+ All Categories
Home > Documents > PILRα negatively regulates mouse inflammatory arthritis

PILRα negatively regulates mouse inflammatory arthritis

Date post: 09-Dec-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
12
of August 8, 2016. This information is current as Inflammatory Arthritis Negatively Regulates Mouse α PILR Zarrin Wenjun Ouyang, Flavius Martin, Wyne P. Lee and Ali A. Balazs, Tao Sai, Benjamin Haley, Philip E. Hass, Min Xu, Mariathasan, Jason DeVoss, Shahram Misaghi, Mercedesz Sophie Lehar, Vida Asghari, WeiYu Lin, Sanjeev Rio, Lu, Zhonghua Lin, Kai Barck, Judy Young, Mariela Del Lesch, Ivan Peng, Andrew Sebrell, Wilman Luk, Yanmei Sarah Kummerfeld, Gabriel Quinones, Kate Senger, Justin Yonglian Sun, Patrick Caplazi, Juan Zhang, Anita Mazloom, http://www.jimmunol.org/content/193/2/860 doi: 10.4049/jimmunol.1400045 2014; 2014; 193:860-870; Prepublished online 16 June J Immunol Material Supplementary 5.DCSupplemental.html http://www.jimmunol.org/content/suppl/2014/06/16/jimmunol.140004 References http://www.jimmunol.org/content/193/2/860.full#ref-list-1 , 19 of which you can access for free at: cites 43 articles This article Subscriptions http://jimmunol.org/subscriptions is online at: The Journal of Immunology Information about subscribing to Permissions http://www.aai.org/ji/copyright.html Submit copyright permission requests at: Email Alerts http://jimmunol.org/cgi/alerts/etoc Receive free email-alerts when new articles cite this article. Sign up at: Print ISSN: 0022-1767 Online ISSN: 1550-6606. Immunologists, Inc. All rights reserved. Copyright © 2014 by The American Association of 9650 Rockville Pike, Bethesda, MD 20814-3994. The American Association of Immunologists, Inc., is published twice each month by The Journal of Immunology by guest on August 8, 2016 http://www.jimmunol.org/ Downloaded from by guest on August 8, 2016 http://www.jimmunol.org/ Downloaded from
Transcript

of August 8, 2016.This information is current as

Inflammatory Arthritis Negatively Regulates MouseαPILR

ZarrinWenjun Ouyang, Flavius Martin, Wyne P. Lee and Ali A.Balazs, Tao Sai, Benjamin Haley, Philip E. Hass, Min Xu, Mariathasan, Jason DeVoss, Shahram Misaghi, MercedeszSophie Lehar, Vida Asghari, WeiYu Lin, Sanjeev

Rio,Lu, Zhonghua Lin, Kai Barck, Judy Young, Mariela Del Lesch, Ivan Peng, Andrew Sebrell, Wilman Luk, YanmeiSarah Kummerfeld, Gabriel Quinones, Kate Senger, Justin Yonglian Sun, Patrick Caplazi, Juan Zhang, Anita Mazloom,

http://www.jimmunol.org/content/193/2/860doi: 10.4049/jimmunol.14000452014;

2014; 193:860-870; Prepublished online 16 JuneJ Immunol 

MaterialSupplementary

5.DCSupplemental.htmlhttp://www.jimmunol.org/content/suppl/2014/06/16/jimmunol.140004

Referenceshttp://www.jimmunol.org/content/193/2/860.full#ref-list-1

, 19 of which you can access for free at: cites 43 articlesThis article

Subscriptionshttp://jimmunol.org/subscriptions

is online at: The Journal of ImmunologyInformation about subscribing to

Permissionshttp://www.aai.org/ji/copyright.htmlSubmit copyright permission requests at:

Email Alertshttp://jimmunol.org/cgi/alerts/etocReceive free email-alerts when new articles cite this article. Sign up at:

Print ISSN: 0022-1767 Online ISSN: 1550-6606. Immunologists, Inc. All rights reserved.Copyright © 2014 by The American Association of9650 Rockville Pike, Bethesda, MD 20814-3994.The American Association of Immunologists, Inc.,

is published twice each month byThe Journal of Immunology

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

The Journal of Immunology

PILRa Negatively Regulates Mouse Inflammatory Arthritis

Yonglian Sun,* Patrick Caplazi,† Juan Zhang,* Anita Mazloom,‡ Sarah Kummerfeld,x

Gabriel Quinones,‡ Kate Senger,* Justin Lesch,* Ivan Peng,* Andrew Sebrell,{

Wilman Luk,‖ Yanmei Lu,‖ Zhonghua Lin,* Kai Barck,# Judy Young,‖ Mariela Del Rio,**

Sophie Lehar,†† Vida Asghari,** WeiYu Lin,* Sanjeev Mariathasan,†† Jason DeVoss,*

Shahram Misaghi,‡‡ Mercedesz Balazs,* Tao Sai,{ Benjamin Haley,‡ Philip E. Hass,‡

Min Xu,* Wenjun Ouyang,* Flavius Martin,* Wyne P. Lee,* and Ali A. Zarrin*

Paired Ig-like type 2 receptor (PILR)a inhibitory receptor and its counterpart PILRb activating receptor are coexpressed on

myeloid cells. In this article, we report that PILRa, but not PILRb, is elevated in human rheumatoid arthritis synovial tissue and

correlates with inflammatory cell infiltration. Pilra2/2 mice produce more pathogenic cytokines during inflammation and are

prone to enhanced autoimmune arthritis. Correspondingly, engaging PILRa with anti-PILRa mAb ameliorates inflammation in

mouse arthritis models and suppresses the production of proinflammatory cytokines. Our studies suggest that PILRa mediates an

important inhibitory pathway that can dampen inflammatory responses. The Journal of Immunology, 2014, 193: 860–870.

Immune responses are modulated by a network of positive-and negative-regulatory mechanisms. Paired receptors consistof highly related activating and inhibitory receptors that

are widely involved in the regulation of the immune system (1).Both inhibitory and activating receptors share high similarityin their extracellular domain, whereas their intracellular signal-ing domains are divergent (2, 3). Paired Ig-like type 2 receptor(PILR)a belongs to the Ig superfamily. Its intracellular domaincontains two ITIMs that recruit SHP-1 and SHP-2 to trigger aninhibitory signaling cascade, resulting in reduced intracellularcalcium mobilization (4, 5). PILRa and its gene-linked activatingcounterpart, PILRb, share highly similar extracellular domains,suggesting that they may recognize the same ligands (5–7). PILRbhas a truncated cytoplasmic domain and a charged amino acidresidue in its transmembrane region that associates with the

ITAM-bearing DAP12 adaptor molecule to deliver activating si-gnals (8). Human and mouse PILRa share only ∼40% homology(7), yet conserved residues mediate ligand interactions (9).PILRa and PILRb transcripts show similar tissue expression,

with high levels in spleen, liver, and lung and lower levels inthe small intestine (8). PILRa and PILRb are predominantlyexpressed in cells of the myelomonocytic lineage, includingmonocytes, macrophages, granulocytes, and monocyte-deriveddendritic cells (DCs) (4, 8). Additionally, PILRb is expressed inNK cells (8). It was shown that PILRa binds to mouse CD99 (8),PILR-associating neural protein (10), and HSV-1 glycoprotein B(11). Specific sialylated O-linked glycans on ligands are requiredfor their binding to PILRa (6, 10, 12). We recently identified twomore binding partners of PILRa, neural proliferation differentia-tion and control-1 and collectin-12, and found that an evolution-arily conserved PILRa domain mediates its interaction with thesediverse sialylated ligands (9). This suggests that a complex net-work of ligands might modulate cellular functions via PILRa. Itwas shown that PILRa binds to HSV-1 glycoprotein B and servesas a virus entry coreceptor (11, 13). Previous studies showed thatPILRb-deficient and agonist anti-PILRa–treated mice show im-proved clearance of infectious reagents and improved survival (14,15). A recent study showed that PILRa negatively regulatesneutrophil recruitment during TLR-mediated inflammatory responses(16). However, the role of PILRa or PILRb in autoimmunity islargely unknown.Myeloid cells play a critical role in the pathophysiology of

inflammation and autoimmunity. Rheumatoid arthritis (RA) isa systemic, inflammatory, autoimmune disorder manifested bychronic polyarthritis with synovial hyperplasia and joint destruc-tion, resulting in pain, loss of joint function, and concomitantreduction of life quality (17, 18). Myeloid cells, includingmonocytes/macrophages and neutrophils, play an important rolein various stages of arthritis development (19, 20). To investigatethe role of PILRa in myeloid cell–mediated immune responsesand its effects on autoimmune diseases, we studied its functionin myeloid-driven models of RA, including collagen Ab–inducedarthritis (CAIA) and K/BxN serum–transfer arthritis, using PILRa-deficient mice and PILRa-specific mAbs. Murine CAIA is in-

*Department of Immunology, Genentech, South San Francisco, CA 94080;†Department of Pathology, Genentech, South San Francisco, CA 94080;‡Department of Protein Chemistry, Genentech, South San Francisco, CA 94080;xDepartment of Bioinformatics and Computational Biology, Genentech, South SanFrancisco, CA 94080; {Department of Antibody Engineering, Genentech, South SanFrancisco, CA 94080; ‖Department of Biochemical and Cellular Pharmacology, Gen-entech, South San Francisco, CA 94080; #Department of Biomedical Imaging, Gen-entech, South San Francisco, CA 94080; **Department of Transgenic Technology,Genentech, South San Francisco, CA 94080; ††Department of Microbial Pathogen-esis, Genentech, South San Francisco, CA 94080; and ‡‡Department of Early StageCell Culture, Genentech, South San Francisco, CA 94080

Received for publication January 9, 2014. Accepted for publication May 19, 2014.

The microarray data presented in this article have been submitted to the NationalCenter for Biotechnology Information’s Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE48780) under accession number GSE48780.

Address correspondence and reprint requests to Dr. Ali A. Zarrin, Genentech, 1 DNAWay, South San Francisco, CA 94080. E-mail address: [email protected]

The online version of this article contains supplemental material.

Abbreviations used in this article: BM, bone marrow; BMDC, BM-derived DC;BMDM, BM-derived monocyte; CAIA, collagen Ab–induced arthritis; CLEC4G,C-type lectin domain family 4, member G; DC, dendritic cell; ES, embryonic stem;h, human; JCBV, joint cortical bone volume; m, mouse; micro-CT, microcomputedtomography; MPO, myeloperoxidase; OA, osteoarthritis; PILR, paired Ig-like type 2receptor; PNBV, periosteal new bone volume; RA, rheumatoid arthritis; WT, wild-type.

Copyright� 2014 by TheAmericanAssociation of Immunologists, Inc. 0022-1767/14/$16.00

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1400045

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

duced by i.v. injection of mAbs against type II collagen, fol-lowed by i.p. injection of LPS. This model is widely used tostudy the pathogenesis of autoimmune arthritis and to determineefficacy of therapeutics (21–24). Myeloid cells, FcgRs (22), andproinflammatory cytokines, especially TNF-a and IL-1b (21), areindispensable for the development and maintenance of arthritis inthis model (23). The K/BxN serum–transfer arthritis model isinduced by transferring K/BxN serum into normal mice; LPS isnot needed in this model. The K/BxN serum–transfer arthritismodel shares multiple features with human RA, including sym-metrical involvement of peripheral joints, pannus formation, sy-novial hyperplasia, and bone and cartilage degradation (25). Weshow that Pilra2/2 mice developed more severe arthritis than didwild-type (WT) mice in these two models, corroborating the in-hibitory role of PILRa during inflammation. Accordingly, wefound that administration of an anti-PILRa mAb attenuatedmouse inflammatory arthritis and reduced the production of sev-eral proinflammatory cytokines, indicating that PILRa is part ofa critical inflammatory axis regulating myeloid cell function.

Materials and MethodsMice

BALB/c mice were purchased from Charles River Laboratory. Pilra-defi-cient mice were generated by genOway and were maintained in-house.Mice were used at 6–10 wk of age. All animal experiments were per-formed in accordance with institutional animal care and use committee–approved protocols.

Generation of Pilra2/2 mice

The genomic region containing the murine PILRa locus was isolated byPCR from 129Sv BAC genomic DNA (genOway, Lyon, France). The6050-bp 59 homology arm corresponds to nt 137843309–137837259cluster NC_000071.6 contig GRCm38.p2 in chromosome 5 of C57BL/6J.The 1872-bp 39 homology arm corresponds to nt 137830963–137829091of the same region. This strategy allows for deletion of a 6.3-kb regioncomprising exons 1 and 2 of PILRa (containing ATG, Ig V-set domain,and extracellular domain). Neomycin-resistant clones were first screenedby PCR using primers 59-ATGCTCCAGACTGCCTTGGGAAAAG-39(neocassette) and antisense 59-CACTTCACAGTCTAGTCACTTCCGC-ACC-39 (outside 39 homology arm). Final clones were confirmed usingprimers 59-GCACCATGTCCTGAGGAAGAGTAGTTTGA-39 (within 59homology arm) and 59-CACTTCACAGTCTAGTCACTTCCGCACC-39(within 39 homology arm), which generates a 7.0-kb fragment in WTembryonic stem (ES) cells or a 3.8-kb fragment in neo-containing targetedES cells. The expected neo-excised fragment of 2.1 kb was cloned andsequenced to confirm the desired recombination event for all ES clones.Gene targeting was further confirmed by Southern blot using a neomycininternal probe (SacI digest generating an 8.6-kb fragment) and a 39 ex-ternal probe (SphI digest changing 11.0 kb to 6.0 kb). The 39 probe con-tains a 450-bp fragment between the 39 end of the targeting arm and theSphI site. A total of 6 of 646 clones carried correct integration. Two in-dependent clones were microinjected into C57BL/6J-Tyrc-2J/J blastocyststo generate chimeras, which were bred to C57BL/6N mice (Charles River)to generate PILRa-knockout mice. Subsequently, the neomycin cassettewas removed by breeding PILRa-knockout mice carrying the neomycincassette with C57BL/6N Rosa-26-cre knockin mice (Taconic). The fol-lowing primers were used to confirm the cre recombination and genotype:forward primer-1: 59-TGCAAACCACAGTTGGGTTGGCAACGGCTG-39(located in exon 2, which is deleted in the knockout) and reverse primer:59-GGAGGAGGCCAGAAGCCAACCTTTGCTATC-39 (within 39 target-ing arm, which generates a 470-bp fragment in WT mice) or forwardprimer-2: 59-ATGACAGTTTGTGTCTGGATCAGAAC-39 (upstream ofthe loxp site) and the same reverse primer, which generates a 600-bpproduct from the knockout allele.

Generation of mAbs

Anti-PILRa mAbs were generated from Armenian hamsters and Pilra2/2

mice using immunogenic fusion proteins consisting of the extracellulardomain of the mouse Pilra gene (GenBank accession number: NM_153510),including amino acid positions Met 1 to Val 197, as previously described(9). Mouse anti-PILRa mAbs are IgG1 isotype. The specificity of anti-PILRa

mAbs was confirmed by ELISA and flow cytometry analysis. These mAbs donot cross-react with mouse PILRb. Hamster anti-PILRa mAb was used forstaining, and mouse anti-PILRa mAb was used for in vivo and in vitrofunctional studies.

Gene expression analysis

For gene expression analysis, total RNAwas isolated using TRIzol reagent(Invitrogen). Total RNA (50 ng) was subjected to RT-PCR using Access RT-PCR System (Promega). Primers used for amplification were as follows:b-actin, sense primer: 59-TACCTCATGAAGATCCTCA-39 and antisenseprimer: 59-TTCGTGGATGCCACAGGAC-39; PILRa, sense primer: 59-GACTCCTGAGCACTCCAGTGTCCCTAGACAG-39 and antisense primer59-AAGGTGACTCTAAGAGTGTCATCCAGACCA-39; and PILRb, senseprimer: 59-GGAAATTCAGAAAGATACAACA-39 and antisense primer: 59-CAGGTTCATCAGTGAAGGAT-39. For quantitative RT-PCR, total RNA(50 ng) was subjected to RT and real-time quantitative PCR assay usingAccess RT-PCR System (Promega), Perkin Elmer SYBR Green, and an ABI7500 instrument. Gene-specific unlabeled primers used were as follows:b-actin, sense primer: 59-TACCTCATGAAGATCCTCA-39 and antisenseprimer: 59-TTCGTGGATGCCACAGGAC-39) PILRb, sense primer: 59-CACCATGAGGAGCCCCTTCATCGTC-39 and antisense primer: 59-TAGGACTATGACCAAAACTTTAGCCAGGAG-39.

Cell extraction and Western blot analysis

Bone marrow (BM) cells and splenocytes were harvested, and RBCs werelysed with ACK lysing buffer. Cells were washed twice with ice-cold PBS(pH 7.4) and then treated in Cell Extraction Buffer (Invitrogen) containing1 mM PMSF and protease inhibitor mixture (Sigma). Lysates were incu-bated at 4˚C for 30 min, with vortexing at 10-min intervals, and the in-soluble material was removed by centrifugation at 13,000 rpm for 10 minat 4˚C. Soluble extracts with Novex Tris-Glycine SDS sample buffer wereseparated by SDS-PAGE using 4–12% Bis-Tris gel and transferred toa PVDF/Filter Paper Sandwich (all from Invitrogen). Blots were blockedwith 5% nonfat dried milk, 0.1% Tween-20 in TBS buffer and incubatedwith hamster anti-mouse PILRa mAb, followed by HRP-conjugated anti-hamster IgG (Jackson ImmunoResearch) for 1 h. Proteins were detected bychemiluminescence with an Amersham ECL Plus Western blotting system(GE Healthcare).

Immunohistochemistry

Immunohistochemistry for PILRa was performed on formalin-fixed,paraffin-embedded tissue using in-house–generated Armenian hamsteranti-mPILRa mAb and an automated system (VENTANA DISCOVERYXT, Ventana Medical Systems, Tucson, AZ). Briefly, 4-mm sections weredeparaffinized, rehydrated, and subjected to Ag retrieval (Ventana Protease2). Primary Ab (5 mg/ml) was incubated at 37˚C for 60 min. Binding wasvisualized using secondary anti-hamster IgG, Ventana Rabbit OmniMap,and Ventana DAB reagents (Ventana Medical Systems). Immunohisto-chemistry for myeloperoxidase (MPO) and F4/80 was performed onformalin-fixed, acid-decalcified, paraffin-embedded sections of murinearthritic paws to visualize infiltration by neutrophils or macrophages,respectively. For MPO, a rabbit polyclonal Ab A95-1 (LabVision/Neomarker, Fremont, CA) was used on an automated platform (Ven-tana) using Ventana Rabbit OmniMap and DAB for visualization. For F4/80, a commercial rat Ab (Cl:A3-1; AbD Serotec, Raleigh, NC) was usedon an automated platform (Leica Bond) and visualized with DAB using anABC Peroxidase protocol.

Flow cytometry

Single-cell suspensions were prepared from BM and peritoneal lavage fluid.After RBC lysis, cells were incubated with rat anti-mouse CD16/CD32(Mouse BD Fc Block) and stained with in-house–generated hamster anti-mouse PILRa mAb, followed by allophycocyanin-conjugated anti-hamsterIgG (R&D Systems). BM cells were further stained with PE-conjugatedLy6G and FITC-conjugated anti-CD11b. Peritoneal cells were stained withPE-conjugated Gr-1 and FITC-conjugated anti-CD11b or FITC-conjugatedCD117 and PE-conjugated FcεRI (all from BD Pharmingen). Cell acquisi-tion was performed on a FACSCalibur (Becton Dickinson, Mountain View,CA), and data were analyzed with FlowJo software.

Induction of CAIA

Arthritis was induced in 7–8-wk-old (age-matched for a given study) fe-male C57BL/6N or BALB/c mice with an arthritogenic anti–type II col-lagen mAb mixture purchased from Chondrex. Pilra2/2 and WT littermatemice received 4 mg a mixture of anti–type II collagen mAbs i.v. in 400 ml

The Journal of Immunology 861

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

PBS on day 0, followed by 50 mg LPS i.p. in 100 ml PBS on day 3. To testthe effect of anti-PILRa mAb on the development of CAIA, BALB/c micereceived 2 mg Arthrogen-CIA Ab mixture i.v. in 200 ml PBS on day 0,followed by 50 mg LPS i.p. in 100 ml PBS on day 3. Mice were treatedwith 100 mg mouse anti-PILRa mAb or anti-ragweed mIgG1 isotypecontrol i.p. daily in 100 ml sterile saline, starting on day 21. For K/BxNserum–transfer arthritis model, BALB/c mice were given 20 ml arthrito-genic K/BxN serum on days 0 and 2 by i.v. injection and were treatedwith 100 mg mouse anti-PILRa mAb or anti-ragweed mIgG1 isotypecontrol i.p. daily in 100 ml sterile saline starting on day21. Swelling of thepaws was monitored for 11–12 d. Each paw was assigned a score based onthe following scale: 0 = no evidence of erythema and swelling; 1 = erythemaand mild swelling confined to the midfoot (tarsal) or ankle; 2 = erythema andmild swelling extending from the ankle to the midfoot; 3 = erythema andmoderate swelling extending from the ankle to the metatarsal joints; and 4 =erythema and severe swelling encompass the ankle, foot, and digits. Theclinical score of each mouse is the sum of the four paw scores.

Histopathological assessment

Paws were removed, fixed in formalin, decalcified, and processed toparaffin sections stained with H&E. Sagittal sections were examined bylight microscopy and scored on arbitrary scales ranging from 0 to 5 forfeatures of arthritis, including number of affected joints, extent of in-flammation, extent of soft tissue proliferation including pannus, cartilageinjury, and bone remodeling. Immunoreactivity for MPO or F4/80 wasevaluated on arthritic paws by microscopic examination of affected sy-novial structures (joints, tendon sheaths). Intensity of infiltration withimmunoreactive cells was estimated using an arbitrary scoring schemeranging from 0 to 3, according to the following criteria: 0 = nonedetected near or in synovial structures; 1 = few, scattered positive cellsnear or in synovial structures; 2 = frequent disseminated or looselyclustered positive cells near or in synovial structures; and 3 = locallyextensive infiltration with numerous positive cells near or in synovialstructures.

Microcomputed tomography imaging

Microcomputed tomography (micro-CT) imaging was performed usinga previously described micro-CT and automated analysis technique (26) toquantify bone destruction in the arthritis mouse model. Briefly, the pawswere scanned ex vivo on a micro-CT 40 scanner (Scanco Medical,Br€uttisellen, Switzerland). The analysis algorithm automatically locatesfive metatarsophalangeal joints and three metacarpophalangeal joints andsegments the cortical bone and periosteal new bone. Joint cortical bonevolume (JCBV) is sensitive to cortical bone erosion at the joints, andperiosteal new bone volume (PNBV) estimates the amount of periostealnew bone formation that results from a repair response to the bone erosion.The analysis technique was modified in such way that JCBV, as well as thePNBV estimate, was limited to the joints.

Measurement of cytokine concentration in arthritic hind paws

Hind footpads were obtained from mice by cutting at the borderline offur growth. The footpads were hammer smashed and shaken at 20 Hz for30–60 min with 5-mm steel beads at 4˚C in ice-cold cell lysis buffer(Cell Signaling Technology) containing Protease Inhibitor Cocktail Set I(Calbiochem). Homogenized tissue was kept on ice for 30 min, vor-texed occasionally, and then spun down at high speed for 15 min at 4˚C.Supernatants were collected, and all samples were frozen at 280˚C andthawed to room temperature before analysis. Cytokine levels were ana-lyzed by ELISA (R&D System). The concentration of total protein in thesupernatants was measured using a Pierce BCA kit and was normalizedagainst the concentration of BSA. The concentration of cytokines wasexpressed as pg/mg of protein.

In vitro stimulation of BM-derived DCs

BM-derived DCs (BMDCs) were generated as described elsewhere (27). Inbrief, femurs of mice were removed, and BM was flushed out with PBScontaining 10% FCS. RBCs were lysed with ACK lysing buffer. Cells werecultivated in bacterial culture dishes for 9–10 d at 37˚C in a humidified

FIGURE 1. Pilra2/2 mice generation and char-

acterization. (A) Schematic diagram of PILRa-tar-

geting construct. Exons 1 and 2 were replaced with

loxp sites (triangles) flanked by the neomycin re-

sistance gene (neo). The neomycin cassette was

deleted by crossing the mice with the Rosa-26-cre

knockin line. (B) Genomic DNA from WT ES cells

or targeted ES cells were digested with SacI and

probed with a neomycin internal probe (left panel).

All independent clones detected a single band of an

8.6-kb fragment corresponding to a single integra-

tion event at the 59 homology arm. SphI genomic

DNA digest was used to confirm expected homol-

ogous recombination at the 39 end using a genomic

probe located between the 39 end of the targeting

arm and the SphI site, which generated a shorter

fragment of 6.0 kb compared with 11.0 kb in WT

ES cells (right panel). (C) PILRa and PILRb ex-

pression in spleen and BM of WT and Pilra2/2

(KO) mice by RT-PCR (n = 3). (D) PILRb ex-

pression in spleen and BM of WT and Pilra2/2

(KO) mice by real-time PCR. (E) PILRa expression

in spleen and BM of WT, Pilra+/2 (HET), and

Pilra2/2 (KO) mice by Western blot. Data are

representative of two or three experiments.

862 PILRa REGULATES INFLAMMATION

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

atmosphere containing 5% CO2 using RPMI 1640 supplemented with 10%heat-inactivated FCS, 2 mM L-glutamine, 100 U/ml penicillin, 100 mg/mlstreptomycin, 50 mM 2-ME (Sigma; Taufkirchen, Germany), and 20 ng/mlrecombinant mouse (m)GM-CSF (R&D Systems). At days 9–10, thenonadherent cells were harvested and stimulated with mIgG (60 mg/ml;Jackson ImmunoResearch Laboratories) plus various concentrations ofmouse anti-mouse PILRa mAb coated on Protein G Agarose beads (Roche)for 18 h. Cytokine production in supernatants was detected by ELISA(R&D) or Luminex (Bio-Rad).

PILRa and PILRb expression in synovial tissues of RA andosteoarthritis patients

Patients and synovial tissues. Synovial tissues were obtained from RAsubjects undergoing arthroplasty and/or synovectomy of affected joints(University of Michigan, two sequential cohorts). RA was diagnosed basedupon the 1987American College of Rheumatology criteria (28). Patients weretreated using the standard of care for RA (nonsteroidal anti-inflammatorydrugs and disease-modifying antirheumatic drugs), and some patients alsowere treated with biologics (adalimumab, etanercept, infliximab, anakinra,and rituximab). Patients were diagnosed with RA $3 y before surgery, and70% of patients for whom data were available were positive for rheumatoidfactor. All procedures to collect human specimens were performed undera protocol approved by the University of Michigan Institutional ReviewBoard. Excised tissues were immediately snap-frozen in liquid nitrogen andstored at 280˚C. Each tissue was used for both histology and RNA ex-traction. For cryosectioning, samples were brought briefly to 220˚C, sec-tioned, and immediately returned to 280˚C to maintain RNA integrity. Alltissues used for downstream studies were prospectively randomized duringprocessing and sectioning, prior to expression analysis, to minimize technicalbatch effects in the data.

RNA isolation. Frozen samples were weighed and homogenized in RLT(QIAGEN, Valencia, CA) + 2-ME (10 ml/ml) at a concentration of 100 mg/ml. RNA was isolated using an RNeasy Mini Kit (QIAGEN) with on-column DNase digestion.

Microarray hybridization. The protocols for preparation of cRNA and forarray hybridization were followed as recommended by Affymetrix (Santa

Clara, CA). Samples were hybridized to GeneChip Human Genome U133Plus 2.0 Arrays (Affymetrix). Arrays were washed and stained in the Af-fymetrix Fluidics station and scanned on a GeneChip scanner 3000. Ex-pression signals were obtained using the Affymetrix GeneChip operatingsystem and analysis software. Data have been deposited in the NationalCenter for Biotechnology Information’s Gene Expression Omnibus underaccession no. GSE48780 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE48780).

Computational analysis. Computational analysis was carried out using the Rprogramming language (29) and libraries from the open-source Bioconductorproject (30). Quality assessment of microarray data was conducted using theBioconductor affy package (31), and the expression data were normalizedusing the robust multiarray average method (32, 33). PILRa and PILRb areeach represented by two probes on the Affymetrix U133Plus chips.

To compare the expression levels of PILRa and PILRb in RA andosteoarthritis (OA) samples, we used a linear model with terms for diag-nosis (OA or RA) and inflammation status (inflamed or noninflamed) basedon histology. Conceptually, this can be thought of as a t test comparing OAand RA expression with correction for inflammation status. The limmaBioconductor package was used for the computation (34).

PILRa ligand screen

Recombinant human PILRa-Fc was screened for interacting partners usingthe Genentech human full-length cDNA expression library. COS-7 cellswere transfected with 6 ng/ml individual cDNA expression vectors andFuGene 6 (Roche) in a 384-well format. Forty-eight hours after transfec-tion, COS-7 cells were exposed to 10 ng/ml PILRa-Fc, followed by fix-ation in 4% formaldehyde. Protein–protein interactions were detected uponbinding of an Alexa Fluor 488–conjugated anti-mIgG2a secondary Ab(Invitrogen) to the cell surface using an MD Isocyte plate scanner or, infollow-up experiments, using a GE IN Cell 2000 Analyzer.

To confirm the binding of screening hit to PILRa, HEK 293T cellswere transfected with C-type lectin domain family 4 (CLEC4G) cDNA,and transfectants were stained with human (h)PILRa-Fc/mPILRa-Fc,hPILRaR126A-Fc/mPILRaR133A-Fc, followed by FITC-conjugated anti-mIgG2a. Cell acquisition was performed on a FACSCalibur (Becton

FIGURE 2. Expression of PILRa in various tis-

sues and cells. (A) Immunohistochemistry of nor-

mal murine tissues. Tissue sections from WT

mouse spleen, liver, lung, and BM were stained

with anti-PILRa Ab and compared with control

tissues from Pilra2/2 mice. Spleen red pulp and

tissue-resident macrophages, such as Kupffer cells

in liver (inset) or exfoliated alveolar macrophages

(inset) in lung, express PILRa. In BM, most

PILRa+ cells are identified as granulocytes or their

precursors. Scale bars, spleen = 500 mm; liver and

lung = 50 mm; BM = 20 mm. (B) PILRa is

expressed on murine granulocytes and monocytes

from BM and peritoneal macrophages, neutrophils,

and mast cells, but it is not expressed on spleen

lymphocytes. Data are representative of three

experiments.

The Journal of Immunology 863

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

Dickinson, Mountain View, CA), and data were analyzed with FlowJosoftware.

Statistical analysis

Statistical significance was determined by the parametric Dunnett test for thecomparison of joint cytokine concentrations, by the nonparametric Dunnetttest for the comparison of arthritis scores and histopathological as well asmicro-CT results, and by Student t test for the comparison of in vitro cytokinedata. The p values , 0.05 were considered significant.

ResultsGeneration and characterization of Pilra2/2 mice

To generate the PILRa-knockout mouse, exon 1 and exon 2 ofthe PILRa gene were deleted by homologous recombination inC57BL/6J ES cells (Fig. 1A), and the targeted locus was con-firmed by Southern blot analysis using internal (Fig. 1B, leftpanel) or external (Fig. 1B, right panel) probes, as described in

Materials and Methods. Chimeric mice were bred to C57BL/6Nmice to generate PILRa-deficient mice on a pure genetic back-ground. Given the close proximity of PILRb, the neo-pgk cassettewas removed by breeding PILRa-knockout mice to Rosa-26-creknockin mice to avoid nonspecific effects on PILRb or other sur-rounding genes (35). RT-PCR analysis revealed that PILRa mRNAproduction was abolished in the spleen and BM of Pilra2/2 mice,whereas PILRb expression was not affected (Fig. 1C). Real timeRT-PCR was done to better quantitate PILRb levels in WT andPilra2/2 mice. These expression studies showed that PILRb ex-pression in spleen and BM was not affected (Fig. 1D). PILRaprotein was abundant in WT BM and was detected in spleen atlower levels. However, in Pilra2/2 mice, no PILRa protein wasdetected in either BM or spleen, and Pilra+/2 heterozygous micedisplayed intermediate levels of protein (Fig. 1E). Pilra2/2 miceare viable, fertile, and have a normal lifespan. They did not show

FIGURE 3. Pilra2/2 mice are more

susceptible to arthritis. (A) Immunohis-

tochemistry of inflamed joint tissue

(CAIA model, day 11). Within the

cavity of an inflamed joint, PILRa is

expressed on neutrophils (arrows), the

predominant cell type in CAIA. (B–E)

C57BL/6N WT (n = 6) and Pilra2/2

mice (n = 8) were given arthritogenic

anti–type II collagen mAb mixture to

induce arthritis. (B) Pilra2/2 mice de-

velop more severe CAIA. Pilra2/2 mice

showed increased average clinical score

(left panel), average daily clinical score

(days 4–12, middle panel), and end

point clinical score (day 12, right panel)

compared with WT mice. (C) Hind

paws, sagittal H&E. Representative

images of the most severe lesions per

group. Periarticular infiltration with in-

flammatory cells and formation of intra-

articular exudate are obvious in Pilra2/2

mice (arrows) compared with WT mice,

where these features are essentially ab-

sent. Scale bar, 200 mm. (D) Lesion

scores for features of arthritis, including

infiltration with inflammatory cells,

fibroplasia, cartilage injury, and bone

remodeling score per group on day 12,

are lower in WT mice compared with

Pilra2/2 mice. Data points represent

average scores for two paws/mouse.

Two limbs per animal, four to six sec-

tions/animal, and a total of 14 mice were

examined. (E) Hind paws from mice

harvested on day 12 were subjected to

micro-CT analysis. Images show high

resolution three-dimensional rendering

of micro-CT scans. Quantification of

bone integrity, including JCBV and

PNBV are shown. Results are represen-

tative of two experiments. The micro-

CT parameter estimates for Pilra2/2

mice were compared with the control

group using the Dunnett test.

864 PILRa REGULATES INFLAMMATION

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

any appreciable developmental defects in myeloid or lymphoidcell populations (Supplemental Table I).Previous work showed that PILRa transcripts are relatively high

in spleen, liver, and lung (8). We tested PILRa protein expressionin these tissues by immunohistochemistry (Fig. 2A) using Pilra2/2

mice as a negative control and in-house–generated Armenianhamster anti-mPILRa mAb, which did not show cross-reactivityto mPILRb. In the spleen, PILRa+ cells are mainly localized inthe red pulp, consistent with expression predominantly by myeloidcells. In liver and lung, PILRa is expressed in tissue-residentmacrophages, such as Kupffer cells or exfoliated alveolar mac-rophages, respectively. In BM, most PILRa+ cells are identifiedas granulocytes or their precursors. These data corroborate thenotion that PILRa is expressed primarily in cells of the myeloidlineage. Correspondingly, PILRa protein is detected on granulo-cytes (CD11b+Ly6G+) and monocytes (CD11b+Ly6G2) from BM,as well as peritoneal cavity macrophages (CD11b+Gr-12), neu-trophils (CD11b+Gr-1+), and mast cells (CD117+FcεRI+), but noton spleen lymphocytes, including T, B, NKT, and NK cells(Fig. 2B).

Pilra2/2 mice develop enhanced autoimmune arthritis

To investigate the biological function of PILRa in myeloid cell–mediated immune responses and its role in autoimmune diseases,we made use of a CAIA model, which is a myeloid-dependentpreclinical disease model. We observed an infiltration of PILRa-producing cells in mouse arthritic tissues, specifically periarticularsoft tissue obtained from CAIA (Fig. 3A). We first tested howCAIA is affected by PILRa deficiency. As shown in Fig. 3B (left

panel), littermate WT mice started to develop clinical signs ofarthritis 5 d after injection of the type II collagen–specific Abmixture, 2 d after LPS administration, and reached a maximumclinical score of 3.8 6 2.6 (mean 6 SD) between days 7 and 9before gradually resolving disease. Pilra2/2 mice showed similarkinetics of disease development as did WT mice, but theyexhibited more severe disease, with maximum clinical scores of8.0 6 2.6 (mean 6 SD). Pilra2/2 mice also had significantlyincreased average clinical scores and end point clinical scorescompared with WT mice (Fig. 3B, middle and right panels).On termination of the experiment at day 12, histopathology

confirmed the clinical observations. Compared with WT mice,Pilra2/2 mice had more severe disease, as shown by all evaluatedparameters, including number of affected joints/paw, periarticularinfiltration with mixed inflammatory cells, soft tissue proliferation(including pannus formation), cartilage injury, and bone remod-eling (Fig. 3C, 3D). Consistent with the histological evidence formore extensive bone remodeling, micro-CT revealed more severeosteolysis (i.e., smaller JCBV), as well as increased new boneformation (i.e., greater PNBV), in Pilra2/2 mice compared withWT mice (Fig. 3E). These results support the regulatory role ofPILRa in downregulating myeloid-dependent inflammation.

PILRa deficiency results in an increase in intra-articularproinflammatory cytokine levels at the onset of arthritis

To better understand how PILRa deficiency impacts CAIA, wetested inflammatory cytokine levels in joint tissue at the onset ofdisease. Onset of disease was similar in WT and Pilra2/2 mice,with the start of clinical signs of arthritis on day 5 following in-

FIGURE 4. Elevated levels of inflammatory cytokines were detected in joints of Pilra2/2 mice at the onset of disease. (A) C57BL/6N WT (n = 9) and

Pilra2/2 mice (n = 9) were given arthritogenic anti–type II collagen mAb mixture to induce arthritis (CAIA). On day 5, hind footpads were obtained from

mice for homogenization in cell lysis buffer containing protease inhibitors. Cytokine levels in lysate supernatants were analyzed by ELISA. The cytokine

concentrations are expressed as pg/mg of protein. (B) Kinetics of neutrophils and macrophages in the early phase of CAIA show a similar degree of

infiltration in arthritic joints of Pilra2/2 and WT mice. WT (dark gray bars, n = 4) and Pilra2/2 mice (light gray bars, n = 4) were given arthritogenic anti–

type II collagen mAb mixture to induce arthritis. On days 3, 4, and 5, paws were collected for immunohistochemical staining of MPO and F4/80. Intensity

of infiltration was estimated using an arbitrary scoring scheme ranging from 0 to 3. Bars represent mean of four arthritic paws. Error bars are SEM. Results

are representative of two or three separate experiments. The p values are based on the parametric Dunnett test.

The Journal of Immunology 865

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

jection of type II collagen–specific Abs. At this time point, hindpaw lysates of Pilra2/2 mice exhibited significantly increased pro-duction of proinflammatory cytokines, including IL-1b and IL-6,as well as neutrophil and monocyte chemokines KC and MCP-1(Fig. 4A). The increased inflammatory cytokine and chemokineproduction could be due to either increased inflammatory cellnumber or activity. It was shown that PILRa plays an importantrole during acute inflammatory responses in regulating neutro-philic migration (16). We then asked how cellular infiltrates areimpacted in the ankle joints at early time points each day from day3 to day 5 using immunohistochemistry MPO to stain neutrophilsor F4/80 to localize macrophages (Fig. 4B). Neutrophils wereslightly reduced in PILRa-knockout joints on day 3; however,their numbers were transiently increased on day 4 compared withWT mice. Macrophages in PILRa-knockout joints also showeda similar trend on day 3; however, their numbers were slightlyincreased on day 5. Overall, the changes in macrophage andneutrophil cellularities were not statistically significant betweenWT and Pilra2/2 mice (Fig. 4B). The slight reduction in neu-trophil/macrophages in PILRa-knockout mice on day 3 mightbe secondary to the systemic effect of LPS on the increased mi-gration of these cells to other tissues, such as liver, in these mice(16). In addition, we did not observe any difference between WTand Pilra2/2 neutrophil and macrophage cellular infiltrates in anacute localized inflammatory model of Staphylococcus aureus(Supplemental Fig. 1). These observations support the notion thatthe lack of PILRa has a global effect on the severity of arthritis,mainly through enhanced cytokine production. In summary, ourdata suggest that the joint-infiltrating Pilra2/2 myeloid cells arefunctionally hyperresponsive, further highlighting the regulatoryrole of PILRa in myeloid cells.

Engagement of PILRa suppresses the production ofinflammatory cytokines and chemokines

To confirm the regulatory role of PILRa in modulating cellularfunction, we tested how the production of cytokines and chemo-kines is affected in Pilra2/2 cells or upon its engagement in vitro.We used various PILRa-expressing cell types, such as BMDCs,BM-derived monocytes (BMDMs), and neutrophils, which areimplicated in the pathology of anti-CAIA. First, we tested howthese cells respond to LPS, which is used to activate myeloidcells in the same CAIA model. The results showed that Pilra2/2

BMDCs produced increased TNF-a and MCP-1 in response toLPS but not IL-1b, IL-6, or KC (Supplemental Fig. 2). A subtletrend of cytokine elevation was seen in neutrophils and BMDMs(Supplemental Fig. 2). The state of cellular hyperresponsiveness,as measured by cytokines/chemokines, appears to vary in differ-ent PILRa-expressing cells. BMDCs were more sensitive to thispathway, which might be explained by the high expression ofPILRa (Fig. 5A) in these cells.We then asked how engagement of PILRa receptor in BMDCs

modulated cytokine production. To couple stimulatory and puta-tive inhibitory pathways, we used mIgG-coated beads as the stim-ulus. WT and Pilra2/2 BMDCs produced similar levels of theproinflammatory cytokine TNF-a and chemokine MCP-1 in re-sponse to mIgG-coated beads (Supplemental Fig. 2). However,stimulation of BMDCs with beads coated with anti-mPILRamAb,but not isotype control, showed a dose-dependent inhibition of TNFand MCP-1 production in WT cells. The inhibitory effect of anti-PILRa Ab was not seen in the BMDCs derived from Pilra2/2 cells,again confirming that the inhibitory effect is PILRa dependent (Fig.5B). This result indicates that engagement of PILRa downregulatesproinflammatory cytokine and chemokine production when it isproximal to an activating receptor, such as FcgR.

A recent study showed that PILRa associates with its ligands incis on neutrophils (16). Because BMDCs express both PILRaand its ligands (Supplemental Fig. 3A), we predicted that PILRawould interact with its ligands in cis, and this may play a role innegatively regulating BMDC function. If this were the case, dis-sociation of the interaction between PILRa and its ligands wouldincrease BMDC cytokine production. We showed previously thatsialic acid is an essential component of PILRa ligands in pri-mary cells, and treatment of primary cells with sialidase A, whichcleaves sialic acid from surface proteins, abolished their bindingto PILRa (9). Therefore, we used sialidase A treatment to disso-ciate PILRa–ligand interactions on BMDCs. We found that sial-idase A treatment increased cytokine production by BMDCs inresponse to mIgG coated on protein G beads, suggesting that theinteraction of sialylated ligands with their receptors inhibits cel-lular function (Supplemental Fig. 3B). Given the fact that PILRais able to interact with a network of ligands expressed by differenttissues, including hematopoietic cells other than myeloid cells (9),cis and trans interactions may both be involved in the regulationof PILRa-expressing myeloid cell function.

Anti-PILRa mAb treatment attenuates arthritis

We then tested whether treatment with anti-PILRa mAb affectsinflammation in vivo using the CAIA model. Because C57BL/6NWT mice develop only mild disease, we used BALB/c mice in-stead for anti-PILRa mAb treatment. We found that anti-PILRamAb treatment significantly reduced inflammation compared withisotype control treatment (Fig. 6A). Isotype control–treated micestarted to develop clinical signs of arthritis 4 d after injection oftype II collagen–specific Ab mixture or 2 d after LPS admin-istration and reached maximum clinical scores of 14.6 6 2.2(mean 6 SD) between days 5 and 7. By contrast, anti-PILRa mAb–

FIGURE 5. Engaging PILRa on BMDCs reduces cytokine production.

(A) BMDCs express PILRa. WT and Pilra2/2 BMDCs were stained with

anti-mPILRa mAb or isotype control. (B) WT and Pilra2/2 BMDCs were

stimulated with mIgG plus various concentrations of mouse anti-mPILRa

mAb–coated Protein G Agarose beads for 18 h, and cytokine production in

the supernatants was measured. Results are representative of three experi-

ments. *p , 0.05, Student t test.

866 PILRa REGULATES INFLAMMATION

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

treated mice exhibited delayed disease onset, which graduallyincreased to a much reduced maximum clinical score of 4.4 6 4.3(mean 6 SD) by the end of the study. Anti-PILRa mAb–treatedmice also showed significantly reduced average clinical scoresand end point clinical scores compared with isotype control–treated mice (Fig. 6A, middle and right panels). In accordancewith in vivo findings, histopathological assessment of hind paws atthe termination of the study at day 11 showed significantly de-creased scores for all scored features in anti-PILRa mAb–treatedmice compared with isotype control Ab–treated mice (Fig. 6B).Finally, micro-CT imaging of paws at day 11 showed that anti-PILRa mAb–treated mice exhibited less severe bone remodelingcompared with isotype control Ab–treated mice (Fig. 6C), asmeasured by JCBV and PNBV (p , 0.01 for both parameters).The observation that anti-PILRa treatment attenuates arthritisdevelopment, whereas Pilra2/2 mice show enhanced disease, sug-gests that the anti-PILRa mAb does not block the receptor butrather engages it. To measure the impact of PILRa modulation onother arthritis models, we tested the effect of anti-PILRa treat-ment on mouse K/BxN serum–transfer arthritis. The K/BxN se-rum–transfer model is a widely accepted model of inflammatoryarthritis that shares features of human RA (25). In K/BxN mice,the induction of disease does not require LPS, and this diseasedepends highly on the innate cells in a TNF-independent fashion(36–39). Mice started to develop clinical signs of arthritis 1 dafter K/BxN serum transfer. Isotype control–treated mice reachedmaximum clinical scores of 14.7 6 1.0 (mean 6 SD), whereas

anti-PILRa mAb–treated mice exhibited a reduced maximumclinical score of 8.5 6 4.0 (mean 6 SD) between days 5 and 7(Fig. 6D, left panel). Anti-PILRa mAb–treated mice also showedsignificantly reduced average clinical scores and terminal clinicalscores compared with isotype control–treated mice (Fig. 6D,middle and right panels). Additional studies are required to un-derstand how PILRa is relevant in the context of additional in-flammatory diseases.

PILRa is expressed in human inflammatory arthritic tissuesand correlates with monocyte and DC levels

To test whether PILRa is involved in human inflammatory ar-thritis, we assessed the expression patterns of PILRa and its coun-terpart PILRb using mRNA microarrays on human synovial tissue.Samples were obtained during joint resection surgery from OA andRA patients. Sections were taken for both histological observationand mRNA extraction. The Affymetrix expression microarrays in-cluded two probes each for PILRa and PILRb. PILRa consistentlyshowed significantly higher expression in the RA patients com-pared with the OA patients, including a correction for inflamma-tion status (p , 1e27) (Fig. 7A, upper panels). In contrast, PILRbshowed a modestly higher level of expression in OA patients (p ,0.02) (Fig. 7A, lower panels). The differential expression of PILRaand PILRb in inflammatory RA suggests that PILRa might func-tion as a negative-feedback regulatory loop to dampen inflammation.Cell-specific gene signatures can be used as an estimate of cellularity(40). For each cell type, we calculated a summary gene set score

FIGURE 6. Anti-PILRa mAb treatment attenu-

ates arthritis development. (A–C) BALB/c mice

were given arthritogenic anti–type II collagen mAb

mixture to induce arthritis (CAIA) and were treated

with anti-PILRa mAb (n = 5). (A) Anti-PILRa

mAb–treated mice exhibited reduced disease de-

velopment compared with isotype control. Anti-

PILRa–treated mice showed decreased average

clinical score (left panel), average daily clinical

score (days 4–11, middle panel), and end point

clinical score (day 11, right panel) compared with

isotype control–treated mice. (B) Pathological

scores of isotype control and anti-PILRa mAb–

treated mice on day 11. Data points represent av-

erage scores for two paws/mouse. Two limbs per

animal, four to six sections/animal, and a total of

10 mice were examined. (C) Hind paws from mice

harvested on day 11 were subjected to micro-CT

analysis. Images show high-resolution three-di-

mensional rendering of micro-CT scans. Quantifi-

cation of bone integrity, including JCBVand PNBV.

(D) Anti-PILRa mAb treatment inhibited disease

development in a K/BxN serum–transfer arthritis

model. BALB/c mice were given 20 ml of arthro-

genic K/BxN serum on days 0 and 2 by i.v. injec-

tion and were treated with anti-PILRa mAb (n = 6).

Mean clinical score (left panel), average daily

clinical score (middle panel), and day-7 clinical

score (right panel) are shown. In vivo arthritis

scores and micro-CT parameter estimates for the

treatment groups were compared with the control

group using the Dunnett test. Results are repre-

sentative of two experiments.

The Journal of Immunology 867

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

using a quartile-trimmed mean of the normalized probe set valuespresent in the gene set. This summary score provides an estimate forthe relative abundance of the given cell type across samples. Cor-respondingly, we found that PILRa expression was highly correlatedwith the tissue-specific gene sets obtained from different inflam-matory cell types, such as DCs (Spearman r = +0.7, p , 2e216),neutrophils (r = +0.5, p, 2e25), and monocytes (r = 0.8, p, 2e216) in RA samples (Fig. 7B, upper panels). In contrast, PILRbexpression was minimally inversely correlated with DC-specific andmonocyte-specific genes (r = 20.2 for both, p , 0.01), and it was

not significantly correlated with neutrophil-specific genes (r =20.1,p , 0.3) (Fig. 7B, lower panels). These trends held true when weconsidered all samples together or RA and OA samples in isolation.Our human expression study shows that PILRa expression maydissociate from PILRb, and this inhibitory pathway is overrepre-sented in myeloid cells in human RA.

Diverse ligands are involved in PILRa pathway

We (9) and other investigators (8, 10, 41) reported that PILRaengaged multiple ligands with common molecular determinants

FIGURE 7. PILRa-inhibitory versus PILRb-activating pathways in human RA joints and their correlation with pathogenic cell types. Synovial samples

were taken from RA (n = 81) and OA (n = 121) patients during joint resection surgery. (A) The Affymetrix expression microarrays included two probes each

for PILRa and PILRb. PILRa expression is significantly higher (p, 1e207) in RA patients compared with OA patients; this also holds when corrected for

inflammation status. PILRb expression is modestly higher in OA patients compared with RA patients (p , 0.02), correcting for inflammation status. (B)

The Affymetrix expression microarrays of PILRa and PILRb. In RA patients, PILRa expression is significantly correlated with DC-specific genes (r =

+0.7, p , 2e216), neutrophil-specific genes (r = +0.5, p , 2e25), and monocytes (r = +0.8, p , 2e216). This trend also holds true for all of the in-

flammatory RA subset of patients. In contrast, PILRb expression was not positively correlated with DC, neutrophil, or monocyte gene expression (r =20.2,

p = 0.01; r = 20.1, p = 0.2; and r = 20.2, p , 0.01). Scores were calculated using a quartile-trimmed mean of the normalized probe set values present in

the gene set.

868 PILRa REGULATES INFLAMMATION

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

(9). Using recombinant hPILRa-Fc to screen a high-throughputhuman full-length cDNA expression library, we identified an ad-ditional PILRa ligand CLEC4G [previously named LSECtin (42)](Fig. 8A). We confirmed its binding to both human and mousePILRa by transfecting HEK 293T cells with this putative ligandand staining with human and mouse PILRa-Fc for analysis byflow cytometry (Fig. 8B). Consistent with our previous find-ings that an evolutionarily conserved PILRa arginine site(hPILRaR126A and mPILRaR133A) is essential for its interac-tion with diverse sialylated ligands, we found that hPILRaR126A-Fc and mPILRaR133A-Fc lost their binding activity to theseligands (Fig. 8B), suggesting that they all share the same sialy-lated PILRa binding domain. mPILRb-Fc also bound to CLEC4G(Fig. 8B, far right panel). Thus, a complex network of ligandsusing a common molecular determinant may modulate the PILRapathway in different tissues.

DiscussionIn this study, we reported that PILRa represents a potent inhibitoryreceptor that negatively regulates the function of myeloid cellsto control inflammation in mouse arthritis models. PILRa ispredominantly expressed by macrophages, DCs, and neutrophils,all of which are important in the pathogenesis of arthritis. UsingPILRa-knockout mice in a CAIA model, we demonstrate that theabsence of PILRa leads to dysregulation of inflammatory pro-cesses in inflamed tissues, resulting in increased inflammatorycytokine production and severity of disease. Conversely, engagingPILRa with an anti-PILRa mAb reduces disease severity in bothCAIA and K/BxN serum–transfer models.In preclinical arthritis models, as well as in human inflammatory

arthritis, PILRa gene expression profiling supports the notion thatthis receptor acts in a counter-regulatory fashion to dampen in-flammation. It is interesting that PILRa (inhibitory receptor), butnot PILRb (activating receptor), expression is positively corre-lated with monocytes or neutrophil gene signatures in human RA,suggesting that this pathway is highly regulated via its ligands topossibly keep these inflammatory cells in check. The usefulnessof the PILRa pathway in human RA or additional inflammatorymodels requires addition studies.A complex network of ligands expressed in a variety of tissues

has been implicated in PILRa interactions (8–10). Although we donot fully understand the consequence of the interaction of PILRa

with individual ligands, it is interesting that a conserved domainin PILRa is critical for its interaction with all known ligands,a property shared with the Siglec family of inhibitory receptors(9, 43). A recent study showed that PILRa interacting domain incis engaged its ligands in the same cell (16). The new ligandCLEC4G is expressed in macrophages, it might be able to interactin cis with PILRa. CLEC4G is implicated in Ebola virus entryreceptor, as well as clearance and presentation of various Ags(44). Given the complexity of the PILRa receptor/ligand net-work, PILRa appears to have a diverse function in that it finetunes the immune response depending on inflammatory cues,similar to other paired receptors (45). Additional studies are re-quired to determine how cis/trans interactions of PILRa ligandsare modulated during inflammation where this pathway is up-regulated.In the context of inflammatory arthritis, the main effect of

PILRa knockout is identified as an enhanced production of pro-inflammatory cytokines, resulting in more severe disease. At theonset of arthritis, PILRa-knockout mice produced higher levelsof proinflammatory cytokines, such as IL-6 and IL-1b, as well aschemokines, such as KC and MCP-1, in joint tissues, even thoughboth WT and Pilra2/2 cellular infiltrates in arthritic joints weresimilar in WT and Pilra2/2 mice by histopathology. This suggeststhat, in the absence of PILRa, myeloid cells infiltrating thejoints are hyperresponsive, supporting a regulatory role for PILRain these cells. Although the myeloid cells in joint tissues at theonset of disease are hyperresponsive in terms of proinflamma-tory cytokine production, we did not detect robust hyperrespon-siveness of PILRa-expressing cells in the absence of PILRain vitro. Pilra2/2 BMDCs produced mildly, but consistently, in-creased levels of TNF and MCP-1 in response to LPS, whereas WTand Pilra2/2 BMDCs produced similar levels of these cytokines inresponse to mIgG binding to Protein G beads (Supplemental Fig. 2).For BMDMs, Pilra2/2 mice showed a trend toward increasedproinflammatory cytokine and chemokine production in response toLPS (Supplemental Fig. 2). Therefore, the cumulative effect of thisaugmented response may explain the robust phenotype in the contextof experimental arthritis. The inhibitory effect of PILRa receptormediated by Ab cross-linking in vitro suggests that, when proximal,stimulatory pathways, such as FcgR, can be diminished. PILRb wasshown to promote cytokine production in myeloid cells, andPILRb2/2 mice and BMDMs produced fewer proinflammatorycytokines (14). In addition, PILRa is able to regulate neutrophilinfiltration during acute inflammation via modulation of integrinactivation in an acute inflammatory peritonitis model (16), inwhich PILRa-knockout mice show increased infiltration of neu-trophils upon thioglycollate challenge (16). In contrast, in CAIAwe did not see a difference between WTand PILRa-knockout micewith regard to the number of neutrophils and macrophages in ar-thritic joints. In support of this finding, we did not observe anydifference between WT and Pilra2/2 neutrophil and macrophagecellular infiltrates in an acute localized inflammatory model ofS. aureus (Supplemental Fig. 1). Despite these differences, bothstudies suggest that PILRa plays a negative regulatory role in theinflammatory process, likely by complex mechanisms under dif-ferent inflammatory cues. Our findings suggest that, in the contextof sterile inflammation, PILRa is a regulatory receptor that mod-ulates inflammatory cytokine responses; however, we cannot ruleout that PILRa might have additional functions outside of cytokineproduction to affect inflammatory responses.Anti-PILRa treatment reduced inflammatory arthritis. Consis-

tent with this observation, engaging PILRa on BMDCs, whichhighly express PILRa on their surface, inhibits their functionwhen a stimulating receptor, such as FcgR, is coengaged. Thus,

FIGURE 8. CLEC4G is a novel ligand of PILRa. (A) Recombinant

hPILRa-Fc was screened for interacting partners using a Genentech hu-

man full-length cDNA expression library. COS-7 cells were transfected

with individual cDNA expression vector and then exposed to PILRa-Fc,

followed by fixation. Protein–protein interactions were detected with an

Alexa Fluor 488–conjugated anti-mouse IgG2a secondary Ab and scanned

with an MD Isocyte plate scanner. (B) HEK 293T cells were transfected

with expression vectors for CLEC4G and then stained with h/mPILRa-Fc,

hPILRaR126A/mPILRa-R133A-Fc, or mPILRb-Fc (black line) or isotype

control IgG (gray line).

The Journal of Immunology 869

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from

PILRa-knockout and anti-PILRa–agonizing studies complementeach other in demonstrating the regulatory effect of this factorin two rodent models of RA. Similarly, engagement of PILRareduces serum or bronchoalveolar lavage fluid levels of proin-flammatory cytokines, including IL-1b, TNF, and IL-6, in a modelof S. aureus–induced pneumonia (14). Further studies are re-quired to understand what counter-regulatory receptors might spe-cifically interact with PILRa to modulate its function in myeloidcells and how much this pathway contributes to other inflam-matory diseases. The contribution of the PILRa pathway mayvary, depending on the type of myeloid cell, inflammation typeand stage, stimulation strength, and the ligand involved. Thus,PILRa is emerging as an important regulatory pathway in myeloidcells and, therefore, is an attractive target in arthritis or potentiallyother inflammatory diseases.

DisclosuresThe authors have no financial conflicts of interest.

References1. Kuroki, K., A. Furukawa, and K. Maenaka. 2012. Molecular recognition of

paired receptors in the immune system. Front. Microbiol. 3: 429.2. Lanier, L. L. 2001. Face off—the interplay between activating and inhibitory

immune receptors. Curr. Opin. Immunol. 13: 326–331.3. Taylor, L. S., S. P. Paul, and D. W. McVicar. 2000. Paired inhibitory and acti-

vating receptor signals. Rev. Immunogenet. 2: 204–219.4. Fournier, N., L. Chalus, I. Durand, E. Garcia, J. J. Pin, T. Churakova, S. Patel,

C. Zlot, D. Gorman, S. Zurawski, et al. 2000. FDF03, a novel inhibitory receptorof the immunoglobulin superfamily, is expressed by human dendritic and my-eloid cells. J. Immunol. 165: 1197–1209.

5. Mousseau, D. D., D. Banville, D. L’Abbe, P. Bouchard, and S. H. Shen. 2000.PILRalpha, a novel immunoreceptor tyrosine-based inhibitory motif-bearingprotein, recruits SHP-1 upon tyrosine phosphorylation and is paired with thetruncated counterpart PILRbeta. J. Biol. Chem. 275: 4467–4474.

6. Tabata, S., K. Kuroki, J. Wang, M. Kajikawa, I. Shiratori, D. Kohda, H. Arase,and K. Maenaka. 2008. Biophysical characterization of O-glycosylated CD99recognition by paired Ig-like type 2 receptors. J. Biol. Chem. 283: 8893–8901.

7. Wilson, M. D., J. Cheung, D. W. Martindale, S. W. Scherer, and B. F. Koop.2006. Comparative analysis of the paired immunoglobulin-like receptor (PILR)locus in six mammalian genomes: duplication, conversion, and the birth of newgenes. Physiol. Genomics 27: 201–218.

8. Shiratori, I., K. Ogasawara, T. Saito, L. L. Lanier, and H. Arase. 2004. Activationof natural killer cells and dendritic cells upon recognition of a novel CD99-likeligand by paired immunoglobulin-like type 2 receptor. J. Exp. Med. 199: 525–533.

9. Sun, Y., K. Senger, T. K. Baginski, A. Mazloom, Y. Chinn, H. Pantua,K. Hamidzadeh, S. R. Ramani, E. Luis, I. Tom, et al. 2012. Evolutionarilyconserved paired immunoglobulin-like receptor a (PILRa) domain mediates itsinteraction with diverse sialylated ligands. J. Biol. Chem. 287: 15837–15850.

10. Kogure, A., I. Shiratori, J. Wang, L. L. Lanier, and H. Arase. 2011. PANP isa novel O-glycosylated PILRa ligand expressed in neural tissues. Biochem.Biophys. Res. Commun. 405: 428–433.

11. Satoh, T., J. Arii, T. Suenaga, J. Wang, A. Kogure, J. Uehori, N. Arase,I. Shiratori, S. Tanaka, Y. Kawaguchi, et al. 2008. PILRalpha is a herpes simplexvirus-1 entry coreceptor that associates with glycoprotein B. Cell 132: 935–944.

12. Wang, J., Q. Fan, T. Satoh, J. Arii, L. L. Lanier, P. G. Spear, Y. Kawaguchi, andH. Arase. 2009. Binding of herpes simplex virus glycoprotein B (gB) to pairedimmunoglobulin-like type 2 receptor alpha depends on specific sialylatedO-linked glycans on gB. J. Virol. 83: 13042–13045.

13. Fan, Q., and R. Longnecker. 2010. The Ig-like v-type domain of paired Ig-liketype 2 receptor alpha is critical for herpes simplex virus type 1-mediatedmembrane fusion. J. Virol. 84: 8664–8672.

14. Banerjee, A., F. Stevenaert, K. Pande, E. Haghjoo, S. Antonenko, D. M. Gorman,M. Sathe, T. K. McClanahan, R. Pierce, S. P. Turner, et al. 2010. Modulation ofpaired immunoglobulin-like type 2 receptor signaling alters the host response toStaphylococcus aureus-induced pneumonia. Infect. Immun. 78: 1353–1363.

15. Tato, C. M., B. Joyce-Shaikh, A. Banerjee, Y. Chen, M. Sathe, S. E. Ewald,M. R. Liu, D. Gorman, T. K. McClanahan, J. H. Phillips, et al. 2012. The my-eloid receptor PILRb mediates the balance of inflammatory responses throughregulation of IL-27 production. PLoS ONE 7: e31680.

16. Wang, J., I. Shiratori, J. Uehori, M. Ikawa, and H. Arase. 2013. Neutrophil in-filtration during inflammation is regulated by PILRa via modulation of integrinactivation. Nat. Immunol. 14: 34–40.

17. Lee, D. M., and M. E. Weinblatt. 2001. Rheumatoid arthritis. Lancet 358: 903–911.18. Klareskog, L., A. I. Catrina, and S. Paget. 2009. Rheumatoid arthritis. Lancet

373: 659–672.

19. Gordon, S., and P. R. Taylor. 2005. Monocyte and macrophage heterogeneity.Nat. Rev. Immunol. 5: 953–964.

20. Nemeth, T., and A. Mocsai. 2012. The role of neutrophils in autoimmune dis-eases. Immunol. Lett. 143: 9–19.

21. Kagari, T., H. Doi, and T. Shimozato. 2002. The importance of IL-1 beta andTNF-alpha, and the noninvolvement of IL-6, in the development of monoclonalantibody-induced arthritis. J. Immunol. 169: 1459–1466.

22. Kagari, T., D. Tanaka, H. Doi, and T. Shimozato. 2003. Essential role of Fcgamma receptors in anti-type II collagen antibody-induced arthritis. J. Immunol.170: 4318–4324.

23. Tanaka, D., T. Kagari, H. Doi, and T. Shimozato. 2006. Essential role of neu-trophils in anti-type II collagen antibody and lipopolysaccharide-induced ar-thritis. Immunology 119: 195–202.

24. Banda, N. K., J. M. Thurman, D. Kraus, A. Wood, M. C. Carroll, W. P. Arend,and V. M. Holers. 2006. Alternative complement pathway activation is essentialfor inflammation and joint destruction in the passive transfer model of collagen-induced arthritis. J. Immunol. 177: 1904–1912.

25. Ditzel, H. J. 2004. The K/BxN mouse: a model of human inflammatory arthritis.Trends Mol. Med. 10: 40–45.

26. Barck, K. H., W. P. Lee, L. J. Diehl, J. Ross, P. Gribling, Y. Zhang, K. Nguyen,N. van Bruggen, S. Hurst, and R. A. Carano. 2004. Quantification of corticalbone loss and repair for therapeutic evaluation in collagen-induced arthritis, bymicro-computed tomography and automated image analysis. Arthritis Rheum.50: 3377–3386.

27. Lutz, M. B., N. Kukutsch, A. L. Ogilvie, S. Rossner, F. Koch, N. Romani, andG. Schuler. 1999. An advanced culture method for generating large quantities ofhighly pure dendritic cells from mouse bone marrow. J. Immunol. Methods 223:77–92.

28. Arnett, F. C., S. M. Edworthy, D. A. Bloch, D. J. McShane, J. F. Fries,N. S. Cooper, L. A. Healey, S. R. Kaplan, M. H. Liang, H. S. Luthra, et al. 1988.The American Rheumatism Association 1987 revised criteria for the classifi-cation of rheumatoid arthritis. Arthritis Rheum. 31: 315–324.

29. R Core Team. 2012. R: A Language and Environment for Statistical Computing.R Foundation for Statistical Computing. Vienna, Austria.

30. Gentleman, R. C., V. J. Carey, D. M. Bates, B. Bolstad, M. Dettling, S. Dudoit,B. Ellis, L. Gautier, Y. Ge, J. Gentry, et al. 2004. Bioconductor: open softwaredevelopment for computational biology and bioinformatics. Genome Biol. 5:R80.

31. Gautier, L., L. Cope, B. M. Bolstad, and R. A. Irizarry. 2004. affy—analysis ofAffymetrix GeneChip data at the probe level. Bioinformatics 20: 307–315.

32. Bolstad, B. M., R. A. Irizarry, M. Astrand, and T. P. Speed. 2003. A comparisonof normalization methods for high density oligonucleotide array data based onvariance and bias. Bioinformatics 19: 185–193.

33. Irizarry, R. A., B. Hobbs, F. Collin, Y. D. Beazer-Barclay, K. J. Antonellis,U. Scherf, and T. P. Speed. 2003. Exploration, normalization, and summaries ofhigh density oligonucleotide array probe level data. Biostatistics 4: 249–264.

34. Smyth, G. K. 2005. Limma: linear models for microarray data. Springer, NewYork.

35. Cogne, M., R. Lansford, A. Bottaro, J. Zhang, J. Gorman, F. Young, H. L. Cheng,and F. W. Alt. 1994. A class switch control region at the 39 end of the immu-noglobulin heavy chain locus. Cell 77: 737–747.

36. Wipke, B. T., Z. Wang, W. Nagengast, D. E. Reichert, and P. M. Allen. 2004.Staging the initiation of autoantibody-induced arthritis: a critical role for im-mune complexes. J. Immunol. 172: 7694–7702.

37. Bruhns, P., A. Samuelsson, J. W. Pollard, and J. V. Ravetch. 2003. Colony-stimulating factor-1-dependent macrophages are responsible for IVIG protec-tion in antibody-induced autoimmune disease. Immunity 18: 573–581.

38. Ji, H., K. Ohmura, U. Mahmood, D. M. Lee, F. M. Hofhuis, S. A. Boackle,K. Takahashi, V. M. Holers, M. Walport, C. Gerard, et al. 2002. Arthritis criti-cally dependent on innate immune system players. Immunity 16: 157–168.

39. Ji, H., A. Pettit, K. Ohmura, A. Ortiz-Lopez, V. Duchatelle, C. Degott,E. Gravallese, D. Mathis, and C. Benoist. 2002. Critical roles for interleukin 1and tumor necrosis factor alpha in antibody-induced arthritis. J. Exp. Med. 196:77–85.

40. Abbas, A. R., D. Baldwin, Y. Ma, W. Ouyang, A. Gurney, F. Martin, S. Fong,M. van Lookeren Campagne, P. Godowski, P. M. Williams, et al. 2005. Immuneresponse in silico (IRIS): immune-specific genes identified from a compendiumof microarray expression data. Genes Immun. 6: 319–331.

41. Satoh, T., and H. Arase. 2008. HSV-1 infection through inhibitory receptor,PILRalpha. Uirusu 58: 27–36.

42. Liu, W., L. Tang, G. Zhang, H. Wei, Y. Cui, L. Guo, Z. Gou, X. Chen, D. Jiang,Y. Zhu, et al. 2004. Characterization of a novel C-type lectin-like gene, LSECtin:demonstration of carbohydrate binding and expression in sinusoidal endothelialcells of liver and lymph node. J. Biol. Chem. 279: 18748–18758.

43. Crocker, P. R., J. C. Paulson, and A. Varki. 2007. Siglecs and their roles in theimmune system. Nat. Rev. Immunol. 7: 255–266.

44. Dominguez-Soto, A., L. Aragoneses-Fenoll, E. Martin-Gayo, L. Martinez-Prats,M. Colmenares, M. Naranjo-Gomez, F. E. Borras, P. Munoz, M. Zubiaur,M. L. Toribio, et al. 2007. The DC-SIGN-related lectin LSECtin mediates an-tigen capture and pathogen binding by human myeloid cells. Blood 109: 5337–5345.

45. Akkaya, M., and A. N. Barclay. 2013. How do pathogens drive the evolution ofpaired receptors? Eur. J. Immunol. 43: 303–313.

870 PILRa REGULATES INFLAMMATION

by guest on August 8, 2016

http://ww

w.jim

munol.org/

Dow

nloaded from


Recommended