Clinical Therapeutics/Volume 38, Number 5, 2016
Review Article
Potential Therapeutic Aspects of Alarmin Cytokine Interleukin 33 or Its Inhibitors in Various Diseases Muhammad Imran Arshad, PhD1,2,3,4; Hilal Ahmad Khan, MBBS4; Gregory Noel, PhD1,2,3; Claire Piquet-Pellorce, PhD1,2,3; and Michel Samson, PhD1,2,3 1
Institut National de la Sante´ et de la Recherche Me´dicale (Inserm), Institut de Recherche Sante´ Environnement Travail (IRSET), Rennes, France; 2Universite´ de Rennes 1, Rennes, France; 3Structure Fe´de´rative BioSit UMS 3480 CNRS-US18 Inserm, Rennes, France; and 4Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan ABSTRACT Purpose: The purpose of this review was to examine the comprehensively accumulated data regarding potential therapeutic aspects of exogenous administration of interleukin 33 (IL-33) or its antagonists in allergic, cancerous, infectious, and inflammatory diseases. Methods: A selected review was undertaken of publications that examined the protective and exacerbating effects of IL-33 or its inhibitors in different diseases. Mechanisms of action are summarized to examine the putative role of IL-33 in various diseases. Findings: IL-33 promoted antibacterial, antiviral, anti-inflammatory, and vaccine adjuvant functions. However, in TH2-biased respiratory, allergic, parasitic, and inflammatory conditions, IL-33 exhibited disease-sensitizing effects. The alarmin cytokine IL-33 induced protective effects in diseases via recruitment of regulatory T cells; antiviral CD8þ cells, natural killer cells, γδ T cells, and nuocytes; antibacterial and antifungal neutrophils or macrophages; vaccineassociated B/T cells; and inhibition of nuclear factor–κB–mediated gene transcription. In contrast, IL-33 exacerbated the disease process by increasing TH2 cytokines, IgE and eosinophilic immune responses, and inhibition of leukocyte recruitment in various diseases.
Scan the QR Code with your phone to obtain FREE ACCESS to the articles featured in the Clinical Therapeutics topical updates or text GS2C65 to 64842. To scan QR Codes your phone must have a QR Code reader installed.
1000
Implications: The protective or exacerbated aspects of use of IL-33 or its inhibitors are dependent on the type of infection or inflammatory condition, duration of disease (acute or chronic), organ involved, cytokine microenvironment, dose or kinetics of IL-33, and genetic predisposition. The alarmin cytokine IL-33 acts at cellular, molecular, and transcriptional levels to mediate pluripotent functions in various diseases and has potential therapeutic value to mitigate the disease process. (Clin Ther. 2016;38:1000–1016) & 2016 Elsevier HS Journals, Inc. All rights reserved. Key words: diseases, IL-33, immunity, therapeutic aspects.
INTRODUCTION The alarmins or danger signals are defined as the class of molecules that alert the immune system to circumvent the invading antigens in the host. The term alarmin was first described in 20051 as a class of structurally diverse and pluripotent host proteins.2 Defensins, eosinophil-derived neurotoxin, cathelicidins, interleukin (IL) 1α, cytosolic calcium-binding proteins of the S100 family, heat-shock proteins, and HMGB1 protein were known to be the classic examples of alarmins.1,3
Accepted for publication February 17, 2016. http://dx.doi.org/10.1016/j.clinthera.2016.02.021 0149-2918/$ - see front matter & 2016 Elsevier HS Journals, Inc. All rights reserved.
Volume 38 Number 5
M.I. Arshad et al. The cytokine IL-33 is the 11th described member of the IL-1 family and is designated as IL-1F11. IL-33 was originally identified as DVS27, a gene that was up-regulated in vasospastic cerebral arteries after subarachnoid hemorrhage in canines,4 and as a nuclear factor from high endothelial venules, which is expressed in endothelial cells nuclei.5 In 2005, the DVS27 gene was rediscovered as IL-33 by using computational tools on the basis of sequences that contained the 12 β-trefoil structure seen in IL-1/ fibroblast growth factor–like proteins.2 The cytokine IL-33 was described as an alarmin with a dual function protein that acts as a cytokine when released from cells and a nuclear factor–regulating gene transcription.6 Once released from cells, the cytokine functions of IL-33 are mediated by interaction with its specific receptors, including ST2 (IL-1 receptor–like 1) and IL-1RAcP (IL-1 receptor accessory protein). The cellular sources of IL-33 include endothelial cells, epithelial cells, smooth muscle cells, keratinocytes, astrocytes, adipocytes, fibroblasts, hepatic and pancreatic stellate cells, monocytes, macrophages, and hepatocytes.7–10 The target cells of IL-33 include B cells, TH2 cells, CD8þT cells, macrophages, dendritic cells, mast cells, basophils, and a recently identified population of innate lymphoid cells called nuocytes in different tissues, including lungs, gut, liver, spleen, or skin.2,11–16 Extracellular IL-33 induces autocrine, paracrine, and juxtacrine actions that are important in autoimmune or inflammatory diseases, immune defense, and repair mechanisms.16,17 The underlying signaling pathway of the IL-33/ST2 axis is dependent on activation of cytoplasmic TIR domain, such as MyD88 and the serine threonine kinases IRAK1, IRAK4, and TRAF6, leading to activation of nuclear factor–κB (NF-κB), AP-1, and mitogen-activated protein kinase (MAPK) pathways. The nuclear functions of IL-33 are associated with down-regulation of NFκB–dependent gene transcription.18 IL-33 was used as a promising mucosal vaccine adjuvant and induced protective immunity (i.e., primary and memory immune responses).19 The mouse Il33 gene revealed the existence of two transcripts, IL-33a and IL-33b, with different 5'UTRs but coding for the same protein. The IL-33a and IL-33b mRNAs started with two different noncoding first exons, distant by 20 kb. A consensus TATA-like sequence was found 29 bp upstream of each of these transcription start sites, evidencing that IL-33a and
May 2016
IL-33b are transcribed from classic TATA box–containing promoters.20 IL-33 was found to be synthesized as a 270–amino acid protein precursor containing an N-terminal nuclear localization sequence, a helix-turn helix motif, and a C-terminal region with structural homology to other IL-1 cytokines (IL-1α, IL-1β, and IL18).2,5 The human and mouse IL-33 shared a 55% homology at the amino acid level. The translated fulllength IL-33 protein (30.7 kDa) was found to be a nuclear factor associated with heterochromatin in vivo and mitotic chromosomes in living cells, which possesses potent transcriptional-repressor properties.6,21 The protective functions of IL-33 plausibly attributed to its binding to acidic pocket of a dimeric histone, H2AH2B, on the surface of nucleosomes, resulting in suppression of gene transcription.21,22 Accordingly, IL-33 interacts with transcription factor NF-κB (p65 subunit) and impaired its DNA binding, resulting in diminished NF-κB–dependent proinflammatory gene transcription.18 IL-33 was described as an alarmin in different pathologic conditions or infectious diseases,23 and it induces multivalent functions, resulting in proinflammatory or anti-inflammatory effects in various conditions.24 The full-length bioactive form of IL-33 was released during cell necrosis and act as an endogenous danger signal or alarmin.6,25,26 It was found that apoptotic caspases cleave IL-33 and rendered it inactive or nonfunctional.25,27 IL-33 was also cleaved by neutrophil elastase and cathepsin G, mast cell chymase enzyme, and calpain enzyme as previously described.28–31 Recently, IL-33 was found to be associated with a form of cell death called necroptosis as inhibitors of necroptosis (necrostatin-1 and PJ34) down-regulated IL-33 expression in liver injury.32 These studies described the activity of endogenous IL-33, but exogenous administration of IL-33 induced dual edge functions, depending on the immune pathology or organ involved. Although IL-33 has been extensively studied in various diseases, a comprehensive review on therapeutic implications of IL-33 and its mechanism of action in different conditions remained obscure. Therefore, based on our and other published data, we summarized a review on the effect of therapeutic administration of IL-33 or blocking of IL-33/ST2 during infectious, allergic, and inflammatory diseases (Figure). The protective and exacerbating or sensitizing role of IL-33 administration in immunopathologic animal disease models and respective mechanism of action is elaborated comprehensively in this review.
1001
Clinical Therapeutics
Exogenous IL-33 administration
IL-33
IL-33
IL-33 IL-33 IL-33
ST2 IL-33
IL-33
IL-33 or ST2 blocking antibodies
IL-33
IL-33
IL33
IL-3 3
ST2s
IL-33
IL-1RAcP
ST2L
ST2L
IL-1RAcP
IL-33
PROTECTIVE EFFECTS EXACERBATUNG EFFECTS Airway allergy Lung fibrosis Influenza virus lung infection Respiratory syncytial virus infection Acute kindeny injury UIcerative colitis Intestinal mucositis Eosinophilic esophagitis Exoperimental autoimmune encephalomyelitis Graft-versus-host disease Periodontal disease Schistosoma japonicum infection Leishmaniasis (Leishmania donovani) Formalin-induced pain
Tumor growth and metastasis Adipose tissue inflammation Atherosclerosis
Joint inflammation/arthritis LCMV infection and hepatitis Adenovital hepatitis Ischemia/reperfusion hepatitis Concanvalin A−induced hepatitis Skin infection by Staphylococcus auerus Keratitis by Pseudomonas aeruginosa Fungal peritonitis Pancreatitis Intestinal inflammation Colitis Intestinal nematode infection Experimental sepsis by cecal ligation puncture Ischemic brain injury/stroke HDM-induced airway inflammation Influenza virus vaccine Immune-mediated experimetnal arthritis Rheumatoid arhritis Nippostrongylus brasiliensis intestinal infection Leishmaniasis (Leishmania major) Meningitis Systemic lupus erythematosus HDM asthma Ovalbumin-induced airway inflammation Allergic rhinitis Fungal asthma Cigarette smoke-induced lung inflammation
Figure. Potential protective or sensitizing effects of exogenous administration of interleukin (IL)-33 or blocking of IL-33/ST2 in diverse diseases. HDM, house dust mite; LMCV, lymphocytic choriomeningitis virus.
Protective Therapeutic Role of IL-33 or Its Inhibitors in Various Diseases
Beneficial Effects of IL-33 Administration in Different Diseases
The dichotomous role of IL-33 in various diseases is evident from previous studies; the exogenous administration of IL-33 induced protective effects during cancerous, infectious, allergic, and inflammatory diseases (Table I and Figure).
IL-33 favored antitumor immune response through enhancement and activation of effector cytotoxic T lymphocyte, natural killer (NK) cells, and interferon (IFN)-γ production, resulting in regression of B16 melanoma, Lewis lung carcinoma, and human
1002
Volume 38 Number 5
M.I. Arshad et al.
Table I. Protective effects of IL-33 or its inhibitors in cancerous, infectious, allergic, and inflammatory diseases. Disease
Role of IL-33 or ST2 Inhibitors
Beneficial effects of exogenous IL-33 in different diseases Tumor growth and metastasis Protective (B16 melanoma and Lewis lung carcinoma metastatic model)
HPV-associated cancer model
Protective, vaccine adjuvant– enhancing and antitumor effect
Adipose tissue inflammation
Protective, in genetically obese mice
Atherosclerosis
Protective
Joint inflammation or arthritis
Protective
LCMV hepatitis
Protective (in acute phase at 16 hours)
Mechanism of Action
Reference
Activation of CD8þ T cells and NK cells with increased infiltration in tumor tissues. Increased cytotoxicity of CD8þ T cells and NK cells in vitro via NF-κB and up-regulated expression of CD69. Enhanced antigen-specific effector and memory T cells (CD4þ and CD8þ) in vivo. Increased antiviral IFN-γ production and HPV induced tumor regression. Decreased adiposity and deceased fasting glucose level. Improvement in glucose level and insulin tolerance. Accumulation of TH2 cells in adipose tissue and polarization of macrophages toward M2 activated phenotype (CD206þ). Decreased inflamed atherosclerotic plaques. Decreased IFN-γ level and enhanced TH2 cytokines (IL-4, IL-5 and IL-13). Increased serum IgA, IgE, and IgG1 level and antioxidized lipoprotein antibodies. Inhibition of cartilage destruction, systemic bone loss, and osteoclast differentiation. IL-33 increased antiosteoclastogenic cytokines, such as GM-CSF, IL-4 and IFN-γ in serum. Increased IFN-γ–producing γδ T cells and NK cells and inhibition of IL-17þ γδ T cells, dendritic cells proliferation, and cytokine production. The above protective effects were reversed in IL-33 knockout mice.
33
34
35
36
37
38
(continued)
May 2016
1003
Clinical Therapeutics
Table I. (continued). Disease LCMV infection
Adenoviral hepatitis
Warm ischemia or reperfusion liver injury
Ischemia/reperfusion hepatitis (acute)
ConA–induced hepatitis (acute)
Skin infection by methicillinresistant Staphylococcus aureus
Keratitis (Pseudomonas aeruginosa)
Role of IL-33 or ST2 Inhibitors
Mechanism of Action
Protective, antiviral, and vaccine Enhanced antigen-specific CD8þ or cytotoxic T-cell functions. adjuvant effect IL-33 favored immunomodulatory effect. Protective Decreased serum ALT, TNF-α level, and councilman bodies in liver. Decreased infiltration of macrophages, dendritic cells, and NK cells in liver and enhanced number of Treg cells and nuocytes in liver. Protective, mouse IL-33 injection Decreased AST, ALT, and IFN-γ in male Balb/C mice level. Increased IL-4, IL-5, and IL-13 production. Treatment with anti-ST2 antibody worsens the disease severity. Protective, i.p. injection in Increased activation of NFκB, C57Bl/6 male mice MAPK, cyclin D, and BCl2 genes in liver. Dampen the CXC chemokines and liver neutrophil accumulation. Protective, mouse IL-33 injection Decreased serum AST, ALT, and (1 μg per mouse i.p.) in TH1/TH17 cytokines. Downregulation of infiltration of Balb/C mice mononuclear cells in liver and increased number of Treg cells and IL-4 producing CD4þ T lymphocytes in liver. ST2 knockout mice had exacerbated liver injury. IL-33 knockout mice were more susceptible to ConA liver injury than WT mice. Protective Enhanced bacterial clearance and wound healing by upregulation of CXCR2 expression on neutrophils and their recruitment to site of infection. Protective Better bacterial control by increased polarization of M2 macrophages and TH2 cytokines (IL-4, IL-5, and IL-10) expression in C57Bl/6 mice.
Reference 39
75
40
41
42,56
43
44
(continued)
1004
Volume 38 Number 5
M.I. Arshad et al.
Table I. (continued). Disease Fungal peritonitis (Candida albicans)
Role of IL-33 or ST2 Inhibitors Protective
Mechanism of Action
Enhanced fungal clearance and survival of mice. IL-33 upregulated expression of CXCL1 and CXCL2 on neutrophils, increased complement receptor-3, and ROS production, leading to yeast phagocytosis and killing. Pancreatitis (Coxsackie B virus) Protective Prevented pancreatitis by enhancing number of CD8þ T cells, NK cells, and upregulation of IL-4–, TNF-α–, and IFN-γ–dependent antiviral immune response. Intestinal inflammation Protective IL-33 administration ameliorated intestinal inflammation in amphiregulin-dependent manner. Experimental colitis Protective IL-33 increased the expression of (trinitrobenzene sulfonic TH2 cytokines, increase in FoxP3 acid–induced acute colitis) expression in Treg cells. Switched TH1 cytokine signature to TH2 cytokines via Treg cells. Enhanced number of CD103þ dendritic cells in intestine. DSS colitis (chronic) Protective, repeated mouse IL-33 Decreased inflammation and TH1 treatment at intermediate and cytokines (IFN-γ) and enhanced after DSS challenge TH2 cytokine expression. Enhanced Ly6g mRNA expression and myeloperoxidase activity in colon. Intestinal nematode infection Protective Increased expulsion of parasites (Trichuris muris) from gut. Decreased TH1 cytokines and pro-TH2 (IL-4, IL-9, and IL-13) response in intestine via overexpression of thymic stromal lymphopoietin in intestinal epithelium. Experimental sepsis by cecal Protective, intravenous mouse Increased neutrophil influx at site ligation puncture IL-33 administration (1 μg per of infection and efficient mouse, Balb/C) bacterial clearance. IL-33 prevented down-regulation of CXCR2 expression and chemotaxis, peritonitis, and mortality in mice.
Reference 45
46
47
48
49
50
51
(continued)
May 2016
1005
Clinical Therapeutics
Table I. (continued). Disease
Role of IL-33 or ST2 Inhibitors
Ischemic brain injury or stroke Protective role of recombinant (acute phase) IL-33 administration in C57Bl/6 male mice
HDM-induced airway inflammation
Protective role of recombinant IL-33 administration in Balb/C mice
Influenza virus vaccine
Protective, adjuvant, and immunostimulatory effect
Beneficial effects of blocking IL-33 or its receptor ST2 in different Immune-mediated experimental No effect of genetic IL-33 arthritis (antigen and deficiency collagen mediated by injection of methylated bovine serum) Rheumatoid arthritis (collagen Protective effect of anti-ST2 induced) antibody Nippostrongylus brasiliensis intestinal infection
Limited protective effect due to genetic deficiency of IL-33
Leishmaniasis (Leishmania major) No effect of anti–ST2-Fc blocking antibody
Meningitis (angiostrongyliasis)
Systemic lupus erythematosus (chronic disease)
Protective effect of anti-ST2 antibody, 3 days after infection Protective effect of anti–IL-33 antibody for 6 weeks (i.p.) in MRL/lpr lupus-susceptible mice
Mechanism of Action
Reference
IL-33 ameliorated ischemic brain damage at 24-72 hours, inflammation, and neurologic deficit. IL-33 decreased TH17 cytokines and promoted TH2 immune response. IL-33 favored antibody production and augmented neutralizing humoral immune response. IL-33 adjuvanted with influenza virus vaccine enhanced IgG, IgA, TH1, and TH2 cytokines. diseases IL-33 deficiency in mice did not impair or affect antigen- and collagen-mediated arthritis. May enhance the antigenspecific IFN-γ and IgG2a production. Decreased IFN-γ production and collagen-induced arthritis or inflammation. Limited N brasiliensis clearance and decreased IL-13 production in IL-33 knockout mice. Impaired RELMβ expression and eosinophil recruitment. Enhanced TH1 response (IFN-γ) by CD4þ T cells without impairment of TH2 cytokine production. Decreased IL-5 in circulation and decreased eosinophilic influx in meninges. Decreased proteinuria, anti–doublestranded DNA antibodies and prevented antigen-antibody complex formation. IL-33– blocking antibody decreased TH17 cytokines, IL-1β, and IL-6 and enhanced the number of Treg and myeloid derived suppressor cells.
52
53
19
54
55
57
58
59
60
(continued)
1006
Volume 38 Number 5
M.I. Arshad et al.
Table I. (continued). Disease HDM asthma mouse model (Balb/C)
Airway inflammation (ovalbumin-mediated, chronic condition) Airway inflammation (ovalbumin mediated)
Allergic rhinitis (ovalbumin induced)
Fungal asthma (Aspergillus fumigatus)
Cigarette smoke–induced lung inflammation
Role of IL-33 or ST2 Inhibitors Anti–IL-33 priming in mice by s.c. route
Mechanism of Action
Anti–IL-33 IgG decreased airway hyperresponsiveness by decreasing eosinophils in BALF and decrease of inflammatory cytokine production (IL-17A, IL-25, and IL-33) in lungs Protective effect of anti-ST2 Decreased IgE-mediated airway antibody or anti–IL-33 inflammation or remodeling antibody treatment by control of IL-33–producing alveolar macrophages. Protective effect of anti–IL-33 Reduced eosinophils and antibody (150 μg per mouse) lymphocytes in lungs and or anti-ST2 monoclonal decreased IL-4, IL-5, and IL-13 antibody treatment in Balb/C in BALF. Decreased level of female mice ovalbumin-specific IgE antibodies in serum. Protective effect of anti–IL-33 Anti–IL-33 antibody decreased antibody treatment rhinitis, improved skin denudation and nociception, decreased IgE level, and reduced eosinophilia and IL-4, IL-5, and IL-13 production in bronchoalveolar lavage or in nasal cavity. Switch from proinflammatory Protective effect of anti-ST2L TH2 response to protective monoclonal antibody with TH1 response by activation of CpG Toll-like receptor 9 on innate immune cells. Protective effect of anti–IL-33 Blocking IL-33 led to decrease antibody by intranasal route neutrophil and macrophage infiltration in lungs and decreased expression of MCP-1, IL-1β, TNF-α, IL-17, and mucin-5.
Reference 61
63
62,65
64
66
67
ALT ¼ alanine aminotransferase; AST ¼ aspartate aminotransferase; ConA ¼ concanavalin A; DSS ¼ dextran sulfate sodium; GM-CSF ¼ granulocyte-macrophage colony-stimulating factor; HDM ¼ house dust mite; HPV ¼ human papillomavirus; IFN-γ ¼ interferon-γ; IL ¼ interleukin; LCMV ¼ lymphocytic choriomeningitis virus; NF-κB ¼ nuclear factor–κB; NK ¼ natural killer; TNF-α ¼ tumor necrosis factor-α; Treg ¼ T regulatory; WT ¼ wild type.
papillomavirus (HPV)–associated tumors.33,34 As a vaccine adjuvant, IL-33 enhanced specific cellular immune responses (effector and memory T-cell
May 2016
functions) against HPV-associated cancer model.34 In genetically obese mice, IL-33 promoted a pro-TH2 cellular immune response, macrophage polarization,
1007
Clinical Therapeutics and decreased adipose tissue inflammation, revealing a protective effect of IL-33 during obesity.35 IL-33 was found to be cardioprotective because IL-33 inhibited formation of atherosclerotic plaques and favored a TH2 response or antioxidized lipoprotein humoral immune response.36 The exogenous administration of IL-33 inhibited joint inflammation by upregulation of anticlastogenic cytokines, such as IL-4, granulocyte-macrophage colony-stimulating factor, and IFN-γ.37 These findings suggested a divergent role of IL-33 in arthritis. In liver disease, the IL-33/ST2 axis was known to be hepatoprotective. In lymphocytic choriomeningitis virus–induced acute hepatitis, IL-33 induced hepatoprotection via increased infiltration of IFN-γ–producing γδ T cells and NK cells in liver, which mediated antiviral immune response.38 IL-33 favored antiviral and vaccine adjuvant effect during lymphocytic choriomeningitis virus infection by activation of cytotoxic T lymphocytes and immune-modulatory functions.39 In the adenoviral hepatic model, IL-33 diminished liver injury marker (ALT), tumor necrosis factor-α, liver infiltrate cells, and an increased number of regulatory T (Treg) cells and innate lymphoid cells and nuocytes.38 The protective functions of IL-33 were found in ischemia/reperfusion liver injury, which were associated with decreased aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels, dampened chemokine and neutrophil infiltration, and pro-TH2 and NF-κB/MAPK responses in the liver.40,41 In concanavalin A (ConA)–mediated immune hepatitis, IL-33 ameliorated liver injury by decreasing AST and ALT, TH1 and TH17 cytokines, and promotion of Treg cell or IL-4–producing CD4þ cells in the liver.42 Hence, IL-33 orchestrated immune functions in the liver at cellular, molecular, and gene transcriptional levels to protect hepatitis. IL-33 induces protective functions in bacterial, fungal, and parasitic infections. The methicillinresistant Staphylococcus aureus–associated skin infection was improved by IL-33 by promoting bacterial clearance, wound healing, and neutrophil recruitment at the site of infection.43 Better control of Pseudomonas aeruginosa–induced keratitis was found after IL-33 treatment, which enhanced macrophage polarization and functions and pro-TH2 immune response.44 In the fungal peritonitis infection model by Candida albicans, IL-33 promoted fungal clearance via increased neutrophil infiltration and its killing
1008
by the ROS-dependent pathway or oxidative burst mechanism.45 In gastrointestinal diseases, IL-33 mediated a protective therapeutic effect. In Coxsackie B virus–induced pancreatitis, IL-33 abrogated pancreatic inflammation via up-regulation of antiviral cytokine response (IL-4, tumor necrosis factor-α, and IFN-γ dependent) and CD8þ cells, NK cells dependent mechanism.46 A protective mechanism dependent of amphiregulin was boosted by IL-33 administration to dampen the intestinal inflammation.47 In an animal model of experimental colitis, single or repeated injections of IL-33 proved protective in acute and chronic colitis.48,49 The underlying mechanism was switching of TH1 cytokine responses to TH2 biased immune responses, recruitment of Treg cells, and increased myeloperoxidase activity.48,49 IL-33 was protective by expelling Trichuris muris nematodes from the gastrointestinal tract in a TH2 cytokine– dependent mechanism.50 In the septic condition, IL-33 enhanced influx of neutrophils and prevented cecal ligation puncture–induced sepsis in mice.51 In acute ischemic brain injury or stroke, IL-33 provided protection by decreasing inflammatory cytokines (TH17) or inflammation.52 IL-33 administration promoted humoral immune response against house dust mite– induced airway inflammation as recently described.53 As a vaccine adjuvant in influenza virus infection, IL-33 enhanced antiviral humoral and cytokinic immune response.19
Beneficial Effects of Blocking IL-33 or ST2 in Different Diseases The potent role of IL-33 during arthritis was evidenced in previous studies; however, the genetic deficiency of IL-33 did not affect collagen-induced arthritis,54 and anti–ST2 antibody treatment protected mice against collagen-induced arthritis.55 In liver disease, the genetic ablation of IL-33 or ST2 sensitized the mice to develop enhanced ConA liver injury compared with WT mice.42,56 In the context of parasitic infections, the genetic deficiency of IL-33 led to impaired clearance of Nippostrongylus brasiliensis intestinal infection,57 whereas in an antagonistic approach, the use of ST2-Fc blocking antibody did not affect leishmaniasis infection in mice.58 In another antagonistic strategy, anti–ST2 antibody decreased eosinophilic response and meningitis caused by Angiostrongylus systemic parasitic infection.59
Volume 38 Number 5
M.I. Arshad et al. Blockade of IL-33 prevented chronic systemic lupus erythematosus in lupus-susceptible mice by inhibition of TH17, proinflammatory cytokines, and recruitment of Treg cells.60 In respiratory diseases, blocking of IL-33 activity proved to be more effective. In the house dust mite asthma model, anti–IL-33 priming was protective by decreasing airway hyperresponsiveness, eosinophilia, and inflammatory cytokines.61 Antagonizing the IL-33 (anti-ST2 or anti–IL-33 injection) induced protective effects in ovalbumin-mediated airway inflammation and allergic rhinitis,62–65 the underlying mechanism was dependent on decreased IgE and TH2 cytokine production. Anti–IL-33 treatment ameliorated fungal asthma by Aspergillus fumigatus in a TH2-dependent manner.66 Similarly, the cigarette smoke–mediated experimental lung inflammation was inhibited by blocking IL-33, which was associated with decreased neutrophil and macrophage infiltration in lungs and down-regulation of inflammatory mediators.67 Conclusively, IL-33 or its inhibitors induced protective effects in a wide range of diseases mainly by mediation of cellular and molecular immune responses. Mechanistically, IL-33 promoted infiltration of regulatory T cells; antiviral CD8þ cells, NK cells, γδ T cells, and nuocytes; neutrophils and macrophagemediated antibacterial and antifungal responses, and vaccine adjuvanted humoral and memory lymphocytic immune responses. IL-33 inhibited proinflammatory cytokines and inflammatory mediators in various organs and tissues after injury or infection and regulated NF-κB gene transcription to control inflammation. However, in allergic or infectious diseases of the respiratory tract, IL-33 blockade induced protective effects by decreasing IgE, eosinophils, and TH2 cytokine production.
EXACERBATING ROLE OF IL-33 IN VARIOUS DISEASES In contrast to protective functions or effects induced by IL-33, the administration of exogenous IL-33 may have exacerbating aspects in different diseases (Table II and Figure). IL-33 aggravated ovalbumin- or Alternaria alternata–mediated airway allergy or inflammation by enhancement of TH2 cytokine (IL-4, IL-5, IL-13)– dependent allergic response.68,69 Profibrotic effect of IL-33 was observed in bleomycin-induced lung fibrosis with augmented IL-13 and transforming
May 2016
growth factor-β1 production.70 In viral lung infections (by influenza virus or respiratory syncytial virus), IL-33 caused the mice to develop severe inflammation, increased airway hyperresponsiveness, TH2 cytokines, and eosinophilic immune responses in lungs of infected mice.71,72 IL-33 exacerbated acute cisplatin-induced kidney injury by increasing apoptotic and necrotic tubular cell death and cellular infiltration.73 Although IL-33 induced hepatoprotective effects (Table I), Chen et al found that increased ConAinduced liver injury (AST and ALT) with a higher dose of IL-33 (10 μg per mouse) and blocking IL-33 (by anti–IL-33 or anti–ST2-Fc antibody) abrogated the liver injury markers.74 Our previous data revealed that deficiency of IL-33 caused the mice to enhanced ConA liver injury, indicating a protective effect of endogenous IL-33 in liver disease.56 The plausible differences of the study by Chen et al with other data may be attributed to the dose of IL-33 or ConA used in these studies, genetic background of mice, and kinetics of liver injury. However, it is evident from most of the studies that the IL-33/ST2 axis induced hepatoprotective effects during viral, ischemic, and immune-mediated hepatitis.38,40–42,75 In contrast to chronic colitis, IL-33 exacerbated dextran sulfate sodium–induced acute colitis with upregulation of proinflammatory immune responses.76,77 Similarly, IL-33 administration sensitized the mice to irinotecan-induced intestinal mucositis with decreased leukocyte number. The blockade of ST2 or IL-33 ameliorated the disease severity by decreasing chemokine production and restricting neutrophil infiltration.78 In acute esophagitis, IL-33 favored pro-TH2 and eosinophilic response to exacerbate the disease.79 Moreover, IL-33 sensitized the mice to increased autoimmune encephalomyelitis via enhanced IFN-γ and IL-17 production.80 In graft-versus-host disease, IL-33 caused the mice to have a severe inflammatory response, and antagonistic IL-33 approaches reversed the inflammatory condition.81 IL-33 enhanced periodontal bone loss in Porphyromonas gingivalis infected mice through up-regulated receptor activator of NF-κB ligand expression in B and T lymphocytes in gingival mucosa,82 which was abolished in ST2 knockout mice. In parasitic infections, IL-33 sensitized the mice to increased TH2 response and liver disease caused by Schistosoma japonicum,83 and antagonizing the IL-33 proved protective. The visceral leishmaniasis by Leishmania
1009
Clinical Therapeutics
Table II. Exacerbating effects of IL-33 in different diseases. Disease
Role of IL-33
Airway allergy (by ovalbumin or Alternaria alternata extract sensitization)
Exacerbating
Airway inflammation
Sensitizing
Lung fibrosis (bleomycin induced)
Exacerbating or profibrotic
Influenza virus lung infection
Exacerbating
Respiratory syncytial virus infection
Exacerbating
Acute kidney injury (cisplatin induced)
Exacerbating
ConA-induced hepatitis (acute)
Exacerbating, mouse IL-33 injection (10 μg per mouse) in C57Bl/6 mice
Mechanism of action
Reference
IL-33 administration abrogated Heligmosomoides polygyrus mediated suppression of Alternaria and ovalbumininduced airway allergic response (IL-4, IL-5, IL-13 production and localized eosinophilia). Enhanced allergic response or airway hyperresponsiveness and goblet cell hyperplasia in lungs accompanied by IL-4, IL-5, and IL-13 secretion in lungs after IL-33 administration. IL-33 promoted lung inflammation and profibrogenic response (increase in IL-13 and TGF-β1 expression). Blocking IL-33 antibody or use of ST2 knockout mice revealed antifibrotic effects. Impaired lung function and repair in IL-5, IL-9, IL-13, and amphiregulin-dependent manner. Enhanced the disease severity and inflammation of the respiratory tract. Anti–IL-33 antibody treatment inhibited airway hyperresponsiveness, TH2 cytokines, eosinophilia, and mucous hyperproduction in airways. IL-33 markedly exacerbated acute tubular necrosis and apoptosis, serum creatinine level, and infiltration of CD4þ T cells. Increased serum AST and ALT levels in mouse IL-33 and ConA–treated mice. Anti–IL-33 antibody (300 μg per mouse) or anti–ST2-Fc antibody
69
68
70
71
72
73
74
(continued)
1010
Volume 38 Number 5
M.I. Arshad et al.
Table II. (continued). Disease
Role of IL-33
DSS colitis (acute), ulcerative colitis
Exacerbating
DSS-induced colitis (acute)
Exacerbating (in acute phase)
Intestinal mucositis (irinotecan Exacerbating, IL-33 injection Camptothecin-II induced) (200 ng per mouse) in Balb/C mice
Eosinophilic esophagitis (acute Exacerbating (use of onset) recombinant IL-33 for 1 week)
Experimental autoimmune Sensitizing or exacerbating encephalomyelitis, myelin oligodendrocyte glycoprotein (MOG 35-55) peptide mouse model Graft-versus-host disease
Sensitizing or exacerbating
Periodontal disease (periodontitis)
Exacerbating
Mechanism of action (50 μg per mouse) decreased liver injury markers (AST and ALT). Augmented colon inflammation in an IL-4–dependent manner with increased IL-1, IL-4, IL-6, IL-13, and IL-17 cytokines and VEGF. Decreased CXCL9 and CXCL10 expression. Decreased IFN-γ and IL-17A producing lymphocytes in lamina propria of colon. Increased IL-5 and IL-13 production. Increased mucositis associated with severe systemic leucopenia, shortened villi, and increased plasma leakage. The mucositis was decreased in ST2 knockout mice or in mice treated with anti–IL-33 antibody or sST2 with reduced neutrophil accumulation and the production of CXCL1, CXCL2, and CCL2 chemokines. Eosinophilia, mucosal hyperproliferation, increased TH2 cytokines (eg, IL-13) and decreased T-regulatory signature. Exacerbated disease severity associated with increased IFN-γ and IL-17 production. Blocking anti–IL-33 antibody reversed the above phenomenon and disease. Increased inflammation after recombinant IL-33 injection. The lethality and inflammation were decreased in IL-33–/ST2deficient mice or by ST2-Fc antibody injection. Porphyromonas gingivalis infected mice had enhanced alveolar
Reference
76
77
78
79
80
81
82 (continued)
May 2016
1011
Clinical Therapeutics
Table II. (continued). Disease
Schistosoma japonicum infection
Leishmaniasis (Leishmania donovani)
Formalin-induced pain
Role of IL-33
Mechanism of action
bone loss or periodontal bone destruction. Increased receptor activator of NF-κB ligand expression in B and T lymphocytes in gingival tissues after mouse IL-33 treatment. Decreased periodontal loss was found in ST2 knockout mice. Exacerbating IL-33 increased expression of TH2 cytokines (IL-5, IL-10, and IL13) at 6 weeks after infection with increased liver disease. Anti–IL-33 treatment reversed the above parameters to normal. Exacerbating with increased Decreased TH1 cytokines inhibit monocyte ad neutrophil susceptibility, i.p., 2-week recruitment in liver (decreased treatment in Balb/C mice KC/CXCL1 and CXCL2 expression). The parasitic load and granuloma formation in liver were diminished in ST2 knockout mice. Sensitizing (administration of s. IL-33 administration enhanced c. mouse IL-33, 300 ng per pain sensation with increased mouse, and intrathecal paw lifting and licking time in mouse IL-33, 3 ng per mouse) normal and formalin-injected mice. ST2 antibody treatment or ST2 deficiency in mice alleviated pain sensation and behavior.
Reference
83
84
85
ALT ¼ alanine aminotransferase; AST ¼ aspartate aminotransferase; ConA ¼ concanavalin A; DSS ¼ dextran sulfate sodium; IFN-γ ¼ interferon-γ; IL ¼ interleukin; NF-κB ¼ nuclear factor–κB; TGF-β1 ¼ transforming growth factor-β1; VEGF ¼ vascular endothelial growth factor; WT ¼ wild type.
donovani was enhanced by IL-33 administration by blocking TH1 cytokines and liver infiltration of leukocytes.84 Finally, IL-33 augmented pain sensation or nociception in mice challenged with formalin85 and blocking of ST2 receptor alleviated the pain sensation.
1012
In summary, IL-33 produced disease exacerbating effects mainly by increasing TH2 cytokines, IgE and eosinophilic immune responses, and inhibition of leukocyte recruitment in respiratory, allergic, parasitic, and inflammatory diseases. The sensitizing effects of
Volume 38 Number 5
M.I. Arshad et al. IL-33 were found in TH2-prone conditions; therefore, antagonistic IL-33 approaches may prove helpful to mitigate such diseases or inflammatory conditions.
CONCLUSION Although most of the previous studies used an active form of recombinant IL-33, considerations should be taken for dose, route, duration, half-life, pharmacokinetic properties, presence of IL-33 cleaving enzymes, and soluble ST2 to completely comprehend the dual biological activity of IL-33 and rational therapeutic use of IL-33 in various diseases. Because most of the above mentioned effects of IL-33 have been reported in murine disease models, there are some limitations in extrapolation of murine data to humans.
ACKNOWLEDGMENT This work was supported by Inserm, the Ministère de l’Education Nationale de la Recherche et de la Technologie, the University of Rennes 1, and the Région Bretagne. Dr Arshad is supported by research grant 20-4613/NRPU/R&D/HEC/14/45 on hepatoprotective role of IL-33 funded by the Higher Education Commission under the National Research Programme for Universities (NRPU) scheme at University of Agriculture, Faisalabad, Pakistan.
CONFLICTS OF INTEREST The authors have indicated that they have no conflicts of interest regarding the content of this article.
SUPPLEMENTARY MATERIALS Supplementary material cited in this article is available online at http://dx.doi.org/10.1016/j.clinthera.2016. 02.021.
REFERENCES 1. Oppenheim JJ, Yang D. Alarmins: chemotactic activators of immune responses. Curr Opin Immunol. 2005;17:359–365. 2. Schmitz J, Owyang A, Oldham E, et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity. 2005;23:479–490. 3. Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol. 2007;81:1–5.
May 2016
4. Onda H, Kasuya H, Takakura K, et al. Identification of genes differentially expressed in canine vasospastic cerebral arteries after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 1999;19:1279–1288. 5. Baekkevold ES, Roussigne M, Yamanaka T, et al. Molecular characterization of NF-HEV, a nuclear factor preferentially expressed in human high endothelial venules. Am J Pathol. 2003;163:69–79. 6. Moussion C, Ortega N, Girard JP. The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: a novel ‘alarmin’? PLoS One. 2008;3:e3331. 7. Arshad MI, Rauch M, L’Helgoualc’h A, et al. NKT cells are required to induce high IL-33 expression in hepatocytes during ConA-induced acute hepatitis. Eur J Immunol. 2011;41:2341–2348. 8. Marvie P, Lisbonne M, L’Helgoualc’h A, et al. Interleukin33 overexpression is associated with liver fibrosis in mice and humans. J Cell Mol Med. 2010;14:1726–1739. 9. Murphy GE, Xu D, Liew FY, McInnes IB. Role of interleukin 33 in human immunopathology. Ann Rheum Dis. 2010;69(Suppl 1):i43–i47. 10. Arshad MI, Guihard P, Danger Y, et al. Oncostatin M induces IL-33 expression in liver endothelial cells in mice and expands ST2þCD4þ lymphocytes. Am J Physiol Gastrointest Liver Physiol. 2015;309:G542–G553. 11. Barlow JL, Bellosi A, Hardman CS, et al. Innate IL-13producing nuocytes arise during allergic lung inflammation and contribute to airways hyperreactivity. J Allergy Clin Immunol. 2012;129:e191–e194. 12. Besnard AG, Togbe D, Guillou N, et al. IL-33-activated dendritic cells are critical for allergic airway inflammation. Eur J Immunol. 2011;41:1675–1686. 13. Komai-Koma M, Gilchrist DS, McKenzie AN, et al. IL-33 activates B1 cells and exacerbates contact sensitivity. J Immunol. 2011;186:2584–2591. 14. Smithgall MD, Comeau MR, Yoon BR, et al. IL-33 amplifies both Th1- and Th2-type responses through its activity on human basophils, allergen-reactive Th2 cells, iNKT and NK cells. Int Immunol. 2008;20:1019–1030. 15. Saluja R, Khan M, Church MK, Maurer M. The role of IL33 and mast cells in allergy and inflammation. Clin Transl Allergy. 2015;5:33. 16. Theoharides TC, Petra AI, Taracanova A, et al. Targeting IL-33 in autoimmunity and inflammation. J Pharmacol Exp Ther. 2015;354:24–31. 17. Martin NT, Martin MU. Interleukin 33 is a guardian of barriers and a local alarmin. Nat Immunol. 2016;17:122– 131. 18. Ali S, Mohs A, Thomas M, et al. The dual function cytokine IL-33 interacts with the transcription factor NFkappaB to dampen NF-kappaB-stimulated gene transcription. J Immunol. 2011;187:1609–1616.
1013
Clinical Therapeutics 19. Kayamuro H, Yoshioka Y, Abe Y, et al. Interleukin-1 family cytokines as mucosal vaccine adjuvants for induction of protective immunity against influenza virus. J Virol. 2010; 84:12703–12712. 20. Talabot-Ayer D, Calo N, Vigne S, et al. The mouse interleukin (Il)33 gene is expressed in a cell type- and stimulus-dependent manner from two alternative promoters. J Leukoc Biol. 2012;91:119–125. 21. Carriere V, Roussel L, Ortega N, et al. IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc Natl Acad Sci U S A. 2007;104:282–287. 22. Roussel L, Erard M, Cayrol C, Girard JP. Molecular mimicry between IL-33 and KSHV for attachment to chromatin through the H2A-H2B acidic pocket. EMBO Rep. 2008;9:1006–1012. 23. Rostan O, Arshad MI, PiquetPellorce C, et al. Crucial and diverse role of the interleukin-33/ST2 axis in infectious diseases. Infect Immun. 2015;83:1738–1748. 24. Liew FY, Pitman NI, McInnes IB. Disease-associated functions of IL33: the new kid in the IL-1 family. Nat Rev Immunol. 2010;10:103–110. 25. Luthi AU, Cullen SP, McNeela EA, et al. Suppression of interleukin-33 bioactivity through proteolysis by apoptotic caspases. Immunity. 2009; 31:84–98. 26. Arshad MI, Piquet-Pellorce C, Samson M. IL-33 and HMGB1 alarmins: sensors of cellular death and their involvement in liver pathology. Liver Int. 2012;32:1200– 1210. 27. Cayrol C, Girard JP. The IL-1-like cytokine IL-33 is inactivated after maturation by caspase-1. Proc Natl Acad Sci U S A. 2009;106:9021– 9026. 28. Hayakawa M, Hayakawa H, Matsuyama Y, et al. Mature interleukin-33 is produced by calpain-mediated cleavage in vivo.
1014
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
Biochem Biophys Res Commun. 2009; 387:218–222. Lefrancais E, Roga S, Gautier V, et al. IL-33 is processed into mature bioactive forms by neutrophil elastase and cathepsin G. Proc Natl Acad Sci U S A. 2012;109:1673–1678. Ohno T, Oboki K, Kajiwara N, et al. Caspase-1, caspase-8, and calpain are dispensable for IL-33 release by macrophages. J Immunol. 2009;183: 7890–7897. Roy A, Ganesh G, Sippola H, et al. Mast cell chymase degrades the alarmins heat shock protein 70, biglycan, HMGB1, and interleukin33 (IL-33) and limits dangerinduced inflammation. J Biol Chem. 2014;289:237–250. Arshad MI, Piquet-Pellorce C, Filliol A, et al. The chemical inhibitors of cellular death, PJ34 and Necrostatin-1, down-regulate IL-33 expression in liver. J Mol Med (Berl). 2015;93:867–878. Gao K, Li X, Zhang L, et al. Transgenic expression of IL-33 activates CD8(þ) T cells and NK cells and inhibits tumor growth and metastasis in mice. Cancer Lett. 2013;335:463–471. Villarreal DO, Wise MC, Walters JN, et al. Alarmin IL-33 acts as an immunoadjuvant to enhance antigen-specific tumor immunity. Cancer Res. 2014;74:1789–1800. Miller AM, Asquith DL, Hueber AJ, et al. Interleukin-33 induces protective effects in adipose tissue inflammation during obesity in mice. Circ Res. 2010;107:650–658. Miller AM, Xu D, Asquith DL, et al. IL-33 reduces the development of atherosclerosis. J Exp Med. 2008; 205:339–346. Zaiss MM, Kurowska-Stolarska M, Bohm C, et al. IL-33 shifts the balance from osteoclast to alternatively activated macrophage differentiation and protects from TNF-alpha-mediated bone loss. J Immunol. 2011;186:6097–6105. Liang Y, Jie Z, Hou L, et al. IL-33 promotes innate IFN-gamma
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
production and modulates dendritic cell response in LCMVinduced hepatitis in mice. Eur J Immunol. 2015;45:3052–3063. Villarreal DO, Svoronos N, Wise MC, et al. Molecular adjuvant IL-33 enhances the potency of a DNA vaccine in a lethal challenge model. Vaccine. 2015;33:4313–4320. Li S, Zhu FX, Zhang HB, et al. Pretreatment with interleukin-33 reduces warm hepatic ischemia/reperfusion injury in mice. Chin Med J (Engl). 2013;126:1855–1859. Sakai N, Van Sweringen HL, Quillin RC, et al. Interleukin-33 is hepatoprotective during liver ischemia/reperfusion in mice. Hepatology. 2012; 56:1468–1478. Volarevic V, Mitrovic M, Milovanovic M, et al. Protective role of IL-33/ST2 axis in Con A-induced hepatitis. J Hepatol. 2012;56:26–33. Yin H, Li X, Hu S, et al. IL-33 promotes Staphylococcus aureusinfected wound healing in mice. Int Immunopharmacol. 2013;17:432–438. Hazlett LD, McClellan SA, Barrett RP, et al. IL-33 shifts macrophage polarization, promoting resistance against Pseudomonas aeruginosa keratitis. Invest Ophthalmol Vis Sci. 2010;51:1524–1532. Le HT, Tran VG, Kim W, et al. IL-33 priming regulates multiple steps of the neutrophil-mediated anti-Candida albicans response by modulating TLR and dectin-1 signals. J Immunol. 2012;189:287–295. Sesti-Costa R, Silva GK, ProencaModena JL, et al. The IL-33/ST2 pathway controls coxsackievirus B5induced experimental pancreatitis. J Immunol. 2013;191:283–292. Monticelli LA, Osborne LC, Noti M, et al. IL-33 promotes an innate immune pathway of intestinal tissue protection dependent on amphiregulin-EGFR interactions. Proc Natl Acad Sci U S A. 2015;112: 10762–10767. Duan L, Chen J, Zhang H, et al. Interleukin-33 ameliorates experimental
Volume 38 Number 5
M.I. Arshad et al.
49.
50.
51.
52.
53.
54.
55.
56.
57.
colitis through promoting Th2/ Foxp3(þ) regulatory T-cell responses in mice. Mol Med. 2012;18: 753–761. Grobeta P, Doser K, Falk W, et al. IL-33 attenuates development and perpetuation of chronic intestinal inflammation. Inflamm Bowel Dis. 2012;18:1900–1909. Humphreys NE, Xu D, Hepworth MR, et al. a potent inducer of adaptive immunity to intestinal nematodes. J Immunol. 2008;180: 2443–2449. Alves-Filho JC, Sonego F, Souto FO, et al. Interleukin-33 attenuates sepsis by enhancing neutrophil influx to the site of infection. Nat Med. 2010; 16:708–712. Luo Y, Zhou Y, Xiao W, et al. Interleukin-33 ameliorates ischemic brain injury in experimental stroke through promoting Th2 response and suppressing Th17 response. Brain Res. 2015;1597:86–94. Canbaz D, Utsch L, Logiantara A, et al. IL-33 promotes the induction of immunoglobulin production after inhalation of house dust mite extract in mice. Allergy. 2015;70: 522–532. Talabot-Ayer D, Martin P, Seemayer CA, et al. Immune-mediated experimental arthritis in IL-33 deficient mice. Cytokine. 2014;69:68–74. Palmer G, Talabot-Ayer D, Lamacchia C, et al. Inhibition of interleukin-33 signaling attenuates the severity of experimental arthritis. Arthritis Rheum. 2009;60: 738–749. Arshad MI, Piquet-Pellorce C, L’Helgoualc’h A, et al. TRAIL but not FasL and TNFalpha, regulates IL-33 expression in murine hepatocytes during acute hepatitis. Hepatology. 2012;56:2353–2362. Hung LY, Lewkowich IP, Dawson LA, et al. IL-33 drives biphasic IL-13 production for noncanonical Type 2 immunity against hookworms. Proc Natl Acad Sci U S A. 2013;110:282– 287.
May 2016
58. Kropf P, Herath S, Klemenz R, Muller I. Signaling through the T1/ ST2 molecule is not necessary for Th2 differentiation but is important for the regulation of type 1 responses in nonhealing Leishmania major infection. Infect Immun. 2003;71: 1961–1971. 59. Chuang CC, Chen CW, Huang YT, Du WY. Anti-ST2 monoclonal antibody inhibits eosinophil infiltration in Angiostrongylus cantonensisinfected mice. J Microbiol Immunol Infect. 2016;49:91–96. 60. Li P, Lin W, Zheng X. IL-33 neutralization suppresses lupus disease in lupus-prone mice. Inflammation. 2014; 37:824–832. 61. Lei Y, Boinapally V, Zoltowska A, et al. Vaccination against IL-33 Inhibits Airway Hyperresponsiveness and Inflammation in a House Dust Mite Model of Asthma. PLoS One. 2015;10:e0133774. 62. Liu X, Li M, Wu Y, et al. Anti-IL-33 antibody treatment inhibits airway inflammation in a murine model of allergic asthma. Biochem Biophys Res Commun. 2009;386:181–185. 63. Mizutani N, Nabe T, Yoshino S. Interleukin-33 and alveolar macrophages contribute to the mechanisms underlying the exacerbation of IgE-mediated airway inflammation and remodelling in mice. Immunology. 2013;139:205–218. 64. Kim YH, Yang TY, Park CS, et al. Anti-IL-33 antibody has a therapeutic effect in a murine model of allergic rhinitis. Allergy. 2012;67: 183–190. 65. Oboki K, Ohno T, Kajiwara N, et al. IL-33 and IL-33 receptors in host defense and diseases. Allergol Int. 2010;59:143–160. 66. Ramaprakash H, Shibata T, Duffy KE, et al. Targeting ST2L potentiates CpG-mediated therapeutic effects in a chronic fungal asthma model. Am J Pathol. 2011;179:104– 115. 67. Qiu C, Li Y, Li M, et al. Antiinterleukin-33 inhibits cigarette
68.
69.
70.
71.
72.
73.
74.
75.
76.
77.
smoke-induced lung inflammation in mice. Immunology. 2013;138:76–82. Kondo Y, Yoshimoto T, Yasuda K, et al. Administration of IL-33 induces airway hyperresponsiveness and goblet cell hyperplasia in the lungs in the absence of adaptive immune system. Int Immunol. 2008;20:791– 800. McSorley HJ, Blair NF, Smith KA, et al. Blockade of IL-33 release and suppression of type 2 innate lymphoid cell responses by helminth secreted products in airway allergy. Mucosal Immunol. 2014;7:1068– 1078. Li D, Guabiraba R, Besnard AG, et al. IL-33 promotes ST2dependent lung fibrosis by the induction of alternatively activated macrophages and innate lymphoid cells in mice. J Allergy Clin Immunol. 2014;134:e1411. Monticelli LA, Sonnenberg GF, Artis D. Innate lymphoid cells: critical regulators of allergic inflammation and tissue repair in the lung. Curr Opin Immunol. 2012;24:284– 289. Saravia J, You D, Shrestha B, et al. Respiratory syncytial virus disease is mediated by age-variable IL-33. PLoS Pathog. 2015;11:e1005217. Akcay A, Nguyen Q, He Z, et al. IL-33 exacerbates acute kidney injury. J Am Soc Nephrol. 2011;22: 2057–2067. Chen J, Duan L, Xiong A, et al. Blockade of IL-33 ameliorates Con A-induced hepatic injury by reducing NKT cell activation and IFN-gamma production in mice. J Mol Med (Berl). 2012;90:1505–1515. Liang Y, Jie Z, Hou L, et al. IL-33 induces nuocytes and modulates liver injury in viral hepatitis. J Immunol. 2013;190:5666–5675. Pushparaj PN, Li D, Komai-Koma M, et al. Interleukin-33 exacerbates acute colitis via interleukin-4 in mice. Immunology. 2013;140:70–77. Zhu J, Yang F, Sang L, et al. IL-33 Aggravates DSS-induced acute
1015
Clinical Therapeutics
78.
79.
80.
81.
82.
83.
84.
85.
colitis in mouse colon lamina propria by enhancing Th2 cell responses. Mediators Inflamm. 2015; 2015:913041. Guabiraba R, Besnard AG, Menezes GB, et al. IL-33 targeting attenuates intestinal mucositis and enhances effective tumor chemotherapy in mice. Mucosal Immunol. 2014;7: 1079–1093. Judd LM, Heine RG, Menheniott TR, et al. Elevated IL-33 expression is associated with paediatric eosinophilic esophagitis, and exogenous IL-33 promotes eosinophilic esophagitis development in mice. Am J Physiol Gastrointest Liver Physiol. 2016;310:G13–G25. Li M, Li Y, Liu X, et al. IL-33 blockade suppresses the development of experimental autoimmune encephalomyelitis in C57BL/6 mice. J Neuroimmunol. 2012;247: 25–31. Reichenbach DK, Schwarze V, Matta BM, et al. The IL-33/ST2 axis augments effector T-cell responses during acute GVHD. Blood. 2015;125:3183–3192. Malcolm J, Awang RA, Oliver-Bell J, et al. IL-33 exacerbates periodontal disease through induction of RANKL. J Dent Res. 2015;94:968– 975. Yu Y, Deng W, Lei J. Interleukin-33 promotes Th2 immune responses in infected mice with Schistosoma japonicum. Parasitol Res. 2015;114: 2911–2918. Rostan O, Gangneux JP, PiquetPellorce C, et al. The IL-33/ST2 axis is associated with human visceral leishmaniasis and suppresses Th1 responses in the livers of BALB/c mice infected with Leishmania donovani. MBio. 2013;4: e00383–e00313. Han P, Zhao J, Liu SB, et al. Interleukin-33 mediates formalininduced inflammatory pain in mice. Neuroscience. 2013;241:59– 66.
1016
Address correspondence to: Michel Samson, PhD, INSERM-U1085, IRSET, Université de Rennes 1, 2 Avenue du Professeur Léon Bernard, 35043 Rennes Cedex, France.. E-mail:
[email protected]
Volume 38 Number 5
M.I. Arshad et al.
GLOSSARY OF ABBREVIATIONS AP-1: Activator protein-1 AHR: Airway hyper responsiveness AREG: Amphiregulin AST: Aspartate aminotransferase ALT: Alanine aminotransferase BALF: Broncho alveolar lavage fluid ConA: Concanavalin A CLP: Cecal ligation puncture CTLs: Cytotoxic T-cells DSS: Dextran sulphate sodium HMGB1: High-mobility group box 1 HSPs: Heat shock proteins HDM: House dust mite HPV: Human papilloma virus IL-33: Interleukin-33 IL-1RAcP: Interleukin-1 receptor accessory protein IRAK: Inteleukin-1 receptor (IL-1R) associated kinase
May 2016
ILCs: Innate lymphoid cells I/R: Ischemia-reperfusion TRAF6: Tumor necrosis factor receptor (TNFR) associated factor-6 LCMV: Lymphocytic chorio-meningitis virus LLC: Lewis lung carcinoma MAPK: Mitogen activated protein kinase MPO: Myeloperoxidase MRSA: Methicillin resistant Staphylococcus aureus MyD88: Myeloid differentiation primary response protein 88 NF-HEV: Nuclear factor of high endothelial venules NF-κB: Nuclear factor- κB OVA: Ovalbumin ROS: Reactive oxygen species RSV: Respiratory syncytial virus SLE: Systemic lupus erythematosus
1016.e1