1
|
Xu WD, Wang DC, Zhao M, Huang AF. An updated advancement of bifunctional IL-27 in inflammatory autoimmune diseases. Front Immunol 2024; 15:1366377. [PMID: 38566992 PMCID: PMC10985211 DOI: 10.3389/fimmu.2024.1366377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Interleukin-27 (IL-27) is a member of the IL-12 family. The gene encoding IL-27 is located at chromosome 16p11. IL-27 is considered as a heterodimeric cytokine, which consists of Epstein-Barr virus (EBV)-induced gene 3 (Ebi3) and IL-27p28. Based on the function of IL-27, it binds to receptor IL-27rα or gp130 and then regulates downstream cascade. To date, findings show that the expression of IL-27 is abnormal in different inflammatory autoimmune diseases (including systemic lupus erythematosus, rheumatoid arthritis, Sjogren syndrome, Behcet's disease, inflammatory bowel disease, multiple sclerosis, systemic sclerosis, type 1 diabetes, Vogt-Koyanagi-Harada, and ankylosing spondylitis). Moreover, in vivo and in vitro studies demonstrated that IL-27 is significantly in3volved in the development of these diseases by regulating innate and adaptive immune responses, playing either an anti-inflammatory or a pro-inflammatory role. In this review, we comprehensively summarized information about IL-27 and autoimmunity based on available evidence. It is hoped that targeting IL-27 will hold great promise in the treatment of inflammatory autoimmune disorders in the future.
Collapse
Affiliation(s)
- Wang-Dong Xu
- Department of Evidence-Based Medicine, School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Da-Cheng Wang
- Department of Evidence-Based Medicine, School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Ming Zhao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
2
|
McCormick ET, Draganski A, Chalmers S, Zahn J, Garcia S, Nussbaum D, Friedman A, Putterman C, Friedman J. Nano-encapsulated anandamide reduces inflammatory cytokines in vitro and lesion severity in a murine model of cutaneous lupus erythematosus. Exp Dermatol 2023; 32:2072-2083. [PMID: 37726950 DOI: 10.1111/exd.14935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023]
Abstract
Cutaneous lupus erythematosus (CLE) is a heterogeneous autoimmune skin disease which occurs independently and in conjunction with systemic lupus erythematosus. Drug development for CLE is severely lacking. Anandamide (AEA) is a primary endocannabinoid which exhibits immunomodulatory effects through mixed cannabinoid receptor agonism. We evaluated AEA as topical treatment for CLE and assessed benefits of nanoparticle encapsulation (AEA-NP) on cutaneous drug penetration, delivery and biological activity. Compared to untreated controls, AEA-NP decreased IL-6 and MCP-1 in UVB-stimulated keratinocytes (p < 0.05) in vitro. In BALB/c mice, AEA-NP displayed improved cutaneous penetration, extended release and persistence of AEA in the follicular unit extending to the base after 24 h. Utilizing the MRL-lpr lupus murine model, twice weekly treatment of lesions with topical AEA-NP for 10 weeks led to decreased clinical and histologic lesion scores compared to unencapsulated AEA and untreated controls (p < 0.05). Prophylactic application of AEA-NP to commonly involved areas on MRL-lpr mice similarly resulted in decreased clinical and histologic scores when compared to controls (p < 0.05), and reduced C3 and IBA-1 in lesional tissue (p < 0.05). The demonstrated clinical and immunomodulatory effects of treatment with AEA support its potential as therapy for CLE. This work also suggests that encapsulation of AEA improves penetration and treatment efficacy. Future studies will be conducted to assess full therapeutic potential.
Collapse
Affiliation(s)
- Erika T McCormick
- George Washington University Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | | | - Samantha Chalmers
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, Bronx, USA
| | - Joseph Zahn
- George Washington University Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Sayra Garcia
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, Bronx, USA
| | - Dillon Nussbaum
- George Washington University Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Adam Friedman
- George Washington University Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Chaim Putterman
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, Bronx, USA
- Azrieli Faculty of Medicine of Bar-Ilan University, Zefat, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
| | - Joel Friedman
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, Bronx, USA
| |
Collapse
|
3
|
Shahi A, Afzali S, Salehi S, Aslani S, Mahmoudi M, Jamshidi A, Amirzargar A. IL-27 and autoimmune rheumatologic diseases: The good, the bad, and the ugly. Int Immunopharmacol 2020; 84:106538. [PMID: 32361567 DOI: 10.1016/j.intimp.2020.106538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/09/2020] [Accepted: 04/20/2020] [Indexed: 12/24/2022]
Abstract
The footprint of cytokines is evident in almost every biological process, such as development, as well as the pathogenesis of the different diseases, immune responses to pathogens, etc. These small proteins are categorized into different functional classes; for instance, they can play a pro-inflammatory or anti-inflammatory role in different situations, or they can confer a polarization to the immune system. Interleukin (IL)-27 is a member of the IL-12 family. Antigen-presenting cells are the primary source of IL-27 production, which exerts its effects by bindings to the IL-27 receptor expressed on the surface of target cells. Interaction of IL-27 and IL-27 receptor leads to activation of the JAK-STAT and p38 MAPK signaling pathways. Most studies focused on the inflammatory effects of this cytokine, but gradually anti-inflammatory effects were also revealed for this cytokine, which changed the traditional perception of the function of this cytokine. The functionality of IL-27 in the pathogenesis of rheumatic diseases has been attributed to a double-blade sword. Hence, novel therapeutic approaches have been devised targeting IL-12 family that has been accompanied with promising results. In this review, we focused on the inflammatory and anti-inflammatory properties of IL-27 in different autoimmune rheumatologic diseases and its plausible therapeutic potentials.
Collapse
Affiliation(s)
- Abbas Shahi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Afzali
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeedeh Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Aslani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran; Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliakbar Amirzargar
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Abstract
The involvement of the interleukin (IL)-17 axis in many inflammatory and autoimmune diseases is now well established, and this has led to the development of successful targeted therapies. Its role in systemic lupus erythematosus (SLE) is less described, since SLE is characterized by the impairment of many other immune actors. However, results from animal models and patients strongly suggest that IL-17 and its producing cells are involved in SLE pathogenesis. Circulating levels of IL-17 are increased in lupus, and tissue staining shows the presence of IL-17-producing cells in organ lesions. Through different mechanisms, the IL-17 axis promotes autoantibody production, immune complex deposition, complement activation and then tissue damage. There are also many interactions with other immune and non-immune actors, which account for the broad spectrum of clinical manifestations and disease heterogeneity. SLE treatment faces challenges with many disappointing trials and persistent unmet needs. The identification of subsets of SLE patients with an IL-17-driven disease now constitutes the key priority before starting trials. More preclinical studies are needed to improve the selection of the right patients able to respond and tolerate the many inhibitors that are already available.
Collapse
Affiliation(s)
- M Robert
- Department of Clinical Immunology and Rheumatology, Immunogenomics and Inflammation Research Unit EA 4130, University of Lyon 1, Hôpital Edouard Herriot, Lyon, France
| | - P Miossec
- Department of Clinical Immunology and Rheumatology, Immunogenomics and Inflammation Research Unit EA 4130, University of Lyon 1, Hôpital Edouard Herriot, Lyon, France
| |
Collapse
|
5
|
Zhang YP, Wu J, Han YF, Shi ZR, Wang L. Pathogenesis of cutaneous lupus erythema associated with and without systemic lupus erythema. Autoimmun Rev 2017; 16:735-742. [PMID: 28483542 DOI: 10.1016/j.autrev.2017.05.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 04/14/2017] [Indexed: 12/11/2022]
Abstract
Cutaneous lupus erythematosus (CLE) can be an individual disease only involving skin, or presents as part of the manifestations of SLE. A small proportion of CLE may progress into SLE, however, the underlying pathogenic mediators remain elusive. By only including researches that clearly described if the subtypes of CLE presented by enrolled subjects was associated with or without SLE, we provided an overview of antibodies, inflammatory cells and inflammatory molecular mediators identified in blood and skin that were possibly involved in lupus skin damages. IgG autoantibodies are crucial for the development of CLE associated with SLE, but the circulating inflammatory cells and molecular mediators require further studies to provide definitive proof for their association with skin damages. Discoid lupus erythematosus (DLE) is the most common subtype of CLE. For DLE without associated with SLE (CDLE), it is lack of evidences if autoantibodies and circulating inflammatory cells are involved in the pathogenesis or not, but is clear that the cutaneous inflammatory infiltrates are dominated by Th1, but not Th17 cells in contrast to the various complex profile in SLE. As the major target cells in skin, keratinocytes may participate the pathophysiological process by increase cell apoptosis and the production of proinflammatory cytokines in SLE and CDLE. Insights into the similarities and differences of the pathogenesis of CLE and CLE associated with SLE will also improve our therapeutic strategies for CLE that is currently adopted from SLE, and prevent the progression of CLE to SLE by providing interventions within an appropriate window of disease development.
Collapse
Affiliation(s)
- Yu-Ping Zhang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jian Wu
- Guangdong Provincial Institute of Geriatrics, Guangdong General Hospital, Guangdong Academy of Medical Science, 510080, China
| | - Yan-Fang Han
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhen-Rui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Liangchun Wang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
6
|
Vijayan D, Mohd Redzwan N, Avery DT, Wirasinha RC, Brink R, Walters G, Adelstein S, Kobayashi M, Gray P, Elliott M, Wong M, King C, Vinuesa CG, Ghilardi N, Ma CS, Tangye SG, Batten M. IL-27 Directly Enhances Germinal Center B Cell Activity and Potentiates Lupus in Sanroque Mice. THE JOURNAL OF IMMUNOLOGY 2016; 197:3008-3017. [PMID: 27619997 DOI: 10.4049/jimmunol.1600652] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/09/2016] [Indexed: 11/19/2022]
Abstract
Germinal centers (GC) give rise to high-affinity and long-lived Abs and are critical in immunity and autoimmunity. IL-27 supports GCs by promoting survival and function of T follicular helper cells. We demonstrate that IL-27 also directly enhances GC B cell function. Exposure of naive human B cells to rIL-27 during in vitro activation enhanced their differentiation into CD20+CD38+CD27lowCD95+CD10+ cells, consistent with the surface marker phenotype of GC B cells. This effect was inhibited by loss-of-function mutations in STAT1 but not STAT3 To extend these findings, we studied the in vivo effects of IL-27 signals to B cells in the GC-driven Roquinsan/san lupus mouse model. Il27ra-/-Roquinsan/san mice exhibited significantly reduced GCs, IgG2a(c)+ autoantibodies, and nephritis. Mixed bone marrow chimeras confirmed that IL-27 acts through B cell- and CD4+ T cell-intrinsic mechanisms to support GCs and alter the production of pathogenic Ig isotypes. To our knowledge, our data provide the first evidence that IL-27 signals directly to B cells promote GCs and support the role of IL-27 in lupus.
Collapse
Affiliation(s)
- Dipti Vijayan
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Norhanani Mohd Redzwan
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Danielle T Avery
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Rushika C Wirasinha
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Giles Walters
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory 2601, Australia.,Department of Renal Medicine, Canberra Hospital, Canberra, Australian Capital Territory 2605, Australia.,Australian National University Medical School, Canberra, Australian Capital Territory 2601, Australia
| | - Stephen Adelstein
- Clinical Immunology, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
| | - Masao Kobayashi
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima 734-8553, Japan
| | - Paul Gray
- University of New South Wales School of Women's and Children's Health, Sydney, New South Wales 2031, Australia
| | - Michael Elliott
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia.,Chris O'Brien Lifehouse Cancer Centre, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
| | - Melanie Wong
- Children's Hospital at Westmead, Westmead, New South Wales 2145, Australia; and
| | - Cecile King
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Carola G Vinuesa
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Nico Ghilardi
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Cindy S Ma
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Stuart G Tangye
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Marcel Batten
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia; .,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
7
|
d'Almeida SM, Kauffenstein G, Roy C, Basset L, Papargyris L, Henrion D, Catros V, Ifrah N, Descamps P, Croue A, Jeannin P, Grégoire M, Delneste Y, Tabiasco J. The ecto-ATPDase CD39 is involved in the acquisition of the immunoregulatory phenotype by M-CSF-macrophages and ovarian cancer tumor-associated macrophages: Regulatory role of IL-27. Oncoimmunology 2016; 5:e1178025. [PMID: 27622030 DOI: 10.1080/2162402x.2016.1178025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 12/18/2022] Open
Abstract
Tumor-associated macrophages (TAM) are immunosuppressive cells that can massively accumulate in the tumor microenvironment. In patients with ovarian cancer, their density is correlated with poor prognosis. Targeting mediators that control the generation or the differentiation of immunoregulatory macrophages represents a therapeutic challenge to overcome tumor-associated immunosuppression. The ectonucleotidase CD39 hydrolyzes ATP into extracellular adenosine that exhibits potent immunosuppressive properties when signaling through the A2A adenosine receptor. We report here that CD14(+) CD163(+) TAM isolated from ovarian cancer patients and macrophages generated in vitro with M-CSF, express high levels of the membrane ectonucleotidase CD39 compared to classically activated macrophages. The CD39 inhibitor POM-1 and adenosine deaminase (ADA) diminished some of the immunosuppressive functions of CD14(high) CD163(high) CD39(high) macrophages, such as IL-10 secretion. We identified the cytokine IL-27, secreted by tumor-infiltrating neutrophils, located close to infiltrating CD163(+) macrophages, as a major rheostat of CD39 expression and consequently, on the acquisition of immunoregulatory properties by macrophages. Accordingly, the depletion of IL-27 downregulated CD39 and PD-L1 expression as well as IL-10 secretion by M-CSF-macrophages. Collectively, these data suggest that CD39, drived by IL-27 and CD115 ligands in ovarian cancer, maintains the immunosuppressive phenotype of TAM. This work brings new information on the acquisition of immunosuppressive properties by tumor-infiltrating macrophages.
Collapse
Affiliation(s)
- Sènan M d'Almeida
- CRCNA, INSERM, CNRS, Université de Nantes, Université d'Angers, Angers, France; LabEx ImmunoGraftOnco, Angers, France; Equipe labellisée Ligue contre le Cancer, Angers, France
| | | | - Charlotte Roy
- BNMI, INSERM, CNRS, Université d'Angers , Angers, France
| | - Laetitia Basset
- CRCNA, INSERM, CNRS, Université de Nantes, Université d'Angers, Angers, France; LabEx ImmunoGraftOnco, Angers, France; Equipe labellisée Ligue contre le Cancer, Angers, France
| | - Loukas Papargyris
- CRCNA, INSERM, CNRS, Université de Nantes, Université d'Angers, Angers, France; LabEx ImmunoGraftOnco, Angers, France; Equipe labellisée Ligue contre le Cancer, Angers, France
| | - Daniel Henrion
- BNMI, INSERM, CNRS, Université d'Angers , Angers, France
| | - Véronique Catros
- INSERM, Université de Rennes 1, CRB santé de Rennes , Rennes, France
| | - Norbert Ifrah
- CRCNA, INSERM, CNRS, Université de Nantes, Université d'Angers, Angers, France; LabEx ImmunoGraftOnco, Angers, France; Equipe labellisée Ligue contre le Cancer, Angers, France; Services des maladies du sang, CHU Angers, Angers, France
| | | | - Anne Croue
- Laboratoire de Pathologie Cellulaire et Tissulaire, CHU Angers , Angers, France
| | - Pascale Jeannin
- CRCNA, INSERM, CNRS, Université de Nantes, Université d'Angers, Angers, France; LabEx ImmunoGraftOnco, Angers, France; Equipe labellisée Ligue contre le Cancer, Angers, France; Laboratoire d'Immunologie et d'Allergologie, CHU Angers, Angers, France
| | - Marc Grégoire
- LabEx ImmunoGraftOnco, Angers, France; CRCNA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Yves Delneste
- CRCNA, INSERM, CNRS, Université de Nantes, Université d'Angers, Angers, France; LabEx ImmunoGraftOnco, Angers, France; Equipe labellisée Ligue contre le Cancer, Angers, France; Laboratoire d'Immunologie et d'Allergologie, CHU Angers, Angers, France
| | - Julie Tabiasco
- CRCNA, INSERM, CNRS, Université de Nantes, Université d'Angers, Angers, France; LabEx ImmunoGraftOnco, Angers, France; Equipe labellisée Ligue contre le Cancer, Angers, France
| |
Collapse
|
8
|
Zhou LW, Ma N, Li Z, Feng BS. Role of interleukin-27 in inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2016; 24:549-557. [DOI: 10.11569/wcjd.v24.i4.549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD), which is characterized by chronic or recurrent relapsing gastrointestinal inflammation, includes ulcerative colitis (UC) and Crohn's disease (CD). The pathogenesis of IBD remains obscure, however, abnormal immune responses are regarded as the major component of IBD pathogenesis. Interleukin-27 (IL-27) is a new member of the IL-12 family, and it is produced by activated antigen-presenting cells and plays an important role in the differentiation and function of different T cell subsets. IL-27 has various immunoregulatory functions and is implicated in the pathogenesis of many infectious and autoimmune diseases. Recent studies have showed that IL-27 is strongly associated with the genesis and development of IBD. Here we provide an overview of the role of IL-27 in the pathogenesis of IBD.
Collapse
|
9
|
Meka RR, Venkatesha SH, Dudics S, Acharya B, Moudgil KD. IL-27-induced modulation of autoimmunity and its therapeutic potential. Autoimmun Rev 2015; 14:1131-1141. [PMID: 26253381 PMCID: PMC4628569 DOI: 10.1016/j.autrev.2015.08.001] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/01/2015] [Indexed: 11/26/2022]
Abstract
Interleukin-27 (IL-27) is a new member of the IL-12 family. It is produced by activated antigen-presenting cells and plays an important role in the regulation of CD4+ T cell differentiation and immune response. IL-27 activates multiple signaling cascades, including the JAK-STAT and p38 MAPK pathways. Several studies have revealed that IL-27 promotes the differentiation of Th1 and Tr1, but inhibits Th2, Th17, and Treg cells. However, a few studies have shown an opposite effect on certain T cell subsets, such as Treg. IL-27 displays both pro- and anti- inflammatory activities in different autoimmune diseases. Here, we have discussed the role of IL-27 in rheumatoid arthritis, multiple sclerosis, colitis, lupus, psoriasis, type 1 diabetes, and uveitis. Most of this information is derived from experimental models of these autoimmune diseases. The mechanistic basis of the dual role of IL-27 in inflammation and autoimmunity is still not fully defined. In general, the pro-/anti-inflammatory activity of IL-27 is influenced by the underlying immune effector pathways, the phase of the disease, the presence or absence of counter-regulatory cytokines/T cell subsets, and the tissue/cell type under study. Despite a spectrum of outcomes in various autoimmune diseases, mostly anti-inflammatory and immunomodulatory effects of IL-27 have been observed in this category of diseases. Accordingly, IL-27 represents a novel, promising target/agent for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Rakeshchandra R. Meka
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Shivaprasad H. Venkatesha
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Steven Dudics
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Bodhraj Acharya
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Kamal D. Moudgil
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
- Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
10
|
Choi EW, Shin IS, Song JW, Yun TW, Yang J, Choi KS, Seong JK. Transplantation of Adipose Tissue-Derived Mesenchymal Stem Cells Prevents the Development of Lupus Dermatitis. Stem Cells Dev 2015; 24:2041-51. [PMID: 25941899 DOI: 10.1089/scd.2015.0021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
MRL/lpr mice spontaneously develop high titers of anti-dsDNA antibodies and symptoms such as glomerular nephritis and organ weight gain. They also develop spontaneous skin inflammation similar to the cutaneous lesions common in human lupus erythematosus. This study aimed to compare the effects of long-term serial administration of human adipose tissue-derived mesenchymal stem cells (ASCs), CTLA4Ig-overexpressing ASCs, and cyclophosphamide treatment in MRL/lpr mice. MRL/lpr mice were divided into saline (C), cyclophosphamide (Y), ASC early (E), ASC late (L), and CTLA4Ig-overexpressing ASC (CT) treatment groups. Background-matched control MRL/MPJ mice treated with saline (N) were also compared. The treatment period was 5-23 weeks, except for the L group (15-23 weeks). Blood and tissue samples were collected when the mice were 24 weeks old. Organ weight, anti-dsDNA antibodies, urine protein, skin and kidney histologic abnormalities, and trabecular bone volume were evaluated. The Y group showed the greatest decrease in anti-dsDNA antibodies, organ weight, degree of kidney inflammation and glomerular infiltration of C3, and incidence rate of severe proteinuria; the E, L, and CT treatment groups showed better results than the C group. ASC transplantation reduced anti-dsDNA antibody levels significantly. Mice treated with ASCs or CTLA4Ig-ASCs starting from the early disease stage did not show dermatitis upon gross examination; they demonstrated significant improvement in hyperkeratosis, acanthosis, and inflammatory cell infiltration scores in histopathology. Micro-CT analysis revealed that cyclophosphamide treatment significantly decreased bone volume and increased bone spacing in the trabecular bone. Thus, we found that ASC and CTLA4-ASC treatments prevent lupus dermatitis development in MRL/lpr mice without adverse effects.
Collapse
Affiliation(s)
- Eun Wha Choi
- 1 Laboratory Animal Research Center, Samsung Biomedical Research Institute , Seoul, Republic of Korea.,2 School of Medicine, Sungkyunkwan University , Seoul, Republic of Korea
| | - Il Seob Shin
- 3 Biostar Stem Cell Research Center, K-STEMCELL , Seoul, Republic of Korea
| | - Ji Woo Song
- 1 Laboratory Animal Research Center, Samsung Biomedical Research Institute , Seoul, Republic of Korea
| | - Tae Won Yun
- 1 Laboratory Animal Research Center, Samsung Biomedical Research Institute , Seoul, Republic of Korea
| | - Jehoon Yang
- 1 Laboratory Animal Research Center, Samsung Biomedical Research Institute , Seoul, Republic of Korea.,2 School of Medicine, Sungkyunkwan University , Seoul, Republic of Korea
| | - Kyu-Sil Choi
- 1 Laboratory Animal Research Center, Samsung Biomedical Research Institute , Seoul, Republic of Korea.,2 School of Medicine, Sungkyunkwan University , Seoul, Republic of Korea
| | - Je Kyung Seong
- 4 Laboratory of Developmental Biology and Genomics, BK21 Program for Veterinary Science, Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| |
Collapse
|
11
|
Affiliation(s)
- Hiroki Yoshida
- Department of Biomolecular Sciences, Division of Molecular and Cellular Immunoscience, Saga University Faculty of Medicine, Saga 849-8501, Japan;
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-4539;
| |
Collapse
|
12
|
Clarke EV, Weist BM, Walsh CM, Tenner AJ. Complement protein C1q bound to apoptotic cells suppresses human macrophage and dendritic cell-mediated Th17 and Th1 T cell subset proliferation. J Leukoc Biol 2015; 97:147-60. [PMID: 25381385 PMCID: PMC4377823 DOI: 10.1189/jlb.3a0614-278r] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/15/2014] [Accepted: 09/01/2014] [Indexed: 12/17/2022] Open
Abstract
A complete genetic deficiency of the complement protein C1q results in SLE with nearly 100% penetrance in humans, but the molecular mechanisms responsible for this association have not yet been fully determined. C1q opsonizes ACs for enhanced ingestion by phagocytes, such as Mϕ and iDCs, avoiding the extracellular release of inflammatory DAMPs upon loss of the membrane integrity of the dying cell. We previously showed that human monocyte-derived Mϕ and DCs ingesting autologous, C1q-bound LALs (C1q-polarized Mϕ and C1q-polarized DCs), enhance the production of anti-inflammatory cytokines, and reduce proinflammatory cytokines relative to Mϕ or DC ingesting LAL alone. Here, we show that C1q-polarized Mϕ have elevated PD-L1 and PD-L2 and suppressed surface CD40, and C1q-polarized DCs have higher surface PD-L2 and less CD86 relative to Mϕ or DC ingesting LAL alone, respectively. In an MLR, C1q-polarized Mϕ reduced allogeneic and autologous Th17 and Th1 subset proliferation and demonstrated a trend toward increased Treg proliferation relative to Mϕ ingesting LAL alone. Moreover, relative to DC ingesting AC in the absence of C1q, C1q-polarized DCs decreased autologous Th17 and Th1 proliferation. These data demonstrate that a functional consequence of C1q-polarized Mϕ and DC is the regulation of Teff activation, thereby "sculpting" the adaptive immune system to avoid autoimmunity, while clearing dying cells. It is noteworthy that these studies identify novel target pathways for therapeutic intervention in SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- Elizabeth V Clarke
- Department of Molecular Biology and Biochemistry, Institute for Immunology, University of California-Irvine, Irvine, California, USA; and
| | - Brian M Weist
- Department of Molecular & Cell Biology, University of California-Berkeley, Berkeley, California, USA
| | - Craig M Walsh
- Department of Molecular Biology and Biochemistry, Institute for Immunology, University of California-Irvine, Irvine, California, USA; and
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, Institute for Immunology, University of California-Irvine, Irvine, California, USA; and
| |
Collapse
|
13
|
Martin JC, Baeten DL, Josien R. Emerging role of IL-17 and Th17 cells in systemic lupus erythematosus. Clin Immunol 2014; 154:1-12. [DOI: 10.1016/j.clim.2014.05.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/10/2014] [Accepted: 05/14/2014] [Indexed: 12/14/2022]
|
14
|
|
15
|
Shinsuke N, Hiroshi I. Overexpression of Epstein-Barr virus-induced gene 3 protein (EBI3) in MRL/lpr mice suppresses their lupus nephritis by activating regulatory T cells. Autoimmunity 2013; 46:446-54. [PMID: 23845089 DOI: 10.3109/08916934.2013.809422] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
To identify the effect of an imbalance of Th1/Th2 cytokines on the development of autoimmune glomerulonephritis (lupus nephritis), we studied the modification of pathological changes in diffuse proliferative glomerulonephritis (DPGN) and membranous glomerulonephritis (MGN) in MRL/lpr mice, which are animal models of systemic lupus erythematosus (SLE). Transgenic MRL/lpr mice (Tg) that overexpressed Epstein--Barr virus-induced gene 3 (EBI3) showed almost normal renal function, which was demonstrated by healing of glomerulonephritis upon renal histology, as compared to the wild-type MRL/lpr (Wt) mice. The levels of anti-dsDNA antibodies and IgE decreased in the Tg mice compared to Wt mice. Quantitative real-time PCR indicated an increase in the mRNA levels of FoxP3, and a decrease in that of IFNγ in the splenocytes of Tg mice as compared to Wt mice. In addition, flow cytometric analysis showed an increase in CD4(+)CD25(+)FoxP3(+)-T cells in the former, as compared to the latter. Our findings suggest that EBI3-overexpression in MRL/lpr mice induces generation of regulatory T cells, which causes suppression of autoimmune and inflammatory reactions by affecting the Th1/Th2 cytokine balance.
Collapse
|
16
|
Abstract
It has been more than 15 years since the identification of individual interleukin-27 (IL-27) and IL-27 receptor components. The last decade has seen the description of the signaling pathways engaged by IL-27, and an appreciation has emerged that this cytokine can modulate the intensity and duration of many classes of T cell responses. Here we provide an overview of the immunobiology of IL-27 and review advances in understanding the functions of individual IL-27 and IL-27 receptor subunits and the role of IL-27 in dictating the balance between protective and pathological immunity. Additionally, this cytokine has been proposed as a therapy to modify inflammatory conditions or to promote antitumor responses, and situations where experimental and clinical data sets implicate IL-27 in the outcome of disease are highlighted.
Collapse
|
17
|
Immunologic and genetic considerations of cutaneous lupus erythematosus: a comprehensive review. J Autoimmun 2013; 41:34-45. [PMID: 23380467 DOI: 10.1016/j.jaut.2013.01.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 01/04/2013] [Indexed: 12/20/2022]
Abstract
Cutaneous lupus erythematosus (CLE) refers to those subtypes of lupus erythematosus (LE) that have predominantly skin manifestations. Discoid lupus erythematosus (DLE), subacute cutaneous lupus erythematosus (SCLE), LE panniculitis (LEP) and lupus erythematosus tumidus (LET) all fall into the category of CLE. The pathogenesis of CLE is likely multifactorial. UV irradiation has been shown to induce keratinocyte apoptosis. Impaired clearance of apoptotic cells is a potential mechanism for the development of CLE. UV irradiation can also induce externalization of autoantigens such as Ro/SSA, exposing them to circulating autoantibodies. Some drugs have been associated with CLE. Possible mechanisms include stimulation of an immune response through disruption of central tolerance and altered T cell function. T17 cells may also play a role in the pathogenesis of CLE as they have been detected in skin lesions of LE. Treg cells have been found to be decreased in LE lesions, which may contribute to the breakdown of self-tolerance. Epidermal Langerhans cells are reduced in CLE while plasmacytoid DCs are increased in the lesions of CLE, suggesting that DCs may also play an important role in the pathogenesis of CLE. Type I IFN- and TNF-α are both upregulated in lesions of CLE. Other cytokines such as IL-6 and IL-17 are also implicated in the pathogenesis of CLE. Cellular and cytokine networks can be impacted by environmental factors and genetic variations and this can result in an increased risk of developing autoimmune diseases such as CLE.
Collapse
|
18
|
Immunoregulatory function of IL-27 and TGF-β1 in cardiac allograft transplantation. Transplantation 2012; 94:226-33. [PMID: 22790384 DOI: 10.1097/tp.0b013e31825b0c38] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Deciphering the mechanisms of tolerance represents a crucial aim of research in transplantation. We previously identified by DNA chip interleukin (IL)-27 p28 and transforming growth factor (TGF)-β1 as overexpressed in a model of rat cardiac allograft tolerance mediated by regulatory CD4CD25 T cells. The role of these two molecules on the control of the inflammatory response remains controversial. However, both are involved in the regulation of the T helper 17/Treg axis, suggesting their involvement in tolerance. METHODS We analyzed regulation of IL-27 and TGF-β1 expression in allograft response and their role in tolerance by using blocking anti-TGF-β antibody and by generating an adeno-associated virus encoding IL-27. RESULTS Here, we confirmed the overexpression of IL-27 and TGF-β1 in tolerated cardiac allografts in two different rodent models. We observed that their expression correlates with inhibition of T helper 17 differentiation and with expansion of regulatory CD4CD25 T cells. We showed in a rat model that anti-TGF-β treatment abrogates infectious tolerance mediated by the transfer of regulatory CD4CD25 T cells. Moreover, overexpression of IL-27 by adeno-associated virus administration in combination with a short-term immunosuppression allows prolongation of cardiac allograft survival and one tolerant recipient. We found that IL-27 overexpression did not induce Foxp3CD4CD25 T-cell expansion but rather IL-10-expressing CD4 T cells in the tolerant recipient. CONCLUSIONS Taken together, these data suggest that both TGF-β1 and IL-27 play a role in the mechanisms of tolerance. However, in contrast to TGF-β1, IL-27 seems not to be involved in regulatory CD4CD25 T-cell expansion but rather in their mode of action.
Collapse
|
19
|
Tojo GI, Fujimura T, Kambayashi Y, Aiba S. Systemic Lupus Erythematosus Accompanied by Psoriasis Induces IL-27-Producing Cells in Both Affected Areas of the Skin. Case Rep Dermatol 2012; 4:181-5. [PMID: 23071464 PMCID: PMC3457044 DOI: 10.1159/000342803] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
IL-27 is mainly produced by activated antigen-presenting cells (APCs), and has been reported in various Th1/Th17-mediated inflammatory diseases, including psoriasis, and even in Th2-complexed disease, such as systemic lupus erythematosus (SLE). We describe a 51-year-old Japanese man with SLE accompanied by psoriasis. Interestingly, immunohistochemical staining revealed the existence of IL-27-producing cells in biopsy specimens from both SLE and psoriasis. Our findings might suggest relationships between IL-27 and the pathogenesis of these inflammatory diseases.
Collapse
Affiliation(s)
- Gen-Ichi Tojo
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | |
Collapse
|
20
|
Abstract
Like many cytokines, IL-27 has pleiotropic properties that can limit or enhance ongoing immune responses depending on context. Thus, under certain circumstances, IL-27 can promote TH1 differentiation and has been linked to the activation of CD8(+) T cells and enhanced humoral responses. However, IL-27 also has potent inhibitory properties and mice that lack IL-27 mediated signaling develop exaggerated inflammatory responses in the context of infection or autoimmunity. This chapter reviews in depth the biology of IL-27, including the initial discovery, characterization, and signaling mediated by IL-27 as well as more recent insights into the molecular and cellular basis for its pleiotropic effects. Many of these advances are relevant to human diseases and highlight the potential of therapies that harness the regulatory properties of IL-27.
Collapse
Affiliation(s)
- Aisling O'Hara Hall
- Department of Pathobiology, School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
21
|
Apostolidis SA, Lieberman LA, Kis-Toth K, Crispín JC, Tsokos GC. The dysregulation of cytokine networks in systemic lupus erythematosus. J Interferon Cytokine Res 2011; 31:769-79. [PMID: 21877904 PMCID: PMC3189553 DOI: 10.1089/jir.2011.0029] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 06/16/2011] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease associated with chronic immune activation and tissue damage. Organ damage in SLE results from the deposition of immune complexes and the infiltration of activated T cells into susceptible organs. Cytokines are intimately involved in every step of the SLE pathogenesis. Defective immune regulation and uncontrolled lymphocyte activation, as well as increased antigen presenting cell maturation are all influenced by cytokines. Moreover, expansion of local immune responses as well as tissue infiltration by pathogenic cells is instigated by cytokines. In this review, we describe the main cytokine abnormalities reported in SLE and discuss the mechanisms that drive their aberrant production as well as the pathogenic pathways that their presence promotes.
Collapse
Affiliation(s)
- Sokratis A Apostolidis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|