1
|
Han X, Wang Y, Zhang K, Na T, Wu T, Hao X, Jin Y, Wang Y, Wang H, Meng S. Dosage and organic acid residue of myelin oligodendrocyte glycoprotein 35-55 peptide influences immunopathology and development of Bacillus Calmette-Guérin induced experimental autoimmune encephalomyelitis. Exp Anim 2025; 74:16-30. [PMID: 38987201 PMCID: PMC11742474 DOI: 10.1538/expanim.24-0012] [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: 02/07/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) serves as a model for studying multiple sclerosis, with immunization strategies utilizing myelin oligodendrocyte glycoprotein (MOG)35-55 peptide, emulsified in adjuvant enriched with Mycobacterium tuberculosis (Mtb). This study examined the effects of Bacillus Calmette-Guérin (BCG) as an adjuvant, alongside the impact of MOG35-55 peptide doses and their residual counter ions on EAE development. We found that BCG can be effectively used to induce EAE with similar incidence and severity as heat-killed H37Ra, contingent upon the appropriate MOG35-55 peptide dose. Different immunization doses of MOG35-55 peptide significantly affect EAE development, with higher doses leading to a paradoxical reduction in disease activity, probably due to peripheral tolerance mechanisms. Furthermore, doses of MOG35-55 peptides with acetate showed a more pronounced effect on disease development compared to those containing trifluoroacetic acid (TFA), suggesting the potential influence of residual counter ions on EAE activity. We highlighted the feasibility of applying BCG to the establishment of EAE for the first time. Our findings emphasized the importance of MOG35-55 peptide dosage and composition in modulating EAE development, offering insights into the mechanisms of autoimmunity and tolerance. This could have implications for autoimmune disease research and the design of therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoyan Han
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Ying Wang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Kehua Zhang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Tao Na
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Tingting Wu
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Xiaofang Hao
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Yuxuan Jin
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Yuchun Wang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Haohan Wang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| | - Shufang Meng
- Cell Collection and Research Center, National Institutes for Food and Drug Control, 31 Huatuo Road, Daxing District, Beijing, 102629, P.R. China
| |
Collapse
|
2
|
Haghmorad D, Yousefi B, Eslami M, Rashidy-Pour A, Tarahomi M, Tavaf MJ, Soltanmohammadi A, Zargarani S, Kamyshnyi A, Oksenych V. Oral Administration of Myelin Oligodendrocyte Glycoprotein Attenuates Experimental Autoimmune Encephalomyelitis through Induction of Th2/Treg Cells and Suppression of Th1/Th17 Immune Responses. Curr Issues Mol Biol 2022; 44:5728-5740. [PMID: 36421672 PMCID: PMC9688797 DOI: 10.3390/cimb44110388] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 10/28/2024] Open
Abstract
Multiple Sclerosis (MS) is a demyelinating autoimmune disorder of the central nervous system (CNS). Experimental autoimmune encephalomyelitis (EAE) has been widely used to determine the pathogenesis of the disease and evaluate new treatment strategies for MS. Therefore, we investigated the efficacy of oral administration of a Myelin Oligodendrocyte Glycoprotein (MOG) in the treatment of EAE. Female C57BL/6 mice were utilized in three groups (Control group, received PBS orally; prevention group, oral administration of MOG35-55 two weeks before EAE induction; treatment group, oral administration of MOG35-55 after EAE induction). MOG administration, both as prevention and treatment, significantly controlled clinical score, weight loss, CNS inflammation, and demyelination, mainly through the modulation of T cell proliferation, and reduction in pro-inflammatory cytokines and transcription factors, including TNF-α, IFN-γ, IL-17, T-bet, and ROR-γt. MOG administration, both as prevention and treatment, also induced anti-inflammatory cytokines and transcription factors, including IL-4, TGF-β, GATA-3, and Foxp3. The results showed that oral administration of MOG, both as prevention and treatment, could efficiently control EAE development. Immunomodulatory mechanisms include the induction of Th2 and Treg cells and the suppression of pro-inflammatory Th1 and Th17 cells.
Collapse
Affiliation(s)
- Dariush Haghmorad
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35131, Iran
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Bahman Yousefi
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35131, Iran
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Mahdieh Tarahomi
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Maryam Jadid Tavaf
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | | | - Simin Zargarani
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35131, Iran
| | - Aleksandr Kamyshnyi
- Department of Microbiology, Virology and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Valentyn Oksenych
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| |
Collapse
|
3
|
Chen W, Wang J, Yang H, Sun Y, Chen B, Liu Y, Han Y, Shan M, Zhan J. Interleukin 22 and its association with neurodegenerative disease activity. Front Pharmacol 2022; 13:958022. [PMID: 36176437 PMCID: PMC9514046 DOI: 10.3389/fphar.2022.958022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/26/2022] [Indexed: 11/29/2022] Open
Abstract
It is worth noting that neuroinflammation is well recognized as a symptom of neurodegenerative diseases (NDs). The regulation of neuroinflammation becomes an attractive focus for innovative ND treatment technologies. There is evidence that IL-22 is associated with the development and progression of a wide assortment of NDs. For example, IL-22 can activate glial cells, causing them to generate pro-inflammatory cytokines and encourage lymphocyte infiltration in the brain. IL-22 mRNA is highly expressed in Alzheimer’s disease (AD) patients, and a high expression of IL-22 has also been detected in the brains of patients with other NDs. We examine the role of IL-22 in the development and treatment of NDs in this review, and we believe that IL-22 has therapeutic potential in these diseases.
Collapse
Affiliation(s)
- Wenjian Chen
- Department of Orthopaedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Jianpeng Wang
- School of First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Huaizhi Yang
- School of First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Yuankai Sun
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Bangjie Chen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuchen Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanxun Han
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ming Shan
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- *Correspondence: Ming Shan, ; Junfeng Zhan,
| | - Junfeng Zhan
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Ming Shan, ; Junfeng Zhan,
| |
Collapse
|
4
|
Moorman CD, Sohn SJ, Phee H. Emerging Therapeutics for Immune Tolerance: Tolerogenic Vaccines, T cell Therapy, and IL-2 Therapy. Front Immunol 2021; 12:657768. [PMID: 33854514 PMCID: PMC8039385 DOI: 10.3389/fimmu.2021.657768] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases affect roughly 5-10% of the total population, with women affected more than men. The standard treatment for autoimmune or autoinflammatory diseases had long been immunosuppressive agents until the advent of immunomodulatory biologic drugs, which aimed at blocking inflammatory mediators, including proinflammatory cytokines. At the frontier of these biologic drugs are TNF-α blockers. These therapies inhibit the proinflammatory action of TNF-α in common autoimmune diseases such as rheumatoid arthritis, psoriasis, ulcerative colitis, and Crohn's disease. TNF-α blockade quickly became the "standard of care" for these autoimmune diseases due to their effectiveness in controlling disease and decreasing patient's adverse risk profiles compared to broad-spectrum immunosuppressive agents. However, anti-TNF-α therapies have limitations, including known adverse safety risk, loss of therapeutic efficacy due to drug resistance, and lack of efficacy in numerous autoimmune diseases, including multiple sclerosis. The next wave of truly transformative therapeutics should aspire to provide a cure by selectively suppressing pathogenic autoantigen-specific immune responses while leaving the rest of the immune system intact to control infectious diseases and malignancies. In this review, we will focus on three main areas of active research in immune tolerance. First, tolerogenic vaccines aiming at robust, lasting autoantigen-specific immune tolerance. Second, T cell therapies using Tregs (either polyclonal, antigen-specific, or genetically engineered to express chimeric antigen receptors) to establish active dominant immune tolerance or T cells (engineered to express chimeric antigen receptors) to delete pathogenic immune cells. Third, IL-2 therapies aiming at expanding immunosuppressive regulatory T cells in vivo.
Collapse
Affiliation(s)
| | | | - Hyewon Phee
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| |
Collapse
|
5
|
Canto E, Isobe N, Didonna A, Hauser SL, Oksenberg JR. Aberrant STAT phosphorylation signaling in peripheral blood mononuclear cells from multiple sclerosis patients. J Neuroinflammation 2018. [PMID: 29514694 PMCID: PMC5840794 DOI: 10.1186/s12974-018-1105-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Multiple sclerosis (MS) is characterized by increased activation of peripheral blood mononuclear cells (PBMCs), linked to perturbations in the phosphorylation of signaling proteins. Methods We developed a phosphoflow cytometry protocol to assess the levels of 11 phosphorylated nuclear proteins at baseline conditions and after cell activation in distinct PBMC populations from 41 treatment-naïve relapsing-remitting (RR) MS subjects and 37 healthy controls, and in a second cohort of 9 untreated RRMS patients and 10 secondary progressive (SP) MS patients. Levels of HLA-ABC, HLA-E, and HLA-DR were also assessed. Phosphorylation levels of selected proteins were also assessed in mouse splenocytes isolated from myelin oligodendrocyte glycoprotein (MOG)35–55-induced experimental autoimmune encephalomyelitis (EAE). Results Modest differences were observed at baseline between patients and controls, with general lower phosphorylation levels in cells from affected individuals. Conversely, a dramatic increase in phosphorylated p38MAPK and STAT proteins was observed across all cell types in MS patients compared to controls after in vitro activation. A similar phosphorylation profile was observed in mouse lymphocytes primed in vivo with MOG. Furthermore, levels of all p-STAT proteins were found directly correlated with HLA expression in monocytes. Levels of phosphorylated proteins did not differ between relapsing-remitting and secondary progressive MS patients either in baseline conditions or after stimulation. Lastly, phosphorylation levels appear to be independent of the genotype. Conclusion The response to IFN-α through STAT proteins signaling is strongly dysregulated in MS patients irrespective of disease stage. These findings suggest that the aberrant activation of this pathway could lead to changes in the expression of HLA molecules in antigen presenting cells, which are known to play important roles in the regulation of the immune response in health and disease. Electronic supplementary material The online version of this article (10.1186/s12974-018-1105-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ester Canto
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Noriko Isobe
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Alessandro Didonna
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, Nelson Rising Lane, San Francisco, CA, 94158, USA
| | | | - Stephen L Hauser
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Jorge R Oksenberg
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, Nelson Rising Lane, San Francisco, CA, 94158, USA.
| |
Collapse
|
6
|
Liu Y, Gibson SA, Benveniste EN, Qin H. Opportunities for Translation from the Bench: Therapeutic Intervention of the JAK/STAT Pathway in Neuroinflammatory Diseases. Crit Rev Immunol 2018; 35:505-27. [PMID: 27279046 DOI: 10.1615/critrevimmunol.2016015517] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathogenic CD4+ T cells and myeloid cells play critical roles in the pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), an animal model of MS. These immune cells secrete aberrantly high levels of pro-inflammatory cytokines that pathogenically bridge the innate and adaptive immune systems and damage neurons and oligodendrocytes. These cytokines include interleukin-2 (IL-2), IL-6, IL-12, IL-21, IL-23, granulocyte macrophage-colony stimulating factor (GM-CSF), and interferon-γ (IFN-γ). It is, therefore, not surprising that both the dysregulated expression of these cytokines and the subsequent activation of their downstream signaling cascades is a common feature in MS/EAE. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway is utilized by numerous cytokines for signal transduction and is essential for the development and regulation of immune responses. Unbridled activation of the JAK/STAT pathway by pro-inflammatory cytokines has been demonstrated to be critically involved in the pathogenesis of MS/EAE. In this review, we discuss recent advancements in our understanding of the involvement of the JAK/STAT signaling pathway in the pathogenesis of MS/EAE, with a particular focus on therapeutic approaches to target the JAK/STAT pathway.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294; Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Sara A Gibson
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| |
Collapse
|
7
|
Thomé R, Moore JN, Mari ER, Rasouli J, Hwang D, Yoshimura S, Ciric B, Zhang GX, Rostami AM. Induction of Peripheral Tolerance in Ongoing Autoimmune Inflammation Requires Interleukin 27 Signaling in Dendritic Cells. Front Immunol 2017; 8:1392. [PMID: 29163476 PMCID: PMC5663690 DOI: 10.3389/fimmu.2017.01392] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/09/2017] [Indexed: 01/08/2023] Open
Abstract
Peripheral tolerance to autoantigens is induced via suppression of self-reactive lymphocytes, stimulation of tolerogenic dendritic cells (DCs) and regulatory T (Treg) cells. Interleukin (IL)-27 induces tolerogenic DCs and Treg cells; however, it is not known whether IL-27 is important for tolerance induction. We immunized wild-type (WT) and IL-27 receptor (WSX-1) knockout mice with MOG35–55 for induction of experimental autoimmune encephalomyelitis and intravenously (i.v.) injected them with MOG35–55 after onset of disease to induce i.v. tolerance. i.v. administration of MOG35–55 reduced disease severity in WT mice, but was ineffective in Wsx−/− mice. IL-27 signaling in DCs was important for tolerance induction, whereas its signaling in T cells was not. Further mechanistic studies showed that IL-27-dependent tolerance relied on cooperation of distinct subsets of spleen DCs with the ability to induce T cell-derived IL-10 and IFN-γ. Overall, our data show that IL-27 is a key cytokine in antigen-induced peripheral tolerance and may provide basis for improvement of antigen-specific tolerance approaches in multiple sclerosis and other autoimmune diseases.
Collapse
Affiliation(s)
- Rodolfo Thomé
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Jason N Moore
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Elisabeth R Mari
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Javad Rasouli
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Daniel Hwang
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Satoshi Yoshimura
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States.,Department of Neurology, Graduate School of Medical Sciences, Neurological Institute, Kyushu University, Fukuoka, Japan
| | - Bogoljub Ciric
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Guang-Xian Zhang
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Abdolmohamad M Rostami
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
8
|
Aryanpour R, Pasbakhsh P, Zibara K, Namjoo Z, Beigi Boroujeni F, Shahbeigi S, Kashani IR, Beyer C, Zendehdel A. Progesterone therapy induces an M1 to M2 switch in microglia phenotype and suppresses NLRP3 inflammasome in a cuprizone-induced demyelination mouse model. Int Immunopharmacol 2017; 51:131-139. [PMID: 28830026 DOI: 10.1016/j.intimp.2017.08.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/14/2017] [Accepted: 08/09/2017] [Indexed: 12/14/2022]
Abstract
Demyelination of the central nervous system (CNS) has been associated to reactive microglia in neurodegenerative disorders, such as multiple sclerosis (MS). The M1 microglia phenotype plays a pro-inflammatory role while M2 is involved in anti-inflammatory processes in the brain. In this study, CPZ-induced demyelination mouse model was used to investigate the effect of progesterone (PRO) therapy on microglia activation and neuro-inflammation. Results showed that progesterone therapy (CPZ+PRO) decreased neurological behavioral deficits, as demonstrated by significantly decreased escape latencies, in comparison to CPZ mice. In addition, CPZ+PRO caused a significant reduction in the mRNA expression levels of M1-markers (iNOS, CD86, MHC-II and TNF-α) in the corpus callosum region, whereas the expression of M2-markers (Trem-2, CD206, Arg-1 and TGF-β) was significantly increased, in comparison to CPZ mice. Moreover, CPZ+PRO resulted in a significant decrease in the number of iNOS+ and Iba-1+/iNOS+ cells (M1), whereas TREM-2+ and Iba-1+/TREM-2+ cells (M2) significantly increased, in comparison to CPZ group. Furthermore, CPZ+PRO caused a significant decrease in mRNA and protein expression levels of NLRP3 and IL-18 (~2-fold), in comparison to the CPZ group. Finally, CPZ+PRO therapy was accompanied with reduced levels of demyelination, compared to CPZ, as confirmed by immunofluorescence to myelin basic protein (MBP) and Luxol Fast Blue (LFB) staining, as well as transmission electron microscopy (TEM) analysis. In summary, we reported for the first time that PRO therapy causes polarization of M2 microglia, attenuation of M1 phenotype, and suppression of NLRP3 inflammasome in a CPZ-induced demyelination model of MS.
Collapse
Affiliation(s)
- Roya Aryanpour
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kazem Zibara
- ER045, Laboratory of Stem Cells, DSST, Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Zeinab Namjoo
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Fatemeh Beigi Boroujeni
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Shahbeigi
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | - Adib Zendehdel
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany; Giulan Neuroscience Research Center, Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
9
|
Ushio A, Arakaki R, Yamada A, Saito M, Tsunematsu T, Kudo Y, Ishimaru N. Crucial roles of macrophages in the pathogenesis of autoimmune disease. World J Immunol 2017; 7:1-8. [DOI: 10.5411/wji.v7.i1.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 11/18/2016] [Accepted: 12/09/2016] [Indexed: 02/05/2023] Open
Abstract
Macrophages are key players in various immune responses. In addition to functions in innate immunity such as antigen phagocytosis and cytokine production, antigen presentation by macrophage represents a link between innate and acquired immunity. During inflammatory processes, naïve monocytes differentiate into pro-inflammatory M1 and anti-inflammatory M2 macrophages. Resident monocytes/macrophages contribute to immune response that maintains tissue-specific homeostasis. In the target organs of autoimmune diseases, macrophages have dual functions in both the induction and suppression of autoimmune responses, which are mediated by production of various cytokines and chemokines, or by interaction with other immune cells. This review focuses on selected autoimmune diseases, such as systemic lupus erythematosus, multiple sclerosis, rheumatoid arthritis, and Sjögren’s syndrome, to illustrate the key roles of macrophages in the cellular or molecular pathogenesis of autoimmunity. In addition, the contribution of macrophages to each autoimmune disease is compared.
Collapse
|
10
|
IL22 in Egyptian SLE patients, could it reflect disease activity, skin or renal involvement or is it only an expensive ESR? EGYPTIAN RHEUMATOLOGIST 2017. [DOI: 10.1016/j.ejr.2016.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
11
|
Challa DK, Mi W, Lo ST, Ober RJ, Ward ES. Antigen dynamics govern the induction of CD4 + T cell tolerance during autoimmunity. J Autoimmun 2016; 72:84-94. [DOI: 10.1016/j.jaut.2016.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 11/16/2022]
|
12
|
Antigen-oriented T cell migration contributes to myelin peptide induced-EAE and immune tolerance. Clin Immunol 2016; 169:36-46. [PMID: 27327113 DOI: 10.1016/j.clim.2016.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 05/05/2016] [Accepted: 06/07/2016] [Indexed: 02/08/2023]
Abstract
Treatment with soluble myelin peptide can efficiently and specifically induce tolerance to demyelination autoimmune diseases including multiple sclerosis, however the mechanism underlying this therapeutic effect remains to be elucidated. In actively induced mouse model of experimental autoimmune encephalomyelitis (EAE) we analyzed T cell and innate immune cell responses in the central nervous system (CNS) and spleen after intraperitoneal (i.p.) infusion of myelin oligodendrocyte glycoprotein (MOG). We found that i.p. MOG infusion blocked effector T cell recruitment to the CNS and protected mice from EAE and lymphoid organ atrophy. Innate immune CD11b(+) cells preferentially recruited MOG-specific effector T cells, particularly when activated to become competent antigen presenting cells (APCs). During EAE development, mature APCs were enriched in the CNS rather than in the spleen, attracting effector T cells to the CNS. Increased myelin antigen exposure induced CNS-APC maturation, recruiting additional effector T cells to the CNS, causing symptoms of disease. MOG triggered functional maturation of splenic APCs. MOG presenting APCs interacted with MOG-specific T cells in the spleen, aggregating to cluster around CD11b(+) cells, and were trapped in the periphery. This process was MHC II dependent as an MHC II directed antibody blocked CD4(+) T cell cluster formation. These findings highlight the role of myelin peptide-loaded APCs in myelin peptide-induced EAE and immune tolerance.
Collapse
|
13
|
Mari ER, Rasouli J, Ciric B, Moore JN, Conejo-Garcia JR, Rajasagi N, Zhang GX, Rabinovich GA, Rostami A. Galectin-1 is essential for the induction of MOG35-55 -based intravenous tolerance in experimental autoimmune encephalomyelitis. Eur J Immunol 2016; 46:1783-96. [PMID: 27151444 DOI: 10.1002/eji.201546212] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 04/04/2016] [Accepted: 05/02/2016] [Indexed: 11/06/2022]
Abstract
In experimental autoimmune encephalomyelitis (EAE), intravenous (i.v.) injection of the antigen, myelin oligodendrocyte glycoprotein-derived peptide, MOG35-55 , suppresses disease development, a phenomenon called i.v. tolerance. Galectin-1, an endogenous glycan-binding protein, is upregulated during autoimmune neuroinflammation and plays immunoregulatory roles by inducing tolerogenic dendritic cells (DCs) and IL-10 producing regulatory type 1 T (Tr1) cells. To examine the role of galectin-1 in i.v. tolerance, we administered MOG35-55 -i.v. to wild-type (WT) and galectin-1 deficient (Lgals1(-/-) ) mice with ongoing EAE. MOG35-55 suppressed disease in the WT, but not in the Lgals1(-/-) mice. The numbers of Tr1 cells and Treg cells were increased in the CNS and periphery of tolerized WT mice. In contrast, Lgals1(-/-) MOG-i.v. mice had reduced numbers of Tr1 cells and Treg cells in the CNS and periphery, and reduced IL-27, IL-10, and TGF-β1 expression in DCs in the periphery. DCs derived from i.v.-tolerized WT mice suppressed disease when adoptively transferred into mice with ongoing EAE, whereas DCs from Lgals1(-/-) MOG-i.v. mice were not suppressive. These findings demonstrate that galectin-1 is required for i.v. tolerance induction, likely via induction of tolerogenic DCs leading to enhanced development of Tr1 cells, Treg cells, and downregulation of proinflammatory responses.
Collapse
Affiliation(s)
- Elisabeth R Mari
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Javad Rasouli
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jason N Moore
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | - José R Conejo-Garcia
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Naveen Rajasagi
- Comparative and Experimental Medicine, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN, USA
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina.,School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
14
|
Yoshida Y, Mikami N, Matsushima Y, Otani F, Miyawaki M, Takatsuji M, Banno R, Tsuji T, Fujita T, Tsujikawa K, Kohno T. Functional Mechanism(s) of the Inhibition of Disease Progression by Combination Treatment with Fingolimod Plus Pathogenic Antigen in a Glucose-6-phosphate Isomerase Peptide-Induced Arthritis Mouse Model. Biol Pharm Bull 2015; 38:1120-5. [PMID: 25994913 DOI: 10.1248/bpb.b14-00873] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that combination treatment with fingolimod (FTY720) plus antigenic peptide of glucose-6-phosphate isomerase (residues 325-339) (GPI325-339) from the onset of symptoms significantly inhibited disease progression in a mouse model of GPI325-339-induced arthritis. In this study, we investigated the mechanism(s) involved. The model mice were treated from arthritis onset with FTY720 alone, GPI325-339 alone, or the combination of FTY720 plus GPI325-339. At the end of treatment, inguinal lymph nodes (LNs) were excised and examined histologically and in flow cytometry. Levels of apoptotic cells, programmed death-1-expressing CD4(+)forkhead box P3(-) nonregulatory T cells (non-Tregs), and cytotoxic T-lymphocyte antigen 4-expressing non-Tregs in inguinal LNs were markedly increased in the combination treatment group mice. Regulatory T cells (Tregs) were also increased. These results indicate that combination treatment with FTY720 plus GPI325-339 inhibits the progression of arthritis by inducing clonal deletion and anergy of pathogenic T cells and also by immune suppression via Tregs.
Collapse
Affiliation(s)
- Yuya Yoshida
- Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Mari ER, Moore JN, Zhang GX, Rostami A. Mechanisms of immunological tolerance in central nervous system inflammatory demyelination. ACTA ACUST UNITED AC 2015; 6:264-274. [PMID: 26425145 DOI: 10.1111/cen3.12196] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Multiple sclerosis is a complex autoimmune disease of the central nervous system that results in a disruption of the balance between pro-inflammatory and anti-inflammatory signals in the immune system. Given that central nervous system inflammation can be suppressed by various immunological tolerance mechanisms, immune tolerance has become a focus of research in the attempt to induce long-lasting immune suppression of pathogenic T cells. Mechanisms underlying this tolerance induction include induction of regulatory T cell populations, anergy and the induction of tolerogenic antigen-presenting cells. The intravenous administration of encephalitogenic peptides has been shown to suppress experimental autoimmune encephalomyelitis and induce tolerance by promoting the generation of regulatory T cells and inducing apoptosis of pathogenic T cells. Safe and effective methods of inducing long-lasting immune tolerance are essential for the treatment of multiple sclerosis. By exploring tolerogenic mechanisms, new strategies can be devised to strengthen the regulatory, anti-inflammatory cell populations thereby weakening the pathogenic, pro-inflammatory cell populations.
Collapse
Affiliation(s)
- Elisabeth R Mari
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jason N Moore
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Efficacy of the selective progesterone receptor agonist Nestorone for chronic experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 276:89-97. [DOI: 10.1016/j.jneuroim.2014.08.619] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/14/2014] [Accepted: 08/19/2014] [Indexed: 11/17/2022]
|
17
|
Yoshida Y, Mikami N, Tsuji T, Takada Y, Nakazawa Y, Dan R, Takatsuji M, Fujita T, Tsujikawa K, Kohno T. Mechanism of induction of immune tolerance in experimental autoimmune encephalomyelitis by combination treatment with fingolimod plus pathogenic autoantigen. ACTA ACUST UNITED AC 2014. [DOI: 10.1111/cen3.12140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Yuya Yoshida
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
| | - Norihisa Mikami
- Laboratory of Molecular and Cellular Physiology; Graduate School of Pharmaceutical Sciences; Osaka University; Osaka Japan
| | - Takumi Tsuji
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
| | - Yuki Takada
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
| | - Yuka Nakazawa
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
| | - Rie Dan
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
| | - Miku Takatsuji
- Laboratory of Molecular and Cellular Physiology; Graduate School of Pharmaceutical Sciences; Osaka University; Osaka Japan
| | - Tetsuro Fujita
- Research Institute for Production and Development; Kyoto Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology; Graduate School of Pharmaceutical Sciences; Osaka University; Osaka Japan
| | - Takeyuki Kohno
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
- Research Institute for Production and Development; Kyoto Japan
| |
Collapse
|
18
|
Macrophages: a double-edged sword in experimental autoimmune encephalomyelitis. Immunol Lett 2014; 160:17-22. [PMID: 24698730 DOI: 10.1016/j.imlet.2014.03.006] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/28/2014] [Accepted: 03/17/2014] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS) is a debilitating neurological disorder of the central nervous system (CNS), characterized by activation and infiltration of leukocytes and dendritic cells into the CNS. In the initial phase of MS and its animal model, experimental autoimmune encephalomyelitis (EAE), peripheral macrophages infiltrate into the CNS, where, together with residential microglia, they participate in the induction and development of disease. During the early phase, microglia/macrophages are immediately activated to become classically activated macrophages (M1 cells), release pro-inflammatory cytokines and damage CNS tissue. During the later phase, microglia/macrophages in the inflamed CNS are less activated, present as alternatively activated macrophage phenotype (M2 cells), releasing anti-inflammatory cytokines, accompanied by inflammation resolution and tissue repair. The balance between activation and polarization of M1 cells and M2 cells in the CNS is important for disease progression. Pro-inflammatory IFN-γ and IL-12 drive M1 cell polarization, while IL-4 and IL-13 drive M2 cell polarization. Given that polarized macrophages are reversible in a well-defined cytokine environment, macrophage phenotypes in the CNS can be modulated by molecular intervention. This review summarizes the detrimental and beneficial roles of microglia and macrophages in the CNS, with an emphasis on the role of M2 cells in EAE and MS patients.
Collapse
|
19
|
Abstract
Interleukin-22 (IL-22) is an IL-10 family cytokine that was recently discovered to be released by T helper 17 (Th17) cells, Th22 cells, etc. Recently, there is emerging evidence that IL-22 is involved in the development and pathogenesis of psoriasis. For instance, IL-22 can inhibit keratinocyte terminal differentiation and can induce psoriasis-like epidermis alterations; serum IL-22 levels were correlated with the disease severity of psoriasis patients, and IL-22 mRNA was positively expressed in the psoriatic skin lesions, but negatively expressed in the normal controls. All these findings suggest that IL-22 may be implicated in psoriasis; therapeutics targeting IL-22 may have promise as a potential therapeutic target for treating psoriasis. In the present review, we summarize recent advances on the role of IL-22 in the pathogenesis and treatment of psoriasis.
Collapse
Affiliation(s)
- Ji-Qing Hao
- Department of Oncology, The Fist Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui, 230022, China,
| |
Collapse
|
20
|
Papadopoulou A, Kappos L, Sprenger T. Teriflunomide for oral therapy in multiple sclerosis. Expert Rev Clin Pharmacol 2013; 5:617-28. [PMID: 23234322 DOI: 10.1586/ecp.12.56] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Teriflunomide, the active metabolite of an approved antirheumatic drug, is an emerging oral therapy for multiple sclerosis (MS). Next to the inhibition of pyrimidine biosynthesis and proliferation of activated lymphocytes, it seems to have multiple anti-inflammatory and immunomodulating effects. Phase II and III clinical trials in relapsing MS demonstrated favorable safety and tolerability of the drug, as well as clinical efficacy, with a significant reduction of relapse rate, comparable with those of the available injectable immunomodulatory agents. While multiple other studies with teriflunomide are currently ongoing, its exact place in future treatment algorithms for MS is difficult to predict. It may be a good alternative for patients wishing to have an oral treatment with relatively large data regarding long-term safety.
Collapse
|
21
|
Bergman P, James T, Kular L, Ruhrmann S, Kramarova T, Kvist A, Supic G, Gillett A, Pivarcsi A, Jagodic M. Next-generation sequencing identifies microRNAs that associate with pathogenic autoimmune neuroinflammation in rats. THE JOURNAL OF IMMUNOLOGY 2013; 190:4066-75. [PMID: 23514736 DOI: 10.4049/jimmunol.1200728] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
MicroRNAs (miRNAs) are known to regulate most biological processes and have been found dysregulated in a variety of diseases, including multiple sclerosis (MS). In this study, we characterized miRNAs that associate with susceptibility to develop experimental autoimmune encephalomyelitis (EAE) in rats, a well-established animal model of MS. Using Illumina next-generation sequencing, we detected 544 miRNAs in the lymph nodes of EAE-susceptible Dark Agouti and EAE-resistant Piebald Virol Glaxo rats during immune activation. Forty-three miRNAs were found differentially expressed between the two strains, with 81% (35 out of 43) showing higher expression in the susceptible strain. Only 33% of tested miRNAs displayed differential expression in naive lymph nodes, suggesting that a majority of regulated miRNAs are EAE dependent. Further investigation of a selected six miRNAs indicates differences in cellular source and kinetics of expression. Several of the miRNAs, including miR-146a, miR-21, miR-181a, miR-223, and let-7, have previously been implicated in immune system regulation. Moreover, 77% (33 out of 43) of the miRNAs were associated with MS and other autoimmune diseases. Target genes likely regulated by the miRNAs were identified using computational predictions combined with whole-genome expression data. Differentially expressed miRNAs and their targets involve functions important for MS and EAE, such as immune cell migration through targeting genes like Cxcr3 and cellular maintenance and signaling by regulation of Prkcd and Stat1. In addition, we demonstrated that these three genes are direct targets of miR-181a. Our study highlights the impact of multiple miRNAs, displaying diverse kinetics and cellular sources, on development of pathogenic autoimmune inflammation.
Collapse
Affiliation(s)
- Petra Bergman
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17176, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Pan HF, Li XP, Zheng SG, Ye DQ. Emerging role of interleukin-22 in autoimmune diseases. Cytokine Growth Factor Rev 2013; 24:51-7. [PMID: 22906768 PMCID: PMC4003867 DOI: 10.1016/j.cytogfr.2012.07.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 07/24/2012] [Accepted: 07/26/2012] [Indexed: 02/08/2023]
Abstract
Interleukin-22 (IL-22) is an IL-10 family cytokine member that was recently discovered to be mainly produced by Th17 cells. Previous studies have indicated the importance of IL-22 in host defense against Gram-negative bacterial organisms (in gut and lung). Recently, there is emerging evidence that IL-22 is involved in the development and pathogenesis of several autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), multiple sclerosis (MS), Sjögren's syndrome (SS) and psoriasis. Therapeutics targeting IL-22 therefore may have promise for treating various autoimmune diseases. In this review, we discuss the recent progression of the involvement of IL-22 in the development and pathogenesis of autoimmune diseases, as well as its clinical implications and therapeutic potential.
Collapse
Affiliation(s)
- Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui provincial laboratory of population health & major disease screening and diagnosis, Anhui Medical University, Hefei, China
| | - Xiang-Pei Li
- Department of Rheumatology, Anhui Provincial Hospital, Hefei, China
| | - Song Guo Zheng
- Division of Rheumatology/Immunology, Department of Medicine, University of Southern California
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Anhui provincial laboratory of population health & major disease screening and diagnosis, Anhui Medical University, Hefei, China
| |
Collapse
|
23
|
Wu WX, Zuo L, Dine KE, Shindler KS. Decreased signal transducers and activators of transcription (STAT) protein expression in lymphatic organs during EAE development in mice. ACTA ACUST UNITED AC 2013; 1. [PMID: 25346854 DOI: 10.7243/2053-213x-1-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is mediated by myelin-specific CD4+ T cells secreting Th1 and/or Th17 cytokines. Signal transducer and activator of transcription (STAT) family proteins have essential roles in transmitting Th1 and/or Th17 cytokine-mediated signals. However, most studies demonstrating the importance of the STAT signaling system in EAE have focused on distinct members of this family, often looking at their role specifically in the central nervous system, or in vitro. There is limited information available regarding the temporal and spatial expression patterns of each STAT protein and interplay between STAT proteins over the course of EAE development in critical lymphatic organs in vivo. In the present study, we demonstrate dramatic and progressive decrease of all six STAT family members (STAT1, STAT2, STAT3, STAT4, STAT5, STAT6) in the spleen and lymph nodes through the course of EAE development in SJL/J mice, in contrast with almost steady expression of thymic STAT proteins. Decreased splenic and lymphatic STAT expression was accompanied by significant enlargement of the spleen and lymph nodes, and histological proliferation of T cell areas with remodeling of the splenic microstructure in EAE mice. All STAT family members except STAT2 were mainly confined in T cell areas in spleen, whereas they were distributed in a protein specific manner in thymus. We present here a comprehensive analysis of all six members of the STAT family in spleen, lymph nodes and thymus through the development phase of EAE. Results suggest that EAE induced inflammatory T cells may develop distinct biological features different from normal splenic T cells due to altered STAT signaling.
Collapse
Affiliation(s)
- Wen Xuan Wu
- Scheie Eye Institute and F.M. Kirby Center for Molecular Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Zuo
- Department of Ophthalmology, Second Hospital of Jilin University, Jilin, China
| | - Kimberly E Dine
- Scheie Eye Institute and F.M. Kirby Center for Molecular Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth S Shindler
- Scheie Eye Institute and F.M. Kirby Center for Molecular Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
O'Brien K, Gran B, Rostami A. T-cell based immunotherapy in experimental autoimmune encephalomyelitis and multiple sclerosis. Immunotherapy 2010; 2:99-115. [PMID: 20231863 DOI: 10.2217/imt.09.61] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
One of the reasons multiple sclerosis (MS) has been considered a T-cell mediated autoimmune disease is that a similar experimental disease can be induced in certain rodents and primates by immunization with myelin antigens, leading to T-cell-mediated inflammatory demyelination in the CNS. In addition, most if not all pharmacological treatments available for MS are biologically active on T cells. In this article we review the principles of T-cell-based immunotherapies and the specific actions of current and novel treatments on T-cell functions, when these are known. For both licensed and innovative agents, we also discuss biological actions on other immune cell types. Finally, we offer a brief perspective on expected changes in the use of MS immunotherapies in the near future.
Collapse
Affiliation(s)
- Kate O'Brien
- Division of Clinical Neurology, University of Nottingham, UK
| | | | | |
Collapse
|