101
|
Petereit HF, Reske D, Pukrop R, Maas-Enriquez M, Japp G, Jongen PJH, Kölmel HW, Merkelbach S, Hartung HP, Heiss WD, Hommes OR. No effect of intravenous immunoglobulins on cytokine-producing lymphocytes in secondary progressive multiple sclerosis. Mult Scler 2016; 12:66-71. [PMID: 16459721 DOI: 10.1191/135248506ms1246oa] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Intravenous immunoglobulins (IVIG) have been effective in reducing multiple sclerosis (MS) disease activity and improving disability scores. However, the mechanism by which this beneficial effect is achieved remains unclear. An effect of IVIG on pro- and anti-inflammatory cytokines- which are thought to play a role in the disease process- has been postulated in a number of animal and ex vivo studies. Hence, we performed a study on 34 patients with secondary progressive (SP) MS being treated with monthly IVIG or placebo for two years according to the protocol of the ESIMS study. Clinical outcome measures and cytokine production (interferon gamma, tumour necrosis factor alpha, interleukin-4 and -10) were recorded in all patients and compared with respect to the treatment group. Against our expectations, IVIG did not reduce the relapse rate or the progression of disability or cytokine production. Our data argue against an enduring immunomodulating effect of IVIG, at least in SPMS.
Collapse
Affiliation(s)
- H F Petereit
- Department of Neurology and Psychiatry, University of Cologne, Josef-Stelzmann-Str. 9, 50924 Kö1n, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Bergkvist M, Olsson M, Sandberg-Wollheim M. No evidence for genetic linkage between development of multiple sclerosis and components of the IFN system and the JA K-STAT pathway. Mult Scler 2016; 10:87-8. [PMID: 14760959 DOI: 10.1191/1352458504ms976sr] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating disease of the central nervous system (CNS). Several observations suggest that the interferon system may be of interest in the study of MS development. To investigate whether polymorphism in components of the IFN system and the JA K-STAT pathway influence susceptibility to MS, we performed a linkage analysis between polymorphic loci in or close to the IFN gamma, IFN gamma recepto r, IFN alpha/beta recepto r, JA K 1, STAT 1 and STAT 3 genes in 27 Swedish families with at least two members having MS. Tests for transmission disequilibrium and nonparametric linkage analysis gave negative results. We found no evidence for linkage between MS and any of these loci.
Collapse
Affiliation(s)
- M Bergkvist
- Department of Neurology, University Hospital, SE-221 85 Lund, Sweden.
| | | | | |
Collapse
|
103
|
Wang Q, Meng J, Dong A, Yu JZ, Zhang GX, Ma CG. The Pharmacological Effects and Mechanism ofTripterygium wilfordiiHook F in Central Nervous System Autoimmunity. J Altern Complement Med 2016; 22:496-502. [DOI: 10.1089/acm.2016.0004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Qing Wang
- 2011 Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Shanxi Province, People's Republic of China
| | - Jian Meng
- Institute of Brain Science, Shanxi Datong University, Datong, People's Republic of China
| | - Aiguo Dong
- 2011 Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Shanxi Province, People's Republic of China
| | - Jie-zhong Yu
- Institute of Brain Science, Shanxi Datong University, Datong, People's Republic of China
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA
| | - Cun-Gen Ma
- 2011 Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Shanxi Province, People's Republic of China
- Institute of Brain Science, Shanxi Datong University, Datong, People's Republic of China
| |
Collapse
|
104
|
Abstract
Until recently, the treatment of multiple sclerosis (MS) was restricted to symptomatic therapies. Advances in our understanding of the pathogenesis of MS are now resulting in the rapid proliferation of treatment strategies to slow or stop the progression of this disease. Clearly, immunological therapies can improve outcomes in MS and offer hope that this crippling disease can be controlled before patients develop major neurological disabilities. Immunological therapies under investigation for the treatment of MS are taking advantage of dramatic improvements in our understanding of immunoregulation. In addition, immunological treatment of MS is becoming selective relative to myelin antigens, enhancing efficacy and reducing toxicity. The Neuroscientist 2:127-136, 1996
Collapse
Affiliation(s)
- Timothy L. Vollmer
- Department of Neurology Yale University School of Medicine
New Haven, Connecticut
| |
Collapse
|
105
|
Totaro R, Passacantando A, Russo T, Parzanese I, Rascente M, Marini C, Tonietti G, Carolei A. Effects of Interferon Beta, Cyclophosphamide and Azathioprine on Cytokine Profile in Patients with Multiple Sclerosis. Int J Immunopathol Pharmacol 2016; 18:377-83. [PMID: 15888259 DOI: 10.1177/039463200501800219] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We assessed the in vitro effects of interferon beta-1b (IFNβ-1b), cyclophosphamide (CY), and azathioprine (AZA) alone and of the combination of IFNβ-1b with CY or AZA on the production of Th1 and Th2 cytokines in 10 patients with multiple sclerosis. Cytokine levels were determined at baseline and after stimulation with IFNβ-1b, CY, and AZA alone or with the combination of IFNβ-1b with CY or AZA. The combination of IFNβ-1b with CY resulted in a statistically significant decrease in the production of interleukin-2 (IL-2) (P=0.003) and tumor necrosis factor alfa (TNF-α) (P=0.03). An additive effect on the production of interferon gamma (IFN-γ) (P=0.2) and interleukin-10 (IL-10) (P=0.6), and a positive interaction on the production of interleukin-4 (IL-4) (P=0.08) were observed although the findings were not statistically significant. The combination of IFNβ-1b with AZA resulted in a significant negative effect on the production of IL-2 (P=0.006), whereas TNF-α (P=0.02), IFN-γ (P=0.03), IL-4 (P=0.2), and IL-10 (P=0.3) were not statistically impacted. Our data show that CY was able to improve the effects of IFNβ-1b on the ratio of Th1/Th2 cytokines.
Collapse
Affiliation(s)
- R Totaro
- Department of Neurology, University of L'Aquila, L'Aquila, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
106
|
Association between seasonal factors and multiple sclerosis. Eur J Epidemiol 2016; 31:1081-1089. [DOI: 10.1007/s10654-016-0165-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/18/2016] [Indexed: 02/06/2023]
|
107
|
Brown KA, Brown GA, Lewis SM, Beale R, Treacher DF. Targeting cytokines as a treatment for patients with sepsis: A lost cause or a strategy still worthy of pursuit? Int Immunopharmacol 2016; 36:291-299. [PMID: 27208433 DOI: 10.1016/j.intimp.2016.04.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 04/26/2016] [Indexed: 12/25/2022]
Abstract
Despite often knowing the aetiology of sepsis and its clinical course there has not been the anticipated advances in treatment strategies. Cytokines are influential mediators of immune/inflammatory reactions and in patients with sepsis high circulating levels are implicated in the onset and perpetuation of organ failure. Antagonising the activities of pro-inflammatory cytokines enhances survival in animal models of sepsis but, so far, such a therapeutic strategy has not improved patient outcome. This article addresses the questions of why encouraging laboratory findings have failed to be translated into successful treatments of critically ill patients and whether modifying cytokine activity still remains a promising avenue for therapeutic advance in severe sepsis. In pursuing this task we have selected reports that we believe provide an incisive, critical and balanced view of the topic.
Collapse
Affiliation(s)
- K Alun Brown
- Intensive Care Unit, Guy's and St.Thomas' Hospitals, London, UK; Division of Asthma Allergy and Lung Biology, King's College London, UK.
| | | | - Sion M Lewis
- Intensive Care Unit, Guy's and St.Thomas' Hospitals, London, UK; Division of Asthma Allergy and Lung Biology, King's College London, UK
| | - Richard Beale
- Intensive Care Unit, Guy's and St.Thomas' Hospitals, London, UK; Division of Asthma Allergy and Lung Biology, King's College London, UK
| | - David F Treacher
- Intensive Care Unit, Guy's and St.Thomas' Hospitals, London, UK; Division of Asthma Allergy and Lung Biology, King's College London, UK
| |
Collapse
|
108
|
NEAGU MONICA, CARUNTU CONSTANTIN, CONSTANTIN CAROLINA, BODA DANIEL, ZURAC SABINA, SPANDIDOS DEMETRIOSA, TSATSAKIS ARISTIDISM. Chemically induced skin carcinogenesis: Updates in experimental models (Review). Oncol Rep 2016; 35:2516-28. [PMID: 26986013 PMCID: PMC4811393 DOI: 10.3892/or.2016.4683] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/16/2016] [Indexed: 02/06/2023] Open
Abstract
Skin cancer is one of the most common malignancies affecting humans worldwide, and its incidence is rapidly increasing. The study of skin carcinogenesis is of major interest for both scientific research and clinical practice and the use of in vivo systems may facilitate the investigation of early alterations in the skin and of the mechanisms involved, and may also lead to the development of novel therapeutic strategies for skin cancer. This review outlines several aspects regarding the skin toxicity testing domain in mouse models of chemically induced skin carcinogenesis. There are important strain differences in view of the histological type, development and clinical evolution of the skin tumor, differences reported decades ago and confirmed by our hands‑on experience. Using mouse models in preclinical testing is important due to the fact that, at the molecular level, common mechanisms with human cutaneous tumorigenesis are depicted. These animal models resemble human skin cancer development, in that genetic changes caused by carcinogens and pro‑inflammatory cytokines, and simultaneous inflammation sustained by pro‑inflammatory cytokines and chemokines favor tumor progression. Drugs and environmental conditions can be tested using these animal models. keeping in mind the differences between human and rodent skin physiology.
Collapse
Affiliation(s)
- MONICA NEAGU
- 'Victor Babes' National Institute of Pathology, Bucharest 050096, Romania
- Faculty of Biology, University of Bucharest, Bucharest 76201, Romania
| | - CONSTANTIN CARUNTU
- Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Dermatology, 'Prof. N. Paulescu' National Institute of Diabetes, Nutrition and Metabolic Diseases, Bucharest 79811, Romania
| | | | - DANIEL BODA
- Department of Dermatology, 'Prof. N. Paulescu' National Institute of Diabetes, Nutrition and Metabolic Diseases, Bucharest 79811, Romania
| | - SABINA ZURAC
- Department of Pathology, 'Colentina' Clinical Hospital, Bucharest 72202, Romania
| | - DEMETRIOS A. SPANDIDOS
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion 71409, Greece
| | - ARISTIDIS M. TSATSAKIS
- Department of Forensic Sciences and Toxicology, Medical School, University of Crete, Heraklion 71003, Greece
| |
Collapse
|
109
|
Wang D, Lu Z, Zhang H, Jin SF, Yang H, Li YM, Shi LY. Daphnetin Alleviates Experimental Autoimmune Encephalomyelitis via Regulating Dendritic Cell Activity. CNS Neurosci Ther 2016; 22:558-67. [PMID: 27013083 DOI: 10.1111/cns.12537] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/16/2016] [Accepted: 02/18/2016] [Indexed: 12/13/2022] Open
Abstract
AIMS Daphnetin, a coumarin derivative extracted from Daphne odora var. marginata, has been reported to have antiinflammatory and immunosuppressive properties. Our previous study indicated that it was able to remarkably suppress the neuroinflammation and suggested its potential application in treating neuroinflammatory diseases. Multiple sclerosis (MS), a Th cell-mediated autoimmune disease, is the most common inflammatory demyelinating disease of the central nervous system (CNS). We examined whether daphnetin treatment can protect mice against experimental autoimmune encephalomyelitis (EAE), an animal model for MS. METHODS To assess the effect of daphnetin in neuroinflammatory diseases, the EAE mice were established and treated with daphnetin at 8 mg/kg for 28 days. The severity of neuroinflammation and demyelination in the spinal cords was examined histopathologically. Infiltration of CD4(+) T cells into the CNS was assessed by immunohistochemistry, and the cytokine production was determined by ELISA. Meanwhile, the effect of daphnetin on the activity of dendritic cells (DCs) was evaluated, as assessed by DCs' capability to express surface markers, secrete cytokines, and activate naïve CD4(+) T cells. Furthermore, we explored the molecular mechanisms whereby DAPH regulated DCs' activity and thereby CD4(+) T cell responses. RESULTS The administration of daphnetin markedly alleviated the clinical symptoms of EAE and reduced the CNS inflammation and demyelination in experimental mice. Th1 and Th17 cell responses were profoundly repressed in daphnetin-treated EAE mice. Mechanistically, daphnetin treatment significantly repressed the activation, maturation, and antigen-presenting capability of DCs. NF-κB signaling was significantly reduced in daphnetin-treated DCs, along with a concomitant induction of heme oxygenase-1, a negative regulator of inflammatory signaling. CONCLUSIONS Our findings for the first time demonstrate the property of daphnetin in regulating DCs' function and subsequently Th development. Given the low or absent toxicity associated with daphnetin, our data may suggest a novel safe and effective approach to control autoimmune neuroinflammation.
Collapse
Affiliation(s)
- Dan Wang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, China.,Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Zhe Lu
- Department of Basic Medical Science, Key Lab of Immunology and Molecular Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Hang Zhang
- Department of Basic Medical Science, Key Lab of Immunology and Molecular Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Su-Feng Jin
- Department of Basic Medical Science, Key Lab of Immunology and Molecular Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Hao Yang
- Department of Basic Medical Science, Key Lab of Immunology and Molecular Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Yun-Man Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, China
| | - Li-Yun Shi
- Department of Basic Medical Science, Key Lab of Immunology and Molecular Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, China.,Department of Microbiology and Immunology, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
110
|
't Hart BA, Dunham J, Jagessar SA, Kap YS. The common marmoset (<i>Callithrix jacchus</i>): a relevant preclinical model of human (auto)immune-mediated inflammatory disease of the brain. Primate Biol 2016. [DOI: 10.5194/pb-3-9-2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Abstract. The increasing prevalence of chronic autoimmune-mediated inflammatory disorders (AIMIDs) in aging human populations creates a high unmet need for safe and effective medications. However, thus far the translation of pathogenic concepts developed in animal models into effective treatments for the patient has been notoriously difficult. The main reason is that currently used mouse-based animal models for the pipeline selection of promising new treatments were insufficiently predictive for clinical success. Regarding the high immunological similarity between human and non-human primates (NHPs), AIMID models in NHPs can help to bridge the translational gap between rodent and man. Here we will review the preclinical relevance of the experimental autoimmune encephalomyelitis (EAE) model in common marmosets (Callithrix jacchus), a small-bodied neotropical primate. EAE is a generic AIMID model projected on the human autoimmune neuro-inflammatory disease multiple sclerosis (MS).
Collapse
|
111
|
Xia XJ, Gao YY, Zhang J, Wang L, Zhao S, Che YY, Ao CJ, Yang HJ, Wang JQ, Lei LC. Autophagy mediated by arginine depletion activation of the nutrient sensor GCN2 contributes to interferon-γ-induced malignant transformation of primary bovine mammary epithelial cells. Cell Death Discov 2016; 2:15065. [PMID: 27551491 PMCID: PMC4979444 DOI: 10.1038/cddiscovery.2015.65] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/19/2015] [Accepted: 11/22/2015] [Indexed: 12/26/2022] Open
Abstract
Autophagy has been linked to the regulation of both the prevention and progression of cancer. IFN-γ has been shown to induce autophagy in multiple cell lines in vitro. However, whether IFN-γ can induce autophagy and whether autophagy promotes malignant transformation in healthy lactating bovine mammary epithelial cells (BMECs) remain unclear. Here, we provide the first evidence of the correlation between IFN-γ treatment, autophagy and malignant transformation and of the mechanism underlying IFN-γ-induced autophagy and subsequent malignant transformation in primary BMECs. IFN-γ levels were significantly increased in cattle that received normal long-term dietary corn straw (CS) roughage supplementation. In addition, an increase in autophagy was clearly observed in the BMECs from the mammary tissue of cows expressing high levels of IFN-γ. In vitro, autophagy was clearly induced in primary BMECs by IFN-γ within 24 h. This induced autophagy could subsequently promote dramatic primary BMEC transformation. Furthermore, we found that IFN-γ promoted arginine depletion, activated the general control nonderepressible-2 kinase (GCN2) signalling pathway and resulted in an increase in autophagic flux and the amount of autophagy in BMECs. Overall, our findings are the first to demonstrate that arginine depletion and kinase GCN2 expression mediate IFN-γ-induced autophagy that may promote malignant progression and that immunometabolism, autophagy and cancer are strongly correlated. These results suggest new directions and paths for preventing and treating breast cancer in relation to diet.
Collapse
Affiliation(s)
- X-J Xia
- College of Veterinary Medicine, Jilin University , Changchun, PR China
| | - Y-Y Gao
- College of Animal Science, Jilin University , Changchun, PR China
| | - J Zhang
- Changchun University of Chinese Medicine , Changchun, PR China
| | - L Wang
- College of Animal Science, Henan Institute of Science and Technology , Xinxiang, PR China
| | - S Zhao
- College of Animal Science, Jilin University , Changchun, PR China
| | - Y-Y Che
- College of Veterinary Medicine, Jilin University , Changchun, PR China
| | - C-J Ao
- College of Animal Science, Inner Mongolian Agricultural University , Hohhot, PR China
| | - H-J Yang
- College of Animal Science and Technology, China Agricultural University , Beijing, PR China
| | - J-Q Wang
- Institute of Animal Science, Chinese Academy of Agricultural Science , Beijing, PR China
| | - L-C Lei
- College of Veterinary Medicine, Jilin University , Changchun, PR China
| |
Collapse
|
112
|
Rasenack M, Rychen J, Andelova M, Naegelin Y, Stippich C, Kappos L, Lindberg RLP, Sprenger T, Derfuss T. Efficacy and Safety of Fingolimod in an Unselected Patient Population. PLoS One 2016; 11:e0146190. [PMID: 26734938 PMCID: PMC4703383 DOI: 10.1371/journal.pone.0146190] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 12/13/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Fingolimod is a first in class oral compound approved for the treatment of relapsing-remitting multiple sclerosis (RR-MS). The aim of this study was to evaluate clinical and neuroradiological responses to fingolimod as well as the safety and tolerability in RR-MS patients in clinical practice. In addition, a panel of pro-inflammatory serum cytokines was explored as potential biomarker for treatment response. METHODS We conducted a retrospective, non-randomized, open-label, observational study in 105 patients with RR-MS and measured cytokines in longitudinal serum samples. RESULTS Compared to the year before fingolimod start the annualized relapse rate was reduced by 44%. Also, the percentage of patients with a worsening of the EDSS decreased. Accordingly, the fraction of patients with no evidence of disease activity (no relapse, stable EDSS, no new active lesions in MRI) increased from 11% to 38%. The efficacy and safety were comparable between highly active patients or patients with relevant comorbidities and our general patient population. CONCLUSIONS The efficacy in reducing relapses was comparable to that observed in the phase III trials. In our cohort fingolimod was safe and efficacious irrespective of comorbidities and previous treatment.
Collapse
Affiliation(s)
- Maria Rasenack
- Department of Neurology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Jonathan Rychen
- Department of Neurology, University Hospital Basel, Basel, Switzerland
| | - Michaela Andelova
- Department of Neurology, University Hospital Basel, Basel, Switzerland.,Department of Radiology, Division of Neuroradiology, Basel, Switzerland
| | - Yvonne Naegelin
- Department of Neurology, University Hospital Basel, Basel, Switzerland
| | | | - Ludwig Kappos
- Department of Neurology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Raija L P Lindberg
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Till Sprenger
- Department of Neurology, DKD Helios Klinik Wiesbaden, Wiesbaden, Germany
| | - Tobias Derfuss
- Department of Neurology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
113
|
Sato F, Omura S, Jaffe S, Tsunoda I. Role of CD4+ T Cells in the Pathophysiology of Multiple Sclerosis. MULTIPLE SCLEROSIS 2016. [PMCID: PMC7150304 DOI: 10.1016/b978-0-12-800763-1.00004-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Although the precise etiology of MS remains unclear, CD4+ T cells have been proposed to play not only effector but also regulatory roles in MS. CD4+ T cells can be divided into four subsets: pro-inflammatory helper T (Th) 1 and Th17 cells, anti-inflammatory Th2 cells and regulatory T cells (Tregs). The roles of CD4+ T cells in MS have been clarified by either “loss-of-function” or “gain-of-function” methods, which have been carried out mainly in autoimmune and viral models of MS: experimental autoimmune encephalomyelitis and Theiler's murine encephalomyelitis virus infection, respectively. Observations in MS patients were consistent with the mechanisms found in the MS models, that is, increased pro-inflammatory Th1 and Th17 activity is associated with disease exacerbation, while anti-inflammatory Th2 cells and Tregs appear to play a protective role.
Collapse
|
114
|
Avau A, Matthys P. Therapeutic Potential of Interferon-γ and Its Antagonists in Autoinflammation: Lessons from Murine Models of Systemic Juvenile Idiopathic Arthritis and Macrophage Activation Syndrome. Pharmaceuticals (Basel) 2015; 8:793-815. [PMID: 26610523 PMCID: PMC4695810 DOI: 10.3390/ph8040793] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/09/2015] [Accepted: 11/18/2015] [Indexed: 01/05/2023] Open
Abstract
Interferon-γ (IFN-γ) affects immune responses in a complex fashion. Its immunostimulatory actions, such as macrophage activation and induction of T helper 1-type responsiveness, are widely acknowledged, however, as documented by a large body of literature, IFN-γ has also the potential to temper inflammatory processes via other pathways. In autoimmune and autoinflammatory disorders, IFN-γ can either play a disease-enforcing role or act as protective agent, depending on the nature of the disease. In animal models of any particular autoimmune disease, certain changes in the induction procedure can reverse the net outcome of introduction or ablation of IFN-γ. Here, we review the role of endogenous IFN-γ in inflammatory disorders and related murine models, with a focus on systemic juvenile idiopathic arthritis (sJIA) and macrophage activation syndrome (MAS). In particular, we discuss our recent findings in a mouse model of sJIA, in which endogenous IFN-γ acts as a regulatory agent, and compare with results from mouse models of MAS. Also, we elaborate on the complexity in the activity of IFN-γ and the resulting difficulty of predicting its value or that of its antagonists as treatment option.
Collapse
Affiliation(s)
- Anneleen Avau
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute, KU Leuven - University of Leuven, Leuven B-3000, Belgium.
| | - Patrick Matthys
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute, KU Leuven - University of Leuven, Leuven B-3000, Belgium.
| |
Collapse
|
115
|
van Noort JM, Bsibsi M, Nacken PJ, Verbeek R, Venneker EH. Therapeutic Intervention in Multiple Sclerosis with Alpha B-Crystallin: A Randomized Controlled Phase IIa Trial. PLoS One 2015; 10:e0143366. [PMID: 26599332 PMCID: PMC4657879 DOI: 10.1371/journal.pone.0143366] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 10/30/2015] [Indexed: 12/14/2022] Open
Abstract
As a molecular chaperone and activator of Toll-like receptor 2-mediated protective responses by microglia and macrophages, the small heat shock protein alpha B-crystallin (HspB5) exerts therapeutic effects in different animal models for neuroinflammation, including the model for multiple sclerosis (MS). Yet, HspB5 can also stimulate human antigen-specific memory T cells to release IFN-γ, a cytokine with well-documented detrimental effects during MS. In this study, we explored in a Phase IIa randomized clinical trial the therapeutic application of HspB5 in relapsing-remitting MS (RR-MS), using intravenous doses sufficient to support its protective effects, but too low to trigger pathogenic memory T-cell responses. These sub-immunogenic doses were selected based on in vitro analysis of the dose-response profile of human T cells and macrophages to HspB5, and on the immunological effects of HspB5 in healthy humans as established in a preparatory Phase I study. In a 48-week randomized, placebo-controlled, double-blind Phase IIa trial, three bimonthly intravenous injections of 7.5, 12.5 or 17.5 mg HspB5 were found to be safe and well tolerated in RR-MS patients. While predefined clinical endpoints did not differ significantly between the relatively small groups of MS patients treated with either HspB5 or placebo, repeated administration especially of the lower doses of HspB5 led to a progressive decline in MS lesion activity as monitored by magnetic resonance imaging (MRI), which was not seen in the placebo group. Exploratory linear regression analysis revealed this decline to be significant in the combined group receiving either of the two lower doses, and to result in a 76% reduction in both number and total volumes of active MRI lesions at 9 months into the study. These data provide the first indication for clinical benefit resulting from intervention in RR-MS with HspB5. Trial Registration: ClinicalTrials.gov Phase I: NCT02442557; Phase IIa: NCT02442570
Collapse
|
116
|
Simvastatin ameliorates experimental autoimmune encephalomyelitis by inhibiting Th1/Th17 response and cellular infiltration. Inflammopharmacology 2015; 23:343-54. [PMID: 26559850 DOI: 10.1007/s10787-015-0252-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/20/2015] [Indexed: 12/14/2022]
Abstract
AIM Experimental autoimmune encephalomyelitis (EAE) is a CD4(+)-mediated autoimmune pathology of the central nervous system (CNS) that is used as a model for the study of the human neuroinflammatory disease, multiple sclerosis. During the development of EAE, auto-reactive Th1 and Th17 CD4(+) T cells infiltrate the CNS promoting inflammatory cells recruitment, focal inflammation and tissue destruction. In this sense, statins, agents used to lower lipid levels, have recently shown to exert interesting immunomodulatory function. In fact, statins promote a bias towards a Th2 response, which ameliorates the clinical outcome of EAE. Additionally, simvastatin can inhibit Th17 differentiation. However, many other effects exerted on the immune system by statins have yet to be clarified, in particular during neuroinflammation. Thus, the aim of this study was to investigate the effects of simvastatin on the development of experimental autoimmune encephalomyelitis. METHODS Mice were immunized with MOG(35-55) and EAE severity was assessed daily and scored using a clinical scale. Cytokine secretion by mononuclear cells infiltrating the CNS was evaluated by flow cytometry. RESULTS Simvastatin (5 mg/kg/day) improved clinical outcome, induced an increase in TGF-β mRNA expression and inhibited IL-6, IL-12p40, IL-12p70, RANTES and MIP-1β secretion (p < 0.05). This was accompanied by a significant decrease in CNS inflammatory mononuclear cell infiltration, with reduced frequencies of both Th1 and Th17 cells. Simvastatin inhibited the proliferation of T lymphocytes co-cultured with primary microglial cells. CONCLUSIONS Simvastatin treatment promotes EAE clinical amelioration by inhibiting T cell proliferation and CNS infiltration by pathogenic Th1 and Th17 cells.
Collapse
|
117
|
Álvarez-Sánchez N, Cruz-Chamorro I, López-González A, Utrilla JC, Fernández-Santos JM, Martínez-López A, Lardone PJ, Guerrero JM, Carrillo-Vico A. Melatonin controls experimental autoimmune encephalomyelitis by altering the T effector/regulatory balance. Brain Behav Immun 2015; 50:101-114. [PMID: 26130320 DOI: 10.1016/j.bbi.2015.06.021] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 06/26/2015] [Accepted: 06/26/2015] [Indexed: 02/01/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE), the experimental model for multiple sclerosis (MS), is triggered by myelin-specific Th1 and Th17 cells. The immunomodulatory activities of melatonin have been shown to be beneficial under several conditions in which the immune system is exacerbated. Here, we sought to elucidate the basis of the melatonin protective effect on EAE by characterizing the T effector/regulatory responses, particularly those of the memory cell subsets. Melatonin was tested for its effect on Th1, Th17 and T regulatory (Treg) cells in the lymph nodes and CNS of immunodominant peptide of myelin oligodendrocyte glycoprotein (pMOG)-immunized and EAE mice, respectively. The capacity of melatonin to ameliorate EAE as well as modifying both T cell response and effector/regulatory balance was surveyed. T cell memory subsets and CD44, a key activation marker involved in the EAE pathogenesis, were also examined. Melatonin protected from EAE by decreasing peripheral and central Th1/Th17 responses and enhancing both the Treg frequency and IL-10 synthesis in the CNS. Melatonin reduced the T effector memory population and its pro-inflammatory response and regulated CD44 expression, which was decreased in T effector cells and increased in Tregs. The alterations in the T cell subpopulations were associated with a reduced mononuclear infiltration (CD4 and CD11b cells) of the melatonin-treated mice CNS. For the first time, we report that melatonin protects against EAE by controlling peripheral and central T effector/regulatory responses, effects that might be partially mediated by CD44. This immunomodulatory effect on EAE suggests that melatonin may represent an effective treatment option for MS.
Collapse
Affiliation(s)
- Nuria Álvarez-Sánchez
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Ivan Cruz-Chamorro
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Spain
| | - Antonio López-González
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Neurosurgery, Virgen Macarena & Virgen del Rocío University Hospitals, Seville, Spain
| | - José C Utrilla
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, Spain
| | - José M Fernández-Santos
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, Spain
| | - Alicia Martínez-López
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Spain
| | - Patricia J Lardone
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Spain
| | - Juan M Guerrero
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Spain; Department of Clinical Biochemistry, Virgen del Rocío University Hospital, Seville, Spain
| | - Antonio Carrillo-Vico
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Spain.
| |
Collapse
|
118
|
López de Padilla CM, Niewold TB. The type I interferons: Basic concepts and clinical relevance in immune-mediated inflammatory diseases. Gene 2015; 576:14-21. [PMID: 26410416 DOI: 10.1016/j.gene.2015.09.058] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 09/04/2015] [Accepted: 09/22/2015] [Indexed: 02/08/2023]
Abstract
There is increasing scientific and clinical interest in elucidating the biology of type I Interferons, which began approximately 60 years ago with the concept of "viral interference", a property that reduces the ability of a virus to infect cells. Although our understanding of the multiple cellular and molecular functions of interferons has advanced significantly, much remains to be learned and type I Interferons remain an active and fascinating area of inquiry. In this review, we cover some general aspects of type I interferon genes, with emphasis on interferon-alpha, and various aspects of molecular mechanisms triggered by type I interferons and toll-like receptor signaling by the Janus activated kinase/signal transducer activation of transcription (JAK-STAT) pathway and interferon regulatory factor pathway. We will also describe the role of type I interferons in autoimmune and inflammatory diseases, and its potential use as therapeutic agent.
Collapse
Affiliation(s)
| | - Timothy B Niewold
- Division of Rheumatology and Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
119
|
Yshii L, Gebauer C, Bernard-Valnet R, Liblau R. Neurons and T cells: Understanding this interaction for inflammatory neurological diseases. Eur J Immunol 2015; 45:2712-20. [DOI: 10.1002/eji.201545759] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 08/26/2015] [Accepted: 09/02/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Lidia Yshii
- INSERM U1043 - CNRS UMR 5282; Centre de Physiopathologie Toulouse-Purpan; Toulouse France
- Université Toulouse III; Toulouse France
- Institute of Biomedical Sciences I; University of Sao Paulo; Sao Paulo Brazil
| | - Christina Gebauer
- INSERM U1043 - CNRS UMR 5282; Centre de Physiopathologie Toulouse-Purpan; Toulouse France
- Université Toulouse III; Toulouse France
| | - Raphaël Bernard-Valnet
- INSERM U1043 - CNRS UMR 5282; Centre de Physiopathologie Toulouse-Purpan; Toulouse France
- Université Toulouse III; Toulouse France
| | - Roland Liblau
- INSERM U1043 - CNRS UMR 5282; Centre de Physiopathologie Toulouse-Purpan; Toulouse France
- Université Toulouse III; Toulouse France
| |
Collapse
|
120
|
Edwards SC, McGinley AM, McGuinness NC, Mills KHG. γδ T Cells and NK Cells - Distinct Pathogenic Roles as Innate-Like Immune Cells in CNS Autoimmunity. Front Immunol 2015; 6:455. [PMID: 26441960 PMCID: PMC4561808 DOI: 10.3389/fimmu.2015.00455] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 08/24/2015] [Indexed: 12/31/2022] Open
Affiliation(s)
- Sarah C Edwards
- Immune Regulation Research Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin , Dublin , Ireland
| | - Aoife M McGinley
- Immune Regulation Research Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin , Dublin , Ireland
| | - Niamh C McGuinness
- Immune Regulation Research Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin , Dublin , Ireland ; Trinity College Institute of Neuroscience, Trinity College Dublin , Dublin , Ireland
| | - Kingston H G Mills
- Immune Regulation Research Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
121
|
Miller NM, Wang J, Tan Y, Dittel BN. Anti-inflammatory mechanisms of IFN-γ studied in experimental autoimmune encephalomyelitis reveal neutrophils as a potential target in multiple sclerosis. Front Neurosci 2015; 9:287. [PMID: 26347600 PMCID: PMC4539553 DOI: 10.3389/fnins.2015.00287] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 07/30/2015] [Indexed: 01/01/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) mediated by T helper (h)1 and/or Th17 CD4 T cells that drive inflammatory lesion development along with demyelination and neuronal damage. Defects in immune regulatory mechanisms are thought to play a role in the pathogenesis of MS. While an early clinical trial indicated that IFN-γ administration was detrimental to MS, studies in the mouse model of MS, experimental autoimmune encephalomyelitis (EAE), indicated that IFN-γ exhibits a number of anti-inflammatory properties within the CNS. These mechanisms include inhibition of IL-17 production, induction of regulatory T cells, T cell apoptosis and regulation of chemokine production. Mice deficient in IFN-γ or its receptor were instrumental in deciphering the anti-inflammatory properties of IFN-γ in the CNS. In particular, they revealed that IFN-γ is a major regulator of neutrophil recruitment into the CNS, which by a variety of mechanisms including disruption of the blood-brain-barrier (BBB) and production of reactive oxygen species are thought to contribute to the onset and progression of EAE. Neutrophils were also shown to be instrumental in EAE relapses. To date neutrophils have not been appreciated as a driver of MS, but more recently based largely on strong EAE data this view is being reevaluated by some investigators in the field.
Collapse
Affiliation(s)
- Nichole M Miller
- BloodCenter of Wisconsin, Blood Research Institute Milwaukee, WI, USA
| | - Jun Wang
- BloodCenter of Wisconsin, Blood Research Institute Milwaukee, WI, USA
| | - Yanping Tan
- BloodCenter of Wisconsin, Blood Research Institute Milwaukee, WI, USA
| | - Bonnie N Dittel
- BloodCenter of Wisconsin, Blood Research Institute Milwaukee, WI, USA
| |
Collapse
|
122
|
Khaiboullina SF, Gumerova AR, Khafizova IF, Martynova EV, Lombardi VC, Bellusci S, Rizvanov AA. CCL27: Novel Cytokine with Potential Role in Pathogenesis of Multiple Sclerosis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:189638. [PMID: 26295034 PMCID: PMC4532821 DOI: 10.1155/2015/189638] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 12/11/2014] [Indexed: 02/08/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune and neurodegenerative disease of unknown etiology. Leukocyte infiltration of brain tissue and the subsequent inflammation, demyelination, axonal damage, and formation of sclerotic plaques is a hallmark of MS. Upregulation of proinflammatory cytokines has been suggested to play an essential role in regulating lymphocyte migration in MS. Here we present data on serum cytokine expression in MS cases. Increased serum levels of IL-17 and IL-23 were observed, suggesting activation of the Th17 population of immune effector cells. Additionally, increased levels of IL-22 were observed in the serum of those with acute phase MS. Unexpectedly, we observed an upregulation of the serum chemokine CCL27 in newly diagnosed and acute MS cases. CCL27 is an inflammatory chemokine associated with homing of memory T cells to sites of inflammation. Therefore, its upregulation in association with MS suggests a potential role in disease pathogenesis. Our data supports previous reports showing IL-17 and -23 upregulation in association with MS and potentially identify a previously unknown involvement for CCL27.
Collapse
Affiliation(s)
- Svetlana F. Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
- WP Institute, Reno, NV 89557, USA
| | - Aigul R. Gumerova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia
- Kazan State Medical University, 49 Butlerova Street, Kazan, Tatarstan 420012, Russia
| | - Irina F. Khafizova
- Kazan State Medical University, 49 Butlerova Street, Kazan, Tatarstan 420012, Russia
| | - Ekaterina V. Martynova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia
- Kazan State Medical University, 49 Butlerova Street, Kazan, Tatarstan 420012, Russia
| | - Vincent C. Lombardi
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
- WP Institute, Reno, NV 89557, USA
| | - Saverio Bellusci
- Excellence Cluster Cardio-Pulmonary System, Justus Liebig University, Aulweg 130, 35392 Giessen, Germany
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia
| |
Collapse
|
123
|
Affiliation(s)
- Yukio Takeshita
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Ube Yamaguchi Japan
| | | |
Collapse
|
124
|
Ramos I, Fernandez-Sesma A. Modulating the Innate Immune Response to Influenza A Virus: Potential Therapeutic Use of Anti-Inflammatory Drugs. Front Immunol 2015; 6:361. [PMID: 26257731 PMCID: PMC4507467 DOI: 10.3389/fimmu.2015.00361] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/04/2015] [Indexed: 12/27/2022] Open
Abstract
Infection by influenza A viruses (IAV) is frequently characterized by robust inflammation that is usually more pronounced in the case of avian influenza. It is becoming clearer that the morbidity and pathogenesis caused by IAV are consequences of this inflammatory response, with several components of the innate immune system acting as the main players. It has been postulated that using a therapeutic approach to limit the innate immune response in combination with antiviral drugs has the potential to diminish symptoms and tissue damage caused by IAV infection. Indeed, some anti-inflammatory agents have been shown to be effective in animal models in reducing IAV pathology as a proof of principle. The main challenge in developing such therapies is to selectively modulate signaling pathways that contribute to lung injury while maintaining the ability of the host cells to mount an antiviral response to control virus replication. However, the dissection of those pathways is very complex given the numerous components regulated by the same factors (i.e., NF kappa B transcription factors) and the large number of players involved in this regulation, some of which may be undescribed or unknown. This article provides a comprehensive review of the current knowledge regarding the innate immune responses associated with tissue damage by IAV infection, the understanding of which is essential for the development of effective immunomodulatory drugs. Furthermore, we summarize the recent advances on the development and evaluation of such drugs as well as the lessons learned from those studies.
Collapse
Affiliation(s)
- Irene Ramos
- Department of Microbiology, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| | - Ana Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| |
Collapse
|
125
|
Scolding N. The best clinical paper on multiple sclerosis in 2014. Mult Scler 2015; 21:854-5. [DOI: 10.1177/1352458515584416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Neil Scolding
- Bristol Institute of Clinical Neurosciences, Bristol, UK
| |
Collapse
|
126
|
Abstract
The molecular mechanisms governing T helper (Th) cell differentiation and function have revealed a complex network of transcriptional and protein regulators. Cytokines not only initiate the differentiation of CD4 Th cells into subsets but also influence the identity, plasticity and effector function of a T cell. Of the subsets, Th17 cells, named for producing interleukin 17 (IL-17) as their signature cytokine, secrete a cohort of other cytokines, including IL-22, IL-21, IL-10, IL-9, IFNγ, and GM-CSF. In recent years, Th17 cells have emerged as key players in host defense against both extracellular pathogens and fungal infections, but they have also been implicated as one of the main drivers in the pathogenesis of autoimmunity, likely mediated in part by the cytokines that they produce. Advances in high throughput genomic sequencing have revealed unexpected heterogeneity in Th17 cells and, as a consequence, may have tremendous impact on our understanding of their functional diversity. The assortment in gene expression may also identify different functional states of Th17 cells. This review aims to understand the interplay between the cytokine regulators that drive Th17 cell differentiation and functional states in Th17 cells.
Collapse
Affiliation(s)
- Youjin Lee
- Evergrande Center for Immumnologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Vijay Kuchroo
- Evergrande Center for Immumnologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA; Genomic and Biotechnology Section, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
127
|
T helper 9 cells induced by plasmacytoid dendritic cells regulate interleukin-17 in multiple sclerosis. Clin Sci (Lond) 2015; 129:291-303. [DOI: 10.1042/cs20140608] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We have established a novel role in multiple sclerosis for a molecule, called IL-9, produced by immune cells. IL-9 reduces inflammation, and its expression in the cerebrospinal fluid of patients inversely correlates with the severity of multiple sclerosis.
Collapse
|
128
|
Multiple Sclerosis and T Lymphocytes: An Entangled Story. J Neuroimmune Pharmacol 2015; 10:528-46. [PMID: 25946987 DOI: 10.1007/s11481-015-9614-0] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/29/2015] [Indexed: 12/17/2022]
Abstract
Multiple sclerosis (MS) is the prototypic inflammatory disease of the central nervous system (CNS) characterized by multifocal areas of demyelination, axonal damage, activation of glial cells, and immune cell infiltration. Despite intensive years of research, the etiology of this neurological disorder remains elusive. Nevertheless, the abundance of immune cells such as T lymphocytes and their products in CNS lesions of MS patients supports the notion that MS is an immune-mediated disorder. An important body of evidence gathered from MS animal models such as experimental autoimmune encephalomyelitis (EAE), points to the central contribution of CD4 T lymphocytes in disease pathogenesis. Both Th1 (producing interferon-γ) and Th17 (producing interleukin 17) CD4 T lymphocytes targeting CNS self-antigens have been implicated in MS and EAE pathobiology. Moreover, several publications suggest that CD8 T lymphocytes also participate in the development of MS lesions. The migration of activated T lymphocytes from the periphery into the CNS has been identified as a crucial step in the formation of MS lesions. Several factors promote such T cell extravasation including: molecules (e.g., cell adhesion molecules) implicated in the T cell-blood brain barrier interaction, and chemokines produced by neural cells. Finally, once in the CNS, T lymphocytes need to be reactivated by local antigen presenting cells prior to enter the parenchyma where they can initiate damage. Further investigations will be necessary to elucidate the impact of environmental factors (e.g., gut microbiota) and CNS intrinsic properties (e.g., microglial activation) on this inflammatory neurological disease.
Collapse
|
129
|
Song ZY, Yamasaki R, Kawano Y, Sato S, Masaki K, Yoshimura S, Matsuse D, Murai H, Matsushita T, Kira JI. Peripheral blood T cell dynamics predict relapse in multiple sclerosis patients on fingolimod. PLoS One 2015; 10:e0124923. [PMID: 25919001 PMCID: PMC4412716 DOI: 10.1371/journal.pone.0124923] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/10/2015] [Indexed: 12/27/2022] Open
Abstract
Background Fingolimod efficiently reduces multiple sclerosis (MS) relapse by inhibiting lymphocyte egress from lymph nodes through down-modulation of sphingosine 1-phosphate (S1P) receptors. We aimed to clarify the alterations in peripheral blood T cell subsets associated with MS relapse on fingolimod. Methods/Principal Findings Blood samples successively collected from 23 relapsing-remitting MS patients before and during fingolimod therapy (0.5 mg/day) for 12 months and 18 healthy controls (HCs) were analysed for T cell subsets by flow cytometry. In MS patients, the percentages of central memory T (CCR7+CD45RO+) cells (TCM) and naïve T (CCR7+CD45RO-) cells decreased significantly, while those of effector memory T (CCR7-CD45RA-) and suppressor precursor T (CD28-) cells increased in both CD4+T and CD8+T cells from 2 weeks to 12 months during fingolimod therapy. The percentages of regulatory T (CD4+CD25highCD127low) cells in CD4+T cells and CCR7-CD45RA+T cells in CD8+T cells also increased significantly. Eight relapsed patients demonstrated greater percentages of CD4+TCM than 15 non-relapsed patients at 3 and 6 months (p=0.0051 and p=0.0088, respectively). The IL17-, IL9-, and IL4-producing CD4+T cell percentages were significantly higher at pre-treatment in MS patients compared with HCs (p<0.01 for all), while the IL17-producing CD4+T cell percentages tended to show a transient increase at 2 weeks of fingolimod therapy (pcorr=0.0834). Conclusions The CD4+TCM percentages at 2 weeks to 12 months during fingolimod therapy are related to relapse.
Collapse
Affiliation(s)
- Zi-Ye Song
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Yamasaki
- Department of Neurological Therapeutics, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuji Kawano
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinya Sato
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoshi Yoshimura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dai Matsuse
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Murai
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takuya Matsushita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun-ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- * E-mail:
| |
Collapse
|
130
|
Kostic M, Stojanovic I, Marjanovic G, Zivkovic N, Cvetanovic A. Deleterious versus protective autoimmunity in multiple sclerosis. Cell Immunol 2015; 296:122-32. [PMID: 25944389 DOI: 10.1016/j.cellimm.2015.04.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/18/2015] [Accepted: 04/22/2015] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disorder of central nervous system, in which myelin specific CD4(+) T cells have a central role in orchestrating pathological events involved in disease pathogenesis. There is compelling evidence that Th1, Th9 and Th17 cells, separately or in cooperation, could mediate deleterious autoimmune response in MS. However, the phenotype differences between Th cell subpopulations initially employed in MS pathogenesis are mainly reflected in the different patterns of inflammation introduction, which results in the development of characteristic pathological features (blood-brain barrier disruption, demyelination and neurodegeneration), clinically presented with MS symptoms. Although, autoimmunity was traditionally seen as deleterious, some studies indicated that autoimmunity mediated by Th2 cells and T regulatory cells could be protective by nature. The concept of protective autoimmunity in MS pathogenesis is still poorly understood, but could be of great importance in better understanding of MS immunology and therefore, creating better therapeutic strategies.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia.
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia
| | - Goran Marjanovic
- Department of Immunology, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia
| | - Ana Cvetanovic
- Clinic of Oncology, Clinical Centre, Blvd. Dr. Zorana Djindjica 48, 18000 Nis, Serbia
| |
Collapse
|
131
|
Racke MK, Yang Y, Lovett-Racke AE. Is T-bet a potential therapeutic target in multiple sclerosis? J Interferon Cytokine Res 2015; 34:623-32. [PMID: 25084179 DOI: 10.1089/jir.2014.0020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Treatments for multiple sclerosis (MS) have changed over the past years as our understanding of immunology and neuroscience has evolved. Experimental autoimmune encephalomyelitis (EAE) continues to remain the major model for MS and has been a major vehicle in the development of new therapeutic targets for MS, including new agents such as natalizumab, fingolimod, and dimethyl fumarate. As progress in the molecular understanding of immunology continues, many observations in EAE are pursued with the ultimate goal of defining the pathophysiology of MS and development of innovative treatments for the disease. Although many consider MS to be a T cell-mediated autoimmune disease directed against myelin antigens, the exact cause of the disease is still unknown. For many years, it was thought that myelin-specific T cells that secreted interferon-γ and were proinflammatory were the major T cell subset that mediated the disease, but recent studies on the cytokine phenotype of pathogenic T cells in EAE and MS have opened debate on this issue. Work over the past several years suggests that the transcription factor T-bet appears to be an important factor in T cell encephalitogenicity; however, recent data suggest that it is also dispensable in certain situations, particularly for Th17 cells. Understanding the molecular mechanisms responsible for T cell encephalitogenicity in MS and other autoimmune diseases will be essential in the development of specific therapies for these inflammatory diseases.
Collapse
Affiliation(s)
- Michael K Racke
- 1 Department of Neurology, The Ohio State University Wexner Medical Center , Columbus, Ohio
| | | | | |
Collapse
|
132
|
Exacerbation of experimental autoimmune encephalomyelitis in ceramide synthase 6 knockout mice is associated with enhanced activation/migration of neutrophils. Immunol Cell Biol 2015; 93:825-36. [PMID: 25833068 DOI: 10.1038/icb.2015.47] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/17/2015] [Accepted: 03/28/2015] [Indexed: 01/03/2023]
Abstract
Ceramides are mediators of inflammatory processes. In experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), we observed that CerS6 mRNA expression was upregulated 15-fold in peripheral blood leukocytes before the onset of EAE symptoms. In peripheral blood leukocytes from MS patients, a 3.9-fold upregulation was found. Total genetic deletion of CerS6 and the selective deletion of CerS6 in peripheral blood leucocytes exacerbated the progression of clinical symptoms in EAE mice. This was associated with enhanced leukocyte, predominantly neutrophil infiltration and enhanced demyelination in the lumbar spinal cord of EAE mice. Interferon-gamma/tumor necrosis factor alpha (IFN-γ/TNF-α) and granulocyte colony-stimulating factor (G-CSF) both drive EAE development and induce expression of the integrin CD11b and the chemokine receptor C-X-C motif chemokine receptor 2 (CXCR2), and we found they also induce CerS6 expression. In vivo, the genetic deletion of CerS6 enhanced the activation/migration of neutrophils, as reflected by an enhanced upregulation of CD11b and CXCR2. In vitro, the genetic deletion of CerS6 enhanced the activation status of IFN-γ/TNF-α-stimulated neutrophils, as shown by increased expression of nitric oxide and CD11b and an increased adhesion capacity. In G-CSF-stimulated neutrophils, the migration status was enhanced, as reflected by an elevated level of CXCR2 and an increased migration capacity. These data suggest that CerS6/C16-Cer mediates feedback regulation by inhibiting the formation of CD11b and CXCR2, which are induced either by IFN-γ/TNF-α or by G-CSF, respectively. We conclude that CerS6/C16-Cer mediates anti-inflammatory effects during the development of EAE and MS possibly by suppressing the migration and deactivation of neutrophils.
Collapse
|
133
|
|
134
|
Abstract
The brain under immunological attack does not surrender quietly. Investigation of brain lesions in multiple sclerosis (MS) reveals a coordinated molecular response involving various proteins and small molecules ranging from heat shock proteins to small lipids, neurotransmitters, and even gases, which provide protection and foster repair. Reduction of inflammation serves as a necessary prerequisite for effective recovery and regeneration. Remarkably, many lesion-resident molecules activate pathways leading to both suppression of inflammation and promotion of repair mechanisms. These guardian molecules and their corresponding physiologic pathways could potentially be exploited to silence inflammation and repair the injured and degenerating brain and spinal cord in both relapsing-remitting and progressive forms of MS and may be beneficial in other neurologic and psychiatric conditions.
Collapse
|
135
|
Capobianchi MR, Uleri E, Caglioti C, Dolei A. Type I IFN family members: similarity, differences and interaction. Cytokine Growth Factor Rev 2015; 26:103-11. [PMID: 25466633 PMCID: PMC7108279 DOI: 10.1016/j.cytogfr.2014.10.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/22/2014] [Indexed: 02/07/2023]
Abstract
Interferons (IFN) are key cytokines with multifaceted antiviral and cell-modulatory properties. Three distinct types of IFN are recognized (I-III) based on structural features, receptor usage, cellular source and biological activities. The action of IFNs is mediated by a complex, partially overlapping, transcriptional program initiated by the interaction with specific receptors. Genetic diversity, with polymorphisms and mutations, can modulate the extent of IFN responses and the susceptibility to infections. Almost all viruses developed mechanisms to subvert the IFN response, involving both IFN induction and effector mechanisms. Interactions between IFN types may occur, for both antiviral and cell-modulatory effects, in a complex interplay, involving both synergistic and antagonistic effects. Interferon-associated diseases, not related to virus infections may occur, some of them frequently observed in IFN-treated patients. On the whole, IFNs are pleiotropic biologic response modifiers, that, upon activation of thousands genes, induce a broad spectrum of activities, regulating cell cycle, differentiation, plasma membrane molecules, release of mediators, etc., that can be relevant for cell proliferation, innate and adaptive immunity, hematopoiesis, angiogenesis and other body functions.
Collapse
Affiliation(s)
- Maria Rosaria Capobianchi
- Laboratory of Virology, National Institute for Infectious Diseases "L. Spallanzani", Via Portuense 292, Rome, Italy
| | - Elena Uleri
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Claudia Caglioti
- Laboratory of Virology, National Institute for Infectious Diseases "L. Spallanzani", Via Portuense 292, Rome, Italy
| | - Antonina Dolei
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.
| |
Collapse
|
136
|
Ding X, Yan Y, Li X, Li K, Ciric B, Yang J, Zhang Y, Wu S, Xu H, Chen W, Lovett-Racke AE, Zhang GX, Rostami A. Silencing IFN-γ binding/signaling in astrocytes versus microglia leads to opposite effects on central nervous system autoimmunity. THE JOURNAL OF IMMUNOLOGY 2015; 194:4251-64. [PMID: 25795755 DOI: 10.4049/jimmunol.1303321] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/18/2015] [Indexed: 01/02/2023]
Abstract
IFN-γ, the hallmark cytokine of Th1 cells, plays an important role in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Thus far, the role of IFN-γ in EAE has been largely studied through its effects on immune cells, whereas much less is known about its effects on CNS cells, especially in vivo. In this study, we dissected the in vivo effects and mechanisms of IFN-γ binding/signaling in astrocytes and microglia, and found that IFN-γ signaling in these cell types has opposite effects in EAE pathogenesis. Silencing IFN-γ binding/signaling in astrocytes alleviated EAE, whereas in microglia, and likely in some infiltrating macrophages, it increased disease severity. Silencing IFN-γ signaling in astrocytes resulted in diminished expression of chemokines and fewer inflammatory cells infiltrating into the CNS, whereas blocking IFN-γ binding/signaling in microglia, probably infiltrating macrophages as well, increased disease severity through augmented activation and proliferation of microglia. Further, blocking IFN-γ binding/signaling in astrocytes alleviated both Th1- and Th17-mediated adoptive EAE, indicating an important role for IFN-γ signaling in astrocytes in autoimmune CNS inflammation. Thus, our study defines novel mechanisms of action of IFN-γ in EAE pathogenesis, and also highlights an opportunity for development of multiple sclerosis therapies directed at CNS cells.
Collapse
Affiliation(s)
- Xiaoli Ding
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107; Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Yaping Yan
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107; Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Xing Li
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Ke Li
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jingxian Yang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107; Department of Pharmacology, Liaoning University of Traditional Chinese Medicine, Dalian 110847, China
| | - Yuan Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Shuai Wu
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Hui Xu
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Wanjun Chen
- Mucosal Immunology Section, Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892; and
| | - Amy E Lovett-Racke
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107;
| | | |
Collapse
|
137
|
Pharmaceutical integrated stress response enhancement protects oligodendrocytes and provides a potential multiple sclerosis therapeutic. Nat Commun 2015; 6:6532. [PMID: 25766071 PMCID: PMC4360920 DOI: 10.1038/ncomms7532] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/05/2015] [Indexed: 01/21/2023] Open
Abstract
Oligodendrocyte death contributes to the pathogenesis of the inflammatory demyelinating disease multiple sclerosis (MS). Nevertheless, current MS therapies are mainly immunomodulatory and have demonstrated limited ability to inhibit MS progression. Protection of oligodendrocytes is therefore a desirable strategy for alleviating disease. Here we demonstrate that enhancement of the integrated stress response using the FDA-approved drug guanabenz increases oligodendrocyte survival in culture and prevents hypomyelination in cerebellar explants in the presence of interferon-γ, a pro-inflammatory cytokine implicated in MS pathogenesis. In vivo, guanabenz treatment protects against oligodendrocyte loss caused by CNS-specific expression of interferon-γ. In a mouse model of MS, experimental autoimmune encephalomyelitis, guanabenz alleviates clinical symptoms, which correlates with increased oligodendrocyte survival and diminished CNS CD4+ T cell accumulation. Moreover, guanabenz ameliorates relapse in relapsing-remitting experimental autoimmune encephalomyelitis. Our results provide support for a MS therapy that enhances the integrated stress response to protect oligodendrocytes against the inflammatory CNS environment. Current multiple sclerosis treatments focus on prevention of immune attack on oligodendrocytes and myelin. Here the authors show a different strategy to ameliorate disease in several mouse models, protecting oligodendrocytes from inflammation-induced death with an FDA-approved drug, guanabenz.
Collapse
|
138
|
Abstract
Multiple sclerosis (MS) has been thought to be a complex and indecipherable disease, and poorly understood with regards to aetiology. Here, we suggest an emphatically positive view of progress over several decades in the understanding and treatment of MS, particularly focusing on advances made within the past 20 years. As with virtually all complex disorders, MS is caused by the interaction of genetic and environmental factors. In recent years, formidable biochemical, bioinformatic, epidemiological and neuroimaging tools have been brought to bear on research into the causes of MS. While susceptibility to the disease is now relatively well accounted for, disease course is not and remains a salient challenge. In the therapeutic realm, numerous agents have become available, reflecting the fact that the disease can be attacked successfully at many levels and using varied strategies. Tailoring therapies to individuals, risk mitigation and selection of first-line as compared with second-line medications remain to be completed. In our view, the MS landscape has been comprehensively and irreversibly transformed by this progress. Here we focus on MS therapeutics-the most meaningful outcome of research efforts.
Collapse
Affiliation(s)
| | - David A Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, 15 York Street, New Haven, CT 06520, USA
| | - Claudia F Lucchinetti
- Department of Neurology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
139
|
Secor McVoy JR, Oughli HA, Oh U. Liver X receptor-dependent inhibition of microglial nitric oxide synthase 2. J Neuroinflammation 2015; 12:27. [PMID: 25889344 PMCID: PMC4332930 DOI: 10.1186/s12974-015-0247-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 01/14/2015] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The nuclear receptor liver X receptor (LXR) exerts transcriptional control over lipid metabolism and inflammatory response in cells of the myeloid lineage, suggesting that LXR may be a potential target in a number of chronic neuroinflammatory and neurodegenerative diseases where persistent microglial activation has been implicated in the pathogenesis. METHODS The effect of LXR activation on microglia and central nervous system (CNS) inflammation was studied using a synthetic LXR agonist in cultured microglia, a microglial cell line and experimental allergic encephalomyelitis (EAE), an animal model of CNS inflammation. RESULTS LXR activation inhibited nitric oxide synthase 2, inducible (Nos2) expression and nitric oxide production in lipopolysaccharide (LPS)-stimulated microglia. Inhibition of microglial activation in response to interferon-γ was less reliable. In LPS-stimulated cells, LXR activation did not inhibit nuclear translocation of NF-kappaB1 p50. Instead, LXR-dependent Nos2 repression was associated with inhibition of histone 4 acetylation and inhibition of NF-kappaB1 p50 binding at the Nos2 promoter. Histone acetylation and NF-kappaB1 p50 binding were mechanistically linked, and histone deacetylase (HDAC) activity appeared to be important for LXR-dependent transcriptional repression of Nos2. Analysis of CNS gene expression in animals undergoing EAE showed that the expressions of Lxr and LXR-dependent genes were downregulated during CNS inflammation. Nevertheless, administration of LXR agonist GW3965 during the effector phase of EAE delayed the onset of clinical disease and reversed the diminished expression of LXR-dependent reverse cholesterol transport genes. However, the CNS expressions of Nos2 and other inflammatory genes were not significantly inhibited by LXR activation in EAE, and clinical disease severity was comparable to vehicle controls at later time points in LXR agonist treated animals. CONCLUSIONS LXR can be targeted to modulate microglial activation. LXR-dependent repression of inflammatory genes may be stimulus-dependent and impaired by HDAC inhibition. Endogenous LXR activity does not appear to modulate CNS inflammation, but LXR activity can be partially restored in the CNS by administration of exogenous LXR agonist with an impact on clinical disease severity at early, but not late, time points in EAE.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Benzoates/therapeutic use
- Benzylamines/therapeutic use
- Cells, Cultured
- Chromatin Immunoprecipitation
- Cytokines/metabolism
- Deoxyribonucleases/metabolism
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Gene Expression Profiling
- Histones/metabolism
- Lipopolysaccharides/pharmacology
- Liver X Receptors
- Mice
- Mice, Inbred C57BL
- Mice, Inbred ICR
- Microglia/drug effects
- Microglia/metabolism
- Myelin-Oligodendrocyte Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Nitric Oxide Synthase Type II/metabolism
- Orphan Nuclear Receptors/antagonists & inhibitors
- Orphan Nuclear Receptors/metabolism
- Peptide Fragments/immunology
- Peptide Fragments/toxicity
- Protein Binding/drug effects
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
Collapse
Affiliation(s)
- Julie R Secor McVoy
- Department of Neurology, Virginia Commonwealth University School of Medicine, PO Box 980599, VA, 23298, Richmond, USA.
| | - Hanadi Ajam Oughli
- Department of Neurology, Virginia Commonwealth University School of Medicine, PO Box 980599, VA, 23298, Richmond, USA.
| | - Unsong Oh
- Department of Neurology, Virginia Commonwealth University School of Medicine, PO Box 980599, VA, 23298, Richmond, USA.
| |
Collapse
|
140
|
Belogurov A, Kuzina E, Kudriaeva A, Kononikhin A, Kovalchuk S, Surina Y, Smirnov I, Lomakin Y, Bacheva A, Stepanov A, Karpova Y, Lyupina Y, Kharybin O, Melamed D, Ponomarenko N, Sharova N, Nikolaev E, Gabibov A. Ubiquitin-independent proteosomal degradation of myelin basic protein contributes to development of neurodegenerative autoimmunity. FASEB J 2015; 29:1901-13. [PMID: 25634956 PMCID: PMC4415016 DOI: 10.1096/fj.14-259333] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 12/22/2014] [Indexed: 11/18/2022]
Abstract
Recent findings indicate that the ubiquitin–proteasome system is involved in the pathogenesis of cancer as well as autoimmune and several neurodegenerative diseases, and is thus a target for novel therapeutics. One disease that is related to aberrant protein degradation is multiple sclerosis, an autoimmune disorder involving the processing and presentation of myelin autoantigens that leads to the destruction of axons. Here, we show that brain-derived proteasomes from SJL mice with experimental autoimmune encephalomyelitis (EAE) in an ubiquitin-independent manner generate significantly increased amounts of myelin basic protein peptides that induces cytotoxic lymphocytes to target mature oligodendrocytes ex vivo. Ten times enhanced release of immunogenic peptides by cerebral proteasomes from EAE-SJL mice is caused by a dramatic shift in the balance between constitutive and β1ihigh immunoproteasomes in the CNS of SJL mice with EAE. We found that during EAE, β1i is increased in resident CNS cells, whereas β5i is imported by infiltrating lymphocytes through the blood–brain barrier. Peptidyl epoxyketone specifically inhibits brain-derived β1ihigh immunoproteasomes in vitro (kobs/[I] = 240 M−1s−1), and at a dose of 0.5 mg/kg, it ameliorates ongoing EAE in vivo. Therefore, our findings provide novel insights into myelin metabolism in pathophysiologic conditions and reveal that the β1i subunit of the immunoproteasome is a potential target to treat autoimmune neurologic diseases.—Belogurov Jr., A., Kuzina, E., Kudriaeva, A., Kononikhin, A., Kovalchuk, S., Surina, Y., Smirnov, I., Lomakin, Y., Bacheva, A., Stepanov, A., Karpova, Y., Lyupina, Y., Kharybin, O., Melamed, D., Ponomarenko, N., Sharova, N., Nikolaev, E., Gabibov, A. Ubiquitin-independent proteosomal degradation of myelin basic protein contributes to development of neurodegenerative autoimmunity.
Collapse
Affiliation(s)
- Alexey Belogurov
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Ekaterina Kuzina
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Anna Kudriaeva
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Alexey Kononikhin
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Sergey Kovalchuk
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Yelena Surina
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Ivan Smirnov
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Yakov Lomakin
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Anna Bacheva
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Alexey Stepanov
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Yaroslava Karpova
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Yulia Lyupina
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Oleg Kharybin
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Dobroslav Melamed
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Natalia Ponomarenko
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Natalia Sharova
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Eugene Nikolaev
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| | - Alexander Gabibov
- *Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Kazan Federal University, Kazan, Republic of Tatarstan, Russia; Institute of Gene Biology, Russian Acedemy of Sciences, Moscow, Russia; Chemistry Department of Moscow State University, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; **Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia; Institute of Biomedical Chemistry, Russian Academy of Medical Sciences, Moscow, Russia; and Assaf Harofeh Medical Center, Zerifin, Israel
| |
Collapse
|
141
|
Autophagy-independent functions of UVRAG are essential for peripheral naive T-cell homeostasis. Proc Natl Acad Sci U S A 2015; 112:1119-24. [PMID: 25583492 DOI: 10.1073/pnas.1423588112] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
UV radiation resistance-associated gene (UVRAG) encodes a tumor suppressor with putative roles in autophagy, endocytic trafficking, and DNA damage repair but its in vivo role in T cells is unknown. Because conditional homozygous deletion of Uvrag in mice results in early embryonic lethality, we generated T-cell-specific UVRAG-deficient mice that lacked UVRAG expression specifically in T cells. This loss of UVRAG led to defects in peripheral homeostasis that could not be explained by the increased sensitivity to cell death and impaired proliferation observed for other autophagy-related gene knockout mice. Instead, UVRAG-deficient T-cells exhibited normal mitochondrial clearance and activation-induced autophagy, suggesting that UVRAG has an autophagy-independent role that is critical for peripheral naive T-cell homeostatic proliferation. In vivo, T-cell-specific loss of UVRAG dampened CD8(+) T-cell responses to LCMV infection in mice, delayed viral clearance, and impaired memory T-cell generation. Our data provide novel insights into the control of autophagy in T cells and identify UVRAG as a new regulator of naïve peripheral T-cell homeostasis.
Collapse
|
142
|
Wong N, Nguyen T, Brenu EW, Broadley S, Staines D, Marshall-Gradisnik S. A Comparison of Cytokine Profiles of Chronic Fatigue Syndrome/Myalgic Encephalomyelitis and Multiple Sclerosis Patients. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/ijcm.2015.610103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
143
|
Resop RS, Uittenbogaart CH. Human T-Cell Development and Thymic Egress: An Infectious Disease Perspective. FORUM ON IMMUNOPATHOLOGICAL DISEASES AND THERAPEUTICS 2015; 6:33-49. [PMID: 28670486 PMCID: PMC5489135 DOI: 10.1615/forumimmundisther.2015014226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Emigration of mature naïve CD4 SP T cells from the human thymus to the periphery is not fully understood, although elucidation of the mechanisms that govern egress of T cells is crucial to understanding both basic immunology and the immune response in diseases such as HIV infection. Recent work has brought to light the requirement for sphingosine-1-phosphate (S1P) and its receptors in a variety of fields including mature naïve T-cell egress from the thymus of mice. We are examining the expression and function of this novel requisite T-cell egress receptor within the human thymus, characterizing changes observed in the expression and function of this receptor in infectious diseases. To perform this work, we use a variety of humanized murine models reviewed in this article. Future work in the field of T-cell egress, especially as it pertains to S1P receptors, should advance the fields of basic T-cell immunology and immunopathology and open new avenues for exploration into novel therapeutics.
Collapse
Affiliation(s)
- Rachel S. Resop
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
- Department of Pediatrics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
| | - Christel H. Uittenbogaart
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
- Department of Pediatrics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
- University of California at Los Angeles AIDS Institute, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, David Geffen Medical School at UCLA, Los Angeles, CA 90095
| |
Collapse
|
144
|
Sex-Based Differences in Multiple Sclerosis (Part I): Biology of Disease Incidence. Curr Top Behav Neurosci 2015; 26:29-56. [PMID: 25690593 DOI: 10.1007/7854_2015_371] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease that leads to neuron damage and progressive disability. One major feature of multiple sclerosis (MS) is that it affects women three times more often than men. In this chapter, we overview the evidence that the autoimmune component of MS, which predominates in the early stages of this disease, is more robust in women than in men and undergoes a sharp increase with the onset of puberty. In addition, we discuss the common rodent models of MS that have been used to study the sex-based differences in the development of central nervous system (CNS) autoimmunity. We then address the biological underpinnings of this enhanced MS risk in women by first reviewing the autoimmune mechanisms that are thought to lead to the initiation of this disease and then honing in on how these mechanisms differ between the sexes. Finally, we review what is known about the hormonal and genetic basis of these sex differences in CNS autoimmunity.
Collapse
|
145
|
Tang SC, Fan XH, Pan QM, Sun QS, Liu Y. Decreased expression of IL-27 and its correlation with Th1 and Th17 cells in progressive multiple sclerosis. J Neurol Sci 2014; 348:174-80. [PMID: 25498842 DOI: 10.1016/j.jns.2014.11.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/14/2014] [Accepted: 11/25/2014] [Indexed: 10/24/2022]
Abstract
Progressive multiple sclerosis (MS) is an immune-mediated demyelinating disease in which both imbalanced T helper (Th) subsets and aberrant cytokine profiles have been found. Interleukin-27 (IL-27), a cytokine with pro-inflammatory and anti-inflammatory effects, plays pleiotropic roles in immunomodulation. In the present study, plasma levels of IL-27, interferon-gamma (IFN-γ), IL-17 and frequencies of peripheral Th1, Th17 cells were determined by enzyme-linked immunosorbent assay (ELISA) and flow cytometry in 45 progressive MS and 25 healthy controls. mRNA expression levels of IL-27, IFN-γ, T-bet, IL-17 and RAR-related orphan receptor gamma t (RORγt) in peripheral blood mononuclear cells (PBMCs) were also quantified by real-time polymerase chain reaction. Plasma and mRNA levels of IL-27 in progressive MS patients were significantly lower than those in healthy controls, while plasma concentrations of IL-17, frequencies of circulating Th17, and mRNA expression levels of IL-17 as well as RORγt were all increased remarkably compared with healthy controls. No statistical significance was observed in IFN-γ and T-bet mRNA expression or plasma IFN-γ levels between progressive MS patients and healthy controls. Moreover, plasma levels of IL-27 were found to be negatively correlated to the percentages of circulating Th17 or plasma IL-17 concentrations in patients with progressive MS. Our data showed that progressive MS patients had decreased plasma and mRNA expression levels of IL-27, suggesting that it might be involved in the pathophysiological process of MS.
Collapse
Affiliation(s)
- Shao-can Tang
- Department of Rehabilitation Medicine, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, PR China; Department of Internal Neurology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, PR China
| | - Xiao-hua Fan
- Department of Rehabilitation Medicine, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, PR China
| | - Qing-min Pan
- Department of Rehabilitation Medicine, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, PR China; Department of Ophthalmology, Affiliated Hospital of Taishan Medical University, 706 Taishanda Street, Taian, Shandong, PR China
| | - Qiang-san Sun
- Department of Rehabilitation Medicine, Second Hospital of Shandong University, 247 Beiyuanda Street, Jinan, PR China.
| | - Yu Liu
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan, PR China.
| |
Collapse
|
146
|
Annibali V, Mechelli R, Romano S, Buscarinu MC, Fornasiero A, Umeton R, Ricigliano VAG, Orzi F, Coccia EM, Salvetti M, Ristori G. IFN-β and multiple sclerosis: from etiology to therapy and back. Cytokine Growth Factor Rev 2014; 26:221-8. [PMID: 25466632 DOI: 10.1016/j.cytogfr.2014.10.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 10/22/2014] [Indexed: 01/09/2023]
Abstract
Several immunomodulatory treatments are currently available for relapsing-remitting forms of multiple sclerosis (RRMS). Interferon beta (IFN) was the first therapeutic intervention able to modify the course of the disease and it is still the most used first-line treatment in RRMS. Though two decades have passed since IFN-β was introduced in the management of MS, it remains a valid approach because of its good benefit/risk profile. This is witnessed by new efforts of pharmaceutical industry to improve this line: a PEGylated form of subcutaneous IFN-β 1a, (Plegridy(®)) with a longer half-life, has been recently approved in RRMS. This review will survey the various stages of the use of type I IFN in MS, with special attention to the effect of the treatment on the supposed viral etiologic factors associated to the disease. The antiviral activities of IFN (that initially prompted its use as immunomodulatory agent in MS), and the mounting evidences in favor of a viral etiology in MS, allowed us to outline a re-appraisal from etiology to therapy and back.
Collapse
Affiliation(s)
- V Annibali
- Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy
| | - R Mechelli
- Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy
| | - S Romano
- Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy
| | - M C Buscarinu
- Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy
| | - A Fornasiero
- Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy
| | - R Umeton
- Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy
| | - V A G Ricigliano
- Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy; Neuroimmunology Unit, Fondazione Santa Lucia-I.R.C.C.S., Rome, Italy
| | - F Orzi
- Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy
| | - E M Coccia
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - M Salvetti
- Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy.
| | - G Ristori
- Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy
| |
Collapse
|
147
|
Ferreira TB, Barros PO, Teixeira B, Cassano T, Centurião N, Kasahara TM, Hygino J, Vasconcelos CCF, Filho HA, Alvarenga R, Wing AC, Andrade RM, Andrade AF, Bento CAM. Dopamine favors expansion of glucocorticoid-resistant IL-17-producing T cells in multiple sclerosis. Brain Behav Immun 2014; 41:182-90. [PMID: 24882215 DOI: 10.1016/j.bbi.2014.05.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 05/14/2014] [Accepted: 05/21/2014] [Indexed: 01/10/2023] Open
Abstract
Dopamine (DA) is a neurotransmitter produced mainly in the central nervous system (CNS) that has immunomodulatory actions on T cells. As the multiple sclerosis (MS) has long been regarded as an autoimmune disease of CNS mediated by T cells, the objective of this study was to evaluate the impact of DA on in vitro functional status of T cells from relapsing-remitting (RR)-MS patients. Peripheral T-cells from RR-MS patients were activated by mitogens and cell proliferation and cytokine production were assayed by [(3)H]-thymidine uptake and ELISA, respectively. Our results demonstrated that DA enhanced in vitro T cell proliferation and Th17-related cytokines in MS-derived cell cultures. In addition, this catecholamine reduced Treg-related cytokines (IL-10 and TGF-β) release by activated CD4(+) T cells. These DA-induced effects on T cells were mainly dependent on IL-6 production by both polyclonally-activated CD4(+) T cells and LPS-stimulated monocytes. Furthermore, the production of IL-17 and IL-6 by MS-derived T cells was directly related with neurological disability (EDSS score), and the release of these cytokines was less sensitive to glucocorticoid inhibition in MS patients than in control group, mainly after DA addition. In conclusion, our data suggest that DA amplifies glucocorticoid-resistant Th17 phenotype in MS patients, and this phenomenon could be, at least in part, due to its ability to induce IL-6 production by monocytes and CD4(+) T cells.
Collapse
Affiliation(s)
- Thais B Ferreira
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscila O Barros
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruna Teixeira
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiane Cassano
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Newton Centurião
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Taissa M Kasahara
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joana Hygino
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Helcio Alvarenga Filho
- Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regina Alvarenga
- Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Cristina Wing
- Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regis M Andrade
- Department of General Medicine, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Arnaldo F Andrade
- Department of Microbiology, Immunology and Parasitology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cleonice A M Bento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
148
|
Ferreira LRP, Frade AF, Baron MA, Navarro IC, Kalil J, Chevillard C, Cunha-Neto E. Interferon-γ and other inflammatory mediators in cardiomyocyte signaling during Chagas disease cardiomyopathy. World J Cardiol 2014; 6:782-790. [PMID: 25228957 PMCID: PMC4163707 DOI: 10.4330/wjc.v6.i8.782] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 03/29/2014] [Accepted: 06/03/2014] [Indexed: 02/06/2023] Open
Abstract
Chagas disease cardiomyopathy (CCC), the main consequence of Trypanosoma cruzi (T.cruzi) infection, is an inflammatory cardiomyopathy that develops in up to 30% of infected individuals. The heart inflammation in CCC patients is characterized by a Th1 T cell-rich myocarditis with increased production of interferon (IFN)-γ, produced by the CCC myocardial infiltrate and detected at high levels in the periphery. IFN-γ has a central role in the cardiomyocyte signaling during both acute and chronic phases of T.cruzi infection. In this review, we have chosen to focus in its pleiotropic mode of action during CCC, which may ultimately be the strongest driver towards pathological remodeling and heart failure. We describe here the antiparasitic protective and pathogenic dual role of IFN-γ in Chagas disease.
Collapse
|
149
|
Dungan LS, McGuinness NC, Boon L, Lynch MA, Mills KHG. Innate IFN-γ promotes development of experimental autoimmune encephalomyelitis: A role for NK cells and M1 macrophages. Eur J Immunol 2014; 44:2903-17. [DOI: 10.1002/eji.201444612] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/09/2014] [Accepted: 07/18/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Lara S. Dungan
- Immune Regulation Research Group; School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin; Dublin Ireland
| | - Niamh C. McGuinness
- Immune Regulation Research Group; School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin; Dublin Ireland
- Trinity College Institute of Neuroscience; Trinity College Dublin; Dublin Ireland
| | - Louis Boon
- Bioceros Holding BV; Utrecht The Netherlands
| | - Marina A. Lynch
- Trinity College Institute of Neuroscience; Trinity College Dublin; Dublin Ireland
| | - Kingston H. G. Mills
- Immune Regulation Research Group; School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin; Dublin Ireland
| |
Collapse
|
150
|
IL-17 and related cytokines involved in the pathology and immunotherapy of multiple sclerosis: Current and future developments. Cytokine Growth Factor Rev 2014; 25:403-13. [DOI: 10.1016/j.cytogfr.2014.07.013] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|