12
Microenvironment and Immunology Genetic and Pharmacological Inactivation of the Purinergic P2RX7 Receptor Dampens Inammation but Increases Tumor Incidence in a Mouse Model of Colitis-Associated Cancer Paul Hofman 1,2,3,4 , Julien Cherls-Vicini 1,2,4 , Marie Bazin 1,2,4 , Marius Ilie 1,2,3,4 , Thierry Juhel 1,2,4 , Xavier H ebuterne 1,2,5 , Eric Gilson 1,2,4,6 , Annie Schmid-Alliana 2,7 , Olivier Boyer 8,9 , Sahil Adriouch 8,9 , and Val erie Vouret-Craviari 1,2,4 Abstract Colitis-associated cancer (CAC) is a complication of inamma- tory bowel disease (IBD). Binding of extracellular ATP to the purinergic receptor P2RX7 has emerged as a critical event in controlling intestinal inammation, acting to limit elevation of proinammatory mast cells and cytokines and promote survival of regulatory T cells (Treg) and enteric neurons. In this study, we investigated the effect of P2RX7 blockade in an established mouse model of CAC. Using genetic and pharmacologic tools, we found unexpectedly that while P2RX7 mediated inammatory responses, it also acted at an early time to suppress CAC development. P2RX7 blockade enhanced proliferation of intestinal epithelial cells and protected them from apoptosis. The proliferative effects of P2RX7 blockade were associated with an increased production of TGFb1 that was sufcient to stimulate the proliferation of intestinal epithelial cells. Finally, P2RX7 blockade also altered immune cell inltration and promoted Treg accumulation within lesions of the digestive system. Taken together, our ndings reveal an unexpected role for P2RX7 in preventing CAC, suggesting cautions in the use of P2RX7 inhibitors to treat IBD given the possibility of increasing risks CAC as a result. Cancer Res; 75(5); 83545. Ó2015 AACR. Introduction Purinergic receptors form a family of adenosine (P1) and ATP (P2) receptors involved in a complex signaling network that affects various cellular functions such as cell proliferation, cell differentiation and cell death, but also exocrine and endocrine secretion, immune responses, and inammation (1). They are widely expressed throughout the digestive tract, where they are involved in the regulation of intestinal motility. Moreover, it was shown that this family of receptors regulated several neuronal and non-neuronal gut functions, visceral sensation, and the immune cell activity (2). Therefore, it is now widely accepted that pur- inergic receptors actively cooperate in the maintenance of gut homeostasis. P2X receptors (P2RX) display a conserved topology with two transmembrane spanning regions, a large extracellular region sensitive to the ligand and intracellular amino- and carboxyl- termini that contain signaling binding motifs. Upon gating by eATP, P2RX7, also referred to as P2X7R, forms a poorly selective channel leading to membrane depolarization, potassium efux, and calcium and sodium inux (3). P2RX7 triggering also induces the formation of larger membrane pores that allow the passage of molecules reaching up to 900 Da. This particular feature has been linked to the presence of a long cytoplasmic C-terminal tail and to the possible activation of a pore-forming molecular partner suggested to be pannexin-1 (4, 5). Impor- tantly, both P2RX7 activation and pannexin-1 mediate NLRP3 inammasome assembly, caspase-1 (casp-1) activation, and maturation of the proinammatory cytokines IL1B (IL1b) and IL18 (6). At the level of the gastrointestinal tract, P2RX7 is expressed on immune and nonimmune cells (7, 8). Its low level of sensitivity to eATP limits its activation within inammatory sites and in the tumor microenvironment where eATP concen- trations are in the 100 mm range (9). We previously reported that P2RX7 is differentially expressed in the mucosa of patients with active and quiescent inammatory bowel disease (IBD) and that its activation could be triggered in response to neu- trophil transepithelial migration (8). These results, together with published data based on the use of P2RX7 antagonists or P2rx7 / mice, support the notion that P2RX7 participates in the initiation as well as in the regulation of the inammatory 1 Institute for Research on Cancer and Aging, Nice, France; IRCAN U1081 UMR CNRS 7284, Nice Cedex, France. 2 University of Nice- Sophia Antipolis, Nice, France. 3 Laboratory of Clinical and Experimen- tal Pathology and Biobank, Pasteur Hospital, Nice, France. 4 Centre Antoine Lacassagne, Nice, France. 5 Laboratory of Gastroenterology, Archet II Hospital, Nice, France. 6 Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, Nice, France. 7 iBV, UMR7277 CNRS- UMR1091 INSERM, Nice, France. 8 INSERM U905, Rouen, France. 9 Insti- tute for Research and Innovation in Biomedicine (IRIB), Normandy University, Rouen, France. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: Val erie Vouret-Craviari, IRCAN, Inserm U1081, UMR 7284 CNRS/Universit e Nice-Sophia Antipolis, 28 Avenue de Valombrose, Nice 06108, France. Phone: 33-492-031-223; Fax: 33-492-031-241; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-14-1778 Ó2015 American Association for Cancer Research. Cancer Research www.aacrjournals.org 835 on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

Microenvironment and Immunology

Genetic and Pharmacological Inactivation of thePurinergic P2RX7 Receptor DampensInflammation but Increases Tumor Incidence in aMouse Model of Colitis-Associated CancerPaul Hofman1,2,3,4, Julien Cherfils-Vicini1,2,4, Marie Bazin1,2,4, Marius Ilie1,2,3,4,Thierry Juhel1,2,4, Xavier H�ebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7,Olivier Boyer8,9, Sahil Adriouch8,9, and Val�erie Vouret-Craviari1,2,4

Abstract

Colitis-associated cancer (CAC) is a complication of inflamma-tory bowel disease (IBD). Binding of extracellular ATP to thepurinergic receptor P2RX7 has emerged as a critical event incontrolling intestinal inflammation, acting to limit elevation ofproinflammatorymast cells and cytokines andpromote survival ofregulatory T cells (Treg) and enteric neurons. In this study, weinvestigated the effect of P2RX7 blockade in an established mousemodel of CAC. Using genetic and pharmacologic tools, we foundunexpectedly thatwhile P2RX7mediated inflammatory responses,it also acted at an early time to suppress CACdevelopment. P2RX7

blockade enhanced proliferation of intestinal epithelial cells andprotected them from apoptosis. The proliferative effects of P2RX7blockade were associated with an increased production of TGFb1that was sufficient to stimulate the proliferation of intestinalepithelial cells. Finally, P2RX7 blockade also altered immune cellinfiltration and promoted Treg accumulation within lesions of thedigestive system. Taken together, ourfindings reveal anunexpectedrole for P2RX7 inpreventingCAC, suggesting cautions in the use ofP2RX7 inhibitors to treat IBD given the possibility of increasingrisks CAC as a result. Cancer Res; 75(5); 835–45. �2015 AACR.

IntroductionPurinergic receptors form a family of adenosine (P1) and ATP

(P2) receptors involved in a complex signaling network thataffects various cellular functions such as cell proliferation, celldifferentiation and cell death, but also exocrine and endocrinesecretion, immune responses, and inflammation (1). They arewidely expressed throughout the digestive tract, where they areinvolved in the regulation of intestinal motility. Moreover, it wasshown that this family of receptors regulated several neuronal andnon-neuronal gut functions, visceral sensation, and the immunecell activity (2). Therefore, it is now widely accepted that pur-

inergic receptors actively cooperate in the maintenance of guthomeostasis.

P2X receptors (P2RX) display a conserved topology with twotransmembrane spanning regions, a large extracellular regionsensitive to the ligand and intracellular amino- and carboxyl-termini that contain signaling binding motifs. Upon gating byeATP, P2RX7, also referred to as P2X7R, forms a poorly selectivechannel leading to membrane depolarization, potassiumefflux, and calcium and sodium influx (3). P2RX7 triggeringalso induces the formation of larger membrane pores that allowthe passage of molecules reaching up to 900 Da. This particularfeature has been linked to the presence of a long cytoplasmicC-terminal tail and to the possible activation of a pore-formingmolecular partner suggested to be pannexin-1 (4, 5). Impor-tantly, both P2RX7 activation and pannexin-1 mediate NLRP3inflammasome assembly, caspase-1 (casp-1) activation, andmaturation of the proinflammatory cytokines IL1B (IL1b) andIL18 (6).

At the level of the gastrointestinal tract, P2RX7 is expressedon immune and nonimmune cells (7, 8). Its low level ofsensitivity to eATP limits its activation within inflammatorysites and in the tumor microenvironment where eATP concen-trations are in the 100 mm range (9). We previously reportedthat P2RX7 is differentially expressed in the mucosa of patientswith active and quiescent inflammatory bowel disease (IBD)and that its activation could be triggered in response to neu-trophil transepithelial migration (8). These results, togetherwith published data based on the use of P2RX7 antagonists orP2rx7�/� mice, support the notion that P2RX7 participates inthe initiation as well as in the regulation of the inflammatory

1Institute for Research on Cancer and Aging, Nice, France; IRCANU1081 UMR CNRS 7284, Nice Cedex, France. 2University of Nice-SophiaAntipolis, Nice, France. 3Laboratoryof Clinical andExperimen-tal Pathology and Biobank, Pasteur Hospital, Nice, France. 4CentreAntoine Lacassagne, Nice, France. 5Laboratory of Gastroenterology,Archet II Hospital, Nice, France. 6Department of Medical Genetics,Archet 2 Hospital, CHU of Nice, Nice, France. 7iBV, UMR7277 CNRS-UMR1091 INSERM, Nice, France. 8INSERMU905, Rouen, France. 9Insti-tute for Research and Innovation in Biomedicine (IRIB), NormandyUniversity, Rouen, France.

Note: Supplementary data for this article are available at Cancer ResearchOnline (http://cancerres.aacrjournals.org/).

Corresponding Author: Val�erie Vouret-Craviari, IRCAN, Inserm U1081, UMR7284 CNRS/Universit�e Nice-Sophia Antipolis, 28 Avenue de Valombrose, Nice06108, France. Phone: 33-492-031-223; Fax: 33-492-031-241; E-mail:[email protected]

doi: 10.1158/0008-5472.CAN-14-1778

�2015 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 835

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 2: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

response notably via the processing and release of IL1B(10–12).

It is widely accepted that chronic inflammation can promotetumor development (13). In this scenario, the proinflammatoryrole of P2RX7 in immuneand epithelial cellswouldbe expected tofavor carcinogenesis. However, P2RX7 also regulates cell prolif-eration and/or cell death (14–17). Moreover, adding to thecomplex interplay between inflammation and carcinogenesis, astudy has revealed the role of the proinflammatory P2RX7/NLRP3/casp-1 cascade in the priming of dendritic cells and inthe immunogenicity of dying tumor cells following chemother-apy, which favors the emergence of an endogenous antitumorimmune response (18). Finally, P2RX7 appears to be overex-pressed in a variety of neoplastic disorders and tumors (19–21)and P2RX7 expression increases the tumorigenic properties ofhuman embryonic kidney cells (16). Hence, based on the appar-ently contradictory evidence, the precise role of P2RX7 in vivo inthe context of inflammation-associated carcinogenesis needs tobe carefully addressed.

In this study, we investigated the role of P2RX7 in CAC andwe showed, using P2rx7�/� mice as well as the P2RX7-specificpharmacologic antagonists, that the P2RX7 activity suppressedtumor development. While deciphering the cellular and molec-ular events leading to this suppressive function, we demon-strated that P2RX7 inactivation leads to: (i) overproduction ofTGFB1, (ii) recruitment of Tregs within inflammatory lesions,and (iii) TGFB1-mediated proliferation of colonic epithelialcells.

Materials and MethodsAnimal strains

This study was approved by the Institutional Care and UseCommittee of the University of Nice-Sophia Antipolis (Nice,France). Animal protocols were approved by the committee forResearch and Ethics of the PACA region (CIEPAL azur, #PEA 12-125) and followed the European directive 2010/63/UE. Thegeneration of C57BL/6 mice harboring targeted disruption of theP2rx7 gene has been described previously (10). Control C57BL/6Jmice were supplied by Harlan Laboratories.

Induction of colitis and inflammation-driven tumor formationAcute inflammation was induced as followed: intraperitoneal

injection of azoxymethane at day 0 before 5% dextran sulfate(DSS) treatment fromday 1 to day 5. CACwas induced by a singleintraperitoneal injection of azoxymethane followed by 3 cycles of3% DSS in the drinking water. Each cycle lasted 5 days and wasseparated by 2 weeks, as illustrated in Fig. 4A. We used thefollowing semiquantitative clinical score, which was adaptedfrom ref. 22. Body weight loss was scored as follows: 0, no loss;1, loss < 5%; 2, loss < 10%; 3, loss < 20%; and 4, loss > 20%. Thescores for stool consistency were measured as 0, normal; 1, loosestools; 2, watery diarrhea; or 3, severe watery diarrhea. Rectalbleeding was scored as 0, no blood; 1, presence of petechia; 2,stools with a trace of blood; or 3, bleeding. Inflammation scorewas performed by trained pathologists. Briefly, the severity ofinflammation (none, mild, moderate, severe), extent of inflam-mation (none, mucosa, mucosa, and submucosa, transmural),crypt damage (none, basal to 1/3, basal to 2/3, crypt loss, crypts,and epithelium loss), and percentage of tissue affected by inflam-mation (0, 25, 50, 75, and 100%) were scored.

Macroscopic polyp analysis and histopathologyColons (cecum to rectum) were removed from animals and

processed for histopathology as described in the SupplementaryMaterials and Methods.

Serum, colon organ cultures, and ELISABlood was collected with a heparin-treated needle from the tail

vein and serum was obtained by centrifugation (15 minutes,3,000 rpm at 4�C). After resection, colon tissues were culturedovernight inDMEM, supernatants were collected, and productionof IL1B, TGFB1, CXCL1, and CXCL2 assayed by an ELISA asdescribed by the manufacturers (see Supplementary Materialsand Methods).

Quantitative real-time PCR and protein gel blottingTotal RNA and protein were isolated from colonic tissues using

TRI Reagent following manufacturer's instructions and processedas described by manufacturers (see Supplementary Materials andMethods). We used the following antibodies: phospho-STAT3(#9131), STAT3 (#9139), and BCL2 (Bcl2, #50E3) antibodieswere from Cell Signaling Technology, anti-BCL2L1 (Bcl-XS/L, sc-634), and anti-BAX (Bax, sc-526) were from Santa Cruz Biotech-nology and the anti-ACTB (Actin, clone AC40) antibodywas fromSigma Aldrich.

Immunohistochemical analyses of mouse colon tissuesResected mouse colon tissues were fixed in 10% formalin,

paraffin-embedded, and processed for immunohistochemicalanalyses, as described previously (8) and SupplementaryMaterialsand Methods. We used the following antibodies: anti-PCNA (Epi-tomics), anti-cleaved-caspase-3 (Imgenex), anti-CD3 (Abcam),anti-Foxp3-biotin (eBioscience), anti-F4/80 (Abcam), and anti-Ly6G (Abcam).

Treatment with the P2RX7 antagonists and depletingantibodies

Competitive P2RX7 antagonists A438079 and A740003 anddepleting antibodies were given intraperitoneally during the DSStreatment as indicated in the Supplementary Materials andMethods.

Statistical analysisAll data are represented asmean values and error bars represent

SEM. The unpaired Mann–Whitney t test and ANOVA were usedto evaluate the statistical significance between groups.

ResultsGenetic and pharmacologic inactivation of P2RX7 dampenDSS-induced colonic inflammation

The inflammatory environment generated by DSS is associatedwith the release of eATP. We reasoned that P2RX7 inactivationshould impact the overall inflammatory response andwe used theacute DSS colitis model to test this hypothesis. After AOMinjection, WT and P2rx7�/� mice were fed a 5% DSS solutionduring 5 days and then clean drinking water for 4 or 9 days. Asexpected, when considering that P2RX7 triggered inflammasomeassembly and proinflammatory cytokine release, P2rx7�/� micewere less susceptible to this treatment (Fig. 1A). P2rx7�/� micesurvived better and lost less weight. Furthermore, they showedmilder signs of colitis with less diarrhea and less rectal bleeding.

Hofman et al.

Cancer Res; 75(5) March 1, 2015 Cancer Research836

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 3: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

We also observed a less dramatic shortening of the colon length atnecropsy. Such protection required themutation of the two allelessince heterozygous P2rx7þ/�mice showed the same phenotype asWTmice (Supplementary Fig. S1). Interestingly, these results werereproduced on WT mice treated with competitive P2RX7 antago-nists (Fig. 1B and Supplementary Fig. S2). Systemic blockade ofP2RX7 with A438079 or A740003 improved the survival of themice and reduced all the hallmarks of colitis. Indeed, 9 days afterDSS treatment, the loss of body weight and the disease activityscore decreased by around 25%, whereas the colon lengthincreased, as observed with P2rx7�/� mice.

Histologic analyses of colon tissues were performed to evaluatethe degree of inflammation. In the nontreated condition, bothWTand P2rx7�/� mice showed a normal colonic mucosa, with thecrypts being straight, well defined, and sitting on the muscularismucosa (Fig. 2A). At the end of the DSS regimen (day 5),histologic colonic transversal tissue sections of both WT andP2rx7�/� mice showed diffuse hemorrhagic walls with multipleulcerations, mucosal edema, transmural neutrophil infiltration,and the presence of large areas of erosion. Scoring for inflamma-tion, which demonstrated no difference between WT and KOmice, nearly reached the maximum (which was scored as 14). Incontrast, 4 days after the end of the DSS challenge (day 9),histologic mid colon tissue sections of WT mice still showed ahigh level of inflammation (12 � 0.7), whereas that of P2rx7�/�

mice showed signs of re-epithelialization, as indicated by better

delineated crypts and milder inflammation of the mucosa(inflammatory index of 5.1 � 1.5). Finally, most of the colonicmucosa of P2rx7�/� mice presented a normal aspect 9 days afterDSS treatment (day 14). In contrast, recovery of WT mice waspartial with a mean inflammatory index of 9 � 0.9 as comparedwith 2.2� 0.6 for KOmice. These differences were reproduced inWT mice treated with the P2RX7 antagonists (Fig. 2B and Sup-plementary Fig. S2). In particular, mice that received the compet-itive antagonist showed weak inflammatory infiltrates with nomucosal erosion and no epithelial cell defects within the crypts.Furthermore, histologic scoring of mid colon tissue sections ofantagonist-treated mice demonstrated a very low inflammatoryindex, which was statistically significant as compared with thecontrol group.

Taken together, these results demonstrate that both genetic andpharmacologic inactivation of P2RX7 dampen DSS-inducedcolonic inflammation. Notably, a time course analysis demon-strated that mice deficient in P2RX7 activity recovered morerapidly from inflammatory lesions than WT mice.

P2rx7�/� mice exhibit increased colonic epithelial cellproliferation and decreased apoptosis after AOM/DSStreatment

The prompt recovery of P2rx7�/� mice to acute AOM/DSSchallenge led us to evaluate whether P2RX7 regulated colonicmucosal epithelial cell proliferation. For that, colon tissue

Figure 1.Genetic and pharmacologic inactivation of P2RX7 decreases susceptibility to develop colitis. A, survival [P � 0.01, log-rank (Mantel–Cox) test], body weightloss, stool consistency, intestinal bleeding, and colon length from cohorts of 8–10 WT and P2rx7�/� mice subjected to colitis. Data are presented as means� SEM; � , P � 0.05; �� , P � 0.01. B, effect of selective P2RX7 antagonist A438079 on survival [P � 0.01, log-rank (Mantel–Cox) test], body weight loss, stoolconsistency, intestinal bleeding, and colon length. Data are presented as means � SEM; � , P � 0.05.

P2RX7 in Colitis and Colon Cancer

www.aacrjournals.org Cancer Res; 75(5) March 1, 2015 837

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 4: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

sections were stained for proliferating cell nuclear antigen(PCNA). In nontreated WT and P2rx7�/� animals, quantificationof PCNA-positive cells per 30 to50well-formed crypts showed5%to 10% PCNA-positive cells (Supplementary Fig. S3A). Five daysafter acute AOM/DSS treatment, no well-defined crypts werevisible (Fig. 2B). Therefore, we evaluated cell proliferation at days9 and 14. The proliferation indices of AOM/DSS-treated animalswere significantly higher in P2rx7�/� as compared with WT mice(Fig. 3A). Such an effect could directly depend on P2RX7expressed by colonic epithelial cells (8) and (Supplementary Fig.S3B), or alternatively, depends on infiltrating immune cells. Toaddress this point, we evaluated whether this effect could bereproduced in vitroon cultivated colonic epithelial cells expressingP2RX7. Indeed, blockade of P2RX7 enhanced the in vitro prolif-eration of the T84 colonic epithelial cell line (Supplementary Fig.S3C). The effect of P2RX7 blockade on epithelial cell proliferationwas further confirmed in vivo as A438079 administration toAOM/DSS-treated mice increased colonic epithelial cell proliferation(Supplementary Fig. S3D).Wenext evaluatedwhether P2RX7alsoregulates apoptosis of intestinal epithelial cells. For this, inflam-matory tissue lesions were stained with cleaved caspase-3 anti-body. We observed significantly less apoptotic cells in P2rx7�/�

mice at day 9 and their complete absence at day 14 after AOM/DSS

treatment (Fig. 3B). We then examined the expression of anti-apoptotic genes. No changes were observed in the expression ofBAX. In contrast, a significant increase in the expression of thegenes encoding BCL2 and BCL2L1 (Bcl-xL) was observed in thewhole colon of P2rx7�/� mice at day 14 after AOM/DSS admin-istration (Fig. 3C). Finally, the increase in epithelial cell prolifer-ation correlated with dysplasia in the colonic mucosa of P2rx7�/�

mice. We consistently found abnormal crypts characterized bymisalignment of the epithelial nuclei and a partial or total loss ofmucosal secretion (Fig. 3D). P2rx7�/� mice were particularlyprone to develop dysplasia, as they were also present in micethat received a short and low dose ofDSS (Supplementary Fig. S4)that induced no signs of colitis and only a very low inflammatoryindex. Taken together, these results demonstrate that the func-tional activity of the P2RX7 receptor is required to control the levelof intestinal epithelial cell proliferation and apoptosis in responseto an inflammatory insult and suggest that alteration in P2RX7functionality may favor possibly tumor progression.

P2RX7 mediates gastrointestinal tumorigenesis in the colitis-associated cancer model

To investigate the effect of P2RX7 on inflammation-inducedcolorectal cancer, azoxymethane-challenged mice received 3

Figure 2.Mice deficient in the P2RX7 activity recovered more rapidly from inflammatory lesions than WT mice. A, WT and P2rx7�/� were submitted to colitis. Colonswere removed either at the end of the DSS treatment (day 5), 4 days after (day 9), or 9 days after (day 14) treatment. Tissue sections were stained with hematoxylinand eosin and analyzed microscopically (scale bar, 100 mm). Histologic scoring of mid colon tissue sections was calculated as indicated in the SupplementaryMaterials and Methods. Data are presented as means � SEM; � , P � 0.05. B, nontreated and A438079-treated WT mice were submitted to colitis. Colontissue sections were removed and stained with hematoxylin and eosin either at the end of the DSS treatment or 4 days after (day 9) treatment. Histologic scoring ofmid colon tissue sections is shown. Data are presented as means � SEM; �� , P � 0.01.

Hofman et al.

Cancer Res; 75(5) March 1, 2015 Cancer Research838

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 5: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

cycles of DSS to mimic chronic colitis (Fig. 4A). Both WT andP2rx7�/� mice survived azoxymethane-DSS treatment andshowed comparable disease activity indices during the first 10weeks of treatment. However, P2rx7�/� mice displayed a higherdisease activity scores at the end of the protocol (SupplementaryFig. S5). Unexpectedly, despite their lower susceptibility to acuteDSS-induced inflammation, P2rx7�/� mice developed signifi-cantly higher numbers of macroscopic polyps (mean of 9 polypsper mouse) as compared with WT mice (mean of 2 polyps permouse). As expected for the CAC model, tumors were located atthe distal end of the colon inWTmice, whereas they invaded up tothe medial part of the colon in P2rx7�/� mice (Fig. 4A). Inaddition, the 4-fold increase in tumor burden in P2rx7�/� micewas accompanied by a 2-fold increase in polyp size (Fig. 4B).Histologic examination of colons from WT and P2RX7-deficientmice showed that 80% of the KOmice developed colonic lesionsranging fromhyperplasia and dysplasia to adenocarcinoma (Sup-plementary Fig. S6A). Adenocarcinoma was observed in morethan 60% of KO mice, whereas only 25% of the WT cohortdisplayed a carcinoma.

Furthermore, we investigated the pharmacologic effect ofP2RX7 blockade in the CAC model using the A438079-compet-itive antagonist. Changes in bodyweightwere followedduring theentire protocol and the colonic tumor burden was monitored 10weeks after AOM injection. As observed for WT and P2rx7�/�

mice, antagonist-treated and nontreated mice displayed compa-rable body weight variations during the course of the treatment(Supplementary Fig. S5, bottom). However, A438079-treatedmice harbored higher numbers of macroscopic polyps andtumors were significantly larger in size than in control mice (Fig.4C). Collectively, these results demonstrate that alteration of theP2RX7 functionality enhanced tumor promotion and/or progres-sion. Interestingly, tumor progression was also found to becontrolled by P2RX7 in an unrelated tumor mouse model basedon subcutaneous injection of Lewis Lung Carcinoma (LLC) cells.As shown in Supplementary Fig. S6B, mice injected with LLC cellsdeveloped tumors within 2 weeks. Importantly, excised tumorsfrom P2rx7�/� mice were bigger than tumors from WT miceconfirming that P2RX7 behaved as a tumor suppressor in thistumorigenic mouse model.

Figure 3.P2RX7-dependent signaling prevented early excessive colonic epithelial cell proliferation following AOM/DSS treatment. A, cohorts of 8–10 WT and P2rx7�/� micewere treated for colitis and epithelial cell proliferation was evaluated by PCNA staining performed on colon tissue 4 and 9 days after DSS treatment (day 9and day 14, respectively). Representative results are shown on the panels on the left and the number of proliferating cells (PCNA positive, arrows) per cryptwas determined at day 9 and 14 (right). Arrows, crypts containing epithelial cells with PCNA nuclear staining. A higher magnification is shown in the inset. B,detection of the cleaved form of caspase-3 (arrows). Representative pictures are shown on the left. Quantification of the number of apoptotic cells at day 9 and 14 isshown on the right. A higher magnification is shown in the insets. C, immunoblot analysis of BCL2, BCL2L1 (Bcl-XL), and BAX expression in the colon tissues ofWT andP2rx7�/� mice, treated for colitis, were performed at day 14. The total actin (ACTB) level was monitored as a control for equal protein loading. D, quantificationof the number of low dysplasia in animals. Data, shown as arbitrary unit, are representative of two independent experiments (mean� SEM). � , P� 0.05; �� , P� 0.01.

P2RX7 in Colitis and Colon Cancer

www.aacrjournals.org Cancer Res; 75(5) March 1, 2015 839

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 6: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

P2RX7 shapes the inflammatory microenvironmentP2RX7 stimulation is known to trigger inflammasome assem-

bly and casp1 activation and to favor the release of the processedmature form of IL1B, thereby initiating the inflammatoryresponse (4, 10). As the early level of inflammation can influencethe quality and the quantity of recruited immune cells, we nextcharacterized the inflammatory microenvironment before tumoronset.We first checked the capacity of colonic explants from acuteDSS-treatedmice to secrete IL1B. After in vivoDSS treatment, IL1Bwas readily produced ex vivo in colon tissue cultures derived fromWT mice (Fig. 5A). In contrast, and not surprisingly, very littleIL1B was produced by colonic explants from DSS-treatedP2rx7�/� mice. In agreement with their lowered inflammatoryresponse, we also observed significantly lower activation of STAT3in P2RX7-deficient mice at day 14 after DSS treatment as com-pared with WT control mice (Fig. 5B).

We next evaluated how the lower P2RX7-dependent IL1Bproduction influenced the composition of the inflammatoryinfiltrate in the colon of DSS-treated mice. For that, colon tissuesections from acute DSS-treated mice were stained for markersspecific for T lymphocytes (CD3), Treg lymphocytes [FOXP3(Foxp3)], macrophages [EMR1 (F4/80)], and neutrophils(LY6G). Colonic-lamina propria tissue sections from P2rx7�/�

mice displayed statistically more CD3þ (9% vs. 3%), FOXP3þ

(15% vs. 3%), and EMR1þ (F4/80) (16% vs. 5%) cells as com-pared with WT animals (Fig. 5C). In contrast, no differencebetween the two groups of mice was observed in regard to

LY6G-positive cells. However, we noticed that colon tissue sec-tions from both groups harbored a significantly high number ofneutrophils (reaching 8% to 10% of total immune cells). Thisobservation is particularly relevant to the field of IBD pathogen-esis. Indeed, acute inflammatory lesions of patients with IBD arecharacterized by a large influx of neutrophils within the intestinalmucosa (23). Keeping in mind the recent concept of the role ofneutrophils in tumor biology (24), we wondered whether theP2RX7 activity participated in the polarization of tumor-associ-ated neutrophils (TAN). To assess this question, we analyzed theexpression of genes encoding chemokines that have been used todistinguish TAN from other tumor-associated myeloid cells (25).As illustrated in Fig. 5D, the P2RX7 deficiency resulted inincreased expression of the Ccl17 transcript and decreased expres-sion of Cxcl10, a molecular profile that has been associated withgenuine TAN (24, 25).

Enhanced production of TGFB1 in the colonic mucosa ofAOM/DSS-treated P2rx7�/� mice

TGFB1 is frequently associated with poor prognosis in patientswith cancer (26). This immunoregulatory cytokine can be pro-duced by immune as well as tumor cells and has been implicatedin the polarization of various immune cell subsets but also in theestablishment of a suppressive microenvironment that favorstumor growth (27). The accumulation of immunosuppressivecells within the inflammatory lesions of mutant mice could

Figure 4.The P2RX7 activity impeded tumorigenesis in the colitis-associated cancer model. A, schematic representation of CAC. B, CAC-treated WT and P2rx7�/� micewere euthanized and colons were resected and opened longitudinally to visualize angiogenic (arrows) and nonangiogenic polyps (star). Macroscopic polypswere then counted, sized, and colon tissue sections were fixed in formalin and stained with hematoxylin/eosin for histologic examination. C, the CAC protocol wasapplied to cohorts of ten nontreated and A438079-treated mice. Colons were resected and macroscopic polyps were counted and sized. Data are presentedas means � SEM; � , P � 0.05; �� , P � 0.01.

Hofman et al.

Cancer Res; 75(5) March 1, 2015 Cancer Research840

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 7: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

possibly be associated with the local production of TGFB1.Indeed, a 3-fold induction of TGFB1 production was observedin acute inflammatory of colon explant culture from P2rx7�/�

versus WT mice (Fig. 6A). These results were confirmed at latertime points, as P2rx7�/� mice subjected to the chronic AOM/DSStreatment displayed an overexpression of Tgfb1 and Tgfb2 tran-scripts, and their cultured explanted colonic tissue showedincreased secretion of the TGFB1 cytokine (Fig. 6B).

As TGFB1 is known to promote the suppressive activity ofFOXP3þ Tregs, we further characterized the composition of thecellular infiltrate in colon tissue biopsies from chronic AOM/DSS-treated mice. As described before for the inflammatory colonicmucosa, a significant increase in the content of FOXP3þ cells wasobserved within tumors derived from P2rx7�/� mice (Fig. 6C),whereas there was no difference in the numbers of macrophages(EMR1, F4/80þ cells) nor total T lymphocytes (CD3þ cells). AsTGFB1 has been proposed to control differentiation of TAN inmice (24), we next investigated the presence of neutrophil infil-tration in their colonic mucosa. We found an enrichment in theproportion of LY6Gþ cells in P2rx7�/� mice (Fig. 6C), in agree-ment with the higher concentration of CXCL1 and CXCL2, twochemokines involved in neutrophil recruitment, in the serum ofthese animals (Supplementary Fig. S7A). Interestingly, neutrophilenrichment appeared to be restricted to colonic tumor tissues asanalysis of bone marrow and spleen cells did not show anydifference in the composition of immune cells (SupplementaryFig. S7B). Moreover, the cytokine profile observed in tumor

lesions of P2rx7�/�mice is compatible with the molecular profileassociated with genuine TAN (Supplementary Fig. S7C; ref. 25).

In addition to its immunosuppressive function, TGFB1 hasbeen reported to increase proliferation of colonic epithelial cells(28). Therefore, overproduction of TGFB1 in inflammatory andtumor lesions of P2rx7�/� mice may account for the increasedproliferation of epithelial cells observed in this strain (Fig. 3). Totest this hypothesis, neutralizing anti-TGFB antibody was injectedin AOM/DSS-treated P2rx7�/� mice. As shown in Fig. 7A, bodyweight and colon length were not significantly different in micetreated with isotype control or with the anti-TGFB antibodies. Incontrast, the number of colonic epithelial cells positive for PCNAstaining was reduced by 2-fold in mice that received the neutral-izing anti-TGFB antibody (Fig. 7B). Indeed, while 35% � 4% ofintestinal epithelial cells were PCNAþ in mice treated with theisotype control antibody, only 15%� 2%of cells were detected tobe positive in mice treated with the neutralizing antibody, a levelcomparable to what was observed in WT mice (see Fig. 3).

Taken together, these results suggest that enhanced secretion ofTGFB1 in P2RX7�/� favors the establishment of an immunosup-pressive microenvironment and stimulates epithelial cell prolif-eration, thereby enhancing the development of colon cancer.

DiscussionP2RX7 is an ATP gated ion channel that can induce multiple

cellular responses. In the early 1990s, studies demonstrated cell-permeabilizing and cytolytic effects of P2RX7 in response to high

Figure 5.P2RX7 shapes the inflammatorymicroenvironment. A, cohorts of 8–10WT and P2rx7�/� mice were treatedfor colitis. Expression levels of Il1bmRNA and of the correspondingsecreted protein (IL1B) weredetermined by RT-qPCR and ELISA. B,proteins were extracted from the midto distal colon tissue sections of DSS-treated WT and P2rx7�/� mice. Theactivation of STAT3 was analyzed byimmunoblotting using an anti-phospho-STAT3 antibody. The totalactin (ACTB) level was monitored as acontrol for equal protein loading. Dataare presented as means � SEM;� , P � 0.05. C, immunohistochemistrywas performed on day 9 after DSStreatment. The quality of infiltratingcells was evaluated using antibodiesdirected against characteristic cellmarkers: anti-CD3 (T cells), anti-FOXP3 (Foxp3, Tregs), EMR1 (F4/80,macrophages), and anti-LY6G(Ly6G, neutrophils). Results arepresented as means � SEM;� , P � 0.05; �� , P � 0.01. D, theexpression of Ccl17 and Cxcl10(IP-10) was examined by RT-qPCR.Data are presented as means� SEM; � , P � 0.05.

P2RX7 in Colitis and Colon Cancer

www.aacrjournals.org Cancer Res; 75(5) March 1, 2015 841

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 8: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

doses of eATP (29, 30). A few years later, it appeared thatstimulation of P2RX7 by low doses of eATP enhanced prolifer-ation of human primary cells (31). This observation was furtherconfirmed in vitro using heterologous expression systems (32) aswell as in vivo using xenograft tumor models (16). P2RX7appeared to be overexpressed in a variety of malignant tumors(reviewed in ref. 33).However, P2RX7-dependent signaling couldalso favor an antitumor effect in vivo. Indeed, P2RX7 participatesin inflammasome assembly and may thereby alert the immunesystem through processing and liberation of the proinflammatoryIL1B cytokine (6). It has been demonstrated that P2RX7 plays acrucial role in the immunogenicity of dying tumor cells inresponse to chemotherapy (a process named immunogenic celldeath) through the activation of dendritic cells and the conse-quent promotion of an adaptive antitumor immune responsethat prevents tumor relapse (18). Of particular interest, in spo-radic breast cancer treated with anthracyclines, patients bearingloss-of-function alleles of P2RX7 (rs3751143, NM_002562.4:

c.1487A>C) are more prone to develop metastases than patientswithout mutation. Such results fuel the hypothesis that P2RX7plays an important role in the antitumor response.

In this study, we conducted experiments in the context of acuteand chronic colitis with the aim of examining the global in vivocontribution of P2XR7 signaling to inflammation and cancerprogression. Using a mouse model of acute AOM/DSS-inducedcolonic inflammation, we found that P2rx7�/� mice displayedreduced signs of inflammation. This finding is in agreement withrecent studies demonstrating that prophylactic systemic blockadeof the P2RX7 activity prevented TNBS-induced colitis in rats (12)and that overexpression of P2RX7 in the intestinal mucosa wasassociated with the pathogenesis of CD (34). In the current study,we found that P2RX7 inactivation using selective antagonists aswell as genetic deletion of the P2rx7 gene attenuated the weightloss and the overall disease activity score (Fig. 1). However,whereas the histologic inflammatory score ofDSS-treated animalswas similar in bothWT and P2rx7�/�mice at day 5 (i.e., at the end

Figure 6.Increased expression of TGFB1 in colontumor biopsies from P2rx7�/�mice. A,cohorts of 8–10WT and P2rx7�/�micewere treated for colitis. Secretion ofTGFB1 was assayed by ELISA onsupernatants from organocultures.Data are expressed as mean� SEM; � , P� 0.05. B, Tgfb1 and Tgfb2transcripts were analyzed by RT-qPCRand the levels of secreted TGFB1 byELISA from CAC-treated cohorts of8–10 WT and P2rx7�/� mice. Data areexpressed asmean� SEM; � , P� 0.05.B, the number of CD3-, FOXP3(Foxp3)-, EMR1 (F4/80)-, and LY6G(Ly6G)-positive cells was determined.Arrows, positive cells. Data arepresented as means � SEM;� , P � 0.05 (scale bar, 100 mm).

Hofman et al.

Cancer Res; 75(5) March 1, 2015 Cancer Research842

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 9: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

of DSS treatment), we paradoxically observed a faster re-epithe-lialization of the digestive mucosa of P2rx7�/� mice during therecovery phase (Fig. 2).

Considering that the faster recovery of P2rx7�/�mice correlatedwith enhanced epithelial cell proliferation, reduced apoptosis,and higher expression of BCL2L1 (Bcl-xL) and BCL2 survivalfactors (Fig. 3), our results support the notion that P2RX7 playsa role in the control of epithelial cell proliferation and/or apo-ptosis in response to injury. We indeed confirmed in vivo inP2rx7�/� mice as well as in WT mice treated with P2RX7 phar-macologic inhibitors, that P2RX7 inactivation increases epithelialcells proliferative index (Fig. 3 and Supplementary Fig. S3D).Concordantly, we confirmed in vitro using the human T84 epi-thelial cell line derived from a colon cancer that pharmacologicinhibition of P2RX7 significantly stimulates cell proliferation(Supplementary Fig. S3C). This results support the notion thatP2RX7 activity directly inhibit epithelial cell proliferation.Wheth-er immune cells, that we found to be recruited at higher level inP2RX7-inactivated mice (i.e, FOXP3þ Tregs and LY6Gþ neutro-

phils) also contribute to the control of epithelial cell proliferationneed to be further address in future studies.

Our finding, which demonstrated that a lack of P2RX7 is linkedto increased epithelial cell proliferation, may explain, at leastpartly, the increased tumor burden observed in chronic AOM/DSS–treated P2rx7�/� mice (Fig. 4). Another nonexclusive mech-anistic explanation may reside downstream of IL1B signaling. Asexpected, little if any IL1B was detected from the colonic mucosaof P2rx7�/� mice at early time points following AOM/DSS treat-ment (Fig. 5). This was associated with reduced STAT3 phosphor-ylation, whichmay indicate lower activation of immune cells andmay favor the establishment of an immunosuppressive microen-vironment prone to tumor development. However, downregula-tion of IL1B and STAT3 phosphorylation could not represent thesolemechanismas it has been reported that genetic inactivation ofIL1BR or of STAT3 in the intestinal epithelial cell compartment(DIECStat3 mice) decreased tumor development (35, 36).

In addition,we found that FOXP3þ Tregs, a cell population thathas been associated with colon cancer progression (37), was

Figure 7.Treatment with anti-TGFB antibodyinhibited colon epithelial cellproliferation in AOM/DSS–treatedP2rx7�/� mice. Cohorts of 6 P2rx7�/�

mice were injected with depletingantibodies and treated for colitis. A,body weight loss and colon lengthwere analyzed as described in thelegend of Fig. 1. Data are presented asmeans � SEM. Results are notstatistically different. B, epithelial cellproliferation was evaluated by PCNA-staining performed on colon tissuesections 9 days after DSS treatment.Representative results are shown onthe panels on the left. The number ofproliferating cells is shownon the rightpanel. A highermagnification is shownon the insets. Data are presented asmeans � SEM. �� , P � 0.01.

P2RX7 in Colitis and Colon Cancer

www.aacrjournals.org Cancer Res; 75(5) March 1, 2015 843

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 10: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

increased in the colonic mucosa of acute AOM/DSS-treatedP2rx7�/� mice (Fig. 5C) in association with lower STAT3 phos-phorylation (Fig. 5B) and higher TGFB1 production (Fig. 6).Interestingly, we previously demonstrated that P2RX7 activationat the surface of Tregs inhibited their suppressive function andinduced cell death (38, 39). Therefore, inactivation of P2RX7should protect Tregs from the deleterious effect of endogenousP2RX7 ligands (i.e., eATP and NAD) and should promote theirearly accumulation in the damaged colonic mucosa, as observedin the current study (Fig. 5C). In addition, local production ofTGFBmay further enhance their suppressive activity andmay evenparticipate in the conversion of conventional CD4þ T cells toinduced Tregs (iTregs). Of note, several reports have linked TGFB1to negative regulation of tumor-specific cytotoxic T lymphocyteresponses (40–44). Therefore, upregulation of TGFB productionin the colonic mucosa of AOM/DSS-treated P2rx7�/� mice mayalso inhibit tumor-specific cytotoxic T cells activity and therebyfurther participates in tumor promotion. In addition to its immu-nosuppressive role, TGFB has been reported to enhance prolifer-ation of epithelial cells (28). Concordantly, we show here thattreatment of DSS-treated P2rx7�/� mice with an anti-TGFB anti-body significantly reduced the proliferation of colonic epithelialcells (Fig. 7B).

During the course of this study, we also observed the recruit-ment of LY6Gþ neutrophils within tumor lesions of P2rx7�/�

mice. These cells shared the characteristics of genuine TAN (Sup-plementary Fig. S7), a population of cell that has been recentlyassociated to tumor progression (24). Therefore, we cannotexclude that TAN also participate in the reinforcement of theimmunosuppressive environment in the colonic mucosa ofAOM/DSS-treated P2rx7�/�mice and in the stimulation of tumorgrowth.

We demonstrate in our study that P2RX7 inactivation dampensinflammation but paradoxically enhances tumor susceptibility.Interestingly, we reproduced this finding in an independenttumor mouse model consisting of subcutaneous injection of LLCcells into C57BL/6 mice (Supplementary Fig. S6B). Several poly-morphisms have been described for the P2RX7 gene and theirimpact on the P2RX7 function, the mechanisms and relationshipwith diseases were characterized for a number of mutants (45–47). Surprisingly, none of these variants were found in a meta-analysis of IBD (CD and UC) that included a Genome-WideAssociation study (48). However, this analysis highlighted theimportance of TGFB signaling in IBD and reinforced the link withcolorectal cancer. Furthermore, P2RX7 was reported to be overexpressed in chronic B lymphocytic leukemia (19), prostate cancer

and adjacent tissue (21), papillary thyroid carcinoma (49) andgastric, colon, kidney, and ovarian cancers (16). However, despitethe large number of P2RX7 mutants and of P2RX7 variants, noattempts were made to characterize their functionality in tumorlesions, highlighting the need for additional studies to provide abetter understanding of the role of P2RX7 in cancer onset.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: P. Hofman, X. H�ebuterne, S. Adriouch, V. Vouret-CraviariDevelopment of methodology: P. Hofman, T. Juhel, E. Gilson, V. Vouret-CraviariAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): J. Cherfils-Vicini, M. Bazin, O. BoyerAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): P. Hofman, J. Cherfils-Vicini, M. Bazin, M. Ilie,V. Vouret-CraviariAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): M. Bazin, E. GilsonWriting, review, and/or revision of the manuscript: P. Hofman, J. Cherfils-Vicini, M. Ilie, X. H�ebuterne, O. Boyer, S. Adriouch, V. Vouret-CraviariAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): A. Schmid-Allilana, V. Vouret-CraviariStudy supervision: P. Hofman, V. Vouret-Craviari

AcknowledgmentsThe authors thank Salom�e Lalvee and Oliver Bordone (LPCE, Nice, France)

for their expert technical assistance and Ludovic Cervera (Cytomed platform,funded by the CG06, INSERM and FEDER) for his expertise in flow cytometry.The authors thank Sandrine Marchetti for helpful discussion. The authorsacknowledge the IRCAN's PICMI, CYTOMED, and Animal core facilities.

Grant SupportThis workwas supported by grants from "InstitutNational de la Sant�e et de la

RechercheM�edicale," the "Associationpour la Recherche contre leCancer" (ARCGrants SL220110603478), the Canc�eropole PACA, the "Centre National de laRecherche Scientifique," the "Ligue Nationale contre le Cancer (E. Gibson:Equipe lab�elis�ee), and the FrenchGovernment (National ResearchAgency, ANRthrough the "Investments for the Future" LABEX SIGNALIFE: program reference#ANR-11-LABX-0028-01).

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received June 13, 2014; revised November 5, 2014; accepted November 26,2014; published OnlineFirst January 6, 2015.

References1. Burnstock G, Williams M. P2 purinergic receptors: modulation of cell

function and therapeutic potential. J Pharmacol Exp Ther 2000;295:862–9.

2. Antonioli L, Colucci R, Pellegrini C, Giustarini G, Tuccori M, Blandizzi C,et al. The role of purinergic pathways in the pathophysiology of gutdiseases: pharmacological modulation and potential therapeutic applica-tions. Pharmacol Ther 2013;139:157–88.

3. North RA. Molecular physiology of P2X receptors. Physiol Rev2002;82:1013–67.

4. Pelegrin P, Surprenant A. Pannexin-1 mediates large pore formation andinterleukin-1beta release by the ATP-gated P2 � 7 receptor. EMBO J2006;25:5071–82.

5. Locovei S, Scemes E,Qiu F, SprayDC,Dahl G. Pannexin1 is part of the poreforming unit of the P2X(7) receptor death complex. FEBS Lett 2007;581:483–8.

6. Ferrari D, Pizzirani C, Adinolfi E, Lemoli RM, Curti A, IdzkoM, et al. The P2� 7 receptor: a key player in IL-1 processing and release. J Immunol2006;176:3877–83.

7. Trautmann A. Extracellular ATP in the immune system: more than just a"danger signal." Sci Signal 2009;2:pe6.

8. Cesaro A, Brest P, Hofman V, Hebuterne X, Wildman S, Ferrua B, et al.Amplification loop of the inflammatory process is induced by P2 � 7Ractivation in intestinal epithelial cells in response to neutrophil transepithe-lial migration. Am J Physiol Gastrointest Liver Physiol 2010;299:G32–42.

Cancer Res; 75(5) March 1, 2015 Cancer Research844

Hofman et al.

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 11: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

9. Pellegatti P, Raffaghello L, Bianchi G, Piccardi F, Pistoia V, Di Virgilio F.Increased level of extracellular ATP at tumor sites: in vivo imaging withplasma membrane luciferase. PLoS ONE 2008;3:e2599.

10. Solle M, Labasi J, Perregaux DG, Stam E, Petrushova N, Koller BH, et al.Altered cytokine production inmice lacking P2X(7) receptors. J Biol Chem2001;276:125–32.

11. Labasi JM, PetrushovaN, Donovan C,McCurdy S, Lira P, PayetteMM, et al.Absence of the P2� 7 receptor alters leukocyte function and attenuates aninflammatory response. J Immunol 2002;168:6436–45.

12. Marques CC, Castelo-Branco MT, Pacheco RG, Buongusto F, do Rosario AJr, Schanaider A, et al. Prophylactic systemic P2 � 7 receptor blockadeprevents experimental colitis. Biochim Biophys Acta 2014;1842:65–78.

13. de Visser KE, Eichten A, Coussens LM. Paradoxical roles of the immunesystem during cancer development. Nat Rev Cancer 2006;6:24–37.

14. Mackenzie AB, Young MT, Adinolfi E, Surprenant A. Pseudoapoptosisinduced by brief activation of ATP-gated P2 � 7 receptors. J Biol Chem2005;280:33968–76.

15. Adinolfi E, Cirillo M,Woltersdorf R, Falzoni S, Chiozzi P, Pellegatti P, et al.Trophic activity of a naturally occurring truncated isoform of the P2 � 7receptor. FASEB J 2010;24:3393–404.

16. Adinolfi E, Raffaghello L, Giuliani AL, Cavazzini L, Capece M, Chiozzi P,et al. Expression of P2� 7 receptor increases in vivo tumor growth. CancerRes 2012;72:2957–69.

17. Wiley JS, Dao-Ung LP, Gu BJ, Sluyter R, Shemon AN, Li C, et al. A loss-of-function polymorphic mutation in the cytolytic P2 � 7 receptor gene andchronic lymphocytic leukaemia: a molecular study. Lancet 2002;359:1114–9.

18. Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, et al.Activation of theNLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med 2009;15:1170–8.

19. Adinolfi E,Melchiorri L, Falzoni S, Chiozzi P,Morelli A, Tieghi A, et al. P2�7 receptor expression in evolutive and indolent forms of chronicB lymphocytic leukemia. Blood 2002;99:706–8.

20. Greig AV, Linge C, Healy V, Lim P, Clayton E, Rustin MH, et al. Expressionof purinergic receptors in non-melanoma skin cancers and their functionalroles in A431 cells. J Invest Dermatol 2003;121:315–27.

21. Slater M, Danieletto S, Gidley-Baird A, Teh LC, Barden JA. Early prostatecancer detected using expression of non-functional cytolytic P2 � 7receptors. Histopathology 2004;44:206–15.

22. Carvalho FA, BarnichN, SivignonA,DarchaC,ChanCH, StannersCP, et al.Crohn's disease adherent-invasive Escherichia coli colonize and inducestrong gut inflammation in transgenic mice expressing human CEACAM.J Exp Med 2009;206:2179–89.

23. Chin AC, Parkos CA. Neutrophil transepithelial migration and epithelialbarrier function in IBD: potential targets for inhibiting neutrophil traffick-ing. Ann N Y Acad Sci 2006;1072:276–87.

24. Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, et al. Polarizationof tumor-associated neutrophil phenotype by TGF-beta: "N1" versus "N2"TAN. Cancer Cell 2009;16:183–94.

25. Fridlender ZG, Sun J, Mishalian I, Singhal S, Cheng G, Kapoor V, et al.Transcriptomic analysis comparing tumor-associated neutrophils withgranulocytic myeloid-derived suppressor cells and normal neutrophils.PLoS ONE 2012;7:e31524.

26. Massague J. TGFbeta in cancer. Cell 2008;134:215–30.27. Flavell RA, Sanjabi S, Wrzesinski SH, Licona-Limon P. The polarization of

immune cells in the tumour environment by TGFbeta. Nat Rev Immunol2010;10:554–67.

28. Yan Z, Kim GY, Deng X, Friedman E. Transforming growth factor beta 1induces proliferation in colon carcinoma cells by Ras-dependent,smad-independent down-regulation of p21cip1. J Biol Chem 2002;277:9870–9.

29. Filippini A, Taffs RE, Agui T, SitkovskyMV. Ecto-ATPase activity in cytolyticT-lymphocytes. Protection from the cytolytic effects of extracellular ATP.J Biol Chem 1990;265:334–40.

30. Di Virgilio F, Bronte V, Collavo D, Zanovello P. Responses of mouselymphocytes to extracellular adenosine 50-triphosphate (ATP). Lympho-cytes with cytotoxic activity are resistant to the permeabilizing effects ofATP. J Immunol 1989;143:1955–60.

31. Baricordi OR, Ferrari D,Melchiorri L, Chiozzi P, Hanau S, Chiari E, et al. AnATP-activated channel is involved in mitogenic stimulation of humanT lymphocytes. Blood 1996;87:682–90.

32. Adinolfi E, Callegari MG, Ferrari D, Bolognesi C, Minelli M, WieckowskiMR, et al. Basal activation of the P2 � 7 ATP receptor elevates mitochon-drial calcium and potential, increases cellular ATP levels, and promotesserum-independent growth. Mol Biol Cell 2005;16:3260–72.

33. Di Virgilio F, Ferrari D, Adinolfi E. P2X(7): a growth-promoting receptor-implications for cancer. Purinergic Signal 2009;5:251–6.

34. Neves AR, Castelo-Branco MT, Figliuolo VR, Bernardazzi C, Buongusto F,Yoshimoto A, et al.Overexpression of ATP-activated P2� 7 receptors in theintestinal mucosa is implicated in the pathogenesis of Crohn's disease.Inflamm Bowel Dis 2014;20:444–57.

35. Salcedo R, Worschech A, Cardone M, Jones Y, Gyulai Z, Dai RM, et al.MyD88-mediated signaling prevents development of adenocarcinomas ofthe colon: role of interleukin 18. J Exp Med 2010;207:1625–36.

36. Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabhapurapu S, et al.IL-6 and Stat3 are required for survival of intestinal epithelial cells anddevelopment of colitis-associated cancer. Cancer Cell 2009;15:103–13.

37. Chaput N, Louafi S, Bardier A, Charlotte F, Vaillant JC, Menegaux F, et al.Identification of CD8þCD25þFoxp3þ suppressive T cells in colorectalcancer tissue. Gut 2009;58:520–9.

38. Hubert S, Rissiek B, Klages K, Huehn J, Sparwasser T, Haag F, et al.Extracellular NADþ shapes the Foxp3þ regulatory T cell compartmentthrough the ART2-P2 � 7 pathway. J Exp Med 2010;207:2561–8.

39. Adriouch S, Haag F, Boyer O, Seman M, Koch-Nolte F. Extracellular NAD(þ): a danger signal hindering regulatory T cells. Microbes Infect 2012;14:1284–92.

40. Shull MM, Ormsby I, Kier AB, Pawlowski S, Diebold RJ, Yin M, et al.Targeted disruption of the mouse transforming growth factor-beta 1 generesults in multifocal inflammatory disease. Nature 1992;359:693–9.

41. Kulkarni AB, Huh CG, Becker D, Geiser A, Lyght M, Flanders KC, et al.Transforming growth factor beta 1 null mutation in mice causes excessiveinflammatory response and early death. Proc Natl Acad Sci U S A 1993;90:770–4.

42. Thomas DA,Massague J. TGF-beta directly targets cytotoxic T cell functionsduring tumor evasion of immune surveillance. Cancer Cell 2005;8:369–80.

43. Li MO, Wan YY, Flavell RA. T cell-produced transforming growth factor-beta1 controls T cell tolerance and regulates Th1- and Th17-cell differen-tiation. Immunity 2007;26:579–91.

44. Donkor MK, Sarkar A, Li MO. Tgf-beta1 produced by activated CD4(þ) TCells Antagonizes T Cell Surveillance of Tumor Development. Oncoim-munology 2012;1:162–71.

45. Jiang LH, Baldwin JM, Roger S, Baldwin SA. Insights into the molecularmechanisms underlying mammalian P2 � 7 receptor functions and con-tributions in diseases, revealed by structural modeling and single nucle-otide polymorphisms. Front Pharmacol 2013;4:55.

46. Miller CM, Boulter NR, Fuller SJ, Zakrzewski AM, Lees MP, Saunders BM,et al. The role of the P2X(7) receptor in infectious diseases. PLoS Pathog2011;7:e1002212.

47. Fuller SJ, Stokes L, Skarratt KK, Gu BJ, Wiley JS. Genetics of the P2 � 7receptor and human disease. Purinergic Signal 2009;5:257–62.

48. Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al.Host-microbe interactions have shaped the genetic architecture of inflam-matory bowel disease. Nature 2012;491:119–24.

49. Solini A, Cuccato S, Ferrari D, Santini E, Gulinelli S, Callegari MG, et al.Increased P2 � 7 receptor expression and function in thyroid papillarycancer: a new potential marker of the disease? Endocrinology 2008;149:389–96.

www.aacrjournals.org Cancer Res; 75(5) March 1, 2015 845

P2RX7 in Colitis and Colon Cancer

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778

Page 12: Genetic and Pharmacological Inactivation of the Purinergic ... · Thierry Juhel1,2,4, Xavier Hebuterne1,2,5, Eric Gilson1,2,4,6, Annie Schmid-Alliana2,7, Olivier Boyer8,9, Sahil Adriouch8,9,

2015;75:835-845. Published OnlineFirst January 6, 2015.Cancer Res   Paul Hofman, Julien Cherfils-Vicini, Marie Bazin, et al.   a Mouse Model of Colitis-Associated CancerReceptor Dampens Inflammation but Increases Tumor Incidence in Genetic and Pharmacological Inactivation of the Purinergic P2RX7

  Updated version

  10.1158/0008-5472.CAN-14-1778doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2015/01/06/0008-5472.CAN-14-1778.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/75/5/835.full#ref-list-1

This article cites 49 articles, 19 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/75/5/835.full#related-urls

This article has been cited by 3 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/75/5/835To request permission to re-use all or part of this article, use this link

on November 17, 2020. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 6, 2015; DOI: 10.1158/0008-5472.CAN-14-1778