1
|
Ottobre M, Van Snick J, Aparicio JL. Interleukin-17A (IL-17A) is involved in antibody specificity to conformational epitopes. Biochem Biophys Res Commun 2024; 739:150588. [PMID: 39191146 DOI: 10.1016/j.bbrc.2024.150588] [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: 05/28/2024] [Revised: 07/29/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
The specificity of antibodies (Ab) is essential for the precise recognition of foreign or dangerous molecules. We have shown that mice infected with non-pathogenic Lactate Dehydrogenase Elevating Virus (LDV) inoculated with human growth hormone (hGH) or Ovalbumin (OVA), exhibit modified specificity of anti-hGH or anti-OVA Ab associated with the secretion of IFN-γ, IL-13, and IL-17. Cytokines are directly or indirectly involved in the isotypes, specificity, and affinity of Ab. Accordingly, here we investigated the effect of IL-17 neutralization on Ab specificities to OVA and Diphtheria Toxoid (DTx) in a mouse model of viral infection. Thereby, we employed an anti-cytokine "auto-vaccination" with an OVA/IL-17A complex or a Monoclonal Ab (MAb) anti-IL-17A (MM17/F3). Competitive ELISA assays were used to estimate the quality of the humoral immune response and the amount of Abs to conformational versus linear antigenic determinants. Results indicated that the OVA/IL-17A complex increased Abs levels to conformational epitopes of OVA, while LDV prolonged antibodies for a longer period. Mice treated with MM17F3 MAb showed an increase in Abs to conformational epitopes of OVA. A similar effect, confirmed by a competitive Western-blot assay, was produced by LDV. Moreover, an increased level of IgM, IgG1, and IgG2a was found in infected animals. Similarly, MAb anti-IL-17A treatment increased the proportion of Ab to conformational epitopes of DTx in uninfected mice, while LDV decreased this parameter. In conclusion, our findings demonstrate a correlation between IL-17A neutralization and a change in Ab specificity to OVA or DTx, presenting a novel strategy for obtaining Abs with higher specificity.
Collapse
Affiliation(s)
- Macarena Ottobre
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Jacques Van Snick
- Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
| | - José L Aparicio
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina.
| |
Collapse
|
2
|
Bai L, Li Y, Lu C, Yang Y, Zhang J, Lu Z, Huang K, Xian S, Yang X, Na N, Huang F, Zeng Z. Anti-IL-17 Inhibits PINK1/Parkin Autophagy and M1 Macrophage Polarization in Rheumatic Heart Disease. Inflammation 2024:10.1007/s10753-024-02094-3. [PMID: 38977539 DOI: 10.1007/s10753-024-02094-3] [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: 04/13/2024] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
Rheumatic heart disease (RHD) is an important and preventable cause of cardiovascular death and disability, but the lack of clarity about its exact mechanisms makes it more difficult to find alternative methods or prevention and treatment. We previously demonstrated that increased IL-17 expression plays a crucial role in the development of RHD-related valvular inflammatory injury. Macrophage autophagy/polarization may be a pro-survival strategy in the initiation and resolution of the inflammatory process. This study investigated the mechanism by which IL-17 regulates autophagy/polarization activation in macrophages. A RHD rat model was generated, and the effects of anti-IL-17 and 3-methyladenine (3-MA) were analyzed. The molecular mechanisms underlying IL-17-induced macrophage autophagy/polarization were investigated via in vitro experiments. In our established RHD rat model, the activation of the macrophage PINK1/Parkin autophagic pathway in valve tissue was accompanied by M1 macrophage infiltration, and anti-IL-17 treatment inhibited autophagy and reversed macrophage inflammatory infiltration, thereby attenuating endothelial-mesenchymal transition (EndMT) in the valve tissue. The efficacy of 3-MA treatment was similar to that of anti-IL-17 treatment. Furthermore, in THP-1 cells, the pharmacological promotion of autophagy by IL-17 induced M1-type polarization, whereas the inhibition of autophagy by 3-MA reversed this process. Mechanistically, silencing PINK1 in THP-1 blocked autophagic flux. Moreover, IL-17-induced M1-polarized macrophages promoted EndMT in HUVECs. This study revealed that IL-17 plays an important role in EndMT in RHD via the PINK1/Parkin autophagic pathway and macrophage polarization, providing a potential therapeutic target.
Collapse
Affiliation(s)
- Ling Bai
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Yuan Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Chuanghong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Yiping Yang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Jie Zhang
- Emergency Office, Nanning Center for Disease Control and Prevention, Nanning , Guangxi, China
| | - Zirong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Keke Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Shenglin Xian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Xi Yang
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Na Na
- Department of Neuroscience, The Scripps Research Institute, La Jolla, USA
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China.
| | - Zhiyu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China.
| |
Collapse
|
3
|
Hayashi H, Sun J, Yanagida Y, Yoshida S, Baba S, Tenma A, Toyoura M, Kawabata S, Ehara T, Asaki R, Sakaguchi M, Tomioka H, Shimamura M, Morishita R, Rakugi H, Tomita T, Nakagami H. Peptide-based vaccine targeting IL17A attenuates experimental spondyloarthritis in HLA-B27 transgenic rats. RMD Open 2023; 9:rmdopen-2022-002851. [PMID: 36737108 PMCID: PMC9900070 DOI: 10.1136/rmdopen-2022-002851] [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: 11/09/2022] [Accepted: 01/19/2023] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Spondyloarthritis (SpA) is known as series of immune-mediated inflammatory disease of the axial and peripheral joints. Human leucocyte antigen (HLA)-B27 is a genetic risk factor for SpA. Recent evidence suggests that the interleukin -17 (IL17) axis strongly contributes to SpA. This study aimed to assess the efficacy of an IL17A peptide-based vaccine on SpA manifestations in model rats. METHODS HLA-B27/human β2-microglobulin (hβ2M) transgenic rats were immunised with heat-inactivated Mycobacterium tuberculosis (MT) to develop spondylitis and arthritis as an experimental SpA model after immunisation with a keyhole limpet hemocyanin-conjugated IL17A peptide-based vaccine with an alum adjuvant three times. The IL17A antibody titre was assessed using ELISA, and arthritis score and joint thickness were monitored two times a week. Enzyme-linked immunospot (ELISpot) assays for IL4- and interferon-γ-secreting splenocytes were conducted to evaluate IL17A-specific T cell activation. We also evaluated the effect of IL17A vaccine in SpA therapeutic model. RESULTS The IL17A peptide-based vaccine with alum adjuvant successfully induced antibody production and suppressed the arthritis score and joint thickness. X-ray and histological analyses showed that enthesitis, bone destruction and new bone formation were inhibited by the IL17A vaccine. The ELISpot assay showed that the IL17A peptide-based vaccine did not elicit any IL17A-reactive T cell responses. IL17A vaccine tends to mitigate, but not significant, in SpA treatment model. These data showed that the peptide-based vaccine targeting IL17A alleviated the SpA phenotype in a heat-inactivated MT-induced SpA model in HLA-B27/hβ2M transgenic rats. CONCLUSIONS IL17A peptide-based vaccine may be a therapeutic option for SpA treatment.
Collapse
Affiliation(s)
- Hiroki Hayashi
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jiao Sun
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuka Yanagida
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shota Yoshida
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan,Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Satoshi Baba
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan,Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | - Munehisa Shimamura
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan,Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tetsuya Tomita
- Graduate School of Health Science, Morinomiya University of Medical Sciences, Osaka, Osaka, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
4
|
Gaignage M, Zhang X, Stockis J, Dedobbeleer O, Michiels C, Cochez P, Dumoutier L, Coulie PG, Lucas S. Blocking GARP-mediated activation of TGF-β1 did not alter innate or adaptive immune responses to bacterial infection or protein immunization in mice. Cancer Immunol Immunother 2022; 71:1851-1862. [PMID: 34973084 PMCID: PMC9294018 DOI: 10.1007/s00262-021-03119-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022]
Abstract
Abstract Transmembrane protein GARP binds latent TGF-β1 to form GARP:(latent)TGF-β1 complexes on the surface of several cell types including Tregs, B-cells, and platelets. Upon stimulation, these cells release active TGF-β1. Blocking TGF-β1 activation by Tregs with anti-GARP:TGF-β1 mAbs overcomes resistance to PD1/PD-L1 blockade and induces immune-mediated regressions of murine tumors, indicating that Treg-derived TGF-β1 inhibits anti-tumor immunity. TGF-β1 exerts a vast array of effects on immune responses. For example, it favors differentiation of TH17 cells and B-cell switch to IgA production, two important processes for mucosal immunity. Here, we sought to determine whether treatment with anti-GARP:TGF-β1 mAbs would perturb immune responses to intestinal bacterial infection. We observed no aggravation of intestinal disease, no systemic dissemination, and no alteration of innate or adaptative immune responses upon oral gavage of C. rodentium in highly susceptible Il22r−/− mice treated with anti-GARP:TGF-β1 mAbs. To examine the effects of GARP:TGF-β1 blockade on Ig production, we compared B cell- and TH cell- responses to OVA or CTB protein immunization in mice carrying deletions of Garp in Tregs, B cells, or platelets. No alteration of adaptive immune responses to protein immunization was observed in the absence of GARP on any of these cells. Altogether, we show that antibody-mediated blockade of GARP:TGF-β1 or genetic deletion of Garp in Tregs, B cells or platelets, do not alter innate or adaptive immune responses to intestinal bacterial infection or protein immunization in mice. Anti-GARP:TGF-β1 mAbs, currently tested for cancer immunotherapy, may thus restore anti-tumor immunity without severely impairing other immune defenses. Précis Immunotherapy with GARP:TGF-β1 mAbs may restore anti-tumor immunity without impairing immune or inflammatory responses required to maintain homeostasis or host defense against infection, notably at mucosal barriers. Supplementary Information The online version contains supplementary material available at 10.1007/s00262-021-03119-8.
Collapse
Affiliation(s)
- Mélanie Gaignage
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Xuhao Zhang
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Julie Stockis
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Olivier Dedobbeleer
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Camille Michiels
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Perrine Cochez
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Laure Dumoutier
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Pierre G Coulie
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium
| | - Sophie Lucas
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium.
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium.
| |
Collapse
|
5
|
Regen T, Isaac S, Amorim A, Núñez NG, Hauptmann J, Shanmugavadivu A, Klein M, Sankowski R, Mufazalov IA, Yogev N, Huppert J, Wanke F, Witting M, Grill A, Gálvez EJC, Nikolaev A, Blanfeld M, Prinz I, Schmitt-Kopplin P, Strowig T, Reinhardt C, Prinz M, Bopp T, Becher B, Ubeda C, Waisman A. IL-17 controls central nervous system autoimmunity through the intestinal microbiome. Sci Immunol 2021; 6:6/56/eaaz6563. [PMID: 33547052 DOI: 10.1126/sciimmunol.aaz6563] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/24/2020] [Indexed: 12/17/2022]
Abstract
Interleukin-17A- (IL-17A) and IL-17F-producing CD4+ T helper cells (TH17 cells) are implicated in the development of chronic inflammatory diseases, such as multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). TH17 cells also orchestrate leukocyte invasion of the central nervous system (CNS) and subsequent tissue damage. However, the role of IL-17A and IL-17F as effector cytokines is still confused with the encephalitogenic function of the cells that produce these cytokines, namely, TH17 cells, fueling a long-standing debate in the neuroimmunology field. Here, we demonstrated that mice deficient for IL-17A/F lose their susceptibility to EAE, which correlated with an altered composition of their gut microbiota. However, loss of IL-17A/F in TH cells did not diminish their encephalitogenic capacity. Reconstitution of a wild-type-like intestinal microbiota or reintroduction of IL-17A specifically into the gut epithelium of IL-17A/F-deficient mice reestablished their susceptibility to EAE. Thus, our data demonstrated that IL-17A and IL-17F are not encephalitogenic mediators but rather modulators of intestinal homeostasis that indirectly alter CNS-directed autoimmunity.
Collapse
Affiliation(s)
- Tommy Regen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sandrine Isaac
- Department of Genomics and Health, Center for Advanced Research in Public Health, FISABIO, Valencia, Spain
| | - Ana Amorim
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Judith Hauptmann
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Arthi Shanmugavadivu
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Roman Sankowski
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.,Berta-Ottenstein-Program for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ilgiz A Mufazalov
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Nir Yogev
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Jula Huppert
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Florian Wanke
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michael Witting
- Research Unit Analytical BioGeoChemistry, Helmholtz Center Munich, Munich, Germany.,Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University Munich, Munich, Germany
| | - Alexandra Grill
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Eric J C Gálvez
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Alexei Nikolaev
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michaela Blanfeld
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Center Munich, Munich, Germany.,Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University Munich, Munich, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.,Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center for NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Carles Ubeda
- Department of Genomics and Health, Center for Advanced Research in Public Health, FISABIO, Valencia, Spain.,Center of Biomedical Research Network (CIBER), Epidemiology and Public Health, Madrid, Spain
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany. .,Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
6
|
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: 45] [Impact Index Per Article: 11.3] [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
|
7
|
Shores LS, Kelly SH, Hainline KM, Suwanpradid J, MacLeod AS, Collier JH. Multifactorial Design of a Supramolecular Peptide Anti-IL-17 Vaccine Toward the Treatment of Psoriasis. Front Immunol 2020; 11:1855. [PMID: 32973764 PMCID: PMC7461889 DOI: 10.3389/fimmu.2020.01855] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022] Open
Abstract
Current treatments for chronic immune-mediated diseases such as psoriasis, rheumatoid arthritis, or Crohn's disease commonly rely on cytokine neutralization using monoclonal antibodies; however, such approaches have drawbacks. Frequent repeated dosing can lead to the formation of anti-drug antibodies and patient compliance issues, and it is difficult to identify a single antibody that is broadly efficacious across diverse patient populations. As an alternative to monoclonal antibody therapy, anti-cytokine immunization is a potential means for long-term therapeutic control of chronic inflammatory diseases. Here we report a supramolecular peptide-based approach for raising antibodies against IL-17 and demonstrate its efficacy in a murine model of psoriasis. B-cell epitopes from IL-17 were co-assembled with the universal T-cell epitope PADRE using the Q11 self-assembling peptide nanofiber system. These materials, with or without adjuvants, raised antibody responses against IL-17. Exploiting the modularity of the system, multifactorial experimental designs were used to select formulations maximizing titer and avidity. In a mouse model of psoriasis induced by imiquimod, unadjuvanted nanofibers had therapeutic efficacy, which could be enhanced with alum adjuvant but reversed with CpG adjuvant. Measurements of antibody subclass induced by adjuvanted and unadjuvanted formulations revealed strong correlations between therapeutic efficacy and titers of IgG1 (improved efficacy) or IgG2b (worsened efficacy). These findings have important implications for the development of anti-cytokine active immunotherapies and suggest that immune phenotype is an important metric for eliciting therapeutic anti-cytokine antibody responses.
Collapse
Affiliation(s)
- Lucas S Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Sean H Kelly
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Kelly M Hainline
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Jutamas Suwanpradid
- Department of Dermatology, Duke University School of Medicine, Durham, NC, United States
| | - Amanda S MacLeod
- Department of Dermatology, Duke University School of Medicine, Durham, NC, United States.,Department of Immunology, Duke University School of Medicine, Durham, NC, United States.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, United States.,Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
8
|
Abbaspour Sani N, Hasani M, Kianmehr A, Mohammadi S, Sheikh Arabi M, Yazdani Y. Enhanced nuclear translocation and activation of aryl hydrocarbon receptor (AhR) in THP-1 monocytic cell line by a novel niosomal formulation of indole-3-carbinol. J Liposome Res 2020; 30:117-125. [PMID: 30917715 DOI: 10.1080/08982104.2019.1600545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although niosomes structurally resemble liposomes, they are composed of nonionic surfactants which result in less toxicity and more stability. Here, we developed a novel niosomal formulation of I3C and investigated the nuclear translocation and activation of AhR among human acute myeloid leukaemia (AML) monocytic THP-1 cell line. Niosomal vesicles comprised of nonionic surfactants, cholesterol and I3C were prepared using thin film hydration (TFH) method and characterized according to the entrapment efficiency (EE %), size and zeta potential, by Dynamic light scattering method (DLS), and the surface morphology visualized by Transmission electron microscopy (TEM). In vitro release of I3C was evaluated and MTS assay was used to evaluate the viability of THP-1 cells. The nuclear translocation of AhR was assessed by immunocytochemistry (ICC) and Real-time RT-PCR was conducted using AhR target genes. The ratio of Cholesterol:Span 60 (1:1) niosomal formulations with the highest significant EE% were selected. I3C exerted cytotoxic effects on THP-1 cells in a dose- and time-dependent manner, while administration of niosomal I3C reduced these effects. Both niosomal and free I3C formulations facilitated the nuclear translocation of AhR. CYP1A1 was overexpressed in response to both free and niosomal I3C treatments, while IL1β was overexpressed merely in niosomal I3C-treated THP-1 cells. Niosomal formulation of I3C resulted in reduced cytotoxicity effects by enhancing the functional effects of I3C on AhR in THP-1 cells, including its nuclear translocation and overexpression of the target genes.
Collapse
Affiliation(s)
- Neda Abbaspour Sani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahsa Hasani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Anvarsadat Kianmehr
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Saeed Mohammadi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehdi Sheikh Arabi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Yaghoub Yazdani
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Laboratory Science Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
9
|
Yang MG, Sun L, Han J, Zheng C, Liang H, Zhu J, Jin T. Biological characteristics of transcription factor RelB in different immune cell types: implications for the treatment of multiple sclerosis. Mol Brain 2019; 12:115. [PMID: 31881915 PMCID: PMC6935142 DOI: 10.1186/s13041-019-0532-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/04/2019] [Indexed: 12/22/2022] Open
Abstract
Transcription factor RelB is a member of the nuclear factror-kappa B (NF-κB) family, which plays a crucial role in mediating immune responses. Plenty of studies have demonstrated that RelB actively contributes to lymphoid organ development, dendritic cells maturation and function and T cells differentiation, as well as B cell development and survival. RelB deficiency may cause a variety of immunological disorders in both mice and humans. Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system which involves a board of immune cell populations. Thereby, RelB may exert an impact on MS by modulating the functions of dendritic cells and the differentiation of T cells and B cells. Despite intensive research, the role of RelB in MS and its animal model, experimental autoimmune encephalomyelitis, is still unclear. Herein, we give an overview of the biological characters of RelB, summarize the updated knowledge regarding the role of RelB in different cell types that contribute to MS pathogenesis and discuss the potential RelB-targeted therapeutic implications for MS.
Collapse
Affiliation(s)
- Meng-Ge Yang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Jinming Han
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.,Present address: Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Chao Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Hudong Liang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.
| |
Collapse
|
10
|
Yi JS, Russo MA, Raja S, Massey JM, Juel VC, Shin J, Hobson-Webb LD, Gable K, Guptill JT. Inhibition of the transcription factor ROR-γ reduces pathogenic Th17 cells in acetylcholine receptor antibody positive myasthenia gravis. Exp Neurol 2019; 325:113146. [PMID: 31838097 DOI: 10.1016/j.expneurol.2019.113146] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/27/2019] [Accepted: 12/10/2019] [Indexed: 12/25/2022]
Abstract
IL-17 producing CD4 T cells (Th17) cells increase significantly with disease severity in myasthenia gravis (MG) patients. To suppress the generation of Th17 cells, we examined the effect of inhibiting retinoic acid receptor-related-orphan-receptor-C (RORγ), a Th17-specific transcription factor critical for differentiation. RORγ inhibition profoundly reduced Th17 cell frequencies, including IFN-γ and IL-17 co-producing pathogenic Th17 cells. Other T helper subsets were not affected. In parallel, CD8 T cell subsets producing IL-17 and IL-17/IFN-γ were increased in MG patients and inhibited by the RORγ inhibitor. These findings provide rationale for exploration of targeted Th17 therapies, including ROR-γ inhibitors, to treat MG patients.
Collapse
Affiliation(s)
- John S Yi
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, 915 S., LaSalle Street, Box 2926, Durham, NC 27710, USA.
| | - Melissa A Russo
- Neuromuscular Section, Department of Neurology, Duke University Medical Center, Box 3403, Durham, NC 27710, USA
| | - Shruti Raja
- Neuromuscular Section, Department of Neurology, Duke University Medical Center, Box 3403, Durham, NC 27710, USA
| | - Janice M Massey
- Neuromuscular Section, Department of Neurology, Duke University Medical Center, Box 3403, Durham, NC 27710, USA
| | - Vern C Juel
- Neuromuscular Section, Department of Neurology, Duke University Medical Center, Box 3403, Durham, NC 27710, USA
| | - Jay Shin
- Duke University, Durham, NC 27710, USA
| | - Lisa D Hobson-Webb
- Neuromuscular Section, Department of Neurology, Duke University Medical Center, Box 3403, Durham, NC 27710, USA
| | - Karissa Gable
- Neuromuscular Section, Department of Neurology, Duke University Medical Center, Box 3403, Durham, NC 27710, USA
| | - Jeffrey T Guptill
- Neuromuscular Section, Department of Neurology, Duke University Medical Center, Box 3403, Durham, NC 27710, USA
| |
Collapse
|
11
|
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: 7] [Impact Index Per Article: 1.4] [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
|
12
|
Danneskiold-Samsøe NB, Dias de Freitas Queiroz Barros H, Santos R, Bicas JL, Cazarin CBB, Madsen L, Kristiansen K, Pastore GM, Brix S, Maróstica Júnior MR. Interplay between food and gut microbiota in health and disease. Food Res Int 2019; 115:23-31. [DOI: 10.1016/j.foodres.2018.07.043] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/01/2018] [Accepted: 07/28/2018] [Indexed: 12/14/2022]
|
13
|
Rueda N, Vidal V, García-Cerro S, Narcís JO, Llorens-Martín M, Corrales A, Lantigua S, Iglesias M, Merino J, Merino R, Martínez-Cué C. Anti-IL17 treatment ameliorates Down syndrome phenotypes in mice. Brain Behav Immun 2018; 73:235-251. [PMID: 29758264 DOI: 10.1016/j.bbi.2018.05.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/02/2018] [Accepted: 05/07/2018] [Indexed: 12/12/2022] Open
Abstract
Down syndrome (DS) is characterized by structural and functional anomalies that are present prenatally and that lead to intellectual disabilities. Later in life, the cognitive abilities of DS individuals progressively deteriorate due to the development of Alzheimer's disease (AD)-associated neuropathology (i.e., β-amyloid (Aβ) plaques, neurofibrillary tangles (NFTs), neurodegeneration, synaptic pathology, neuroinflammation and increased oxidative stress). Increasing evidence has shown that among these pathological processes, neuroinflammation plays a predominant role in AD etiopathology. In AD mouse models, increased neuroinflammation appears earlier than Aβ plaques and NFTs, and in DS and AD models, neuroinflammation exacerbates the levels of soluble and insoluble Aβ species, favoring neurodegeneration. The Ts65Dn (TS) mouse, the most commonly used murine model of DS, recapitulates many alterations present in both DS and AD individuals, including enhanced neuroinflammation. In this study, we observed an altered neuroinflammatory milieu in the hippocampus of the TS mouse model. Pro-inflammatory mediators that were elevated in the hippocampus of this model included pro-inflammatory cytokine IL17A, which has a fundamental role in mediating brain damage in neuroinflammatory processes. Here, we analyzed the ability of an anti-IL17A antibody to reduce the neuropathological alterations that are present in TS mice during early neurodevelopmental stages (i.e., hippocampal neurogenesis and hypocellularity) or that are aggravated in later-life stages (i.e., cognitive abilities, cholinergic neuronal loss and increased cellular senescence, APP expression, Aβ peptide expression and neuroinflammation). Administration of anti-IL17 for 5 months, starting at the age of 7 months, partially improved the cognitive abilities of the TS mice, reduced the expression of several pro-inflammatory cytokines and the density of activated microglia and normalized the APP and Aβ1-42 levels in the hippocampi of the TS mice. These results suggest that IL17-mediated neuroinflammation is involved in several AD phenotypes in TS mice and provide a new therapeutic target to reduce these pathological characteristics.
Collapse
Affiliation(s)
- Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Verónica Vidal
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Susana García-Cerro
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Josep Oriol Narcís
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - María Llorens-Martín
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa", CBMSO, CSICUAM, Madrid, Spain; Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - Andrea Corrales
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Sara Lantigua
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Marcos Iglesias
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, USA
| | - Jesús Merino
- Department of Molecular Biology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Ramón Merino
- Institute of Biomedicine and Biotechnology of Cantabria, Consejo Superior de Investigaciones Científicas-University of Cantabria, Santander, Spain.
| | - Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain.
| |
Collapse
|
14
|
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: 8] [Impact Index Per Article: 1.3] [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
|
15
|
Zhang S. The role of transforming growth factor β in T helper 17 differentiation. Immunology 2018; 155:24-35. [PMID: 29682722 PMCID: PMC6099164 DOI: 10.1111/imm.12938] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/11/2018] [Accepted: 04/11/2018] [Indexed: 12/12/2022] Open
Abstract
T helper 17 (Th17) cells play critical roles in inflammatory and autoimmune diseases. The lineage-specific transcription factor RORγt is the key regulator for Th17 cell fate commitment. A substantial number of studies have established the importance of transforming growth factor β (TGF-β) -dependent pathways in inducing RORγt expression and Th17 differentiation. TGF-β superfamily members TGF-β1 , TGF-β3 or activin A, in concert with interleukin-6 or interleukin-21, differentiate naive T cells into Th17 cells. Alternatively, Th17 differentiation can occur through TGF-β-independent pathways. However, the mechanism of how TGF-β-dependent and TGF-β-independent pathways control Th17 differentiation remains controversial. This review focuses on the perplexing role of TGF-β in Th17 differentiation, depicts the requirement of TGF-β for Th17 development, and underscores the multiple mechanisms underlying TGF-β-promoted Th17 generation, pathogenicity and plasticity. With new insights and comprehension from recent findings, this review specifically tackles the involvement of the canonical TGF-β signalling components, SMAD2, SMAD3 and SMAD4, summarizes diverse SMAD-independent mechanisms, and highlights the importance of TGF-β signalling in balancing the reciprocal conversion of Th17 and regulatory T cells. Finally, this review includes discussions and perspectives and raises important mechanistic questions about the role of TGF-β in Th17 generation and function.
Collapse
Affiliation(s)
- Song Zhang
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
| |
Collapse
|
16
|
Effects of interleukin 17A (IL-17A) neutralization on murine hepatitis virus (MHV-A59) infection. Eur Cytokine Netw 2018; 28:111-119. [PMID: 29187338 PMCID: PMC7099234 DOI: 10.1684/ecn.2017.0399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mice infected with mouse hepatitis virus A59 (MHV-A59) develop hepatitis and autoantibodies (autoAb) to liver and kidney fumarylacetoacetate hydrolase (FAH), a fact closely related to the release of alarmins such as uric acid and/or high-mobility group box protein 1 (HMGB1). We studied the effect of neutralizing monoclonal antibodies (MAb) against IL-17A in our model of mouse MHV-A59-infection. MAb anti-IL-17F and anti-IFNγ were used to complement the study. Results showed that transaminase levels markedly decreased in MHV-A59-infected mice treated with MAb anti-IL-17A whereas plasmatic Ig concentration sharply increased. Conversely, MAb anti-IL-17F enhanced transaminase liberation and did not affect Ig levels. Serum IFNγ was detected in mice infected with MHV-A59 and its concentration increased after MAb anti-IL-17A administration. Besides, MAb anti-IFNγ greatly augmented transaminase plasmatic levels. IL-17A neutralization did not affect MHV-A59-induction of HMGB1 liberation and slightly augmented plasmatic uric acid concentration. However, mice treated with the MAb failed to produce autoAb to FAH. The above results suggest a reciprocal regulation of Th1 and Th17 cells acting on the different MHV-A59 effects. In addition, it is proposed that IL-17A is involved in alarmins adjuvant effects leading to autoAb expression.
Collapse
|
17
|
Handono K, Pratama MZ, Sari DK, Hermawan HO, Agdana HM, Kawuningan KB, Nur'aini N, Hasanah D, Kalim H. Effect of active immunization with IL-17A on B cell function and infection risk in pristane-induced lupus model. Int J Rheum Dis 2018; 21:1277-1286. [DOI: 10.1111/1756-185x.13325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Kusworini Handono
- Department of Clinical Pathology; Universitas Brawijaya; Malang Indonesia
| | - Mirza Zaka Pratama
- Rheumatology and Immunology Division; Department of Internal Medicine; Universitas Brawijaya; Malang Indonesia
| | - Dita Kartika Sari
- Master Degrees of Biomedical Sciences; Faculty of Medicine; Universitas Brawijaya; Malang Indonesia
| | - Hanestya Oky Hermawan
- Master Degrees of Biomedical Sciences; Faculty of Medicine; Universitas Brawijaya; Malang Indonesia
| | | | | | - Nafisah Nur'aini
- Master Degrees of Biomedical Sciences; Faculty of Medicine; Universitas Brawijaya; Malang Indonesia
| | - Dian Hasanah
- Rheumatology and Immunology Division; Department of Internal Medicine; Universitas Brawijaya; Malang Indonesia
| | - Handono Kalim
- Rheumatology and Immunology Division; Department of Internal Medicine; Universitas Brawijaya; Malang Indonesia
| |
Collapse
|
18
|
Smith PA, Schmid C, Zurbruegg S, Jivkov M, Doelemeyer A, Theil D, Dubost V, Beckmann N. Fingolimod inhibits brain atrophy and promotes brain-derived neurotrophic factor in an animal model of multiple sclerosis. J Neuroimmunol 2018. [PMID: 29530550 DOI: 10.1016/j.jneuroim.2018.02.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Longitudinal brain atrophy quantification is a critical efficacy measurement in multiple sclerosis (MS) clinical trials and the determination of No Evidence of Disease Activity (NEDA). Utilising fingolimod as a clinically validated therapy we evaluated the use of repeated brain tissue volume measures during chronic experimental autoimmune encephalomyelitis (EAE) as a new preclinical efficacy measure. Brain volume changes were quantified using magnetic resonance imaging (MRI) at 7 Tesla and correlated to treatment-induced brain derived neurotrophic factor (BDNF) measured in blood, cerebrospinal fluid, spinal cord and brain. Serial brain MRI measurements revealed slow progressive brain volume loss in vehicle treated EAE mice despite a stable clinical score. Fingolimod (1 mg/kg) significantly ameliorated brain tissue atrophy in the cerebellum and striatum when administered from established EAE disease onwards. Fingolimod-dependent tissue preservation was associated with induction of BDNF specifically within the brain and co-localized with neuronal soma. In contrast, therapeutic teriflunomide (3 mg/kg) treatment failed to inhibit CNS autoimmune mediated brain degeneration. Finally, weekly anti-IL-17A antibody (15 mg/kg) treatment was highly efficacious and preserved whole brain, cerebellum and striatum volume. Fingolimod-mediated BDNF increases within the CNS may contribute to limiting progressive tissue loss during chronic neuroinflammation.
Collapse
Affiliation(s)
- Paul A Smith
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| | - Cindy Schmid
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| | - Stefan Zurbruegg
- Neurosciences, Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| | - Magali Jivkov
- Preclinical Safety, Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| | - Arno Doelemeyer
- Musculoskeletal Diseases, Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| | - Diethilde Theil
- Preclinical Safety, Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| | - Valérie Dubost
- Preclinical Safety, Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| | - Nicolau Beckmann
- Musculoskeletal Diseases, Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| |
Collapse
|
19
|
Neutralization of either IL-17A or IL-17F is sufficient to inhibit house dust mite induced allergic asthma in mice. Clin Sci (Lond) 2017; 131:2533-2548. [PMID: 29026003 DOI: 10.1042/cs20171034] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/24/2017] [Accepted: 08/29/2017] [Indexed: 01/09/2023]
Abstract
T helper (Th)17 immune response participates in allergic lung inflammation and asthma is reduced in the absence of interleukin (IL)-17 in mice. Since IL-17A and IL-17F are induced and bind the shared receptor IL-17RA, we asked whether both IL-17A and IL-17F contribute to house dust mite (HDM) induced asthma. We report that allergic lung inflammation is attenuated in absence of either IL-17A or IL-17F with reduced airway hyperreactivity, eosinophilic inflammation, goblet cell hyperplasia, cytokine and chemokine production as found in absence of IL-17RA. Furthermore, specific antibody neutralization of either IL-17A or IL-17F given during the sensitization phase attenuated allergic lung inflammation and airway hyperreactivity. In vitro activation by HDM of primary dendritic cells revealed a comparable induction of CXCL1 and IL-6 expression and the response to IL-17A and IL-17F relied on IL-17RA signaling via the adaptor protein act1 in fibroblasts. Therefore, HDM-induced allergic respiratory response depends on IL-17RA via act1 signaling and inactivation of either IL-17A or IL-17F is sufficient to attenuate allergic asthma in mice.
Collapse
|
20
|
Thorenz A, Völker N, Bräsen JH, Chen R, Jang MS, Rong S, Haller H, Kirsch T, Vieten G, Klemann C, Gueler F. IL-17A blockade or deficiency does not affect progressive renal fibrosis following renal ischaemia reperfusion injury in mice. J Pharm Pharmacol 2017; 69:1125-1135. [DOI: 10.1111/jphp.12747] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/18/2017] [Indexed: 12/31/2022]
Abstract
Abstract
Objectives
IL-17A contributes to acute kidney injury and fibrosis. Therefore, we asked whether IL-17A deficiency or treatment with a IL-17A blocking antibody impacts severe renal ischaemia reperfusion injury (IRI) and the progression to chronic kidney disease (CKD).
Methods
IL-17A-deficient and wild-type (WT) mice underwent transient unilateral renal pedicle clamping for 45 min to induce IRI and subsequent renal fibrosis. Furthermore, a neutralizing anti-IL-17A antibody (mAb) was injected into WT mice before induction of renal IRI intravenously. On days 1, 7 and 21, inflammation, fibrosis, leukocyte infiltration and pro-inflammatory and pro-fibrotic cytokine expression were assessed in kidneys using histology, qPCR and flow cytometry.
Key findings
IL-17A was significantly increased after renal IRI in WT kidneys. Levels of pro-inflammatory (MCP-1) cytokine and pro-fibrotic (collagen 1α1, fibronectin) transcripts were similar in the experimental groups studied. IL-17A deficiency had no effect on renal T-cell influx or the number, inflammatory phenotype, or spatial distribution of macrophages. Similarly, administration of an IL-17A blocking antibody did not attenuate inflammation.
Conclusions
Despite the effects of IL-17 in other inflammation models, neither genetic IL-17A deficiency nor treatment with an IL-17A blocking antibody attenuated IRI and progression to CKD. We conclude that in severe renal IRI IL-17A is not crucially involved in disease progression.
Collapse
Affiliation(s)
- Anja Thorenz
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Nicole Völker
- Nephrology, Hannover Medical School, Hannover, Germany
| | | | - Rongjun Chen
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Mi-Sun Jang
- Nephrology, Hannover Medical School, Hannover, Germany
| | - Song Rong
- Nephrology, Hannover Medical School, Hannover, Germany
- The Transplantation Center of the Affiliated Hospital, Zunyi Medical College, Zunyi, China
| | | | - Torsten Kirsch
- Nephrology, Hannover Medical School, Hannover, Germany
- Core Facility Quality Management and Health Technology Assessment in Transplantation, Hannover Medical School, Hannover, Germany
| | - Gertrud Vieten
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Christian Klemann
- Center of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Faikah Gueler
- Nephrology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
21
|
Veldhoen M. Interleukin 17 is a chief orchestrator of immunity. Nat Immunol 2017; 18:612-621. [DOI: 10.1038/ni.3742] [Citation(s) in RCA: 307] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/03/2017] [Indexed: 12/11/2022]
|
22
|
Clemente N, Comi C, Raineri D, Cappellano G, Vecchio D, Orilieri E, Gigliotti CL, Boggio E, Dianzani C, Sorosina M, Martinelli-Boneschi F, Caldano M, Bertolotto A, Ambrogio L, Sblattero D, Cena T, Leone M, Dianzani U, Chiocchetti A. Role of Anti-Osteopontin Antibodies in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis. Front Immunol 2017; 8:321. [PMID: 28386258 PMCID: PMC5362623 DOI: 10.3389/fimmu.2017.00321] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/07/2017] [Indexed: 11/23/2022] Open
Abstract
Osteopontin (OPN) is highly expressed in demyelinating lesions in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). OPN is cleaved by thrombin into N- (OPN-N) and C-terminal (OPN-C) fragments with different ligands and functions. In EAE, administering recombinant OPN induces relapses, whereas treatment with anti-OPN antibodies ameliorates the disease. Anti-OPN autoantibodies (autoAbs) are spontaneously produced during EAE but have never been detected in MS. The aim of the study was to evaluate anti-OPN autoAbs in the serum of MS patients, correlate them with disease course, and recapitulate the human findings in EAE. We performed ELISA in the serum of 122 patients collected cross-sectionally, and 50 patients with relapsing–remitting (RR) disease collected at diagnosis and followed longitudinally for 10 years. In the cross-sectional patients, the autoAb levels were higher in the RR patients than in the primary- and secondary-progressive MS and healthy control groups, and they were highest in the initial stages of the disease. In the longitudinal group, the levels at diagnosis directly correlated with the number of relapses during the following 10 years. Moreover, in patients with active disease, who underwent disease-modifying treatments, autoAbs were higher than in untreated patients and were associated with low MS severity score. The autoAb displayed neutralizing activity and mainly recognized OPN-C rather than OPN-N. To confirm the clinical effect of these autoAbs in vivo, EAE was induced using myelin oligodendrocyte glycoprotein MOG35–55 in C57BL/6 mice pre-vaccinated with ovalbumin (OVA)-linked OPN or OVA alone. We then evaluated the titer of antibodies to OPN, the clinical scores and in vitro cytokine secretion by spleen lymphocytes. Vaccination significantly induced antibodies against OPN during EAE, decreased disease severity, and the protective effect was correlated with decreased T cell secretion of interleukin 17 and interferon-γ ex vivo. The best effect was obtained with OPN-C, which induced significantly faster and more complete remission than other OPN vaccines. In conclusion, these data suggest that production of anti-OPN autoAbs may favor remission in both MS and EAE. Novel strategies boosting their levels, such as vaccination or passive immunization, may be proposed as a future strategy in personalized MS therapy.
Collapse
Affiliation(s)
- Nausicaa Clemente
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO) , Novara , Italy
| | - Cristoforo Comi
- Department of Translational Medicine, IRCAD, Neurology Unit, University of Piemonte Orientale (UPO) , Novara , Italy
| | - Davide Raineri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO) , Novara , Italy
| | - Giuseppe Cappellano
- Division for Experimental Pathophysiology and Immunology, Biocenter, Medical University of Innsbruck , Innsbruck , Austria
| | - Domizia Vecchio
- Department of Translational Medicine, IRCAD, Neurology Unit, University of Piemonte Orientale (UPO) , Novara , Italy
| | - Elisabetta Orilieri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO) , Novara , Italy
| | - Casimiro L Gigliotti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO) , Novara , Italy
| | - Elena Boggio
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO) , Novara , Italy
| | - Chiara Dianzani
- Department of Drug Science and Technology, University of Turin , Torino , Italy
| | - Melissa Sorosina
- Laboratory of Human Genetics of Neurological Disorders, CNS Inflammatory Unit, Division of Neuroscience, Institute of Experimental Neurology (INSPE), San Raffaele Scientific Institute , Milano , Italy
| | - Filippo Martinelli-Boneschi
- Laboratory of Human Genetics of Neurological Disorders, CNS Inflammatory Unit, Division of Neuroscience, Institute of Experimental Neurology (INSPE), San Raffaele Scientific Institute , Milano , Italy
| | - Marzia Caldano
- Neurology Unit 2, Centro Riferimento Regionale Sclerosi Multipla (CRESM), Azienda Ospedaliero-Universitaria San Luigi , Orbassano , Italy
| | - Antonio Bertolotto
- Neurology Unit 2, Centro Riferimento Regionale Sclerosi Multipla (CRESM), Azienda Ospedaliero-Universitaria San Luigi , Orbassano , Italy
| | - Luca Ambrogio
- ASO Neurologia, Azienda Ospedaliera S. Croce e Carle , Cuneo , Italy
| | | | - Tiziana Cena
- Department of Translational Medicine, Medical Statistics Unit, University of Piemonte Orientale (UPO) , Novara , Italy
| | - Maurizio Leone
- IRCAD, Neurology Unit, Scientific Institute, Hospital "Casa Sollievo della Sofferenza" , San Giovanni Rotondo , Italy
| | - Umberto Dianzani
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO) , Novara , Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO) , Novara , Italy
| |
Collapse
|
23
|
Qiu AW, Liu QH, Wang JL. Blocking IL-17A Alleviates Diabetic Retinopathy in Rodents. Cell Physiol Biochem 2017; 41:960-972. [PMID: 28222445 DOI: 10.1159/000460514] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/27/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND/AIMS Interleukin (IL)-17A, a proinflammatory cytokine, has been implicated in several autoimmune diseases. However, it is unclear whether IL-17A is involved in diabetic retinopathy (DR), one of the most serious complications of autoimmune diabetes. This study aimed to demonstrate that IL-17A exacerbates DR by affecting retinal Müller cell function. METHODS High glucose (HG)-treated rat Müller cell line (rMC-1) was exposed to IL-17A, anti-IL-17A-neutralizing monoclonal antibody (mAb) or/and anti-IL-17 receptor (R)A-neutralizing mAb for 24 h. For in vivo study, DR was induced by intraperitoneal injections of streptozotocin (STZ). DR model mice were treated with anti-IL-17A mAb or anti-IL-17RA mAb in the vitreous cavity. Mice that were prepared for retinal angiography were sacrificed two weeks after intravitreal injection, while the rest were sacrificed two days after intravitreal injection. RESULTS IL-17A production and IL-17RA expression were increased in both HG-treated rMC-1 and DR retina. HG induced rMC-1 activation and dysfunction, as determined by the increased GFAP, VEGF and glutamate levels as well as the downregulated GS and EAAT1 expression. IL-17A exacerbated the HG-induced rMC-1 functional disorders, whereas either anti-IL-17A mAb or anti-IL-17RA mAb alleviated the HG-induced rMC-1 disorders. Intravitreal injections with anti-IL-17A mAb or anti-IL-17RA mAb in DR model mice reduced Müller cell dysfunction, vascular leukostasis, vascular leakage, tight junction protein downregulation and ganglion cell apoptosis in the retina. CONCLUSIONS IL-17A aggravates DR-like pathology at least partly by impairing retinal Müller cell function. Blocking IL-17A is a potential therapeutic strategy for DR.
Collapse
|
24
|
Assier E, Bessis N, Zagury JF, Boissier MC. IL-1 Vaccination Is Suitable for Treating Inflammatory Diseases. Front Pharmacol 2017; 8:6. [PMID: 28197099 PMCID: PMC5281538 DOI: 10.3389/fphar.2017.00006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/04/2017] [Indexed: 01/25/2023] Open
Affiliation(s)
- Eric Assier
- UMR 1125 Institut National de la Santé et de la Recherche MédicaleBobigny, France; Sorbonne Paris Cité Université Paris 13Bobigny, France
| | - Natacha Bessis
- UMR 1125 Institut National de la Santé et de la Recherche MédicaleBobigny, France; Sorbonne Paris Cité Université Paris 13Bobigny, France
| | | | - Marie-Christophe Boissier
- UMR 1125 Institut National de la Santé et de la Recherche MédicaleBobigny, France; Sorbonne Paris Cité Université Paris 13Bobigny, France; Assistance Publique-Hôpitaux de Paris, HUPSSD, Service de RhumatologieBobigny, France
| |
Collapse
|
25
|
Haruta K, Otaki N, Nagamine M, Kayo T, Sasaki A, Hiramoto S, Takahashi M, Hota K, Sato H, Yamazaki H. A Novel PEGylation Method for Improving the Pharmacokinetic Properties of Anti-Interleukin-17A RNA Aptamers. Nucleic Acid Ther 2016; 27:36-44. [PMID: 27827561 PMCID: PMC5312557 DOI: 10.1089/nat.2016.0627] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The obstacles to the development of therapeutic aptamers for systemic inflammatory diseases, such as nuclease degradation and renal clearance, have not been fully overcome. Here, we report a novel PEGylation method, sbC-PEGylation, which improves the pharmacokinetic properties of RNA aptamers that act against interleukin-17A (IL-17A) in mice and monkeys. sbC-PEGylated aptamers were synthesized by coupling the symmetrical branching molecule 2-cyanoethyl-N,N-diisopropyl phosphoroamidite to the 5′ end of the aptamer, before conjugating two polyethylene glycol (PEG) molecules to the aptamer. Pharmacokinetic studies showed that compared with conventionally PEGylated aptamers, the sbC-PEGylated aptamer exhibited excellent stability in the blood circulation of mice and monkeys. In addition, one of the sbC-PEGylated aptamers, 17M-382, inhibited the interleukin-6 (IL-6) production induced by IL-17A in NIH3T3 cells in a concentration-dependent manner, and the half-maximal inhibitory concentration of sbC-PEGylated 17M-382 was two times lower than that of non-PEGylated 17M-382. Furthermore, the intraperitoneal administration of sbC-PEGylated 17M-382 significantly inhibited the IL-6 production induced by IL-17A in a mouse air pouch model. Our findings suggest that the novel PEGylation method described in this study, sbC-PEGylation, could be used to develop anti-IL-17A aptamers as a therapeutic option for systemic inflammatory disease.
Collapse
Affiliation(s)
| | - Natsuki Otaki
- 1 R&D Center, Zenyaku Kogyo Co., Ltd. , Tokyo, Japan
| | | | | | - Asako Sasaki
- 1 R&D Center, Zenyaku Kogyo Co., Ltd. , Tokyo, Japan
| | | | | | | | | | - Hiroaki Yamazaki
- 3 Prescription Products Development, Zenyaku Kogyo Co., Ltd. , Tokyo, Japan
| |
Collapse
|
26
|
Xu W, Chen L, Guo S, Wu L, Zhang J. Intranasal Administration of Recombinant Mycobacterium smegmatis Inducing IL-17A Autoantibody Attenuates Airway Inflammation in a Murine Model of Allergic Asthma. PLoS One 2016; 11:e0151581. [PMID: 26974537 PMCID: PMC4790942 DOI: 10.1371/journal.pone.0151581] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/01/2016] [Indexed: 11/30/2022] Open
Abstract
Asthma is a chronic inflammatory disorder, previous studies have shown that IL-17A contributes to the development of asthma, and there is a positive correlation between the level of IL-17A and the severity of disease. Here, we constructed recombinant Mycobacterium smegmatis expressing fusion protein Ag85A-IL-17A (rMS-Ag85a-IL-17a) and evaluated whether it could attenuate allergic airway inflammation, and further investigated the underlying mechanism. In this work, the murine model of asthma was established with ovalbumin, and mice were intranasally vaccinated with rMS-Ag85a-IL-17a. Autoantibody of IL-17A in sera was detected, and the airway inflammatory cells infiltration, the local cytokines and chemokines production and the histopathological changes of lung tissue were investigated. We found that the administration of rMS-Ag85a-IL-17a induced the autoantibody of IL-17A in sera. The vaccination of rMS-Ag85a-IL-17a remarkably reduced the infiltration of inflammatory cells and the secretion of mucus in lung tissue and significantly decreased the numbers of the total cells, eosinophils and neutrophils in BALF. Th1 cells count in spleen, Th1 cytokine levels in BALF and supernatant of splenocytes and mediastinal lymph nodes, and T-bet mRNA in lung tissue were significantly increased with rMS-Ag85a-IL-17a administration. Meanwhile, rMS-Ag85a-IL-17a vaccination markedly decreased Th2 cells count, Th2 cytokine and Th17 cytokine levels in BALF and supernatant of splenocytes and mediastinal lymph nodes, and chemokines mRNA expression in lung tissue. These data confirmed that recombinant Mycobacterium smegmatis in vivo could induce autoantibody of IL-17A, which attenuated asthmatic airway inflammation.
Collapse
Affiliation(s)
- Wanting Xu
- Department of Paediatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ling Chen
- Department of Paediatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Sheng Guo
- Department of Endocrinology, Shanghai Jiao Tong University Affiliated Children’s Hospital, Shanghai, China
| | - Liangxia Wu
- Department of Paediatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jianhua Zhang
- Department of Paediatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
27
|
Klemann C, Schröder A, Dreier A, Möhn N, Dippel S, Winterberg T, Wilde A, Yu Y, Thorenz A, Gueler F, Jörns A, Tolosa E, Leonhardt J, Haas JD, Prinz I, Vieten G, Petersen C, Kuebler JF. Interleukin 17, Produced by γδ T Cells, Contributes to Hepatic Inflammation in a Mouse Model of Biliary Atresia and Is Increased in Livers of Patients. Gastroenterology 2016; 150:229-241.e5. [PMID: 26404950 DOI: 10.1053/j.gastro.2015.09.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 09/06/2015] [Accepted: 09/15/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Biliary atresia (BA) is a rare disease in infants, with unknown mechanisms of pathogenesis. It is characterized by hepatobiliary inflammatory, progressive destruction of the biliary system leading to liver fibrosis, and deterioration of liver function. Interleukin (IL) 17A promotes inflammatory and autoimmune processes. We studied the role of IL17A and cells that produce this cytokine in a mouse model of BA and in hepatic biopsy samples from infants with BA. METHODS We obtained peripheral blood and liver tissue specimens from 20 patients with BA, collected at the time of Kasai portoenterostomy, along with liver biopsies from infants without BA (controls). The tissue samples were analyzed by reverse transcription quantitative polymerase chain reaction (PCR), in situ PCR, and flow cytometry analyses. BA was induced in balb/cAnNCrl mice by rhesus rotavirus infection; uninfected mice were used as controls. Liver tissues were collected from mice and analyzed histologically and by reverse transcriptase PCR; leukocytes were isolated, stimulated, and analyzed by flow cytometry and PCR analyses. Some mice were given 3 intraperitoneal injections of a monoclonal antibody against IL17 or an isotype antibody (control). RESULTS Livers from rhesus rota virus-infected mice with BA had 7-fold more Il17a messenger RNA than control mice (P = .02). γδ T cells were the exclusive source of IL17; no T-helper 17 cells were detected in livers of mice with BA. The increased number of IL17a-positive γδ T cells liver tissues of mice with BA was associated with increased levels of IL17A, IL17F, retinoid-orphan-receptor C, C-C chemokine receptor 6, and the IL23 receptor. Mice that were developing BA and given antibodies against IL17 had lower levels of liver inflammation and mean serum levels of bilirubin than mice receiving control antibodies (191 μmol/L vs 78 μmol/L, P = .002). Liver tissues from patients with BA had 4.6-fold higher levels of IL17 messenger RNA than control liver tissues (P = .02). CONCLUSIONS In livers of mice with BA, γδ T cells produce IL17, which is required for inflammation and destruction of the biliary system. IL17 is up-regulated in liver tissues from patients with BA, compared with controls, and might serve as a therapeutic target.
Collapse
Affiliation(s)
- Christian Klemann
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Arne Schröder
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Anika Dreier
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Nora Möhn
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Stephanie Dippel
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Thomas Winterberg
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Anne Wilde
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Yi Yu
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Anja Thorenz
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Faikah Gueler
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Leonhardt
- Department of Pediatric Surgery, St Bernward Hospital, Hildesheim, Germany
| | - Jan D Haas
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Gertrud Vieten
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Claus Petersen
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Joachim F Kuebler
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
28
|
Role of IL-4 receptor α-positive CD4(+) T cells in chronic airway hyperresponsiveness. J Allergy Clin Immunol 2015; 137:1852-1862.e9. [PMID: 26688514 DOI: 10.1016/j.jaci.2015.10.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND TH2 cells and their cytokines are associated with allergic asthma in human subjects and with mouse models of allergic airway disease. IL-4 signaling through the IL-4 receptor α (IL-4Rα) chain on CD4(+) T cells leads to TH2 cell differentiation in vitro, implying that IL-4Rα-responsive CD4(+) T cells are critical for the induction of allergic asthma. However, mechanisms regulating acute and chronic allergen-specific TH2 responses in vivo remain incompletely understood. OBJECTIVE This study defines the requirements for IL-4Rα-responsive CD4(+) T cells and the IL-4Rα ligands IL-4 and IL-13 in the development of allergen-specific TH2 responses during the onset and chronic phase of experimental allergic airway disease. METHODS Development of acute and chronic ovalbumin (OVA)-induced allergic asthma was assessed weekly in CD4(+) T cell-specific IL-4Rα-deficient BALB/c mice (Lck(cre)IL-4Rα(-/lox)) and respective control mice in the presence or absence of IL-4 or IL-13. RESULTS During acute allergic airway disease, IL-4 deficiency did not prevent the onset of TH2 immune responses and OVA-induced airway hyperresponsiveness or goblet cell hyperplasia, irrespective of the presence or absence of IL-4Rα-responsive CD4(+) T cells. In contrast, deficiency of IL-13 prevented allergic asthma, irrespective of the presence or absence of IL-4Rα-responsive CD4(+) T cells. Importantly, chronic allergic inflammation and airway hyperresponsiveness were dependent on IL-4Rα-responsive CD4(+) T cells. Deficiency in IL-4Rα-responsive CD4(+) T cells resulted in increased numbers of IL-17-producing T cells and, consequently, increased airway neutrophilia. CONCLUSION IL-4-responsive T helper cells are dispensable for acute OVA-induced airway disease but crucial in maintaining chronic asthmatic pathology.
Collapse
|
29
|
Yamagata T, Skepner J, Yang J. Targeting Th17 Effector Cytokines for the Treatment of Autoimmune Diseases. Arch Immunol Ther Exp (Warsz) 2015; 63:405-14. [PMID: 26358867 DOI: 10.1007/s00005-015-0362-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 07/28/2015] [Indexed: 12/14/2022]
Abstract
Interleukin (IL)-17-producing T cells, especially T helper (Th)17 cells, play a critical role in the pathogenesis of a variety of autoimmune inflammatory diseases. The pathogenic function of Th17 cells results from their production of Th17 effector cytokines, namely IL-17 (or IL-17A), IL-17F, IL-22 and IL-26. The importance of IL-17 has been demonstrated by antibody neutralization studies in both animal models of autoimmune diseases as well as in human clinical trials. This review highlights the current knowledge of the clinical aspects of the Th17 cytokines as well as therapeutic antibodies against IL-17, IL-17F, IL-17 receptor, IL-22, IL-26 and granulocyte macrophage colony-stimulating factor for the future treatment of autoimmune inflammatory diseases.
Collapse
Affiliation(s)
| | - Jill Skepner
- Tempero, GlaxoSmithKline, Cambridge, MA, 02139, USA
| | - Jianfei Yang
- Tempero, GlaxoSmithKline, Cambridge, MA, 02139, USA.
| |
Collapse
|
30
|
Oral administration of PDX1 confers protection against insulitis in the non-obese diabetic (NOD) mice. Biochem Biophys Res Commun 2015; 466:656-63. [PMID: 26403969 DOI: 10.1016/j.bbrc.2015.09.098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 09/17/2015] [Indexed: 12/22/2022]
Abstract
Type 1 diabetes is a T cell-mediated organ-specific autoimmune disease. Antigen-specific immune intervention allows the selective targeting of autoreactive T cell, while leaving the remainder of the immune system intact. However, immune intervention for type 1 diabetes has not yielded perfect results clinically. In our paper published previously, we asked whether pancreatic duodenal home box 1 (PDX1) is a target of anti-islet autoimmunity in type 1 diabetes. In this experiment, we assessed the therapeutic effect of oral administration of PDX1 on diabetes development of 4-week-old non-obese diabetic (NOD) mice. The results indicate that PDX1 immunization is an effective intervention strategy for delaying the onset of diabetes in NOD mice in association with: 1) reduced insulitis; 2) suppression of destructive autoreactive T cells; 3) augmentation of regulatory T cells; 4) a shift in cytokine production. The present observations suggest that immunization with PDX1 modulates immune cell responses in NOD mice, raising the possibility that it is beneficial in ameliorating autoimmune destruction of beta-cells and delaying type 1 diabetes development clinically.
Collapse
|
31
|
Rodgers DT, McGrath MA, Pineda MA, Al-Riyami L, Rzepecka J, Lumb F, Harnett W, Harnett MM. The parasitic worm product ES-62 targets myeloid differentiation factor 88-dependent effector mechanisms to suppress antinuclear antibody production and proteinuria in MRL/lpr mice. Arthritis Rheumatol 2015; 67:1023-35. [PMID: 25546822 PMCID: PMC4409857 DOI: 10.1002/art.39004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 12/16/2014] [Indexed: 12/30/2022]
Abstract
Objective The hygiene hypothesis suggests that parasitic helminths (worms) protect against the development of autoimmune disease via a serendipitous side effect of worm-derived immunomodulators that concomitantly promote parasite survival and limit host pathology. The aim of this study was to investigate whether ES-62, a phosphorylcholine-containing glycoprotein secreted by the filarial nematode Acanthocheilonema viteae, protects against kidney damage in an MRL/lpr mouse model of systemic lupus erythematosus (SLE). Methods MRL/lpr mice progressively produce high levels of autoantibodies, and the resultant deposition of immune complexes drives kidney pathology. The effects of ES-62 on disease progression were assessed by measurement of proteinuria, assessment of kidney histology, determination of antinuclear antibody (ANA) production and cytokine levels, and flow cytometric analysis of relevant cellular populations. Results ES-62 restored the disrupted balance between effector and regulatory B cells in MRL/lpr mice by inhibiting plasmablast differentiation, with a consequent reduction in ANA production and deposition of immune complexes and C3a in the kidneys. Moreover, by reducing interleukin-22 production, ES-62 may desensitize downstream effector mechanisms in the pathogenesis of kidney disease. Highlighting the therapeutic importance of resetting B cell responses, adoptive transfer of purified splenic B cells from ES-62–treated MRL/lpr mice mimicked the protection afforded by the helminth product. Mechanistically, this reflects down-regulation of myeloid differentiation factor 88 expression by B cells and also kidney cells, resulting in inhibition of pathogenic cross-talk among Toll-like receptor–, C3a-, and immune complex–mediated effector mechanisms. Conclusion This study provides the first demonstration of protection against kidney pathology by a parasitic worm–derived immunomodulator in a model of SLE and suggests therapeutic potential for drugs based on the mechanism of action of ES-62.
Collapse
|
32
|
Mangan PR, Su LJ, Jenny V, Tatum AL, Picarillo C, Skala S, Ditto N, Lin Z, Yang X, Cotter PZ, Shuster DJ, Song Y, Borowski V, Thomas RL, Heimrich EM, Devaux B, Das Gupta R, Carvajal I, McIntyre KW, Xie J, Zhao Q, Struthers M, Salter-Cid LM. Dual Inhibition of Interleukin-23 and Interleukin-17 Offers Superior Efficacy in Mouse Models of Autoimmunity. J Pharmacol Exp Ther 2015; 354:152-65. [PMID: 26015463 DOI: 10.1124/jpet.115.224246] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/22/2015] [Indexed: 01/13/2023] Open
Abstract
Therapies targeting either interleukin (IL)-23 or IL-17 have shown promise in treating T helper 17 (Th17)-driven autoimmune diseases. Although IL-23 is a critical driver of IL-17, recognition of nonredundant and independent functions of IL-23 and IL-17 has prompted the notion that dual inhibition of both IL-23 and IL-17 could offer even greater efficacy for treating autoimmune diseases relative to targeting either cytokine alone. To test this hypothesis, we generated selective inhibitors of IL-23 and IL-17 and tested the effect of either treatment alone compared with their combination in vitro and in vivo. In vitro, using a novel culture system of murine Th17 cells and NIH/3T3 fibroblasts, we showed that inhibition of both IL-23 and IL-17 completely suppressed IL-23-dependent IL-22 production from Th17 cells and cooperatively blocked IL-17-dependent IL-6 secretion from the NIH/3T3 cells to levels below either inhibitor alone. In vivo, in the imiquimod induced skin inflammation model, and in the myelin oligodendrocyte glycoprotein peptide-induced experimental autoimmune encephalomyelitis model, we demonstrated that dual inhibition of IL-17 and IL-23 was more efficacious in reducing disease than targeting either cytokine alone. Together, these data support the hypothesis that neutralization of both IL-23 and IL-17 may provide enhanced benefit against Th17 mediated autoimmunity and provide a basis for a therapeutic strategy aimed at dual targeting IL-23 and IL-17.
Collapse
Affiliation(s)
- Paul R Mangan
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Linhui Julie Su
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Victoria Jenny
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Andrea L Tatum
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Caryn Picarillo
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Stacey Skala
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Noah Ditto
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Zheng Lin
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - XiaoXia Yang
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Pete Z Cotter
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - David J Shuster
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Yunling Song
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Virna Borowski
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Rochelle L Thomas
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Elizabeth M Heimrich
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Brigitte Devaux
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Ruchira Das Gupta
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Irvith Carvajal
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Kim W McIntyre
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Jenny Xie
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Qihong Zhao
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Mary Struthers
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| | - Luisa M Salter-Cid
- Discovery Biology, Immunoscience (P.R.M., S.S., X.Y., D.J.S., Y.S., V.B., R.L.T., E.M.H., K.W.M., J.X., Q.Z., M.S., L.M.S.-C.) Selection Technologies (L.J.S., V.J.), Immunogenicity Prediction (C.P.), Pharmacology (P.Z.C., I.C.), Discovery Assays (R.D.G.), Bristol-Myers Squibb Research and Development, Waltham, Massachusetts; Protein Science and Structure, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (Z.L.); Hybridoma Research, Bristol-Myers Squibb Research and Development, Redwood City, California (A.L.T., B.D.); and Wasatch Microfluidics, Salt Lake City, Utah (N.D.)
| |
Collapse
|
33
|
Mari ER, Moore JN, Zhang GX, Rostami A. Mechanisms of immunological tolerance in central nervous system inflammatory demyelination. ACTA ACUST UNITED AC 2015; 6:264-274. [PMID: 26425145 DOI: 10.1111/cen3.12196] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Multiple sclerosis is a complex autoimmune disease of the central nervous system that results in a disruption of the balance between pro-inflammatory and anti-inflammatory signals in the immune system. Given that central nervous system inflammation can be suppressed by various immunological tolerance mechanisms, immune tolerance has become a focus of research in the attempt to induce long-lasting immune suppression of pathogenic T cells. Mechanisms underlying this tolerance induction include induction of regulatory T cell populations, anergy and the induction of tolerogenic antigen-presenting cells. The intravenous administration of encephalitogenic peptides has been shown to suppress experimental autoimmune encephalomyelitis and induce tolerance by promoting the generation of regulatory T cells and inducing apoptosis of pathogenic T cells. Safe and effective methods of inducing long-lasting immune tolerance are essential for the treatment of multiple sclerosis. By exploring tolerogenic mechanisms, new strategies can be devised to strengthen the regulatory, anti-inflammatory cell populations thereby weakening the pathogenic, pro-inflammatory cell populations.
Collapse
Affiliation(s)
- Elisabeth R Mari
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jason N Moore
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
34
|
Schmidt T, Paust H, Krebs CF, Turner J, Kaffke A, Bennstein SB, Koyro T, Peters A, Velden J, Hünemörder S, Haag F, Steinmetz OM, Mittrücker H, Stahl RAK, Panzer U. Function of the Th17/Interleukin‐17A Immune Response in Murine Lupus Nephritis. Arthritis Rheumatol 2015; 67:475-87. [DOI: 10.1002/art.38955] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/06/2014] [Indexed: 01/09/2023]
Affiliation(s)
| | | | | | | | - Anna Kaffke
- Universitätsklinikum Hamburg‐EppendorfHamburgGermany
| | | | - Tobias Koyro
- Universitätsklinikum Hamburg‐EppendorfHamburgGermany
| | - Anett Peters
- Universitätsklinikum Hamburg‐EppendorfHamburgGermany
| | | | | | | | | | | | | | - Ulf Panzer
- Universitätsklinikum Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
35
|
Dallenbach K, Maurer P, Röhn T, Zabel F, Kopf M, Bachmann MF. Protective effect of a germline, IL-17-neutralizing antibody in murine models of autoimmune inflammatory disease. Eur J Immunol 2015; 45:1238-47. [PMID: 25545966 DOI: 10.1002/eji.201445017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/19/2014] [Accepted: 12/22/2014] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies (mAbs) inhibiting cytokines have recently emerged as new drug modalities for the treatment of chronic inflammatory diseases. Interleukin-17 (IL-17) is a T-cell-derived central mediator of autoimmunity. Immunization with Qβ-IL-17, a virus-like particle based vaccine, has been shown to produce autoantibodies in mice and was effective in ameliorating disease symptoms in animal models of autoimmunity. To characterize autoantibodies induced by vaccination at the molecular level, we generated mouse mAbs specific for IL-17 and compared them to germline Ig sequences. The variable regions of a selected hypermutated high-affinity anti-IL-17 antibody differed in only three amino acid residues compared to the likely germline progenitor. An antibody, which was backmutated to germline, maintained a surprisingly high affinity (0.5 nM). The ability of the parental hypermutated antibody and the derived germline antibody to block inflammation was subsequently tested in murine models of multiple sclerosis (experimental autoimmune encephalomyelitis), arthritis (collagen-induced arthritis), and psoriasis (imiquimod-induced skin inflammation). Both antibodies were able to delay disease onset and significantly reduced disease severity. Thus, the mouse genome unexpectedly encodes for antibodies with the ability to functionally neutralize IL-17 in vivo.
Collapse
|
36
|
Sie C, Korn T, Mitsdoerffer M. Th17 cells in central nervous system autoimmunity. Exp Neurol 2014; 262 Pt A:18-27. [DOI: 10.1016/j.expneurol.2014.03.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 03/12/2014] [Accepted: 03/19/2014] [Indexed: 01/08/2023]
|
37
|
Bavoso A, Ostuni A, De Vendel J, Bracalello A, Shcheglova T, Makker S, Tramontano A. Aldehyde modification and alum coadjuvancy enhance anti-TNF-α autovaccination and mitigate arthritis in rat. J Pept Sci 2014; 21:400-7. [PMID: 25424319 DOI: 10.1002/psc.2718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/17/2014] [Accepted: 10/27/2014] [Indexed: 12/21/2022]
Abstract
Experimental vaccination to induce antibodies (Abs) capable of cytokine antagonism shows promise as a novel immunotherapy for chronic inflammatory disease. We prepared a hybrid antigen consisting of residues 141-235 of rat TNF-α fused to the C-terminus of glutathione-S-transferase (GST), chemically modified to incorporate aldehyde residues, for development of an auto-vaccine eliciting anti-rTNF-α Abs. In rat immunization the soluble aldehyde-modified fusion protein did not generate observable Ab responses. By contrast, vaccination with the aldehyde-modified fusion protein adsorbed on alum induced anti-TNF-α autoAbs with high titer and neutralizing activity. Induction of adjuvant arthritis in rats pre-immunized with unmodified fusion protein or a control protein in alum resulted in severe inflammation and joint damage, whereas the disease induced in rats immunized with the aldehyde-bearing fusion protein in alum was markedly attenuated. Similar results were obtained in a collagen-induced rat arthritis model. Anti-collagen II IgG Ab titers did not deviate significantly in groups pre-immunized with modified fusion protein and control protein, suggesting that anti-TNF vaccination did not skew the immune response related to disease induction. This study demonstrates synergy between particulate alum and protein bound carbonyl residues for enhancement of protein immunogenicity. The antigen-specific co-adjuvant system could prove advantageous for breaking tolerance in emerging auto-vaccination therapies targeting inflammatory cytokines as well as for enhancing a broader category of subunit vaccines. Aldehyde adduction introduces a minimal modification which, together with the established use of alum as a safe adjuvant for human use, could be favorable for further vaccine development.
Collapse
Affiliation(s)
- Alfonso Bavoso
- Department of Sciences, University of Basilicata, 85100, Potenza, Italy
| | | | | | | | | | | | | |
Collapse
|
38
|
Song L, Weng D, Dai W, Tang W, Chen S, Li C, Chen Y, Liu F, Chen J. Th17 can regulate silica-induced lung inflammation through an IL-1β-dependent mechanism. J Cell Mol Med 2014; 18:1773-84. [PMID: 25091058 PMCID: PMC4196653 DOI: 10.1111/jcmm.12341] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 05/14/2014] [Indexed: 12/16/2022] Open
Abstract
Silicosis is an occupational lung disease caused by the inhalation of silica dust and characterized by lung inflammation and fibrosis. Interleukin (IL)-1β is induced by silica and functions as the key pro-inflammatory cytokine in this process. The Th17 response, which is induced by IL-1β, has been reported very important in chronic human lung inflammatory diseases. To elucidate the underlying mechanisms of IL-1β and IL-17 in silicosis, we used anakinra and an anti-IL-17 monoclonal antibody (mAb) to block the receptor of IL-1β (IL-RI) and IL-17, respectively, in a mouse model of silicosis. We observed increased IL-1β expression and an enhanced Th17 response after silica instillation. Treatment with an IL-1 type I receptor (IL-1RI) antagonist anakinra substantially decreased silica-induced lung inflammation and the Th17 response. Lung inflammation and the accumulation of inflammatory cells were attenuated in the IL-17-neutralized silicosis group. IL-17 may promote lung inflammation by modulating the differentiation of Th1 and regulatory T cells (Tregs) and by regulating the production of IL-22 and IL-1β during the lung inflammation of silicosis. Silica may induce IL-1β production from alveolar macrophages and promote inflammation by initiating a Th17 response via an IL-1β/IL-1RI-dependent mechanism. The Th17 response could induce lung inflammation during the pathogenesis of silicosis by regulating the homoeostasis of the Th immune responses and affecting the production of IL-22 and IL-1β. This study describes a potentially important inflammatory mechanism of silicosis that may bring about novel therapies for this inflammatory and fibrotic disease.
Collapse
Affiliation(s)
- Laiyu Song
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
- Department of Clinical Immunology, Dalian Medical UniversityDalian, China
| | - Dong Weng
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Wujing Dai
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Wen Tang
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Shi Chen
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Chao Li
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Ying Chen
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Fangwei Liu
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Jie Chen
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| |
Collapse
|
39
|
Treatment with a hybrid between the synapsin ABC domains and the B subunit of E. coli heat-labile toxin reduces frequency of proinflammatory cells and cytokines in the central nervous system of rats with EAE. Neuroscience 2014; 277:217-28. [DOI: 10.1016/j.neuroscience.2014.06.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/27/2014] [Accepted: 06/28/2014] [Indexed: 01/07/2023]
|
40
|
Avau A, Mitera T, Put S, Put K, Brisse E, Filtjens J, Uyttenhove C, Van Snick J, Liston A, Leclercq G, Billiau AD, Wouters CH, Matthys P. Systemic juvenile idiopathic arthritis-like syndrome in mice following stimulation of the immune system with Freund's complete adjuvant: regulation by interferon-γ. Arthritis Rheumatol 2014; 66:1340-51. [PMID: 24470407 DOI: 10.1002/art.38359] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 01/09/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Systemic juvenile idiopathic arthritis (JIA) is unique among the rheumatic diseases of childhood, given its distinctive systemic inflammatory character. Inappropriate control of innate immune responses following an initially harmless trigger is thought to account for the excessive inflammatory reaction. The aim of this study was to generate a similar systemic inflammatory syndrome in mice by injecting a relatively innocuous, yet persistent, immune system trigger: Freund's complete adjuvant (CFA), containing heat-killed mycobacteria. METHODS Given the central role of interferon-γ (IFNγ) in immune regulation, we challenged wild-type (WT) and IFNγ-knockout (KO) BALB/c mice with CFA, and analyzed their clinical symptoms and biologic characteristics. The production of cytokines and the effects of anticytokine antibodies were investigated. RESULTS In WT mice, CFA injection resulted in splenomegaly, lymphadenopathy, neutrophilia, thrombocytosis, and increased cytokine expression. In the absence of IFNγ, these symptoms were more pronounced and were accompanied by weight loss, arthritis, anemia, hemophagocytosis, abundance of immature blood cells, and increased levels of interleukin-6 (IL-6), all of which are reminiscent of the symptoms of systemic JIA. CFA-challenged IFNγ-KO mice showed increased expression of IL-17 by CD4+ T cells and by innate γ/δ T cells. Inflammatory and hematologic changes were prevented by treatment with anti-IL-12/IL-23p40 and anti-IL-17 antibodies. CONCLUSION Immune stimulation of IFNγ-KO mice with CFA produces a systemic inflammatory syndrome reflecting the clinical, biologic, and histopathologic picture of systemic JIA. The protective function of IFNγ in preventing anemia and overall systemic inflammation is a striking observation. The finding that both adaptive and innate T cells are important sources of IL-17 may be of relevance in the pathogenesis of systemic JIA.
Collapse
|
41
|
Targeting IL-1β and IL-17A driven inflammation during influenza-induced exacerbations of chronic lung inflammation. PLoS One 2014; 9:e98440. [PMID: 24918427 PMCID: PMC4053370 DOI: 10.1371/journal.pone.0098440] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 05/04/2014] [Indexed: 12/05/2022] Open
Abstract
For patients with chronic lung diseases, such as chronic obstructive pulmonary disease (COPD), exacerbations are life-threatening events causing acute respiratory distress that can even lead to hospitalization and death. Although a great deal of effort has been put into research of exacerbations and potential treatment options, the exact underlying mechanisms are yet to be deciphered and no therapy that effectively targets the excessive inflammation is available. In this study, we report that interleukin-1β (IL-1β) and interleukin-17A (IL-17A) are key mediators of neutrophilic inflammation in influenza-induced exacerbations of chronic lung inflammation. Using a mouse model of disease, our data shows a role for IL-1β in mediating lung dysfunction, and in driving neutrophilic inflammation during the whole phase of viral infection. We further report a role for IL-17A as a mediator of IL-1β induced neutrophilia at early time points during influenza-induced exacerbations. Blocking of IL-17A or IL-1 resulted in a significant abrogation of neutrophil recruitment to the airways in the initial phase of infection or at the peak of viral replication, respectively. Therefore, IL-17A and IL-1β are potential targets for therapeutic treatment of viral exacerbations of chronic lung inflammation
Collapse
|
42
|
Marillier RG, Uyttenhove C, Goriely S, Marbaix E, Van Snick J. IL-27p28 is essential for parent-to-F1 acute graft-versus-host disease. Eur J Immunol 2014; 44:2064-73. [PMID: 24700119 DOI: 10.1002/eji.201444491] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 02/14/2014] [Accepted: 03/26/2014] [Indexed: 12/29/2022]
Abstract
Acute graft versus host disease (aGVHD) remains a life-threatening complication of bone marrow transplantation. Here we show that IL-27, a member of the IL-12 cytokine family, plays an essential role in a parent-to-F1 murine aGVHD model, using B6 mice as parents and B6D2 mice as F1 recipients. IL-27 is transiently detectable in the serum of B6D2 recipients of B6 spleen cells, with a peak at day 10. Treatment with anti-IL-27p28 mAb MM27.7B1 (αp28Ab), at the time of and six days after B6 cell transfer, blocked GVHD. Protection was associated with host cell survival and undiminished engraftment of donor cells, lack of host B-cell depletion, increased Th2-type immunoglobulin production, a decrease in serum IFN-γ, a drop in anti-H-2D(d) cytotoxic T lymphocyte activity and an increase in Foxp3(+) T cells. We therefore conclude that IL-27 plays a critical role in the parent-to-F1 model of aGVHD and that blocking IL-27 could have therapeutic relevance.
Collapse
Affiliation(s)
- Reece G Marillier
- Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium; de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | | | | | | |
Collapse
|
43
|
Reduction of IL-17A might suppress the Th1 response and promote the Th2 response by boosting the function of Treg cells during silica-induced inflammatory response in vitro. Mediators Inflamm 2014; 2014:570894. [PMID: 24692850 PMCID: PMC3945471 DOI: 10.1155/2014/570894] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 12/30/2013] [Accepted: 01/03/2014] [Indexed: 02/07/2023] Open
Abstract
Silica inhalation can induce chronic lung inflammation and fibrosis. Upon silica stimulation, activated macrophages trigger the T-lymphocyte which can differentiate into many different types of Th cells, including the recently discovered Th17 cells. IL-17A, the typical Th17 cytokine, is reported in some inflammatory diseases. However, the role of IL-17A in silica-induced inflammatory response is still not clear. The regulatory mechanism of silica-induced Th17 response also needs to be investigated. So we established a mice primary cell coculture system (macrophage and lymphocyte) to investigate the role of IL-17A in silica-induced inflammatory response in vitro, by using anti-IL-17A mAb and IL-1Ra. Both anti-IL-17A mAb and IL-1Ra decreased the level of IL-17A and increased the function of Treg cells. The Th1 response was suppressed and the Th2 response was promoted by the addition of anti-IL-17A mAb or IL-1Ra. IL-1Ra treatment decreased the level of IL-6, whereas the levels of IL-23 and ROR- γ t were increased. Our study demonstrated that IL-17A reduction altered the pattern of silica-induced Th responses by boosting the function of Treg cells in vitro. Blocking the function of IL-1 signal pathway could suppress the level of IL-17A, which played the major role in modulating silica-induced Th responses in vitro.
Collapse
|
44
|
Clinical consequences of targeting IL-17 and TH17 in autoimmune and allergic disorders. Curr Allergy Asthma Rep 2014; 13:587-95. [PMID: 23760974 DOI: 10.1007/s11882-013-0361-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The TH17 lineage of T cells and its canonical cytokine IL-17 have been the focus of many recent studies in autoimmune, allergic, and infectious disease. In this review, we will briefly discuss the current knowledge about the role of these cells and IL-17 in a spectrum of disorders. It is clear that IL-17 plays pathogenic roles in certain conditions while the same pathway is critically important to immunity in others. Targeting of TH17 cells or IL-17 therapeutically may impart many benefits, but this approach is not without potentially serious implications regarding host defense. These issues will be discussed herein as we evaluate pharmacological approaches targeting this pathway that are just beginning to be fully tested in human disease.
Collapse
|
45
|
Noack M, Miossec P. Th17 and regulatory T cell balance in autoimmune and inflammatory diseases. Autoimmun Rev 2014; 13:668-77. [PMID: 24418308 DOI: 10.1016/j.autrev.2013.12.004] [Citation(s) in RCA: 647] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 12/31/2013] [Indexed: 01/01/2023]
Abstract
This review focuses on the biology of T helper 17 (Th17) and regulatory T (Treg) cells and their role in inflammatory diseases, such as rheumatoid arthritis. Th17 cells represent a pro-inflammatory subset whereas Treg cells have an antagonist effect. Their developmental pathways are reciprocally interconnected and there is an important plasticity between Th17 and Treg cells. These features implicate that the Th17/Treg balance plays a major role in the development and the disease outcomes of animal model and human autoimmune/inflammatory diseases. During these diseases, this balance is disturbed and this promotes the maintenance of inflammation. Targeting the Th17/Treg imbalance can be performed at different levels such as inhibition of pro-inflammatory cytokines and their receptors, of pathogenic cells or their specific signaling pathways. Conversely, direct effects include administration or induction of protective cells, or stimulation of their specific pathways. Several clinical trials are underway and some positive results have been obtained.
Collapse
Affiliation(s)
- Mélissa Noack
- Immunogenomics and Inflammation Research Unit EA 4130, University of Lyon 1, Department of Immunology and Rheumatology, Hospital Edouard Herriot, 5 Place d'Arsonval, 69437 Lyon Cedex 03, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit EA 4130, University of Lyon 1, Department of Immunology and Rheumatology, Hospital Edouard Herriot, 5 Place d'Arsonval, 69437 Lyon Cedex 03, France.
| |
Collapse
|
46
|
Koussoroplis SJ, Heywood S, Uyttenhove C, Barilly C, Van Snick J, Vanbever R. Production, purification and biological characterization of mono-PEGylated anti-IL-17A antibody fragments. Int J Pharm 2013; 454:107-15. [DOI: 10.1016/j.ijpharm.2013.06.077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/19/2013] [Accepted: 06/27/2013] [Indexed: 11/26/2022]
|
47
|
Fogli LK, Sundrud MS, Goel S, Bajwa S, Jensen K, Derudder E, Sun A, Coffre M, Uyttenhove C, Van Snick J, Schmidt-Supprian M, Rao A, Grunig G, Durbin J, Casola S, Casola SS, Rajewsky K, Koralov SB. T cell-derived IL-17 mediates epithelial changes in the airway and drives pulmonary neutrophilia. THE JOURNAL OF IMMUNOLOGY 2013; 191:3100-11. [PMID: 23966625 DOI: 10.4049/jimmunol.1301360] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Th17 cells are a proinflammatory subset of effector T cells that have been implicated in the pathogenesis of asthma. Their production of the cytokine IL-17 is known to induce local recruitment of neutrophils, but the direct impact of IL-17 on the lung epithelium is poorly understood. In this study, we describe a novel mouse model of spontaneous IL-17-driven lung inflammation that exhibits many similarities to asthma in humans. We have found that STAT3 hyperactivity in T lymphocytes causes an expansion of Th17 cells, which home preferentially to the lungs. IL-17 secretion then leads to neutrophil infiltration and lung epithelial changes, in turn leading to a chronic inflammatory state with increased mucus production and decreased lung function. We used this model to investigate the effects of IL-17 activity on airway epithelium and identified CXCL5 and MIP-2 as important factors in neutrophil recruitment. The neutralization of IL-17 greatly reduces pulmonary neutrophilia, underscoring a key role for IL-17 in promoting chronic airway inflammation. These findings emphasize the role of IL-17 in mediating neutrophil-driven pulmonary inflammation and highlight a new mouse model that may be used for the development of novel therapies targeting Th17 cells in asthma and other chronic pulmonary diseases.
Collapse
Affiliation(s)
- Laura K Fogli
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Guan Q, Weiss CR, Qing G, Ma Y, Peng Z. An IL-17 peptide-based and virus-like particle vaccine enhances the bioactivity of IL-17 in vitro and in vivo. Immunotherapy 2013; 4:1799-807. [PMID: 23240747 DOI: 10.2217/imt.12.129] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIMS To develop an IL-17 peptide-based virus-like particle vaccine that elicits autoantibodies to IL-17 and to evaluate the effects of the vaccine in mice with experimental colitis. MATERIALS & METHODS Recombinant IL-17 vaccines were constructed by inserting selected peptides derived from mouse IL-17 into the carrier protein, hepatitis B core antigen, using molecular engineering methods. To evaluate the in vivo effects of the vaccine, mice with 2,4,6-trinitrobenzene sulfonic acid-induced chronic colitis were injected three times with the vaccine, carrier or saline after the second delivery of 2,4,6-trinitrobenzene sulfonic acid. Colon inflammation and fibrosis were evaluated by histological examination. Serum IL-17-specific IgG and colon-tissue cytokine levels were measured by ELISA. In vitro inhibition tests of sera from vaccine-immunized mice were performed using IL-17-induced IL-6 production by NIH 3T3 cells and IL-17-induced TNF production by macrophages. RESULTS Immunization with the vaccine without the use of adjuvants induced high-titered and long-lasting antibodies to IL-17. Unexpectedly, vaccinated mice exhibited increases in colon inflammation, collagen deposition, levels of TNF and IL-17 cytokines compared with carrier and saline groups. Furthermore, in vitro study revealed that serum IL-17-specific IgG from vaccine-immunized mice significantly enhanced IL-17-induced IL-6 production and IL-17-induced TNF production dose-dependently. CONCLUSION The IL-17 peptide-based vaccine enhances the bioactivity of IL-17 in vitro and in vivo, providing a potential immunotherapy for treatment of diseases associated with insufficient IL-17 production, such as hyper-IgE syndrome.
Collapse
Affiliation(s)
- Qingdong Guan
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | |
Collapse
|
49
|
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
|
50
|
Role of Th17 cells in the pathogenesis of CNS inflammatory demyelination. J Neurol Sci 2013; 333:76-87. [PMID: 23578791 DOI: 10.1016/j.jns.2013.03.002] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 02/19/2013] [Accepted: 03/04/2013] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS). The etiology of MS is not well understood, but it is believed that myelin-specific CD4(+) T cells play a central role in initiating and orchestrating CNS inflammation. In this scenario, CD4(+) T cells, activated in the periphery, infiltrate the CNS, where, by secreting cytokines and chemokines, they start an inflammatory cascade. Given the central role of CD4(+) T cells in CNS autoimmunity, they have been studied extensively, principally by using experimental autoimmune encephalomyelitis (EAE), an animal model of MS. In the late 1980s, CD4(+) T cells, based on their cytokine production, were divided into two helper lineages, Th1 and Th2 cells. It was postulated that Th1 cells, which produce IFN-γ, mediate inflammation of the CNS in MS/EAE, while Th2 cells, which produce IL-4, have a beneficial effect in disease, because of their antagonistic effect on Th1 cells. The Th1/Th2 paradigm remained the prevailing view of MS/EAE pathogenesis until 2005, when a new lineage, Th17, was discovered. In a relatively short period of time it became apparent that Th17 cells, named after their hallmark cytokine, IL-17A, play a crucial role in many inflammatory diseases, including EAE, and likely in MS as well. The Th17 paradigm developed rapidly, initiating the debate of whether Th1 cells contribute to EAE/MS pathogenesis at all, or if they might even have a protective role due to their antagonistic effects on Th17 cells. Numerous findings support the view that Th17 cells play an essential role in autoimmune CNS inflammation, perhaps mainly in the initial phases of disease. Th1 cells likely contribute to pathogenesis, with their role possibly more pronounced later in disease. Hence, the current view on the role of Th cells in MS/EAE pathogenesis can be called the Th17/Th1 paradigm. It is certain that Th17 cells will continue to be the focus of intense investigation aimed at elucidating the pathogenesis of CNS autoimmunity.
Collapse
|