This information is current as of March 25, 2014.

References Subscriptions Permissions Email Alerts

Jason M. Schenkel, Kathryn A. Fraser and David Masopust J Immunol 2014; 192:2961-2964; Prepublished online 5 March 2014; doi: 10.4049/jimmunol.1400003 http://www.jimmunol.org/content/192/7/2961

This article cites 21 articles, 6 of which you can access for free at: http://www.jimmunol.org/content/192/7/2961.full#ref-list-1 Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscriptions Submit copyright permission requests at: http://www.aai.org/ji/copyright.html Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/cgi/alerts/etoc

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 9650 Rockville Pike, Bethesda, MD 20814-3994. Copyright © 2014 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606.

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 25, 2014

Cutting Edge: Resident Memory CD8 T Cells Occupy Frontline Niches in Secondary Lymphoid Organs

The

Cutting Edge

Journal of

Immunology

Cutting Edge: Resident Memory CD8 T Cells Occupy Frontline Niches in Secondary Lymphoid Organs Resident memory CD8 T cells (TRM) are a nonrecirculating subset positioned in nonlymphoid tissues to provide early responses to reinfection. Although TRM are associated with nonlymphoid tissues, we asked whether they populated secondary lymphoid organs (SLO). We show that a subset of virus-specific memory CD8 T cells in SLO exhibit phenotypic signatures associated with TRM , including CD69 expression. Parabiosis revealed that SLO CD69+ memory CD8 T cells do not circulate, defining them as TRM. SLO TRM were overrepresented in IL-15–deficient mice, suggesting independent regulation compared with central memory CD8 T cells and effector memory CD8 T cells. These cells were positioned at SLO entry points for peripheral Ags: the splenic marginal zone, red pulp, and lymph node sinuses. Consistent with a potential role in guarding SLO pathogen entry points, SLO TRM did not vacate their position in response to peripheral alarm signals. These data extend the range of tissue resident memory to SLO. The Journal of Immunology, 2014, 192: 2961–2964.

M

emory CD8 T cells are widely distributed throughout the body and are partitioned into subsets based on trafficking properties (1, 2). Central memory CD8 T cells (TCM) recirculate through secondary lymphoid organs (SLO). Effector memory CD8 T cells (TEM) recirculate through nonlymphoid tissues (NLT). Resident memory CD8 T cells (TRM) are a newly recognized subset that patrols nonlymphoid frontline sites of pathogen exposure without recirculating (3–5). CD69 is expressed upon TCR stimulation by early effectors and T cells responding to chronic infections. CD69 is also constitutively expressed by TRM after Ag clearance, suggesting a separate pathway for regulation (6, 7). Tissue microenvironments induce constitutive CD69 expression by TRM, tying this phenotype to location (4, 6–9). Moreover, CD69 expression is associated with retention in tissues, suggesting that this marker plays a defining functional role in TRM differentiation (8). Department of Microbiology, University of Minnesota, Minneapolis, MN 55455; and Center for Immunology, University of Minnesota, Minneapolis, MN 55455 1

J.M.S. and K.A.F. contributed equally to this work.

Received for publication January 3, 2014. Accepted for publication February 3, 2014. This work was supported by National Institutes of Health Grants R01AI084913-01 (to D.M.), DP2OD006467 (by the Office of the Director) (to D.M.), T32AI007313, and F30DK100159-01 (both to J.M.S.).

TRM play a specialized sentinel role and are positioned to rapidly alert the host in the event of reinfection and accelerate pathogen clearance (4, 5, 10, 11). Thus, their anatomic localization at initial sites of Ag exposure is critical for the conceptualization of TRM function. Although these early responders are tightly associated with NLT distribution, SLO are compartmentalized and also contain “frontline” sites of initial Ag encounter (12). For instance, the subcapsular and medullary sinuses of lymph nodes (LN) capture lymph-borne pathogens derived from tissues (13, 14). In spleen, hematogenous pathogens are first encountered in the red pulp or trapped in the marginal zone (15). Thus, we hypothesized that TRM also may exist in the “frontline” portions of lymphoid tissue.

Materials and Methods Mice and infections All mice were used in accordance with the guidelines of the Institutional Animal Care and Use Committees at the University of Minnesota. C57BL/6J mice were purchased from The Jackson Laboratory. Thy1.1+ P14, CD45.1+ OT-I, and CD45.2+ OT-I mice were fully backcrossed to C57BL/6J mice and maintained in our animal colony. P14 immune chimeras were generated by transferring 5 3 104 naive transgenic Thy1.1+ P14 T cells into naive wildtype or IL-152/2 C57BL/6J mice and infecting with 2 3 105 PFU lymphocytic choriomeningitis virus (LCMV, Armstrong strain) i.p. the next day. Memory OT-I cells were generated by transferring 5 3 104 naive transgenic CD45.1+ OT-I T cells into memory P14 chimeras. The next day, recipients were infected with 1 3 106 PFU recombinant vesicular stomatitis virus expressing OVA (VSV-OVA) i.v., as described (16). Naive CD45.2+ OT-I T cells were injected into CD45.1+ RAG2/2 mice, as indicated. For local rechallenge experiments, 50 mg gp33 peptide was delivered transcervically, as described (10), in a volume of 35 ml delivered by modified gel loading pipette.

Parabiosis surgery Parabiosis surgeries were performed as described (10). Briefly, mice were shaved along opposite lateral flanks. Skin was wiped clean of fur with alcohol prep pads and further cleaned with Betadine solution and 70% isopropyl alcohol. Mirrored incisions were made on lateral aspects of both mice, and 5-0 VICRYL thread was used to suture skin to conjoin the mice. Additional sutures were placed through the olecranon and knee joints to secure the legs.

Immunofluorescence microscopy and flow cytometry For immunofluorescence microscopy, tissues were frozen in 2-methylbutane surrounded by dry ice. Frozen blocks were cut into 7-mm sections, fixed in acetone, blocked in a 5% BSA PBS solution for 1 h, and stained with DAPI (Invitrogen) and Abs specific for Thy1.1 (OX-7; eBioscience), CD45.1 (A20;

Address correspondence and reprint requests to Dr. David Masopust, Department of Microbiology and Center for Immunology, University of Minnesota, 2-182, 2101 6th Street SE, Minneapolis, MN 55455. E-mail address: [email protected] Abbreviations used in this article: LCMV, lymphocytic choriomeningitis virus; LN, lymph node; NLT, nonlymphoid tissue; SLO, secondary lymphoid organ; TCM, central memory CD8 T cell; TEM, effector memory CD8 T cell; TRM, resident memory CD8 T cell; VSV-OVA, vesicular stomatitis virus expressing OVA. Copyright Ó 2014 by The American Association of Immunologists, Inc. 0022-1767/14/$16.00

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1400003

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 25, 2014

Jason M. Schenkel,1 Kathryn A. Fraser,1 and David Masopust

2962

CUTTING EDGE: RESIDENT MEMORY CD8 T CELLS GUARD SLO

eBioscience), CD169 (AbD Serotec), CD69 (goat polyclonal; R&D Systems), and Collagen IV (rabbit polyclonal; Acris). Jackson ImmunoResearch secondary Abs conjugated to various fluorochromes were used to stain unconjugated Abs. Tiled images were acquired with an automated Leica DM5500B microscope, and analysis was performed with ImageJ and Adobe Photoshop. Cell isolations and flow cytometry were performed as previously described (17). Quantification of microscopy images was performed as previously described (10).

Statistical analysis The p values were determined by a two-tailed, unpaired Student t test. Differences between groups were considered significant if p # 0.05.

Results and Discussion A subset of memory CD8 T cells in SLO phenotypically resembles TRM

To determine whether a population of memory CD8 T cells within SLO expressed canonical markers of TRM, we analyzed

Thy1.1+ memory P14 CD8 T cells 8 wk after LCMV Armstrong infection (see Materials and Methods). P14 small intestine intraepithelial lymphocytes were used as a positive control because they were shown to be TRM in this infection model (3). Flow cytometric analysis revealed a subset of CD69+ memory CD8 T cells within both the LN (data not shown) and the spleen (Fig. 1A). CD69 + memory CD8 T cells within the SLO lacked CD62L, a cardinal feature of recirculating TCM. This unusual SLO population was distinct from canonical TEM (CD692 CD62L2) and TCM (CD692 CD62L+) but was similar to NLT-derived TRM, by expression of Ly6C, KLRG1, and Eomes (Fig. 1B). Unlike TRM that have been characterized in or near epithelial surfaces, CD69+ memory CD8 T cells within the SLO did not express CD103

FIGURE 2. CD69+ SLO memory CD8 T cells are TRM positioned at common Ag entry sites. (A) P14 LCMV-immune chimeric (black bars) and naive (white bars) mice were surgically conjoined. Fifteen days later, the fraction of P14 that expressed CD69 was enumerated histologically within the SLO of both parabionts. (B) The proportion of CD69+ P14 (black) and CD692 P14 (white) memory CD8 T cells present in each SLO subcompartment of the immune parabiont was determined histologically. (C) Representative staining showing CD69+ memory CD8 T cells in the LN sinus. Scale bars, 20 mm. (D) Representative CD69 staining in the red pulp, white pulp, and marginal zone of the spleen. Original magnification 320, (C) and (D). Scale bars in (D), 20 mm. (E) The fraction of P14 memory CD8 T cells within each SLO compartment of the immune parabiont that expressed CD69. Data are from six mice from two independent experiments. *p , 0.01, **p , 0.001.

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 25, 2014

FIGURE 1. A subset of memory CD8 T cells in SLO phenotypically resembles TRM. (A) Eight weeks after LCMV infection, P14 memory CD8 T cells in spleen were analyzed for the indicated markers by flow cytometry. All plots gated on Thy1.1+ P14 memory CD8 T cells. (B) As in (A), however, P14 splenocytes were split into CD69+ CD62L2 (black line), CD692 CD62L2 (blue line), and CD692 CD62L+ (red line) subsets. P14, isolated from small intestinal epithelium (green line), were included as a positive control for previously defined TRM. (C) P14 splenocyte phenotype was compared on days 7 and 40 after LCMV infection. (D) A total of 5 3 104 naive CD45.2+ OT-I splenocytes were transferred to CD45.1+ RAG2/2 C57BL/6 mice. Sixty days later, OT-I splenocytes were analyzed for the indicated markers by flow cytometry. Data are representative of six mice from two independent experiments.

The Journal of Immunology

2963

(i.e., aeb7 integrin; data not shown). It should be noted that CD69 expression was upregulated after clearance of LCMV, suggesting that persistent foreign Ag was not driving expression (Fig. 1C). To support this interpretation, we transferred naive Rag2/2 OT-I CD8 T cells to lymphopenic RAG2/2 mice, which results in homeostatically expanded memory-like T cell differentiation without pathogenderived Ag (6). Under these conditions, we also observed a CD8 T cell subset within SLO that exhibited a TRM-like phenotype (Fig. 1D). CD69+ SLO memory CD8 T cells are TRM positioned at common Ag entry sites

We wished to test whether CD69+ memory CD8 T cells within SLO were TRM. The vascular systems of LCMVimmune mice (generated as in Fig. 1) were surgically conjoined to naive mice via parabiosis, which equilibrates TCM and TEM between hosts without equilibrating TRM (5, 18). Fig. 2A shows that the CD69+ SLO memory CD8 T cell population was specifically absent in the naive parabiont, demonstrating that this population was indeed resident within SLO. Immunofluorescence microscopy localized these TRM preferentially to the marginal zone and red pulp of the spleen and the subcapsular and medullary sinuses of the LN (Fig. 2B–E), which represent the primary initial sites of Ag entry into these SLO.

SLO TRM do not redistribute in response to peripheral alarm signals

Upon Ag recognition within the female reproductive mucosa, reactivated local memory CD8 TRM precipitate alarm signals (10). These signals recruit bystander resting memory CD8 T cells, which redistribute from spleen and blood to the site of reactivation in the mucosa (10). We asked whether this redistribution function was restricted to recirculating memory CD8 T cells or whether CD8 TRM in the SLO also would mobilize to the female reproductive tract in this context. To this end, we transferred naive OT-I CD8 T cells into P14 LCMV-immune chimeras. The next day, we infected these mice with VSV-OVA. Thirty days later, these mice contained populations of both OT-I and P14 memory CD8 T cells. They were then rechallenged transcervically with gp33 peptide, as described (10), to reactivate P14 within the female reproductive tract and precipitate local alarm signals that induce redistribution of memory CD8 T cells. CD69+ and CD692 OT-I CD8 T cells were enumerated within the spleen (2 d after gp33 peptide rechallenge) (Fig. 4). Unlike recirculating memory OT-I CD8 T cells, many of which vacated SLO to respond to the site of peripheral P14 CD8 T cell reactivation, the number of CD69+ memory OT-I CD8 T cells was unaffected. This indicates that, unlike recirculating memory CD8 T cell subsets, TRM maintain their

Reduced IL-15 dependence of CD8 TRM in SLO

Previous reports showed that maintenance of T CM and TEM is critically dependent on IL-15 (19). We found that CD69+ TRM within SLO express reduced levels of CD122 (IL-15Rb, Fig. 3A), similar to what was reported for TRM located within the epithelium of the small intestine (17). We hypothesized that a distinguishing feature between SLO TRM and recirculating memory CD8 T cells could be dependence upon IL-15. To test this hypothesis, we compared memory P14 differentiation and maintenance in wild-type and IL-15–deficient LCMV-immune chimeras (see Materials and Methods). Sixty days after LCMV infection, we found that CD69+ SLO TRM were overrepresented in the absence of IL-15 (Fig. 3B, 3C). These data suggest that SLO TRM are less dependent upon IL-15 for their survival compared with recirculating primary TCM and TEM, which further defines them as a unique subset.

FIGURE 4. SLO TRM do not redistribute in response to peripheral inflammation. Both P14 and OT-I memory CD8 T cells were established in mice following LCMV and VSV-OVA infection. Mice were rechallenged transcervically with 50 mg gp33 peptide. Two days later, CD692 (left panel) and CD69+ (right panel) OT-I memory CD8 T cells were enumerated within spleen. Data are from three mice/group representative of two independent experiments. *p , 0.01.

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 25, 2014

FIGURE 3. Reduced IL-15 dependence of CD8 TRM in SLO. (A) CD122 (IL-15Rb) expression was analyzed by flow cytometry on CD69+ (black line) and CD692 (gray line) memory P14 CD8 T cells isolated from spleen 60 d after LCMV infection. (B and C) P14 immune chimeras were generated in wild-type (WT; black) and IL-152/2 (white) mice. Sixty days later, CD69+ and CD692 P14 were enumerated in LN and spleen. Data are from five mice/group from two independent experiments representative of four different experiments. *p , 0.05, **p , 0.01, ***p , 0.001.

2964

CUTTING EDGE: RESIDENT MEMORY CD8 T CELLS GUARD SLO

Disclosures The authors have no financial conflicts of interest.

References 1. Mueller, S. N., T. Gebhardt, F. R. Carbone, and W. R. Heath. 2013. Memory T cell subsets, migration patterns, and tissue residence. Annu. Rev. Immunol. 31: 137–161. 2. Masopust, D., and J. M. Schenkel. 2013. The integration of T cell migration, differentiation and function. Nat. Rev. Immunol. 13: 309–320. 3. Masopust, D., D. Choo, V. Vezys, E. J. Wherry, J. Duraiswamy, R. Akondy, J. Wang, K. A. Casey, D. L. Barber, K. S. Kawamura, et al. 2010. Dynamic T cell migration program provides resident memory within intestinal epithelium. J. Exp. Med. 207: 553–564. 4. Gebhardt, T., L. M. Wakim, L. Eidsmo, P. C. Reading, W. R. Heath, and F. R. Carbone. 2009. Memory T cells in nonlymphoid tissue that provide enhanced

5. 6.

7.

8.

9. 10. 11. 12. 13.

14.

15. 16.

17. 18. 19. 20. 21.

22.

local immunity during infection with herpes simplex virus. Nat. Immunol. 10: 524– 530. Jiang, X., R. A. Clark, L. Liu, A. J. Wagers, R. C. Fuhlbrigge, and T. S. Kupper. 2012. Skin infection generates non-migratory memory CD8+ T(RM) cells providing global skin immunity. Nature 483: 227–231. Casey, K. A., K. A. Fraser, J. M. Schenkel, A. Moran, M. C. Abt, L. K. Beura, P. J. Lucas, D. Artis, E. J. Wherry, K. Hogquist, et al. 2012. Antigen-independent differentiation and maintenance of effector-like resident memory T cells in tissues. J. Immunol. 188: 4866–4875. Mackay, L. K., A. T. Stock, J. Z. Ma, C. M. Jones, S. J. Kent, S. N. Mueller, W. R. Heath, F. R. Carbone, and T. Gebhardt. 2012. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc. Natl. Acad. Sci. USA 109: 7037–7042. Mackay, L. K., A. Rahimpour, J. Z. Ma, N. Collins, A. T. Stock, M.-L. Hafon, J. Vega-Ramos, P. Lauzurica, S. N. Mueller, T. Stefanovic, et al. 2013. The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat. Immunol. 14: 1294–1301. Skon, C. N., J.-Y. Lee, K. G. Anderson, D. Masopust, K. A. Hogquist, and S. C. Jameson. 2013. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat. Immunol. 14: 1285–1293. Schenkel, J. M., K. A. Fraser, V. Vezys, and D. Masopust. 2013. Sensing and alarm function of resident memory CD8⁺ T cells. Nat. Immunol. 14: 509–513. Teijaro, J. R., D. Turner, Q. Pham, E. J. Wherry, L. Lefranc¸ois, and D. L. Farber. 2011. Cutting edge: Tissue-retentive lung memory CD4 T cells mediate optimal protection to respiratory virus infection. J. Immunol. 187: 5510–5514. Junt, T., E. Scandella, and B. Ludewig. 2008. Form follows function: lymphoid tissue microarchitecture in antimicrobial immune defence. Nat. Rev. Immunol. 8: 764–775. Junt, T., E. A. Moseman, M. Iannacone, S. Massberg, P. A. Lang, M. Boes, K. Fink, S. E. Henrickson, D. M. Shayakhmetov, N. C. Di Paolo, et al. 2007. Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells. Nature 450: 110–114. Gonzalez, S. F., V. Lukacs-Kornek, M. P. Kuligowski, L. A. Pitcher, S. E. Degn, Y.-A. Kim, M. J. Cloninger, L. Martinez-Pomares, S. Gordon, S. J. Turley, and M. C. Carroll. 2010. Capture of influenza by medullary dendritic cells via SIGN-R1 is essential for humoral immunity in draining lymph nodes. Nat. Immunol. 11: 427–434. Aoshi, T., J. A. Carrero, V. Konjufca, Y. Koide, E. R. Unanue, and M. J. Miller. 2009. The cellular niche of Listeria monocytogenes infection changes rapidly in the spleen. Eur. J. Immunol. 39: 417–425. Kim, S. K., D. S. Reed, S. Olson, M. J. Schnell, J. K. Rose, P. A. Morton, and L. Lefranc¸ois. 1998. Generation of mucosal cytotoxic T cells against soluble protein by tissue-specific environmental and costimulatory signals. Proc. Natl. Acad. Sci. USA 95: 10814–10819. Masopust, D., V. Vezys, E. J. Wherry, D. L. Barber, and R. Ahmed. 2006. Cutting edge: gut microenvironment promotes differentiation of a unique memory CD8 T cell population. J. Immunol. 176: 2079–2083. Klonowski, K. D., K. J. Williams, A. L. Marzo, D. A. Blair, E. G. Lingenheld, and L. Lefranc¸ois. 2004. Dynamics of blood-borne CD8 memory T cell migration in vivo. Immunity 20: 551–562. Surh, C. D., and J. Sprent. 2008. Homeostasis of naive and memory T cells. Immunity 29: 848–862. Hofmann, M., A. Oschowitzer, S. R. Kurzhals, C. C. Kr€ uger, and H. Pircher. 2013. Thymus-resident memory CD8+ T cells mediate local immunity. Eur. J. Immunol. 43: 2295–2304. Sathaliyawala, T., M. Kubota, N. Yudanin, D. Turner, P. Camp, J. J. C. Thome, K. L. Bickham, H. Lerner, M. Goldstein, M. Sykes, et al. 2013. Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets. Immunity 38: 187–197. Turner, D. L., K. L. Bickham, J. J. Thome, C. Y. Kim, F. D’Ovidio, E. J. Wherry, and D. L. Farber. 2013. Lung niches for the generation and maintenance of tissueresident memory T cells. Mucosal Immunol. DOI: 10.1038/mi.2013.67.

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 25, 2014

presence in the SLO even in the context of peripheral alarm signals. TRM have been identified in many NLT, as well as the thymus, where they are positioned to rapidly provide protection against pathogens re-encountered at anatomic barriers (4, 5, 7, 11, 20). TRM express unique phenotypic signatures, including CD69 expression, that distinguish them from recirculating TCM and TEM (4, 8, 17). TRM differentiation is induced by local environmental cues; thus, TRM ontogeny is intrinsically coupled with location (6, 8). Therefore, it is surprising that we defined a population of bona fide TRM via parabiosis experiments, which also could be identified by CD69 expression, present within lymphoid tissues. It should be noted that CD69+ CD8 T cells also have been detected in human SLO (21, 22). It remains possible that local inductive cues also regulate TRM ontogeny within SLO microenvironments. Indeed, SLO TRM occupied specific niches within lymphoid organs, particularly the subcapsular and medullary sinuses of LN and the marginal zones of spleen, which showed minimal overlap with TCM and TEM. These compartments also represent major portals for pathogen and Ag entry into lymphoid organs (12). In light of these findings, there may be operational similarities between SLO TRM and those in NLT tissues: as guardians of frontline sites of pathogen exposure. It is telling that, unlike CD692 recirculating memory CD8 T cell populations, SLO TRM did not abandon the lymphoid tissue in response to distant inflammatory cues. Perhaps they fulfill a specialized immunosurveillance role that, like NLT TRM, accelerates alarm signals and pathogen control in response to reinfection.

Cutting edge: resident memory CD8 T cells occupy frontline niches in secondary lymphoid organs.

Resident memory CD8 T cells (TRM) are a nonrecirculating subset positioned in nonlymphoid tissues to provide early responses to reinfection. Although ...
1MB Sizes 3 Downloads 3 Views