IRAK-M Associates with Susceptibility to Adult-Onset Asthma and Promotes Chronic Airway Inflammation

This information is current as Yi Liu, Mingqiang Zhang, Lili Lou, Lun Li, Youming of September 28, 2021. Zhang, Wei Chen, Weixun Zhou, Yan Bai and Jinming Gao J Immunol published online 7 January 2019 http://www.jimmunol.org/content/early/2019/01/04/jimmun ol.1800712 Downloaded from

Supplementary http://www.jimmunol.org/content/suppl/2019/01/04/jimmunol.180071 Material 2.DCSupplemental http://www.jimmunol.org/ Why The JI? Submit online.

• Rapid Reviews! 30 days* from submission to initial decision

• No Triage! Every submission reviewed by practicing scientists

• Fast Publication! 4 weeks from acceptance to publication

by guest on September 28, 2021 *average

Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2019 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published January 7, 2019, doi:10.4049/jimmunol.1800712 The Journal of Immunology

IRAK-M Associates with Susceptibility to Adult-Onset Asthma and Promotes Chronic Airway Inflammation

Yi Liu,*,†,1 Mingqiang Zhang,*,1 Lili Lou,* Lun Li,* Youming Zhang,‡ Wei Chen,x Weixun Zhou,{ Yan Bai,‖ and Jinming Gao*

IL-1R–associated (IRAK)-M regulates lung immunity during asthmatic airway inflammation. However, the regulatory effect of IRAK-M differs when airway inflammation persists. A positive association between IRAK-M polymorphisms with childhood asthma has been reported. In this study, we investigated the role of IRAK-M in the susceptibility to adult-onset asthma and in chronic airway inflammation using an animal model. Through genetic analysis of IRAK-M polymorphisms in a cohort of adult-onset asthma patients of Chinese Han ethnicity, we identified two IRAK-M single nucleotide polymorphisms, rs1624395 and rs1370128, genetically associated with adult-onset asthma. Functionally, the top-associated rs1624395, with an enhanced affinity to

the transcription factor c-Jun, was associated with a higher expression of IRAK-M mRNA in blood monocytes. In contrast to the Downloaded from protective effect of IRAK-M in acute asthmatic inflammation, we found a provoking impact of IRAK-M on chronic asthmatic inflammation. Following chronic OVA stimulation, IRAK-M knockout (KO) mice presented with significantly less inflammatory cells, a lower Th2 cytokine level, a higher IFN-g concentration, and increased percentage of Th1 cells in the lung tissue than wild type mice. Moreover, lung dendritic cells (DC) from OVA-treated IRAK-M KO mice expressed a higher percentage of costim- ulatory molecules PD-L1 and PD-L2. Mechanistically, in vitro TLR ligation led to a greater IFN-g production by IRAK-M KO

DCs than wild type DCs. These findings demonstrated a distinctive role of IRAK-M in maintaining chronic Th2 airway inflam- http://www.jimmunol.org/ mation via inhibiting the DC-mediated Th1 activation and indicated a complex role for IRAK-M in the initiation and progression of experimental allergic asthma. The Journal of Immunology, 2019, 202: 000–000.

sthma is a heterogeneous disease characterized by of IL-1R–associated kinase (IRAK) on human chronic airway inflammation and airway remodeling 12q14.2 is one of the most consistently replicated regions linked A (1, 2), affecting patients of all ages (3, 4). Genome-wide to asthma or asthma-related traits in diverse populations (9, 10). A association studies and positional cloning have identified mul- positive association between IRAK-M SNPs and early-onset tiple single nucleotide polymorphisms (SNPs) of several asthma- asthma was reported in an Italian population (11). However, ge- by guest on September 28, 2021 associated expressing in airway epithelium, illustrating the netic studies on IRAK-M SNPs in asthma or atopy have critical role of the respiratory epithelial barrier in the pathogenesis yielded inconsistent results in children and adult patients in of asthma (5, 6). Epithelial cells, in conjunction with type 2 innate different populations (12–14). In addition to asthma, several lymphoid cells, dendritic cells (DCs), macrophages, and other cell IRAK-M SNPs have been implicated in other acute inflammatory types, constitute an innate immune network in the airway. Acti- diseases, such as acute lung injury (15). vation of this immune network regulates the Th2/Th17–dominant IRAK-M, primarily expressed by DCs, macrophages, and airway adaptive immune response (7). A predisposition to asthma was epithelium, is an intracellular negative regulator of TLRs that shown to be related to the impaired host innate immunity (8). regulates the innate immune homeostasis (11, 16). Evolving evi- Moreover, genome-wide association studies reveal that the locus dence has shown IRAK-M also participates in asthmatic immune

*Department of Respiratory Diseases, Peking Union Medical College Hospital, Chinese tions; Y.B. reviewed the experimental data and edited the manuscript for important Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; intellectual contents; Y.Z. designed the experiments, reviewed the experimental data, †Department of Respiratory Medicine, Civil Aviation General Hospital, Beijing 100123, and edited the manuscript for important intellectual contents; and J.G. designed and China; ‡Genomics Medicine Section, National Heart and Lung Institute, Imperial College supervised this study, interpreted the experimental findings, and drafted the manu- London, London SW3 6LY, United Kingdom; xDepartment of Cardiology, Peking Union script. All authors were involved in experimental design, data interpretation, and Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union manuscript preparation. All authors have read and approved the final version of this Medical College, Beijing 100730, China; {Department of Pathology, Peking Union Med- manuscript. ical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical ‖ Address correspondence and reprint requests to Prof. Jinming Gao, Department College, Beijing 100730, China; and Division of Pulmonary and Critical Care Medicine, of Respiratory Diseases, Peking Union Medical College Hospital, Number 1 Department of Internal Medicine, Brigham and Women’s Hospital, Harvard Medical Shuaifuyuan, Dongcheng District, Beijing 100730, China. E-mail address: School, Boston, MA 02115 [email protected] 1Y.L. and M.Z. equally contributed to this work. The online version of this article contains supplemental material. ORCIDs: 0000-0001-6226-1773 (L. Li); 0000-0001-5403-4044 (Y.Z.); 0000-0002- Abbreviations used in this article: a-SMA, a-smooth muscle actin; BAL, 7459-7343 (W.Z.); 0000-0002-8393-3062 (Y.B.). bronchoalveolar lavage; BMDC, bone marrow–derived DC; BMDM, bone marrow– Received for publication May 21, 2018. Accepted for publication November 30, derived macrophage; DC, dendritic cell; FEV1, forced expiratory volume in the first 2018. second; IRAK, IL-1R–associated kinase; KO, knockout; Mch, methacholine; MT, Masson trichrome; PAS, periodic acid–Schiff; PD-L1, programmed death–ligand 1; This work was supported by grants from National Natural Sciences Foundation of PD-L2, programmed death–ligand 2; qRT-PCR, quantitative real-time–PCR; SNP, single China (81170040, 81470229, and 30470767, all to J.G.) and National Institutes of nucleotide polymorphism; Treg, regulatory T; TSLP, thymic stromal lymphopoietin; WT, Health Grant K08HL135443 (to Y.B.). The sponsor had no role in study design, data wild type. collection and interpretation, and manuscript draft. Y.L., M.Z., and L. Lou performed the experiments; L. Li handled experimental Copyright Ó 2019 by The American Association of Immunologists, Inc. 0022-1767/19/$37.50 animals; W.C. facilitated the experiments; W.Z. performed the pathological evalua-

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1800712 2 IRAK-M AND CHRONIC ASTHMA responses. For instance, the expression of IRAK-M was upregu- Quantitative RT-PCR analysis for mRNA expression of lated in peripheral blood leukocytes in asthmatic children (8), IRAK-M in circulating monocytes in asthmatics and high mRNA expression of IRAK-M in sputum was a marker Total RNA was extracted from PBMCs in asthmatics using TRIzol reagents for frequent exacerbations of asthma (17). A unique feature of (Invitrogen) and then reverse transcribed into cDNA using M-MLV Reverse IRAK-M–mediated regulation is that it varies in a disease stage– Transcription System (Promega). Quantitative real-time PCR (qRT-PCR) dependent manner (18). Accumulating evidence from us and was employed to analyze mRNA expression of IRAK-M by a Bio-Rad others indicates that IRAK-M may exert contradictory roles, in- CFX96 Touch Real-Time PCR system using PowerUp SYBR Green Master Mix according to the manufacturer’s instructions (Applied Bio- hibitory or activating, in inflammatory and immune responses, systems). The housekeeping gene GAPDH was used as internal control to depending on the duration of stimulation and the kind of stimuli. normalize IRAK-M mRNA expression. All reactions were carried out in In our previous study using an animal model, we demonstrated triplicate. The qRT-PCR primers were as follows: IRAK-M transcript that IRAK-M knockout (KO) enhanced the acute airway inflam- variant 1 and 2, 59-TCAGAGAAGAGTTATCAGGAAGGTG-39 and 39- AAGCTGATGGAAGATTTAAGCAGTA-59; IRAK-M transcript variant 2, mation in response to 3 days of cigarette smoking but attenuated 59-TGGCTGGATGTTCGTCATATTG-39 and 39-CTCCTGGAGGACC- the chronic inflammatory responses to 7 weeks of cigarette TGTAAAAGGT-59; and GAPDH,59-GGAGCGAGATCCCTCCAAAAT- smoking via differentially modulating the function of regulatory T 39 and 39-GGCTGTTGTCATACTTCTCATGG-59. (Treg) cells, Th17 cells, DCs, and macrophages (19). Similarly, EMSA others have demonstrated IRAK-M KO reduced the acute in- flammatory responses to a noninfectious injury and subsequent Allele-specific alteration in rs1624395 bindingtotranscriptionfactorc-Junwas fibrosis (20, 21). By contrast, overexpression of airway epithelial assayed by using EMSA. Nuclear protein c-Jun was prepared from HeLa cells transfected with an expression pET-28a-His-MBP vector for c-Jun. EMSA were Downloaded from IRAK-M by IL-13 stimulation impaired the innate immunity and performed using dsDNA probes. Sequences of the oligonucleotides used were increased susceptibility to respiratory infection (22, 23). In par- as follows: for IRAK-M rs1624395 allele A, forward 59-ACAGAGCACA- ticular, we have studied the regulatory effect of IRAK-M on the GAGGAGGAAATGATAACTTGGG-39 and reverse 59-CCCAAGTTATCA- asthmatic airway inflammation using the acute OVA mouse model TTTCCTCCTCTGTGCTCTGT-39;forIRAK-M rs1624395 allele G, forward and found that IRAK-M ablation exacerbated airway inflamma- 59-ACAGAGCACAGAGGAGGGAATGATAACTTGGG-39 and reverse 59- CCCAAGTTATCATTCCCTCCTCTGTGCTCTGT-39.The59-biotin–labeled tion by enhancing Th2 and Th17 differentiation (24).These results probes were synthesized by Invitrogen. support the multifactorial roles of IRAK-M signaling in modu- To ensure specificity of DNA–protein binding, an unlabeled probe was http://www.jimmunol.org/ lating inflammatory responses (25). However, the impact of used in a 30-fold molar excess as a binding competitor before incubation IRAK-M on chronic asthmatic inflammation remains unclear. of nuclear protein with biotin-labeled probes. A LightShift Chemilumi- nescent EMSA Kit was used according to manufacturer’s instructions To fill in the knowledge gaps, in the current study, we assessed (Thermo Fisher Scientific). the role of IRAK-M in adult-onset asthma and the development of chronic asthmatic inflammation. First, using samples from a Animals hospital-based asthma cohort with Chinese Han ethnicity, we IRAK-M KO mice have been described previously elsewhere (16). IRAK-M explored the genetic association between IRAK-M SNPs and hu- KO mice (B6.129S1-Irak3tmlFlv/J) were a kind gift by Dr. N.G. Frango- man adult-onset asthma and the functionalities of susceptible giannis (Albert Einstein College of Medicine), and wild type (WT) mice IRAK-M SNPs. Next, using a chronic OVA mouse model, we in- (purchased from Experimental Animal Research Center, Beijing, China) by guest on September 28, 2021 with C57BL/6 background were maintained in a pathogen-free mouse vestigated the regulation of IRAK-M expression following re- facility at Peking Union Medical College Hospital Animal Care Center. peated allergen exposure and the influence of IRAK-M loss on the Ten- to twelve-week-old mice were used for experiments. chronic allergic inflammation and airway remodeling. All experiments were performed according to international and insti- tutional guidelines for animal care and approved by Peking Union Medical Materials and Methods College Hospital Ethics Committee for Animal Experimentation. Participants Chronic asthma mouse model There were a total of 1348 subjects with Han ethnicity from the northern The chronic asthma model was established according to that previ- region of China enrolled in this study. Six hundred and seventy-nine patients ously reported, with minor modifications (24, 27). Briefly, mice were given with adult-onset asthma were recruited from two medical centers. Adult- 50 mg of OVA (Grade V, Sigma-Aldrich) absorbed to 2.25 mg aluminum onset asthma was defined as previously described (26). The asthma pa- hydroxide in 200 ml of sterile saline on days 0 and 13 by i.p. injection, then tients, who had their first symptoms at the age of 18 y or older, were fully challenged with 1% aerosolized OVA via a PARI nebulizer for 30 min characterized by clinical presentation, functional status, and inflammatory daily from day 19 to day 24, then three times per week from day 26 to markers. Asthma patients with forced expiratory volume in the first second day 60. Sham mice received aluminum hydroxide and were exposed to (FEV1) $70% of predicted underwent the methacholine (Mch) challenge 0.9% NaCl solution alone. Mice were sacrificed 24 h after the last test. Patients whose FEV1 was ,70% of predicted underwent an airway challenge for further analysis. reversibility test by b2 agonist. Six hundred and sixty-nine age- and ethnicity-matched controls were selected from nonasthmatic, nonatopic, Airway responsiveness healthy individuals with normal lung function. All participants were un- related. Detailed characteristics of these individuals were listed in Table I. Airway responsiveness to inhaled Mch was determined in mice 24 h after The study protocols were approved by the Ethics Committee for Human the final aerosol challenge, as we have previously described (24, 28). Research of Peking Union Medical College Hospital (S-767) and Beijing Lung histology Aviation General Hospital (MHZYY 2014-05-01), and all subjects gave informed written consent. Mice were sacrificed, and left lungs were taken out and inflated to 25 cm H2O with 10% formalin, fixed overnight, then embedded in paraffin and Genotyping sectioned to 3-mm thickness. H&E, periodic acid–Schiff (PAS), and Masson trichrome (MT) stains were performed to evaluate the severity of Genomic DNA from peripheral blood leukocytes was extracted by stan- airway inflammation, mucus secretion, and collagen deposit. dard protocols. A total of nine markers were genotyped. SNPs were gen- otyped using SNaPshot. The primers for PCR amplification and extension Histopathological scoring of IRAK-M gene are listed in Supplemental Tables 1 and 2. The PCR products were sequenced on an 3730xl DNA Analyzer (Applied Bio- Semiquantitative scoring for airway inflammation in H&E slides was systems) and analyzed using the GeneMapper 4 software. evaluated in a blind manner by counting the inflammatory cells that The genotyping efficiency for IRAK-M SNPs was .95%. Minor allele infiltrated around airways and vessels according to the method of a pre- frequency was .5%. All SNPs tested were in Hardy–Weinberg equilib- viously reported study (29). The scores were as follows: 0 = normal; 1 = rium (p cutoff-value = 0.05) in asthma cases and controls. ,3 cells thick in diameter; 2 = 4–10 cells thick; 3 = .10 cells thick. The Journal of Immunology 3

Table I. Characteristics of subjects included in this study 39-CCACCAAGGCAAGCAAGAG-59;IFN-g,59-GCTGATCCTTTGGACCCT- CT-39 and 39-AGGCTTTCAATGACTGTGCC-59,CTLA-4,59-TCCGGAGG- Clinical Phenotypes Asthma Patients Controls TACAAAGCTCAACTG-39 and 39-ACCATGGCTGCTAGCCAACAC-59;TGF- b1, 59-AATGGTGGACCGCAACAAC-39 and 39-GCACTGCTTCCCGAATGT- Number of subjects 679 669 C-59; and GAPDH, 59-TTGTCTCCTGCGACTTCAACA-39 and 39-TGGTCC- Mean age (y) 42.3 6 14.4 39.4 6 11.9 AGGGTTTCTTACTCC-59. Male, n (%) 285 (41.9) 284 (42.5) Commercial ELISA kits were employed to detect concentrations of IL-4, Smoke history (%) 30.8 16.4 IL-13, and IFN-g in bronchoalveolar lavage (BAL) fluid (R&D Systems) and Blood Eos (%) 5.6 6 4.4* 2.3 6 1.5 levels of IL-17A, IL-33, and thymic stromal lymphopoietin (TSLP) in the su- Serum LnIgE 5.0 6 1.5* 3.2 6 1.3 pernatant of lung homogenates (eBioscience). Measurements of IL-10, IL-12p40, Atopy, n (%) 498 (73.3) N/A IL-23, and IL-27 were analyzed by LEGENDplex software (BioLegend). Allergic rhinitis, n (%) 467 (68.8) N/A Lung function Hydroxyproline assay parameters Total collagen contents in the lung were determined by analysis of FEV1 (%) 70.2 6 23.9** 101.9 6 12.8 hydroxyproline. Hydroxyproline contents in the lung were measured using a FEV1/FVC (%) 65.2 6 15.1*** 85.4 6 8.2 commercial (Jiancheng Bioengineering Company, Nanjing, China). Positive for airway 155 (22.8) N/A challenge test, n (%) Western blotting Positive for airway 524 (77.2) N/A reversibility test, n (%) The whole protein from the lung was prepared as previously described, with , , , modifications (31). The lungs were harvested and homogenized in lysis *p 0.01, **p 0.0001,***p 0.001. buffer containing a protease inhibitor mixture (Sigma-Aldrich). Cell debris Eos, eosinophil; FVC, forced vital capacity; LnLgE: loge of total serum IgE; 3 Downloaded from N/A, not applicable. was removed by centrifugation at 12,000 g for 10 min at 4˚C. The supernatants were eluted by adding an equal volume of sample loading buffer, then run on 12% SDS–PAGE gels before transferring to PVDF A score of 0–3 on each section was used to reflect overall extent of airway membranes. Membranes were incubated in the presence of the indicated inflammation (0: normal, 1: ,25% of each section, 2: 25–75%, 3: .75%). Abs. The Abs were as follows: anti-mouse IRAK-M (Abcam) and anti- The index was calculated by multiplying severity by overall extent with a mouse a-smooth muscle actin (a-SMA) (Boster Biological Technology, maximal score of 9. Wuhan, China). Primary Ab application was followed by incubation with Mucus scoring was evaluated based on the previously published method HRP-conjugated secondary Abs (rabbit anti-mouse IgG) (Bioleaf Biotech, http://www.jimmunol.org/ (30). Mucous scores were as follows: 0, no mucous; 1, a few cells se- Shanghai, China). Immunopositive blots were visualized using an ECL with creting mucous; 2, many cells secreting mucous; and 3, extensive mucous AlphaEaseFC imaging system (Alpha Innotech, San Leandro, CA) and production. semiquantitatively analyzed with AlphaEase FC software (Alpha Innotech).

Bronchoalveolar lavage fluid collection Flow cytometry analysis Mouse trachea were cannulated with a 20-gauge catheter, through which Flow cytometry analysis was performed using single cell suspension. 0.8 ml of ice-cold PBS (pH 7.4) was used to lavage the lung three times. Single-cell suspension was prepared according to the method in a previ- Therecoveredfluidwascentrifugedat1500rpmfor5minat4˚C. ously described study (24). Briefly, instilled lungs and spleens were Cytospins were prepared from cell pellets. minced and digested with collagenase type 1A and type IV bovine pan-

Cytokine analysis creatic DNase, then passed through a cell strainer to obtain cell suspension. by guest on September 28, 2021 The mononuclear cells from lung and spleen were isolated by centrifu- qRT-PCR was used to reflect mRNA expression of cytokines, including IL-4, gation with Ficoll solution. IL-13, IFN-g, cytotoxic lymphocyte Ag 4 (CTLA-4), and TGF-b1. Total Cells were incubated with fluorochrome (FITC, PE, PerCP-Cyanine5.5, RNA was prepared from the right lung of the animal using TRIzol reagent and and allophycocyanin)–conjugated Abs (anti-Mouse CD4, CD8, ST2, MHC reverse transcribed into cDNA using a commercial kit (Takara Bio). qRT-PCR class II, CD80, CD86, OX40L, Siglec-F, Ly-6G, CD3, OX40L, programmed was performed on the 7500 Fast Real-Time PCR System (Applied Biosystems) death–ligand 1 [PD-L1], and programmed death–ligand 2 [PD-L2]) at room in a 20-ml reaction that contained 2 ml of cDNA and SYBR Premix Ex Taq temperature for 30 to 45 min. Cytokine intracellular staining was performed (Takara Bio). The expression level of GAPDH was used as an internal control. on cells that had been stimulated with 50 ng/ml PMA (Sigma-Aldrich) and The relative expression was calculated with the comparative cycle threshold 1 mM of ionomycin (Sigma-Aldrich) in the presence of GolgiStop (BD method using Applied Biosystems 7500 Real-Time PCR Software v2.0.6 (Ap- Biosciences) for 6 h. The activated cells were fixed, permeabilized, and plied Biosystems) and was expressed as the fold change compared with the stained with mAbs (anti-mouse IFN-g, IL-4, IL-17A, T-bet, Foxp3, and control. The murine primers used to amplify genes were as follows: IRAK-M, ROR-g). All samples were analyzed with Accuri C6 (BD Biosciences). The 59-GGGGCATCAACGAGCTATCC-39 and 39-GGTTCCAGAGGTCCAGG- fluorochrome-conjugated Abs were purchased from eBioscience, BD Bio- GTC-59;IL-4,59-TCGAATGTACCAGGAGCCATAT-39 and 39-GAAGCA- sciences, or BioLegend, and the corresponding isotype control Abs were CCTTGGAAGCCCTA-59;IL-13,59-AGGCCAGCCCACAGTTCTA-39 and added to “isotype samples” at the same concentrations as the Abs of interest.

Table II. Distribution of alleles and genotypes of IRAK-M gene in asthma patients and controls

Alternative versus Alternative Frequency Reference Analysis Genotype Analysisa

95% Odds Confidence Marker Ref./Alt. Case Control p Value Ratio Interval Case Control p Value rs7970350 T/C 0.227 0.244 0.298 1.100 0.919–1.316 394/231/34 373/264/31 0.240 rs6581660 T/G 0.174 0.166 0.598 0.947 0.774–1.158 454/209/13 459/196/13 0.820 rs11836463 C/A 0.093 0.107 0.204 1.179 0.914–1.520 555/104/10 527/115/13 0.468 rs2870784 G/T 0.085 0.093 0.440 1.110 0.852–1.448 571/99/8 553/107/9 0.742 rs2141709 G/A 0.374 0.389 0.444 1.063 0.909–1.243 270/290/104 253/312/104 0.512 rs2701652 G/C 0.112 0.124 0.338 1.122 0.887–1.419 531/142/5 505/146/9 0.447 rs1821777 C/T 0.110 0.129 0.120 1.203 0.953–1.519 540/129/10 509/147/13 0.300 rs1624395 A/G 0.565 0.514 0.008 1.227 1.054–1.428 127/336/215 152/346/171 0.025 rs1370128 T/C 0.550 0.507 0.027 1.187 1.020–1.381 100/407/169 142/375/152 0.009 Bold represents significant value. aThe three values represent the number of individuals carrying major allele homozygote, heterozygote, and mutant allele homozygote. Ref./Alt., reference/alternative. 4 IRAK-M AND CHRONIC ASTHMA

Table III. Association between haplotypes of IRAK-M and asthma

Haplotype rs2701652 rs1821777 rs1624395 rs1370128 Frequency Case: Control Ratio x2 p Value H1 G C G C 0.394 552.8:803.2, 508.0:830.0 2.209 0.1372 H2 G C A T 0.323 417.2:938.8, 454.0:884.0 3.085 0.079 H3 C C G C 0.109 142.9:1213.1, 149.6:1188.4 0.284 0.594 H4 G T A T 0.105 121.5:1234.5, 162.0:1176.0 0.787 0.0079 Bold represents significant value.

Culture and purification of bone marrow–derived DCs and (Table III). This haplotype, therefore, appears to confer protection bone marrow–derived macrophages against the risk of having asthma. Bone marrow was obtained from C57BL/6 mice. The harvested bone Influence of rs1624395 on mRNA expression of IRAK-M in marrow cells, depleted of RBCs, were incubated for 7 d with RPMI 1640 circulating monocytes containing 10% FBS, 100 U/ml penicillin, 100 U/ml streptomycin, 20 ng/ml GM-CSF (for bone marrow–derived DCs [BMDCs]) or 50 ng/ml GM-CSF The asthma-susceptible genes contain polymorphisms that modify (for bone marrow–derived macrophages [BMDMs]). Purification of function. We next studied the functionality of the risk for SNPs BMDCs was used a CD11c magnetic cell-sorting kit (Miltenyi Biotec, of IRAK-M influencing asthma susceptibility. We chose the top- Gladbach, Germany). BMDMs were recovered from the adherent cells by digestion with 5% trypsin. Stimulation of BMDCs/BMDMs was per- associated polymorphism rs1624395, which is in linkage dis- Downloaded from 2 formed with LPS (1 mg/ml) on day 8 for 24 h. equilibrium with rs1370128 (r = 0.9) (Supplemental Fig. 1). We found that rs1624395 influenced the mRNA expression of the Statistical analysis IRAK-M transcript variant 1 in peripheral monocytes from asthma For association between IRAK-M SNPs and asthma susceptibility, all patients (n = 60, AA = 30, AG + GG = 30). As shown in Fig. 1, genotype and phenotype data were formatted using PLINK. Genotype and asthmatics carrying AG/GG genotypes had significantly higher allele frequencies in cases and controls were compared by contingency table analysis. Genotype relative risk was calculated according to the statistical mRNA levels in blood monocytes than the AA genotype carriers. http://www.jimmunol.org/ method described by Lathrop and colleagues (17). Quantile normalization Transcription factor binding property modulated by rs1624395 was applied to all quantitative traits. Linkage disequilibrium and haplo- types were assessed using the Haploview version 4.0 (32). All markers To define the possible molecular mechanism underlying the dif- were included in these analyses on a gene-by-gene basis. ference in mRNA expression of IRAK-M by PBMCs in asthmatics Comparisons of the animal data were performed using one-way ANOVA carrying the rs1624395 A or G allele, we used EMSA analysis to with a Tukey post hoc test for multiple groups and Student t test for two groups with GraphPad Prism Version 5.01 (GraphPad Software, La Jolla, detect the binding properties of rs1624395 G allele and A allele to CA). Data are expressed as means 6 SEM, and a p value ,0.05 was NF C-Jun. c-Jun has been demonstrated to directly bind the human considered significant. IRAK-M promoter to upregulate epithelial IRAK-M expression

(22). Using the 59-biotinylated probe against rs1624395 to coin- by guest on September 28, 2021 Results cubate with NF c-Jun protein from human HeLa cells, we dem- IRAK-M SNPs and susceptibility to adult-onset asthma onstrated significantly greater affinity for allele G than allele A Given the positive association of IRAK-M polymorphism with the (Fig. 2A, 2B). susceptibility to early-onset asthma in children (11), we examined the impact of IRAK-M SNPs on the risk of developing adult-onset asthma (Table I). We selected nine IRAK-M SNPs that were studied in early-onset asthma (11). After correction for multiple tests, we found two IRAK-M SNPs, rs1624395 and rs1370128, were signif- icantly associated with adult-onset asthma (odds ratio = 1.227, 95% confidence interval = 1.054 –1.428, p = 0.008 for rs1624395; odds ratio = 1.187, 95% confidence interval = 1.020 –1.381, p = 0.027 for rs1370128). Homozygote G/G for rs1624395 and homozygote C/C for rs1370128 were more often seen in adult-onset asthma patients than in controls (p values were 0.025 and 0.009, respectively) (Table II). We also demonstrated the associations between poly- morphic markers in IRAK-M locus and numerous quantitative asthma-related traits, for instance the marker rs7970350 with high blood eosinophil counts (p , 0.05), rs2141709 with airflow ob- struction (FEV1% and FEV1/forced vital capacity%, all p , 0.05), rs1821777 with FEV1% (p , 0.01) and serum total IgE level (p , 0.05), and rs1624395 with serum total IgE level (p , 0.05). Haplotypes analysis and linkage disequilibrium tests Haplotype analyses of all markers on a gene-by-gene basis (in- cluding haplotypes present in $5% of the genotyped population) FIGURE 1. mRNA expression of IRAK-M carrying rs1624395 in cir- revealed the significant association between one IRAK-M haplo- 2 culating monocytes of asthmatics. The allele-specific RNA expression of type and asthma after 1000 permutations (GTAT, x = 0.787, two transcriptional variants of IRAK-M carrying rs1624395 was quantified permutation p = 0.0079). This haplotype was significantly under- in PBMCs from asthma patients. Each sample was analyzed in triplicate. represented in asthmatic cases in comparison with controls, with The results were normalized to the expression level of GADPH. For AA case and control frequencies being 0.098 and 0.138, respectively group, n = 30; for AG/GG, n = 30. Data were expressed as mean 6 SEM. The Journal of Immunology 5 Downloaded from

FIGURE 2. Enhanced binding of rs1642395 allele G to the nuclear protein c-Jun. (A). A representative image showing the A or G allele of rs1642395 bound to the c-Jun from HeLA cells in the absence of competitor (Lane 3 and 7) or in the presence of cold probe (Lane 4 and 8) or mutant probe (Lane 5 and 9) by EMSA analysis. (B). The summary plot of lane intensity to reflect the quantity of binding complex of c-Jun and allele A or G. The result was normalized to the intensity of the total signal in the corresponding lanes. Data were expressed as mean 6 SEM and quantified for three replicates from HeLA cells in each column. http://www.jimmunol.org/

IRAK-M expression in the mouse lungs following chronic and chronic inflammatory phases (25). In this study, we further OVA exposure assessed the impact of IRAK-M on chronic asthma, with a sim- In an acute asthma mouse model, we have shown significantly ulated model established by repeated OVA administration higher mRNA expression of IRAK-M in the lungs exposed to OVA (Fig. 3A) to WT or IRAK-M KO mice. We first assessed the (around 50-fold) (24). We further examined the change of IRAK-M airway inflammation by examining the epithelial thickness and the expression in a chronic asthma mouse model sensitized and chal- inflammatory cell infiltration in the airway walls. We found no lenged by OVA for 2 mo (Fig. 3A). The expression of IRAK-M inflammation around the airways in PBS-treated WT or IRAK-M by guest on September 28, 2021 mRNA was significantly higher (by ∼100-fold) in mouse lungs KO mice (Fig. 4A, left panel). Following chronic OVA challenge, with chronic OVA treatment than those with PBS treatment WT mice showed worse pathological characteristics of airway (Fig. 3B, p , 0.01). Consistently, a significantly higher level of inflammation than IRAK-M KO mice did, such as thicker airway lung IRAK-M protein was observed in chronically OVA-treated epithelia, heavier mucus production, and more inflammatory cells mice than PBS-treated mice (Fig. 3C, p , 0.05). around the bronchioles and vessels (Fig. 4A, right panel). We semiquantitatively scored the histopathological findings. The in- Effect of IRAK-M deficiency on chronic airway inflammation flammation scores of WT mice were significantly higher than induced by OVA those of IRAK-M KO mice (3.28 6 0.40 versus 2.47 6 0.39, p , In our previous study, we have demonstrated a clear inhibitory role 0.05) (Fig. 4B). Consistently, the number of total inflammatory of IRAK-M on the acute allergic airway inflammation with OVA cells, eosinophils, neutrophils, lymphocytes, but not macrophages asthmatic mouse model (24). However, this conclusion cannot be in BAL fluid was significantly more in WT mice than IRAK-M KO simply extrapolated to the regulation of chronic allergic inflam- mice (Fig. 4C). Using flow cytometry analysis, we found a sig- mation, as the role of IRAK-M is known to vary between the acute nificantly higher percentage of Th2 cells in the BAL fluid from

FIGURE 3. Increased expression of IRAK-M in lungs of mice after chronic OVA induction. (A). Schematic representation of the protocol to establish the chronic asthma mouse model with OVA stimulation. (B). mRNA expression of IRAK-M in the lung of WT mice following chronic PBS or OVA treatment. IRAK-M gene expression was normalized to GAPDH expres- sion (n = 6–8 mice in each group). *p , 0.01. (C). A representative Western blot image (left panel) and summary plot (right panel) of IRAK-M protein expression in the lung of WT mice fol- lowing chronic OVA or PBS stimulation. IRAK-M protein expression normalized to b-actin expression (n = 3–6 mice in each group). *p , 0.05. 6 IRAK-M AND CHRONIC ASTHMA Downloaded from http://www.jimmunol.org/ by guest on September 28, 2021

FIGURE 4. Attenuated airway inflammation in IRAK-M KO mice after chronic induction of OVA. (A) Representative photomicrographs of H&E-stained lung tissues showing that OVA-sensitized and -challenged WT mice exhibited the typical pathological characteristics of chronic airway inflammation evidenced by thickened airway epithelium and more inflammatory cells in the peribronchial area and around vessels compared with the similarly-treated IRAK-M KO mice. Original magnification 3200. (B) Semiquantitative histopathological scores from PBS- or OVA-treated WT (Figure legend continues) The Journal of Immunology 7 Downloaded from

FIGURE 5. Reduction in airway hyperresponsiveness, mucus production, and airway remodeling in IRAK-M KO mice after chronic induction of OVA. (A) Sensitized and challenged IRAK-M KO mice showed significantly lower airway reactivity reflected by airway resistance in response to increasing doses of inhaled Mch than similarly treated WT mice (n = 8 animals in each group). *p , 0.05. (B) Representative photomicrographs of PAS-stained lung tissues http://www.jimmunol.org/ showing that OVA-sensitized and -challenged IRAK-M KO mice had less mucus hypersecretion than the similarly treated WT mice. Original magnification 3200. Semiquantitative analysis of PAS scoring using the previously described method (n = 6 animals each group). *p , 0.05. (C) Representative images of MT-stained lung tissue showing fewer deposits of mature collagen around the airways of IRAK-M KO mice chronically treated with OVA (left panel). Original magnification 3200. Lung hydroxyproline contents were significantly lower in IRAK-M KO mice chronically treated with OVA than those in similarly challenged WT mice (n = 6 mice per group). *p , 0.05. (D) Representative image of Western blotting for lung a-SMA expression after chronic OVA induction (upper panel). Semiquantitative analysis showing significantly lower expression of a-SMA protein in the lungs from IRAK-M KO after chronic OVA induction (lower panel) (n = 3–6 mice per group). *p , 0.05.

WT mice than that in IRAK-M KO mice (Fig. 4D), even though Impact of IRAK-M deficiency on T cell subsets following the percentages of total T cells or B cells were not different be- chronic OVA stimulation by guest on September 28, 2021 tween the two groups. Asthma is typically characterized by Th2-type inflammation in the Effect of IRAK-M ablation on airway remodeling after chronic airways (7). However, the immune regulation of asthmatic inflam- OVA stimulation mation involves a complex entity of T cell subtypes. Therefore, we further characterized the influence of IRAK-M KO on the compo- Airway remodeling is one of characteristics of chronic asthma and sition of T subsets in mouse lung homogenates and spleens of the is related to airway obstruction (2, 33). We subsequently examined chronic OVA model. As shown in Fig. 6A, chronic OVA challenge the impact of IRAK-M KO on airway reactivity and airway resulted in an increased Th2 cell percentage and a reduced Th1 cell remodeling in mice of the chronic OVA model. Following chronic percentage in both WT and IRAK-M KO mouse lungs. However, the OVA challenge, WT and IRAK-M KO mice developed increased changes in Th1 cell and Th2 cell percentage were significantly less airway resistance in response to the increasing concentrations of in IRAK-M KO mouse lungs than WT mouse lungs. Additionally, we Mch. Of note, the airway resistance was significantly higher in observed significantly higher percentage of Th1 cells in the spleens WT mice as compared with the IRAK-M KO mice (p , 0.05) from OVA-treated IRAK-M KO mice than WT mice (Fig. 6B). (Fig. 5A). Moreover, chronic OVA treatment significantly increased the Th17 To reflect the effect of IRAK-M deficiency on airway pathology and Treg cell percentage in both IRAK-M KO and WT mouse lungs, (33), we performed PAS scorings, MT staining, and a-SMA ex- with no significant intergroup difference (Fig. 6A, 6B). pression. We found the following after chronic OVA challenge: 1) more severe mucus hypersecretion in the airways from the WT mice than that from IRAK-M KO mice (Fig. 5B), 2) significant Effect of IRAK-M deficiency on cytokine production in higher PAS scores in WT mice than IRAK-M KO mice (2.78 6 response to chronic OVA stimulation 0.44 versus 1.84 6 0.38, p , 0.05) (Fig. 5B), 3) more collagen Asthmatic airway inflammation is mediated by cytokines produced deposition in the peribronchiolar area by MT staining and higher from structural and immune cells (1). To understand the mechanisms by hydroxyproline content in the WT lungs than IRAK-M KO lungs which IRAK-M supports the Th2 immune responses following chronic (0.60 6 0.11 mg/mg versus 0.41 6 0.09 mg/mg, p , 0.05) OVA stimulation, we examined the levels of both proinflammatory and (Fig. 5C), and 4) higher expression of a-SMA in WT lungs than anti-inflammatory cytokines in BAL fluid and lungs of chronically IRAK-M KO lungs by Western blotting assay (Fig. 5D). immunized and challenged WT and IRAK-M KO mice.

and IRAK-M KO mice (n = 6 mice in each group). *p , 0.05. (C) Total inflammatory cells and differential subpopulations in BAL fluids. *p , 0.05. (D)A representative image (left panel) and summary plots (right panel) showing the flow cytometry analysis of T cells, B cells, and Th2 cell percentage in the BAL fluid from WT or IRAK-M KO mice following chronic OVA stimulation (n = 7–10 mice in each group). *p , 0.05. 8 IRAK-M AND CHRONIC ASTHMA Downloaded from http://www.jimmunol.org/ by guest on September 28, 2021

FIGURE 6. Decreased Th2-mediated immunity in IRAK-M KO mice following chronic OVA stimulation. Representative image (left panel) and summary plots (right panel) of flowcytometry analysis of Th1, Th2, Th17, and Treg cell percentage in the lungs (A) and spleen (B)ofWTorIRAK-M KO mice following chronic PBS or OVA stimulation (n = 10 animals in each group). *p , 0.05. The Journal of Immunology 9

The airway epithelium constitutes the first line of defense against we found that chronic OVA-stimulation caused significantly the environmental stimuli. Upon exposure to an inhaled allergen, higher levels of IL-12p40, IL-23, and IL-27 in BAL fluid from airway epithelial cells release the cytokines to induce resident DC IRAK-M KO mice than those from WT mice. At mRNA level, maturation and further skew the naive T cells differentiation to Th2 lung expression of IL-4 and IL-13 was significantly increased in type (1, 34). In the acute mouse model of asthma, we have dem- WT mouse lungs but not in IRAK-M KO mouse lungs by chronic onstrated OVA-induced elevation of epithelium-derived Th2 in- OVA stimulation. By contrast, mRNA expression of IFN-g and structive cytokines, IL-17A, IL-33, and TSLP, with a higher level in CLTA-4 in the lungs was significantly elevated in IRAK-M KO the lungs of IRAK-M KO mice than WT mice (24). However, we mice (Fig. 7C). TGF-b1, increased in the airways of patients didn’t find a significant difference in the elevated concentrations of with asthma (35, 36), mediates the association between airway these cytokines in the lung homogenates between WT and IRAK-M remodeling and airway hyperresponsiveness during asthmatic pro- KO mice following chronic OVA administration (Fig. 7A). cess (37). In supporting of this finding, we found that lung ex- At protein level, the concentrations of Th2 cytokines IL-4 and pression of TGF-b1 mRNA was significantly enhanced by chronic IL-10 but not IL-13 were significantly higher in BAL fluid from OVA challenge in WT mice but not in IRAK-M KO mice chronically OVA-treated WT mice, whereas the concentration of (Fig. 7C). Together, these data suggested IRAK-M KO suppressed Th1 cytokine IFN-g was significantly higher in BAL fluid from the Th2 inflammation and promoted Th1 phenotype exampled by chronically OVA-treated IRAK-M KO mice (Fig. 7B). Moreover, a greater INF-g production. Downloaded from http://www.jimmunol.org/ by guest on September 28, 2021

FIGURE 7. IRAK-M KO influence on produc- tion of Th1/Th2 cytokines in the mouse lungs after chronic OVA induction. (A) ELISA analysis for levels of epithelial cytokines IL-33, IL-25, and TSLP in the lung homogenates (n = 6–8 animals in each group). (B) ELISA analysis for levels of various Th1/Th2 cytokines in BAL fluid from WT or IRAK-M KO mice following repeti- tive OVA or PBS stimulation (n = 5–8 animals in each group). *p , 0.05. (C) qRT-PCR analysis for mRNA expression of IL-4, IL-13, TGF-b1, IFN-g, and CLTA-4 in mouse lungs following chronic PBS or OVA treatment (n = 6–10 animals in each group). *p , 0.05. 10 IRAK-M AND CHRONIC ASTHMA

Effect of IRAK-M deficiency on expression of costimulatory not shown). These data support that loss function of IRAK-M in molecules on DCs and macrophages DCs or macrophages favors Th1 promotion. To further understand the mechanism underlying the suppression of chronic allergic airway inflammation by IRAK-M KO, we further Discussion evaluated the function of IRAK-M KO on APCs in the chronic In the present study, we reported an association of two IRAK-M SNPs, asthmatic inflammation. rs1642395 and rs1370128, with susceptibility to adult-onset asthma Airway APCs, mainly DCs and macrophages, play crucial roles and one haplotype (GTAT) of IRAK-M potentially protecting against not only to initiate the specific Th2 cell immune responses but also the risk of asthma. These findings provide evidence for the hy- to restimulate local residential T (memory) lymphocytes and re- pothesis that IRAK-M plays a role in the pathogenesis of adult-onset cruit effector cells following the repeated allergen exposure (1, 34). asthma. It is by far, to our knowledge, the first genetic study of We have previously demonstrated that IRAK-M KO led to a IRAK-M as an adult-onset asthma susceptible gene in Han Chinese downregulation of costimulatory molecules, such as OX40L, ethnicity and provides invaluable supplementary information to the CD80, CD86, and CD124, on the airway APCs in acute asthma comprehensive role of IRAK-M in the pathogenesis of asthma. model of mice (24). However, in the current study of chronic OVA The susceptible IRAK-M SNPs reported in this study are dif- model of asthma, we found that IRAK-M KO did not make a ferent from the original findings from early-onset persistent difference on the expression of costimulatory molecules MHC asthmatic patients (11). Several factors could attribute to the class II, CD80, and CD86 on lung DCs despite a higher expression discrepancy. First, the patient age, ethnicity, and disease stage are of CD80 by airway macrophages of IRAK-M KO mice (Supplemental different among the study groups. Asthma susceptibility is the Fig. 2). Notably, the expression of PD-L1 (CD274) and PD-L2 result of interaction of genetic profiles and environmental factors Downloaded from (CD273) was significantly higher on the lung DCs from IRAK-M at various times throughout the entire life. Some susceptible genes KO mice than WT mice after chronic OVA exposure (Fig. 8A, were reported to cause early-onset mild or intermittent asthma p , 0.05). Similarly, a higher surface expression of PD-L1 (CD274) under the risk of environmental exposure (6). Genetic evidence was revealed on the BAL fluid macrophages from IRAK-M KO has shown different gene expression profiles in the airways be- mice as compared with WT mice (Fig. 8B). tween adult- and childhood-onset severe asthma (38). Moreover, case-control association and next-generation sequencing studies http://www.jimmunol.org/ In vitro effect of IRAK-M KO on activity of BMDC and BMDM have revealed significant discrepancies in the frequencies of SNPs To confirm that the absence of IRAK-M modifies the instructive and haplotype blocks for asthma genes between Chinese and other role of APCs on T cell responses, we examined the cytokines ethnicities (39). Second, the inconsistencies among studies could released by activated WT and IRAK-M KO DCs and macrophages. reflect real differences in the genetic effects of IRAK-M variants We used BMDCs and BMDMs and stimulated them with LPS, a or linkage disequilibrium patterns between typed and true causa- specific TLR4 agonist. We found that LPS induced a significant tive variants (11, 14, 40). Also, we found in the current study that elevation of IRAK-M expression on WT BMDCs and BMDM genetic markers in IRAK-M were closely associated with asthma (Fig. 9A). susceptibility but not linked to asthma-related phenotypes. A A lack of IRAK-M did not make a difference in the CD124, similar phenomenon has been reported in the previous genetic by guest on September 28, 2021 CD206, and OX40L expression of BMDC or BMDM to LPS studies of the asthmatic populations (6). stimulation (data not shown). Instead, IRAK-M KO led to a sig- Many asthma-associated SNPs are located in noncoding inter- nificantly higher level of IFN-g, TNF-a, and IL-6 production by genic and intronic regulatory regions. These SNPs may modify BMDCs and BMDMs to LPS stimulation (Fig. 9B, 9C). We did mRNA expression and transcription factor binding (41, 42). In the not find a significant difference in the LPS-induced IL-13 pro- current study, we analyzed the top-associated SNP rs164395 and duction between WT and IRAK-M KO BMDCs or BMDMs (data identified the risk allele G that linked to significantly higher

FIGURE 8. The change induced by IRAK-M KO in the expression of costimulatory mole- cules on DCs and macrophages following a repeated OVA treatment. Representative images (left panel) and summary plots of flow cytom- etry analysis of costimulatory molecules PD-L1 (CD274) and PD-L2 (CD273) on lung DCs (A) and BAL macrophages (B)fromWTor IRAK-M KO mice after chronic OVA or PBS treatment (n = 10 animals in each group). *p , 0.05. MFI, mean fluorescence intensity. The Journal of Immunology 11 Downloaded from http://www.jimmunol.org/ by guest on September 28, 2021

FIGURE 9. The impact of IRAK-M KO on Th1 cytokine production by BMDCs and BMDMs. Expression of IRAK-M mRNA in WT BMDCs and BMDMs stimulated by PBS or LPS (n = 10 in each group). *p , 0.05 (A). ELISA analysis of cytokine production by IRAK-M KO or WT BMDCs (B) and BMDMs (C) following in vitro LPS stimulation (n = 6 independent experiments). *p , 0.05. mRNA expression of IRAK-M in PBMCs from asthmatics. Using IRAK-M is known to regulate the cytokine production of EMSA, we reveal the potential underlying mechanism is that G monocytes/macrophages via intervening the intracellular the TLR/ allele, as compared with A allele, has a higher binding capacity NF-kB signaling pathway (16). We have previously reported that to C-Jun, a transcription factor directly binding to the human IRAK-M deficiency exacerbated the OVA-induced acute airway IRAK-M promoter (22). inflammation through multiple pathways, such as promoting the 12 IRAK-M AND CHRONIC ASTHMA production of proinflammatory cytokines by airway epithelial an overexpression animal model is desired to precisely study the cells, enhancing the activities of DCs and macrophages, and de- complexity of asthma. viating the CD4+ T cells differentiation to Th2 and Th17 cells In conclusion, we demonstrated in the current study that IRAK- (24). But, in the current study, we demonstrated an inhibitory M was genetically linked in adult-onset human asthmatics, and it effect of IRAK-M deficiency on the OVA-induced chronic mouse has a proinflammatory effect in an animal model using mouse lungs airway inflammation. Mechanistically, we found that IRAK-M KO following chronic allergen stimulation. This action of IRAK-M is could reduce the Th2/Th1 imbalance in the chronic allergic in- opposite to its anti-inflammatory effect in the acute phase of flammation by increasing the Th1 cell percentage and Th1 cyto- asthma. The underlying mechanism of these findings is related to kines (IFN-g, IL-12p40, IL-23, and IL-27) production. The promoting the effect of IRAK-M on DC-mediated, Th2-deviated enhanced Th1 function can inhibit the Th2 cell proliferation and immune activation following a repetitive allergen stimulation. activity (1). Indeed, constant overexpression of airway epithelial These findings illustrated the complexity of immune regulation at IRAK-M could impair the innate immunity and compromise the different stages of asthmatic inflammation. Moreover, it implicates capability of clearance of inhaled environmental Ags from the that downregulating IRAK-M and the downstream signaling airways (22, 23, 43). Chronic stimulation of innate immunity is a pathway may provide an alternative therapy to control chronic major contributor to the persistent airway inflammation and irre- asthmatic airway inflammation. versible airway remodeling (1, 34). At the cellular level, our in vitro data demonstrated IRAK-M KO BMDCs and BMDMs Acknowledgments secreted more Th1 cytokines after TLR ligation by LPS. A similar We thank Prof. Nikolaos G. Frangogiannis for kindly gifting IRAK-M KO finding, that IRAK-M inhibited the Th1 cytokine production by mice. We thank all subjects for agreeing to participate in this study. We Downloaded from DCs, has been reported previously (44). Hence, the impact of thank Dr. Kewu Huang for the help in the FlexiVent measurement of IRAK-M KO on the chronic allergic airway inflammation could be airway reactivity. We thank Prof. Roy Pleasants for English editing. attributed to the alternation of DC/macrophage–medicated T cell differentiation. Disclosures The mechanism underlying the distinct regulation of IRAK-M The authors have no financial conflicts of interest. on the DC function at different inflammatory stages is not clear. http://www.jimmunol.org/ Our studies suggested it could act via modulating the costimulatory signaling molecules on the DCs differentially. There is mounting References evidence demonstrating that differentiation of Th cells relies on 1. Lambrecht, B. N., and H. Hammad. 2015. The immunology of asthma. Nat. Immunol. 16: 45–56. particular costimulatory molecules signaling from APCs (45). In 2. Ojiaku, C. A., G. Cao, W. Zhu, E. J. Yoo, M. Shumyatcher, B. E. Himes, the current study, we observed that IRAK-M KO led to a signifi- S. S. An, and R. A. Panettieri, Jr. 2018. TGF-b1 evokes human airway smooth muscle cell shortening and hyperresponsiveness via Smad3. Am J Respir Cell cantly higher expression of PD-L1 (CD274) and PD-L2 (CD273) Mol Biol. 58: 575–584. on the lung DCs or macrophages following chronic OVA stimu- 3. Siroux, V., X. Basagan˜a, A. Boudier, I. Pin, J. Garcia-Aymerich, A. Vesin, lation. The surface molecules PD-L1 (CD274) and PD-L2 R. Slama, D. Jarvis, J. M. Anto, F. Kauffmann, and J. Sunyer. 2011. Identifying adult asthma phenotypes using a clustering approach. Eur. Respir. J. 38: 310–317. by guest on September 28, 2021 (CD273) of DCs were known to present the inhibitory signal to 4. Siroux, V., J. R. Gonza´lez, E. Bouzigon, I. Curjuric, A. Boudier, M. Imboden, T cell activation (46). Particularly, PD-L2 (CD273) was shown to J. M. Anto, I. Gut, D. Jarvis, M. Lathrop, et al. 2014. Genetic heterogeneity of attenuate Th2-predominant allergic response through an IFN-g– asthma phenotypes identified by a clustering approach. Eur. Respir. J. 43: 439–452. 5. Zhang, Y., M. F. Moffatt, and W. O. Cookson. 2012. Genetic and genomic ap- dependent mechanism (47). Previous studies have demonstrated proaches to asthma: new insights for the origins. Curr. Opin. Pulm. Med. 18: 6–13. that CTLA-4 interacting with CD80 inhibited Th2 differentiation 6. Meyers, D. A., E. R. Bleecker, J. W. Holloway, and S. T. Holgate. 2014. Asthma genetics and personalised medicine. Lancet Respir. Med. 2: 405–415. (48, 49). In supporting with these findings, our data showed that 7. Wenzel, S. E. 2016. Emergence of biomolecular pathways to define novel asthma IRAK-M KO mice had a higher expression of CTLA-4 mRNA phenotypes. Type-2 immunity and beyond. Am. J. Respir. Cell Mol. Biol. 55: 1–4. by lung and CD80 by airway macrophages after chronic OVA 8. Stein,M.M.,C.L.Hrusch,J.Gozdz,C.Igartua,V.Pivniouk,S.E.Murray,J.G.Ledford, M. Marques Dos Santos, R. L. Anderson, N. Metwali, et al. 2016. Innate immunity and challenge. asthma risk in amish and Hutterite farm children. N.Engl.J.Med.375: 411–421. In addition, IRAK-M also regulates the tissue remodeling 9. Yokouchi, Y., Y. Nukaga, M. Shibasaki, E. Noguchi, K. Kimura, S. Ito, process accompanying chronic inflammation. In an animal model M. Nishihara, K. Yamakawa-Kobayashi, K. Takeda, N. Imoto, et al. 2000. Significant evidence for linkage of mite-sensitive childhood asthma to chro- of bleomycin-induced lung injury, IRAK-M was shown to promote mosome 5q31-q33 near the interleukin 12 B locus by a genome-wide search in inflammatory responses and collagen deposit in the lungs at the late Japanese families. Genomics 66: 152–160. 10. Raby, B. A., E. K. Silverman, R. Lazarus, C. Lange, D. J. Kwiatkowski, and stage (20). In another study of cardiac remodeling following S. T. Weiss. 2003. Chromosome 12q harbors multiple genetic loci related to myocardial infarction, IRAK-M was demonstrated to prompt the asthma and asthma-related phenotypes. Hum. Mol. Genet. 12: 1973–1979. myofibroblast conversion via inducing the TGF-b1–mediated 11. Balaci, L., M. C. Spada, N. Olla, G. Sole, L. Loddo, F. Anedda, S. Naitza, M. A. Zuncheddu, A. Maschio, D. Altea, et al. 2007. IRAK-M is involved in the a-SMA expression in the fibroblasts (50). Similarly, in the current pathogenesis of early-onset persistent asthma. Am.J.Hum.Genet.80: 1103–1114. study, we found that IRAK-M loss attenuated the airway remod- 12. Nakashima, K., T. Hirota, K. Obara, M. Shimizu, A. Jodo, M. Kameda, S. Doi, eling and reduced the a-SMA and TGF-b1 expression in the lung K. Fujita, T. Shirakawa, T. Enomoto, et al. 2006. An association study of asthma and related phenotypes with polymorphisms in negative regulator molecules of tissue with chronic allergic airway inflammation. the TLR signaling pathway. J. Hum. Genet. 51: 284–291. Our study has a few limitations. For instance, this study only 13. Beygo, J., Q. Parwez, E. Petrasch-Parwez, J. T. Epplen, and S. Hoffjan. 2009. No evidence of an association between polymorphisms in the IRAK-M gene and included adult patients and lacked child asthmatics as replicating atopic dermatitis in a German cohort. Mol. Cell. Probes 23: 16–19. control. Therefore, we cannot conclude if the polymorphisms of 14. Pino-Yanes, M., I. Sanchez-Machin, J. Cumplido, J. Figueroa, M. J. Torres- IRAK-M identified in this study are an age-specific risk of asthma Galvan, R. Gonzalez, A. Corrales, O. Acosta-Fernandez, J. C. Garcia-Robaina, T. Carrillo, et al. 2012. IL-1 receptor-associated kinase 3 gene (IRAK3) variants susceptibility or asthma-related traits. However, this current associate with asthma in a replication study in the Spanish population. J. Allergy study demonstrated the functionalities of the associated IRAK-M Clin. Immunol. 129: 573–575, 575.e1–10. SNPs, suggesting the importance of IRAK-M in asthma patho- 15. Pino-Yanes, M., S. F. Ma, X. Sun, P. Tejera, A. Corrales, J. Blanco, L. Pe´rez- Me´ndez, E. Espinosa, A. Muriel, L. Blanch, et al. 2011. Interleukin-1 receptor- genesis. In addition, the mice used in this study underwent whole- associated kinase 3 gene associates with susceptibility to acute lung injury. Am. body IRAK-M KO, which might introduce some unrecognized J. Respir. Cell Mol. Biol. 45: 740–745. 16. Kobayashi, K., L. D. Hernandez, J. E. Gala´n, C. A. Janeway, Jr., R. Medzhitov, confounding variables to impact the experimental results. An and R. A. Flavell. 2002. IRAK-M is a negative regulator of Toll-like receptor allelic-specific and temporally controllable KO model as well as signaling. Cell 110: 191–202. The Journal of Immunology 13

17. Baines, K. J., J. J. Fu, V. M. McDonald, and P. G. Gibson. 2017. Airway gene 35. Redington, A. E., J. Madden, A. J. Frew, R. Djukanovic, W. R. Roche, expression of IL-1 pathway mediators predicts exacerbation risk in obstructive S. T. Holgate, and P. H. Howarth. 1997. Transforming growth factor-beta 1 in airway disease. Int. J. Chron. Obstruct. Pulmon. Dis. 12: 541–550. asthma. Measurement in bronchoalveolar lavage fluid. Am. J. Respir. Crit. Care 18. Jenkins, B. J. 2016. Multifaceted role of IRAK-M in the promotion of colon Med. 156: 642–647. carcinogenesis via barrier dysfunction and STAT3 oncoprotein stabilization in 36. Vignola, A. M., P. Chanez, G. Chiappara, A. Merendino, E. Pace, A. Rizzo, tumors. Cancer Cell 29: 615–617. A. M. la Rocca, V. Bellia, G. Bonsignore, and J. Bousquet. 1997. Transforming 19. Gong, H., T. Liu, W. Chen, W. Zhou, and J. Gao. 2017. Effect of IRAK-M on growth factor-beta expression in mucosal biopsies in asthma and chronic bron- airway inflammation induced by cigarette smoking. Mediators Inflamm. DOI: chitis. Am. J. Respir. Crit. Care Med. 156: 591–599. 10.1155/2017/6506953. 37. Ojiaku, C. A., E. J. Yoo, and R. A. Panettieri, Jr. 2017. Transforming growth 20. Ballinger, M. N., M. W. Newstead, X. Zeng, U. Bhan, X. M. Mo, S. L. Kunkel, factor b1 function in airway remodeling and hyperresponsiveness. The missing B. B. Moore, R. Flavell, J. W. Christman, and T. J. Standiford. 2015. IRAK-M link? Am. J. Respir. Cell Mol. Biol. 56: 432–442. promotes alternative macrophage activation and fibroproliferation in bleomycin- 38. Hekking, P. P., M. J. Loza, S. Pavlidis, B. de Meulder, D. Lefaudeux, induced lung injury. J. Immunol. 194: 1894–1904. F. Baribaud, C. Auffray, A. H. Wagener, P. I. Brinkman, R. I. Lutter, et al. 2018. 21. Steiger, S., S. V. Kumar, M. Honarpisheh, G. Lorenz, R. Gu¨nthner, S. Romoli, Pathway discovery using transcriptomic profiles in adult-onset severe asthma. R. Gro¨bmayr, H. E. Susanti, J. Potempa, J. Koziel, and M. Lech. 2017. Immu- J Allergy Clin Immunol. 141: 1280–1290. nomodulatory molecule IRAK-M balances macrophage polarization and deter- 39. Leung, T. F., F. W. Ko, H. Y. Sy, S. K. Tsui, and G. W. Wong. 2014. Differences mines macrophage responses during renal fibrosis. J. Immunol. 199: 1440–1452. in asthma genetics between Chinese and other populations. J. Allergy Clin. 22. Wu, Q., D. Jiang, S. Smith, J. Thaikoottathil, R. J. Martin, R. P. Bowler, and Immunol. 133: 42–48. H. W. Chu. 2012. IL-13 dampens human airway epithelial innate immunity 40. Gabriel, S. B., S. F. Schaffner, H. Nguyen, J. M. Moore, J. Roy, B. Blumenstiel, through induction of IL-1 receptor-associated kinase M. J. Allergy Clin. J. Higgins, M. DeFelice, A. Lochner, M. Faggart, et al. 2002. The structure of Immunol. 129: 825–833.e2. haplotype blocks in the . Science 296: 2225–2229. 23. Wu, Q., L. F. van Dyk, D. Jiang, A. Dakhama, L. Li, S. R. White, A. Gross, and 41. Hindorff, L. A., P. Sethupathy, H. A. Junkins, E. M. Ramos, J. P. Mehta, H. W. Chu. 2013. Interleukin-1 receptor-associated kinase M (IRAK-M) pro- F. S. Collins, and T. A. Manolio. 2009. Potential etiologic and functional im- motes human rhinovirus infection in lung epithelial cells via the autophagic plications of genome-wide association loci for human diseases and traits. Proc. pathway. Virology 446: 199–206. Natl. Acad. Sci. USA 106: 9362–9367. 24. Zhang, M., W. Chen, W. Zhou, Y. Bai, and J. Gao. 2017. Critical role of IRAK-M 42. Maurano, M. T., R. Humbert, E. Rynes, R. E. Thurman, E. Haugen, H. Wang, Downloaded from in regulating antigen-induced airway inflammation. Am. J. Respir. Cell Mol. A. P. Reynolds, R. Sandstrom, H. Qu, J. Brody, et al. 2012. Systematic locali- Biol. 57: 547–559. zation of common disease-associated variation in regulatory DNA. Science 337: 25. Hubbard, L. L., and B. B. Moore. 2010. IRAK-M regulation and function in host 1190–1195. defense and immune homeostasis. Infect. Dis. Rep. 2: e9 43. Wu, Q., R. J. Martin, S. Lafasto, B. J. Efaw, J. G. Rino, R. J. Harbeck, and 26. Amelink, M., J. C. de Groot, S. B. de Nijs, R. Lutter, A. H. Zwinderman, H. W. Chu. 2008. Toll-like receptor 2 down-regulation in established mouse P. J. Sterk, A. ten Brinke, and E. H. Bel. 2013. Severe adult-onset asthma: a allergic lungs contributes to decreased mycoplasma clearance. Am. J. Respir. distinct phenotype. J. Allergy Clin. Immunol. 132: 336–341. Crit. Care Med. 177: 720–729.

27. Lee, S. H., M. Eren, D. E. Vaughan, R. P. Schleimer, and S. H. Cho. 2012. A 44. Turnis, M. E., X. T. Song, A. Bear, A. E. Foster, S. Gottschalk, M. K. Brenner, http://www.jimmunol.org/ plasminogen activator inhibitor-1 inhibitor reduces airway remodeling in a S. Y. Chen, and C. M. Rooney. 2010. IRAK-M removal counteracts dendritic cell murine model of chronic asthma. Am. J. Respir. Cell Mol. Biol. 46: 842–846. vaccine deficits in migration and longevity. J. Immunol. 185: 4223–4232. 28. Lin, Y., H. Yan, Y. Xiao, H. Piao, R. Xiang, L. Jiang, H. Chen, K. Huang, Z. Guo, 45. Singh, A. K., P. Stock, and O. Akbari. 2011. Role of PD-L1 and PD-L2 in al- W. Zhou, et al. 2011. Attenuation of antigen-induced airway hyperresponsiveness lergic diseases and asthma. Allergy 66: 155–162. and inflammation in CXCR3 knockout mice. Respir. Res. 12: 123. 46. Freeman, G. J., A. J. Long, Y. Iwai, K. Bourque, T. Chernova, H. Nishimura, 29. Willis, C. R., L. Siegel, A. Leith, D. Mohn, S. Escobar, S. Wannberg, K. Misura, L. J. Fitz, N. Malenkovich, T. Okazaki, M. C. Byrne, et al. 2000. Engagement of E. Rickel, J. B. Rottman, M. R. Comeau, et al. 2015. IL-17RA signaling in the PD-1 immunoinhibitory receptor by a novel B7 family member leads to airway inflammation and bronchial hyperreactivity in allergic asthma. Am. J. negative regulation of lymphocyte activation. J. Exp. Med. 192: 1027–1034. Respir. Cell Mol. Biol. 53: 810–821. 47. Matsumoto, K., H. Inoue, T. Nakano, M. Tsuda, Y. Yoshiura, S. Fukuyama, 30. Wittke, A., V. Weaver, B. D. Mahon, A. August, and M. T. Cantorna. 2004. F. Tsushima, T. Hoshino, H. Aizawa, H. Akiba, et al. 2004. B7-DC regulates Vitamin D receptor-deficient mice fail to develop experimental allergic asthma. asthmatic response by an IFN-gamma-dependent mechanism. J. Immunol. 172:

J. Immunol. 173: 3432–3436. 2530–2541. by guest on September 28, 2021 31. Liu, Z. J., Y. J. Yang, L. Jiang, Y. C. Xu, A. X. Wang, G. H. Du, and J. M. Gao. 48. Oosterwegel, M. A., D. A. Mandelbrot, S. D. Boyd, R. B. Lorsbach, D. Y. Jarrett, 2011. Tyrosine sulfation in N-terminal domain of human C5a receptor is nec- A. K. Abbas, and A. H. Sharpe. 1999. The role of CTLA-4 in regulating Th2 essary for binding of chemotaxis inhibitory protein of Staphylococcus aureus. differentiation. J. Immunol. 163: 2634–2639. Acta Pharmacol. Sin. 32: 1038–1044. 49. Bour-Jordan, H., J. L. Grogan, Q. Tang, J. A. Auger, R. M. Locksley, and 32. Barrett, J. C., B. Fry, J. Maller, and M. J. Daly. 2005. Haploview: analysis and J. A. Bluestone. 2003. CTLA-4 regulates the requirement for cytokine-induced visualization of LD and haplotype maps. Bioinformatics 21: 263–265. signals in T(H)2 lineage commitment. Nat. Immunol. 4: 182–188. 33. Davies, D. E., J. Wicks, R. M. Powell, S. M. Puddicombe, and S. T. Holgate. 50. Saxena, A., A. V. Shinde, Z. Haque, Y. J. Wu, W. Chen, Y. Su, and 2003. Airway remodeling in asthma: new insights. J. Allergy Clin. Immunol. N. G. Frangogiannis. 2015. The role of Associated Kinase 111: 215–225, quiz 226. (IRAK)-M in regulation of myofibroblast phenotype in vitro, and in an experi- 34. Lambrecht, B. N., and H. Hammad. 2014. Dendritic cell and epithelial cell interac- mental model of non-reperfused myocardial infarction. J. Mol. Cell. Cardiol. 89 tions at the origin of murine asthma. Ann. Am. Thorac. Soc. 11(Suppl. 5): S236–S243. (Pt B): 223–231. Supplemental Data

IRAK-M associates with susceptibility to adult-onset asthma and promotes

chronic airway inflammation

Yi Liu, #†*, Mingqiang Zhang, #*, Lili Lou,# Lun Li, #, Youming Zhang, ‡ Wei Chen, §

Weixun Zhou, §§ Yan Bai, ¶¶ Jinming Gao, M.D., Ph.D.#

#Departments of Respiratory Diseases, §Cardiology, and §§Pathology, Peking Union

Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union

Medical College, Beijing 100730, China

†Department of Respiratory Medicine, Civil Aviation General Hospital, Beijing,

100123, China

‡ Genomics Medicine Section, National Heart and Lung Institute, Imperial College

London, London, SW3 6LY, UK

¶¶Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine,

Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts

*These authors equally contributed to this work.

Correspondence to: Jinming Gao, M.D.,

Professor of Pulmonary Medicine,

Department of Respiratory Diseases, 1 Peking Union Medical College Hospital,

#1 Shuaifuyuan, Dongcheng District,

Beijing 100730, China.

E-mail: [email protected]

OR [email protected]

Tel: 861069155049

Fax: 861065124875

2

Supplemental Table 1 Primers for amplification of human IRAK-M gene Rs Number Chr. Location Gene Position (effect) SNPs Amplycation Primers (5' to 3') rs7970350 65966384 HMGA2 downstream 500B C/T F: GCACAATAAACATAACAGCCTC R: CAAAATCTGAAAGGCGAGCT rs6581660 66036672 T/G F: TTCCAGGCAAGTGGTGGAT

R: AACCTGCATTTCAGCAAGATC rs11836463 66056192 C/A F: AGATCACTCAGCTCCCAACAT

R: CTCTTAGAAAGGCAGGCAGAT rs2870784 66088660 5’ region G/T F: CCCCAGCTTCAACAACAACT R: CAGGTGCAAAAGGGTTCTTGT rs2141709 66145121 Intron X G/A F: AAAACTCTGCTCTTGCTAAAC R: TCCCTAAATCAAACTCCATTCT rs2701652 66187097 G/C F: CAGTTGATAGCAACCATGATG

R: ACCTGCTTTACCAGATCTTGT rs1821777 66212448 Intron X C/T F: AGACAGTTGATTGACTGCAGC R: TCATCTCTACATGCTCAGCAC rs1624395 66224436 Intron 6 A/G F: GAT(G/A)CAGCTTTCTCCTCTTAG R: ATCCTTCAGGTCTCAGCTTAG rs1370128 66224858 Intron 6 T/C F: GCATCATTTTGTCTGAACAAG R: AAGTTTGCTGGTGTTGTACCT

3

Supplemental Table 2 Primers for extension of human IRAK-M gene

Markers SNPs Extending Primers (5' to 3')

rs7970350 C/T R: ttttttttttttttttttttttttttttTCAGGGCAGTTGATTGAAG rs6581660 T/G R: tttttttttttttttttttttttttAAGTCCTGTTCCACACTGC rs11836463 C/A R: ttttttttttttttttttttttttttttttttttttCACTGTGTTAGCAAACAGTTTC rs2870784 G/T F: ttttttttttttttttttCAGGATGATAAATGGATTCGTTA rs2141709 G/A F: tttttttttttttttAGCCAACACTGACAATTGC rs2701652 G/C F: tttttttttttttttttttttttttttttttttGACAGTTTTAGGAATAGGAAGG rs1821777 C/T F: ttttttttttAGAGATAGGTCCACACACAAA rs1624395 A/G F: tttttttAAATACAGAGCACAGAGGAGG rs1370128 T/C F: tttttttttttttttttCATTTTGTCTGAACAAGGCAC

4 Supplemental figure S1

Supplemental Figure S1 Pairwise linkage disequilibrium (LD) pattern of IRAK-M in Chinese patients with asthma.

The location of each SNP tested on the chromosome is shown on top panel. The number in each diamond refers to the magnitude of LD measured by r2 (100).

Haplotype block structure was estimated with Haploview, version 4.0.

5 Supplemental figure S2

Supplemental Figure S2 IRAK-M KO changed expression of co-stimulatory molecules on cell surface after chronic treatment of OVA. FACS analysis of cellular surface expression of co-stimulatory molecules MHC II, CD80, CD86, and OX40L (A) lung dendritic cells and (B) BAL macrophages using specific Abs against the indicated costimulatory molecule, mean fluorescence intensity of indicated costimulatory molecules was plotted, n=10 animals in each group, *, p < 0.05.

6