Infection Protective Immunity to Intestinal Helminth Regulatory ...

3 downloads 0 Views 877KB Size Report
shown in murine models of autoimmune hepatitis, auto- immune colitis ..... Erin C. Boyle for critical reading of the manuscript, and Andreas Hahn for assistance ...
This information is current as of September 13, 2017.

Cutting Edge: The BTLA−HVEM Regulatory Pathway Interferes with Protective Immunity to Intestinal Helminth Infection Minka Breloer, Wiebke Hartmann, Birte Blankenhaus, Marie-Luise Eschbach, Klaus Pfeffer and Thomas Jacobs

Supplementary Material References Subscription Permissions Email Alerts

http://www.jimmunol.org/content/suppl/2015/01/16/jimmunol.140251 0.DCSupplemental This article cites 26 articles, 11 of which you can access for free at: http://www.jimmunol.org/content/194/4/1413.full#ref-list-1 Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2015 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606.

Downloaded from http://www.jimmunol.org/ by guest on September 13, 2017

J Immunol 2015; 194:1413-1416; Prepublished online 16 January 2015; doi: 10.4049/jimmunol.1402510 http://www.jimmunol.org/content/194/4/1413

Cutting Edge

The

Journal of

Immunology

Cutting Edge: The BTLA–HVEM Regulatory Pathway Interferes with Protective Immunity to Intestinal Helminth Infection Minka Breloer,* Wiebke Hartmann,* Birte Blankenhaus,* Marie-Luise Eschbach,* Klaus Pfeffer,† and Thomas Jacobs*

T

he mammalian immune system is a complex network of activating and inhibiting regulatory circuits that allow the rapid induction of immune responses to pathogens while facilitating homeostasis in the absence of infection. B and T lymphocyte attenuator (BTLA; CD272) is an Ig domain superfamily protein that delivers inhibitory signals via an intracellular ITIM upon engagement by its ligand, herpes virus entry mediator (HVEM) (1–3). Although BTLA is constitutively expressed on B cells, it is predominantly Foxp32 effector T cells that upregulate BTLA upon activation (4). Maintenance of homeostasis by BTLA-mediated negative coregulation was shown in murine models of autoimmune hepatitis, autoimmune colitis, and graft-versus-host disease (1). The BTLA ligand HVEM is a member of the TNFR superfamily and is

*Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; and † University Hospital D€usseldorf, 40225 D€ usseldorf, Germany Received for publication October 6, 2014. Accepted for publication December 12, 2014. M.B. is supported by Deutsche Forschungsgemeinschaft Grant 3754/2-1. T.J. is supported by Sonderforschungsbereich 841, a special funding tool for large collaborative research by the Deutsche Forschungsgemeinschaft. Address correspondence and reprint requests to Dr. Minka Breloer, Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Strasse 74, 20359 Hamburg, Germany. E-mail address: [email protected] www.jimmunol.org/cgi/doi/10.4049/jimmunol.1402510

expressed on a broad range of hematopoietic and nonhematopoietic cells. While delivering negative signals into BTLAexpressing cells, HVEM engagement leads to costimulatory signaling into the HVEM-expressing cells via NF-kB induction (5). Moreover, HVEM can bind to three ligands in addition to BTLA: one coinhibitory receptor (CD160) and two costimulatory receptors (lymphotoxin-like, exhibits inducible expression, competes with HSV gpD for HVEM, a receptor expressed by T lymphocytes [LIGHT] and lymphotoxin a) (6). Thus, the BTLA–HVEM-mediated network of costimulation and coinhibition is a classic example of the regulatory circuits that enable the immune system to rapidly respond to infections without losing homeostatic control (1, 5, 7). The role of BTLA–HVEM-mediated immune regulation during helminth infection has not been investigated. Helminths are large multicellular parasites that are exposed to the immune system of their host. To survive and to avoid the induction of immune pathology, helminths use existing regulatory elements of the immune system, such as regulatory surface receptors, to suppress the immune response that is directed toward them (8). We used experimental infection of mice with the parasitic nematode Strongyloides ratti to investigate helminthinduced immune modulation. Thereby, S. ratti serves as a model for transient gastrointestinal nematode infections. Infective third-stage larvae (iL3) dwell in the environment and actively penetrate the skin of their host (9). Larvae migrate within 2 d to the head, are swallowed, and reach the small intestine. They molt via a fourth larval stage to become parasitic adults that live embedded in the intestinal mucosa. Five days postinfection (p.i.), adults start to reproduce, and eggs and hatched first-stage larvae leave the host with the feces. Immune-competent mice terminate infection within 30 d and remain semiresistant to subsequent infections (10). To our knowledge, we report for the first time that BTLA–HVEM-mediated regulation dampens the protective The online version of this article contains supplemental material. Abbreviations used in this article: BTLA, B and T lymphocyte attenuator; HVEM, herpes virus entry mediator; iL3, infective third-stage larvae; LIGHT, lymphotoxinlike, exhibits inducible expression, competes with HSV gpD for HVEM, a receptor expressed by T lymphocytes; mcpt-1, mouse mast cell protease-1; mLN, mesenteric lymph node; p.i., postinfection; Treg, regulatory T cell. Copyright Ó 2015 by The American Association of Immunologists, Inc. 0022-1767/15/$25.00

Downloaded from http://www.jimmunol.org/ by guest on September 13, 2017

Helminths exploit intrinsic regulatory pathways of the mammalian immune system to dampen the immune response directed against them. In this article, we show that infection with the parasitic nematode Strongyloides ratti induced upregulation of the coinhibitory receptor B and T lymphocyte attenuator (BTLA) predominantly on CD4+ T cells but also on a small fraction of innate leukocytes. Deficiency of either BTLA or its ligand herpes virus entry mediator (HVEM) resulted in reduced numbers of parasitic adults in the small intestine and reduced larval output throughout infection. Reduced parasite burden in BTLA- and HVEM-deficient mice was accompanied by accelerated degranulation of mucosal mast cells and increased Ag-specific production of the mast cell–activating cytokine IL-9. Our combined results support a model whereby BTLA on CD4+ T cells and additional innate leukocytes is triggered by HVEM and delivers negative signals into BTLA+ cells, thereby interfering with the protective immune response to this intestinal parasite. The Journal of Immunology, 2015, 194: 1413–1416.

1414

CUTTING EDGE: BTLA REGULATES IMMUNITY TO INTESTINAL NEMATODE INFECTION Statistical analysis Statistical analysis was performed with GraphPad Prism software (La Jolla, CA) using two-way ANOVA for repeated experiments. Group sizes in experiments were always $3. The data are presented as mean 6 SEM.

Results and Discussion

immune response to S. ratti infection and promotes survival of parasitic adults in the small intestine. Mice lacking either BTLA or its ligand HVEM displayed decreased intestinal parasite burden in the context of increased IL-9 production and accelerated mast cell degranulation.

Materials and Methods S. ratti life cycle and infections Animal experimentation was conducted at the animal facility of the Bernhard Nocht Institute for Tropical Medicine in agreement with the German animal protection law under the supervision of a veterinarian. BTLA2/2 mice (2), a kind gift of Dr. Kenneth Murphy (Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO), BTLA2/2RAG12/2 mice, and HVEM2/2 mice (11) were cohoused with the C57BL/6 and RAG12/2 control groups for $4 wk prior to infection. For selected experiments (Fig. 1C), heterozygous BTLA2/2 and BTLA+/2 or BTLA+/+ littermates were generated by backcrossing BTLA2/2 mice to C57BL/6 mice. Mice were infected by s.c. injection of 1000 S. ratti iL3 in the footpad, and parasite burden in tissue and intestine, as well as larval output, was quantified as described (10, 12). For IL-9 blockade, mice received 100 mg anti– IL-9 mAb (MM9C1; Bio X Cell, West Lebanon, NH) or isotype control i.p. at days 0 and 3 of S. ratti infection.

Cytokine production and mast cell activation Mice were sacrificed at day 6 p.i., mesenteric lymph node (mLN) cells (5 3 105/well) were stimulated with S. ratti Ag or anti-CD3 (145-2C11), and cytokines in the culture supernatants were measured as described (13). The supernatants of stimulated mLN cell cultures derived from naive mice did not contain detectable concentrations of IL-9 or IL-13. Mouse mast cell protease-1 (mcpt-1) concentration in serum samples was detected using the MCTP-1 ELISA Ready-SET-Go! kit (eBioscience, San Diego, CA), according to the manufacturer’s recommendations.

FIGURE 2. Reduced intestinal parasite burden and larval output throughout infection in HVEM2/2 mice. Wild-type (wt) and HVEM2/2 mice were infected with S. ratti by s.c. injection of 1000 iL3 after 4 wk of cohousing. Migrating larvae in the lung (A) and head (B) were counted at day 2 p.i. (C) Parasitic adults in the small intestine were counted at day 6 p.i. Shown are the combined results of two (A and B) (n = 16) or five (C) (n $ 20) independent experiments. Each symbol represents one mouse, and horizontal lines represent the means. (D) Release of S. ratti DNA in the feces over 24 h was quantified at the indicated time points. Shown are the combined results of two independent experiments (n = 6–12 per group and time point). Error bars represent SEM. **p # 0.01, ***p # 0.001.

Downloaded from http://www.jimmunol.org/ by guest on September 13, 2017

FIGURE 1. Reduced intestinal parasite burden and larval output throughout infection in BTLA2/2 mice. BTLA+/+ or BTLA+/2 wild-type mice (wt) and BTLA2/2 littermates were infected with S. ratti by s.c. injection of 1000 iL3. Migrating larvae in the lung (A) and the head (B) were counted at day 2 p.i. (C) Parasitic adults in the small intestine were counted at day 6 p.i. Shown are the combined results of two (A and B) (n = 10) or 4 (C) (n $ 15) independent experiments. Each symbol represents one mouse, and horizontal lines represent the means. (D) Release of S. ratti DNA in the feces over 24 h was quantified at the indicated time points. Shown are the combined results of two independent experiments (n = 6–12 per group and time point). Error bars represent SEM. **p # 0.01, ***p # 0.001.

Monitoring the expression of regulatory receptors during murine S. ratti infection, we observed increased BTLA expression specifically in the mLNs, but not on leukocytes, in blood, or in the peritoneum, of S. ratti–infected mice (Supplemental Fig. 1). Within the mLNs, BTLA expression was induced predominantly on CD4+ T lymphocytes and to a lesser extent on CD8+ T lymphocytes. Constitutive BTLA expression on B cells was not modulated during infection. BTLA also was induced on a small fraction of CD192 TCR2 cells in the mLNs of S. ratti–infected mice that displayed variable CD11b, CD11c, and Gr-1 expression and were CD49b (DX5) and F4/80 negative. To evaluate a possible biological function for BTLAmediated signaling during S. ratti infection, we compared the parasite burden in BTLA2/2 and BTLA-expressing mice. Although the numbers of tissue-migrating larvae in the lung and head were similar (Fig. 1A, 1B), BTLA2/2 mice displayed significantly reduced numbers of parasitic adults in the small intestine at day 6 p.i. compared with BTLA-expressing wild-type mice (Fig. 1C). Reduced parasite burden in BTLA2/2 mice was reflected by the reduced release of S. ratti– derived DNA in the feces as an indicator of larval output throughout infection until clearance by day 29 p.i. (Fig. 1D).

The Journal of Immunology

HVEM is the only ligand described for BTLA (3). Compared with wild-type mice, HVEM2/2 mice displayed increased numbers of tissue-migrating larvae in lung and head (Fig. 2A, 2B). Interestingly, and despite the initially increased larval burden in the tissue, numbers of parasitic adults in the small intestine were reduced in HVEM2/2 mice (Fig. 2C). Subsequent larval output in the feces also was drastically reduced in the absence of HVEM at all time points examined p.i. (Fig. 2D).

Taken together, our results show no impact of BTLAmediated regulation during the control of tissue-migrating larvae. HVEM apparently promoted eradication of migrating larvae during the first 2 d of infection independently of BTLA in the wild-type state. HVEM, as a signaling receptor itself, may have responded to engagement by alternative HVEM ligands, such as LIGHT, CD160, or lymphotoxin a (5, 7). With regard to intestinal immunity to S. ratti, our results clearly show that BTLA and its ligand HVEM interfere with efficient eradication of parasites from the small intestine of infected mice. Control of Strongyloides infection in the intestine was shown to depend on a functional Th2 response (14, 15) and activated mast cells (16). To analyze the immunological mechanism that reduced parasite burden in BTLA2/2 and HVEM2/2 mice, we quantified the cytokine response during S. ratti infection (Fig. 3A). Although IL-13 and IL-10 production by mLN cells was comparable in all mouse strains, the absence of either BTLA or its ligand HVEM increased IL-9 production in response to both CD3 engagement and S. ratti–specific stimulation (Fig. 3A and data not shown). IL-9 is a cytokine with pleiotropic functions (17) that promotes expulsion of intestinal parasites (18, 19). We demonstrated recently that neutralization of IL-9 increased parasite burden and reduced mast cell degranulation in S. ratti–infected BALB/c and C57BL/6 mice (13). To quantify mast cell degranulation during infection, we measured the serum concentration of mcpt-1 that is specifically released by mucosal mast cells (20). Compared with wild-type mice, both BTLA and HVEM deficiency were accompanied by increased early degranulation of mucosal mast cells at 2 and 3 d post– S. ratti infection (Fig. 3B). Furthermore, in vivo neutralization of IL-9 partially abrogated the improved host defense observed in BTLA2/2 and HVEM2/2 mice (Fig. 3C). Collectively, our data suggest that accelerated IL-9–driven mast cell activation contributed to improved expulsion of S. ratti from the intestine of BTLA2/2 and HVEM2/2 mice. Our results agree with previous studies showing that negative coregulation via BTLA dampens the adaptive immune response during murine Plasmodium infection. Decreased parasitemia and increased cytokine production by BTLA2/2 CD4+ T cells were observed in a model for protective immunity in the absence of BTLA-mediated regulation (21). Conversely, an increase in negative signaling into BTLA-expressing cells via

FIGURE 4. Intestinal parasite burden in BTLA2/2RAG12/2 mice. C57BL/6, BTLA2/2, RAG12/2, and BTLA2/2RAG12/2 mice were infected with S. ratti by s.c. injection of 1000 iL3. Parasitic adults in the small intestine were counted at day 6 p.i. Shown are the combined results of three independent experiments (n $ 12, each symbol represents one mouse, horizontal lines represent the means). *p # 0.05, ***p # 0.001.

Downloaded from http://www.jimmunol.org/ by guest on September 13, 2017

FIGURE 3. Increased IL-9 secretion and accelerated mast cell degranulation in S. ratti–infected BTLA2/2 and HVEM2/2 mice. Wild-type, BTLA2/2, and HVEM2/2 mice were cohoused for $4 wk and infected with S. ratti by s.c. injection of 1000 iL3. (A) Mice were sacrificed at day 6 p.i., and mLN cells were cultured with S. ratti Ag or anti-CD3 for 72 h. IL-13 and IL-9 in the supernatants were quantified by ELISA. Graphs shows the mean of one experiment (n = 4) and are representative of one independent repeat. (B) Serum samples were acquired at the indicated time points, and mcpt-1 was quantified by ELISA. Graph shows the mean of combined results from two independent experiments (n = 7). Error bars represent SEM. (C) BTLA2/2 and HVEM2/2 mice were treated with isotype-control or neutralizing mAb to IL-9. Parasites in the small intestine were counted on day 6 p.i. and compared with isotypetreated wild-type mice. Graph shows combined results from two independent experiments (n $ 8). *p # 0.05, **p # 0.01, ***p # 0.001.

1415

1416

CUTTING EDGE: BTLA REGULATES IMMUNITY TO INTESTINAL NEMATODE INFECTION

Acknowledgments

We thank Dr. Kenneth Murphy for providing the BTLA2/2 mice, Erin C. Boyle for critical reading of the manuscript, and Andreas Hahn for assistance with statistics.

Disclosures The authors have no financial conflicts of interest.

References 1. Murphy, T. L., and K. M. Murphy. 2010. Slow down and survive: Enigmatic immunoregulation by BTLA and HVEM. Annu. Rev. Immunol. 28: 389–411. 2. Watanabe, N., M. Gavrieli, J. R. Sedy, J. Yang, F. Fallarino, S. K. Loftin, M. A. Hurchla, N. Zimmerman, J. Sim, X. Zang, et al. 2003. BTLA is a lymphocyte inhibitory receptor with similarities to CTLA-4 and PD-1. Nat. Immunol. 4: 670–679. 3. Sedy, J. R., M. Gavrieli, K. G. Potter, M. A. Hurchla, R. C. Lindsley, K. Hildner, S. Scheu, K. Pfeffer, C. F. Ware, T. L. Murphy, and K. M. Murphy. 2005. B and T lymphocyte attenuator regulates T cell activation through interaction with herpesvirus entry mediator. Nat. Immunol. 6: 90–98. 4. Hurchla, M. A., J. R. Sedy, M. Gavrieli, C. G. Drake, T. L. Murphy, and K. M. Murphy. 2005. B and T lymphocyte attenuator exhibits structural and expression polymorphisms and is highly Induced in anergic CD4+ T cells. J. Immunol. 174: 3377–3385. 5. Steinberg, M. W., T. C. Cheung, and C. F. Ware. 2011. The signaling networks of the herpesvirus entry mediator (TNFRSF14) in immune regulation. Immunol. Rev. 244: 169–187. 6. Cai, G., and G. J. Freeman. 2009. The CD160, BTLA, LIGHT/HVEM pathway: a bidirectional switch regulating T-cell activation. Immunol. Rev. 229: 244–258. 7. Shui, J. W., and M. Kronenberg. 2014. HVEM is a TNF Receptor with Multiple Regulatory Roles in the Mucosal Immune System. Immune Netw. 14: 67–72. 8. McSorley, H. J., and R. M. Maizels. 2012. Helminth infections and host immune regulation. Clin. Microbiol. Rev. 25: 585–608. 9. Viney, M.E. and Lok J.B. Strongyloides spp. (May 23, 2007), WormBook, ed. The C. elegans Research Community, WormBook, doi/10.1895/wormbook.1.141.1, http://www.wormbook.org. 10. Eschbach, M. L., U. Klemm, J. Kolbaum, B. Blankenhaus, N. Brattig, and M. Breloer. 2010. Strongyloides ratti infection induces transient nematode-specific Th2 response and reciprocal suppression of IFN-gamma production in mice. Parasite Immunol. 32: 370–383. 11. Wang, Y., S. K. Subudhi, R. A. Anders, J. Lo, Y. Sun, S. Blink, Y. Wang, J. Wang, X. Liu, K. Mink, et al. 2005. The role of herpesvirus entry mediator as a negative regulator of T cell-mediated responses. J. Clin. Invest. 115: 711–717. 12. Nouir, N. B., M. L. Eschbach, M. Pie´davent, A. Osterloh, M. T. Kingsley, K. Erttmann, N. Brattig, E. Liebau, B. Fleischer, and M. Breloer. 2012. Vaccination with Strongyloides ratti heat shock protein 60 increases susceptibility to challenge infection by induction of Th1 response. Vaccine 30: 862–871. 13. Blankenhaus, B., M. Reitz, Y. Brenz, M. L. Eschbach, W. Hartmann, I. Haben, T. Sparwasser, J. Huehn, A. K€uhl, T. B. Feyerabend, et al. 2014. Foxp3+ regulatory T cells delay expulsion of intestinal nematodes by suppression of IL-9-driven mast cell activation in BALB/c but not in C57BL/6 mice. PLoS Pathog. 10: e1003913. 14. Machado, E. R., D. Carlos, E. V. Lourenc¸o, C. A. Sorgi, E. V. Silva, S. G. Ramos, M. T. Ueta, D. M. Aronoff, and L. H. Faccioli. 2009. Counterregulation of Th2 immunity by interleukin 12 reduces host defenses against Strongyloides venezuelensis infection. Microbes Infect. 11: 571–578. 15. Yoshimoto, T., T. Yoshimoto, K. Yasuda, J. Mizuguchi, and K. Nakanishi. 2007. IL-27 suppresses Th2 cell development and Th2 cytokines production from polarized Th2 cells: a novel therapeutic way for Th2-mediated allergic inflammation. J. Immunol. 179: 4415–4423. 16. Lantz, C. S., J. Boesiger, C. H. Song, N. Mach, T. Kobayashi, R. C. Mulligan, Y. Nawa, G. Dranoff, and S. J. Galli. 1998. Role for interleukin-3 in mast-cell and basophil development and in immunity to parasites. Nature 392: 90–93. 17. Noelle, R. J., and E. C. Nowak. 2010. Cellular sources and immune functions of interleukin-9. Nat. Rev. Immunol. 10: 683–687. 18. Licona-Limo´n, P., J. Henao-Mejia, A. U. Temann, N. Gagliani, I. Licona-Limo´n, H. Ishigame, L. Hao, D. R. Herbert, and R. A. Flavell. 2013. Th9 Cells Drive Host Immunity against Gastrointestinal Worm Infection. Immunity 39: 744–757. 19. Richard, M., R. K. Grencis, N. E. Humphreys, J. C. Renauld, and J. Van Snick. 2000. Anti-IL-9 vaccination prevents worm expulsion and blood eosinophilia in Trichuris muris-infected mice. Proc. Natl. Acad. Sci. USA 97: 767–772. 20. Reynolds, D. S., R. L. Stevens, W. S. Lane, M. H. Carr, K. F. Austen, and W. E. Serafin. 1990. Different mouse mast cell populations express various combinations of at least six distinct mast cell serine proteases. Proc. Natl. Acad. Sci. USA 87: 3230–3234. 21. Adler, G., C. Steeg, K. Pfeffer, T. L. Murphy, K. M. Murphy, J. Langhorne, and T. Jacobs. 2011. B and T lymphocyte attenuator restricts the protective immune response against experimental malaria. J. Immunol. 187: 5310–5319. 22. Lepenies, B., K. Pfeffer, M. A. Hurchla, T. L. Murphy, K. M. Murphy, J. Oetzel, B. Fleischer, and T. Jacobs. 2007. Ligation of B and T lymphocyte attenuator prevents the genesis of experimental cerebral malaria. J. Immunol. 179: 4093–4100. 23. Steinberg, M. W., O. Turovskaya, R. B. Shaikh, G. Kim, D. F. McCole, K. Pfeffer, K. M. Murphy, C. F. Ware, and M. Kronenberg. 2008. A crucial role for HVEM and BTLA in preventing intestinal inflammation. J. Exp. Med. 205: 1463–1476. 24. Schaer, C., S. Hiltbrunner, B. Ernst, C. Mueller, M. Kurrer, M. Kopf, and N. L. Harris. 2011. HVEM signalling promotes colitis. PLoS ONE 6: e18495. 25. Sun, Y., N. K. Brown, M. J. Ruddy, M. L. Miller, Y. Lee, Y. Wang, K. M. Murphy, K. Pfeffer, L. Chen, J. Kaye, and Y. X. Fu. 2009. B and T lymphocyte attenuator tempers early infection immunity. J. Immunol. 183: 1946–1951. 26. Kobayashi, Y., A. Iwata, K. Suzuki, A. Suto, S. Kawashima, Y. Saito, T. Owada, M. Kobayashi, N. Watanabe, and H. Nakajima. 2013. B and T lymphocyte attenuator inhibits LPS-induced endotoxic shock by suppressing Toll-like receptor 4 signaling in innate immune cells. Proc. Natl. Acad. Sci. USA 110: 5121–5126. 27. Shubin, N. J., C. S. Chung, D. S. Heffernan, L. R. Irwin, S. F. Monaghan, and A. Ayala. 2012. BTLA expression contributes to septic morbidity and mortality by inducing innate inflammatory cell dysfunction. J. Leukoc. Biol. 92: 593–603.

Downloaded from http://www.jimmunol.org/ by guest on September 13, 2017

BTLA ligation prevented the genesis of cerebral malaria in a model of immune pathology (22). The BTLA–HVEM pathway also plays a central role in the regulation of T cell– mediated intestinal inflammation in the absence of infection. HVEM expressed by intestinal epithelial cells ameliorated T cell transfer colitis by engagement of BTLA on the adoptively transferred CD4+ T cells (23). HVEM expression on T cells apparently promoted colitis, because HVEM deficiency in transferred T cells resulted in less severe colitis (24), thus emphasizing the dual nature of HVEM-BTLA–mediated regulation (7). Although negative regulation of T and B cells and NKT cells by BTLA is well established (1), accumulating evidence suggests that BTLA may regulate innate immune cells in Plasmodium (21) or Listeria infection (25) or during experimental sepsis (26, 27). We observed BTLA upregulation on CD11b+ and CD11c+ but not on F4/801 non-B non-T cells (Supplemental Fig. 1). To elucidate the contribution of these BTLA+ innate leukocytes to the regulation of acute nematode infection, we analyzed the impact of BTLA deficiency in the absence of adaptive immunity using RAG12/2 and BTLA2/2RAG12/2 mice. Although RAG12/2 mice are unable to terminate infection (data not shown), initial control of parasite burden can be maintained independently of B and T cells. Thus, RAG12/2 and wild-type mice displayed comparable numbers of parasitic adults in the small intestine at day 6 p.i. (Fig. 4). BTLA deficiency in the presence of B and T cells strongly reduced parasite burden. In the absence of adaptive immunity (i.e., comparing RAG12/2 mice and BTLA2/2RAG12/2 mice), the advantage of BTLA deficiency was still significant but less pronounced. Our combined results could be explained by a model whereby BTLA, predominantly on CD4+ T cells, is triggered by HVEM and delivers negative coregulation into BTLA+ cells, thus interfering with the protective immune response to the intestinal parasite. BTLA expression on innate leukocytes may contribute to this negative coregulation. Although immune-competent wild-type mice clear S. ratti infection within 30 d, even this short survival of parasites in their host critically depends on immune evasion. We showed previously that Foxp3+ regulatory T cells (Tregs) promote survival of S. ratti in the small intestine of BALB/c mice by dampening IL-9–driven mast cell activation (13). Because Foxp3+ Tregs did not play a central role in the survival of S. ratti, within the even more susceptible C57BL/6 mice, we hypothesized that redundant alternative regulatory pathways contributed to immune evasion on this genetic background. In the current study we provide evidence that, in addition to Tregs, the BTLA–HVEM network represents one additional layer of regulation that can be exploited by intestinal parasites to delay immune-mediated expulsion.