1
|
Oertel FC, Hastermann M, Paul F. Delimiting MOGAD as a disease entity using translational imaging. Front Neurol 2023; 14:1216477. [PMID: 38333186 PMCID: PMC10851159 DOI: 10.3389/fneur.2023.1216477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/23/2023] [Indexed: 02/10/2024] Open
Abstract
The first formal consensus diagnostic criteria for myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) were recently proposed. Yet, the distinction of MOGAD-defining characteristics from characteristics of its important differential diagnoses such as multiple sclerosis (MS) and aquaporin-4 antibody seropositive neuromyelitis optica spectrum disorder (NMOSD) is still obstructed. In preclinical research, MOG antibody-based animal models were used for decades to derive knowledge about MS. In clinical research, people with MOGAD have been combined into cohorts with other diagnoses. Thus, it remains unclear to which extent the generated knowledge is specifically applicable to MOGAD. Translational research can contribute to identifying MOGAD characteristic features by establishing imaging methods and outcome parameters on proven pathophysiological grounds. This article reviews suitable animal models for translational MOGAD research and the current state and prospect of translational imaging in MOGAD.
Collapse
Affiliation(s)
- Frederike Cosima Oertel
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Hastermann
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max-Delbrück-Centrum für Molekulare Medizin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
2
|
Gaignage M, Uyttenhove C, Jones LL, Bourdeaux C, Chéou P, Mandour MF, Coutelier JP, Vignali DAA, Van Snick J. Novel antibodies that selectively block mouse IL-12 enable the re-evaluation of the role of IL-12 in immune protection and pathology. Eur J Immunol 2021; 51:1482-1493. [PMID: 33788263 DOI: 10.1002/eji.202048936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/11/2021] [Accepted: 03/23/2021] [Indexed: 01/01/2023]
Abstract
The dimeric cytokine IL-12 is important in the control of various infections but also contributes to the pathology of certain diseases making it a potential target for therapy. However, its specific inhibition with antibodies is complicated by the fact that its two subunits are present in other cytokines: p40 in IL-23 and p35 in IL-35. This has led to erroneous conclusions like the alleged implication of IL-12 in experimental autoimmune encephalomyelitis (EAE). Here, we report the development of a mouse anti-mouse IL-12 vaccine and the production of monoclonal antibodies (mAbs) that do not react with p40 or p35 (in IL-35) but specifically recognize and functionally inhibit the IL-12 heterodimer. Using one of these mAbs, MM12A1.6, that strongly inhibited IFN-γ production and LPS-induced septic shock after viral infection, we demonstrate the critical role played by IL-12 in the rejection of male skin graft by female C57BL/6 syngeneic recipients and in the clearance of an immunogenic mastocytoma tumor variant by DBA/2 mice, but not in a parent to F1 immune aggression model nor in MOG-induced EAE, which was clearly prevented by anti-p40 mAb C17.8. Given this selective inhibition of IL-12, these mAbs provide new options for reassessing IL-12 function in vivo.
Collapse
Affiliation(s)
| | - Catherine Uyttenhove
- de Duve Institute, Université de Louvain, Brussels, Belgium.,Ludwig Cancer Research, Brussels, Belgium
| | - Lindsay L Jones
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Paméla Chéou
- de Duve Institute, Université de Louvain, Brussels, Belgium
| | - Mohamed F Mandour
- de Duve Institute, Université de Louvain, Brussels, Belgium.,Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Dario A A Vignali
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Jacques Van Snick
- de Duve Institute, Université de Louvain, Brussels, Belgium.,Ludwig Cancer Research, Brussels, Belgium
| |
Collapse
|
3
|
Ehteshamfar S, Akhbari M, Afshari JT, Seyedi M, Nikfar B, Shapouri‐Moghaddam A, Ghanbarzadeh E, Momtazi‐Borojeni AA. Anti-inflammatory and immune-modulatory impacts of berberine on activation of autoreactive T cells in autoimmune inflammation. J Cell Mol Med 2020; 24:13573-13588. [PMID: 33135395 PMCID: PMC7754052 DOI: 10.1111/jcmm.16049] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Autoreactive inflammatory CD4+ T cells, such as T helper (Th)1 and Th17 subtypes, have been found to associate with the pathogenesis of autoimmune disorders. On the other hand, CD4+ Foxp3+ T regulatory (Treg) cells are crucial for the immune tolerance and have a critical role in the suppression of the excessive immune and inflammatory response promoted by these Th cells. In contrast, dendritic cells (DCs) and macrophages are immune cells that through their inflammatory functions promote autoreactive T-cell responses in autoimmune conditions. In recent years, there has been increasing attention to exploring effective immunomodulatory or anti-inflammatory agents from the herbal collection of traditional medicine. Berberine, an isoquinoline alkaloid, is one of the main active ingredients extracted from medicinal herbs and has been shown to exert various biological and pharmacological effects that are suggested to be mainly attributed to its anti-inflammatory and immunomodulatory properties. Several lines of experimental study have recently investigated the therapeutic potential of berberine for treating autoimmune conditions in animal models of human autoimmune diseases. Here, we aimed to seek mechanisms underlying immunomodulatory and anti-inflammatory effects of berberine on autoreactive inflammatory responses in autoimmune conditions. Reported data reveal that berberine can directly suppress functions and differentiation of pro-inflammatory Th1 and Th17 cells, and indirectly decrease Th cell-mediated inflammation through modulating or suppressing other cells assisting autoreactive inflammation, such as Tregs, DCs and macrophages.
Collapse
Affiliation(s)
- Seyed‐Morteza Ehteshamfar
- Department of ImmunologyFaculty of MedicineBuAli Research InstituteMashhad University of Medical SciencesMashhadIran
| | - Masoume Akhbari
- Department of Molecular MedicineSchool of MedicineQazvin University of Medical SciencesQazvinIran
| | - Jalil Tavakol Afshari
- Department of ImmunologyFaculty of MedicineBuAli Research InstituteMashhad University of Medical SciencesMashhadIran
| | | | - Banafsheh Nikfar
- Pars Advanced and Minimally Invasive Medical Manners Research CenterPars HospitalIran University of Medical SciencesTehranIran
| | - Abbas Shapouri‐Moghaddam
- Department of ImmunologyFaculty of MedicineBuAli Research InstituteMashhad University of Medical SciencesMashhadIran
| | | | | |
Collapse
|
4
|
Chen J, Liu X, Zhong Y. Interleukin-17A: The Key Cytokine in Neurodegenerative Diseases. Front Aging Neurosci 2020; 12:566922. [PMID: 33132897 PMCID: PMC7550684 DOI: 10.3389/fnagi.2020.566922] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are characterized by the loss of neurons and/or myelin sheath, which deteriorate over time and cause dysfunction. Interleukin 17A is the signature cytokine of a subset of CD4+ helper T cells known as Th17 cells, and the IL-17 cytokine family contains six cytokines and five receptors. Recently, several studies have suggested a pivotal role for the interleukin-17A (IL-17A) cytokine family in human inflammatory or autoimmune diseases and neurodegenerative diseases, including psoriasis, rheumatoid arthritis (RA), Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and glaucoma. Studies in recent years have shown that the mechanism of action of IL-17A is more subtle than simply causing inflammation. Although the specific mechanism of IL-17A in neurodegenerative diseases is still controversial, it is generally accepted now that IL-17A causes diseases by activating glial cells. In this review article, we will focus on the function of IL-17A, in particular the proposed roles of IL-17A, in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Junjue Chen
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohong Liu
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yisheng Zhong
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Zare F, Amiri MM, Hadinedoushan H, Dehghan-Manshadi M, Mansouri F, Fesahat F, Saboor-Yaraghi AA. Contraceptive and molecular function of a novel recombinant vaccine based human leukemia inhibitory factor on Balb/c mice: An experimental in vivo study. J Reprod Immunol 2020; 142:103195. [PMID: 32927320 DOI: 10.1016/j.jri.2020.103195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 08/09/2020] [Accepted: 08/25/2020] [Indexed: 11/24/2022]
Abstract
The functional competence of leukemia inhibitory factor (LIF), as immunocontraceptive vaccine in mice, was investigated. Balb/c mice were divided into two groups of vaccinated and controls. The recombinant human LIF (rhLIF) protein and phosphate buffer saline was emulsified with Freund's adjuvant and injected into vaccinated and control groups, respectively. Theinhibition of implantation was evaluated in mice uterine. The concentration of secreted interferon-γ (IFN-γ) and interleukin (IL)-4 were measured in cultured splenocyte of mice stimulated by rhLIF. The expressions of immune responsive gene 1 (IRG-1), cochlin (COCH), amphiregulin(Ar), and heparin-binding EGF-like growth factor (HB-EGF) genes were determined. Mice were assessed for inhibition of fertility after delivery, reversibility of immune response against rhLIF, and survival rate. Active immunization of mice with rhLIF resulted in reduction of the implantation and fertility rate up to 80.49% and 75%, respectively. All mice produced a high titer of anti-rhLIF antibodies in serums and vaginal fluids washes after 16 weeks; however, these antibodies were cleared from vaginal fluid washes after six months. A significant down-regulation in mRNA levels of IRG-1, Ar and HB-EGF was observed in vaccinated group compared to controls; however, no significant change in the expression profile of cochlin gene was detected. The results showed that rhLIF prevented pregnancy in a high percentage of female mice. Although the immunization of female Balb/c mice with rhLIF inhibited fertility and expression of genes associated with this molecule, further studies are needed to support this protein as a suitable candidate for contraceptive vaccine in mammals.
Collapse
Affiliation(s)
- Fateme Zare
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Hadinedoushan
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahdi Dehghan-Manshadi
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mansouri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Fesahat
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Akbar Saboor-Yaraghi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Alvarez P, Augustín JJ, Tamayo E, Iglesias M, Acinas O, Mendiguren MA, Vázquez JA, Genre F, San Segundo D, Merino J, Merino R. Therapeutic Effects of Anti-Bone Morphogenetic Protein and Activin Membrane-Bound Inhibitor Treatment in Psoriasis and Arthritis. Arthritis Rheumatol 2020; 72:1547-1558. [PMID: 32249544 DOI: 10.1002/art.41272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 03/24/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The transforming growth factor β (TGFβ) inhibitor BAMBI (bone morphogenetic protein and activin membrane-bound inhibitor) has been shown to control differentiation of CD4+ T lymphocytes into either tolerogenic Treg cells or pathogenic Th17 cells, through the regulation of TGFβ and interleukin-2 (IL-2) signaling strength. The present study was undertaken to explore the potential beneficial effects of this strategy of pharmacologic inhibition using novel anti-BAMBI monoclonal antibodies (mAb) in different experimental murine models of chronic skin and joint inflammatory/autoimmune disease. METHODS Development of Saccharomyces cerevisiae mannan-induced psoriatic arthritis (MIP) (n = 18-30 mice per group), imiquimod-induced skin psoriasis (n = 20-30 mice per group), or type II collagen-induced arthritis (CIA) (n = 13-16 mice per group) was analyzed in a total of 2-5 different experiments with either wild-type (WT) or BAMBI-deficient B10.RIII mice that were left untreated or treated with mAb B101.37 (mouse IgG1 anti-BAMBI), a mouse IgG1 anti-TNP isotype control, anti-CD25, or anti-TGFβ mAb. RESULTS Treatment of normal mice with IgG1 anti-BAMBI mAb clone B101.37 led to expansion of Treg cells in vivo, and had both preventive and therapeutic effects in mice with MIP (each P < 0.05 versus controls). The conferred protection against disease progression was found to be mediated by Treg cells, which controlled the activation and expansion of pathogenic IL-17-producing cells, and was dependent on the level of TGFβ activity. Furthermore, treatment with B101.37 mAb blocked both the development of skin psoriasis induced by imiquimod and the development of CIA in mice (each P < 0.05 versus controls). Finally, pharmacologic inhibition of BAMBI with the IgM anti-BAMBI mAb B143.14 also potentiated the suppressive activity of Treg cells in vitro (P < 0.001 versus controls). CONCLUSION These results in murine models identify BAMBI as a promising new therapeutic target for chronic inflammatory diseases and other pathologic conditions modulated by Treg cells.
Collapse
Affiliation(s)
- Pilar Alvarez
- Instituto de Biomedicina y Biotecnología de Cantabria, CSIC-Universidad de Cantabria-SODERCAN, Santander, Spain
| | | | - Esther Tamayo
- IDIVAL and Universidad de Cantabria, Santander, Spain
| | | | - Olga Acinas
- Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | | | | | | | | | - Jesús Merino
- IDIVAL and Universidad de Cantabria, Santander, Spain
| | - Ramón Merino
- IDIVAL and Instituto de Biomedicina y Biotecnología de Cantabria, CSIC-Universidad de Cantabria-SODERCAN, Santander, Spain
| |
Collapse
|
7
|
Karpus WJ. Cytokines and Chemokines in the Pathogenesis of Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2020; 204:316-326. [PMID: 31907274 DOI: 10.4049/jimmunol.1900914] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/13/2019] [Indexed: 12/25/2022]
Abstract
Experimental autoimmune encephalomyelitis is a CD4+ T cell-mediated demyelinating disease of the CNS that serves as a model for multiple sclerosis. Cytokines and chemokines shape Th1 and Th17 effector responses as well as regulate migration of leukocytes to the CNS during disease. The CNS cellular infiltrate consists of Ag-specific and nonspecific CD4+ and CD8+ T cells, neutrophils, B cells, monocytes, macrophages, and dendritic cells. The mechanism of immune-mediated inflammation in experimental autoimmune encephalomyelitis has been extensively studied in an effort to develop therapeutic modalities for multiple sclerosis and, indeed, has provided insight in modern drug discovery. The present Brief Review highlights critical pathogenic aspects of cytokines and chemokines involved in generation of effector T cell responses and migration of inflammatory cells to the CNS. Select cytokines and chemokines are certainly important in the regulatory response, which involves T regulatory, B regulatory, and myeloid-derived suppressor cells. However, that discussion is beyond the scope of this brief review.
Collapse
Affiliation(s)
- William J Karpus
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
8
|
Creyns B, Cremer J, Hoshino T, Geboes K, de Hertogh G, Ferrante M, Vermeire S, Ceuppens JL, Van Assche G, Breynaert C. Fibrogenesis in Chronic DSS Colitis is Not Influenced by Neutralisation of Regulatory T Cells, of Major T Helper Cytokines or Absence of IL-13. Sci Rep 2019; 9:10064. [PMID: 31296924 PMCID: PMC6624199 DOI: 10.1038/s41598-019-46472-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023] Open
Abstract
Mechanisms underlying fibrogenesis in chronic colitis are largely unknown. There is an urgent need for clinical markers and identification of targets to prevent, treat and limit intestinal fibrosis. This study investigated the contribution of major T cell cytokines and T regulatory cells (Tregs) to inflammation and fibrosis induced in a model of experimental colitis by oral intake of dextran sodium sulphate (DSS) in wild type and IL-13 knock-out C57Bl/6 mice. Inflammation and fibrosis were scored by macroscopic and histological examination and fibrosis was quantified by hydroxyproline. Numbers of Tregs and IFN-γ+, IL-13+ and IL-17A+ CD4+ T helper (Th) cells in mesenteric lymph nodes increased during chronic DSS administration and mRNA for IFN-γ and IL-17 in the inflamed colon tissue was upregulated. However, antibody-mediated neutralisation of IFN-γ or IL-17A/F in a therapeutic setting had no effect on chronic intestinal inflammation and fibrosis. Antibody-mediated depletion of Tregs did not enhance fibrosis, nor did IL-13 deficiency have an effect on the fibrotic disease. These data argue against an important contribution of Tregs and of the cytokines IFN-γ, IL-13, IL-17A, IL-17F in the induction and/or control of fibrosis in this Crohn's disease like murine model.
Collapse
Affiliation(s)
- Brecht Creyns
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium.,KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven, Belgium
| | - Jonathan Cremer
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium.,KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven, Belgium
| | - Tomoaki Hoshino
- Division of Respirology, Neurology and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Karel Geboes
- KU Leuven, Department of Imaging and Pathology, Translational Cell & Tissue Research, Leuven, Belgium
| | - Gert de Hertogh
- KU Leuven, Department of Imaging and Pathology, Translational Cell & Tissue Research, Leuven, Belgium
| | - Marc Ferrante
- KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven, Belgium.,University Hospitals Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
| | - Séverine Vermeire
- KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven, Belgium.,University Hospitals Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
| | - Jan L Ceuppens
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium
| | - Gert Van Assche
- KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven, Belgium.,University Hospitals Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
| | - Christine Breynaert
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium. .,University Hospitals Leuven, Department of General Internal Medicine, Leuven, Belgium.
| |
Collapse
|
9
|
Uyttenhove C, Gaignage M, Donckers D, Nasr Z, Cheou P, van Snick J, D'Auria L, van Pesch V. Prophylactic treatment against GM-CSF, but not IL-17, abolishes relapses in a chronic murine model of multiple sclerosis. Eur J Immunol 2018; 48:1883-1891. [PMID: 30216414 DOI: 10.1002/eji.201847580] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/18/2018] [Accepted: 09/12/2018] [Indexed: 01/25/2023]
Abstract
The pathogenic role of IL-17 and GM-CSF has been unravelled in experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS). However, in most models, EAE is characterised by a monophasic attack which is not representative of the relapsing nature nor the chronicity displayed in MS. Here, we used proteolipid protein peptide (PLP139-151 ) to trigger EAE-relapses (EAE-II) in SJL mice that had recovered from a primary-EAE episode (EAE-I). This procedure resulted in severe and irreversible disease that, unlike EAE-I, was not abolished by anti-IL-17-mAb. In contrast, prophylactic anti-GM-CSF-mAb treatment prevented EAE-I and -II. Strikingly, the expression of T-cell transcription factors and cytokines/chemokines in mice treated with anti-GM-CSF during both EAE episodes was silenced. Anti-GM-CSF-mAb treatment administered only during EAE-II did not completely prevent relapses but mice ultimately reached full recovery. Anti-GM-CSF treatment also strongly impaired and ultimately resolved monophasic MOG35-55 -induced EAE in C57Bl/6 mice. In such protected mice, anti-GM-CSF treatment also prevented a further relapse induced by MOG-revaccination. These results underscore the critical role of GM-CSF on pro-inflammatory mediator production. Furthermore, we observed a strong preventive and curative effect of anti-GM-CSF neutralisation in two EAE models, relapsing and chronic. Altogether these findings are relevant for further MS research.
Collapse
Affiliation(s)
- Catherine Uyttenhove
- Ludwig Cancer Research, Brussels Branch, Brussels, Belgium.,de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Mélanie Gaignage
- Ludwig Cancer Research, Brussels Branch, Brussels, Belgium.,de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Dominique Donckers
- Ludwig Cancer Research, Brussels Branch, Brussels, Belgium.,de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Zakia Nasr
- Neurochemistry Unit, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Pamela Cheou
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Jacques van Snick
- Ludwig Cancer Research, Brussels Branch, Brussels, Belgium.,de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Ludovic D'Auria
- Neurochemistry Unit, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Vincent van Pesch
- Neurochemistry Unit, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
10
|
Monoclonal Antibodies in Preclinical EAE Models of Multiple Sclerosis: A Systematic Review. Int J Mol Sci 2017; 18:ijms18091992. [PMID: 28926943 PMCID: PMC5618641 DOI: 10.3390/ijms18091992] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 02/08/2023] Open
Abstract
Monoclonal antibodies (mAb) are promising therapeutics in multiple sclerosis and multiple new candidates have been developed, hence increasing the need for some agreement for preclinical mAb studies. We systematically analyzed publications of experimental autoimmune encephalomyelitis (EAE) studies showing effects of monoclonal antibodies. A PubMed search retrieved 570 records, out of which 122 studies with 253 experiments were eligible based on experimental design, number of animals and presentation of time courses of EAE scores. Analysis of EAE models, treatment schedules, single and total doses, routes of administration, and onset of treatment from pre-immunization up to 35 days after immunization revealed high heterogeneity. Total doses ranged from 0.1 to 360 mg/kg for observation times of up to 35 days after immunization. About half of experiments (142/253) used total doses of 10-70 mg/kg. Employing this range, we tested anti-Itga4 as a reference mAb at varying schedules and got no, mild or substantial EAE-score reductions, depending on the mouse strain and onset of the treatment. The result agrees with the range of outcomes achieved in 10 reported anti-Itga4 experiments. Studies comparing low and high doses of various mAbs or early vs. late onset of treatment did not reveal dose-effect or timing-effect associations, with a tendency towards better outcomes with preventive treatments starting within the first week after immunization. The systematic comparison allows for extraction of some "common" design characteristics, which may be helpful to further assess the efficacy of mAbs and role of specific targets in preclinical models of multiple sclerosis.
Collapse
|
11
|
Rodríguez-Álvarez Y, Morera-Díaz Y, Gerónimo-Pérez H, Castro-Velazco J, Martínez-Castillo R, Puente-Pérez P, Besada-Pérez V, Hardy-Rando E, Chico-Capote A, Martínez-Cordovez K, Santos-Savio A. Active immunization with human interleukin-15 induces neutralizing antibodies in non-human primates. BMC Immunol 2016; 17:30. [PMID: 27671547 PMCID: PMC5036325 DOI: 10.1186/s12865-016-0168-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/12/2016] [Indexed: 02/07/2023] Open
Abstract
Background Interleukin-15 is an immunostimulatory cytokine overexpressed in several autoimmune and inflammatory diseases such as Rheumatoid Arthritis, psoriasis and ulcerative colitis; thus, inhibition of IL-15-induced signaling could be clinically beneficial in these disorders. Our approach to neutralize IL-15 consisted in active immunization with structurally modified human IL-15 (mhIL-15) with the aim to induce neutralizing antibodies against native IL-15. In the present study, we characterized the antibody response in Macaca fascicularis, non-human primates that were immunized with a vaccine candidate containing mhIL-15 in Aluminum hydroxide (Alum), Montanide and Incomplete Freund’s Adjuvant. Results Immunization with mhIL-15 elicited a specific antibodies response that neutralized native IL-15-dependent biologic activity in a CTLL-2 cell proliferation assay. The highest neutralizing response was obtained in macaques immunized with mhIL-15 adjuvanted in Alum. This response, which was shown to be transient, also inhibited the activity of simian IL-15 and did not affect the human IL-2-induced proliferation of CTLL-2 cells. Also, in a pool of synovial fluid cells from two Rheumatoid Arthritis patients, the immune sera slightly inhibited TNF-α secretion. Finally, it was observed that this vaccine candidate neither affect animal behavior, clinical status, blood biochemistry nor the percentage of IL-15-dependent cell populations, specifically CD56+ NK and CD8+ T cells. Conclusion Our results indicate that vaccination with mhIL-15 induced neutralizing antibodies to native IL-15 in non-human primates. Based on this fact, we propose that this vaccine candidate could be potentially beneficial for treatment of diseases where IL-15 overexpression is associated with their pathogenesis.
Collapse
Affiliation(s)
- Yunier Rodríguez-Álvarez
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba.
| | - Yanelys Morera-Díaz
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba
| | - Haydee Gerónimo-Pérez
- Quality Control Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Jorge Castro-Velazco
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Rafael Martínez-Castillo
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Pedro Puente-Pérez
- Animal Facility Department, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Vladimir Besada-Pérez
- Chemistry and Physics Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10600, Cuba
| | - Eugenio Hardy-Rando
- Biotechnology Laboratory, Study Center for Research and Biological Evaluations, Institute of Pharmacy and Foods, Havana University, Avenue 222, PO Box 13600, Havana, 10600, Cuba
| | - Araceli Chico-Capote
- Rheumatology Department, Hermanos Ameijeiras Hospital, San Lazaro 701, PO Box 6122, Havana, 10600, Cuba
| | - Klaudia Martínez-Cordovez
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba
| | - Alicia Santos-Savio
- Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Avenue 31, PO Box 6162, Havana, 10 600, Cuba
| |
Collapse
|
12
|
Liu Y, Zeng M, Liu Z. Th17 response and its regulation in inflammatory upper airway diseases. Clin Exp Allergy 2015; 45:602-12. [PMID: 25048954 DOI: 10.1111/cea.12378] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Allergic rhinitis (AR) and chronic rhinosinusitis (CRS) are two widely prevalent inflammatory diseases in the upper airways. T cell immunity has been suggested to play an important pathogenic role in many chronic inflammatory diseases including inflammatory upper airway diseases. Inappropriate CD4(+) T cell responses, especially the dysregulation of the Th1/Th2 balance leading to excessive Th1 or Th2 cell activation, have been associated with allergic rhinitis and chronic rhinosinusitis. Nevertheless, recent studies suggest that IL-17A and IL-17A-producing Th17 cell subset, a distinct pro-inflammatory CD4(+) T cell lineage, may also play an important role in the pathophysiology of inflammatory upper airway diseases. Th17 cells may promote both eosinophilic and neutrophilic inflammation in AR and CRS. In addition, a few, but accumulating evidence shows that the Th17 responses can be tightly regulated by endogenous and exogenous substances in the context of AR and CRS. This review discusses recent advances in our understanding of the expression and function of the Th17 response and its regulation in inflammatory upper airway diseases, and the perspective for future investigation and clinical utility.
Collapse
Affiliation(s)
- Y Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | |
Collapse
|
13
|
RORγt-specific transcriptional interactomic inhibition suppresses autoimmunity associated with TH17 cells. Proc Natl Acad Sci U S A 2014; 111:18673-8. [PMID: 25527718 DOI: 10.1073/pnas.1413687112] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The nuclear hormone receptor retinoic acid-related orphan receptor gamma t (RORγt) is a transcription factor (TF) specific to TH17 cells that produce interleukin (IL)-17 and have been implicated in a wide range of autoimmunity. Here, we developed a novel therapeutic strategy to modulate the functions of RORγt using cell-transducible form of transcription modulation domain of RORγt (tRORγt-TMD), which can be delivered effectively into the nucleus of cells and into the central nerve system (CNS). tRORγt-TMD specifically inhibited TH17-related cytokines induced by RORγt, thereby suppressing the differentiation of naïve T cells into TH17, but not into TH1, TH2, or Treg cells. tRORγt-TMD injected into experimental autoimmune encephalomyelitis (EAE) animal model can be delivered effectively in the splenic CD4(+) T cells and spinal cord-infiltrating CD4(+) T cells, and suppress the functions of TH17 cells. The clinical severity and incidence of EAE were ameliorated by tRORγt-TMD in preventive and therapeutic manner, and significant reduction of both infiltrating CD4(+) IL-17(+) T cells and inflammatory cells into the CNS was observed. As a result, the number of spinal cord demyelination was also reduced after tRORγt-TMD treatment. With the same proof of concept, tTbet-TMD specifically blocking TH1 differentiation improved the clinical incidence of rheumatoid arthritis (RA). Therefore, tRORγt-TMD and tTbet-TMD can be novel therapeutic reagents with the natural specificity for the treatment of inflammatory diseases associated with TH17 or TH1. This strategy can be applied to treat various diseases where a specific transcription factor has a key role in pathogenesis.
Collapse
|
14
|
Guan Q, Burtnick HA, Qing G, Weiss CR, Ma AG, Ma Y, Warrington RJ, Peng Z. Employing an IL-23 p19 vaccine to block IL-23 ameliorates chronic murine colitis. Immunotherapy 2014; 5:1313-22. [PMID: 24283842 DOI: 10.2217/imt.13.141] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Overexpression of IL-23 has been implicated in the pathogenesis of Crohn's disease. Using vaccines to block overexpressed endogenous cytokines has emerged as a new therapeutic strategy for the long-term treatment of the disease. AIM We sought to develop peptide-based vaccines specific to IL-23 and evaluate their effects in colitis mice. MATERIALS & METHODS The vaccine was developed by inserting a peptide derived from mouse IL-23 p19 into the carrier protein, hepatitis B core antigen, using molecular engineering methods. One vaccine against IL-23 p19 was obtained that induced high-titered and long-lasting antibodies to IL-23 without the use of adjuvants. The inhibitory effect of vaccine-immunized serum was subsequently evaluated in vitro. To evaluate the in vivo effects, mice were subcutaneously injected with the vaccine, carrier or saline three times. Two weeks after the last injection, chronic colitis was induced in mice by seven weekly administrations with 2,4,6-trinitrobenzene sulfonic acid. RESULTS In vitro studies revealed that serum IL-23 p19-specific IgG significantly suppressed IL-23-induced IL-17 production by splenocytes. In vivo evaluation of the effect of the vaccine in mice with chronic colitis indicated that vaccine-immunized mice exhibited a decrease in colon inflammation, collagen deposition and levels of IL-23 and IL-12 cytokines, compared with control groups. CONCLUSION IL-23 p19 vaccine is capable of downregulating inflammatory responses in chronic murine colitis, providing a novel therapeutic approach in Crohn's disease.
Collapse
Affiliation(s)
- Qingdong Guan
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, R3E 3P4, Canada
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Uyttenhove C, van Snick J. [Auto-vaccines: an immunological alternative to gene silencing]. Med Sci (Paris) 2013; 29:425-9. [PMID: 23621939 DOI: 10.1051/medsci/2013294017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Auto-vaccination is a procedure that recently attracted the interest of a growing number of investigators as an alternative to gene inactivation for functional studies of cytokines or other mediators. It is based on the observation that autologous cytokines cross-linked to a foreign protein or peptide are recognized by self-reactive B cells that present foreign peptides, and by doing so attract illicit help from helper T cells that recognize the foreign peptide on the self-reactive B cell MHC Class II complex. This leads to the production of antibodies reacting with self-proteins and thus to neutralization of the targeted factor. Here, we summarize the different techniques that were successful in breaking this self-tolerance and provide several examples of the functional consequences of these auto-vaccines. An additional output of auto-vaccination is the production of mouse monoclonal antibodies against mouse factors. Such antibodies have obvious advantages for long-term use in vivo.
Collapse
Affiliation(s)
- Catherine Uyttenhove
- Institut Ludwig pour la Recherche sur le Cancer et Unité de Génétique Cellulaire, Université Catholique de Louvain, 74 Avenue Hippocrate, 1200 Bruxelles, Belgique
| | | |
Collapse
|
16
|
Feng J, Zhang G, Hu X, Si Chen C, Qin X. Estrogen inhibits estrogen receptor α-mediated rho-kinase expression in experimental autoimmune encephalomyelitis rats. Synapse 2013; 67:399-406. [PMID: 23401288 DOI: 10.1002/syn.21650] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 02/05/2013] [Accepted: 02/06/2013] [Indexed: 12/25/2022]
Affiliation(s)
- Jinzhou Feng
- Department of Neurology; the First Affiliated Hospital of Chongqing Medical University; Chongqing; 400016; China
| | - Guanghui Zhang
- Department of Neurology; the First Affiliated Hospital of Henan University of Science and Technology; Luoyang; 471003; China
| | - Xiao Hu
- Department of Neurology; the Guizhou Provincial People's Hospital; Guizhou; 550002; China
| | - Cindy Si Chen
- Department of Medicine; Drexel University College of Medicine; Philadelphia; Pennsylvania; 19129
| | - Xinyue Qin
- Department of Neurology; the First Affiliated Hospital of Chongqing Medical University; Chongqing; 400016; China
| |
Collapse
|
17
|
Hu X, Qin X. Lentivirus-mediated estrogen receptor α overexpression in the central nervous system ameliorates experimental autoimmune encephalomyelitis in mice. Int J Mol Med 2013; 31:1209-21. [PMID: 23525227 DOI: 10.3892/ijmm.2013.1306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 03/05/2013] [Indexed: 11/05/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory cell infiltration of the central nervous system (CNS) and multifocal demyelination. Clinical data and clinical indicators demonstrate that estrogen improves the relapse-remittance of MS patients. This study aimed to investigate the anti-inflammatory effects and the underlying mechanism(s) of action of estrogen and estrogen receptor α (ERα) in an experimental autoimmune encephalomyelitis (EAE) mouse model of MS. An ERα recombinant lentivirus was constructed. Mouse neurons were cultured in serum-free culture medium, and ERα recombinant lentivirus with a multiplicity of infection (MOI) of 5 was used to infect the neurons. Furthermore, neuronal ERα mRNA and protein expression were detected using real-time quantitative PCR and western blot analysis. We sterotaxically injected ERα recombinant lentivirus into the lateral ventricle of mouse brains, and successfully identified infected neurons using Flag immunofluorescence staining to determine the optimal dose. A total of 75 C57BL/6 mice were ovariectomized. After 2 weeks, EAE was induced with myelin oligodendrocyte glycoprotein (MOG) 35-55 peptide. The EAE mice were divided into 5 groups: the estrogen group (treatment with estradiol), the ERα agonist group (treatment with raloxifene), the ERα recombinant lentivirus group (ERα group, treatment with ERα recombinant lentivirus), the empty virus group and the normal saline (NS) group; clinical symptoms and body weight were compared among the groups. We assessed EAE-related parameters, detected pathological changes with immunohistochemistry and quantified the expression of myelin basic protein (MBP), matrix metalloproteinase-9 (MMP-9), and a subset of EAE-related cytokines using enzyme-linked immunosorbent assay (ELISA). We successfully constructed an ERα recombinant lentivirus. C57BL/6 mouse neurons can survive in culture for at least 8 weeks. During that period, the recombinant lentivirus was able to infect the neurons, while sustaining green fluorescence protein (GFP) expression. ERα recombinant lentivirus also infected the neurons at a MOI of 5. The ERα mRNA and protein expression levels were higher in the infected neurons compared to the uninfected ones. We successfully infected the CNS of C57BL/6 mice by stereotaxically injecting ERα recombinant lentivirus into the lateral ventricle of the mouse brains and induced EAE. The lentivirus-mediated overexpression of ERα reduced the incidence of EAE, ameliorated the clinical symptoms, inhibited inflammatory cell CNS infiltration, and reduced nerve fiber demyelination. MMP-9, tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), interleukin (IL)-17 and IL-23 expression levels were decreased, while those of MBP and IL-4 were increased. These data demonstrate that it is possible to induce the overexpression of ERα using a recombinant lentivirus, and that this novel intervention ameliorates EAE in a mouse model. Mechanistically, estrogen and ERα inhibit inflammatory responses, and ERα alleviates damage to the myelin sheath. Collectively, our findings support the potential use of ERα as a therapeutic target for the treatment of MS.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | | |
Collapse
|
18
|
Zimmermann J, Krauthausen M, Hofer MJ, Heneka MT, Campbell IL, Müller M. CNS-targeted production of IL-17A induces glial activation, microvascular pathology and enhances the neuroinflammatory response to systemic endotoxemia. PLoS One 2013; 8:e57307. [PMID: 23468966 PMCID: PMC3584143 DOI: 10.1371/journal.pone.0057307] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 01/20/2013] [Indexed: 01/13/2023] Open
Abstract
Interleukin-17A (IL-17A) is a key cytokine modulating the course of inflammatory diseases. Whereas effector functions of IL-17A like induction of antimicrobial peptides and leukocyte infiltration could clearly be demonstrated for peripheral organs, CNS specific effects are not well defined and appear controversial. To further clarify the functional significance of IL-17A in the CNS, we generated a transgenic mouse line with astrocyte-restricted expression of the IL-17A gene. GFAP/IL-17A transgenic mice develop normally and do not show any signs of neurological dysfunction. However, histological characterization revealed astrocytosis and activation of microglia. Demyelination, neurodegeneration or prominent tissue damage was not observed but a vascular pathology mimicking microangiopathic features was evident. Histological and flow cytometric analysis demonstrated the absence of parenchymal infiltration of immune cells into the CNS of GFAP/IL-17A transgenic mice. In GFAP/IL-17A mice, LPS-induced endotoxemia led to a more pronounced microglial activation with expansion of a distinct CD45(high)/CD11b(+) population and increased induction of proinflammatory cytokines compared with controls. Our data argues against a direct role of IL-17A in mediating tissue damage during neuroinflammation. More likely IL-17A acts as a modulating factor in the network of induced cytokines. This novel mouse model will be a very useful tool to further characterize the role of IL-17A in neuroinflammatory disease models.
Collapse
Affiliation(s)
| | | | - Markus J. Hofer
- Department of Neuropathology, University Clinic of Marburg and Giessen, Marburg, Germany
| | - Michael T. Heneka
- Department of Neurology, Universitätsklinikum Bonn, Bonn, Germany
- Clinical Neuroscience Unit, University of Bonn, Bonn, Germany
| | - Iain L. Campbell
- School of Molecular Bioscience, University of Sydney, Sydney, Australia
| | - Marcus Müller
- Department of Neurology, Universitätsklinikum Bonn, Bonn, Germany
- School of Molecular Bioscience, University of Sydney, Sydney, Australia
- * E-mail:
| |
Collapse
|
19
|
Yang XF, Yang Y, Lian YT, Wang ZH, Li XW, Cheng LX, Liu JP, Wang YF, Gao X, Liao YH, Wang M, Zeng QT, Liu K. The antibody targeting the E314 peptide of human Kv1.3 pore region serves as a novel, potent and specific channel blocker. PLoS One 2012; 7:e36379. [PMID: 22558454 PMCID: PMC3338681 DOI: 10.1371/journal.pone.0036379] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 04/04/2012] [Indexed: 01/08/2023] Open
Abstract
Selective blockade of Kv1.3 channels in effector memory T (T(EM)) cells was validated to ameliorate autoimmune or autoimmune-associated diseases. We generated the antibody directed against one peptide of human Kv1.3 (hKv1.3) extracellular loop as a novel and possible Kv1.3 blocker. One peptide of hKv1.3 extracellular loop E3 containing 14 amino acids (E314) was chosen as an antigenic determinant to generate the E314 antibody. The E314 antibody specifically recognized 63.8KD protein stably expressed in hKv1.3-HEK 293 cell lines, whereas it did not recognize or cross-react to human Kv1.1(hKv1.1), Kv1.2(hKv1.2), Kv1.4(hKv1.4), Kv1.5(hKv1.5), KCa3.1(hKCa3.1), HERG, hKCNQ1/hKCNE1, Nav1.5 and Cav1.2 proteins stably expressed in HEK 293 cell lines or in human atrial or ventricular myocytes by Western blotting analysis and immunostaining detection. By the technique of whole-cell patch clamp, the E314 antibody was shown to have a directly inhibitory effect on hKv1.3 currents expressed in HEK 293 or Jurkat T cells and the inhibition showed a concentration-dependence. However, it exerted no significant difference on hKv1.1, hKv1.2, hKv1.4, hKv1.5, hKCa3.1, HERG, hKCNQ1/hKCNE1, L-type Ca(2+) or voltage-gated Na(+) currents. The present study demonstrates that the antibody targeting the E314 peptide of hKv1.3 pore region could be a novel, potent and specific hKv1.3 blocker without affecting a variety of closely related K(v)1 channels, KCa3.1 channels and functional cardiac ion channels underlying central nervous system (CNS) disorders or drug-acquired arrhythmias, which is required as a safe clinic-promising channel blocker.
Collapse
Affiliation(s)
- Xiao-Fang Yang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Tian Lian
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Wang
- Department of Geriatrics, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Wei Li
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Long-Xian Cheng
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Ping Liu
- Department of Cardiovascular Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-Fu Wang
- Department of Cardiology, Affiliated Hospital, Jining Medical College, Shandong, China
| | - Xiang Gao
- Department of Geriatrics, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Hua Liao
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qiu-Tang Zeng
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Liu
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
IL-17 producing T cells in mouse models of multiple sclerosis and rheumatoid arthritis. J Mol Med (Berl) 2012; 90:613-24. [PMID: 22231742 DOI: 10.1007/s00109-011-0841-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 11/22/2011] [Accepted: 11/28/2011] [Indexed: 12/11/2022]
Abstract
Multiple Sclerosis (MS) and Rheumatoid Arthritis (RA) are amongst the most common autoimmune diseases in the northern hemisphere. There is mounting evidence that in both afflictions, not only environmental and genetic factors influence disease, but cellular components such as autoreactive T cells also contribute to pathology. Animal models are key in the study and subsequent therapeutic development for human autoimmune diseases. As patient material is often difficult to obtain and in some cases--as in MS, where the central nervous system (CNS) is concerned--even not accessible, animal models provide a multifaceted tool to explore disease-underlying mechanisms. The pro-inflammatory T cell cytokine IL-17 has recently moved to center stage due to its crucial role in autoimmune diseases including MS and RA. A plethora of studies in animal models has sustained the relevance of this cytokine pathway for the development of autoimmunity and shed light on its cellular sources and patho-mechanisms. This review addresses the role of IL-17 producing T lymphocytes, in particular CD4(+) and γδ T cells, in three commonly used mouse models for MS and RA, namely experimental autoimmune encephalomyelitis (EAE), collagen-induced arthritis (CIA), and antigen-induced arthritis (AIA). Comparing and combining knowledge gained from different animal models will broaden our understanding of the IL-17 biology and facilitate the translation to the human diseases.
Collapse
|
21
|
Van Belle AB, de Heusch M, Lemaire MM, Hendrickx E, Warnier G, Dunussi-Joannopoulos K, Fouser LA, Renauld JC, Dumoutier L. IL-22 is required for imiquimod-induced psoriasiform skin inflammation in mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 188:462-9. [PMID: 22131335 DOI: 10.4049/jimmunol.1102224] [Citation(s) in RCA: 230] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Psoriasis is a common chronic autoimmune skin disease of unknown cause that involves dysregulated interplay between immune cells and keratinocytes. IL-22 is a cytokine produced by the TH1, TH17, and TH22 subsets that are functionally implicated in the psoriatic pathology. We assessed the role of IL-22 in a mouse model where psoriasiform skin inflammation is triggered by topical application of the TLR7/8 agonist imiquimod. At the macroscopic level, scaly skin lesions induced by daily applications of imiquimod in wild-type mice were almost totally absent in IL-22-deficient mice or in mice treated with a blocking anti-IL-22 Ab. At the microscopic level, IL-22-deficient mice showed a dramatic decrease in the development of pustules and a partial decrease in acanthosis. At the molecular level, the absence or inhibition of IL-22 strongly decreased the expression of chemotactic factors such as CCL3 and CXCL3 and of biomarkers such as S100A8, S100A7, and keratin 14, which reflect the antimicrobial and hyperproliferative responses of keratinocytes. IL-22 also played a major role in neutrophil infiltration after imiquimod treatment. IL-23 was required for IL-22 production, and γδ TCR lymphocytes represented the major source of IL-22 in lymph nodes from imiquimod-treated mice. However, T cells were not absolutely required for IL-22 production because imiquimod-induced IL-22 expression in the skin is still preserved in Rag2(-/-) mice. Taken together, our data show that IL-22 is required for psoriasis-like lesions in the mouse imiquimod model and is produced by both T cells and innate immune cells.
Collapse
MESH Headings
- Adjuvants, Immunologic/adverse effects
- Adjuvants, Immunologic/pharmacology
- Aminoquinolines/adverse effects
- Aminoquinolines/pharmacology
- Animals
- Antigens, Differentiation/genetics
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- Chemokine CCL3/genetics
- Chemokine CCL3/immunology
- Chemokine CCL3/metabolism
- Dermatitis/etiology
- Dermatitis/immunology
- Dermatitis/metabolism
- Disease Models, Animal
- Gene Expression Regulation/genetics
- Gene Expression Regulation/immunology
- Imiquimod
- Immunity, Innate/drug effects
- Immunity, Innate/genetics
- Immunity, Innate/immunology
- Interleukins/biosynthesis
- Interleukins/genetics
- Interleukins/immunology
- Mice
- Mice, Knockout
- Neutrophil Infiltration/drug effects
- Neutrophil Infiltration/genetics
- Neutrophil Infiltration/immunology
- Neutrophils/immunology
- Neutrophils/metabolism
- Neutrophils/pathology
- Psoriasis/chemically induced
- Psoriasis/immunology
- Psoriasis/metabolism
- Psoriasis/pathology
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Skin/immunology
- Skin/metabolism
- Skin/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- Interleukin-22
Collapse
|
22
|
El-Behi M, Ciric B, Dai H, Yan Y, Cullimore M, Safavi F, Zhang GX, Dittel BN, Rostami A. The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol 2011; 12:568-75. [PMID: 21516111 DOI: 10.1038/ni.2031] [Citation(s) in RCA: 847] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 04/04/2011] [Indexed: 12/12/2022]
Abstract
Interleukin 17 (IL-17)-producing helper T cells (T(H)17 cells) require exposure to IL-23 to become encephalitogenic, but the mechanism by which IL-23 promotes their pathogenicity is not known. Here we found that IL-23 induced production of the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) in T(H)17 cells and that GM-CSF had an essential role in their encephalitogenicity. Our findings identify a chief mechanism that underlies the important role of IL-23 in autoimmune diseases. IL-23 induced a positive feedback loop whereby GM-CSF secreted by T(H)17 cells stimulated the production of IL-23 by antigen-presenting cells. Such cross-regulation of IL-23 and GM-CSF explains the similar pattern of resistance to autoimmunity when either of the two cytokines is absent and identifies T(H)17 cells as a crucial source of GM-CSF in autoimmune inflammation.
Collapse
Affiliation(s)
- Mohamed El-Behi
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Guan Q, Ma Y, Hillman CL, Qing G, Ma AG, Weiss CR, Zhou G, Bai A, Warrington RJ, Bernstein CN, Peng Z. Targeting IL-12/IL-23 by employing a p40 peptide-based vaccine ameliorates TNBS-induced acute and chronic murine colitis. Mol Med 2011; 17:646-56. [PMID: 21424108 DOI: 10.2119/molmed.2010.00252] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 03/10/2011] [Indexed: 12/17/2022] Open
Abstract
Interleukin (IL)-12 and IL-23 both share the p40 subunit and are key cytokines in the pathogenesis of Crohn's disease. Previously, we have developed and identified three mouse p40 peptide-based and virus-like particle vaccines. Here, we evaluated the effects and immune mechanisms of the optimal vaccine in downregulating intestinal inflammation in murine acute and chronic colitis, induced by intrarectal administrations of trinitrobenzene sulfonic acid (TNBS). Mice were injected subcutaneously with vaccine, vaccine carrier or saline three times, and then intrarectally administered TNBS weekly for 2 wks (acute colitis) or 7 wks (chronic colitis). The severity of colitis was evaluated by body weight, histology and collagen and cytokine levels in colon tissue. Th1 and Th17 cells in mesenteric lymph nodes (MLN) were determined. Our results showed the vaccine induced high level and long-lasting specific IgG antibodies to p40, IL-12 and IL-23. After administrations of TNBS, vaccinated mice had significantly less body weight loss and a significant decrease of inflammatory scores, collagen deposition and expression of p40, IL-12, IL-23, IL-17, TNF, iNOS and Bcl-2 in colon tissues, compared with carrier and saline groups. Moreover, vaccinated mice exhibited a trend to lower percentages of Th1 cells in acute colitis and of Th17 cells in chronic colitis in MLN than in controls. In summary, administration of the vaccine induced specific antibodies to IL-12 and IL-23, which was associated with improvement of intestinal inflammation and fibrosis. This suggests that the vaccine may provide a potential approach for the long-term treatment of Crohn's disease.
Collapse
Affiliation(s)
- Qingdong Guan
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Li H, Nourbakhsh B, Ciric B, Zhang GX, Rostami A. Neutralization of IL-9 ameliorates experimental autoimmune encephalomyelitis by decreasing the effector T cell population. THE JOURNAL OF IMMUNOLOGY 2010; 185:4095-100. [PMID: 20805418 DOI: 10.4049/jimmunol.1000986] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Multiple sclerosis is a CD4(+) T cell-mediated autoimmune disease affecting the CNS. Multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), have been thought to be Th1-mediated diseases. However, recent studies provide strong evidence that the major pathogenic T cell subsets in EAE are Th17 cells. IL-9, a hematopoietic growth factor, is considered to be a mediator of Th17 cells, but the precise mechanisms of its action are largely unknown. The present study was designed to investigate the role of IL-9 in autoimmune demyelination. IL-9 blockade with anti-IL-9 mAb inhibited the development of EAE, reduced the serum levels of IL-17, the CNS mRNA expression of IL-17, IL-6, IFN-γ, and TNF-α, and the myelin oligodendrocyte glycoprotein (MOG)-induced IL-17, IFN-γ secretion of lymphocytes. Furthermore, anti-IL-9 mAb in culture suppressed IL-17 production of MOG-reactive T cells and their potency in adoptive transfer EAE. These findings indicate that the protective effect of IL-9 blockade in EAE was likely mediated via inhibition of the development of MOG peptide-specific T cells, which in turn led to reduced infiltration of T cells into the CNS. Thus, anti-IL-9 mAb treatment may provide an effective therapeutic strategy against autoimmune diseases.
Collapse
Affiliation(s)
- Hongmei Li
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | |
Collapse
|
25
|
Qin X, Guo BT, Wan B, Fang L, Lu L, Wu L, Zang YQ, Zhang JZ. Regulation of Th1 and Th17 cell differentiation and amelioration of experimental autoimmune encephalomyelitis by natural product compound berberine. THE JOURNAL OF IMMUNOLOGY 2010; 185:1855-63. [PMID: 20622114 DOI: 10.4049/jimmunol.0903853] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Berberine (BBR), an isoquinoline alkaloid derived from plants, is widely used as an anti-inflammatory remedy in traditional Chinese medicine. In this study, we showed that BBR was efficacious in the amelioration of experimental autoimmune encephalomyelitis (EAE) through novel regulatory mechanisms involving pathogenic Th1 and Th17 cells. BBR inhibited differentiation of Th17 cells and, to a lesser degree, Th1 cells through direct actions on the JAK/STAT pathway, whereas it had no effect on the relative number of CD4(+)Foxp3(+) regulatory T cells. In addition, BBR indirectly influenced Th17 and Th1 cell functions through its effect on the expression and function of costimulatory molecules and the production of IL-6, which was attributable to the inhibition of NF-kappaB activity in CD11b(+) APCs. BBR treatment completely abolished the encephalitogenicity of MOG(35-55)-reactive Th17 cells in an adoptive transfer EAE model, and the same treatment significantly inhibited the ability of MOG(35-55)-reactive Th1 cells to induce EAE. This study provides new evidence that natural compounds, such as BBR, are of great value in the search for novel anti-inflammatory agents and therapeutic targets for autoimmune diseases.
Collapse
Affiliation(s)
- Xia Qin
- Joint Immunology Laboratory, Institute of Health Sciences, Shanghai 200025, China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Ait-Oufella H, Herbin O, Bouaziz JD, Binder CJ, Uyttenhove C, Laurans L, Taleb S, Van Vré E, Esposito B, Vilar J, Sirvent J, Van Snick J, Tedgui A, Tedder TF, Mallat Z. B cell depletion reduces the development of atherosclerosis in mice. ACTA ACUST UNITED AC 2010; 207:1579-87. [PMID: 20603314 PMCID: PMC2916123 DOI: 10.1084/jem.20100155] [Citation(s) in RCA: 345] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
B cell depletion significantly reduces the burden of several immune-mediated diseases. However, B cell activation has been until now associated with a protection against atherosclerosis, suggesting that B cell–depleting therapies would enhance cardiovascular risk. We unexpectedly show that mature B cell depletion using a CD20-specific monoclonal antibody induces a significant reduction of atherosclerosis in various mouse models of the disease. This treatment preserves the production of natural and potentially protective anti–oxidized low-density lipoprotein (oxLDL) IgM autoantibodies over IgG type anti-oxLDL antibodies, and markedly reduces pathogenic T cell activation. B cell depletion diminished T cell–derived IFN-γ secretion and enhanced production of IL-17; neutralization of the latter abrogated CD20 antibody–mediated atheroprotection. These results challenge the current paradigm that B cell activation plays an overall protective role in atherogenesis and identify new antiatherogenic strategies based on B cell modulation.
Collapse
Affiliation(s)
- Hafid Ait-Oufella
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, 75015 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ma Y, Guan Q, Bai A, Weiss CR, Hillman CL, Ma A, Zhou G, Qing G, Peng Z. Targeting TGF-beta1 by employing a vaccine ameliorates fibrosis in a mouse model of chronic colitis. Inflamm Bowel Dis 2010; 16:1040-50. [PMID: 19924805 DOI: 10.1002/ibd.21167] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Intestinal fibrosis and stricture formation are major complications of inflammatory bowel disease (IBD), for which there are currently few effective treatments. We sought to investigate whether targeting transforming growth factor-beta1 (TGF-beta1), a key profibrotic mediator, with a peptide-based virus-like particle vaccine would be effective in suppressing intestinal fibrosis by using a mouse model of 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced chronic colitis. METHODS The vaccine was prepared by inserting a peptide derived from mouse TGF-beta1 into a carrier hepatitis B core antigen using gene recombination methods. Chronic colitis was induced in BALB/c mice by 8 weekly TNBS administrations. Mice were subcutaneously injected with vaccine, carrier, or phosphate-buffered saline (PBS) in 2 separate studies: either before or after acute inflammatory responses commenced. RESULTS Sera from vaccinated mice exhibited significantly elevated levels of TGF-beta1-specific immunoglobulin G (IgG), which inhibited TGF-beta1-induced luciferase production in mink lung epithelial cells. In the chronic colitis model, mice receiving vaccine showed improved body weight gain and significantly reduced colonic collagen deposition. Hematoxylin and eosin staining and semiquantitative scoring indicated that vaccination even ameliorated colonic inflammation. Cytokine profile analysis revealed that levels of TGF-beta1, interleukin (IL)-17, and IL-23 in vaccinated mouse colon tissues were decreased, and that percentages of IL-17-expressing CD4(+) lymphocytes in mesenteric lymph node cells were reduced. Furthermore, Smad3 phosphorylation, a key event in TGF-beta signaling, was decreased in colonic tissue in vaccinated mice. CONCLUSIONS This TGF-beta1 peptide-based vaccine, which suppressed excessive TGF-beta1 bioactivity, may prevent the development of intestinal fibrosis and associated complications, presenting a novel approach in the treatment of IBD.
Collapse
Affiliation(s)
- Yanbing Ma
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Lo Re S, Dumoutier L, Couillin I, Van Vyve C, Yakoub Y, Uwambayinema F, Marien B, van den Brûle S, Van Snick J, Uyttenhove C, Ryffel B, Renauld JC, Lison D, Huaux F. IL-17A-producing gammadelta T and Th17 lymphocytes mediate lung inflammation but not fibrosis in experimental silicosis. THE JOURNAL OF IMMUNOLOGY 2010; 184:6367-77. [PMID: 20421647 DOI: 10.4049/jimmunol.0900459] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
IL-17-producing T lymphocytes play a crucial role in inflammation, but their possible implication in fibrosis remains to be explored. In this study, we examined the involvement of these cells in a mouse model of lung inflammation and fibrosis induced by silica particles. Upregulation of IL-17A was associated with the development of experimental silicosis, but this response was markedly reduced in athymic, gammadelta T cell-deficient or CD4(+) T cell-depleted mice. In addition, gammadelta T lymphocytes and CD4(+) T cells, but not macrophages, neutrophils, NK cells or CD8 T cells, purified from the lungs of silicotic mice markedly expressed IL-17A. Depletion of alveolar macrophages or neutralization of IL-23 reduced upregulation of IL-17A in the lung of silicotic mice. IL-17R-deficient animals (IL-17R(-/-)) or IL-17A Ab neutralization, but not IL-22(-/-) mice, developed reduced neutrophil influx and injury during the early lung response to silica. However, chronic inflammation, fibrosis, and TGF-beta expression induced by silica were not attenuated in the absence of IL-17R or -22 or after IL-17A Ab blockade. In conclusion, a rapid lung recruitment of IL-17A-producing T cells, mediated by macrophage-derived IL-23, is associated with experimental silicosis in mice. Although the acute alveolitis induced by silica is IL-17A dependent, this cytokine appears dispensable for the development of the late inflammatory and fibrotic lung responses to silica.
Collapse
Affiliation(s)
- Sandra Lo Re
- Louvain Centre for Toxicology and Applied Pharmacology, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Chen X, Das R, Komorowski R, van Snick J, Uyttenhove C, Drobyski WR. Interleukin 17 is not required for autoimmune-mediated pathologic damage during chronic graft-versus-host disease. Biol Blood Marrow Transplant 2009; 16:123-8. [PMID: 19772944 DOI: 10.1016/j.bbmt.2009.09.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 09/14/2009] [Indexed: 01/08/2023]
Abstract
The transition from acute to chronic graft-versus-host disease (aGVHD, cGVHD) is characterized by the progressive loss of self-tolerance and the development of autoimmune manifestations. Interleukin 17 (IL-17) is a proinflammatory cytokine that has been shown to play a prominent role in autoimmune disorders in the nontransplant setting, but the extent to which IL-17 is necessary for the autoimmunity that occurs as a consequence of GVHD is not known. In this study, we demonstrate using a combination of antibody-based and genetic approaches that IL-17 is not required for the loss of self-tolerance and resulting CD4(+)T cell-dependent pathologic damage that occurs during the evolution from aGVHD to cGVHD. Rather, T(H)1 cells and other proinflammatory cytokines are fully competent to promote autoimmune-mediated tissue damage. Thus, the selective targeting of IL-17 may not be a viable clinical strategy for preventing the autoimmune manifestations that develop during cGVHD.
Collapse
Affiliation(s)
- Xiao Chen
- Bone Marrow Transplant Program, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | | | | | | | |
Collapse
|
30
|
Taleb S, Romain M, Ramkhelawon B, Uyttenhove C, Pasterkamp G, Herbin O, Esposito B, Perez N, Yasukawa H, Van Snick J, Yoshimura A, Tedgui A, Mallat Z. Loss of SOCS3 expression in T cells reveals a regulatory role for interleukin-17 in atherosclerosis. ACTA ACUST UNITED AC 2009; 206:2067-77. [PMID: 19737863 PMCID: PMC2757872 DOI: 10.1084/jem.20090545] [Citation(s) in RCA: 320] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Atherosclerosis is an inflammatory vascular disease responsible for the first cause of mortality worldwide. Recent studies have clearly highlighted the critical role of the immunoinflammatory balance in the modulation of disease development and progression. However, the immunoregulatory pathways that control atherosclerosis remain largely unknown. We show that loss of suppressor of cytokine signaling (SOCS) 3 in T cells increases both interleukin (IL)-17 and IL-10 production, induces an antiinflammatory macrophage phenotype, and leads to unexpected IL-17–dependent reduction in lesion development and vascular inflammation. In vivo administration of IL-17 reduces endothelial vascular cell adhesion molecule–1 expression and vascular T cell infiltration, and significantly limits atherosclerotic lesion development. In contrast, overexpression of SOCS3 in T cells reduces IL-17 and accelerates atherosclerosis. We also show that in human lesions, increased levels of signal transducer and activator of transcription (STAT) 3 phosphorylation and IL-17 are associated with a stable plaque phenotype. These results identify novel SOCS3-controlled IL-17 regulatory pathways in atherosclerosis and may have important implications for the understanding of the increased susceptibility to vascular inflammation in patients with dominant-negative STAT3 mutations and defective Th17 cell differentiation.
Collapse
Affiliation(s)
- Soraya Taleb
- Institut National de la Santé et de la Recherche Médicale, Unit 970 and Université Paris Descartes, Paris Cardiovascular Research Center, 75015 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Rafei M, Campeau PM, Aguilar-Mahecha A, Buchanan M, Williams P, Birman E, Yuan S, Young YK, Boivin MN, Forner K, Basik M, Galipeau J. Mesenchymal stromal cells ameliorate experimental autoimmune encephalomyelitis by inhibiting CD4 Th17 T cells in a CC chemokine ligand 2-dependent manner. THE JOURNAL OF IMMUNOLOGY 2009; 182:5994-6002. [PMID: 19414750 DOI: 10.4049/jimmunol.0803962] [Citation(s) in RCA: 263] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The administration of ex vivo culture-expanded mesenchymal stromal cells (MSCs) has been shown to reverse symptomatic neuroinflammation observed in experimental autoimmune encephalomyelitis (EAE). The mechanism by which this therapeutic effect occurs remains unknown. In an effort to decipher MSC mode of action, we found that MSC conditioned medium inhibits EAE-derived CD4 T cell activation by suppressing STAT3 phosphorylation via MSC-derived CCL2. Further analysis demonstrates that the effect is dependent on MSC-driven matrix metalloproteinase proteolytic processing of CCL2 to an antagonistic derivative. We also show that antagonistic CCL2 suppresses phosphorylation of AKT and leads to a reciprocal increased phosphorylation of ERK associated with an up-regulation of B7.H1 in CD4 T cells derived from EAE mice. CD4 T cell infiltration of the spinal cord of MSC-treated group was robustly decreased along with reduced plasma levels of IL-17 and TNF-alpha levels and in vitro from restimulated splenocytes. The key role of MSC-derived CCL2 was confirmed by the observed loss of function of CCL2(-/-) MSCs in EAE mice. In summary, this is the first report of MSCs modulating EAE biology via the paracrine conversion of CCL2 from agonist to antagonist of CD4 Th17 cell function.
Collapse
Affiliation(s)
- Moutih Rafei
- Montreal Center for Experimental Therapeutics in Cancer, Lady Davis Institute for Medical Research, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ivanov S, Lindén A. Interleukin-17 as a drug target in human disease. Trends Pharmacol Sci 2009; 30:95-103. [PMID: 19162337 DOI: 10.1016/j.tips.2008.11.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 11/04/2008] [Accepted: 11/10/2008] [Indexed: 12/20/2022]
Abstract
Interleukin (IL)-17 (now synonymous with IL-17A) is an archetype molecule for an entire family of IL-17 cytokines. Currently believed to be produced mainly by a specific subset of CD4 cells, named Th-17 cells, IL-17 is functionally located at the interface of innate and acquired immunity. Specifically, it induces the release of chemokines and growth factors from mesenchymal cells and is now emerging as an important local orchestrator of neutrophil accumulation in several mammalian organs. Furthermore, there is growing evidence that targeting IL-17 signaling might prove useful in a variety of diseases including asthma, Crohn's disease, multiple sclerosis, psoriatric disease and rheumatoid arthritis. Here, we summarize the key aspects of the biology of IL-17 in mammals and scrutinize the potential pharmacological use of targeting IL-17 in humans.
Collapse
|
33
|
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the CNS, characterized pathologically by a perivascular infiltrate consisting predominantly of T cells and macrophages. Although its aetiology remains unknown, several lines of evidence support the hypothesis that autoimmune mechanisms play a major role in the development of the disease. Several widely used disease-modifying agents are approved for the treatment of MS. However, these agents are only partially effective and their ability to attenuate the more progressive phases of the disease is not clear at this time. Therefore, there is a need to develop improved treatment options for MS. This article reviews the role of several novel, selective vaccine strategies that are currently under investigation, including: (i) T-cell vaccination (TCV); (ii) T-cell receptor (TCR) peptide vaccination; (iii) DNA vaccination; and (iv) altered peptide ligand (APL) vaccination. The administration of attenuated autoreactive T cells induces regulatory networks to specifically suppress pathogenic T cells in MS, a strategy named TCV. The concept of TCV was based on the experience of vaccination against aetiological agents of infectious diseases in which individuals are purposely exposed to an attenuated microbial pathogen, which then instructs the immune system to recognize and neutralize it in its virulent form. In regard to TCV, attenuated, pathogenic T cells are similarly used to instruct the immune system to recognize and neutralize disease-inducing T cells. In experimental allergic encephalomyelitis (EAE), an animal model for MS, pathogenic T cells use a strikingly limited number of variable-region elements (V region) to form TCR specific for defined autoantigens. Thus, vaccination with peptides directed against these TCR structures may induce immunoregulatory mechanisms, thereby preventing EAE. However, unlike EAE, myelin-reactive T cells derived from MS patients utilize a broad range of different V regions, challenging the clinical utility of this approach. Subsequently, the demonstration that injection of plasmid DNA encoding a reporter gene into skeletal muscle results in expression of the encoded proteins, as well as in the induction of immune responses in animal models of autoimmunity, was explored as another strategy to re-establish self-tolerance. This approach has promise for the treatment of MS and, therefore, warrants further investigation. APLs are molecules in which the native encephalitogenic peptides are modified by substitution(s) of one or a few amino acids critical for contact with the TCR. Depending on the substitution(s) at the TCR contact residues of the cognate peptide, an APL can induce immune responses that can protect against or reverse EAE. However, the heterogeneity of the immune response in MS patients requires further study to determine which patients are most likely to benefit from APL therapy. Other potential approaches for vaccines in MS include vaccination against axonal growth inhibitors associated with myelin, use of dendritic cells pulsed with specific antigens, and active vaccination against proinflammatory cytokines. Overall, vaccines for MS represent promising approaches for the treatment of this devastating disease, as well as other autoimmune diseases.
Collapse
Affiliation(s)
- Jorge Correale
- Department of Neurology, Raúl Carrea Institute for Neurological Research, Buenos Aires, Argentina.
| | | | | |
Collapse
|
34
|
Abstract
The discovery in mice of a new lineage of CD4+ effector T helper (Th) cells that selectively produce IL-17 has provided exciting new insights into immune regulation, host defence, and the pathogenesis of autoimmune and other chronic inflammatory disorders. This population of CD4+ Th cells, which has been termed 'Th17', indeed plays an apparently critical role in the pathogenesis of some murine models of autoimmunity. Interestingly, murine Th17 cells share a common origin with Foxp3+ T regulatory cells, because both populations are produced in response to transforming growth factor-beta, but they develop into Th17 cells only when IL-6 is simultaneously produced. Initial studies in humans have confirmed the existence of Th17 cells, but they have shown that the origin of these cells in humans differs from that in mice, with IL-1beta and IL-23 being the major cytokines responsible for their development. Moreover, the presence in the circulation and in various tissues of Th cells that can produce both IL-17 and interferon-gamma, as well as the flexibility of human Th17 clones to produce interferon-gamma in addition to IL-17 in response to IL-12, suggests that there may be a developmental relationship between Th17 and Th1 cells, at least in humans. Resolving this issue has great implications in tems of establishing the respective pathogenic roles of Th1 and Th17 cells in autoimmune disorders. In contrast, it is unlikely that Th17 cells contribute to the pathogenesis of human allergic IgE-mediated disorders, because IL-4 and IL-25 (a powerful inducer of IL-4) are both potent inhibitors of Th17 cell development.
Collapse
Affiliation(s)
- Sergio Romagnani
- Department of Internal Medicine, University of Florence, Viale Morgagni, 85 Firenze 50134, Italy.
| |
Collapse
|
35
|
van Puijvelde GHM, van Es T, Habets KLL, Hauer AD, van Berkel TJC, Kuiper J. A vaccine against atherosclerosis: myth or reality? Future Cardiol 2008; 4:125-33. [DOI: 10.2217/14796678.4.2.125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that develops in the context of enhanced serum lipid levels. Nowadays, many studies focus on the modulation of inflammatory responses to reduce atherosclerosis. The most powerful strategy to achieve this is vaccination. In several immune diseases vaccination is shown to be very effective, resulting in a drastic decline in the incidence of the disease. But is vaccination also realistic in atherosclerosis? In this article, several approaches to vaccinate against atherosclerosis are described. Vaccination (based on protein or DNA) against bioactive molecules and disease-related proteins successfully reduces experimental atherosclerosis. In addition, passive immunization with antibodies against atherosclerosis-specific antigens and tolerance induction, in which antigen-specific regulatory T cells are elicited, are described. In the near future, we expect an increased interest in vaccination against atherosclerosis and, maybe, the myth may become reality when the first clinical trials are performed.
Collapse
Affiliation(s)
- Gijs HM van Puijvelde
- Leiden University, LACDR, Division of Biopharmaceutics, Einsteinweg 55, 2300 RA Leiden, The Netherlands
| | - Thomas van Es
- Leiden University, Division of Biopharmaceutics, Leiden, The Netherlands
| | - Kim LL Habets
- Leiden University, Division of Biopharmaceutics, Leiden, The Netherlands
| | - Arnaud D Hauer
- Leiden University, Division of Biopharmaceutics, Leiden, The Netherlands
| | - Theo JC van Berkel
- Leiden University, Division of Biopharmaceutics, Leiden, The Netherlands
| | - Johan Kuiper
- Leiden University, Division of Biopharmaceutics, Leiden, The Netherlands
| |
Collapse
|