1
|
Tan PH, Ji J, Hsing CH, Tan R, Ji RR. Emerging Roles of Type-I Interferons in Neuroinflammation, Neurological Diseases, and Long-Haul COVID. Int J Mol Sci 2022; 23:ijms232214394. [PMID: 36430870 PMCID: PMC9696119 DOI: 10.3390/ijms232214394] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Interferons (IFNs) are pleiotropic cytokines originally identified for their antiviral activity. IFN-α and IFN-β are both type I IFNs that have been used to treat neurological diseases such as multiple sclerosis. Microglia, astrocytes, as well as neurons in the central and peripheral nervous systems, including spinal cord neurons and dorsal root ganglion neurons, express type I IFN receptors (IFNARs). Type I IFNs play an active role in regulating cognition, aging, depression, and neurodegenerative diseases. Notably, by suppressing neuronal activity and synaptic transmission, IFN-α and IFN-β produced potent analgesia. In this article, we discuss the role of type I IFNs in cognition, neurodegenerative diseases, and pain with a focus on neuroinflammation and neuro-glial interactions and their effects on cognition, neurodegenerative diseases, and pain. The role of type I IFNs in long-haul COVID-associated neurological disorders is also discussed. Insights into type I IFN signaling in neurons and non-neuronal cells will improve our treatments of neurological disorders in various disease conditions.
Collapse
Affiliation(s)
- Ping-Heng Tan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 701, Taiwan
- Correspondence: (P.-H.T.); (C.-H.H.)
| | - Jasmine Ji
- Neuroscience Department, Wellesley College, Wellesley, MA 02482, USA
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 701, Taiwan
- Correspondence: (P.-H.T.); (C.-H.H.)
| | - Radika Tan
- Kaohsiung American School, Kaohsiung 81354, Taiwan
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
2
|
Shiri-Shahsavar MR, Mirshafiee A, Parastouei K, Ebrahimi-Kalan A, Yekaninejad S, Soleymani F, Chahardoli R, Mazaheri Nezhad Fard R, Saboor-Yaraghi AA. A Novel Combination of Docosahexaenoic Acid, All-Trans Retinoic Acid, and 1, 25-Dihydroxyvitamin D 3 Reduces T-Bet Gene Expression, Serum Interferon Gamma, and Clinical Scores but Promotes PPARγ Gene Expression in Experimental Autoimmune Encephalomyelitis. J Mol Neurosci 2016; 60:498-508. [PMID: 27647308 DOI: 10.1007/s12031-016-0834-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/30/2016] [Indexed: 12/19/2022]
Abstract
Vitamins are immunologically interesting due to their significant immunomodulatory activities. Experimental autoimmune encephalomyelitis (EAE) is one of the most commonly used experimental models for studying autoimmune disorder in multiple sclerosis (MS). The aim of this study was to evaluate the protective and ameliorative effects of novel combination of all-trans retinoic acid (ATRA), 1,25-dihydroxyvitamin D3 (D3), and docosahexaenoic acid (DHA) on EAE-specific determinants and target gene expressions. Mice were randomly categorized into three groups before EAE induction [non-treated EAE (Group E), treated EAE (Group T), and healthy mice (Group H)]. Encephalomyelitis was induced in female C57BL/6 mice by subcutaneous immunization using commercial kits. Preceding day of EAE induction, combination of ATRA, D3, and DHA was administered with a single IP injection every 48 h and continued until day 26. Findings of present study showed that administration of vitamins A, D, and DHA significantly decreased average clinical scores, cumulative EAE score, and EAE incidence in Group T, compared to Group E (p values <0.001). Interferon γ secretion in serum and T-bet mRNA expression in splenocytes were significantly reduced (p = 0.004, p = 0.029, respectively) while PPARγ mRNA expression was significantly increased in Group T compared to Group E (p = 0.021). These findings highlighted that ATRA, D3, and DHA combination modulated PPARγ and T-bet gene expression and resulted in decrease in Th1 response and lymphocyte invasion into the central nervous system (CNS) and resultant inflammation. In conclusion, the results of this study suggested the potential use of this intervention in treatment and/or prevention of EAE/MS and probably other Th1 cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Mohammad Reza Shiri-Shahsavar
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Mirshafiee
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Karim Parastouei
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Ebrahimi-Kalan
- Neuroscience Department, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Yekaninejad
- Department of Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Soleymani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Chahardoli
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Mazaheri Nezhad Fard
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Akbar Saboor-Yaraghi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, International Campus, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Li J, Yang J, Lu YW, Wu S, Wang MR, Zhu JM. Possible Role of Staphylococcal Enterotoxin B in the Pathogenesis of Autoimmune Diseases. Viral Immunol 2015; 28:354-9. [PMID: 26086678 DOI: 10.1089/vim.2015.0017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
As a member of superantigens (SAgs) produced by Staphylococcus aureus, staphylococcal enterotoxin B (SEB) is a exotoxin superantigen that can regulate the activity of immunomodulatory and pro-inflammatory cell types. In addition, SEB plays a critical role in the pathogenesis of autoimmune disorders either by initiating the autoimmune process or by inducing a relapse in an individual in clinical remission from an autoimmune disorder. SEB can directly activate T lymphocytes, leading to the release of cytokines, superoxides, or other mediators of inflammation either directly or indirectly, because of its unique ability to cross-link human major histocompatibility complex (MHC) class II and T cell receptors (TCR), forming a trimolecular complex. This review discusses the potential effects of SEB in the pathogenesis of autoimmune diseases such as multiple sclerosis, systemic lupus erythematosus, and rheumatoid arthritis, and explores some updated therapeutic medications to neutralize SEB.
Collapse
Affiliation(s)
- Jing Li
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Jie Yang
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Yu-wei Lu
- 2 Department of Information, The Second Hospital of Anhui Medical University , Hefei, China
| | - Song Wu
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Ming-rui Wang
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Ji-min Zhu
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| |
Collapse
|
4
|
Abstract
Despite dramatic advances in surgical techniques, imaging and adjuvant radiotherapy or chemotherapy, the prognosis for patients with malignant glial tumors remains dismal. Based on the current knowledge regarding immune responses in the healthy CNS and glioma-bearing hosts, this review discusses dendritic cell-based immunotherapeutic approaches for malignant gliomas and the relevance of recent clinical trials and their outcomes. It is now recognized that the CNS is not an immunologically tolerated site and clearance of arising glioma cells is a routine physiologic function of the normal, noncompromised immune system. To escape from immune surveillance, however, clinically apparent gliomas develop complex mechanisms that suppress tumoricidal immune responses. Although the use of dendritic cells for the treatment of glioma patients may be the most appropriate approach, an effective treatment paradigm for these tumors may eventually require the use of several types of treatment. Additionally, given the heterogeneity of this disease process and an immune-refractory tumor cell population, the series use of rational multiple modalities that target disparate tumor characteristics may be the most effective therapeutic strategy to treat malignant gliomas.
Collapse
Affiliation(s)
- Yasuharu Akasaki
- Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Suite 800 East, 8631 West 3 Street, Los Angeles, CA 90048, USA
| | | | | |
Collapse
|
5
|
Shachar I, Karin N. The dual roles of inflammatory cytokines and chemokines in the regulation of autoimmune diseases and their clinical implications. J Leukoc Biol 2012; 93:51-61. [PMID: 22949334 DOI: 10.1189/jlb.0612293] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cytokines and chemokines are secreted, small cell-signaling protein molecules, whose receptors are expressed on immune cells. These factors play a critical role in immune cell differentiation, migration, and polarization into functional subtypes and in directing their biological functions. Much attention has been devoted to exploring the role of key inflammatory cytokines and promigratory chemokines in autoimmune, autoinflammatory, and allergic diseases, leading to development of therapeutic strategies that are based on their targeted neutralization. Recent studies, including those coming from our groups, show that several major proinflammatory cytokines and chemokines, including IFN-γ, IL-2, CCL2, and CXCL12, may also function as anti-inflammatory mediators and therefore, may have potential as anti-inflammatory drugs. Likewise, major anti-inflammatory mediators, such as TGF-β, may under certain conditions, in combination with other cytokines, exhibit proinflammatory function and direct the polarization of the highly inflammatory CD4(+) Th17 cells. We show here that the biological function of pro- and anti-inflammatory cytokines is dependent on three key parameters: the local concentration of a given cytokine, the stage of disease in which it is administered, and its combination with other cytokines. The therapeutic implications of these findings are discussed, including two very recent studies summarizing clinical trials, in which low-dose administration of IL-2 was used to successfully suppress HCV and GVHD.
Collapse
Affiliation(s)
- Idit Shachar
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| | | |
Collapse
|
6
|
Kaushansky N, Kerlero de Rosbo N, Zilkha-Falb R, Yosef-Hemo R, Cohen L, Ben-Nun A. 'Multi-epitope-targeted' immune-specific therapy for a multiple sclerosis-like disease via engineered multi-epitope protein is superior to peptides. PLoS One 2011; 6:e27860. [PMID: 22140475 PMCID: PMC3226621 DOI: 10.1371/journal.pone.0027860] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 10/26/2011] [Indexed: 11/26/2022] Open
Abstract
Antigen-induced peripheral tolerance is potentially one of the most efficient and specific therapeutic approaches for autoimmune diseases. Although highly effective in animal models, antigen-based strategies have not yet been translated into practicable human therapy, and several clinical trials using a single antigen or peptidic-epitope in multiple sclerosis (MS) yielded disappointing results. In these clinical trials, however, the apparent complexity and dynamics of the pathogenic autoimmunity associated with MS, which result from the multiplicity of potential target antigens and “epitope spread”, have not been sufficiently considered. Thus, targeting pathogenic T-cells reactive against a single antigen/epitope is unlikely to be sufficient; to be effective, immunospecific therapy to MS should logically neutralize concomitantly T-cells reactive against as many major target antigens/epitopes as possible. We investigated such “multi-epitope-targeting” approach in murine experimental autoimmune encephalomyelitis (EAE) associated with a single (“classical”) or multiple (“complex”) anti-myelin autoreactivities, using cocktail of different encephalitogenic peptides vis-a-vis artificial multi-epitope-protein (designated Y-MSPc) encompassing rationally selected MS-relevant epitopes of five major myelin antigens, as “multi-epitope-targeting” agents. Y-MSPc was superior to peptide(s) in concomitantly downregulating pathogenic T-cells reactive against multiple myelin antigens/epitopes, via inducing more effective, longer lasting peripheral regulatory mechanisms (cytokine shift, anergy, and Foxp3+ CTLA4+ regulatory T-cells). Y-MSPc was also consistently more effective than the disease-inducing single peptide or peptide cocktail, not only in suppressing the development of “classical” or “complex EAE” or ameliorating ongoing disease, but most importantly, in reversing chronic EAE. Overall, our data emphasize that a “multi-epitope-targeting” strategy is required for effective immune-specific therapy of organ-specific autoimmune diseases associated with complex and dynamic pathogenic autoimmunity, such as MS; our data further demonstrate that the “multi-epitope-targeting” approach to therapy is optimized through specifically designed multi-epitope-proteins, rather than myelin peptide cocktails, as “multi-epitope-targeting” agents. Such artificial multi-epitope proteins can be tailored to other organ-specific autoimmune diseases.
Collapse
Affiliation(s)
- Nathali Kaushansky
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | | | - Rina Zilkha-Falb
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Reut Yosef-Hemo
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Lydia Cohen
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Avraham Ben-Nun
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
7
|
Ghazavi A, Mosayebi G. The mechanism of sesame oil in ameliorating experimental autoimmune encephalomyelitis in C57BL/6 mice. Phytother Res 2011; 26:34-8. [PMID: 21538630 DOI: 10.1002/ptr.3515] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 03/22/2011] [Indexed: 01/22/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a Th1 cell-mediated autoimmune disease of the CNS that serves as an animal model for multiple sclerosis (MS). The study investigated the effectiveness of treatment with sesame oil on the development of EAE. EAE was induced in 8 week old C57BL/6 mice with an emulsion of MOG35-55. Therapy with sesame oil (4 mL/kg/day as oral gavage) was started on day 3 before the immunization. IFN-gamma and IL-10 production from cultured spleen supernatants were determined by the ELISA method. The results showed that daily oral gavage of sesame oil significantly reduced the clinical symptoms of EAE in C57BL/6 mice. In addition, sesame oil-treated mice displayed a significantly delayed disease onset. Mononuclear cells isolated from spleen of sesame oil-treated mice showed a significant decrease in the production of IFN-gamma compared with control mice (p = 0.001). IL-10 production was also enhanced in splenic mononuclear cells in sesame oil-treated mice. The ratio of IFN-gamma to IL-10 in sesame oil-treated EAE mice was significantly less than in non-treated EAE mice (p = 0.01). This report indicates that sesame oil therapy protected mice from developing EAE by reducing IFN-gamma secretion. Thus, sesame oil treatment may be effective in MS patients by immunomodulating the Th1 immune response.
Collapse
Affiliation(s)
- A Ghazavi
- Infectious Disease Research Center, Department of Immunology and Microbiology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | | |
Collapse
|
8
|
Reyes JL, Espinoza-Jiménez AF, González MI, Verdin L, Terrazas LI. Taenia crassiceps infection abrogates experimental autoimmune encephalomyelitis. Cell Immunol 2010; 267:77-87. [PMID: 21185554 DOI: 10.1016/j.cellimm.2010.11.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 11/18/2010] [Accepted: 11/29/2010] [Indexed: 12/19/2022]
Abstract
Helminth infections induce strong immunoregulation that can modulate subsequent pathogenic challenges. Taenia crassiceps causes a chronic infection that induces a Th2-biased response and modulates the host cellular immune response, including reduced lymphoproliferation in response to mitogens, impaired antigen presentation and the recruitment of suppressive alternatively activated macrophages (AAMФ). In this study, we aimed to evaluate the ability of T. crassiceps to reduce the severity of experimental autoimmune encephalomyelitis (EAE). Only 50% of T. crassiceps-infected mice displayed EAE symptoms, which were significantly less severe than uninfected mice. This effect was associated with both decreased MOG-specific splenocyte proliferation and IL-17 production and limited leukocyte infiltration into the spinal cord. Infection with T. crassiceps induced an anti-inflammatory cytokine microenvironment, including decreased TNF-α production and high MOG-specific production of IL-4 and IL-10. While the mRNA expression of TNF-α and iNOS was lower in the brain of T. crassiceps-infected mice with EAE, markers for AAMФ were highly expressed. Furthermore, in these mice, there was reduced entry of CD3(+)Foxp3(-) cells into the brain. The T. crassiceps-induced immune regulation decreased EAE severity by dampening T cell activation, proliferation and migration to the CNS.
Collapse
Affiliation(s)
- José L Reyes
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Edo, México 54090, Mexico
| | | | | | | | | |
Collapse
|
9
|
Ulzheimer JC, Meuth SG, Bittner S, Kleinschnitz C, Kieseier BC, Wiendl H. Therapeutic approaches to multiple sclerosis: an update on failed, interrupted, or inconclusive trials of immunomodulatory treatment strategies. BioDrugs 2010; 24:249-74. [PMID: 20623991 DOI: 10.2165/11537160-000000000-00000] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Multiple sclerosis (MS) continues to be a therapeutic challenge, and much effort is being made to develop new and more effective immune therapies. Particularly in the past decade, neuroimmunologic research has delivered new and highly effective therapeutic options, as seen in the growing number of immunotherapeutic agents and biologics in development. However, numerous promising clinical trials have failed to show efficacy or have had to be halted prematurely because of unexpected adverse events. Some others have shown results that are of unknown significance with regard to a reliable assessment of true efficacy versus safety. For example, studies of the highly innovative monoclonal antibodies that selectively target immunologic effector molecules have not only revealed the impressive efficacy of such treatments, they have also raised serious concerns about the safety profiles of these antibodies. These results add a new dimension to the estimation of risk-benefit ratios regarding acute or long-term adverse effects. Therapeutic approaches that have previously failed in MS have indicated that there are discrepancies between theoretical expectations and practical outcomes of different compounds. Learning from these defeats helps to optimize future study designs and to reduce the risks to patients. This review summarizes trials on MS treatments since 2001 that failed or were interrupted, attempts to analyze the underlying reasons for failure, and discusses the implications for our current view of MS pathogenesis, clinical practice, and design of future studies. In order to maintain clarity, this review focuses on anti-inflammatory therapies and does not include studies on already approved and effective disease-modifying therapies, albeit used in distinct administration routes or under different paradigms. Neuroprotective and alternative treatment strategies are presented elsewhere.
Collapse
|
10
|
York NR, Mendoza JP, Ortega SB, Benagh A, Tyler AF, Firan M, Karandikar NJ. Immune regulatory CNS-reactive CD8+T cells in experimental autoimmune encephalomyelitis. J Autoimmun 2010; 35:33-44. [PMID: 20172692 PMCID: PMC2878858 DOI: 10.1016/j.jaut.2010.01.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 01/12/2010] [Accepted: 01/21/2010] [Indexed: 10/19/2022]
Abstract
Immune-based self-recognition and failure to modulate this response are believed to contribute to the debilitating autoimmune pathology observed in multiple sclerosis (MS). Studies from its murine model, experimental autoimmune encephalomyelitis (EAE), have shown that neuroantigen-specific CD4+T cells are capable of inducing disease, while their immune sibling, the CD8+T cells, have largely been ignored. To understand their role in autoimmune demyelination, we first confirmed that, similar to our observations in human MS, there is robust induction of neuroantigen-reactive CD8+T cells in several models, including MOG(35-55)/CFA-induced EAE. However, MOG(35-55)-specific CD8+T-cells, when purified, were unable to adoptively transfer disease into naïve mice (in contrast to CD4+T-cells). In fact, we observed that the transfer of these neuroantigen-specific CD8+T cells was able to suppress the induction of EAE and to inhibit ongoing EAE. These regulatory CD8+T cells produced IFN-gamma and perforin and were able to kill MOG loaded CD4+T-cells as well as CD4-depleted APC, suggesting a cytotoxic/suppressor mechanism. Inhibition of EAE was associated with both the modulation of APC function as well as decreased MOG-specific CD4+T cell responses. Our studies reveal a novel and unexpected immune regulatory function for neuroantigen-specific CD8+T cells and have interesting biologic and therapeutic implications.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cells, Cultured
- Central Nervous System/immunology
- Cytotoxicity, Immunologic
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Glycoproteins/immunology
- Humans
- Interferon-gamma/metabolism
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis/immunology
- Myelin-Oligodendrocyte Glycoprotein
- Peptide Fragments/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
| | | | - Sterling B. Ortega
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Andrew Benagh
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Andrew F. Tyler
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Mihail Firan
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| | - Nitin J. Karandikar
- Department of Pathology, UT Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9072, USA
| |
Collapse
|
11
|
Li M, Liu X, Zhou Y, Su SB. Interferon-lambdas: the modulators of antivirus, antitumor, and immune responses. J Leukoc Biol 2009; 86:23-32. [PMID: 19304895 DOI: 10.1189/jlb.1208761] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
IFN-lambdas, including IFN-lambda1, IFN-lambda2, and IFN-lambda3, also known as IL-29, IL-28A, or IL-28B, are a newly described group of cytokines distantly related to the type I IFNs and IL-10 family members. The IFN-lambdaR complex consists of a unique ligand-binding chain, IFN-lambdaR1 (also designated IL-28Ralpha), and an accessory chain, IL-10R2, which is shared with receptors for IL-10-related cytokines. IFN-lambdas signal through the IFN-lambdaR and activate pathways of JAK-STATs and MAPKs to induce antiviral, antiproliferative, antitumor, and immune responses. In this review, we summarize recent findings about the biology of IFN-lambdas and their pathophysiological roles in viral infection, cancer, and immune responses of the innate and adaptive arms.
Collapse
Affiliation(s)
- Mingcai Li
- Institute of Inflammation and Immune Diseases, Shantou University Medical College, Shantou, China
| | | | | | | |
Collapse
|
12
|
Interferon beta induces mature dendritic cell apoptosis through caspase-11/caspase-3 activation. Blood 2009; 114:1344-54. [PMID: 19531658 DOI: 10.1182/blood-2008-12-196592] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Although interferon beta (IFNbeta) decreases relapse rate and disease activity in multiple sclerosis (MS), the mechanisms involved have not been elucidated. The present study is the first report on the apoptotic effect of IFNbeta in mature, but not immature, myeloid dendritic cells (DCs). Both exogenous IFNbeta added to DCs matured through exposure to proinflammatory cytokines and endogenous IFNbeta secreted after lipopolysaccharide stimulation induced DC cell death. Apoptosis of mature DCs required both NF-kappaB and STAT-1 activation, and was mediated through the induction of caspase-11 expression and activation of caspase-3. In vivo, we observed increased caspase-11 expression and a significant decrease in the number of splenic DCs after lipopolysaccharide administration in wt but not in STAT-1-deficient mice. Since mature DCs are major contributors to the inflammatory response and essential partners in the induction of adaptive immunity, IFNbeta-dependent elimination of activated DCs could play an essential role in re-establishing homeostasis, and might represent a new molecular mechanism for the therapeutic effect of IFNbeta in MS.
Collapse
|
13
|
Wasserman HA, Beal CD, Zhang Y, Jiang N, Zhu C, Evavold BD. MHC variant peptide-mediated anergy of encephalitogenic T cells requires SHP-1. THE JOURNAL OF IMMUNOLOGY 2008; 181:6843-9. [PMID: 18981103 DOI: 10.4049/jimmunol.181.10.6843] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Our lab has demonstrated that encephalitogenic T cells can be effectively anergized by treatment with MHC variant peptides, which are analogues of immunogenic peptides containing an amino acid substitution at an MHC anchor residue. The MHC variant peptide of myelin oligodendrocyte glycoprotein (MOG)(35-55) proves an effective treatment as it does not induce symptoms of experimental autoimmune encephalomyelitis and fails to recruit macrophages or MOG(35-55)-specific T cells to the CNS. In this study, we sought to characterize the signaling pathways required for the induction of anergy by building upon the observations identifying the tyrosine phosphatase SHP-1 as a critical regulator of T cell responsiveness. Motheaten viable heterozygous mice, which contain a mutation in the SHP-1 gene resulting in a reduction in functional SHP-1, were challenged with MOG(35-55) or the MOG(35-55) MHC variant 45D. These mice display symptoms of experimental autoimmune encephalomyelitis upon immunization with MHC variant peptide and have significant CNS infiltration of tetramer-positive CD4(+) cells and macrophages, unlike B6 mice challenged with the variant peptide. The effects of SHP-1 are directly on the T cell as Motheaten viable heterozygous mice autoreactive T cells are not anergized in vitro. Lastly, we demonstrate no distinguishable difference in the initial interaction between the TCR and agonist or MHC variant. Rather, an unstable interaction between peptide and MHC attenuates the T cell response, seen in a decreased half-life relative to MOG(35-55). These results identify SHP-1 as a mediator of T cell anergy induced by destabilized peptide:MHC complexes.
Collapse
Affiliation(s)
- Heather A Wasserman
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30332, USA
| | | | | | | | | | | |
Collapse
|
14
|
Thomson CE, McCulloch M, Sorenson A, Barnett SC, Seed BV, Griffiths IR, McLaughlin M. Myelinated, synapsing cultures of murine spinal cord--validation as an in vitro model of the central nervous system. Eur J Neurosci 2008; 28:1518-35. [PMID: 18793322 PMCID: PMC2777255 DOI: 10.1111/j.1460-9568.2008.06415.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Research in central nervous system (CNS) biology and pathology requires in vitro models, which, to recapitulate the CNS in vivo, must have extensive myelin and synapse formation under serum-free (defined) conditions. However, finding such a model has proven difficult. The technique described here produces dense cultures of myelinated axons, with abundant synapses and nodes of Ranvier, that are suitable for both morphological and biochemical analysis. Cellular and molecular events were easily visualised using conventional microscopy. Ultrastructurally, myelin sheaths were of the appropriate thickness relative to axonal diameter (G-ratio). Production of myelinated axons in these cultures was consistent and repeatable, as shown by statistical analysis of multiple experimental repeats. Myelinated axons were so abundant that from one litter of embryonic mice, myelin was produced in amounts sufficient for bulk biochemical analysis. This culture method was assessed for its ability to generate an in vitro model of the CNS that could be used for both neurobiological and neuropathological research. Myelin protein kinetics were investigated using a myelin fraction isolated from the cultures. This fraction was found to be superior, quantitatively and qualitatively, to the fraction recovered from standard cultures of dissociated oligodendrocytes, or from brain slices. The model was also used to investigate the roles of specific molecules in the pathogenesis of inflammatory CNS diseases. Using the defined conditions offered by this culture system, dose-specific, inhibitory effects of inflammatory cytokines on myelin formation were demonstrated, unequivocally. The method is technically quick, easy and reliable, and should have wide application to CNS research.
Collapse
Affiliation(s)
- C E Thomson
- Comparative Anatomy and Physiology, Institute of Veterinary, Biomedical and Animal Sciences, Massey University, Palmerston North, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
15
|
Zhou WB, Wang R, Deng YN, Ji XB, Huang GX, Xu YZ. Study of Cbl-b dynamics in peripheral blood lymphocytes isolated from patients with multiple sclerosis. Neurosci Lett 2008; 440:336-9. [PMID: 18565657 DOI: 10.1016/j.neulet.2008.05.089] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Revised: 05/21/2008] [Accepted: 05/26/2008] [Indexed: 10/22/2022]
Abstract
E3 ubiquitin ligase Casitas B cell lymphoma-b (Cbl-b) has been recently highlighted as a negative regulator of T-cell activation and which dysfunction usually results in autoimmunity. To present, however, the possible involvement of Cbl-b in multiple sclerosis (MS), an autoimmune demyelinating disease mediated by T-helper 1 (Th1) cells is still unclear. To clarify this, using reverse transcriptase polymerase chain reaction (RT-PCR) and Western blot analyses, we thus investigated the levels of Cbl-b mRNA and protein in peripheral blood T-lymphocytes isolated from 11 MS patients in acute relapse phase and 20 cases in remission phase. 16 healthy subjects were used as normal control. Cbl-b mRNA and protein levels were found both significantly down-regulated in peripheral blood lymphocytes isolated from MS patients (P<0.0001). Interestingly, this decrease of Cbl-b protein but not mRNA levels was significantly more marked in samples of relapsed patients than that of remitted cases (P<0.0001). In addition, it was shown that Cbl-b mRNA levels being inversely correlated with the frequency of MS clinical relapses (P<0.0001). Altogether, the data show for the first time that Cbl-b dynamics in peripheral blood T-lymphocyte subset and which possible relationship with the clinical onsets during MS.
Collapse
Affiliation(s)
- Wen-bin Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | | | | | | | | | | |
Collapse
|
16
|
Penitente R, Nicolò C, Van den Elzen P, Di Sante G, Agrati C, Aloisi F, Sercarz EE, Ria F. Administration of PLP139–151 Primes T Cells Distinct from Those Spontaneously Responsive In Vitro to This Antigen. THE JOURNAL OF IMMUNOLOGY 2008; 180:6611-22. [DOI: 10.4049/jimmunol.180.10.6611] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Sommereyns C, Paul S, Staeheli P, Michiels T. IFN-lambda (IFN-lambda) is expressed in a tissue-dependent fashion and primarily acts on epithelial cells in vivo. PLoS Pathog 2008; 4:e1000017. [PMID: 18369468 PMCID: PMC2265414 DOI: 10.1371/journal.ppat.1000017] [Citation(s) in RCA: 643] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Accepted: 01/30/2008] [Indexed: 12/13/2022] Open
Abstract
Interferons (IFN) exert antiviral, immunomodulatory and cytostatic activities. IFN-alpha/beta (type I IFN) and IFN-lambda (type III IFN) bind distinct receptors, but regulate similar sets of genes and exhibit strikingly similar biological activities. We analyzed to what extent the IFN-alpha/beta and IFN-lambda systems overlap in vivo in terms of expression and response. We observed a certain degree of tissue specificity in the production of IFN-lambda. In the brain, IFN-alpha/beta was readily produced after infection with various RNA viruses, whereas expression of IFN-lambda was low in this organ. In the liver, virus infection induced the expression of both IFN-alpha/beta and IFN-lambda genes. Plasmid electrotransfer-mediated in vivo expression of individual IFN genes allowed the tissue and cell specificities of the responses to systemic IFN-alpha/beta and IFN-lambda to be compared. The response to IFN-lambda correlated with expression of the alpha subunit of the IFN-lambda receptor (IL-28R alpha). The IFN-lambda response was prominent in the stomach, intestine and lungs, but very low in the central nervous system and spleen. At the cellular level, the response to IFN-lambda in kidney and brain was restricted to epithelial cells. In contrast, the response to IFN-alpha/beta was observed in various cell types in these organs, and was most prominent in endothelial cells. Thus, the IFN-lambda system probably evolved to specifically protect epithelia. IFN-lambda might contribute to the prevention of viral invasion through skin and mucosal surfaces.
Collapse
Affiliation(s)
- Caroline Sommereyns
- Université catholique de Louvain, de Duve Institute, MIPA-VIRO 74-49, Brussels, Belgium
| | - Sophie Paul
- Université catholique de Louvain, de Duve Institute, MIPA-VIRO 74-49, Brussels, Belgium
| | - Peter Staeheli
- Department of Virology, University of Freiburg, Freiburg, Germany
| | - Thomas Michiels
- Université catholique de Louvain, de Duve Institute, MIPA-VIRO 74-49, Brussels, Belgium
| |
Collapse
|
18
|
Vaknin-Dembinsky A, Murugaiyan G, Hafler DA, Astier AL, Weiner HL. Increased IL-23 secretion and altered chemokine production by dendritic cells upon CD46 activation in patients with multiple sclerosis. J Neuroimmunol 2008; 195:140-5. [PMID: 18403025 PMCID: PMC2702859 DOI: 10.1016/j.jneuroim.2008.01.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Revised: 01/09/2008] [Accepted: 01/14/2008] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). In MS, myeloid dendritic cells (mDCs) secrete elevated amounts of IL-23, a potent proinflammatory cytokine, compared to healthy donors. Here, we examined the role of CD46, a complement binding factor, in mDCs by analyzing cytokine and chemokine production in healthy donors and patients with MS. There were striking differences between these groups with increased IL-23p19, CCL3 and CCL5 production, but decreased CCL2 levels in patients. This demonstrates major differences of DC activation upon CD46 activation, with a potential role in the pathogenesis of MS.
Collapse
Affiliation(s)
- Adi Vaknin-Dembinsky
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Gopal Murugaiyan
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - David A. Hafler
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Anne L. Astier
- Address reprint requests and correspondence to Howard Weiner or Anne Astier, Phone: (1) 617 525 5300 Fax: (1) 617 525 5252, E-mail address: or
| | - Howard L. Weiner
- Address reprint requests and correspondence to Howard Weiner or Anne Astier, Phone: (1) 617 525 5300 Fax: (1) 617 525 5252, E-mail address: or
| |
Collapse
|
19
|
Shahabi S, Rasmi Y, Jazani NH, Hassan ZM. Protective effects of Helicobacter pylori against gastroesophageal reflux disease may be due to a neuroimmunological anti-inflammatory mechanism. Immunol Cell Biol 2008; 86:175-178. [PMID: 17923849 DOI: 10.1038/sj.icb.7100119] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is some evidence that Helicobacter pylori infection has a protective effect against gastroesophageal reflux disease (GORD) and its complications such as Barrett's oesophagus and oesophageal adenocarcinoma. In this paper, we propose that a neuroimmunological mechanism is responsible for the protective effect of H. pylori on GORD. H. pylori infection of the gastric mucosa induces a T helper1-like immune response and production of pro-inflammatory cytokines. These cytokines can inhibit local sympathetic tone, whereas they increase systemic sympathetic tone. Increased sympathetic tone can induce an anti-inflammatory milieu, which in turn can inhibit inflammation in the oesophagus and lower oesophageal sphincter (LOS). Furthermore, H. pylori infection may stimulate the cholinergic anti-inflammatory pathway. It has been suggested that reflux-induced oesophageal inflammation plays an important role in the pathogenesis of reflux oesophagitis. Reduction of oesophageal inflammation by increased systemic sympathetic tone and vagal activity may lead to a decrease in reflux-induced oesophageal injury and LOS dysfunction in GORD.
Collapse
Affiliation(s)
- Shahram Shahabi
- Department of Microbiology, Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | | | | | | |
Collapse
|
20
|
Gorantla S, Liu J, Wang T, Holguin A, Sneller HM, Dou H, Kipnis J, Poluektova L, Gendelman HE. Modulation of innate immunity by copolymer-1 leads to neuroprotection in murine HIV-1 encephalitis. Glia 2008; 56:223-32. [PMID: 18046731 PMCID: PMC2734453 DOI: 10.1002/glia.20607] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Virus-infected and immune-competent mononuclear phagocytes (MP; perivascular macrophages and microglia) drive the neuropathogenesis of human immunodeficiency virus type 1 (HIV-1) infection. Modulation of the MP phenotype from neurodestructive to neuroprotective underlies adjunctive therapeutic strategies for human disease. We reasoned that, as Copolymer-1 (Cop-1) can induce neuroprotective activities in a number of neuroinflammatory and neurodegenerative disorders, it could directly modulate HIV-1-infected MP neurotoxic activities. We now demonstrate that, in laboratory assays, Cop-1-stimulated virus-infected human monocyte-derived macrophages (MDM) protect against neuronal injury. Severe combined immune-deficient (SCID) mice were stereotactically injected with HIV-1-infected human MDM, into the basal ganglia, to induce HIV-1 encephalitis (HIVE). Cop-1 was administered subcutaneously for 7 days. In HIVE mice, Cop-1 treatment led to anti-inflammatory and neuroprotective responses. Reduced micro- and astrogliosis, and conserved NeuN/MAP-2 levels were observed in virus-affected brain regions in Cop-1-treated mice. These were linked to interleukin-10 and brain-derived neurotrophic factor expression and downregulation of inducible nitric oxide synthase. The data, taken together, demonstrate that Cop-1 can modulate innate immunity and, as such, improve disease outcomes in an animal model of HIVE.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Embryo, Mammalian
- Encephalitis, Viral/etiology
- Encephalitis, Viral/pathology
- Encephalitis, Viral/prevention & control
- Gene Expression Regulation, Viral/drug effects
- Glatiramer Acetate
- Glial Fibrillary Acidic Protein/metabolism
- HIV Infections/complications
- Humans
- Immunity, Innate/drug effects
- Immunity, Innate/physiology
- Immunosuppressive Agents/pharmacology
- In Situ Nick-End Labeling
- Male
- Mice
- Mice, SCID
- Neuroglia/drug effects
- Neuroglia/metabolism
- Neuroglia/virology
- Neurons/drug effects
- Neurons/metabolism
- Neurons/virology
- Peptides/pharmacology
- Time Factors
Collapse
Affiliation(s)
- Santhi Gorantla
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Jianuo Liu
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Tong Wang
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Adelina Holguin
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Hannah M Sneller
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Huanyu Dou
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Jonathan Kipnis
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Larisa Poluektova
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Howard E Gendelman
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5880
| |
Collapse
|
21
|
Gorantla S, Liu J, Sneller H, Dou H, Holguin A, Smith L, Ikezu T, Volsky DJ, Poluektova L, Gendelman HE. Copolymer-1 induces adaptive immune anti-inflammatory glial and neuroprotective responses in a murine model of HIV-1 encephalitis. THE JOURNAL OF IMMUNOLOGY 2007; 179:4345-56. [PMID: 17878329 DOI: 10.4049/jimmunol.179.7.4345] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Copolymer-1 (COP-1) elicits neuroprotective activities in a wide range of neurodegenerative disorders. This occurs, in part, by adaptive immune-mediated suppression of microglial inflammatory responses. Because HIV infection and immune activation of perivascular macrophages and microglia drive a metabolic encephalopathy, we reasoned that COP-1 could be developed as an adjunctive therapy for disease. To test this, we developed a novel animal model system that reflects HIV-1 encephalitis in rodents with both innate and adaptive arms of the immune system. Bone marrow-derived macrophages were infected with HIV-1/vesicular stomatitis-pseudotyped virus and stereotactically injected into the basal ganglia of syngeneic mice. HIV-1 pseudotyped with vesicular stomatitis virus envelope-infected bone marrow-derived macrophages induced significant neuroinflammation, including astrogliosis and microglial activation with subsequent neuronal damage. Importantly, COP-1 immunization reduced astro- and microgliosis while diminishing neurodegeneration. Hippocampal neurogenesis was, in part, restored. This paralleled reductions in proinflammatory cytokines, including TNF-alpha and IL-1beta, and inducible NO synthase, and increases in brain-derived neurotrophic factor. Ingress of Foxp3- and IL-4-expressing lymphocytes into brains of COP-1-immunized animals was observed. We conclude that COP-1 may warrant therapeutic consideration for HIV-1-associated cognitive impairments.
Collapse
Affiliation(s)
- Santhi Gorantla
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). In the recent years, accumulating evidence has supported an immunosuppressive role for regulatory T cells (Tregs). Most studies in the context of autoimmunity have focused on the defects of the CD4+CD25 high Tregs. However, we recently demonstrated an altered function of Tr1 Treg cells in MS, characterized by a lack of IL-10 secretion. Therefore, several major regulatory T cell defects are involved in human autoimmune disease. Hence, the induction of Tregs or the stimulation of Treg activity may be beneficial for the treatment of such diseases.
Collapse
Affiliation(s)
- Anne L Astier
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
23
|
Ke Y, Sun D, Zhang P, Jiang G, Kaplan HJ, Shao H. Suppression of established experimental autoimmune uveitis by anti-LFA-1alpha Ab. Invest Ophthalmol Vis Sci 2007; 48:2667-75. [PMID: 17525198 PMCID: PMC2587054 DOI: 10.1167/iovs.06-1383] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE To identify costimulatory molecules that are important in the effector phase of experimental autoimmune uveitis (EAU). METHODS EAU was induced in C57BL/6 (B6) mice by transfer of activated T cells specific for the interphotoreceptor-binding protein (IRBP) 1-20 peptide. The animals were then treated with and without anti-leukocyte function associated antigen (LFA)-1alpha mAb, at day 0 or 10 (disease onset) after T-cell transfer. Clinical signs of inflammation, ocular histology, and infiltrated inflammatory cells in the eye were compared. The primary and secondary proliferative responses of uveitogenic CD4 and CD8 T cells were tested after treatment with costimulatory blockers in vivo and in vitro. Moreover, the abilities of uveitogenic T cell trafficking and their interaction with retinal astrocytes were examined. RESULTS Anti-LFA-1alpha Abs caused significant suppression of disease when administered either at the time of effector uveitogenic T cell transfer or at disease onset. Studies of the mechanisms by which anti-LFA-1alpha Ab inhibits the effector phase of uveitis demonstrated that it blocks multiple pathogenic events of uveitis mediated by IRBP-specific uveitogenic T cells, including the activation of T cells outside and inside the eye and the trafficking of activated autoreactive T cells into the inflammatory site. In addition, Ab treatment selectively suppressed the activation and expansion of pathogenic, but not regulatory, T cells in vivo. CONCLUSIONS Anti-LFA-1alpha Abs are potent inhibitors of established autoimmune uveitis and that such treatment may be applicable not only for the prevention, but also the treatment, of T-cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Yan Ke
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | | | | | | | |
Collapse
|
24
|
Paul S, Ricour C, Sommereyns C, Sorgeloos F, Michiels T. Type I interferon response in the central nervous system. Biochimie 2007; 89:770-8. [PMID: 17408841 DOI: 10.1016/j.biochi.2007.02.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Accepted: 02/16/2007] [Indexed: 12/25/2022]
Abstract
This review is dedicated to the influence of type I IFNs (also called IFN-alpha/beta) in the central nervous system (CNS). Studies in mice with type I IFN receptor or IFN-beta gene deficiency have highlighted the importance of the type I IFN system against CNS viral infections and non-viral autoimmune disorders. Direct antiviral effects of type I IFNs appear to be crucial in limiting early spread of a number of viruses in CNS tissues. Type I IFNs have also proved to be beneficial in autoimmune disorders like multiple sclerosis or experimental autoimmune encephalitis, probably through immunomodulatory effects. Increasing efforts are done to characterize IFN expression and response in the CNS: to identify type I IFN producing cells, to decipher pathways leading to type I IFN expression in those cells, and to identify responding cells. However, reversible and irreversible damages consecutive to chronic exposure of the CNS to type I IFNs underline the importance of a tightly regulated type I IFN homeostasis in this organ.
Collapse
Affiliation(s)
- Sophie Paul
- Université catholique de Louvain, Christian de Duve Institute of Cellular Pathology, Microbial Pathogenesis Unit, MIPA-VIRO 74-49, 74, avenue Hippocrate, B-1200, Brussels, Belgium
| | | | | | | | | |
Collapse
|
25
|
Chen W, Li H, Jia Y, Lv M, Li M, Feng P, Hu H, Zhang L. In vivo administration of plasmid DNA encoding recombinant immunotoxin DT390-IP-10 attenuates experimental autoimmune encephalomyelitis. J Autoimmun 2007; 28:30-40. [PMID: 17267177 DOI: 10.1016/j.jaut.2006.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2006] [Revised: 10/17/2006] [Accepted: 11/01/2006] [Indexed: 02/07/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a T-cell-mediated autoimmune demyelinating disease. The expression of chemokine receptor CXCR3 on activated T cells is crucial to direct the migration of effector cells into the inflammatory sites and initiate EAE. In this study we tested the effect of a novel recombinant immunotoxin targeting CXCR3(+) cells for EAE prevention. The immunotoxin construct DT390-IP-10-SRalpha consisted of interferon gamma-inducible protein 10 (IP-10), a ligand of CXCR3, as the targeting moiety, and a truncated diphtheria toxin (DT390) as the toxic moiety. In vitro transfection of DT390-IP-10-SRalpha into NIH3T3 cells resulted in expression of DT390-IP-10 which proved highly toxic to activated T cells. To evaluate the effect of DT390-IP-10-SRalpha on EAE prevention in vivo, cationic liposome-embedded DT390-IP-10-SRalpha was injected into the muscle of hind limbs of C57BL/6 mice immunized by myelin basic protein (MBP). DT390-IP-10-SRalpha-treated mice showed a delayed onset of EAE and milder symptoms compared to the mice treated with empty control plasmid or PBS alone. Immunohistochemical staining detected significantly reduced infiltrating CXCR3(+) cells in the inflammatory lesions of CNS from immunotoxin treated mice as compared to the controls. This study suggests that targeting CXCR3(+) T cells with recombinant immunotoxin could be achieved in vivo to delay and ameliorate murine EAE.
Collapse
MESH Headings
- Animals
- Chemokine CXCL10
- Chemokines, CXC/administration & dosage
- Chemokines, CXC/genetics
- Chemokines, CXC/immunology
- DNA/administration & dosage
- DNA/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Female
- Genetic Therapy/methods
- Immunotoxins/administration & dosage
- Immunotoxins/genetics
- Immunotoxins/immunology
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- NIH 3T3 Cells
- Plasmids/administration & dosage
- Plasmids/biosynthesis
- Plasmids/genetics
- Receptors, CXCR3
- Receptors, Chemokine/antagonists & inhibitors
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/immunology
- T-Lymphocytes/immunology
- Transfection
Collapse
Affiliation(s)
- Wenjie Chen
- West China School of Preclinical and Forensic Medicine, Sichuan University, Number 17, Section , Renmin Nan Road, Chengdu, Sichuan 610041, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Carter LL, Leach MW, Azoitei ML, Cui J, Pelker JW, Jussif J, Benoit S, Ireland G, Luxenberg D, Askew GR, Milarski KL, Groves C, Brown T, Carito BA, Percival K, Carreno BM, Collins M, Marusic S. PD-1/PD-L1, but not PD-1/PD-L2, interactions regulate the severity of experimental autoimmune encephalomyelitis. J Neuroimmunol 2007; 182:124-34. [PMID: 17182110 DOI: 10.1016/j.jneuroim.2006.10.006] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2006] [Revised: 10/02/2006] [Accepted: 10/11/2006] [Indexed: 01/10/2023]
Abstract
Interactions between PD-1 and its two differentially expressed ligands, PD-L1 and PD-L2, attenuate T cell activation and effector function. To determine the role of these molecules in autoimmune disease of the CNS, PD-1-/-, PD-L1-/- and PD-L2-/- mice were generated and immunized to induce experimental autoimmune encephalomyelitis (EAE). PD-1-/- and PD-L1-/- mice developed more severe EAE than wild type and PD-L2-/- mice. Consistent with this, PD-1-/- and PD-L1-/- cells produced elevated levels of the pro-inflammatory cytokines IFN-gamma, TNF, IL-6 and IL-17. These results demonstrate that interactions between PD-1/PD-L1, but not PD-1/PDL-2, are crucial in attenuating T cell responses in EAE.
Collapse
MESH Headings
- Animals
- Antigens, Differentiation/metabolism
- B7-1 Antigen/metabolism
- B7-H1 Antigen
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Glycoproteins/immunology
- Humans
- Interferon-gamma/biosynthesis
- Interleukin-17/biosynthesis
- Interleukin-6/biosynthesis
- Lymph Nodes/metabolism
- Lymph Nodes/pathology
- Lymphocyte Activation
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Knockout
- Mice, Transgenic
- Myelin-Oligodendrocyte Glycoprotein
- Peptide Fragments/immunology
- Peptides/deficiency
- Peptides/metabolism
- Programmed Cell Death 1 Ligand 2 Protein
- Programmed Cell Death 1 Receptor
- Severity of Illness Index
- T-Lymphocytes/immunology
- Tumor Necrosis Factor-alpha/biosynthesis
Collapse
Affiliation(s)
- Laura L Carter
- Inflammation, Wyeth Research, 200 CambridgePark Dr., Cambridge, MA 02140, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Trifari S, Sitia G, Aiuti A, Scaramuzza S, Marangoni F, Guidotti LG, Martino S, Saracco P, Notarangelo LD, Roncarolo MG, Dupré L. Defective Th1 cytokine gene transcription in CD4+ and CD8+ T cells from Wiskott-Aldrich syndrome patients. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:7451-61. [PMID: 17082665 DOI: 10.4049/jimmunol.177.10.7451] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Wiskott-Aldrich syndrome (WAS) protein (WASP) plays a key role in TCR-mediated activation and immunological synapse formation. However, the effects of WASP deficiency on effector functions of human CD4+ and CD8+ T cells remain to be determined. In this study, we report that TCR/CD28-driven proliferation and secretion of IL-2, IFN-gamma, and TNF-alpha are strongly reduced in CD8+ T cells from WAS patients, compared with healthy donor CD8+ T cells. Furthermore, WAS CD4+ T cells secrete low levels of IL-2 and fail to produce IFN-gamma and TNF-alpha, while the production of IL-4, IL-5, and IL-10 is only minimally affected. Defective IL-2 and IFN-gamma production persists after culture of naive WAS CD4+ T cells in Th1-polarizing conditions. The defect in Th1 cytokine production by WAS CD4+ and CD8+ T cells is also present at the transcriptional level, as shown by reduced IL-2 and IFN-gamma mRNA transcripts after TCR/CD28 triggering. The reduced transcription of Th1 cytokine genes in WAS CD4+ T cells is associated with a defective induction of T-bet mRNA and a reduction in the early nuclear recruitment of NFAT-1, while the defective activation of WAS CD8+ T cells correlates with reduced nuclear recruitment of both NFAT-1 and NFAT-2. Together, our data indicate that WASP regulates the transcriptional activation of T cells and is required specifically for Th1 cytokine production.
Collapse
Affiliation(s)
- Sara Trifari
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Astier AL, Meiffren G, Freeman S, Hafler DA. Alterations in CD46-mediated Tr1 regulatory T cells in patients with multiple sclerosis. J Clin Invest 2006; 116:3252-7. [PMID: 17099776 PMCID: PMC1635165 DOI: 10.1172/jci29251] [Citation(s) in RCA: 228] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Accepted: 09/19/2006] [Indexed: 12/20/2022] Open
Abstract
Loss of Treg function appears to be a critical factor in the pathogenesis of human autoimmune diseases. Attention has focused on defects of CD4(+)CD25(high) Tregs, and techniques have been developed to determine their function. In contrast, the role of Tr1 regulatory T cells, which secrete the antiinflammatory cytokine IL-10, in autoimmune disease has not been well assessed. CD46 is a newly defined costimulatory molecule for T cell activation, and CD46-costimulated human T cells induce a Tr1 Treg phenotype with considerable amounts of IL-10 secretion. Here, we examined the role of Tr1 cells in patients with multiple sclerosis (MS) by stimulating CD4(+) T cells with anti-CD3 and -CD46 mAbs and measuring IL-10 secretion. There were striking defects in the induction of Tr1 cells with CD46 costimulation as measured by IL-10 but not IFN-gamma secretion in patients with MS compared with healthy subjects. This loss of Tr1 cell-associated IL-10 secretion was specific to CD46 and not CD28 costimulation and was associated with an altered regulation of the CD46-Cy2 isoform that differentially regulates T cell function in a CD46-transgenic murine model. These data demonstrate a second major Treg defect in human autoimmune disease associated with the CD46 pathway.
Collapse
Affiliation(s)
- Anne L. Astier
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
INSERM U503, Laboratory of Fundamental and Clinical Immunobiology, Lyon, France
| | - Gregory Meiffren
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
INSERM U503, Laboratory of Fundamental and Clinical Immunobiology, Lyon, France
| | - Samuel Freeman
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
INSERM U503, Laboratory of Fundamental and Clinical Immunobiology, Lyon, France
| | - David A. Hafler
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
INSERM U503, Laboratory of Fundamental and Clinical Immunobiology, Lyon, France
| |
Collapse
|
29
|
Mirshafiey A, Matsuo H, Nakane S, Rehm BHA, Koh CS, Miyoshi S. Novel immunosuppressive therapy by M2000 in experimental multiple sclerosis. Immunopharmacol Immunotoxicol 2006; 27:255-65. [PMID: 16114509 DOI: 10.1081/iph-200067751] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The therapeutic potency of M2000 (beta-D-mannuronic acid), a novel designed nonsteroidal anti-inflammatory drug with immunosuppressive property in T-cell-mediated autoimmune disease, was tested. The influence of M2000 on myelin basic protein (MBP)-induced experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, was assessed. M2000 at two doses, 40 and 80 mg/kg/day, was administered intraperitoneally (i.p.) to prevention and treatment groups, respectively. Onset of i.p. injections of M2000 to prophylactic and therapeutic groups was day-1 and day-7 postimmunization. The WEHI-164 cell line was used for assaying the tolerability against M2000. The results of this experiment showed that the treatment of EAE with M2000 could significantly suppress disease development both prophylactically and therapeutically; the onset and symptoms of EAE in Lewis rats could be suppressed following the administration of M2000. Clinical improvement was accompanied by a marked decrease in mean numbers of vessels with perivascular cellular infiltration in M2000-treated rats compared with nontreated control. Disease suppression was associated with a marked suppression of MBP-specific T-cell reactivity in vitro, without any evidence for a generalized impairment of T-cell activity. Moreover, M2000 also showed a very high tolerability compared with certain steroidal and nonsteroidal anti-inflammatory drugs. Collectively, our data suggest that M2000 may provide a novel therapeutic option for T-cell-mediated autoimmune diseases in animal models and possibly in humans.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Cell Line, Tumor/drug effects
- Cell Proliferation
- Dose-Response Relationship, Immunologic
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Female
- Hexuronic Acids/administration & dosage
- Hexuronic Acids/pharmacology
- Immunization
- Immunosuppressive Agents/pharmacology
- Lymph Nodes/cytology
- Lymph Nodes/drug effects
- Multiple Sclerosis/drug therapy
- Mycobacterium tuberculosis
- Myelin Basic Protein
- Rats
- Rats, Inbred Lew
- Time Factors
Collapse
Affiliation(s)
- Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | | | |
Collapse
|
30
|
Franco A, Albani S. Translating the concept of suppressor/regulatory T cells to clinical applications. Int Rev Immunol 2006; 25:27-47. [PMID: 16669133 DOI: 10.1080/08830180500544506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The in vivo expansion of suppressor/regulatory T cells (Tregs) is a desirable event in autoimmunity and transplantation. Here we summarize the general rules involved in antigen recognition by T cells and describe Tregs and their requirements, discussing different levels of immune intervention.
Collapse
Affiliation(s)
- Alessandra Franco
- Department of Medicine and Pediatrics, University of California, San Diego, La Jolla, California 92093-0731, USA
| | | |
Collapse
|
31
|
Dubucquoi S. [Physiological and pathological autoimmunity in neurological disease]. Rev Neurol (Paris) 2005; 161:1163-76. [PMID: 16340911 DOI: 10.1016/s0035-3787(05)85189-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Over the last years, the functions of the immune system have radically been revised. It has been illustrated how the different responses of the innate defense and the adaptative system were intimately intricated. STATE OF KNOWLEDGE Innate immunity is an elaborate strategy, switched on by an invariant receptor-based response that is able to develop a specific defense against some pathogens. Moreover, this innate immunity governs T- and B-cell-dependent adaptive immune response, mediated via dendritic cells whose maturation is controlled by immune specificity. The concepts of autoreactivity have also strongly progressed and the functions of the physiological autoimmune response have been highlighted. The notion of T- and B-cell self-antigens not only shapes the immune repertoire, but also the self-recognition process which is a tool for the control of the immune response itself. PERSPECTIVE New concepts of the pathophysiology of autoimmune diseases have emerged from a better understanding of the immune response, balancing between an intrinsic deregulation of the immune system and system deficiency to mount an effective response against an initial injury. CONCLUSION Of course, therapeutic strategies are challenged by these data. We should expect that control of several autoimmune processes will be achieved in a few years, e.g. in rheumatoid arthritis or Crohn's disease controlled with biotherapies.
Collapse
Affiliation(s)
- S Dubucquoi
- Laboratoire d'Immunologie du CHRU de Lille, Pôle Biologie Pathologie, Parc Eurasante, 152, avenue du Docteur-Yersin, 59120 Loos.
| |
Collapse
|