101
|
Schwartz C, O'Grady K, Lavelle EC, Fallon PG. Interleukin 33: an innate alarm for adaptive responses beyond Th2 immunity-emerging roles in obesity, intestinal inflammation, and cancer. Eur J Immunol 2016; 46:1091-100. [DOI: 10.1002/eji.201545780] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 02/20/2016] [Accepted: 03/15/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Christian Schwartz
- School of Medicine, Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin 2 Ireland
| | - Katie O'Grady
- Adjuvant Research Group, School of Biochemistry and Immunology; Trinity Biomedical Sciences Institute, Trinity College Dublin; Dublin 2 Ireland
| | - Ed C. Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology; Trinity Biomedical Sciences Institute, Trinity College Dublin; Dublin 2 Ireland
| | - Padraic G. Fallon
- School of Medicine, Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin 2 Ireland
| |
Collapse
|
102
|
Lu B, Yang M, Wang Q. Interleukin-33 in tumorigenesis, tumor immune evasion, and cancer immunotherapy. J Mol Med (Berl) 2016; 94:535-43. [PMID: 26922618 DOI: 10.1007/s00109-016-1397-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/14/2016] [Accepted: 02/17/2016] [Indexed: 12/20/2022]
Abstract
Interleukin-33 (IL-33) is a member of the IL-1 gene family and mainly expressed in the nucleus of tissue lining cells, stromal cells, and activated myeloid cells. IL-33 is considered a damage-associated molecular pattern (DAMP) molecule and plays an important role in many physiological and pathological settings such as tissue repair, allergy, autoimmune disease, infectious disease, and cancer. The biological functions of IL-33 include maintaining tissue homeostasis, enhancing type 1 and 2 cellular immune responses, and mediating fibrosis during chronic inflammation. IL-33 exerts diverse functions through signaling via its receptor ST2, which is expressed in many types of cells including regulatory T cells (Treg), group 2 innate lymphoid cells (ILC2s), myeloid cells, cytotoxic NK cells, Th2 cells, Th1 cells, and CD8(+) T cells. Tumor development results in downregulation of IL-33 in epithelial cells but upregulation of IL-33 in the tumor stroma and serum. The current data suggest that IL-33 expression in tumor cells increases immunogenicity and promotes type 1 antitumor immune responses through CD8(+) T cells and NK cells, whereas IL-33 in tumor stroma and serum facilitates immune suppression via Treg and myeloid-derived suppressor cell (MDSC). Understanding the role of IL-33 in cancer immunobiology sheds lights on targeting this cytokine for cancer immunotherapy.
Collapse
Affiliation(s)
- Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA, 15261, USA.
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
| | - Min Yang
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
103
|
Apostolova P, Zeiser R. The role of danger signals and ectonucleotidases in acute graft-versus-host disease. Hum Immunol 2016; 77:1037-1047. [PMID: 26902992 DOI: 10.1016/j.humimm.2016.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/09/2016] [Accepted: 02/18/2016] [Indexed: 12/28/2022]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) represents the only curative treatment approach for many patients with benign or malignant diseases of the hematopoietic system. However, post-transplant morbidity and mortality are significantly increased by the development of acute graft-versus-host disease (GvHD). While alloreactive T cells act as the main cellular mediator of the GvH reaction, recent evidence suggests a critical role of the innate immune system in the early stages of GvHD initiation. Danger-associated molecular patterns released from the intracellular space as well as from the extracellular matrix activate antigen-presenting cells and set pro-inflammatory pathways in motion. This review gives an overview about danger signals representing therapeutic targets with a clinical perspective with a particular focus on extracellular nucleotides and ectonucleotidases.
Collapse
Affiliation(s)
- Petya Apostolova
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany.
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany.
| |
Collapse
|
104
|
Kim J, Kim W, Moon UJ, Kim HJ, Choi HJ, Sin JI, Park NH, Cho HR, Kwon B. Intratumorally Establishing Type 2 Innate Lymphoid Cells Blocks Tumor Growth. THE JOURNAL OF IMMUNOLOGY 2016; 196:2410-23. [PMID: 26829987 DOI: 10.4049/jimmunol.1501730] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/22/2015] [Indexed: 12/12/2022]
Abstract
A long-standing question in the field of tumor immunotherapy is how Th2 cytokines block tumor growth. Their antitumor effects are particularly prominent when they are secreted continuously in tumors, suggesting that Th2 cytokines may create a tumor microenvironment unfavorable for tumor growth independently of adaptive immunity. In this study, we show that local production of IL-33 establishes a high number of type 2 innate lymphoid cells (ILC2s) with potent antitumor activity. IL-33 promotes secretion of a massive amount of CXCR2 ligands from ILC2s but creates a tumor microenvironment where tumor cells express CXCR2 through a dysfunctional angiogenesis/hypoxia/reactive oxygen species axis. These two signaling events converge to reinforce tumor cell-specific apoptosis through CXCR2. Our results identify a previously unrecognized antitumor therapeutic pathway wherein ILC2s play a central role.
Collapse
Affiliation(s)
- Juyang Kim
- Biomedical Research Center, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea
| | - Wonyoung Kim
- Biomedical Research Center, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea
| | - U J Moon
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Hyun J Kim
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Hye-Jeong Choi
- Department of Pathology, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea
| | - Jeong-Im Sin
- Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea
| | - Neung H Park
- Biomedical Research Center, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea; Department of Internal Medicine, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea; and
| | - Hong R Cho
- Biomedical Research Center, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea; Department of Surgery, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea
| | - Byungsuk Kwon
- Biomedical Research Center, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea; School of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea;
| |
Collapse
|
105
|
Ottenlinger F, Schwiebs A, Pfarr K, Wagner A, Grüner S, Mayer CA, Pfeilschifter JM, Radeke HH. Fingolimod targeting protein phosphatase 2A differently affects IL-33 induced IL-2 and IFN-γ production in CD8(+) lymphocytes. Eur J Immunol 2016; 46:941-51. [PMID: 26683421 DOI: 10.1002/eji.201545805] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 11/02/2015] [Accepted: 12/10/2015] [Indexed: 01/05/2023]
Abstract
Multiple sclerosis patients are treated with fingolimod (FTY720), a prodrug that acts as an immune modulator. FTY720 is first phosphorylated to FTY720-P and then internalizes sphingosine-1-phosphate receptors, preventing lymphocyte sequestration. IL-33 is released from necrotic endothelial cells and contributes to MS severity by coactivating T cells. Herein we analyzed the influence of FTY720, FTY720-P, and S1P on IL-33 induced formation of IL-2 and IFN-γ, by using IL-33 receptor overexpressing EL4 cells, primary CD8(+) T cells, and splenocytes. EL4-ST2 cells released IL-2 after IL-33 stimulation that was inhibited dose-dependently by FTY720-P but not FTY720. In this system, S1P increased IL-2, and accordingly, inhibition of S1P producing sphingosine kinases diminished IL-2 release. In primary CD8(+) T cells and splenocytes IL-33/IL-12 stimulation induced IFN-γ, which was prevented by FTY720 but not FTY720-P, independently from intracellular phosphorylation. The inhibition of IFN-γ by nonphosphorylated FTY720 was mediated via the SET/protein phosphatase 2A (PP2A) pathway, since a SET peptide antagonist also prevented IFN-γ formation and the inhibition of IFN-γ by FTY720 was reversible by a PP2A inhibitor. While our findings directly improve the understanding of FTY720 therapy in MS, they could also contribute to side effects of FTY720 treatment, like progressive multifocal leukoencephalopathy, caused by an insufficient immune response to a viral infection.
Collapse
Affiliation(s)
- Florian Ottenlinger
- pharmazentrum frankfurt/ZAFES, Hospital of the Goethe University, Frankfurt am Main, Germany
| | - Anja Schwiebs
- pharmazentrum frankfurt/ZAFES, Hospital of the Goethe University, Frankfurt am Main, Germany
| | - Kathrin Pfarr
- pharmazentrum frankfurt/ZAFES, Hospital of the Goethe University, Frankfurt am Main, Germany
| | - Annika Wagner
- pharmazentrum frankfurt/ZAFES, Hospital of the Goethe University, Frankfurt am Main, Germany
| | - Sophia Grüner
- pharmazentrum frankfurt/ZAFES, Hospital of the Goethe University, Frankfurt am Main, Germany
| | - Christoph A Mayer
- Center for Neurology and Neurosurgery, Hospital of the Goethe University, Frankfurt am Main, Germany
| | - Josef M Pfeilschifter
- pharmazentrum frankfurt/ZAFES, Hospital of the Goethe University, Frankfurt am Main, Germany
| | - Heinfried H Radeke
- pharmazentrum frankfurt/ZAFES, Hospital of the Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
106
|
van der Torren CR, Verrijn Stuart AA, Lee D, Meerding J, van de Velde U, Pipeleers D, Gillard P, Keymeulen B, de Jager W, Roep BO. Serum Cytokines as Biomarkers in Islet Cell Transplantation for Type 1 Diabetes. PLoS One 2016; 11:e0146649. [PMID: 26751709 PMCID: PMC4713434 DOI: 10.1371/journal.pone.0146649] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 12/21/2015] [Indexed: 11/19/2022] Open
Abstract
Background Islet cell transplantation holds a potential cure for type 1 diabetes, but many islet recipients do not reach long-lasting insulin independence. In this exploratory study, we investigated whether serum cytokines, chemokines and adipokines are associated with the clinical outcome of islet transplantation. Methods Thirteen islet transplant patients were selected on basis of good graft function (reaching insulin independence) or insufficient engraftment (insulin requiring) from our cohort receiving standardized grafts and immune suppressive therapy. Patients reaching insulin independence were divided in those with continued (>12 months) versus transient (<6 months) insulin independence. A panel of 94 proteins including cytokines and adipokines was measured in sera taken before and at one year after transplantation using a validated multiplex immunoassay platform. Results Ninety serum proteins were detectable in concentrations varying markedly among patients at either time point. Thirteen markers changed after transplantation, while another seven markers changed in a clinical subpopulation. All other markers remained unaffected after transplantation under generalized immunosuppression. Patterns of cytokines could distinguish good graft function from insufficient function including IFN-α, LIF, SCF and IL-1RII before and after transplantation, by IL-16, CCL3, BDNF and M-CSF only before and by IL-22, IL-33, KIM-1, S100A12 and sCD14 after transplantation. Three other proteins (Leptin, Cathepsin L and S100A12) associated with loss of temporary graft function before or after transplantation. Conclusions Distinct cytokine signatures could be identified in serum that predict or associate with clinical outcome. These serum markers may help guiding patient selection and choice of immunotherapy, or act as novel drug targets in islet transplantation.
Collapse
Affiliation(s)
- Cornelis R. van der Torren
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
- Juvenile Diabetes Research Foundation Center for Beta Cell Therapy in Diabetes
| | - Annemarie A. Verrijn Stuart
- Department of Pediatric Immunology, Department of Pediatric Endocrinology and Laboratory of Translational Immunology and Multiplex Core Facility, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - DaHae Lee
- Dept. of Endocrinology, University Hospital, Katholieke Universiteit Leuven, 3000, Leuven, Belgium
- Juvenile Diabetes Research Foundation Center for Beta Cell Therapy in Diabetes
| | - Jenny Meerding
- Department of Pediatric Immunology, Department of Pediatric Endocrinology and Laboratory of Translational Immunology and Multiplex Core Facility, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Ursule van de Velde
- Diabetes Research Center and Academic Hospital, Free University-Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium
- Juvenile Diabetes Research Foundation Center for Beta Cell Therapy in Diabetes
| | - Daniel Pipeleers
- Diabetes Research Center and Academic Hospital, Free University-Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium
| | - Pieter Gillard
- Dept. of Endocrinology, University Hospital, Katholieke Universiteit Leuven, 3000, Leuven, Belgium
- Juvenile Diabetes Research Foundation Center for Beta Cell Therapy in Diabetes
| | - Bart Keymeulen
- Diabetes Research Center and Academic Hospital, Free University-Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium
- Juvenile Diabetes Research Foundation Center for Beta Cell Therapy in Diabetes
| | - Wilco de Jager
- Department of Pediatric Immunology, Department of Pediatric Endocrinology and Laboratory of Translational Immunology and Multiplex Core Facility, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Bart O. Roep
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
- Juvenile Diabetes Research Foundation Center for Beta Cell Therapy in Diabetes
- * E-mail:
| |
Collapse
|
107
|
Okragly AJ, Hamang MJ, Pena EA, Baker HE, Bullock HA, Lucchesi J, Martin AP, Ma YL, Benschop RJ. Elevated levels of Interleukin (IL)-33 induce bone pathology but absence of IL-33 does not negatively impact normal bone homeostasis. Cytokine 2016; 79:66-73. [PMID: 26771472 DOI: 10.1016/j.cyto.2015.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/05/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
Interleukin (IL)-33 is a member of the IL-1 family. IL-33 effects are mediated through its receptor, ST2 and IL-1RAcP, and its signaling induces the production of a number of pro-inflammatory mediators, including TNFα, IL-1β, IL-6, and IFN-γ. There are conflicting reports on the role of IL-33 in bone homeostasis, with some demonstrating a bone protective role for IL-33 whilst others show that IL-33 induces inflammatory arthritis with concurrent bone destruction. To better clarify the role IL-33 plays in bone biology in vivo, we studied IL-33 KO mice as well as mice in which the cytokine form of IL-33 was overexpressed. Mid-femur cortical bone mineral density (BMD) and bone strength were similar in the IL-33 KO mice compared to WT animals during the first 8months of life. However, in the absence of IL-33, we observed higher BMD in lumbar vertebrae and distal femur in female mice. In contrast, overexpression of IL-33 resulted in a marked and rapid reduction of bone volume, mineral density and strength. Moreover, this was associated with a robust increase in inflammatory cytokines (including IL-6 and IFN-γ), suggesting the bone pathology could be a direct effect of IL-33 or an indirect effect due to the induction of other mediators. Furthermore, the detrimental bone effects were accompanied by increases in osteoclast number and the bone resorption marker of C-terminal telopeptide collagen-I (CTX-I). Together, these results demonstrate that absence of IL-33 has no negative consequences in normal bone homeostasis while high levels of circulating IL-33 contributes to pathological bone loss.
Collapse
Affiliation(s)
- Angela J Okragly
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Matthew J Hamang
- Musculoskeletal-Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Emily A Pena
- Musculoskeletal-Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Hana E Baker
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Heather A Bullock
- Musculoskeletal-Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jonathan Lucchesi
- Musculoskeletal-Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Andrea P Martin
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Y Linda Ma
- Musculoskeletal-Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Robert J Benschop
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA.
| |
Collapse
|
108
|
Abstract
Thymic-derived, regulatory T cells (Treg) represent a subset of CD4(+) T cells that are required for normal immune homeostasis and suppression of unwanted responses against self-antigens (Ags) that prevent autoimmunity. Their role as immune regulators and potent ability to suppress T cell responses has been the focus of intense investigations aimed at utilizing these cells therapeutically, particularly in the settings of autoimmunity and transplantation. Many methods for expanding Treg have been described; however, efforts to generate large numbers of Treg for use in vivo often compromise their suppressor function or rely on the induction of Treg rather than their expansion. Our recent studies have focused on the barrier tissue-derived cytokine IL-33, a recently described IL-1 family member. IL-33 has emerged as a multifunctional protein, with reported roles in driving potent Type 1 and Type 2 immunity, as well as facilitating profound Treg expansion in vitro and in vivo. IL-33-expanded Treg express the IL-33 receptor (R) ST2, and express classical markers associated with Treg phenotype and suppressor function. They suppress both CD4(+) and CD8(+) T cell proliferation and effector functions in vitro, and Treg expressing ST2 have been identified as important regulators of detrimental immune responses in vivo. In the present chapter, we detail methods for expanding significant numbers of Treg using IL-33 both in vitro and in vivo that may potentially be used to promote/maintain organ transplant tolerance or suppress autoimmunity.
Collapse
Affiliation(s)
- Benjamin M Matta
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Hēth R Turnquist
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
109
|
O'Donnell C, Mahmoud A, Keane J, Murphy C, White D, Carey S, O'Riordain M, Bennett MW, Brint E, Houston A. An antitumorigenic role for the IL-33 receptor, ST2L, in colon cancer. Br J Cancer 2015; 114:37-43. [PMID: 26679377 PMCID: PMC4716545 DOI: 10.1038/bjc.2015.433] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 11/06/2015] [Accepted: 11/18/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Despite the importance of inflammation in cancer, the role of the cytokine IL-33, and its receptor ST2, in colon cancer is unclear. The aim of this study was to investigate the role of IL-33, and its receptor isoforms (ST2 and ST2L), in colon cancer. METHODS Serum levels of IL-33 and sST2 were determined with ELISA. ST2 and IL-33 expression was detected with quantitative real-time PCR (qRT-PCR), western blotting and immunohistochemistry. ST2 expression in CT26 cells was stably suppressed using ST2-specific shRNA. Cytokine and chemokine gene expression was detected with qRT-PCR. RESULTS Human colon tumours showed lower expression of ST2L as compared with adjacent non-tumour tissue (P<0.01). Moreover, the higher the tumour grade, the lower the expression of ST2L (P=0.026). Colon cancer cells expressed ST2 and IL-33 in vitro. Functional analyses showed that stimulation of tumour cells with IL-33 induced the expression of chemokine (C-C motif) ligand 2 (CCL2). Knockdown of ST2 in murine colon cancer cells resulted in enhanced tumour growth (P<0.05) in BALB/c mice in vivo. This was associated with a decrease in macrophage infiltration, with IL-33-induced macrophage recruitment reduced by antagonising CCL2 in vitro. CONCLUSION The IL-33/ST2 signalling axis may have a protective role in colon carcinogenesis.
Collapse
Affiliation(s)
| | - Amr Mahmoud
- Department of Pathology, University College Cork, Cork, Ireland
| | - Jonathan Keane
- Department of Medicine, University College Cork, Cork, Ireland.,Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Carola Murphy
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
| | - Declan White
- Department of Pathology, University College Cork, Cork, Ireland.,Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Sinead Carey
- Department of Histopathology, Cork University Hospital, Cork, Ireland
| | - Micheal O'Riordain
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland.,Department of Surgery, Mercy University Hospital, Cork, Ireland
| | - Michael W Bennett
- Department of Histopathology, Cork University Hospital, Cork, Ireland
| | - Elizabeth Brint
- Department of Pathology, University College Cork, Cork, Ireland.,Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Aileen Houston
- Department of Medicine, University College Cork, Cork, Ireland.,Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| |
Collapse
|
110
|
Komai-Koma M, Wang E, Kurowska-Stolarska M, Li D, McSharry C, Xu D. Interleukin-33 promoting Th1 lymphocyte differentiation dependents on IL-12. Immunobiology 2015; 221:412-7. [PMID: 26688508 PMCID: PMC4731778 DOI: 10.1016/j.imbio.2015.11.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 11/26/2015] [Accepted: 11/29/2015] [Indexed: 12/14/2022]
Abstract
The pro-Th2 cytokine IL-33 is now emerging as an important Th1 cytokine-IFN-γ inducer in murine CD4+ T cells that is essential for protective cell-mediated immunity against viral infection in mice. However, whether IL-33 can promote human Th1 cell differentiation and how IL-33 polarizes Th1 cells is less understood. We assessed the ability of IL-33 to induce Th1 cell differentiation and IFN-γ production in vitro and in vivo. We report here that IL-33 alone had no ability in Th1 cell polarization. However it potentiated IL-12-mediated Th1 cell differentiation and IFN-γ production in TCR-stimulated murine and human CD4+ T cells in vitro and in vivo. IL-33 promoted Th1 cell development via MyD88 and synergized with IL-12 to enhance St2 and IL-12R expression in CD4+ T cells. These data therefore provide a novel mechanism for Th1 cell differentiation and optimal induction of a Type 1 response. Thus, IL-33 is capable of inducing IL-12-dependent Th1 cell differentiation in human and mouse CD4+ T cells.
Collapse
Affiliation(s)
- Mousa Komai-Koma
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK; Department of Haematology & Immunology, Faculty of Medicine, Umm Al-Qura University, Mecca, KSA, Saudi Arabia
| | - Eryi Wang
- Department of Pharmacology, Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | | | - Dong Li
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Charles McSharry
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Damo Xu
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| |
Collapse
|
111
|
Wagner A, Köhm M, Nordin A, Svenungsson E, Pfeilschifter JM, Radeke HH. Increased Serum Levels of the IL-33 Neutralizing sST2 in Limited Cutaneous Systemic Sclerosis. Scand J Immunol 2015; 82:269-74. [PMID: 26095613 DOI: 10.1111/sji.12317] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 06/07/2015] [Indexed: 01/31/2023]
Abstract
The pathophysiology of both limited cutaneous systemic sclerosis (lcSSc) and diffuse cutaneous SSc (dcSSc), representing two subtypes of an autoimmune disease of the connective tissue, is still enigmatic. Life-limiting, progressive fibrotic changes as a consequence of vasculopathy and autoimmunity are characteristic in varying extent for lcSSc and dcSSc. Previously, an increased IL-33 serum concentration in early phase SSc patients and an elevated tissue expression of its receptor, ST2L, on endothelial cells (EC) were described. While suggested as a biomarker for fibrotic diseases, for example liver fibrosis, the role of soluble ST2 (sST2) in the pathological processes and its contribution to vascular fibrosis in SSc has not been investigated. Here, we showed that sST2 is elevated in late phase limited cutaneous SSc (lcSSc) as compared to patients with shorter disease duration or with the diffuse subtype of SSc. We demonstrated that sST2, not IL-33, is significantly increased in serum of lcSSc patients with disease duration over 9 years. Soluble ST2 was not elevated in healthy controls or in SSc patients with early skin involvement or disease duration shorter than 9 years. Furthermore, we observed that sST2 serum levels were lowered by iloprost (prostacyclin) treatment. After 5 days of iloprost infusion, sST2 serum levels fell in 6 of 7 patients. Therefore, we not only like to propose sST2 as a biomarker for progressive vascular fibrosis, but moreover, suggest that the involvement of sST2 in the pathogenesis of lcSSc may be exploited therapeutically.
Collapse
Affiliation(s)
- A Wagner
- pharmazentrum frankfurt/ZAFES, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - M Köhm
- Division of Rheumatology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Frankfurt am Main, Germany
| | - A Nordin
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - E Svenungsson
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - J M Pfeilschifter
- pharmazentrum frankfurt/ZAFES, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - H H Radeke
- pharmazentrum frankfurt/ZAFES, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
112
|
Tsurutani N, Mittal P, St Rose MC, Ngoi SM, Svedova J, Menoret A, Treadway FB, Laubenbacher R, Suárez-Ramírez JE, Cauley LS, Adler AJ, Vella AT. Costimulation Endows Immunotherapeutic CD8 T Cells with IL-36 Responsiveness during Aerobic Glycolysis. THE JOURNAL OF IMMUNOLOGY 2015; 196:124-34. [PMID: 26573834 DOI: 10.4049/jimmunol.1501217] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 10/17/2015] [Indexed: 01/07/2023]
Abstract
CD134- and CD137-primed CD8 T cells mount powerful effector responses upon recall, but even without recall these dual-costimulated T cells respond to signal 3 cytokines such as IL-12. We searched for alternative signal 3 receptor pathways and found the IL-1 family member IL-36R. Although IL-36 alone did not stimulate effector CD8 T cells, in combination with IL-12, or more surprisingly IL-2, it induced striking and rapid TCR-independent IFN-γ synthesis. To understand how signal 3 responses functioned in dual-costimulated T cells we showed that IL-2 induced IL-36R gene expression in a JAK/STAT-dependent manner. These data help delineate a sequential stimulation process where IL-2 conditioning must precede IL-36 for IFN-γ synthesis. Importantly, this responsive state was transient and functioned only in effector T cells capable of aerobic glycolysis. Specifically, as the effector T cells metabolized glucose and consumed O2, they also retained potential to respond through IL-36R. This suggests that T cells use innate receptor pathways such as the IL-36R/axis when programmed for aerobic glycolysis. To explore a function for IL-36R in vivo, we showed that dual costimulation therapy reduced B16 melanoma tumor growth while increasing IL-36R gene expression. In summary, cytokine therapy to eliminate tumors may target effector T cells, even outside of TCR specificity, as long as the effectors are in the correct metabolic state.
Collapse
Affiliation(s)
- Naomi Tsurutani
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Payal Mittal
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Marie-Clare St Rose
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Soo Mun Ngoi
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Julia Svedova
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Antoine Menoret
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Forrest B Treadway
- Center for Quantitative Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Reinhard Laubenbacher
- Center for Quantitative Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Jenny E Suárez-Ramírez
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Linda S Cauley
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Adam J Adler
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Anthony T Vella
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| |
Collapse
|
113
|
ST2 contributes to T-cell hyperactivation and fatal hemophagocytic lymphohistiocytosis in mice. Blood 2015; 127:426-35. [PMID: 26518437 DOI: 10.1182/blood-2015-07-659813] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/25/2015] [Indexed: 12/19/2022] Open
Abstract
Cytokine storm syndromes, such as familial hemophagocytic lymphohistiocytosis (FHL), are lethal disorders caused by uncontrolled, systemic immune activation. In the murine model of FHL, in which perforin-deficient (Prf1(-/-)) mice are infected with lymphocytic choriomeningitis virus (LCMV), disease is driven by overabundant interferon (IFN)γ-producing LCMV-specific CD8(+) T cells thought to arise from excessive antigen stimulation through the T-cell receptor. However, this paradigm is insufficient to explain several fundamental aspects of FHL, namely, the inability of many pathogenic antigens to induce hyperinflammation, and the previously identified role of MyD88 in the disease. We now show a novel role for the MyD88-dependent interleukin-33 (IL-33) receptor, ST2, in FHL. Expression of IL-33 and ST2 is upregulated in LCMV-infected Prf1(-/-) mice. Blockade of ST2 markedly improves survival of LCMV-infected Prf1(-/-) mice and reduces the severity of multiple disease parameters, including serum levels of IFNγ. This decrease in IFNγ corresponds to a reduction in both the frequency of IFNγ(+) LCMV-specific CD8(+) and CD4(+) T cells and the magnitude of IFNγ expression in these cells. These findings demonstrate that disruption of ST2 signaling in the murine model of FHL reduces T cell-mediated production of IFNγ and suggest a revised paradigm in which danger signals such as IL-33 are crucial amplifiers of immune dysregulation in FHL. Furthermore, this study provides evidence to support blockade of ST2 as a novel therapeutic strategy for FHL.
Collapse
|
114
|
Wang X, Zhao X, Feng C, Weinstein A, Xia R, Wen W, Lv Q, Zuo S, Tang P, Yang X, Chen X, Wang H, Zang S, Stollings L, Denning TL, Jiang J, Fan J, Zhang G, Zhang X, Zhu Y, Storkus W, Lu B. IL-36γ Transforms the Tumor Microenvironment and Promotes Type 1 Lymphocyte-Mediated Antitumor Immune Responses. Cancer Cell 2015; 28:296-306. [PMID: 26321222 PMCID: PMC4573903 DOI: 10.1016/j.ccell.2015.07.014] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 01/11/2015] [Accepted: 07/27/2015] [Indexed: 12/21/2022]
Abstract
Cytokines play a pivotal role in regulating tumor immunogenicity and antitumor immunity. IL-36γ is important for the IL-23/IL-17-dominated inflammation and anti-BCG Th1 immune responses. However, the impact of IL-36γ on tumor immunity is unknown. Here we found that IL-36γ stimulated CD8(+) T cells, NK cells, and γδ T cells synergistically with TCR signaling and/or IL-12. Importantly, IL-36γ exerted profound antitumor effects in vivo and transformed the tumor microenvironment in favor of tumor eradication. Furthermore, IL-36γ strongly increased the efficacy of tumor vaccination. Moreover, IL-36γ expression inversely correlated with the progression of human melanoma and lung cancer. Our study establishes a role of IL-36γ in promoting antitumor immune responses and suggests its potential clinical translation into cancer immunotherapy.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Biochemistry and Molecular Biology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China; Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Xin Zhao
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Chao Feng
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Aliyah Weinstein
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Dermatology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Rui Xia
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Wen Wen
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Quansheng Lv
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Shuting Zuo
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Peijun Tang
- Department of Pulmonary Tuberculosis, The Affiliated Hospital for Infectious Diseases of Soochow University, Suzhou 215007, China
| | - Xi Yang
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; School of Medicine, Tsinghua University, Peking 100084, China
| | - Xiaojuan Chen
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Hongrui Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Shayang Zang
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Lindsay Stollings
- Department of Anesthesiology, University of Pittsburgh Medical Center, 3471 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Timothy L Denning
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Jingting Jiang
- Department of Tumor Biotherapy, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh PA 15240, USA
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xueguang Zhang
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Yibei Zhu
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Walter Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Dermatology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA.
| |
Collapse
|
115
|
Pinto SM, Nirujogi RS, Rojas PL, Patil AH, Manda SS, Subbannayya Y, Roa JC, Chatterjee A, Prasad TSK, Pandey A. Quantitative phosphoproteomic analysis of IL-33-mediated signaling. Proteomics 2015; 15:532-44. [PMID: 25367039 DOI: 10.1002/pmic.201400303] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/25/2014] [Accepted: 10/28/2014] [Indexed: 12/13/2022]
Abstract
Interleukin-33 (IL-33) is a novel member of the IL-1 family of cytokines that plays diverse roles in the regulation of immune responses. IL-33 exerts its effects through a heterodimeric receptor complex resulting in the production and release of proinflammatory cytokines. A detailed understanding of the signaling pathways activated by IL-33 is still unclear. To gain insights into the IL-33-mediated signaling mechanisms, we carried out a SILAC-based global quantitative phosphoproteomic analysis that resulted in the identification of 7191 phosphorylation sites derived from 2746 proteins. We observed alterations in the level of phosphorylation in 1050 sites corresponding to 672 proteins upon IL-33 stimulation. We report, for the first time, phosphorylation of multiple protein kinases, including mitogen-activated protein kinase activated protein kinase 2 (Mapkapk2), receptor (TNFRSF) interacting serine-threonine kinase 1 (Ripk1), and NAD kinase (Nadk) that are induced by IL-33. In addition, we observed IL-33-induced phosphorylation of several protein phosphatases including protein tyrosine phosphatase, nonreceptor-type 12 (Ptpn12), and inositol polyphosphate-5-phosphatase D (Inpp5d), which have not been reported previously. Network analysis revealed an enrichment of actin binding and cytoskeleton reorganization that could be important in macrophage activation induced by IL-33. Our study is the first quantitative analysis of IL-33-regulated phosphoproteome. Our findings significantly expand the understanding of IL-33-mediated signaling events and have the potential to provide novel therapeutic targets pertaining to immune-related diseases such as asthma where dysregulation of IL-33 is observed. All MS data have been deposited in the ProteomeXchange with identifier PXD000984 (http://proteomecentral.proteomexchange.org/dataset/PXD000984).
Collapse
Affiliation(s)
- Sneha M Pinto
- Institute of Bioinformatics, International Technology Park, Bangalore, India; Manipal University, Madhava Nagar, Manipal, India; McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Potential role of the IL-33/ST2 axis in celiac disease. Cell Mol Immunol 2015; 14:285-292. [PMID: 26343805 DOI: 10.1038/cmi.2015.85] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/10/2015] [Accepted: 08/10/2015] [Indexed: 11/09/2022] Open
Abstract
The IL-33/ST2 axis has been implicated in the pathogenesis of several tissue-specific autoimmune diseases. Celiac disease (CD) is the only autoimmune disease in which both the major genetic factors (HLA-DQ2/DQ8) and etiologic ones (dietary gluten) for susceptibility are known. We have measured serum levels and determined intestinal tissue expression of IL-33 and its receptor soluble ST2 in patients with CD to investigate their association with disease activity. Serum and tissue levels of both IL-33 and sST2 were significantly higher in patients with CD compared with those in control patients without CD. We show that toxic peptides extracted from barley and wheat gliadin significantly stimulate the production of IL-33 and ST2 in cultured peripheral blood mononuclear cell from celiac patients, strongly implicating the IL-33/ST2 axis in the pathogenesis of CD. The higher levels of IL-33 and its receptor ST2 in tissue and serum reflect an active inflammatory state and may represent a potential biomarker for disease activity. A better understanding of IL-33/ST2 release, mode of action, and regulation will be crucial to develop therapeutics that target the IL-33/ST2 pathway to treat CD.Cellular & Molecular Immunology advance online publication, 7 September 2015; doi:10.1038/cmi.2015.85.
Collapse
|
117
|
Molofsky AB, Savage AK, Locksley RM. Interleukin-33 in Tissue Homeostasis, Injury, and Inflammation. Immunity 2015; 42:1005-19. [PMID: 26084021 DOI: 10.1016/j.immuni.2015.06.006] [Citation(s) in RCA: 487] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Indexed: 12/12/2022]
Abstract
Interleukin-33 (IL-33) is a nuclear-associated cytokine of the IL-1 family originally described as a potent inducer of allergic type 2 immunity. IL-33 signals via the receptor ST2, which is highly expressed on group 2 innate lymphoid cells (ILC2s) and T helper 2 (Th2) cells, thus underpinning its association with helminth infection and allergic pathology. Recent studies have revealed ST2 expression on subsets of regulatory T cells, and for a role for IL-33 in tissue homeostasis and repair that suggests previously unrecognized interactions within these cellular networks. IL-33 can participate in pathologic fibrotic reactions, or, in the setting of microbial invasion, can cooperate with inflammatory cytokines to promote responses by cytotoxic NK cells, Th1 cells, and CD8(+) T cells. Here, we highlight the regulation and function of IL-33 and ST2 and review their roles in homeostasis, damage, and inflammation, suggesting a conceptual framework for future studies.
Collapse
Affiliation(s)
- Ari B Molofsky
- Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA; Department of Laboratory Medicine, University of California, San Francisco, 94143-0795, USA
| | - Adam K Savage
- Howard Hughes Medical Institute, University of California, San Francisco, 94143-0795, USA; Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA
| | - Richard M Locksley
- Howard Hughes Medical Institute, University of California, San Francisco, 94143-0795, USA; Department of Medicine, University of California, San Francisco, 94143-0795, USA; Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA.
| |
Collapse
|
118
|
Rose WA, Okragly AJ, Patel CN, Benschop RJ. IL-33 released by alum is responsible for early cytokine production and has adjuvant properties. Sci Rep 2015; 5:13146. [PMID: 26272855 PMCID: PMC4536651 DOI: 10.1038/srep13146] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/21/2015] [Indexed: 11/09/2022] Open
Abstract
Human vaccines have used aluminium-based adjuvants (alum) for >80 years despite incomplete understanding of how alum enhances the immune response. Alum can induce the release of endogenous danger signals via cellular necrosis which elicits inflammation-associated cytokines resulting in humoral immunity. IL-33 is proposed to be one such danger signal that is released from necrotic cells. Therefore, we investigated whether there is a role for IL-33 in the adjuvant activity of alum. We show that alum-induced cellular necrosis results in elevated levels of IL-33 following injection in vivo. Alum and IL-33 induce similar increases in IL-5, KC, MCP-1, MIP-1α and MIP-1β; many of which are dependent on IL-33 as shown in IL-33 knockout mice or by using an IL-33-neutralizing recombinant ST2 receptor. Furthermore, IL-33 itself functions as an adjuvant that, while only inducing a marginal primary response, facilitates a robust secondary response comparable to that observed with alum. However, IL-33 is not absolutely required for alum-induced antibody responses since alum mediates similar humoral responses in IL-33 knockout and wild-type mice. Our results provide novel insights into the mechanism of action behind alum-induced cytokine responses and show that IL-33 is sufficient to provide a robust secondary antibody response independently of alum.
Collapse
Affiliation(s)
- William A Rose
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285
| | - Angela J Okragly
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285
| | - Chetan N Patel
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285
| | - Robert J Benschop
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285
| |
Collapse
|
119
|
Gajardo Carrasco T, Morales RA, Pérez F, Terraza C, Yáñez L, Campos-Mora M, Pino-Lagos K. Alarmin' Immunologists: IL-33 as a Putative Target for Modulating T Cell-Dependent Responses. Front Immunol 2015; 6:232. [PMID: 26082774 PMCID: PMC4451696 DOI: 10.3389/fimmu.2015.00232] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/29/2015] [Indexed: 12/23/2022] Open
Abstract
IL-33 is a known member of the IL-1 cytokine superfamily classically named “atypical” due to its diverse functions. The receptor for this cytokine is the ST2 chain (or IL-1RL1), part of the IL-1R family, and the accessory chain IL-1R. ST2 can be found as both soluble and membrane-bound forms, property that explains, at least in part, its wide range of functions. IL-33 has increasingly gained our attention as a potential target to modulate immune responses. At the beginning, it was known as one of the participants during the development of allergic states and other Th2-mediated responses and it is now accepted that IL-33 contributes to Th1-driven pathologies as demonstrated in animal models of experimental autoimmune encephalomyelitis (EAE), collagen-induced arthritis, and trinitrobenzene sulfonic acid-induced experimental colitis, among others. Interestingly, current data are placing IL-33 as a novel regulator of immune tolerance by affecting regulatory T cells (Tregs); although the mechanism is not fully understood, it seems that dendritic cells and myeloid suppressor-derived cells may be cooperating in the generation and/or establishment of IL-33-mediated tolerance. Here, we review the most updated literature on IL-33, its role on T cell biology, and its impact in immune tolerance.
Collapse
Affiliation(s)
- Tania Gajardo Carrasco
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes , Santiago , Chile
| | | | - Francisco Pérez
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Claudia Terraza
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes , Santiago , Chile
| | - Luz Yáñez
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes , Santiago , Chile
| | - Mauricio Campos-Mora
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes , Santiago , Chile
| | - Karina Pino-Lagos
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes , Santiago , Chile
| |
Collapse
|
120
|
Abstract
Atopic dermatitis is a very prevalent disease that affects children as well as adults. The disease has a severe impact on quality of life for the patients and their families. The skin in atopic dermatitis patients is a site of both a severe inflammatory reaction dominated by lymphocytes and decreased skin barrier function. The treatment of the disease is mainly aimed at reducing the inflammation in the skin and/or restoring the skin barrier function. However, most of the treatments used today singularly aim at reducing the inflammation in the skin. Depending on the severity of the disease, the anti-inflammatory treatment may be topical or systemic, but basic treatment, no matter the severity, should always be emollients. In addition, new studies have shown good effects of psychosocial interventions, such as eczema schools, for patients and their families. This review covers the latest trends in the treatment of atopic dermatitis.
Collapse
|
121
|
Li FF, Wang RJ, Pan HX, Wu YL, Li YW. Dynamic evaluation of a mouse model of DSS induced ulcerative colitis. Shijie Huaren Xiaohua Zazhi 2015; 23:1721-1727. [DOI: 10.11569/wcjd.v23.i11.1721] [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
AIM: To develop an experimental mouse model of ulcerative colitis (UC) and to dynamically monitor the colonic injures, pathological changes, and contents of interleukin (IL)-1β, IL-18, and IL-33 in plasma and colonic mucosa to evaluate the model.
METHODS: Sixty-four C57BL/6 mice were randomly divided into either a normal group or a model group. The model group was fed 3% dextran sulfate sodium (DSS) solution for a week, followed by normal drinking water for another 3 wk. We then dynamically observed fecal condition, alterations of colon length and weight, pathological changes, colonic mucosa inflammation score and contents of IL-1β, IL-18, and IL-33 in plasma and colonic mucosa every week.
RESULTS: One week after modeling, the mice developed diarrhea, bloody stools, crouching, and huddled together. Pathological examination showed colon epithelial erosion, bleeding, multifocal ulcers and numerous inflammation cells infiltrating the mucosa and submucosa. Compared to the normal group, the colon weight was markedly increased (P < 0.05), the colon length was significantly shortened (P < 0.01), the colonic mucosa inflammation score was significantly increased (P < 0.01), and the contents of IL-1β, IL-18, and IL-33 in both plasma and mucosa were significantly elevated (P < 0.05, P < 0.01) in the model group. At the end of the second week, compared to the normal group, the model group had gradually decreased bleeding, a large number of inflammatory cells in the submucosa, increased colon weight (P < 0.01), shortened colon length (P < 0.01), increased colonic mucosa inflammation score (P < 0.01), and dramatically elevated contents of IL-1β and IL-18 in plasma and IL-18 in colonic mucosa (P < 0.05). At the third and fourth weeks, the mouse activity and fecal condition returned to normal, inflammation cells could be seen in colonic mucosa, muscle fiber was arranged disorderly; compared to normal group, colon weight was increased (P < 0.01), colon length was shortened (P < 0.01), but colonic mucosa inflammation score and the contents of IL-1β, IL-18, and IL-33 showed no significant changes in the model group (P > 0.05).
CONCLUSION: The dynamic changes in the mouse UC model show a shift from acute to chronic disease process, and this mouse model could be used for studying UC mechanism and treatment in the future.
Collapse
|
122
|
Villarreal DO, Svoronos N, Wise MC, Shedlock DJ, Morrow MP, Conejo-Garcia JR, Weiner DB. Molecular adjuvant IL-33 enhances the potency of a DNA vaccine in a lethal challenge model. Vaccine 2015; 33:4313-20. [PMID: 25887087 DOI: 10.1016/j.vaccine.2015.03.086] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 02/26/2015] [Accepted: 03/25/2015] [Indexed: 12/25/2022]
Abstract
Identifying new molecular adjuvants that elicit effective vaccine-induced CD8(+) T cell immunity may be critical for the elimination of many challenging diseases including Tuberculosis, HIV and cancer. Here, we report that co-administration of molecular adjuvant IL-33 during vaccination enhanced the magnitude and function of antigen (Ag)-specific CD8(+) T cells against a model Ag, LCMV NP target protein. These enhanced responses were characterized by higher frequencies of Ag-specific, polyfunctional CD8(+) T cells exhibiting cytotoxic characteristics. Importantly, these cells were capable of robust expansion upon Ag-specific restimulation in vivo and conferred remarkable protection against a high dose lethal LCMV challenge. In addition, we demonstrate the ability of IL-33 to amplifying the frequency of Ag-specific KLRG1(+) effector CD8(+) T cells. These data show that IL-33 is a promising immunoadjuvant at improving T cell immunity in a vaccine setting and suggest further development and understanding of this molecular adjuvant for strategies against many obstinate infectious diseases and cancer.
Collapse
Affiliation(s)
- Daniel O Villarreal
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nikolaos Svoronos
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Tumor Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Megan C Wise
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Devon J Shedlock
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew P Morrow
- Inovio Pharmaceuticals, Inc., 660 West Germantown Pike, Suite 110, Plymouth Meeting, PA 19462, USA
| | - Jose R Conejo-Garcia
- Tumor Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - David B Weiner
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
123
|
The IL-33/ST2 axis augments effector T-cell responses during acute GVHD. Blood 2015; 125:3183-92. [PMID: 25814531 DOI: 10.1182/blood-2014-10-606830] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/11/2015] [Indexed: 12/19/2022] Open
Abstract
Interleukin (IL)-33 binding to the receptor suppression of tumorigenicity 2 (ST2) produces pro-inflammatory and anti-inflammatory effects. Increased levels of soluble ST2 (sST2) are a biomarker for steroid-refractory graft-versus-host disease (GVHD) and mortality. However, whether sST2 has a role as an immune modulator or only as a biomarker during GVHD was unclear. We show increased IL-33 production by nonhematopoietic cells in the gastrointestinal (GI) tract in mice post-conditioning and patients during GVHD. Exogenous IL-33 administration during the peak inflammatory response worsened GVHD. Conversely, GVHD lethality and tumor necrosis factor-α production was significantly reduced in il33(-/-) recipients. ST2 was upregulated on murine and human alloreactive T cells and sST2 increased as experimental GVHD progressed. Concordantly, st2(-/-) vs wild-type (WT) donor T cells had a marked reduction in GVHD lethality and GI histopathology. Alloantigen-induced IL-18 receptor upregulation was lower in st2(-/-) T cells, and linked to reduced interferon-γ production by st2(-/-) vs WT T cells during GVHD. Blockade of IL-33/ST2 interactions during allogeneic-hematopoietic cell transplantation by exogenous ST2-Fc infusions had a marked reduction in GVHD lethality, indicating a role of ST2 as a decoy receptor modulating GVHD. Together, these studies point to the IL-33/ST2 axis as a novel and potent target for GVHD therapy.
Collapse
|
124
|
Lott JM, Sumpter TL, Turnquist HR. New dog and new tricks: evolving roles for IL-33 in type 2 immunity. J Leukoc Biol 2015; 97:1037-48. [DOI: 10.1189/jlb.3ri1214-595r] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/23/2015] [Indexed: 12/25/2022] Open
|
125
|
Palomo J, Reverchon F, Piotet J, Besnard AG, Couturier-Maillard A, Maillet I, Tefit M, Erard F, Mazier D, Ryffel B, Quesniaux VFJ. Critical role of IL-33 receptor ST2 in experimental cerebral malaria development. Eur J Immunol 2015; 45:1354-65. [PMID: 25682948 DOI: 10.1002/eji.201445206] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 01/19/2015] [Accepted: 02/11/2015] [Indexed: 11/11/2022]
Abstract
Cerebral malaria, a severe complication of Plasmodium falciparum infection, can be modeled in murine Plasmodium berghei ANKA (PbA) infection. PbA-induced experimental cerebral malaria (ECM) is CD8(+) T-cell mediated, and influenced by TH 1/TH 2 balance. Here, we show that IL-33 expression is increased in brain undergoing ECM and we address the role of the IL-33/ST2 pathway in ECM development. ST2-deficient mice were resistant to PbA-induced neuropathology. They survived >20 days with no ECM neurological sign and a preserved cerebral microcirculation, while WT mice succumbed within 10 days with ECM, brain vascular leakage, distinct microvascular pathology obstruction, and hemorrhages. Parasitemia and brain parasite load were similar in ST2-deficient and WT mice. Protection was accompanied by reduced brain sequestration of activated CD4(+) T cells and perforin(+) CD8(+) T cells. While IFN-γ and T-cell-attracting chemokines CXCL9 and CXCL10 were not affected in the absence of functional ST2 pathway, the local expression of ICAM-1, CXCR3, and LT-α, crucial for ECM development, was strongly reduced, and this may explain the diminished pathogenic T-cell recruitment and resistance to ECM. Therefore, IL-33 is induced in PbA sporozoite infection, and the pathogenic T-cell responses with local microvascular pathology are dependent on IL-33/ST2 signaling, identifying IL-33 as a new actor in ECM development.
Collapse
Affiliation(s)
- Jennifer Palomo
- CNRS, UMR7355, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Flora Reverchon
- CNRS, UMR7355, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Julie Piotet
- CNRS, UMR7355, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Anne-Gaelle Besnard
- CNRS, UMR7355, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Aurélie Couturier-Maillard
- CNRS, UMR7355, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Isabelle Maillet
- CNRS, UMR7355, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Maurel Tefit
- CIMI-Paris (UPMC UMRS CR7, Inserm U1135, CNRS ERL 8255), Paris, France
| | - François Erard
- CNRS, UMR7355, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Dominique Mazier
- CIMI-Paris (UPMC UMRS CR7, Inserm U1135, CNRS ERL 8255), Paris, France.,Groupe Hospitalier Pitié-Salpêtrière Service Parasitologie-Mycologie, Paris, France
| | - Bernhard Ryffel
- CNRS, UMR7355, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Valérie F J Quesniaux
- CNRS, UMR7355, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| |
Collapse
|
126
|
T-bet- and STAT4-dependent IL-33 receptor expression directly promotes antiviral Th1 cell responses. Proc Natl Acad Sci U S A 2015; 112:4056-61. [PMID: 25829541 DOI: 10.1073/pnas.1418549112] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
During infection, the release of damage-associated molecular patterns, so-called "alarmins," orchestrates the immune response. The alarmin IL-33 plays a role in a wide range of pathologies. Upon release, IL-33 signals through its receptor ST2, which reportedly is expressed only on CD4(+) T cells of the Th2 and regulatory subsets. Here we show that Th1 effector cells also express ST2 upon differentiation in vitro and in vivo during lymphocytic choriomeningitis virus (LCMV) infection. The expression of ST2 on Th1 cells was transient, in contrast to constitutive ST2 expression on Th2 cells, and marked highly activated effector cells. ST2 expression on virus-specific Th1 cells depended on the Th1-associated transcription factors T-bet and STAT4. ST2 deficiency resulted in a T-cell-intrinsic impairment of LCMV-specific Th1 effector responses in both mixed bone marrow-chimeric mice and adoptive cell transfer experiments. ST2-deficient virus-specific CD4(+) T cells showed impaired expansion, Th1 effector differentiation, and antiviral cytokine production. Consequently, these cells mediated little virus-induced immunopathology. Thus, IL-33 acts as a critical and direct cofactor to drive antiviral Th1 effector cell activation, with implications for vaccination strategies and immunotherapeutic approaches.
Collapse
|
127
|
Kearley J, Silver JS, Sanden C, Liu Z, Berlin AA, White N, Mori M, Pham TH, Ward CK, Criner GJ, Marchetti N, Mustelin T, Erjefalt JS, Kolbeck R, Humbles AA. Cigarette smoke silences innate lymphoid cell function and facilitates an exacerbated type I interleukin-33-dependent response to infection. Immunity 2015; 42:566-79. [PMID: 25786179 DOI: 10.1016/j.immuni.2015.02.011] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 12/29/2014] [Accepted: 01/15/2015] [Indexed: 10/23/2022]
Abstract
Cigarette smoking is a major risk factor for chronic obstructive pulmonary disease and is presumed to be central to the altered responsiveness to recurrent infection in these patients. We examined the effects of smoke priming underlying the exacerbated response to viral infection in mice. Lack of interleukin-33 (IL-33) signaling conferred complete protection during exacerbation and prevented enhanced inflammation and exaggerated weight loss. Mechanistically, smoke was required to upregulate epithelial-derived IL-33 and simultaneously alter the distribution of the IL-33 receptor ST2. Specifically, smoke decreased ST2 expression on group 2 innate lymphoid cells (ILC2s) while elevating ST2 expression on macrophages and natural killer (NK) cells, thus altering IL-33 responsiveness within the lung. Consequently, upon infection and release, increased local IL-33 significantly amplified type I proinflammatory responses via synergistic modulation of macrophage and NK cell function. Therefore, in COPD, smoke alters the lung microenvironment to facilitate an alternative IL-33-dependent exaggerated proinflammatory response to infection, exacerbating disease.
Collapse
MESH Headings
- Animals
- Female
- Gene Expression Regulation
- Humans
- Immunity, Innate/drug effects
- Influenza A virus/immunology
- Interleukin-1 Receptor-Like 1 Protein
- Interleukin-33
- Interleukins/deficiency
- Interleukins/genetics
- Interleukins/immunology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Lung/drug effects
- Lung/immunology
- Lung/pathology
- Lymphocytes/drug effects
- Lymphocytes/immunology
- Lymphocytes/pathology
- Macrophages/immunology
- Macrophages/pathology
- Mice, Transgenic
- Orthomyxoviridae Infections/etiology
- Orthomyxoviridae Infections/genetics
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/pathology
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/immunology
- Pulmonary Disease, Chronic Obstructive/pathology
- Receptors, Interleukin/deficiency
- Receptors, Interleukin/genetics
- Receptors, Interleukin/immunology
- Respiratory Mucosa/drug effects
- Respiratory Mucosa/immunology
- Respiratory Mucosa/pathology
- Signal Transduction
- Smoke/adverse effects
- Nicotiana/chemistry
- Weight Loss
Collapse
Affiliation(s)
- Jennifer Kearley
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Jonathan S Silver
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Caroline Sanden
- Department of Experimental Medical Science, Lund University, Lund 22184, Sweden
| | - Zheng Liu
- Department of Translational Sciences, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Aaron A Berlin
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Natalie White
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Michiko Mori
- Department of Experimental Medical Science, Lund University, Lund 22184, Sweden
| | - Tuyet-Hang Pham
- Department of Translational Sciences, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Christine K Ward
- Department of Translational Sciences, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Gerard J Criner
- Pulmonary and Critical Care Medicine, Department of Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Nathaniel Marchetti
- Pulmonary and Critical Care Medicine, Department of Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Tomas Mustelin
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Jonas S Erjefalt
- Department of Experimental Medical Science, Lund University, Lund 22184, Sweden
| | - Roland Kolbeck
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Alison A Humbles
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, MD 20878, USA.
| |
Collapse
|
128
|
Lu J, Kang J, Zhang C, Zhang X. The role of IL-33/ST2L signals in the immune cells. Immunol Lett 2015; 164:11-7. [DOI: 10.1016/j.imlet.2015.01.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/26/2014] [Accepted: 01/27/2015] [Indexed: 12/18/2022]
|
129
|
Tong X, Lu F. IL-33/ST2 involves the immunopathology of ocular toxoplasmosis in murine model. Parasitol Res 2015; 114:1897-905. [PMID: 25693767 DOI: 10.1007/s00436-015-4377-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 02/04/2015] [Indexed: 12/12/2022]
Abstract
Ocular toxoplasmosis (OT) is the major cause of infective uveitis. Since the eye is a special organ protected by immune privilege, its immune response is different from general organs with Toxoplasma gondii infection. Here, we used Kunming outbred mice to establish OT by intravitreal injection of T. gondii RH strain tachyzoites, IL-33 expression in the eyes was localized by immunostaining, the levels of interleukin (IL)-33 and ST2 (IL-33 receptor) and T-helper (Th)1 and Th2-associated cytokines in the eye and cervical lymph nodes (CLNs) of infected mice were measured, and their correlations were analyzed. Our results showed that the pathologies of the eye and CLN tissues and the IL-33 positive cells in the eye tissues of ocular T. gondii-infected mice were all increased at days 2, 6, and 9 postinfection (p.i.), accompanied with significantly increased transcript levels of IL-33, ST2, IL-1β, IFN-γ, IL-12p40, IL-10, and IL-13 in both the eyes and CLNs, and increased IL-4 expressions in the eyes of T. gondii-infected mice. There were significant correlations between the levels of IFN-γ and ST2, IL-4 and ST2, and IL-13 and ST2 in the eye tissues (P < 0.001), significant correlations between the levels of IFN-γ and ST2 (P < 0.001) as well as between IL-13 and ST2 (P < 0.05) in the CLNs, and significant correlations between the levels of IL-1β and IL-33 in the eyes (P < 0.05) and between IL-1β and IL-33/ST2 in the CLNs (P < 0.001 and P < 0.01, respectively). Our data indicated that IL-33/ST2 may involve the regulation of ocular immunopathology induced by T. gondii infection.
Collapse
Affiliation(s)
- Xinxin Tong
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China
| | | |
Collapse
|
130
|
Abstract
The identification and characterization of cytokine isoforms is likely to provide critical important new insight into immunobiology. Cytokine isoforms can provide additional diversity to their complex biological effects that participate in control and protection against different foreign pathogens. Recently, IL-33 has been identified as a proinflammatory cytokine having several different biologically active isoform products. Originally associated with Th2 immunity, new evidence now supports the role of two IL-33 isoforms to facilitate the generation of protective Th1 and CD8 T cell immunity against specific pathogens. Therefore, a better understanding of the IL-33 isoforms will inform us on how to utilize them to facilitate their development as tools as vaccine adjuvants for immune therapy.
Collapse
Affiliation(s)
- Daniel O Villarreal
- Department of Pathology, University of Pennsylvania, 505A Stellar-Chance Laboratories, 422 Curie Blvd, Philadelphia, PA 19104, USA
| | | |
Collapse
|
131
|
Xia J, Zhao J, Shang J, Li M, Zeng Z, Zhao J, Wang J, Xu Y, Xie J. Increased IL-33 expression in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2015; 308:L619-27. [PMID: 25595648 DOI: 10.1152/ajplung.00305.2014] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/15/2015] [Indexed: 01/13/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an inflammatory lung disease characterized by inflammatory cell activation and the release of inflammatory mediators. Interleukin-33 (IL-33) plays a critical role in various inflammatory and immunological pathologies, but evidence for its role in COPD is lacking. This study aimed to investigate the expression of IL-33 in COPD and to determine whether IL-33 participates in the initiation and progression of COPD. Levels of serum IL-33 and its receptors were measured by ELISA, and serum levels of IL-33, ST2, and IL-1 receptor accessory protein were elevated in patients with COPD compared with control subjects. Flow cytometry analysis further demonstrated an increase in peripheral blood lymphocytes (PBLs) expressing IL-33 in patients with COPD. Immunofluorescence analysis revealed that the main cellular source of IL-33 in lung tissue was human bronchial epithelial cells (HBEs). Cigarette smoke extract and lipopolysaccharide could enhance the ability of PBLs and HBEs to express IL-33. Furthermore, PBLs from patients with COPD showed greater IL-33 release in response to the stimulus. Collectively, these findings suggest that IL-33 expression levels are increased in COPD and related to airway and systemic inflammation. Therefore, IL-33 might contribute to the pathogenesis and progression of this disease.
Collapse
Affiliation(s)
- Jie Xia
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junling Zhao
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Shang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Miao Li
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhilin Zeng
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianmiao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongjian Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
132
|
Naumnik W, Naumnik B, Niklińska W, Ossolińska M, Chyczewska E. Interleukin-33 as a New Marker of Pulmonary Sarcoidosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 866:1-6. [DOI: 10.1007/5584_2015_142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
133
|
Ikutani M, Tsuneyama K, Nakae S, Takatsu K. Emerging roles of IL-33 in inflammation and immune regulation. Inflamm Regen 2015. [DOI: 10.2492/inflammregen.35.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Masashi Ikutani
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Koichi Tsuneyama
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Susumu Nakae
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Takatsu
- Toyama Prefectural Institute for Pharmaceutical Research, Toyama, Japan
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| |
Collapse
|
134
|
Kunes P, Mandak J, Holubcova Z, Kolackova M, Krejsek J. Actual position of interleukin(IL)-33 in atherosclerosis and heart failure: Great Expectations or En attendant Godot? Perfusion 2014; 30:356-74. [DOI: 10.1177/0267659114562269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Atherosclerosis has been recognized as an inflammatory/autoimmune disease. The long-standing low-grade inflammation which fuels its development is primarily focused on the components of the vessel wall. Originally, inflammation in atherogenesis was supposed to be driven by the pro-inflammatory Th1 cellular and cytokine immune response. On the basis of accumulating evidence, this view has been re-evaluated to include the Th17/Th1 axis which is shared by most diseases of sterile inflammation. The anti-inflammatory Th2 cellular and cytokine immune response is initiated concomitantly with the former two, the latter dampening their harmful reactions which culminate in full-blown atherosclerosis. Interleukin-33, a novel member of the IL-1 cytokine superfamily, was suggested to take part in the anti-atherogenic response by mediating the Th1-to-Th2 switch of the immune reactions. However, IL-33 is a multifaceted mediator with both pro- and anti-inflammatory activities, also called a “dual factor” or a “Janus face” interleukin. IL-33 occurs both in an extracellular (cytokine-like) and in a nuclear-bound (transcription factor-like) form, each of them performing distinct activities of their own. This review article presents the latest data relevant to IL-33’s role in atherosclerosis and cardiac diseases as perceived by a cardiologist and a cardiac surgeon.
Collapse
Affiliation(s)
- P Kunes
- Department of Cardiac Surgery, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - J Mandak
- Department of Cardiac Surgery, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - Z Holubcova
- Department of Cardiac Surgery, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - M Kolackova
- Department of Clinical Immunology and Allergology, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - J Krejsek
- Department of Clinical Immunology and Allergology, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| |
Collapse
|
135
|
Gao X, Wang X, Yang Q, Zhao X, Wen W, Li G, Lu J, Qin W, Qi Y, Xie F, Jiang J, Wu C, Zhang X, Chen X, Turnquist H, Zhu Y, Lu B. Tumoral expression of IL-33 inhibits tumor growth and modifies the tumor microenvironment through CD8+ T and NK cells. THE JOURNAL OF IMMUNOLOGY 2014; 194:438-45. [PMID: 25429071 DOI: 10.4049/jimmunol.1401344] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy has shown great promise as a new standard cancer therapeutic modality. However, the response rates are limited for current approach that depends on enhancing spontaneous antitumor immune responses. Therefore, increasing tumor immunogenicity by expressing appropriate cytokines should further improve the current immunotherapy. IL-33 is a member of the IL-1 family of cytokines and is released by necrotic epithelial cells or activated innate immune cells and is thus considered a "danger" signal. The role of IL-33 in promoting type 2 immune responses and tissue inflammation has been well established. However, whether IL-33 drives antitumor immune responses is controversial. Our previous work established that IL-33 promoted the function of CD8(+) T cells. In this study, we showed that the expression of IL-33 in two types of cancer cells potently inhibited tumor growth and metastasis. Mechanistically, IL-33 increased numbers and IFN-γ production by CD8(+) T and NK cells in tumor tissues, thereby inducing a tumor microenvironment favoring tumor eradication. Importantly, IL-33 greatly increased tumor Ag-specific CD8(+) T cells. Furthermore, both NK and CD8(+) T cells were required for the antitumor effect of IL-33. Moreover, depletion of regulatory T cells worked synergistically with IL-33 expression for tumor elimination. Our studies established "alarmin" IL-33 as a promising new cytokine for tumor immunotherapy through promoting cancer-eradicating type 1 immune responses.
Collapse
Affiliation(s)
- Xin Gao
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Xuefeng Wang
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China; Department of Biochemistry and Molecular Biology, Soochow University, Suzhou 215007, People's Republic of China
| | - Qianting Yang
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Key Laboratory of Infection and Immunity, Third People's Hospital, Guangdong Medical College, Shenzhen, Guangdong 518112, China
| | - Xin Zhao
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; The First Affiliated Hospital, Soochow University, Suzhou 215006, People's Republic of China
| | - Wen Wen
- Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Gang Li
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Junfeng Lu
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Haidian District, Beijing 100190, People's Republic of China
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Yuan Qi
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Fang Xie
- Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Jingting Jiang
- The Third Affiliated Hospital, Soochow University, Changzhou 213003, China
| | - Changping Wu
- The Third Affiliated Hospital, Soochow University, Changzhou 213003, China
| | - Xueguang Zhang
- Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Xinchun Chen
- Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Key Laboratory of Infection and Immunity, Third People's Hospital, Guangdong Medical College, Shenzhen, Guangdong 518112, China
| | - Heth Turnquist
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; and
| | - Yibei Zhu
- Department of Immunology, Institute of Medical Biotechnology, Soochow University, Suzhou 215007, People's Republic of China
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232
| |
Collapse
|
136
|
Matta BM, Lott JM, Mathews LR, Liu Q, Rosborough BR, Blazar BR, Turnquist HR. IL-33 is an unconventional Alarmin that stimulates IL-2 secretion by dendritic cells to selectively expand IL-33R/ST2+ regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:4010-20. [PMID: 25217167 DOI: 10.4049/jimmunol.1400481] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
IL-33 is a recently characterized IL-1 family member that is proposed to function as an alarmin, or endogenous signal of cellular damage, as well as act as a pleiotropic cytokine. The ability of IL-33 to potentiate both Th1 and Th2 immunity supports its role in pathogen clearance and disease immunopathology. Yet, IL-33 restrains experimental colitis and transplant rejection by expanding regulatory T cells (Treg) via an undefined mechanism. We sought to determine the influence of IL-33 on hematopoietic cells that drives Treg expansion and underlies the therapeutic benefit of IL-33 administration. In this study, we identify a feedback loop in which conventional mouse CD11c(+) dendritic cells (DC) stimulated by IL-33 secrete IL-2 to selectively expand IL-33R(ST2(+))- suppressive CD4(+)Foxp3(+) Treg. Interestingly, this occurs in the absence of classical DC maturation, and DC-derived (innate) IL-2 increases ST2 expression on both DC and interacting Treg. ST2(+) Treg represent an activated subset of Foxp3(+) cells, demonstrated to be ICOS(high)CD44(high) compared with their ST2(-) counterparts. Furthermore, although studies have shown that IL-33-exposed DC promote Th2 responses, we reveal that ST2(+) DC are required for IL-33-mediated in vitro and in vivo Treg expansion. Thus, we have uncovered a relationship between IL-33 and innate IL-2 that promotes the selective expansion of ST2(+) Treg over non-Treg. These findings identify a novel regulatory pathway driven by IL-33 in immune cells that may be harnessed for therapeutic benefit or for robust expansion of Treg in vitro and in vivo.
Collapse
Affiliation(s)
- Benjamin M Matta
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Jeremy M Lott
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Lisa R Mathews
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261
| | - Quan Liu
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Brian R Rosborough
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Graduate Training Program in Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455; and
| | - Hēth R Turnquist
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
137
|
Wang L, Li H, Liang F, Hong Y, Jiang S, Xiao L. Examining IL-33 expression in the cervix of HPV-infected patients: a preliminary study comparing IL-33 levels in different stages of disease and analyzing its potential association with IFN-γ. Med Oncol 2014; 31:143. [PMID: 25106529 DOI: 10.1007/s12032-014-0143-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 07/26/2014] [Indexed: 12/11/2022]
Abstract
Research has shown the essential role of interleukin-33 (IL-33) in driving protective anti-viral immunity. IFN-γ has been reported to improve IL-33 expression in cultured epithelial cells. The development of cervical intraepithelial neoplasia (CIN) and carcinogenesis was closely related to human papilloma virus (HPV) infection and defective anti-viral immunity. The aim of this study was to investigate IL-33 expression alternation during the disease progress and its association with IFN-γ in HPV-positive patients. IL-33 was detected in endothelial cells and populations of epithelial cells in cervix. Though there was no statistically significant difference of IL-33 levels in cervical lavage and serum among different stages of disease (P > 0.05), the IL-33 protein and mRNA levels in cervical tissues were significantly lower in severe CIN patients than that of mild CIN or no CIN patients (P < 0.05). In addition, IL-33 protein levels were positively correlated with IFN-γ mRNA levels in all groups except cervical cancer (CA) group (r = 0.546, P < 0.01). In vitro, IFN-γ was also found to upregulate IL-33 expression in human epidermal keratinocytes (NHEKs) in a dose-dependent manner. However, CA tissues did not show further reduced IL-33 protein and mRNA levels compared with severe CIN tissues (P > 0.05). IFN-γ mRNA levels were even higher in CA tissues than in severe CIN tissues (P < 0.05). Therefore, in cervical precancerous tissues, IL-33 levels were lower in more severe lesions and that may be related to diminished local IFN-γ.
Collapse
Affiliation(s)
- Lei Wang
- Institute of Laboratory Medicine, Hubei University of Chinese Medicine, Huangjia Lake West Road, Wuhan, 430065, Hubei Province, China,
| | | | | | | | | | | |
Collapse
|
138
|
Striz I, Brabcova E, Kolesar L, Sekerkova A. Cytokine networking of innate immunity cells: a potential target of therapy. Clin Sci (Lond) 2014; 126:593-612. [PMID: 24450743 DOI: 10.1042/cs20130497] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Innate immune cells, particularly macrophages and epithelial cells, play a key role in multiple layers of immune responses. Alarmins and pro-inflammatory cytokines from the IL (interleukin)-1 and TNF (tumour necrosis factor) families initiate the cascade of events by inducing chemokine release from bystander cells and by the up-regulation of adhesion molecules required for transendothelial trafficking of immune cells. Furthermore, innate cytokines produced by dendritic cells, macrophages, epithelial cells and innate lymphoid cells seem to play a critical role in polarization of helper T-cell cytokine profiles into specific subsets of Th1/Th2/Th17 effector cells or regulatory T-cells. Lastly, the innate immune system down-regulates effector mechanisms and restores homoeostasis in injured tissue via cytokines from the IL-10 and TGF (transforming growth factor) families mainly released from macrophages, preferentially the M2 subset, which have a capacity to induce regulatory T-cells, inhibit the production of pro-inflammatory cytokines and induce healing of the tissue by regulating extracellular matrix protein deposition and angiogenesis. Cytokines produced by innate immune cells represent an attractive target for therapeutic intervention, and multiple molecules are currently being tested clinically in patients with inflammatory bowel disease, rheumatoid arthritis, systemic diseases, autoinflammatory syndromes, fibrosing processes or malignancies. In addition to the already widely used blockers of TNFα and the tested inhibitors of IL-1 and IL-6, multiple therapeutic molecules are currently in clinical trials targeting TNF-related molecules [APRIL (a proliferation-inducing ligand) and BAFF (B-cell-activating factor belonging to the TNF family)], chemokine receptors, IL-17, TGFβ and other cytokines.
Collapse
Affiliation(s)
| | - Eva Brabcova
- *Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Videnska 1958/9 Prague, Czech Republic
| | - Libor Kolesar
- *Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Videnska 1958/9 Prague, Czech Republic
| | - Alena Sekerkova
- *Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Videnska 1958/9 Prague, Czech Republic
| |
Collapse
|
139
|
Interleukin 33: a switch-hitting cytokine. Curr Opin Immunol 2014; 28:102-6. [PMID: 24762410 DOI: 10.1016/j.coi.2014.03.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 03/19/2014] [Accepted: 03/19/2014] [Indexed: 11/23/2022]
Abstract
For many years IL-33 has been widely studied in the context of T helper type 2 (Th2)-driven inflammatory disorders. Interestingly, IL-33 has now emerged as a cytokine with a plethora of pleiotropic properties. Depending on the immune cells targeted by IL-33, it is reported to not only promote Th2 immunity, but also to induce T helper type 1 (Th1) immunity. Furthermore, recent studies have revealed that IL-33 can activate CD8(+) T cells. These new studies provide evidence for its beneficial role in antiviral and antitumor immunity. Here we review the evidence of IL-33 to drive protective T cell immunity plus its potential use as an adjuvant in vaccination and tumor therapy.
Collapse
|
140
|
Ryba-Stanisławowska M, Stanisławowski M, Myśliwska J. Effector and regulatory T cell subsets in diabetes-associated inflammation. Is there a connection with ST2/IL-33 axis? Perspective. Autoimmunity 2014; 47:361-71. [PMID: 24547981 DOI: 10.3109/08916934.2014.886198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Type 1 diabetes (DM1) is a chronic inflammatory disease, which when progresses leads to the development of late vascular complications. The disease involves impairments in regulatory and effector subsets of T lymphocytes, which suppress and maintain inflammatory response, respectively. ST2/IL-33 pathway is involved in T-cell-mediated immune response and might regulate the inflammatory process in several diseases. This review presents the latest research findings regarding effector and regulatory T cell subsets in the context of inflammation accompanying DM1 with particular focus on the ST2/IL-33 network and its possible association with T cell-mediated immunity.
Collapse
|
141
|
Villarreal DO, Wise MC, Walters JN, Reuschel EL, Choi MJ, Obeng-Adjei N, Yan J, Morrow MP, Weiner DB. Alarmin IL-33 acts as an immunoadjuvant to enhance antigen-specific tumor immunity. Cancer Res 2014; 74:1789-800. [PMID: 24448242 DOI: 10.1158/0008-5472.can-13-2729] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Studies of interleukin (IL)-33 reveal a number of pleiotropic properties. Here, we report that IL-33 has immunoadjuvant effects in a human papilloma virus (HPV)-associated model for cancer immunotherapy where cell-mediated immunity is critical for protection. Two biologically active isoforms of IL-33 exist that are full-length or mature, but the ability of either isoform to function as a vaccine adjuvant that influences CD4 T helper 1 or CD8 T-cell immune responses is not defined. We showed that both IL-33 isoforms are capable of enhancing potent antigen-specific effector and memory T-cell immunity in vivo in a DNA vaccine setting. In addition, although both IL-33 isoforms drove robust IFN-γ responses, neither elevated secretion of IL-4 or immunoglobulin E levels. Further, both isoforms augmented vaccine-induced antigen-specific polyfunctional CD4(+) and CD8(+) T-cell responses, with a large proportion of CD8(+) T cells undergoing plurifunctional cytolytic degranulation. Therapeutic studies indicated that vaccination with either IL-33 isoform in conjunction with an HPV DNA vaccine caused rapid and complete regressions in vivo. Moreover, IL-33 could expand the magnitude of antigen-specific CD8(+) T-cell responses and elicit effector-memory CD8(+) T cells. Taken together, our results support the development of these IL-33 isoforms as immunoadjuvants in vaccinations against pathogens, including in the context of antitumor immunotherapy.
Collapse
Affiliation(s)
- Daniel O Villarreal
- Authors' Affiliations: Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia; Inovio Pharmaceuticals, Inc., Blue Bell, Pennsylvania; and Korea Food and Drug Administration, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Lee HY, Rhee CK, Kang JY, Byun JH, Choi JY, Kim SJ, Kim YK, Kwon SS, Lee SY. Blockade of IL-33/ST2 ameliorates airway inflammation in a murine model of allergic asthma. Exp Lung Res 2014; 40:66-76. [PMID: 24446582 DOI: 10.3109/01902148.2013.870261] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Interleukin (IL)-33 is involved in the development of lung inflammation by inducing or amplifying Th2 type-mediated responses in various animal models of allergic asthma. The ST2 gene is a member of the IL-1 receptor family, producing a transmembrane form (ST2L) and a soluble secreted form (sST2). sST2 has been shown to block this IL-33/ST2 signaling pathway. This study aimed to investigate whether anti-IL-33 and sST2 reduced airway inflammation in a murine model of asthma. METHODS BALB/c mice were sensitized and challenged with ovalbumin (OVA), and the effect of sST2 and anti-IL-33 antibody on airway inflammation and airway hyperresponsiveness (AHR) was evaluated. Furthermore, we measured changes in various cytokines in the bronchoalveolar lavage (BAL) fluid when treated with sST2 or anti-IL-33. RESULTS We observed that anti-IL-33 antibody and sST2 exert a negative regulation on OVA-mediated allergic airway inflammation. Both treatments reduced total cell counts and eosinophil counts in BAL fluid and AHR to methacholine. The Th2 cytokines, such as IL-4, IL-5, and IL-13 in BAL fluid were also significantly decreased after both treatments. However, there were no changes in the level of TGF- ß1 and IL-10 after each treatment. CONCLUSIONS These results suggest that anti-IL-33 as well as sST2 have therapeutic potential for allergic asthma through inhibition of Th2 cytokine production.
Collapse
Affiliation(s)
- Hea Yon Lee
- 1Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Caserta D, Di Benedetto L, Bordi G, D'Ambrosio A, Moscarini M. Levels of Galectin-3 and Stimulation Expressed Gene 2 in the peritoneal fluid of women with endometriosis: a pilot study. Gynecol Endocrinol 2014; 30:877-80. [PMID: 25069762 DOI: 10.3109/09513590.2014.943728] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Endometriosis is a puzzling disorder with obscure pathogenesis. Several studies suggest that peritoneal fluid is a key inflammatory environment in the development and progression of the disease. This study analyzed the levels of two inflammatory factors - Galectin-3 and Stimulation Expressed Gene 2 - in the peritoneal fluid of 15 women affected by endometriosis and 8 controls. The peritoneal fluid was collected during laparoscopic surgery avoiding any form of contamination and it was properly processed and stored. Gal-3 and ST2 peritoneal concentrations were analyzed using enzyme immunoassay kit. Gal-3 levels were significantly higher in endometriosis group than in controls (64.7 ± 52.34 versus 21.05 ± 20.83 ng/ml, p = 0.044), whereas ST2 concentrations did not differ between the two groups. A significant positive correlation was found between Gal-3 and ST2 levels. Gal-3 levels positively correlated with the stage of endometriosis, the duration of symptoms, Marinoff scale and VAS score, while ST2 levels were positively associated with VAS score. Our results suggest that Gal-3 and ST2 could be implicated in the inflammatory process of the disease. Further studies are needed to identify markers of early diagnosis and to open new therapeutic avenues in endometriosis.
Collapse
Affiliation(s)
- Donatella Caserta
- a Department of Gynecologic-Obstetrical Sciences and Urological Sciences, University of Rome Sapienza, S. Andrea Hospital Rome Italy
| | - Luisa Di Benedetto
- a Department of Gynecologic-Obstetrical Sciences and Urological Sciences, University of Rome Sapienza, S. Andrea Hospital Rome Italy
| | - Giulia Bordi
- a Department of Gynecologic-Obstetrical Sciences and Urological Sciences, University of Rome Sapienza, S. Andrea Hospital Rome Italy
| | - Angelo D'Ambrosio
- b Department of Rare Diseases and Complex Phenotypes, Bambin Gesù Children's Hospital Rome Italy
| | - Massimo Moscarini
- a Department of Gynecologic-Obstetrical Sciences and Urological Sciences, University of Rome Sapienza, S. Andrea Hospital Rome Italy
| |
Collapse
|
144
|
|
145
|
Le H, Kim W, Kim J, Cho HR, Kwon B. Interleukin-33: a mediator of inflammation targeting hematopoietic stem and progenitor cells and their progenies. Front Immunol 2013; 4:104. [PMID: 23653627 PMCID: PMC3644799 DOI: 10.3389/fimmu.2013.00104] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 04/22/2013] [Indexed: 11/30/2022] Open
Abstract
Inflammation is defined as a physiological response initiated by a variety of conditions that cause insult to the body, such as infection and tissue injury. Inflammation is triggered by specialized receptors in the innate immune system, which recognize microbial components known as pathogen-associated molecular patterns or endogenous signals produced by damaged cells (damage-associated molecular patterns). IL-33 is a cytokine that is released predominantly at the epithelial barrier when it is exposed to pathogens, allergens, or injury-inducing stimuli. IL-33 target cells are various, ranging from hematopoietic stem and progenitor cells (HSPCs) and essentially all types of their progeny to many non-hematopoietic cells. The pleiotropic actions of IL-33 suggest that IL-33 is involved in every phase of the inflammatory process. In this review, we discuss recent advances in the understanding of how IL-33 orchestrates inflammatory responses by regulating HSPCs and innate immune cells.
Collapse
Affiliation(s)
- Hongnga Le
- School of Biological Sciences, University of Ulsan Ulsan, Republic of Korea
| | | | | | | | | |
Collapse
|
146
|
Wang X, Podila R, Shannahan JH, Rao AM, Brown JM. Intravenously delivered graphene nanosheets and multiwalled carbon nanotubes induce site-specific Th2 inflammatory responses via the IL-33/ST2 axis. Int J Nanomedicine 2013; 8:1733-48. [PMID: 23662055 PMCID: PMC3647448 DOI: 10.2147/ijn.s44211] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Carbon-based nanomaterials (CBN), such as graphene nanosheets (GNS) and multiwalled carbon nanotubes (MWCNT), have been proposed for potential nanomedicine applications such as biomedical devices and carriers for drug delivery. However, our current understanding regarding the systemic toxicity of these CBN through intravenous (iv) injection is limited. In this study, we compare the immune response resulting from GNS and MWCNT exposure. We hypothesize that iv administration of GNS and MWCNT would result in divergent systemic inflammatory responses due to physicochemical differences between these two CBN. In the lungs of C57BL/6 mice, GNS actuate a Th2 immune response 1 day following iv administration, which consists of neutrophilic influx and a significant increase in interleukin (IL)-5, IL-13, IL-33, and its soluble receptor (sST2) in the bronchoalveolar lavage fluid. MWCNT elicited a significant increase in the messenger ribonucleic acid expression of cytokines in the spleen including IL-4 and IL-33, which are associated with an increase in splenic cell differentiation (CD)4+ and CD8+ T-cells in C57BL/6 mice following iv injection. The observed Th2 responses in both the lung and spleen are absent in ST2−/− mice administrated GNS or MWCNT, suggesting a critical role for IL-33. In conclusion, the use of GNS or MWCNT as nanocarriers for drug delivery may result in Th2 immune responses that are mediated through the IL-33/ST2 axis and therefore may promote adverse allergic reactions.
Collapse
Affiliation(s)
- Xiaojia Wang
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | | | | | | | | |
Collapse
|
147
|
ST2 regulates allergic airway inflammation and T-cell polarization in epicutaneously sensitized mice. J Invest Dermatol 2013; 133:2522-2529. [PMID: 23633023 DOI: 10.1038/jid.2013.195] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 03/21/2013] [Accepted: 03/26/2013] [Indexed: 12/14/2022]
Abstract
IL-33 is an inducer of proinflammatory and T-helper type 2 (Th2) cytokines, which have an important role in atopic dermatitis (AD) and allergic asthma. ST2 is a specific receptor for IL-33 and is expressed on Th2 cells, eosinophils and mast cells. A murine model of AD was used to characterize the role of ST2 in allergen-induced skin inflammation and allergic asthma. ST2-/- and wild-type (WT) mice were epicutaneously sensitized with ovalbumin (OVA) and staphylococcal enterotoxin B, and intranasally challenged with OVA. ST2-/- mice exhibited increased production of IFNγ and increased number of CD8(+) T cells in the sensitized skin and in the airways compared with WT mice. The number of eosinophils was decreased, and Th2 cytokines were downregulated in the airways of epicutaneously sensitized ST2-/- mice compared with WT controls. However, dermal eosinophil numbers were as in WT, and the levels of Th2 cytokines were even elevated in the sensitized skin of ST2-/- mice. ST2-/- mice had elevated numbers of neutrophils and macrophages and increased levels of proinflammatory cytokines in the sensitized skin. The role of ST2 differs between different target tissues: ST2 is dispensable for the development of Th2 response in the sensitized skin, whereas it is a main inducer of Th2 cytokines in asthmatic airways.
Collapse
|
148
|
Liang Y, Jie Z, Hou L, Aguilar-Valenzuela R, Vu D, Soong L, Sun J. IL-33 induces nuocytes and modulates liver injury in viral hepatitis. THE JOURNAL OF IMMUNOLOGY 2013; 190:5666-75. [PMID: 23630360 DOI: 10.4049/jimmunol.1300117] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Molecules containing damage-associated molecular patterns play an important role in many pathogenic processes. In this study, our aim was to investigate the role of IL-33, a damage-associated molecular pattern molecule, in adenovirus (Ad)-induced liver inflammation. Ad-infected mice exhibited a steadily increased IL-33 and its receptor IL-1R-like 1 expression in the liver during the first week of infection. Treatment of exogenous IL-33 resulted in a great decrease in the serum alanine aminotransferase levels and the number of Councilman bodies in the liver. Attenuated liver injury by IL-33 correlated with an increase in T regulatory cells but with a decrease in macrophages, dendritic cells, and NK cells in the liver. IL-33 enhanced both type 1 (IL-2 and IFN-γ) and type 2 (IL-5 and IL-13) immune responses in infected mice. However, IL-33 inhibited TNF-α expression in hepatic T cells and macrophages, and significantly reduced TNF-α levels in the liver. We found that in addition to its direct effects, IL-33 strongly induced novel nuocytes in the livers and spleens of infected mice. When cocultured with nuocytes, hepatic T cells and macrophages expressed lower levels of TNF-α. The IL-33-treated mice also demonstrated a slight delay, but no significant impairment, in eliminating an intrahepatic infection with Ad. In conclusion, this study reveals that IL-33 acts as a potent immune stimulator and a hepatoprotective cytokine in acute viral hepatitis. Its direct immunoregulatory functions and ability to induce novel nuocytes further suggest to us that it may be a potentially promising therapeutic candidate for the management of viral hepatitis.
Collapse
Affiliation(s)
- Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | | | | | | | | |
Collapse
|
149
|
Ishikawa S, Shimizu M, Ueno K, Sugimoto N, Yachie A. Soluble ST2 as a marker of disease activity in systemic juvenile idiopathic arthritis. Cytokine 2013; 62:272-7. [PMID: 23561929 DOI: 10.1016/j.cyto.2013.03.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 02/21/2013] [Accepted: 03/08/2013] [Indexed: 10/27/2022]
Abstract
To assess the role of interleukin (IL)-33 and ST2, the receptor for IL-33, in the pathogenesis of systemic juvenile idiopathic arthritis (s-JIA), we sequentially measured the serum levels of IL-33 and soluble ST2 (sST2) in patients with s-JIA and determined their correlation with measures of disease activity and severity. Twenty-four patients with s-JIA, 5 with rheumatoid factor positive polyarticular JIA (RF+poly-JIA), and 20 age-matched healthy controls (HCs) were analyzed. IL-33 and sST2 levels were quantified in serum by enzyme-linked immunosorbent assays. Serum IL-33 levels in most patients with active s-JIA were below the lowest detection limit. Serum IL-33 levels in patients with RF+poly-JIA were significantly higher than those in patients with s-JIA and HC. Serum sST2 levels in patients during the active phase of s-JIA were much higher than those in patients with poly-JIA and HC. Serum sST2 levels in patients with s-JIA were significantly elevated even in the inactive phase, when other clinical parameters were normalized. Serum sST2 levels correlated positively with the clinical parameters of disease activity. These findings indicate that ST2 may be an important mediator in s-JIA. Serum sST2 levels in patients with s-JIA correlated with disease activity, suggesting a potential role as a promising indicator of disease activity.
Collapse
Affiliation(s)
- Sayaka Ishikawa
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan
| | | | | | | | | |
Collapse
|
150
|
Jiang J, Wu C, Lu B. Cytokine-induced killer cells promote antitumor immunity. J Transl Med 2013; 11:83. [PMID: 23536996 PMCID: PMC3617047 DOI: 10.1186/1479-5876-11-83] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 03/20/2013] [Indexed: 12/20/2022] Open
Abstract
The number of immune cells, especially dendritic cells and cytotoxic tumor infiltrating lymphocytes (TIL), particularly Th1 cells, CD8 T cells, and NK cells is associated with increased survival of cancer patients. Such antitumor cellular immune responses can be greatly enhanced by adoptive transfer of activated type 1 lymphocytes. Recently, adoptive cell therapy based on infusion of ex vivo expanded TILs has achieved substantial clinical success. Cytokine-induced killer (CIK) cells are a heterogeneous population of effector CD8 T cells with diverse TCR specificities, possessing non-MHC-restricted cytolytic activities against tumor cells. Preclinical studies of CIK cells in murine tumor models demonstrate significant antitumor effects against a number of hematopoietic and solid tumors. Clinical studies have confirmed benefit and safety of CIK cell-based therapy for patients with comparable malignancies. Enhancing the potency and specificity of CIK therapy via immunological and genetic engineering approaches and identifying robust biomarkers of response will significantly improve this therapy.
Collapse
Affiliation(s)
- Jingting Jiang
- Department of Tumor Biological treatment, the Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, China.
| | | | | |
Collapse
|