<<

Bacterial modulates host to establish a T-helper cell, type 17 (Th17)-dominant immune response in infection

Annah S. Roliga, J. Elliot Carterb, and Karen M. Ottemannc,1

Departments of aMolecular, Cellular, and Developmental Biology and cMicrobiology and Environmental Toxicology, University of California, Santa Cruz, CA 95064; and bDepartment of Pathology, University of South Alabama College of Medicine, Mobile, AL 36688

Edited by Ralph R. Isberg, Howard Hughes Medical Institute/Tufts University School of Medicine, Boston, MA, and approved October 27, 2011 (received for review March 22, 2011) The host inflammatory response to chronic bacterial infections often of their contact with the , because the APCs produce dictates the disease outcome. In the case of the gastric pathogen that dictate the character of the adaptive immune re- Helicobacter pylori,hostinflammatory responses result in outcomes sponse. Dendritic cells interacting with H. pylori fuel the pro- that range from moderate and asymptomatic to more severe with liferation of particular T cells, including T helper cells, type 1 (Th1 concomitant ulcer or . It was found recently that H. pylori cells) (8), CD25+FoxP3+ T-regulatory cells (T-regs) (8, 9), and T − chemotaxis mutants (Che ), which lack directed but colo- helper cells, type 17 (Th17 cells) (10). nize to nearly wild-type levels, trigger less host inflammation. We The inflammatory response to H. pylori includes all these T-cell used these mutants to observe host immune responses that resulted types. However, the roles of the Th17 and T-reg cell populations in reduced disease states. Here we report that these mutants are during H. pylori infections have been debated recently. The Th17 defective for early gastric recruitment of CD4+ T cells compared with cell is involved in promoting chronic inflammation (11, 12); the T- − wild-type infection. Furthermore, Che mutant infections lack the T- reg cell, in contrast, regulates host immune responses. Th17 and T- helper cell, type 17 (Th17) component of the immune response, as reg cells are developmentally related and exist in a delicate balance measured by mRNA levels in gastric tissue via intracellular (13) that can dictate the outcome of a bacterial infection (14). cytokine staining and immunofluorescence. We additionally find MICROBIOLOGY − Evidence suggests that H. pylori pathogenesis results primarily that a Che mutant infection results in significantly less host cell from the immune response, and thus understanding how this im- apoptosis than does wild-type infection, in accordance with previous mune response is initiated and controlled is critical. Currently it is observations that T-helper cell, type 17 responses in Citrobacter unknown if a Th17 response (12) or a T-reg response (9) underlies rodentium infections are driven by concomitant bacterial and apopto- the ineffective immune response to H. pylori. Therefore, we sought tic cell signals. We propose that bacterial chemotaxis allows H. pylori to understand better how H. pylori promotes gastritis by comparing to access a particular host niche that allows the to express or the host immune cell and cytokine responses to wild-type H. pylori − deliver proapoptotic host cell factors. This report indicates that che- and to a Che mutant. Our studies provide evidence that bacte- motaxis plays a role in enhancing apoptosis, suggesting bacterial rially driven interactions with host tissues alter the nature of the chemotaxis systems might serve as therapeutic targets for infections immune and pathological response generated during infection. whose symptoms arise from host cell apoptosis and tissue damage. Results and Discussion T regulatory cells | adaptive immunity | pathogenesis H. pylori Chemotaxis Increases Inflammation 2 mo After Inoculation. − As stated above, Che H. pylori cause milder inflammation than do nfection with the gastric pathogen Helicobacter pylori leads to wild-type infections after 3–6 mo of colonization (4). To determine Ichronic inflammation, or gastritis, in all individuals. This bac- whether bacterial chemotaxis affected inflammation earlier, we terium colonizes 50% of the world’s population and triggers a examined inflammation at the earliest time inflammation was de- wide range of disease severities; many infected individuals remain tectable, 2 mo after inoculation. For these experiments, we orally − asymptomatic, but others develop peptic or gastric ulcers, gastric infected mice with either wild-type H. pylori or an isogenic Che adenocarcinoma, or mucosa-associated lymphoid tumors (1). mutant lacking a central chemotaxis protein, CheY. H. pylori cheY The pathogenesis of H. pylori-induced inflammation is not well mutants have been characterized extensively and found to retain understood. Inflammation is promoted by both host factors (2) flagella and motility but to lack chemotaxis completely (5, 15). − and H. pylori factors, such as the proteins cytotoxin associated gene Che mutants have early mouse colonization defects but achieve A (CagA) (1, 2) and vacuolating cytotoxin A (VacA) (1, 3) and normal bacterial levels by 1 mo after inoculation (5, 16). All cheY bacterial chemotaxis (4). Chemotaxis is the bacterial ability to mutant-associated phenotypes can be complemented, indicating move toward beneficial environmental signals and away from that loss of cheY is responsible for the chemotaxis and animal- harmful ones. H. pylori genetically altered to lack chemotaxis colonization deficits (5, 15). Using standard inflammation grading − (Che ) retain flagella and motility but cannot migrate toward or that captures the number and distribution of , we away from environmental signals. In mouse models, these mutants found that inflammation was significantly lower in mice infected − have a marginal colonization defect (4–6) but induce less overall for 2 mo with Che H. pylori than in mice infected with wild-type H. − chronic inflammation (4). Specifically, Che mutants localize far pylori but was greater than in the no-H. pylori control (Fig. 1). from the epithelial surface and do not colonize the gastric glands robustly (4, 6), suggesting that chemotaxis-driven contact with epithelial cells, resident dendritic cells, or promotes the Author contributions: A.S.R. and K.M.O. designed research; A.S.R. and J.E.C. performed inflammatory response to H. pylori. research; A.S.R. and K.M.O. analyzed data; and A.S.R. and K.M.O. wrote the paper. Inflammation begins when resident monocytes and epithelial The authors declare no conflict of interest. cells detect injury or a pathogen such as H. pylori (7). Epithelial cells This article is a PNAS Direct Submission. secrete to recruit antigen-presenting cells (APCs) such 1To whom correspondence should be addressed. E-mail: [email protected]. as dendritic cells that will prime T cells (7). The newly recruited This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. APCs definetheimmuneresponsetoH. pylori based on the nature 1073/pnas.1104598108/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1104598108 PNAS Early Edition | 1of6 Downloaded by guest on September 27, 2021 − We infected mice with either wild-type or Che H. pylori and allowed infections to proceed for 2 wk to 3 mo. The total T-cell populations (CD3+CD45+) in infected stomachs rose steadily from week 2 to week 5 and displayed a subtle trend for the wild − type to recruit more T cells than the Che mutant (Fig. S1A). Additionally, the percentage of CD4+ T cells remained at back- − ground levels in Che infections but increased significantly above background in wild-type H. pylori infections (Fig. S1B). T-cytotoxic cells (CD8+CD3+) never increased over the mock-infection con- − trol and were not different between the wild-type and Che infections (Fig. S1B), in agreement with previous reports that this Fig. 1. H. pylori chemotaxis promotes inflammation. Inflammation grade of T-cell type does not play a major role in H. pylori immunity (17). stomach sections taken from C57BL/6 mice 2 mo after infection with wild- When we extended our flow cytometric analysis to 3 mo after − − type or Che H. pylori SS1. n = 6 for wild-type and Che H. pylori infections; inoculation, we observed that the percentage of CD4+ T cells in − n = 3 for mock infections. Error bars represent SEM. At 2 mo after in- the Che infection had risen to match those in the wild-type in- oculation, there was a not a statistically significant difference between the log + − fection, suggesting that CD4 T-cell recruitment is delayed but not cfu/g stomach tissue for wild-type (6.29 ± 1.1) and Che H. pylori (5.72 ± 0.24). − fl *P < 0.05, Student t test. abrogated in Che infections (Fig. 2 A and B). To verify our ow cytometry result, we further used immunohistochemistry at 2 mo − after inoculation and indeed confirmed that wild-type and Che fi Overall scores at this time point are low, con rming that 2 mo after H. pylori recruited similar numbers of CD4+ cells at this later time fl inoculation is the earliest time point at which in ammation is (Fig. 2 C and D). However, we did observe slight differences in the fi + pathologically identi able. Bacterial numbers at 2 mo after in- distribution of CD4 cells between the two infection types, with 6 oculation in both infection groups were ∼1 × 10 cfu/g and did not wild-type infections bearing more widespread inflammatory cells − differ significantly from each other, similar to previous findings (4, (Fig. 2C). Overall, these findings show that early Che infections 5), suggesting that inflammatory differences are not caused by a recruit fewer CD4+ T cells than do wild-type infections. This dif- disparity in bacterial burden. Additionally, there was no correlation ference dissipates sometime between 5 and 8 wk after inoculation, within individual mice between bacterial colonization levels and but significant differences in the inflammation grading remain. The inflammation grade, as seen previously (4). This finding thus sug- inflammation grading method examines density in − − gests that Che inflammatory deficits arise before 2 mo of infection. gastric tissue, and thus our score differences suggest that Che fl − in ammation is more multifocal in character, whereas wild-type in- Che H. pylori Inefficiently Recruits CD4+ T Cells. We were curious fl − ammation is more widespread (Materials and Methods)(Fig.2C). about the exact cell types that are reduced in the Che infection. Numerous studies have found that T cells are the dominant lym- H. pylori Chemotaxis Affects Gastric Cytokine Profiles. CD4+ T cells phocyte in an H. pylori infection (8) so we examined T-cell subsets comprise different subsets that play distinctive roles during infec- in detail using both flow cytometry and immunohistochemistry. tion. Therefore, we examined total gastric tissue for the types of

Fig. 2. H. pylori chemotaxis is not necessary for the presence of CD4+ lymphocytes 2 mo after inoculation. (A) Representative flow cytometry plots showing the percentage of CD3+CD4+ T cells of the CD45+CD3+ gastric lymphocytes from tissue 3 mo after infection. Numbers in quadrants indicate the percentage of positively stained cells. (B) CD4+ cell percentage is presented as the percent of CD4 positively stained cells out of CD3+CD45+ lymphocytes. In total, 50,000 cells were counted. n = 6 mice for wild-type and Che− H. pylori infection; n = 4 mice for mock infection. Results of flow cytometric experiments are representative of two independent experiments. Each bar represents mean ± SEM. (C) Representative images of mouse stomach tissue stained with CD4 from wild- − type H. pylori infection with mild widespread inflammation (Left) or Che H. pylori infection demonstrating mild multifocal infiltration (Right). Arrow indicates a focus of CD4 cells. (Scale bar, 50 μm.) (D) Immunohistochemical enumeration of CD4+ cells in mouse stomach tissue 2 mo postinoculation (PI). An average of 14.1 mm2 of tissue was analyzed per mouse. Data are reported as number of cells per mm2 of tissue ± SD. These data were subjected to Grubbs’ − statistical outlier test, which resulted in the removal of one wild-type and one mock infection data point. Wild-type infection, n = 5 mice; Che infection, n =6 mice; mock infection, n = 2 mice.

2of6 | www.pnas.org/cgi/doi/10.1073/pnas.1104598108 Rolig et al. Downloaded by guest on September 27, 2021 CD4+ T cells present by monitoring transcripts of genes encoding Th17 subset are significantly elevated in the wild-type infection, cytokines and transcription factors associated with Th1 (IFN-γ), T- qPCR of total gastric tissue cannot specifically identify the cellular helper, type 2 (Th2; IL-4), Th17 [IL-6, IL-17A, TGF-β, and related cytokine source. IL-17A, for example, is secreted from Th17 cells orphan -γT(Rorγt)], and T-reg [IL -10, TGF-β,and as well as from γδ T cells (20) and lymphoid tissue-inducer (LTi) forkhead box P3 (Foxp3)] cells. IL-6 and TGF-β are required for cells (21). We therefore performed intracellular cytokine staining Th17 cell differentiation, whereas T-regs require only TGF-β (13). to assess which gastric tissue lymphocytes produce IL-17A. To Examining the induction of the Th17-specific transcription factor accomplish this goal, we sorted CD45+ live cells from gastric tissue Rorγt (13) and the production of IL-17A further identifies IL- of H. pylori-infected mice, stimulated them, and subsequently 17–producing cells. We additionally assessed T-cell activation by stained them with the surface markers CD3, CD4, and CD45 and monitoring IL-2. the cytokine IL-17A. In wild-type H. pylori infections, IL-17A– Two months after inoculation, when both infection types had producing cells were detected within the CD4+ cells after gating equal numbers of CD4+ cells (Fig. 2), stomachs infected with wild- on CD3+CD45+ populations, consistent with Th17 cells (Fig. 4 A type H. pylori displayed statistically significant increases in mRNA and B). A smaller population of IL-17A–producing cells also was − for Il10 and the Rorγt-encoding gene Rorc as well as a strong in- detected with in the CD4 CD3+CD45+ populations, suggesting − crease in Il17a, IL6, and Foxp3 compared with levels in the Che - secretion by CD3+ γδ T cells, given that there are few CD8 T cells infected stomach, which remained at the levels occurring in mock- in these infections (Fig. S1). No IL-17A production was detected − infected stomachs (Fig. 3 A and B). Rorγt, IL-17A, and IL-6 are within the CD3 CD45+ populations from either wild-type– or − associated with IL-17–producing cells, suggesting that the wild- Che -infected mice, suggesting that LTi cells, which lack CD3 type infection skews the response toward IL-17–producing cells. (21), were not the source of IL-17A production at this time point − Tgfβ and Foxp3 (Fig. 3) levels in wild-type and Che H. pylori in- (Fig. S2). Other types of innate lymphoid cells have been shown to fections were not altered significantly from the levels in mock in- drive intestinal pathology in a Helicobacter hepaticus model (22); fection, but Il10 was up-regulated significantly in the wild-type thus it would be interesting to look at earlier time points to see if infection, suggesting that T-regs may be more active in a wild-type LTi cells contribute to the initiation of adaptive responses to H. − than in a Che H. pylori infection. The presence of T-regs in pylori. We also performed an immunofluorescence study looking H. pylori infection has been well established (8), but their role in for the colocalization of CD4 and IL-17A in gastric tissue slices the outcome of the immune response still is debated. Additionally, from mice 2 mo after infection (Fig. 4 C and D). This experiment Th1 and Th2 cells secrete IL-10 as a way to control their responses confirmed the results of intracellular cytokine staining, finding that

(18), and Th17 cells also have been shown to produce IL-10 under CD4 and IL-17A were colocalized to a significant degree in tissue MICROBIOLOGY some circumstances (19). infected with wild-type H. pylori (Fig. 4 C and D). We detected only Because we observed the up-regulation of Il10, a regulatory few cells that, as would γδ T cells, stained solely with IL-17A and cytokine, we additionally determined the ratio of Rorc to Foxp3 to not CD4; this finding is consistent with a low number of γδ T cells − assess whether the immune response is more inflammatory or in wild-type and Che H. pylori infections (Fig. S3) and supports regulatory in character; higher ratios of Rorc:Foxp3 indicate that a principal role for Th17 cells. Together the results from the in- the immune response trends toward inflammatory. This parameter tracellular cytokine staining, the immunofluorescence assay, and also has been examined as the Il17a:Foxp3 ratio (9). We found that the gastric cytokine analysis strongly indicate that the Th17 subset the both the Rorc:Foxp3 ratio and the Il17a:Foxp3 ratio are higher is elevated significantly in stomachs with wild-type H. pylori in- − − in the wild-type infection than in the Che infection and the mock fection as compared with stomachs with either Che H. pylori or infection (Fig. 3C). This finding is consistent with previous evi- mock infection. These findings suggest a role for bacterial che- dence showing IL-17 expression in H. pylori-infected gastric tissue motaxis in eliciting a Th17 response. Th17 responses have been (9, 12, 17). Furthermore, studies have shown that depletion of IL- implicated in Citrobacter rodentium (23) and Salmonella enterica − − 17 or the use of IL-17 / mice lowers both H. pylori colonization serovar Typhimurium infections (24), where these responses seem levels and inflammation grade (12). to underlie pathology.

Chemotactic H. pylori Induces a Th17 Response. Although the quan- Chemotaxis Promotes Apoptosis in H. pylori Infection. Th17 cell titative PCR (qPCR) data for IL6, IL17a,andRorc suggest that differentiation is directed in part when innate immune cells, such key cytokines critical for the development and activation of the as APCs, recognize apoptotic cells combined with bacterial pro-

Fig. 3. H. pylori chemotaxis is necessary to induce an inflammatory response associated with Rorγt expression. (A) Gastric mRNA expression of Ifng, Il4, Il6, − Tgfb, Il17a, Il2, and Il10 in mice infected with wild-type or Che H. pylori for 2 mo. RNA was isolated from total mouse stomach tissue and used for qRT-PCR. mRNA expression levels are presented as the fold increase over mock infection, as determined by the ΔΔ threshold cycle (Ct) method. (B) Gastric mRNA expression of the transcription factors Foxp3 and Rorc (Rorγt) 2 mo after infection. (C) Ratio of Rorc or Il17a to Foxp3. Each bar represents the mean ± SEM based on experiments using five or six mice [wild-type (6.29 ± 1.1 log cfu/g) and Che− (5.72 ± 0.24 log cfu/g) H. pylori infection] or two to five mice (mock infection). All experiments were performed in triplicate. *P < 0.05; Mann–Whitney u test.

Rolig et al. PNAS Early Edition | 3of6 Downloaded by guest on September 27, 2021 Fig. 4. H. pylori chemotaxis promotes a Th17 response. (A) CD45+-positive/ propidium iodide-negative cells were sorted from the entire stomach of each mouse, Yields differed depending on infection type and status; therefore, results are presented as percentages of parent populations. Intracellular cytokines were detected 4 h after restimulation with phosphomolybdic acid/ionomycin. Flow cytometry plots were gated on CD3+CD45+ (Left) to select the CD3+CD45+ cells for additional analysis for CD4 and on IL-17A (Right). The top two boxes show isotype controls to demonstrate the veracity of the sorted cell populations. Numbers in quadrants indicate the percentage of positively stained cells. Data shown are one representative of n = 6 from two experiments for wild-type and Che− H. pylori infections and n = 4 for mock infections. (B) Percentage of CD4+ and CD4− IL17A+ cells. These cells were collected 2 mo after inoculation, when the mice were colonized with 6.91 ± 0.18 log cfu/g or 6.16 ± 0.18 log cfu/g for wild-type and Che− H. pylori infections, respectively. (C) Two representative examples showing colocalization of CD4+ (red) and IL-17A (green) in stomach tissue. (D) Percentage of total cells counted that colocalized IL-17A and CD4+ − (black bars) or stained with IL-17A alone (white bars). Samples from six mice (wild-type H. pylori), five mice (Che H. pylori), or three mice (mock infection) − were examined; 2,029, 1,531, and 145 cells were examined for wild-type, Che , and mock infections, respectively. Each bar represents the mean ± SEM. *P < 0.05; Student’s t test. N.S., not significant. Two independent infections were used to generate the data for A and B and for C and D.

ducts that activate Toll-like receptors (24). In addition to apo- apoptosis of host cells when bound by its , FasL (30). − ptosis, Th17 development also is associated with IL-10 induction Therefore, our results suggest Che bacteria express or deliver − (24); we observed high IL-10 levels in the wild-type infection (Fig. less proapoptotic virulence factors in vivo. Che H. pylori localize 3A). Thus, one possibility is that wild-type H. pylori triggers sig- farther from the gastric epithelial cell surface than does wild type − nificant apoptotic cells and in turn Th17 induction. To probe this (4, 6) suggesting Che either may be too spatially distant from hypothesis, we enumerated apoptotic cells in gastric tissue by the target cells for efficient delivery of virulence factors or may TUNEL staining 2 mo after inoculation, when equal numbers of experience a different microenvironment that does not trigger − wild-type and Che bacteria colonize the stomach and there are appropriate expression of apoptosis-inducing proteins. Support- differences in the degree of Th17 response (Figs. 3 and 4). We ing this latter idea, chemotaxis regulates virulence factor ex- found elevated apoptosis in the corpus associated with wild-type pression in Vibrio cholerae (31). − H. pylori in comparison with Che (Fig. 5 A and B). Although To investigate if different mechanisms leading to apoptosis vary − apoptosis has been demonstrated previously in several bacterial in wild-type and Che H. pylori, we looked at the gastric mRNA infections, including H. pylori (25), the link to Th17 responses has expression of Fas, which is up-regulated and contributes to apo- been demonstrated only in C. rodentium infections (24). C. ptosis in H. pylori infection (30), and its ligand, FasL. We found rodentium strains are generally nonmotile (26), and instead use that after 2 mo of infection the expression of Fas in wild-type and − type-three secretion-based adherence to deliver proapoptotic Che H. pylori infections is similar to that seen in an uninfected − factors (27). We can now link apoptosis in H. pylori infection to mouse (Fig. S4). Additionally, wild-type and Che infections lead bacterial chemotaxis and Th17 responses. equally to up-regulation of FasL (Fig. S4), indicating that Fas/ H. pylori can induce apoptosis in epithelial cells and macro- FasL-induced cell death does not contribute to apoptotic differences phages through secretion of proapoptotic factors including the seen in gastric tissue. We also examined the expression of the H. VacA cytotoxin (3), urease (28), and γ-glutamyl transpeptidase pylori apoptosis-causing virulence factors VacA and urease (subunit) (29) and by up-regulating Fas, a TNF receptor that triggers A (UreA) in mouse stomachs after 2-mo infection (Fig. S5). We

4of6 | www.pnas.org/cgi/doi/10.1073/pnas.1104598108 Rolig et al. Downloaded by guest on September 27, 2021 Fig. 5. H. pylori chemotaxis correlates with increased gastric cell apoptosis. (A) Representative images taken from the corpus of a mouse infected with wild- type or Che− H. pylori 2 mo after inoculation stained for apoptotic cells (brown) using the TUNEL assay. (Scale bars, 50 μm.) (B) Apoptotic cells were counted in 20 randomly selected well-oriented glands of the corpus. Each bar represents the mean ± SEM based on experiments using six mice (wild-type and Che−H. pylori infections) or three mice (mock infections). These tissue samples were collected 2 mo after inoculation, when the mice were colonized with 6.29 ± 1.1 − log cfu/g wild-type or 5.72 ± 0.24 log cfu/g Che H. pylori.*P < 0.05, Student t test. N.S., not significant.

− found that on average Che bacteria have a somewhat lower ex- grown H. pylori. After the infection period, the animals were killed via CO2 pression of VacA but a somewhat higher expression of UreA than narcosis, and the stomach was dissected, opened along the lesser curvature, and wild-type bacteria (Fig. S5). The expression of UreA is regulated divided into longitudinal strips for preservation and analysis. The tissue pieces by the acid-responsive signaling regulator/sensor (ArsRS) two- were (i) homogenized using the Bullet Blender (Next Advance) with 1.0-mm zirconium silicate beads and plated to determine the number of cfu/g of component system, which prompts pH-controlled gene expres- − stomach tissue; (ii) preserved in Optimal Cutting (Tissue-Tek OCT, sion (32). Therefore, because Che bacteria are farther from the Sakura Finetek), snap-frozen in liquid nitrogen, and stored at −80 °C for im- epithelial cells (4, 6) where they may encounter a more acidic pH, munohistochemistry; (iii) frozen in liquid nitrogen and stored at −80 °C for qRT- it is not surprising that they have slightly increased expression of PCR of cytokines; or (iv) stored in cold HBSS (Lonza) to be used in flow

UreA. The slightly lower expression of VacA along with a greater cytometry experiments. MICROBIOLOGY delivery distance may contribute to reduced apoptosis through reduced delivery of VacA to the epithelial cells. Interestingly an- Immunohistochemistry/Immunofluorescence. Detailed information about im- other virulence factor, the cytotoxin gene-associated pathogenicity munohistochemistry and immunofluorescence is provided in SI Materials and fl island (cag-PAI) has been associated with antiapoptotic activity (1). Methods. In brief, samples for immunohistochemistry and immuno uores- cence were prepared from OCT-frozen sections. For immunohistochemistry, The H. pylori strain used here, SS1 (33), is cag-PAI positive, but for 6-μm tissue sections were incubated with monoclonal against CD4 unknown molecular reasons it does not deliver the effector protein fl – or rat IgG2a isotype control. For immuno uorescence, the following anti- CagA and uses cag-PAI independent mechanisms to evoke im- bodies were used sequentially: rat anti-mouse IL-17A (BD Pharmingen), Alexa mune responses (34, 35). Thus we did not examine cagA expression. Fluor 488 goat anti-rat IgG (Invitrogen), rat anti-mouse CD4 (BD Pharmingen), In conclusion, our data suggest a model in which H. pylori uses and TRITC donkey anti-rat IgG (Jackson ImmunoResearch Laboratories). chemotaxis to approach the gastric epithelial cells in such a way as to trigger apoptosis, and the combination of cell death with bac- Gastric Tissue Observation. Pathology. Gastric tissue preserved in OCT was terial products promotes the development of a Th17-skewed im- sectioned, stained with H&E, and evaluated in a blind fashion by a pathol- mune response, as these same signals do in C. rodentium infections ogist. Each slide was evaluated twice. Lymphocytic infiltration was scored as fi (24, 25). Our model fits well with previous H. pylori experiments outlined by Eaton et al. (36). The scores are 0, no in ltrate; 1, mild multifocal infiltration; 2 mild widespread infiltration; 3, mild widespread and moderate that demonstrated chemotaxis is needed for normal bacterial po- multifocal infiltration; 4, moderate widespread infiltration; and 5, moderate sitioning within the gastric epithelium (4, 6). Here we show that widespread and severe multifocal infiltration. appropriate localization allows H. pylori to increase VacA ex- Specific cellular infiltrate. To evaluate immune cell infiltration and colocali- pression (Fig. S5) and perhaps promote efficient delivery of this zation, sections were examined under 200× magnification (Nikon Eclipse and other proapoptotic virulence factors. The simultaneous de- E600 Microscope), and images of the tissue were captured with a Spot In- tection of the apoptotic cells and H. pylori bacterial ligands sight 4 camera (Diagnostic Instruments, Inc). The immunohistochemistry encourages innate immune cells to promote the Th17 cell-domi- images were subjected to analysis with Photoshop CS3 (Adobe), as described nant response that we observed in wild-type infections (Figs. 3 and in SI Materials and Methods. 4). We conclude that one function of the chemotaxis system is to drive H. pylori to interact with the host and induce a pathological Flow Cytometric Characterization of Cells/Intracellular Cytokine Staining. De- tailed methods used for flow cytometry and intracellular cytokine staining Th17 immune response that promotes chronic infection. are provided in SI Materials and Methods. In brief, for flow cytometry, single- cell suspensions of mouse stomach tissue were prepared as in ref. 17 and Materials and Methods then were stained for expression of CD3 and CD45 and CD4 or CD8. The H. pylori Strains and Growth Conditions. Helicobacter pylori strain SS1 (33) and optimal concentrations for the antibodies were determined in prior its isogenic mutants were used for all studies. SS1 was a gift of Jani O’Rourke experiments to be 2 μg/mL for Alexa Fluor 488-CD3 (eBioscience), PeCy7-CD4 (University of New South Wales, Sydney, Australia) and was minimally labo- (eBioscience), APC/Cy-7-CD8 (Biolegend), and for Alexa Fluor 647-CD45 ratory passaged before our experiments. SS1 ΔcheY::cat was created by (Biolegend). An isotype control was included. Before analysis the cells were replacing most of the cheY gene (HP1067) with the Campylobacter cat gene as fixed in 2% paraformaldehyde. Fifty thousand cells were counted on a BD described (5). H. pylori was cultured as described in SI Materials and Methods. LSR II (BD Biosciences) and analyzed using FlowJo software (BD Biosciences); results are presented as a percentage of the 50,000 cells counted. A Mann– Animal Infections. The University of California, Santa Cruz Institutional Animal Whitney u test was used for statistical analysis. For intracellular cytokine Care and UseCommitteeapprovedallanimalprotocolsand experiments.Female staining, gastric cells in a single-cell suspension were stained with Alexa C57BL/6N mice (Helicobacter-free; Taconic Labs) were housed at the University Fluor 647-CD45 and propidium iodide and then were sorted on the BD of California, Santa Cruz animal facility. Mice were 6–8wkoldatthetimeof FACSAria cell sorter (BD Biosciences) for CD45-positive/ propidium iodide- H. pylori infection, and age-matched uninfected mice were included in all negative cells. The entire stomach from each mouse was sorted, resulting in experiments. Animals were intragastrically inoculated orally via a 20-gauge × different yields depending on infection type. After overnight incubation in 1.5 inch feeding needle with 500 μL containing ∼1 × 107 cfu/mL Brucella broth- CellGro DMEM plus 10% FBS (GIBCO), 1× penicillin/streptomycin/glutamine

Rolig et al. PNAS Early Edition | 5of6 Downloaded by guest on September 27, 2021 (GIBCO), 1 M Hepes (Fisher), and 1× nonessential solution (Invi- TUNEL Assay. Samples used for the TUNEL assay were prepared from 14-μm- trogen), cells were stimulated for 4 h with the Leukocyte Activation Mixture thick OCT-frozen sections mounted on glass slides (SuperFrost/Plus; Fisher). fi (BD Biosciences) in a tissue culture CO2 incubator at 37 °C. Surface staining Sections were xed in 3.7% buffered formaldehyde (Fisher) and then were was performed with Alexa Fluor 488-CD3 (eBioscience), PeCy7-CD4 (eBio- stained using the TACS 2 TdT-DAB In Situ Apoptosis Detection Kit (Trevigen) science), and Alexa Fluor6 47-CD45 (Biolegend). After surface staining, cells following the manufacturer’s protocol. Tissue was counterstained with were resuspended in a fixation/permeabilization solution kit (BD Cytofix/ methyl green (Trevigen). To analyze apoptotic cells, numbers of positively cytoperm; BD Biosciences), and intracellular staining was performed with PE- stained cells were counted in 20 well-oriented glands in the corpus from six − IL17A (BD Pharmingen) following the manufacturer’s protocol. mice infected with wild-type H. pylori, six mice infected with Che H. pylori, and three uninfected mice. Student’s t test was used for statistical analysis. qPCR. Quantitative analysis of the cytokines in the mouse stomach was per- formed by real-time PCR. RNA was isolated from mouse stomach samples using ACKNOWLEDGMENTS. We thank Susan Williams and Victoria Auerbuch the TRIzol RNA isolation protocol (GIBCO) and was converted to cDNA using Stone for comments on the manuscript; Martha Zuniga for numerous dis- fl SuperScript III First Strand Synthesis System (Invitrogen). The cDNA was mea- cussions; Holly Scott Algood for advice about gastric sample ow cytometry; anonymous reviewers for insightful experimental suggestions; and Amber sured in a qPCR reaction with SYBR green master mix (SABiosciences). Primers ΔΔ Kofman for assistance with immunohistochemistry. We acknowledge the used for qPCR are given in Table S1. Mammalian data were analyzed by the technical support received from Bari Holm Nazario, California Institute for threshold cycle(Ct) method, as described in SI Materials and Methods.All Regenerative Medicine Shared Stem Cell Facility. This project was supported samples were analyzed in triplicate, along with no-reverse transcriptase con- by National Institute of and Infectious Diseases at the National Insti- trols. The Mann–Whitney u test was used for statistical analysis. tutes of Health Grant AI050000 (to K.M.O.).

1. Peek RM, Jr., Fiske C, Wilson KT (2010) Role of innate immunity in Helicobacter pylori- 18. O’Garra A, Vieira P (2007) T(H)1 cells control themselves by producing -10. induced gastric malignancy. Physiol Rev 90:831–858. Nat Rev Immunol 7:425–428. 2. Polk DB, Peek RM, Jr. (2010) Helicobacter pylori: Gastric and beyond. Nat Rev 19. McGeachy MJ, et al. (2007) TGF-beta and IL-6 drive the production of IL-17 and IL-10 Cancer 10:403–414. by T cells and restrain T(H)-17 cell-mediated pathology. Nat Immunol 8:1390–1397. 3. Cover TL, Blanke SR (2005) Helicobacter pylori VacA, a paradigm for toxin multi- 20. Do JS, Visperas A, Dong C, Baldwin WM, 3rd, Min B; Do J-su (2011) Cutting edge: functionality. Nat Rev Microbiol 3:320–332. Generation of colitogenic Th17 CD4 T cells is enhanced by IL-17+ γδ T cells. J Immunol 4. Williams SM, et al. (2007) Helicobacter pylori chemotaxis modulates inflammation 186:4546–4550. and bacterium-gastric epithelium interactions in infected mice. Infect Immun 75: 21. Sawa S, et al. (2010) Lineage relationship analysis of RORgammat+ innate lymphoid – 3747–3757. cells. Science 330:665 669. 5. Terry K, Williams SM, Connolly L, Ottemann KM (2005) Chemotaxis plays multiple 22. Buonocore S, et al. (2010) Innate lymphoid cells drive interleukin-23-dependent in- – roles during Helicobacter pylori animal infection. Infect Immun 73:803–811. nate intestinal pathology. Nature 464:1371 1375. 6. Howitt MR, et al. (2011) ChePep controls Helicobacter pylori infection of the gastric 23. Symonds EL, et al. (2009) Involvement of T helper type 17 and regulatory T cell activity fl glands and chemotaxis in the Epsilonproteobacteria. mBio 2(4):1–10. in Citrobacter rodentium invasion and in ammatory damage. Clin Exp Immunol 157: – 7. Soehnlein O, Lindbom L (2010) partnership during the onset and resolu- 148 154. tion of inflammation. Nat Rev Immunol 10:427–439. 24. Torchinsky MB, Garaude J, Martin AP, Blander JM (2009) Innate immune recognition – 8. Wilson KT, Crabtree JE (2007) Immunology of Helicobacter pylori: Insights into the of infected apoptotic cells directs T(H)17 cell differentiation. Nature 458:78 82. 25. Brereton CF, Blander JM (2011) The unexpected link between infection-induced ap- failure of the immune response and perspectives on vaccine studies. Gastroenterology optosis and a TH17 immune response. J Leukoc Biol 89:565–576. 133:288–308. 26. Borenshtein D, McBee ME, Schauer DB (2008) Utility of the Citrobacter rodentium 9. Kao JY, et al. (2010) Helicobacter pylori immune escape is mediated by dendritic infection model in laboratory mice. Curr Opin Gastroenterol 24:32–37. cell-induced Treg skewing and Th17 suppression in mice. Gastroenterology 138: 27. Deng W, et al. (2004) Dissecting virulence: Systematic and functional analyses of 1046–1054. a pathogenicity island. Proc Natl Acad Sci USA 101:3597–3602. 10. Khamri W, et al. (2010) Helicobacter pylori stimulates dendritic cells to induce in- 28. Fan X, et al. (2000) Helicobacter pylori urease binds to class II MHC on gastric epi- terleukin-17 expression from CD4+ T lymphocytes. Infect Immun 78:845–853. thelial cells and induces their apoptosis. J Immunol 165:1918–1924. 11. Luzza F, et al. (2000) Up-regulation of IL-17 is associated with bioactive IL-8 expression 29. Shibayama K, et al. (2003) A novel apoptosis-inducing protein from Helicobacter in Helicobacter pylori-infected human gastric mucosa. J Immunol 165:5332–5337. pylori. Mol Microbiol 47:443–451. 12. Shi Y, et al. (2010) Helicobacter pylori-induced Th17 responses modulate Th1 cell 30. Wang J, et al. (2000) Helicobacter pylori modulates lymphoepithelial cell interactions fi responses, bene t bacterial growth, and contribute to pathology in mice. J Immunol leading to epithelial cell damage through Fas/Fas ligand interactions. Infect Immun – 184:5121 5129. 68:4303–4311. 13. Littman DR, Rudensky AY (2010) Th17 and regulatory T cells in mediating and re- 31. Lee SH, Butler SM, Camilli A (2001) Selection for in vivo regulators of bacterial viru- fl – straining in ammation. Cell 140:845 858. lence. Proc Natl Acad Sci USA 98:6889–6894. fi 14. Ivanov II, et al. (2009) Induction of intestinal Th17 cells by segmented lamentous 32. Loh JT, Gupta SS, Friedman DB, Krezel AM, Cover TL (2010) Analysis of protein ex- – bacteria. Cell 139:485 498. pression regulated by the Helicobacter pylori ArsRS two-component signal trans- fl fi 15. McGee DJ, et al. (2005) Colonization and in ammation de ciencies in Mongolian duction system. J Bacteriol 192:2034–2043. gerbils infected by Helicobacter pylori chemotaxis mutants. Infect Immun 73: 33. Lee A, et al. (1997) A standardized mouse model of Helicobacter pylori infection: 1820–1827. Introducing the Sydney strain. Gastroenterology 112:1386–1397. 16. Beier D, Spohn G, Rappuoli R, Scarlato V (1997) Identification and characterization of 34. Arnold IC, et al. (2011) Tolerance rather than immunity protects from Helicobacter an operon of Helicobacter pylori that is involved in motility and stress adaptation. pylori-induced gastric preneoplasia. Gastroenterology 140:199–209. J Bacteriol 179:4676–4683. 35. Eaton KA, et al. (2001) Role of Helicobacter pylori cag region genes in colonization 17. Algood HMS, Gallo-Romero J, Wilson KT, Peek RM, Jr., Cover TL (2007) Host response and gastritis in two animal models. Infect Immun 69:2902–2908. to Helicobacter pylori infection before initiation of the adaptive immune response. 36. Eaton KA, Radin MJ, Krakowka S (1995) An animal model of gastric ulcer due to FEMS Immunol Med Microbiol 51:577–586. bacterial gastritis in mice. Vet Pathol 32:489–497.

6of6 | www.pnas.org/cgi/doi/10.1073/pnas.1104598108 Rolig et al. Downloaded by guest on September 27, 2021