1
|
Izadjoo S, Moritz KE, Khayrullina G, Bergman EM, Melvin BM, Stinson MW, Paulson SG, McCormack NM, Anderson KN, Lewis LA, Rotty JD, Burnett BG. Key features of the innate immune response is mediated by the immunoproteasome in microglia. RESEARCH SQUARE 2024:rs.3.rs-4467983. [PMID: 38883799 PMCID: PMC11177974 DOI: 10.21203/rs.3.rs-4467983/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Microglia are the resident immune cells of the central nervous system (CNS). We and others have shown that the inflammatory response of microglia is partially regulated by the immunoproteasome, an inducible form of the proteasome responsible for the generation of major histocompatibility complex (MHC) class I epitopes. While the role of the proteasome in the adaptive immune system is well established, emerging evidence suggests the immunoproteasome may have discrete functions in the innate immune response. Here, we show that inhibiting the immunoproteasome reduces the IFNγ-dependent induction of complement activator C1q, suppresses phagocytosis, and alters the cytokine expression profile in a microglial cell line and microglia derived from human inducible pluripotent stem cells. Moreover, we show that the immunoproteasome regulates the degradation of IκBα, a modulator of NF-κB signaling. Finally, we demonstrate that NADH prevents induction of the immunoproteasome, representing a potential pathway to suppress immunoproteasome-dependent immune responses.
Collapse
|
2
|
Moorman CD, Sohn SJ, Phee H. Emerging Therapeutics for Immune Tolerance: Tolerogenic Vaccines, T cell Therapy, and IL-2 Therapy. Front Immunol 2021; 12:657768. [PMID: 33854514 PMCID: PMC8039385 DOI: 10.3389/fimmu.2021.657768] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases affect roughly 5-10% of the total population, with women affected more than men. The standard treatment for autoimmune or autoinflammatory diseases had long been immunosuppressive agents until the advent of immunomodulatory biologic drugs, which aimed at blocking inflammatory mediators, including proinflammatory cytokines. At the frontier of these biologic drugs are TNF-α blockers. These therapies inhibit the proinflammatory action of TNF-α in common autoimmune diseases such as rheumatoid arthritis, psoriasis, ulcerative colitis, and Crohn's disease. TNF-α blockade quickly became the "standard of care" for these autoimmune diseases due to their effectiveness in controlling disease and decreasing patient's adverse risk profiles compared to broad-spectrum immunosuppressive agents. However, anti-TNF-α therapies have limitations, including known adverse safety risk, loss of therapeutic efficacy due to drug resistance, and lack of efficacy in numerous autoimmune diseases, including multiple sclerosis. The next wave of truly transformative therapeutics should aspire to provide a cure by selectively suppressing pathogenic autoantigen-specific immune responses while leaving the rest of the immune system intact to control infectious diseases and malignancies. In this review, we will focus on three main areas of active research in immune tolerance. First, tolerogenic vaccines aiming at robust, lasting autoantigen-specific immune tolerance. Second, T cell therapies using Tregs (either polyclonal, antigen-specific, or genetically engineered to express chimeric antigen receptors) to establish active dominant immune tolerance or T cells (engineered to express chimeric antigen receptors) to delete pathogenic immune cells. Third, IL-2 therapies aiming at expanding immunosuppressive regulatory T cells in vivo.
Collapse
Affiliation(s)
| | | | - Hyewon Phee
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| |
Collapse
|
3
|
Recent Advances in Antigen-Specific Immunotherapies for the Treatment of Multiple Sclerosis. Brain Sci 2020; 10:brainsci10060333. [PMID: 32486045 PMCID: PMC7348736 DOI: 10.3390/brainsci10060333] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system and is considered to be the leading non-traumatic cause of neurological disability in young adults. Current treatments for MS comprise long-term immunosuppressant drugs and disease-modifying therapies (DMTs) designed to alter its progress with the enhanced risk of severe side effects. The Holy Grail for the treatment of MS is to specifically suppress the disease while at the same time allow the immune system to be functionally active against infectious diseases and malignancy. This could be achieved via the development of immunotherapies designed to specifically suppress immune responses to self-antigens (e.g., myelin antigens). The present study attempts to highlight the various antigen-specific immunotherapies developed so far for the treatment of multiple sclerosis (e.g., vaccination with myelin-derived peptides/proteins, plasmid DNA encoding myelin epitopes, tolerogenic dendritic cells pulsed with encephalitogenic epitopes of myelin proteins, attenuated autologous T cells specific for myelin antigens, T cell receptor peptides, carriers loaded/conjugated with myelin immunodominant peptides, etc), focusing on the outcome of their recent preclinical and clinical evaluation, and to shed light on the mechanisms involved in the immunopathogenesis and treatment of multiple sclerosis.
Collapse
|
4
|
Duarte-Silva E, Araújo SMDR, Oliveira WH, Lós DBD, França MERD, Bonfanti AP, Peron G, Thomaz LDL, Verinaud L, Nunes AKDS, Peixoto CA. Sildenafil ameliorates EAE by decreasing apoptosis in the spinal cord of C57BL/6 mice. J Neuroimmunol 2018; 321:125-137. [DOI: 10.1016/j.jneuroim.2018.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/16/2018] [Accepted: 06/04/2018] [Indexed: 12/27/2022]
|
5
|
Kim MJ, Sung JJ, Kim SH, Kim JM, Jeon GS, Mun SK, Ahn SW. The Anti-Inflammatory Effects of Oral-Formulated Tacrolimus in Mice with Experimental Autoimmune Encephalomyelitis. J Korean Med Sci 2017; 32:1502-1507. [PMID: 28776347 PMCID: PMC5546971 DOI: 10.3346/jkms.2017.32.9.1502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 05/28/2017] [Indexed: 11/20/2022] Open
Abstract
Multiple sclerosis (MS) is a T-lymphocyte-mediated autoimmune disease that is characterized by inflammation in the central nervous system (CNS). Although many disease-modifying therapies (DMTs) are presumed effective in patients with MS, studies on the efficacy and safety of DMTs for preventing MS relapse are limited. Therefore, we tested the immunosuppressive anti-inflammatory effects of oral-formulated tacrolimus (FK506) on MS in a mouse model of experimental autoimmune encephalomyelitis (EAE). The mice were randomly divided into 3 experimental groups: an untreated EAE group, a low-dose tacrolimus-treated EAE group, and a high-dose tacrolimus-treated EAE group. After autoimmunization of the EAE mice with myelin oligodendrocyte glycoprotein, symptom severity scores, immunohistochemistry of the myelination of the spinal cord, and western blotting were used to evaluate the EAE mice. After the autoimmunization, the symptom scores of each EAE group significantly differed at times. The group treated with the larger tacrolimus dose had the lowest symptom scores. The tacrolimus-treated EAE groups exhibited less demyelination and inflammation and weak immunoreactivity for all of the immunization biomarkers. Our results revealed that oral-formulated tacrolimus inhibited the autoimmunization in MS pathogenesis by inactivating inflammatory cells.
Collapse
Affiliation(s)
- Myung Jin Kim
- Department of Neurology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jung Joon Sung
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Hyun Kim
- Department of Neurology, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Korea
| | - Jeong Min Kim
- Department of Neurology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| | - Gye Sun Jeon
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seog Kyun Mun
- Department of Otorhinolaryngology, Head and Neck Surgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Suk Won Ahn
- Department of Neurology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea.
| |
Collapse
|
6
|
Multiple Sclerosis: Immunopathology and Treatment Update. Brain Sci 2017; 7:brainsci7070078. [PMID: 28686222 PMCID: PMC5532591 DOI: 10.3390/brainsci7070078] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 02/07/2023] Open
Abstract
The treatment of multiple sclerosis (MS) has changed over the last 20 years. All immunotherapeutic drugs target relapsing remitting MS (RRMS) and it still remains a medical challenge in MS to develop a treatment for progressive forms. The most common injectable disease-modifying therapies in RRMS include β-interferons 1a or 1b and glatiramer acetate. However, one of the major challenges of injectable disease-modifying therapies has been poor treatment adherence with approximately 50% of patients discontinuing the therapy within the first year. Herein, we go back to the basics to understand the immunopathophysiology of MS to gain insights in the development of new improved drug treatments. We present current disease-modifying therapies (interferons, glatiramer acetate, dimethyl fumarate, teriflunomide, fingolimod, mitoxantrone), humanized monoclonal antibodies (natalizumab, ofatumumb, ocrelizumab, alentuzumab, daclizumab) and emerging immune modulating approaches (stem cells, DNA vaccines, nanoparticles, altered peptide ligands) for the treatment of MS.
Collapse
|
7
|
Engineered erythrocytes covalently linked to antigenic peptides can protect against autoimmune disease. Proc Natl Acad Sci U S A 2017; 114:3157-3162. [PMID: 28270614 DOI: 10.1073/pnas.1701746114] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Current therapies for autoimmune diseases rely on traditional immunosuppressive medications that expose patients to an increased risk of opportunistic infections and other complications. Immunoregulatory interventions that act prophylactically or therapeutically to induce antigen-specific tolerance might overcome these obstacles. Here we use the transpeptidase sortase to covalently attach disease-associated autoantigens to genetically engineered and to unmodified red blood cells as a means of inducing antigen-specific tolerance. This approach blunts the contribution to immunity of major subsets of immune effector cells (B cells, CD4+ and CD8+ T cells) in an antigen-specific manner. Transfusion of red blood cells expressing self-antigen epitopes can alleviate and even prevent signs of disease in experimental autoimmune encephalomyelitis, as well as maintain normoglycemia in a mouse model of type 1 diabetes.
Collapse
|
8
|
Peng W. G-CSF treatment promotes apoptosis of autoreactive T cells to restrict the inflammatory cascade and accelerate recovery in experimental allergic encephalomyelitis. Exp Neurol 2017; 289:73-84. [DOI: 10.1016/j.expneurol.2016.10.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 09/29/2016] [Accepted: 10/18/2016] [Indexed: 12/11/2022]
|
9
|
Li C, Zhou X, Zhong Y, Li C, Dong A, He Z, Zhang S, Wang B. A Recombinant G Protein Plus Cyclosporine A-Based Respiratory Syncytial Virus Vaccine Elicits Humoral and Regulatory T Cell Responses against Infection without Vaccine-Enhanced Disease. THE JOURNAL OF IMMUNOLOGY 2016; 196:1721-31. [PMID: 26792805 DOI: 10.4049/jimmunol.1502103] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/14/2015] [Indexed: 12/28/2022]
Abstract
Respiratory syncytial virus (RSV) infection can cause severe disease in the lower respiratory tract of infants and older people. Vaccination with a formalin-inactivated RSV vaccine (FI-RSV) and subsequent RSV infection has led to mild to severe pneumonia with two deaths among vaccinees. The vaccine-enhanced disease (VED) was recently demonstrated to be due to an elevated level of Th2 cell responses following loss of regulatory T (Treg) cells from the lungs. To induce high levels of neutralizing Abs and minimize pathogenic T cell responses, we developed a novel strategy of immunizing animals with a recombinant RSV G protein together with cyclosporine A. This novel vaccine induced not only a higher level of neutralizing Abs against RSV infection, but, most importantly, also significantly higher levels of Treg cells that suppressed VED in the lung after RSV infection. The induced responses provided protection against RSV challenge with no sign of pneumonia or bronchitis. Treg cell production of IL-10 was one of the key factors to suppress VED. These finding indicate that G protein plus cyclosporine A could be a promising vaccine against RSV infection in children and older people.
Collapse
Affiliation(s)
- Chaofan Li
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Xian Zhou
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Yiwei Zhong
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Changgui Li
- Division for Respiratory Viral Vaccines of National Institutes for Food and Drug Control, Beijing 100050, China; and
| | - Aihua Dong
- Beijing Advaccine Biotechnology Company, Ltd., Beijing 100085, China
| | - Zhonghuai He
- Beijing Advaccine Biotechnology Company, Ltd., Beijing 100085, China
| | - Shuren Zhang
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Bin Wang
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Medical College, Fudan University, Shanghai 201508, China;
| |
Collapse
|
10
|
Wang S, Yu Y, Geng S, Wang D, Zhang L, Xie X, Wu B, Li C, Xu H, Li X, Hu Y, Zhang L, Kaether C, Wang B. A coimmunization vaccine of Aβ42 ameliorates cognitive deficits without brain inflammation in an Alzheimer's disease model. ALZHEIMERS RESEARCH & THERAPY 2014; 6:26. [PMID: 24987466 PMCID: PMC4075150 DOI: 10.1186/alzrt256] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 04/07/2014] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Vaccination against amyloid-β protein (Aβ42) induces high levels of antibody, making it a promising strategy for treating Alzheimer's disease (AD). One drawback in the past was that clinical trial approval was withheld because of speculation that the Aβ42 vaccine induces CD4(+) T cell infiltrations into the central nervous system. To reduce T-cell activation while concomitantly maintaining high anti-Aβ42 titers is a great challenge in immunology. METHODS We aimed to demonstrate that coimmunization with Aβ42 protein and expression plasmid can be beneficial in a mouse AD model and can prevent inflammation. We immunized the AD mice with the coimmunization vaccine and assessed behavior change and Aβ42 deposition. Furthermore, to determine the safety of the coimmunization vaccine, we used an induced Aβ42-EAE model to mimic the meningoencephalitis that happened in the AN-1792 vaccine clinical phase II trial and tested whether the coimmunization vaccine could ameliorate T-cell-mediated brain inflammation. RESULTS The coimmunization vaccination reduced Aβ plaques and significantly ameliorated cognitive deficit while inhibiting T-cell-mediated brain inflammation and infiltration. These studies demonstrate that the coimmunization strategy that we describe in this article can ameliorate AD pathology without notable adverse effects in mice. CONCLUSIONS A coimmunization strategy leading to the development of a safe immunotherapeutic/preventive protocol against AD in humans is warranted.
Collapse
Affiliation(s)
- Shuang Wang
- Key Laboratory of Medical Molecular Virology of MOH and MOE, Fudan University Shanghai Medical College, 131 Dong An Road, Shanghai 200032, China ; State Key Laboratory for Agro-Biotechnology and College of Biologic Sciences, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Yang Yu
- State Key Laboratory for Agro-Biotechnology and College of Biologic Sciences, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China ; Present address: MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuang Geng
- Key Laboratory of Medical Molecular Virology of MOH and MOE, Fudan University Shanghai Medical College, 131 Dong An Road, Shanghai 200032, China
| | - Dongmei Wang
- Chinese Academy of Medical Sciences & Comparative Medical Center, 5 South Panjiayuan, Beijing 100021, China
| | - Li Zhang
- Chinese Academy of Medical Sciences & Comparative Medical Center, 5 South Panjiayuan, Beijing 100021, China
| | - Xiaoping Xie
- State Key Laboratory for Agro-Biotechnology and College of Biologic Sciences, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Bing Wu
- State Key Laboratory for Agro-Biotechnology and College of Biologic Sciences, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Chaofan Li
- Key Laboratory of Medical Molecular Virology of MOH and MOE, Fudan University Shanghai Medical College, 131 Dong An Road, Shanghai 200032, China
| | - Hanqian Xu
- State Key Laboratory for Agro-Biotechnology and College of Biologic Sciences, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Xiaolin Li
- Institute for Age Research, Fritz Lipmann Institute, Beutenbergstraße 11, Jena D-07745, Germany
| | - Yanxin Hu
- Department of Pathology, College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Lianfeng Zhang
- Chinese Academy of Medical Sciences & Comparative Medical Center, 5 South Panjiayuan, Beijing 100021, China
| | - Christoph Kaether
- Institute for Age Research, Fritz Lipmann Institute, Beutenbergstraße 11, Jena D-07745, Germany
| | - Bin Wang
- Key Laboratory of Medical Molecular Virology of MOH and MOE, Fudan University Shanghai Medical College, 131 Dong An Road, Shanghai 200032, China
| |
Collapse
|
11
|
Gülden E, Ihira M, Ohashi A, Reinbeck AL, Freudenberg MA, Kolb H, Burkart V. Toll-like receptor 4 deficiency accelerates the development of insulin-deficient diabetes in non-obese diabetic mice. PLoS One 2013; 8:e75385. [PMID: 24086519 PMCID: PMC3781027 DOI: 10.1371/journal.pone.0075385] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 08/14/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND/OBJECTIVE Toll-like receptors (TLR) mediate the recognition of microbial constituents and stress-induced endogenous ligands by the immune system. They may also be involved in the maintenance or break down of tolerance against autologous antigens. The aim of our investigation was to study the consequence of TLR4 deficiency on the development of insulin-deficient diabetes in the NOD mouse. METHODS The TLR4 defect of the C57BL/10ScN mouse was backcrossed onto the NOD background and the effect of TLR4 deficiency on diabetes development was analysed by in vivo and in vitro studies. RESULTS Compared to animals with wildtype TLR4 expression (TLR4(+/+)), female NOD mice carrying a homozygous TLR4 defect (TLR4(-/-)), showed significant acceleration of diabetes development, with a younger age at diabetes onset (TLR4 (+/+) 177±22 d, TLR(-/-): 118±21 d; p<0.01). Pancreata of 120 d old TLR4(-/-) NOD mice revealed increased proportions of islets with advanced stages of immune cell infiltration compared to TLR4(+/+) mice (p<0.05). TLR4 deficiency did not affect the susceptibility of islet cells to the beta cell damaging mediators nitric oxide or the inflammatory cytokines tumor necrosis factor alpha, interleukin-1 beta and interferon gamma. The lack of TLR4 further had no effect on the frequency of regulatory T-cells but reduced their capacity to inhibit T-cell proliferation. CONCLUSIONS Our findings demonstrate that TLR4 deficiency results in an acceleration of diabetes development and immune cell infiltration of islets in NOD mice. We conclude that TLR4 is involved in the progression of the insulitis process thereby controlling the development of insulin-deficient diabetes in NOD mice.
Collapse
Affiliation(s)
- Elke Gülden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Member of the German Center for Diabetes Research (DZD), Düsseldorf, Germany
| | - Masaru Ihira
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Member of the German Center for Diabetes Research (DZD), Düsseldorf, Germany
| | - Atsushi Ohashi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Member of the German Center for Diabetes Research (DZD), Düsseldorf, Germany
| | - Anna Lena Reinbeck
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Member of the German Center for Diabetes Research (DZD), Düsseldorf, Germany
| | - Marina A. Freudenberg
- Department of Developmental Immunology, Max-Planck-Institute of Immunbiology and Epigenetics, Freiburg, Germany
| | - Hubert Kolb
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Member of the German Center for Diabetes Research (DZD), Düsseldorf, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Member of the German Center for Diabetes Research (DZD), Düsseldorf, Germany
| |
Collapse
|
12
|
Thomé R, Moraes AS, Bombeiro AL, Farias ADS, Francelin C, da Costa TA, Di Gangi R, dos Santos LMB, de Oliveira ALR, Verinaud L. Chloroquine treatment enhances regulatory T cells and reduces the severity of experimental autoimmune encephalomyelitis. PLoS One 2013; 8:e65913. [PMID: 23799062 PMCID: PMC3683039 DOI: 10.1371/journal.pone.0065913] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 04/30/2013] [Indexed: 12/13/2022] Open
Abstract
Background The modulation of inflammatory processes is a necessary step, mostly orchestrated by regulatory T (Treg) cells and suppressive Dendritic Cells (DCs), to prevent the development of deleterious responses and autoimmune diseases. Therapies that focused on adoptive transfer of Treg cells or their expansion in vivo achieved great success in controlling inflammation in several experimental models. Chloroquine (CQ), an anti-malarial drug, was shown to reduce inflammation, although the mechanisms are still obscure. In this context, we aimed to access whether chloroquine treatment alters the frequency of Treg cells and DCs in normal mice. In addition, the effects of the prophylactic and therapeutic treatment with CQ on Experimental Autoimmune Encephalomyelitis (EAE), an experimental model for human Multiple Sclerosis, was investigated as well. Methodology/Principal Findings EAE was induced in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein (MOG35–55) peptide. C57BL/6 mice were intraperitoneally treated with chloroquine. Results show that the CQ treatment provoked an increase in Treg cells frequency as well as a decrease in DCs. We next evaluated whether prophylactic CQ administration is capable of reducing the clinical and histopathological signs of EAE. Our results demonstrated that CQ-treated mice developed mild EAE compared to controls that was associated with lower infiltration of inflammatory cells in the central nervous system CNS) and increased frequency of Treg cells. Also, proliferation of MOG35–55-reactive T cells was significantly inhibited by chloroquine treatment. Similar results were observed when chloroquine was administrated after disease onset. Conclusion We show for the first time that CQ treatment promotes the expansion of Treg cells, corroborating previous reports indicating that chloroquine has immunomodulatory properties. Our results also show that CQ treatment suppress the inflammation in the CNS of EAE-inflicted mice, both in prophylactic and therapeutic approaches. We hypothesized that the increased number of regulatory T cells induced by the CQ treatment is involved in the reduction of the clinical signs of EAE.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Cells, Cultured
- Central Nervous System/drug effects
- Central Nervous System/immunology
- Central Nervous System/pathology
- Chloroquine/pharmacology
- Chloroquine/therapeutic use
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Humans
- Immunologic Factors/pharmacology
- Immunologic Factors/therapeutic use
- Interferon-gamma/metabolism
- Interleukin-17/metabolism
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis/drug therapy
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/transplantation
Collapse
Affiliation(s)
- Rodolfo Thomé
- Department of Structural and Functional Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Adriel S. Moraes
- Department of Genetics, Evolution and Bioagents, University of Campinas, Campinas, São Paulo, Brazil
| | - André Luis Bombeiro
- Department of Structural and Functional Biology, University of Campinas, Campinas, São Paulo, Brazil
| | | | - Carolina Francelin
- Department of Structural and Functional Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Thiago Alves da Costa
- Department of Structural and Functional Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Rosária Di Gangi
- Department of Structural and Functional Biology, University of Campinas, Campinas, São Paulo, Brazil
| | | | | | - Liana Verinaud
- Department of Structural and Functional Biology, University of Campinas, Campinas, São Paulo, Brazil
- * E-mail:
| |
Collapse
|