homology, regulate inflammatory processes in a molecule-specific manner. Hence, the overarching theme of this review is to highlight the molecular and functional specificity of biglycan-, decorin-, lumican-, and fibromodulin-mediated signaling in inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Jinyang Zeng-Brouwers
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Sony Pandey
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Jonel Trebicka
- Translational Hepatology, Department of Internal Medicine I, University Clinic Frankfurt, Frankfurt, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
- German Center for Lung Research, Giessen, Germany
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
10
|
Yahn SL, Li J, Goo I, Gao H, Brambilla R, Lee JK. Fibrotic scar after experimental autoimmune encephalomyelitis inhibits oligodendrocyte differentiation. Neurobiol Dis 2019; 134:104674. [PMID: 31731043 PMCID: PMC7547849 DOI: 10.1016/j.nbd.2019.104674] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/25/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Remyelination failure is a crucial component of disease progression in the autoimmune demyelinating disease Multiple Sclerosis (MS). The regenerative capacity of oligodendrocyte progenitor cells (OPCs) to replace myelinating oligodendrocytes is likely influenced by many aspects of the lesion environment including inflammatory signaling and extracellular matrix (ECM) deposition. These features of MS lesions are typically attributed to infiltrating leukocytes and reactive astrocytes. Here we demonstrate that fibroblasts also contribute to the inhibitory environment in the animal model of MS, experimental autoimmune encephalomyelitis (EAE). Using Col1α1GFP transgenic mice, we show that perivascular fibroblasts are activated in the spinal cord at EAE onset, and infiltrate the parenchyma by the peak of behavioral deficits where they are closely associated with areas of demyelination, myeloid cell accumulation, and ECM deposition. We further show that both fibroblast conditioned media and fibroblast ECM inhibit the differentiation of OPCs into mature oligodendrocytes. Taken together, our results indicate that the fibrotic scar is a major component of EAE pathology that leads to an inhibitory environment for remyelination, thus raising the possibility that anti-fibrotic mechanisms may serve as novel therapeutic targets for MS.
Collapse
Affiliation(s)
- Stephanie L Yahn
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL 33136, United States of America
| | - Jiajun Li
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL 33136, United States of America
| | - Irene Goo
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL 33136, United States of America
| | - Han Gao
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL 33136, United States of America
| | - Roberta Brambilla
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL 33136, United States of America
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL 33136, United States of America.
| |
Collapse
|
11
|
Qu X, Han J, Zhang Y, Wang X, Fan H, Hua F, Yao R. TLR4-RelA-miR-30a signal pathway regulates Th17 differentiation during experimental autoimmune encephalomyelitis development. J Neuroinflammation 2019; 16:183. [PMID: 31561751 PMCID: PMC6764145 DOI: 10.1186/s12974-019-1579-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/10/2019] [Indexed: 12/13/2022] Open
Abstract
Background Toll-like receptor 4 (TLR4) is well known for activating the innate immune system; however, it is also highly expressed in adaptive immune cells, such as CD4+ T-helper 17 (Th17) cells, which play a key role in multiple sclerosis (MS) pathology. However, the function and governing mechanism of TLR4 in Th17 remain unclear. Methods The changes of TLR4 in CD4+ T cells from MS patients and experimental autoimmune encephalomyelitis (EAE) mice were tested. TLR4-deficient (TLR4−/−) naïve T cells were induced in vitro and transferred into Rag1−/− mice to measure Th17 differentiation and EAE pathology. DNA sequence analyses combining with deletion fragments and mutation analyses, chromatin immunoprecipitation (ChIP), and electrophoretic mobility shift assay (EMSA) were used to explore the mechanism of TLR4 signaling pathway in regulating Th17 differentiation. Results The levels of TLR4 were increased in CD4+ Th17 cells both from MS patients and EAE mice, as well as during Th17 differentiation in vitro. TLR4−/− CD4+ naïve T cells inhibited their differentiation into Th17, and transfer of TLR4−/− CD4+ naïve T cells into Rag1−/− mice was defective in promoting EAE, characterized by less demyelination and Th17 infiltration in the spinal cord. TLR4 signal enhanced Th17 differentiation by activating RelA, downregulating the expression of miR-30a, a negative regulator of Th17 differentiation. Inhibition of RelA activity increased miR-30a level, but decreased Th17 differentiation rate. Furthermore, RelA directly regulated the expression of miR-30a via specific binding to a conserved element of miR-30a gene. Conclusions TLR4−/− CD4+ naïve T cells are inadequate in differentiating to Th17 cells both in vitro and in vivo. TLR4-RelA-miR-30a signal pathway regulates Th17 differentiation via direct binding of RelA to the regulatory element of miR-30a gene. Our results indicate modulating TLR4-RelA-miR-30a signal in Th17 may be a therapeutic target for Th17-mediated neurodegeneration in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Xuebin Qu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, People's Republic of China.
| | - Jingjing Han
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Ying Zhang
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, People's Republic of China
| | - Xingqi Wang
- School of Life Science, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, China
| | - Hongbin Fan
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Fang Hua
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.,Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, People's Republic of China.
| |
Collapse
|
12
|
Role of Immunological Memory Cells as a Therapeutic Target in Multiple Sclerosis. Brain Sci 2017; 7:brainsci7110148. [PMID: 29112130 PMCID: PMC5704155 DOI: 10.3390/brainsci7110148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 12/14/2022] Open
Abstract
Pharmacological targeting of memory cells is an attractive treatment strategy in various autoimmune diseases, such as psoriasis and rheumatoid arthritis. Multiple sclerosis is the most common inflammatory disorder of the central nervous system, characterized by focal immune cell infiltration, activation of microglia and astrocytes, along with progressive damage to myelin sheaths, axons, and neurons. The current review begins with the identification of memory cell types in the previous literature and a recent description of the modulation of these cell types in T, B, and resident memory cells in the presence of different clinically approved multiple sclerosis drugs. Overall, this review paper tries to determine the potential of memory cells to act as a target for the current or newly-developed drugs.
Collapse
|
13
|
Ocular antigen does not cause disease unless presented in the context of inflammation. Sci Rep 2017; 7:14226. [PMID: 29079770 PMCID: PMC5660195 DOI: 10.1038/s41598-017-14618-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/11/2017] [Indexed: 12/14/2022] Open
Abstract
Ocular antigens are sequestered behind the blood-retina barrier and the ocular environment protects ocular tissues from autoimmune attack. The signals required to activate autoreactive T cells and allow them to cause disease in the eye remain in part unclear. In particular, the consequences of peripheral presentation of ocular antigens are not fully understood. We examined peripheral expression and presentation of ocular neo-self-antigen in transgenic mice expressing hen egg lysozyme (HEL) under a retina-specific promoter. High levels of HEL were expressed in the eye compared to low expression throughout the lymphoid system. Adoptively transferred naïve HEL-specific CD4+ T cells proliferated in the eye draining lymph nodes, but did not induce uveitis. By contrast, systemic infection with a murine cytomegalovirus (MCMV) engineered to express HEL induced extensive proliferation of transferred naïve CD4+ T cells, and significant uveoretinitis. In this model, wild-type MCMV, lacking HEL, did not induce overt uveitis, suggesting that disease is mediated by antigen-specific peripherally activated CD4+ T cells that infiltrate the retina. Our results demonstrate that retinal antigen is presented to T cells in the periphery under physiological conditions. However, when the same antigen is presented during viral infection, antigen-specific T cells access the retina and autoimmune uveitis ensues.
Collapse
|
14
|
Caveolin-1 Is Critical for Lymphocyte Trafficking into Central Nervous System during Experimental Autoimmune Encephalomyelitis. J Neurosci 2017; 36:5193-9. [PMID: 27170118 DOI: 10.1523/jneurosci.3734-15.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 04/07/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Multiple sclerosis (MS) is a progressive autoimmune disease of the CNS with its underlying mechanisms not fully understood. In the present study, we tested the hypothesis that caveolin-1, a major membrane scaffolding protein, plays a critical role in the pathogenesis of experimental autoimmune encephalomyelitis, a laboratory murine model of MS. We found increased expression of caveolin-1 in serum and spinal cord tissues in association with disease incidence and severity in wild-type mice with active encephalomyelitis. After immunization, Cav-1 knock-out mice showed remarkable disease resistance with decreased incidence and clinical symptoms. Furthermore, Cav-1 knock-out mice had alleviated encephalitogenic T cells trafficking into the CNS with decreased expressions of adhesion molecules ICAM-1 and VCAM-1 within the lesions. In agreement with in vivo studies, in vitro knockdown of caveolin-1 compromised the upregulation of ICAM-1 in endothelial cells, leading to the amelioration of the transendothelial migration of pathogenic TH1 and TH17 cells. Together, those results indicate that caveolin-1 serves as an active modulator of CNS-directed lymphocyte trafficking and could be a therapeutic target for neuroinflammatory diseases, such as multiple sclerosis. SIGNIFICANCE STATEMENT The hallmark feature of neuroinflammatory diseases is the massive infiltrations of encephalitogenic leukocytes into the CNS parenchyma, a process that remains largely unclear. Our study demonstrates the critical contribution of caveolin-1 to encephalomyelitis pathogenesis and CNS-directed lymphocyte trafficking by modulation of adhesion molecules ICAM-1 and VCAM-1, highlighting the pathological involvement of caveolin-1 in neuroinflammatory diseases.
Collapse
|
15
|
Lee MJ, Choi JH, Lee SJ, Cho IH. Oriental Medicine Samhwangsasim-tang Alleviates Experimental Autoimmune Encephalomyelitis by Suppressing Th1 Cell Responses and Upregulating Treg Cell Responses. Front Pharmacol 2017; 8:192. [PMID: 28458638 PMCID: PMC5394181 DOI: 10.3389/fphar.2017.00192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/24/2017] [Indexed: 01/09/2023] Open
Abstract
Oriental medicine Samhwangsasim-tang (SHSST) has traditionally been used in East Asia to treat hypertension and its complications. However, little is known about its potential value regarding the treatment of chronic inflammatory diseases such as multiple sclerosis (MS). In this study, we investigated whether SHSST has a beneficial effect in treating myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (EAE). Onset-treatment with SHSST was found to alleviate neurological symptoms as well as demyelination and glial activation in the spinal cords from the EAE mice. The SHSST also attenuated the mRNA or protein expression of pro-inflammatory cytokines (interleukin-1beta and tumor necrotic factor-alpha); chemokines (RANTES, monocyte chemotactic protein-1, and macrophage inflammatory protein-1alpha); inducible nitric oxide synthase; and cyclooxygenase-2 in correspondence with the down-regulation of the nuclear factor-kappa B and mitogen-activated protein kinases signal pathways in the spinal cords from EAE mice. Interestingly, the protective effect of the SHSST was related to a decreased number of Th1 cells and an increased number of Treg cells in spinal cords from EAE mice. Taken together, our finding firstly suggested that SHSST could delay or mitigate EAE with a wide therapeutic time-window by suppressing Th1 cell responses and upregulating Treg cell responses. Also, our findings are strong enough to warrant further investigation of SHSST as a treatment for chronic autoimmune diseases including MS.
Collapse
Affiliation(s)
- Min J Lee
- Department of Science in Korean Medicine and Brain Korea 21 Plus Program, Graduate School, Kyung Hee UniversitySeoul, South Korea.,Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee UniversitySeoul, South Korea
| | - Jong H Choi
- Department of Science in Korean Medicine and Brain Korea 21 Plus Program, Graduate School, Kyung Hee UniversitySeoul, South Korea.,Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee UniversitySeoul, South Korea
| | - Sung J Lee
- Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National UniversitySeoul, South Korea
| | - Ik-Hyun Cho
- Department of Science in Korean Medicine and Brain Korea 21 Plus Program, Graduate School, Kyung Hee UniversitySeoul, South Korea.,Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee UniversitySeoul, South Korea.,Institute of Korean Medicine, College of Korean Medicine, Kyung Hee UniversitySeoul, South Korea
| |
Collapse
|
16
|
Haghayegh Jahromi N, Tardent H, Enzmann G, Deutsch U, Kawakami N, Bittner S, Vestweber D, Zipp F, Stein JV, Engelhardt B. A Novel Cervical Spinal Cord Window Preparation Allows for Two-Photon Imaging of T-Cell Interactions with the Cervical Spinal Cord Microvasculature during Experimental Autoimmune Encephalomyelitis. Front Immunol 2017; 8:406. [PMID: 28443093 PMCID: PMC5387098 DOI: 10.3389/fimmu.2017.00406] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/22/2017] [Indexed: 11/13/2022] Open
Abstract
T-cell migration across the blood-brain barrier (BBB) is a crucial step in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Two-photon intravital microscopy (2P-IVM) has been established as a powerful tool to study cell-cell interactions in inflammatory EAE lesions in living animals. In EAE, central nervous system inflammation is strongly pronounced in the spinal cord, an organ in which 2P-IVM imaging is technically very challenging and has been limited to the lumbar spinal cord. Here, we describe a novel spinal cord window preparation allowing to use 2P-IVM to image immune cell interactions with the cervical spinal cord microvascular endothelium during EAE. We describe differences in the angioarchitecture of the cervical spinal cord versus the lumbar spinal cord, which will entail different hemodynamic parameters in these different vascular beds. Using T cells as an example, we demonstrate the suitability of this novel methodology in imaging the post-arrest multistep T-cell extravasation across the cervical spinal cord microvessels. The novel methodology includes an outlook to the analysis of the cellular pathway of T-cell diapedesis across the BBB by establishing visualization of endothelial junctions in this vascular bed.
Collapse
Affiliation(s)
| | - Heidi Tardent
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Naoto Kawakami
- Max Planck Institute of Neurobiology, Martinsried, Germany.,Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians University of Munich, Martinsried, Germany
| | - Stefan Bittner
- Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | - Frauke Zipp
- Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jens V Stein
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | |
Collapse
|
17
|
Druzd D, Matveeva O, Ince L, Harrison U, He W, Schmal C, Herzel H, Tsang AH, Kawakami N, Leliavski A, Uhl O, Yao L, Sander LE, Chen CS, Kraus K, de Juan A, Hergenhan SM, Ehlers M, Koletzko B, Haas R, Solbach W, Oster H, Scheiermann C. Lymphocyte Circadian Clocks Control Lymph Node Trafficking and Adaptive Immune Responses. Immunity 2017; 46:120-132. [PMID: 28087238 PMCID: PMC5263259 DOI: 10.1016/j.immuni.2016.12.011] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/24/2016] [Accepted: 11/18/2016] [Indexed: 12/25/2022]
Abstract
Lymphocytes circulate through lymph nodes (LN) in search for antigen in what is believed to be a continuous process. Here, we show that lymphocyte migration through lymph nodes and lymph occurred in a non-continuous, circadian manner. Lymphocyte homing to lymph nodes peaked at night onset, with cells leaving the tissue during the day. This resulted in strong oscillations in lymphocyte cellularity in lymph nodes and efferent lymphatic fluid. Using lineage-specific genetic ablation of circadian clock function, we demonstrated this to be dependent on rhythmic expression of promigratory factors on lymphocytes. Dendritic cell numbers peaked in phase with lymphocytes, with diurnal oscillations being present in disease severity after immunization to induce experimental autoimmune encephalomyelitis (EAE). These rhythms were abolished by genetic disruption of T cell clocks, demonstrating a circadian regulation of lymphocyte migration through lymph nodes with time-of-day of immunization being critical for adaptive immune responses weeks later. Lymphocyte numbers in lymph nodes and lymph oscillate over the course of the day Rhythmic Ccr7 and S1pr1 expression drives rhythmic lymphocyte homing and egress Adaptive immune responses to immunization and pathogens are time-of-day dependent Loss of circadian clocks in lymphocytes ablates rhythmic adaptive immune responses
Collapse
Affiliation(s)
- David Druzd
- BioMedical Center, Walter-Brendel-Centre for Experimental Medicine, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany
| | - Olga Matveeva
- Medical Department I, University of Lübeck, 23562 Lübeck, Germany
| | - Louise Ince
- BioMedical Center, Walter-Brendel-Centre for Experimental Medicine, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany
| | - Ute Harrison
- Max von Pettenkofer-Institute for Hygiene and Medical Microbiology, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Wenyan He
- BioMedical Center, Walter-Brendel-Centre for Experimental Medicine, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany
| | | | - Hanspeter Herzel
- Institute for Theoretical Biology, Humboldt University of Berlin, 10115 Berlin, Germany
| | - Anthony H Tsang
- Medical Department I, University of Lübeck, 23562 Lübeck, Germany
| | - Naoto Kawakami
- BioMedical Center, Institute of Clinical Neuroimmunology, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany
| | - Alexei Leliavski
- Laboratory of Immunology, Institute for Nutrition Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Olaf Uhl
- Ludwig-Maximilians-University, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337 Munich, Germany
| | - Ling Yao
- Department of Infectious Diseases and Pulmonary Medicine, Charité University Hospital Berlin, 10117 Berlin, Germany
| | - Leif Erik Sander
- Department of Infectious Diseases and Pulmonary Medicine, Charité University Hospital Berlin, 10117 Berlin, Germany
| | - Chien-Sin Chen
- BioMedical Center, Walter-Brendel-Centre for Experimental Medicine, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany
| | - Kerstin Kraus
- BioMedical Center, Walter-Brendel-Centre for Experimental Medicine, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany
| | - Alba de Juan
- BioMedical Center, Walter-Brendel-Centre for Experimental Medicine, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany
| | - Sophia Martina Hergenhan
- BioMedical Center, Walter-Brendel-Centre for Experimental Medicine, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany
| | - Marc Ehlers
- Laboratory of Immunology, Institute for Nutrition Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Berthold Koletzko
- Ludwig-Maximilians-University, Dr. von Hauner Children's Hospital, University of Munich Medical Center, 80337 Munich, Germany
| | - Rainer Haas
- Max von Pettenkofer-Institute for Hygiene and Medical Microbiology, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Werner Solbach
- Center for Infection and Inflammation, University of Lübeck, 23562 Lübeck, Germany
| | - Henrik Oster
- Medical Department I, University of Lübeck, 23562 Lübeck, Germany
| | - Christoph Scheiermann
- BioMedical Center, Walter-Brendel-Centre for Experimental Medicine, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany.
| |
Collapse
|
18
|
Castillo EF, Zheng H, Van Cabanlong C, Dong F, Luo Y, Yang Y, Liu M, Kao WWY, Yang XO. Lumican negatively controls the pathogenicity of murine encephalitic TH17 cells. Eur J Immunol 2016; 46:2852-2861. [PMID: 27682997 DOI: 10.1002/eji.201646507] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/02/2016] [Accepted: 09/27/2016] [Indexed: 11/11/2022]
Abstract
TH17 cells play an essential role in the development of both human multiple sclerosis and animal experimental autoimmune encephalomyelitis (EAE). Nevertheless, it is not well understood how the pathogenicity of TH17 cells is controlled in the autoimmune neuroinflammation. In vitro, we found Lumican (Lum), an extracellular matrix (ECM) protein, is selectively expressed by TH17 cells among tested murine TH subsets. Lum deficiency leads to earlier onset and enhanced severity of EAE. This enhanced disease in Lum-deficient mice is associated with increased production of IL-17 and IL-21 and decreased TH17 cell apoptosis. Dysregulation in cytokine production appears to be specific to TH17 cells as TH1 and TH2 cell polarization and/or cytokine production were unaltered. Furthermore, adoptive transfer of myelin oligodendrocyte glycoprotein specific TH17 cells derived from Lum-deficient mice led to earlier onset and increased severity of disease compared to controls highlighting a TH17-cell-intrinsic effect of Lum. Taken together, our results suggest that Lum negatively regulates encephalitic TH17 cells, implicating a potential therapeutic pathway in TH17 cell mediated autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Eliseo F Castillo
- Department of Molecular Genetics and Microbiology and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Handong Zheng
- Department of Molecular Genetics and Microbiology and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Christian Van Cabanlong
- Department of Molecular Genetics and Microbiology and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Fei Dong
- Deparment of Ophthalmology, University of Cincinnati, Cincinnati, OH, USA
| | - Yan Luo
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Winston W-Y Kao
- Deparment of Ophthalmology, University of Cincinnati, Cincinnati, OH, USA
| | - Xuexian O Yang
- Department of Molecular Genetics and Microbiology and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
19
|
Cekanaviciute E, Buckwalter MS. Astrocytes: Integrative Regulators of Neuroinflammation in Stroke and Other Neurological Diseases. Neurotherapeutics 2016; 13:685-701. [PMID: 27677607 PMCID: PMC5081110 DOI: 10.1007/s13311-016-0477-8] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Astrocytes regulate neuroinflammatory responses after stroke and in other neurological diseases. Although not all astrocytic responses reduce inflammation, their predominant function is to protect the brain by driving the system back to homeostasis after injury. They receive multidimensional signals within the central nervous system and between the brain and the systemic circulation. Processing this information allows astrocytes to regulate synapse formation and maintenance, cerebral blood flow, and blood-brain barrier integrity. Similarly, in response to stroke and other central nervous system disorders, astrocytes detect and integrate signals of neuronal damage and inflammation to regulate the neuroinflammatory response. Two direct regulatory mechanisms in the astrocyte arsenal are the ability to form both physical and molecular barriers that seal the injury site and localize the neuroinflammatory response. Astrocytes also indirectly regulate the inflammatory response by affecting neuronal health during the acute injury and axonal regrowth. This ability to regulate the location and degree of neuroinflammation after injury, combined with the long time course of neuroinflammation, makes astrocytic signaling pathways promising targets for therapies.
Collapse
Affiliation(s)
- Egle Cekanaviciute
- Department of Neurology and Neurological Sciences, Stanford Medical School, Stanford, CA, 94305, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford Medical School, Stanford, CA, 94305, USA.
- Department of Neurosurgery, Stanford Medical School, Stanford, CA, 94305, USA.
- Stanford Stroke Center, Stanford Medical School, Stanford, CA, 94305, USA.
| |
Collapse
|
20
|
Kawakami N. In vivo imaging in autoimmune diseases in the central nervous system. Allergol Int 2016; 65:235-42. [PMID: 26935215 DOI: 10.1016/j.alit.2016.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 11/18/2022] Open
Abstract
Intravital imaging is becoming more popular and is being used to visualize cellular motility and functions. In contrast to in vitro analysis, which resembles in vivo analysis, intravital imaging can be used to observe and analyze cells directly in vivo. In this review, I will summarize recent imaging studies of autoreactive T cell infiltration into the central nervous system (CNS) and provide technical background. During their in vivo journey, autoreactive T cells interact with many different cells. At first, autoreactive T cells interact with endothelial cells in the airways of the lung or with splenocytes, where they acquire a migratory phenotype to infiltrate into the CNS. After arriving at the CNS, they interact with endothelial cells of the leptomeningeal vessels or the choroid plexus before passing through the blood-brain barrier. CNS-infiltrating T cells become activated by recognizing endogenous autoantigens presented by local antigen-presenting cells (APCs). This activation was visualized in vivo by using protein-based sensors. One such sensor detects changes in intracellular calcium concentration as an early marker of T cell activation. Another sensor detects translocation of Nuclear factor of activated T-cells (NFAT) from cytosol to nucleus as a definitive sign of T cell activation. Importantly, intravital imaging is not just used to visualize cellular behavior. Together with precise analysis, intravital imaging deepens our knowledge of cellular functions in living organs and also provides a platform for developing therapeutic treatments.
Collapse
Affiliation(s)
- Naoto Kawakami
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians Universitaet Muenchen, Munich, Germany; Neuroimmunology Group, Max-Planck Institute of Neurobiology, Martinsried, Germany.
| |
Collapse
|
21
|
Abad C, Jayaram B, Becquet L, Wang Y, O’Dorisio MS, Waschek JA, Tan YV. VPAC1 receptor (Vipr1)-deficient mice exhibit ameliorated experimental autoimmune encephalomyelitis, with specific deficits in the effector stage. J Neuroinflammation 2016; 13:169. [PMID: 27357191 PMCID: PMC4928347 DOI: 10.1186/s12974-016-0626-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/14/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Vasoactive intestinal peptide (VIP) and pituitary adenylyl cyclase-activating polypeptide (PACAP) are two highly homologous neuropeptides. In vitro and ex vivo experiments repeatedly demonstrate that these peptides exert pronounced immunomodulatory (primarily anti-inflammatory) actions which are mediated by common VPAC1 and VPAC2 G protein-coupled receptors. In agreement, we have shown that mice deficient in PACAP ligand or VPAC2 receptors exhibit exacerbated experimental autoimmune encephalomyelitis (EAE). However, we observed that VIP-deficient mice are unexpectedly resistant to EAE, suggesting a requirement for this peptide at some stage of disease development. Here, we investigated the involvement of VPAC1 in the development of EAE using a VPAC1-deficient mouse model. METHODS EAE was induced in wild-type (WT) and VPAC1 knockout (KO) mice using myelin oligodendrocyte glycoprotein 35-55 (MOG35-55), and clinical scores were assessed continuously over 30 days. Immune responses in the spinal cords were determined by histology, real-time PCR and immunofluorescence, and in the draining lymph nodes by antigen-recall assays. The contribution of VPAC1 expression in the immune system to the development of EAE was evaluated by means of adoptive transfer and bone marrow chimera experiments. In other experiments, VPAC1 receptor analogs were given to WT mice. RESULTS MOG35-55-induced EAE was ameliorated in VPAC1 KO mice compared to WT mice. The EAE-resistant phenotype of VPAC1 KO mice correlated with reduced central nervous system (CNS) histopathology and cytokine expression in the spinal cord. The immunization phase of EAE appeared to be unimpaired because lymph node cells from EAE-induced VPAC1 KO mice stimulated in vitro with MOG exhibited robust proliferative and Th1/Th17 responses. Moreover, lymph node and spleen cells from KO mice were fully capable of inducing EAE upon transfer to WT recipients. In contrast, WT cells from MOG-immunized mice did not transfer the disease when administered to VPAC1 KO recipients, implicating a defect in the effector phase of the disease. Bone marrow chimera studies suggested that the resistance of VPAC1-deficient mice was only minimally dependent on the expression of this receptor in the immunogenic/hematopoietic compartment. Consistent with this, impaired spinal cord inductions of several chemokine mRNAs were observed in VPAC1 KO mice. Finally, treatment of WT mice with the VPAC1 receptor antagonist PG97-269 before, but not after, EAE induction mimicked the clinical phenotype of VPAC1 KO mice. CONCLUSIONS VPAC1 gene loss impairs the development of EAE in part by preventing an upregulation of CNS chemokines and invasion of inflammatory cells into the CNS. Use of VPAC1 antagonists in WT mice prior to EAE induction also support a critical role for VPAC1 signaling for the development of EAE.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Central Nervous System/immunology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Cytokines/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/complications
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Female
- Freund's Adjuvant/toxicity
- Laminin/metabolism
- Lymph Nodes/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Peptide Fragments/toxicity
- RNA, Messenger/metabolism
- Receptors, Vasoactive Intestinal Polypeptide, Type I/deficiency
- Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Th1 Cells/metabolism
- Th1 Cells/pathology
- Th17 Cells/metabolism
- Th17 Cells/pathology
- Time Factors
Collapse
Affiliation(s)
- Catalina Abad
- />Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, USA
- />Inserm U905, Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, Normandy, France
| | - Bhavaani Jayaram
- />Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Laurine Becquet
- />Inserm U905, Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, Normandy, France
| | - Yuki Wang
- />Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - M Sue O’Dorisio
- />Department of Pediatrics and Holden Comprehensive Cancer Center, RJ and LA Carver College of Medicine, University of Iowa, Iowa City, 52242 IA USA
| | - James A. Waschek
- />Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Yossan-Var Tan
- />Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, USA
- />Inserm U905, Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, Normandy, France
| |
Collapse
|
22
|
Abstract
In autoimmune neurologic disorders, the blood-brain barrier (BBB) plays a central role in immunopathogenesis, since this vascular interface is an entry path for cells and effector molecules of the peripheral immune system to reach the target organ, the central nervous system (CNS). The BBB's unique anatomic structure and the tightly regulated interplay of its cellular and acellular components allow for maintenance of brain homeostasis, regulation of influx and efflux, and protection from harm; these ensure an optimal environment for the neuronal network to function properly. In both health and disease, the BBB acts as mediator between the periphery and the CNS. For example, immune cell trafficking through the cerebral vasculature is essential to clear microbes or cell debris from neural tissues, while poorly regulated cellular transmigration can underlie or worsen CNS pathology. In this chapter, we focus on the specialized multicellular structure and function of the BBB/neurovascular unit and discuss how BBB breakdown can precede or be a consequence of neuroinflammation. We introduce the blood-cerebrospinal fluid barrier and include a brief aside about evolutionary aspects of barrier formation and refinements. Lastly, since restoration of barrier function is considered key to ameliorate neurologic disease, we speculate about new therapeutic avenues to repair a damaged BBB.
Collapse
Affiliation(s)
| | - Ajay Verma
- Biomarkers and Experimental Medicine, Biogen, Cambridge, MA, USA
| | | |
Collapse
|
23
|
Yang X, Yan J, Feng J. Treatment with tanshinone IIA suppresses disruption of the blood-brain barrier and reduces expression of adhesion molecules and chemokines in experimental autoimmune encephalomyelitis. Eur J Pharmacol 2016; 771:18-28. [DOI: 10.1016/j.ejphar.2015.12.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/19/2015] [Accepted: 12/07/2015] [Indexed: 10/22/2022]
|
24
|
Nosratabadi R, Rastin M, Sankian M, Haghmorad D, Tabasi N, Zamani S, Aghaee A, Salehipour Z, Mahmoudi M. St. John’s wort and its component hyperforin alleviate experimental autoimmune encephalomyelitis through expansion of regulatory T-cells. J Immunotoxicol 2015; 13:364-74. [DOI: 10.3109/1547691x.2015.1101512] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Reza Nosratabadi
- School of Medicine, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Rastin
- School of Medicine, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- School of Medicine, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Dariush Haghmorad
- School of Medicine, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nafiseh Tabasi
- School of Medicine, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shahrzad Zamani
- School of Medicine, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azita Aghaee
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zohre Salehipour
- School of Medicine, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- School of Medicine, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
25
|
Choi JH, Lee MJ, Jang M, Kim EJ, Shim I, Kim HJ, Lee S, Lee SW, Kim YO, Cho IH. An Oriental Medicine, Hyungbangpaedok-San Attenuates Motor Paralysis in an Experimental Model of Multiple Sclerosis by Regulating the T Cell Response. PLoS One 2015; 10:e0138592. [PMID: 26444423 PMCID: PMC4596626 DOI: 10.1371/journal.pone.0138592] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 09/01/2015] [Indexed: 12/23/2022] Open
Abstract
The preventive and therapeutic mechanisms in multiple sclerosis are not clearly understood. We investigated whether Hyungbangpaedok-san (HBPDS), a traditional herbal medicine, has a beneficial effect in experimental autoimmune encephalomyelitis (EAE) mice immunized with myelin oligodendrocyte glycoprotein peptide (MOG35-55). Onset-treatment with 4 types of HBPDS (extracted using distilled water and 30%/70%/100% ethanol as the solvent) alleviated neurological signs, and HBPDS extracted within 30% ethanol (henceforth called HBPDS) was more effective. Onset-treatment with HBPDS reduced demyelination and the recruitment/infiltration and activation of microglia/macrophages in the spinal cord of EAE mice, which corresponded to the reduced mRNA expression of pro-inflammatory cytokines (TNF-α, IL–6, and IL–1β), iNOS, and chemokines (MCP–1, MIP–1α, and RANTES) in the spinal cord. Onset-treatment with HBPDS inhibited changes in the components of the blood-brain barrier such as astrocytes, adhesion molecules (ICAM–1 and VCAM–1), and junctional molecules (claudin–3, claudin–5, and zona occludens–1) in the spinal cord of EAE mice. Onset-treatment with HBPDS reduced the elevated population of CD4+, CD4+/IFN-γ+, and CD4+/IL–17+ T cells in the spinal cord of EAE mice but it further increased the elevated population of CD4+/CD25+/Foxp3+ and CD4+/Foxp3+/Helios+ T cells. Pre-, onset-, post-, but not peak-treatment, with HBPDS had a beneficial effect on behavioral impairment in EAE mice. Taken together, HBPDS could alleviate the development/progression of EAE by regulating the recruitment/infiltration and activation of microglia and peripheral immune cells (macrophages, Th1, Th17, and Treg cells) in the spinal cord. These findings could help to develop protective strategies using HBPDS in the treatment of autoimmune disorders including multiple sclerosis.
Collapse
Affiliation(s)
- Jong Hee Choi
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
- Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
| | - Min Jung Lee
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
- Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
| | - Minhee Jang
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
| | - Eun-Jeong Kim
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
- Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
| | - Insop Shim
- Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
- Acupuncture & Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
| | - Hak-Jae Kim
- Department of Clinical Pharmacology, College of Medicine, Soonchunhyang University, Cheonan, 336–745, Republic of Korea
| | - Sanghyun Lee
- Department of Integrative Plant Science, Chung-Ang University, Anseong, 456–756, Republic of Korea
| | - Sang Won Lee
- Department of Medicinal Crop Research Institute, National Institute of Horticultural & Herbal Science, Rural Development Administration, Eumseong, 369–873, Republic of Korea
| | - Young Ock Kim
- Department of Medicinal Crop Research Institute, National Institute of Horticultural & Herbal Science, Rural Development Administration, Eumseong, 369–873, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
- Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
- Institute of Koreran Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 130–701, Republic of Korea
- * E-mail:
| |
Collapse
|
26
|
Reichenbach ZW, Li H, Gaughan JP, Elliott M, Tuma R. IV and IP administration of rhodamine in visualization of WBC-BBB interactions in cerebral vessels. Microsc Res Tech 2015. [PMID: 26207355 DOI: 10.1002/jemt.22552] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Epi-illuminescence intravital fluorescence microscopy has been employed to study leukocyte-endothelial interactions in a number of brain pathologies. Historically, dyes such as Rhodamine 6G have been injected intravenously. However, intravenous injections can predispose experimental animals to a multitude of complications and requires a high degree of technical skill. Here, we study the efficacy of injecting Rhodamine 6G into the peritoneum (IP) for the purpose of analyzing leukocyte-endothelial interactions through a cranial window during real time intravital microscopy. After examining the number of rolling and adherent leukocytes through a cranial window, we found no advantage to the intravenous injection (IV). Additionally, we tested blood from both routes of injection by flow cytometry to gain a very precise picture of the two methods. The two routes of administration failed to show any difference in the ability to detect cells. The study supports the notion that IP Rhodamine 6G works as efficaciously as IV and should be considered a viable alternative in experimental design for investigations employing intravital microscopy. Facilitated intravital studies will allow for more exploration into cerebral pathologies and allow for more rapid translation from the laboratory to the patient with less chance of experimental error from failed IV access.
Collapse
Affiliation(s)
- Zachary Wilmer Reichenbach
- Temple University School of Medicine, Center for Sustance Abuse Research, Philadelphia, Pennsylvania, 19140.,Department of Cellular and Molecular Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Hongbo Li
- Temple University School of Medicine, Center for Sustance Abuse Research, Philadelphia, Pennsylvania, 19140.,Department of Cellular and Molecular Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140
| | - John P Gaughan
- Temple University School of Medicine, Biostatistics Consulting Center, Philadelphia, Pennsylvania, 19140
| | - Melanie Elliott
- Department Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, 19107
| | - Ronald Tuma
- Temple University School of Medicine, Center for Sustance Abuse Research, Philadelphia, Pennsylvania, 19140.,Department of Cellular and Molecular Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140
| |
Collapse
|
27
|
Fasudil regulates T cell responses through polarization of BV-2 cells in mice experimental autoimmune encephalomyelitis. Acta Pharmacol Sin 2014; 35:1428-38. [PMID: 25263338 DOI: 10.1038/aps.2014.68] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/22/2014] [Indexed: 01/22/2023] Open
Abstract
AIM Fasudil, a selective Rho kinase (ROCK) inhibitor, has been shown to alleviate the severity of experimental autoimmune encephalomyelitis (EAE) via attenuating demyelination and neuroinflammation. The aim of this study was to investigate the effects of fasudil on interactions between macrophages/microglia and T cells in a mice EAE model. METHODS Mouse BV-2 microglia were treated with IFN-γ and fasudil. Cell viability was detected with MTT assay. BV-2 microglia polarization was analyzed using flow cytometry. Cytokines and other proteins were detected with ELISA and Western blotting, respectively. Mice were immunized with MOG35-55 to induce EAE, and then treated with fasudil (40 mg/kg, ip) every other day from d 3 to d 27 pi. Encephalomyelitic T cells were prepared from the spleen of mice immunized with MOG35-55 on d 9 pi. RESULTS Treatment of mouse BV-2 microglia with fasudil (15 μg/mL) induced significant phenotype polarization and functional plasticity, shifting M1 to M2 polarization. When co-cultured with the encephalomyelitic T cells, fasudil-treated BV-2 microglia significantly inhibited the proliferation of antigen-reactive T cells, and down-regulated IL-17-expressing CD4(+) T cells and IL-17 production. Furthermore, fasudil-treated BV-2 microglia significantly up-regulated CD4(+)CD25(high) and CD4(+)IL-10(+) regulatory T cells (Tregs) and IL-10 production, suggesting that the encephalomyelitic T cells had converted to Tregs. In EAE mice, fasudil administration significantly decreased both CD11b(+)iNOS(+) and CD11b(+)TNF-α(+) M1 microglia, and increased CD11b(+)IL-10(+) M2 microglia. CONCLUSION Fasudil polarizes BV-2 microglia into M2 cells, which convert the encephalomyelitic T cells into Tregs in the mice EAE model.
Collapse
|
28
|
Affiliation(s)
- Wakiro Sato
- Department of Immunology; National Institute of Neuroscience; National Center of Neurology and Psychiatry (NCNP); Tokyo Japan
| |
Collapse
|
29
|
Abstract
Multiple sclerosis (MS) is a complex autoimmune disease of the CNS. At present, MS etiology remains unknown, but it is believed to be caused by environmental factors acting on a genetic predisposition. Several studies suggest that different microorganisms could play a role in triggering autoimmunity, through immunological cross-reactivity or molecular mimicry. An overview of the knowledge regarding the bacteria involved in MS is given, placing emphasis on the newest candidate proposed: Mycobacterium avium subsp. paratuberculosis. This review will focus on discussing several arguments that might support a causal role for Mycobacterium avium subsp. paratuberculosis as an etiologic agent in MS. Additionally, a possible mechanism is postulated attempting to explain how the bacteria could initiate autoimmunity.
Collapse
Affiliation(s)
- Davide Cossu
- Department of Biomedical Sciences, Division of Experimental & Clinical Microbiology, University of Sassari, Sassari, Italy
| | | | | |
Collapse
|
30
|
Kan QC, Zhu L, Liu N, Zhang GX. Matrine suppresses expression of adhesion molecules and chemokines as a mechanism underlying its therapeutic effect in CNS autoimmunity. Immunol Res 2013; 56:189-96. [DOI: 10.1007/s12026-013-8393-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Ahmad A, Seguias L, Ban K. Diagnosis and treatment of pediatric acute transverse myelitis. Pediatr Ann 2012; 41:477-82. [PMID: 23814937 DOI: 10.3928/00904481-20121022-15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Ayesha Ahmad
- University of Texas Southwestern Medical Center, 5151 Harry Hines Boulevard, Dallas, TX 75390, USA
| | | | | |
Collapse
|
32
|
Steinman L. Nostalgia: the similarities between immunological and neurological memory. Immunol Rev 2012; 248:5-9. [DOI: 10.1111/j.1600-065x.2012.01139.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|