Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Nov 15.
Published in final edited form as: Immunity. 2021 Oct 28;54(11):2547–2564.e7. doi: 10.1016/j.immuni.2021.10.002

Commensal Cryptosporidium colonization elicits a cDC1-dependent Th1 response that promotes intestinal homeostasis and restricts other infections

Emilie V Russler-Germain 1, Jisun Jung 1, Aidan T Miller 1, Shannon Young 1, Jaeu Yi 1, Alec Wehmeier 1, Lindsey E Fox 1, Kristen J Monte 1, Jiani N Chai 1, Devesha H Kulkarni 2, Lisa J Funkhouser-Jones 3, Georgia Wilke 3, Vivek Durai 4, Bernd H Zinselmeyer 4, Rafael S Czepielewski 4, Suellen Greco 5, Kenneth M Murphy 4, Rodney D Newberry 2, L David Sibley 3,*, Chyi-Song Hsieh 1,*,
PMCID: PMC8716016  NIHMSID: NIHMS1749093  PMID: 34715017

Summary

Cryptosporidium can cause severe diarrhea and morbidity, but many infections are asymptomatic. Here, we studied the immune response to a commensal strain of Cryptosporidium tyzzeri (Ct-STL) serendipitously discovered when conventional type 1 dendritic cell (cDC1)-deficient mice developed cryptosporidiosis. Ct-STL was vertically transmitted without negative health effects in wild-type mice. Yet, Ct-STL provoked profound changes in the intestinal immune system, including induction of an IFNγ-producing Th1 response. T cell receptor sequencing coupled with in vitro and in vivo analysis of common Th1 TCRs revealed that Ct-STL elicited a dominant antigen-specific Th1 response. By contrast, deficiency in cDC1s skewed the Ct-STL CD4 T cell response toward Th17 and regulatory T cells. Although Ct-STL predominantly colonized the small intestine, colon Th1 responses were enhanced and associated with protection against Citrobacter rodentium infection and exacerbation of dextran sodium sulfate and anti-IL10R-triggered colitis. Thus, Ct-STL represents a commensal pathobiont that elicits Th1-mediated intestinal homeostasis that may reflect asymptomatic human Cryptosporidium infection.

eTOC Blurb

Cryptosporidium are protozoan parasites that cause gastrointestinal illness in humans and animals worldwide. We identified a commensal strain of Cryptosporidium tyzzeri endemic in our mouse colony. This strain of Cryptosporidium does not cause disease in wild-type mice but changes the composition and function of the intestinal immune system.

Introduction

Cryptosporidium species are environmentally ubiquitous protozoan parasites that infect gastrointestinal epithelial cells of a broad range of animals (Fayer et al., 2010). Humans are mainly infected by Cryptosporidium hominis (Morgan-Ryan et al., 2002) and Cryptosporidium parvum (Cp) (Upton and Current, 1985). Infection starts with ingestion of oocysts, which contain 4 sporozoites that invade enterocytes and form parasitophorous vacuoles just below the apical enterocyte membrane. Asexual and then sexual development within enterocytes culminates in the generation of thick-walled, environmentally hardy oocysts that are excreted into the lumen to facilitate fecal-oral transmission.

Cryptosporidium is the fourth leading cause of death globally in children under the age of 5 (12.1%) (Collaborators, 2017), and is the second or third cause of diarrhea in in young children in Southeast Asia and Africa (Kotloff et al., 2019). Within the United States there are ~750,000 cases of cryptosporidiosis annually (Gharpure et al., 2019; Scallan et al., 2011). Cryptosporidiosis is also one of the original opportunistic infections characteristic of AIDS (Current et al., 1983). Moreover, therapies for cryptosporidiosis are limited, with only a single FDA-approved drug (nitazoxanide) that is ineffective in immunocompromised individuals and not approved in young children (Checkley et al., 2015). One issue for controlling transmission is that oocysts are resistant to bleach and other common disinfectants (Checkley et al., 2015). Thus, Cryptosporidium infection of humans remains an important health problem.

For many years, Cryptosporidium was thought to be a commensal (Flanigan and Soave, 1993). Cryptosporidium is now studied primarily as a pathogen due to its ability to cause human disease. One limitation of these studies is that mouse models typically rely on Cp, which naturally infects cows and humans but not wild-type (WT) mice (Upton and Current, 1985), thus requiring studies in immunocompromised mice such as IFNγ signaling-deficient (Chen et al., 1993a) or neonatal mice (Urban et al., 1996). A mouse-tropic species, Cryptosporidium tyzzeri (Ct, formerly mouse genotype I) (Morgan et al., 1999; Ren et al., 2012), infects the jejunum and ileum in mice and is closely related to Cp (Ren et al., 2012). Infection in wild mice is common with a worldwide distribution (Condlova et al., 2019; Osman et al., 2017b), and is reported to affect 2% of lab mice in China (Lv et al., 2009). Adult mice shed Ct oocysts for 24–28 days post colonization, after which oocysts become undetectable. However, the mice remain infected as steroid administration at day 65 elicits oocyst shedding (Ren et al., 2012). A different strain of Ct isolated from wild mice from a farm (Sateriale et al., 2019) also infects adult C57BL/6 mice, which shed oocysts for up to 3 weeks. In this case, the oocysts are no longer detectable by qPCR after the initial infection.

We serendipitously discovered that our laboratory’s mouse colony at Washington University in St. Louis has likely been colonized for years with a strain of Ct (Ct-STL) when conventional type 1 dendritic cell (cDC1)-deficient mice developed spontaneous cryptosporidiosis. Notably, the perspective of our study is different than studies that assessed Ct as an infection (Ren et al., 2012; Sateriale et al., 2019). In contrast to these reports, we found that Ct-STL acted like a commensal pathobiont, undergoing vertical transmission with colonization at weaning and oocyst shedding that persisted for the lifetime of the mouse without immunosuppression. As Cryptosporidium was detected in ~7% of animal rooms sampled in our building, this suggests that Ct may be common yet remain undetected in routine analyses of the microbiome given that such analyses tend to focus on prokaryotes through 16S rRNA sequencing. Thus, the immune system effects of Ct colonization may represent a major hidden variable in the study of gastrointestinal immune function.

Results

cDC1-deficient mice develop fulminant cryptosporidiosis

During the course of our studies of host:commensal interactions, we utilized Irf8 +32 5’−/− mice that have a 149 base pair enhancer deletion in Irf8 that efficiently eliminates cDC1s (Durai et al., 2019). Binding of BATF3 to this enhancer induces IRF8 expression in cDC1s, but does not affect IRF8 expression in other cell lineages. We bred Irf8 +32 5’−/− mice to human Langerin-DTA (hLangDTA) mice, which express diphtheria toxin (DTA) under the control of the human langerin promoter, to also delete CD103+ CD11b+ (double positive, DP) cDC2s in the mesenteric lymph nodes (MLN) (Welty et al., 2013). As neither of these cDC-subset-deficient strains were reported to develop spontaneous disease, we were surprised to observe that approximately 10% of homozygous Irf8 +32 5’−/− mice failed to thrive and died between 5–20 weeks of age. hLangDTA Irf8 +32 5’−/− mice, which lack most CD103+ cDCs (Figure S1A), seemed more prone to this spontaneous illness, although still with variable penetrance.

Necropsy analysis of a sick hLangDTA Irf8 +32 5’−/− mouse revealed pancytopenia with low white blood cells (WBC), red blood cells (RBC), and platelets, as well as small secondary lymphoid organs and hyposplenism (Figure 1AB). However, bone marrow histology only showed mild hypocellularity with reduced megakaryocytes (Figure 1C), which would not account for the profound pancytopenia observed. Notably, small intestine (SI) histology was pathognomonic for cryptosporidiosis (Figure 1C). Laboratory values such as low total protein were consistent with malabsorption secondary to cryptosporidiosis, which could also explain pancytopenia (Figure 1A). Thus, the presumptive cause of death of sick cDC1-deficient mice in our colony was fulminant cryptosporidiosis.

Figure 1. cDC1-deficient mice develop fulminant cryptosporidiosis.

Figure 1.

(A) Complete blood cell counts, total protein, and glucose from peripheral blood of a spontaneously sick 7-week-old cDC1-deficient mouse (hLangDTA Irf8 +32 5’−/−) compared with a non-sick littermate (Irf8 +32 5’−/−). (B) Spleen size of a spontaneously sick 6-week-old cDC1-deficient mouse (hLangDTA Irf8 +32 5’−/−) compared with a non-sick littermate. (C) Bone marrow and SI histology from mice in (A). Arrowheads indicate Cryptosporidium parasites in the SI epithelium. (D) Alignment and clustering of Ct-STL Gapdh sequence with Gapdh sequences from other Cryptosporidium species (left). The number of nucleotide differences in Gapdh between Cp-AUCP-1, Ct-UGA55, and Ct-STL are shown (right).

Ct-STL behaves like a commensal in WT mice

To characterize the species of Cryptosporidium in these mice, we analyzed parasite Gapdh (Huang et al., 2004) and Gp60 genes (Xiao, 2010) by PCR amplification from fecal DNA. Gapdh sequencing revealed a high degree of similarity with Ct-UGA55 versus Cp-AUCP-1 (Sateriale et al., 2019; Wilke et al., 2019) (Figure 1D). Parasite Gp60 also showed the greatest similarity with Ct amongst the Cryptosporidium genus (Figure S1B). Based on these genetic similarities, we refer to the strain in our St. Louis colony as Ct-STL.

We suspected that Ct-STL originated from WT Foxp3IRES-GFP mice bred in our colony for a decade. To quantify oocyst shedding, we performed flow cytometry (Barbosa et al., 2008) using a commercial antibody used to detect Cryptosporidium in human clinical samples (Crypt-a-Glo), which we found cross reacts with Ct-STL (Figure S2A). We found that our WT mice shed Ct-STL oocysts, while mice recently purchased from Charles River (CR) or Jackson (JAX) Laboratories showed no evidence of Cryptosporidium, consistent with monitoring and exclusion of this organism in their facilities (Figure 2A). Histological analysis of jejunums from our WT mice revealed characteristic signs of Cryptosporidium parasites in the brush border of epithelial cells (Figure 2B). Some strains in our colony were negative for Ct-STL, suggesting that it does not always spread between cages within a barrier facility. Cryptosporidium was not unique to our colony, however, as a PCR survey of air dust swabbed from the exhaust plenums of individually ventilated cage racks revealed that 7 out of 101 rooms in our facility were positive. Although we cannot ascertain when we acquired Ct-STL, our impression based on these data is that Ct-STL has been endemic in our colony for many years.

Figure 2. Ct-STL behaves like a commensal in WT mice.

Figure 2.

(A) Quantification of Ct-STL oocysts in terminal pellets by flow cytometry (FC) using Crypto-a-Glo of a Cryptosporidium-free Charles River (CR) vs a WT mouse from our colony (STL Wild-Type). 105 total events are displayed on the left; oocysts in the gate represent total oocysts in 105 stool events. (B) Ct-STL parasites within the jejunum brush border (arrows) in STL WT mice by histology. (C) Ct-STL quantification by FC and qPCR of stool DNA in whole luminal colon contents from STL WT mice before and after weaning (14–21 days and 3–6 weeks old, respectively). FC and qPCR samples were from different mice. Dotted line in FC represents the limit of specific detection determined using Cryptosporidium-free mice (expt.=2). (D) Ct-STL quantification by FC and qPCR in the terminal pellets of 9–13 month old mice with JAX or CR microbiota versus similarly-aged STL WT mice. FC and qPCR samples were from identical mice (expt.=2). (E) FC quantification of Ct-STL in terminal pellets of WT CR mice colonized at 3.5 weeks of age with 250, 4×103, or 2.5×104 Ct-STL oocysts (expt.=3). (F) Normalized (left) and absolute (right) weights of CR mice colonized with 4×103 Ct-STL oocysts at 3.5 weeks of age (expt.=2). (G) Fecundity of Ct-STL colonized mice. Littermate CR mice were gavaged with 4×103 Ct-STL oocysts or PBS (control) at 3.5 weeks of age and set up as breeding pairs at 6 weeks of age. Number of litters per breeding pair (far left), pups born per litter (mid left), number of pups dead or alive at 21 days of life (mid right), and pup weights at 21 days (far right) were quantified over 6 months (expt.=5). (H) Fecal water weight of terminal pellets (left), transit velocity of gavaged dye (mid), and total gastrointestinal (GI) transit time (right) in Ct-STL colonized vs. control CR mice. WT CR mice were gavaged with 4×103 Ct-STL oocysts or PBS (control) at 3.5 weeks old and mice were assessed after 4 weeks (expt.=2). Each dot represents data from an individual host. Geometric mean ± geometric S.D. (C–E) or mean ± S.E.M. (F right, G, H) are shown. Student’s t-test (C, D, F right, G, H), repeated measures ANOVA (E, F left), or Fisher’s exact test (G mid right) p-values are shown.

The presence of Ct-STL in multiple strains implies that it is vertically transmitted and maintained in the colony. To determine at what point during ontogeny WT mice become naturally colonized with Ct-STL, we compared oocyst numbers in the colon luminal contents of mice pre-weaning (14–21 days old) and post-weaning (3–6 weeks old) by flow cytometry. We also utilized Ct-STL Gapdh qPCR from fecal DNA to improve sensitivity of detection, with the caveat that this is not specific for DNA within oocysts. These analyses revealed that oocyst shedding became easily detectable around weaning (Figure 2C), likely via the fecal-oral route. Additionally, 9- to 13-month-old WT mice from our colony shed oocysts, albeit at low levels compared to younger mice (Figure 2D). These data are consistent with our conclusion that Ct-STL has been present in our colony for years, as transient infection would limit opportunities for vertical and horizontal transmission and likely lead to elimination from our colony.

To determine the infectious dose of Ct-STL, we generated a Ct-STL oocyst preparation free of bacterial constituents by treating cecal and colon contents from infected mice with 40% bleach, which rendered aerobic bacterial colony forming units (CFUs) undetectable. In contrast to Cp, which requires upward of 106 oocysts to cause transient infection of WT mice (Theodos et al., 1997), transfer of as few as 250 Ct-STL oocysts at weaning infected WT mice (Figures 2E and S2B). In fact, 250, 4×103, and 2.5×104 oocysts all produced similar levels of colonization over time as measured by oocyst shedding, which remained stable for many weeks before decreasing at week 12 post-infection (Figures 2E and S2B). However, qPCR analysis revealed an inverse relationship between initial Ct-STL dose and Ct-STL DNA in stool at 12 weeks (Figure S2B), suggesting that large inoculations may trigger increased sterilizing immunity. Based on these data, we transferred 4×103 oocysts in Cryptosporidium-free mice at weaning to mimic homeostatic vertical infection unless otherwise noted. FACS-purified oocysts from this preparation also transferred Ct-STL, suggesting that oocysts alone were sufficient for colonization (data not shown). These data show that Ct-STL has a low infectious dose in WT mice and persists over 3 months, consistent with observations from naturally-colonized mice.

We next assessed whether Ct-STL colonization affected the overall health of the animal. Colonization with Ct-STL at weaning did not affect weight gain over 7 weeks (Figure 2F). 1 year after colonization, Ct-STL was still detected in the stool of these mice (Figure S2C). Colonization with Ct-STL also did not affect fecundity or cause signs of diarrhea or increased intestinal permeability (Figures 2GH and S2D). Therefore, in contrast to other Ct strains (Ren et al., 2012; Sateriale et al., 2019), Ct-STL acts less like a pathogen and more like a commensal.

Both innate and adaptive immunity are required to limit Ct-STL

IFNγ signaling is important in the control of Cp in mice (Chen et al., 1993b; Pollok et al., 2001; Tessema et al., 2009). Consistent with these observations, we found that anti-IFNγ antibody treatment increased Ct-STL oocyst shedding by 2 weeks, an effect also seen with Cp (Chen et al., 1993a) (Figure S3A). Additionally, Ifngr1−/− mice lost weight starting 1–2 weeks after Ct-STL colonization, with some mice succumbing to infection over the 4 week observation period (Figure S3B). Taken together, these data support a pivotal role for IFNγ signaling for controlling Ct-STL.

Multiple innate and adaptive cell types secrete IFNγ, including NK, ILC1, NKT, CD4+ T, and CD8+ T cells (Cortez and Colonna, 2016; Schoenborn and Wilson, 2007). Cryptosporidium control requires adaptive immunity (Current et al., 1983; Sateriale et al., 2019; Tandel et al., 2019). To assess whether T and B cells are necessary for the control of Ct-STL, we colonized Rag1+/− and Rag1−/− littermates with Ct-STL and tracked stool oocysts. Differences in oocyst shedding began at 3–4 weeks post-colonization, when oocyst shedding decreased in Rag1+/− mice (Figures 3A and S3C). Notably, Rag1−/− mice colonized with Ct-STL did not appear ill or lose weight (Figure 3B). Histologic analysis of jejunums revealed that Rag1−/− mice had decreased villus height and villus:crypt length ratio indicative of crypt hyperplasia, and a trend toward more Ct-STL organisms per villus than Rag1+/− littermates (Figures 3C and S3D). Thus, these data from mice deficient in T and B cells highlight the low pathogenicity of Ct-STL and confirm that adaptive immunity is required to limit Ct-STL infection.

Figure 3. Both innate and adaptive immunity are required to limit Ct-STL.

Figure 3.

(A-C) Ct-STL oocyst shedding in terminal pellets by FC (A), weight loss (B), and jejunum histology at 13 weeks (C, arrows indicate Ct-STL parasites) in littermate Rag1+/− and Rag1−/− mice after colonization with 4×103 Ct-STL oocysts at 3 weeks of age (expt.=2). Weights were normalized to starting weights. (D) Quantification of Ct-STL oocysts by FC after depletion of innate immune cells with anti-NK1.1 and anti-Thy1.2 or isotype control treatment in Rag1−/− mice colonized with Ct-STL at weaning (expt=2). (E) IL-12p40YFP expression in SILP cDC (MHCII+ CD11c+ F4/80) subsets 1 week after colonization with Ct-STL. DP = double positive (CD103+ CD11b+), SP = single positive (expt.=2). (F) Expression of IFNγYFP IL17AGFP Foxp3IRES-Thy1.1 in SILP CD4+ T cells 3 weeks after colonization with Ct-STL (expt.=3). Each dot represents data from an individual host. Geometric mean ± geometric S.D. (A, D) or mean ± S.E.M. (B, C, E, F) are shown. P-values from Student’s t-test (C, F), ANOVA with Sidak’s correction (E), repeated measures ANOVA (A, D), or repeated measures mixed effects analysis (B), are shown.

As Rag1−/− mice only showed a defect in late control of Ct-STL colonization, we wondered whether components of innate immunity also participate in control of Ct-STL. We depleted NK and ILC cells with weekly anti-NK1.1 and anti-Thy1.2 antibodies, respectively. Rag1−/− mice shed significantly more Ct-STL oocysts with depletion of innate lymphocytes (Figure 3D). Altogether, these data highlight the critical role of type I immunity from both innate and adaptive immune cells for the control of Ct-STL.

To better understand the requirement for cDC1s to prevent fulminant cryptosporidiosis, we asked whether Ct-STL colonization alters cDC subsets. At 1 week post-colonization, cDC1 frequencies were unchanged and the percentage of MLN migratory DP cDC2s was slightly decreased (Figure S3E). We also asked whether Ct-STL induces a signal for Th1 development, IL-12 (Hsieh et al., 1993) using Cryptosporidium-free IL-12p40YFP mice. At 1 week post-colonization, IL-12p40YFP was selectively induced in a small fraction of cDC1s in the small intestine lamina propria (SILP) (Figures 3E and S3F). IL-12p40YFP was not enhanced in cDC1s in the MLN, although the baseline frequency of expression was substantial (Figure S3F). Macrophages also secrete IL-12 (Cooper and Khader, 2007), but induction of IL-12p40YFP expression was not observed in SILP macrophages (Figure S3F). The observation that IL-12p40 was only induced in cDC1s provides an explanation for why cDC1-deficient mice were susceptible to Ct-STL (Figure 1).

The induction of IL-12p40 in cDC1s suggests that Ct-STL induces an adaptive Th1 response, consistent with responses to other Cryptosporidium (Urban et al., 1996). Colonization of Cryptosporidium-free IFNγYFP IL17AGFP Foxp3IRES-Thy1.1 triple reporter mice with Ct-STL revealed increased frequency and mean fluorescent intensity (MFI) of IFNγYFP expression in CD4+ T cells in the SILP (Figure 3F) with similar trends in the proximal MLN (pMLN, all but distal MLN; Figure S3G). Additionally, the increase in CD4+ T cell IFNγ corresponded with a loss of IL-17AGFP+ Th17, but not Foxp3IRES-Thy1.1+ regulatory T (Treg) cells (Figures 3F and S3G). Consistent with an adaptive immune response to Ct-STL, increased CD4+ T cell numbers were also observed in the SILP (Figure S3G). CD8+ T cells play a role in Cp clearance in mice, although not as important a role as CD4+ T cells (McDonald et al., 1994). While CD3ε + CD4T cells, which included both CD8+ T cells but also a small fraction of other T cell subsets such as γδ T cells, were increased in numbers in the SILP by Ct-STL colonization, Ct-STL induced a decrease in IL17AGFP but no change in IFNγYFP expression in CD4 T cells (Figures S3HI). Thus, in contrast to other commensals, Ct-STL elicits a strong adaptive Th1 response.

Ct-STL induces an antigen-specific Th1 response

T cell responses after Ct-STL colonization could be specific to Cryptosporidium vs. other gut antigens (Hand et al., 2012). To address the question regarding TCR specificity, we analyzed the T cell receptor α (TCRα) repertoire (Table S1) of fixed TCRβ transgenic mice treated with anti-IFNγ to increase parasite burden (Figure S3A). Here, we used CXCR3 as a surrogate for Tbet-expressing Th1 cells in the MLN (Groom, 2019). A unique TCR was designated by its TRAV designation and CDR3 amino acid sequence.

Comparison of TCRα repertoires using non-metric multidimensional scaling (NMDS) of Bray-Curtis distances revealed that the effector T (Teff) cell repertoires in the MLN and SILP underwent substantial changes with Ct-STL infection in comparison with the Treg TCR repertoire (Figure 4A). The differences in TCR repertoire were greater between Treg and Teff cells (axis 1 of NMDS) than the effect of Ct-STL (axis 2 of NMDS), suggesting that convergence of the Treg and Teff TCR repertoires does not occur, in contrast to DSS plus anti-IL-10R induced colitis (Chai et al., 2017). The top Teff clones in Ct-STL infected mice in the presence of anti-IFNγ were virtually absent in control mice (Figures 4B and S4A, Table S2). By contrast, the most abundant Treg TCRs in Ct-STL mice were present at relatively high frequencies in control mice.

Figure 4. Ct-STL induces an antigen-specific Th1 response.

Figure 4.

(A-C) CD4+ T cell TCR repertoire changes in anti-IFNγ-treated Ct-STL colonized mice. Fixed TCRβ Foxp3GFP Tcra+/− mice were gavaged with 2.5–5×104 Ct-STL oocysts or PBS and treated weekly with anti-IFNγ to increase Ct-STL burden. TCRα chain sequencing was performed on CD4+ Teff (CD62Llo CD44hi Foxp3) and Treg (Foxp3+) populations after 2–3 weeks (expt.=3). (A) Bray-Curtis NMDS analysis of TCRα sequences from the MLN (top) or SILP (bottom). (B) Heatmaps representing the mean frequencies of the top 10 Teff (top) or Treg (bottom) TCRs in the SILP in Ct-STL colonized mice. TCRs selected for functional studies are noted (RG1–3, LA3). RG1-like TCRs with TRAV differences are indicated by (RG1) (n=5 for each group). (C) TRAV and CDR3 amino acid sequences for Ct-STL specific TCRs are shown. (D) RG1 and RG2 TCR-transduced T cell hybridoma cells were cultured together with the indicated antigen presented by splenic Flt3L-induced CD11c+ cells. Anti-CD3 was used as a positive control. NFAT-GFP expression was analyzed after 40 hours (expt.=3). (E-H) In vivo analysis of RG1 TCR. 2×104 (E-G) or 105 (H) retroviral RG1-expressing TCli-αβ Foxp3IRES-GFP T cells were transferred into Ct-STL colonized mice and analyzed 1 week later (expt.=2). (E) CTV-labeled RG1 T cell expansion in mice colonized with Ct-STL for 3 days. (F) RG1 T cell expression of CXCR3, CD25, and Foxp3 in Ct-STL colonized mice for 3 days. CXCR3 and CD25 on host polyclonal MLN CD4+ T cells are also shown. (G) CTV-labeled RG1 T cell expansion and expression of CXCR3 and Foxp3 in mice colonized with Ct-STL for 4–6 weeks. (H) RG1 T cell expression of Tbet and FoxP3 transcription factor staining in mice colonized with Ct-STL for 3 days. Each dot represents data from an individual host (A, E-H) or mean of 2 in vitro co-cultures (D). Mean ± S.E.M. is shown. Student’s t-test (E-H) or ANOVA with Dunnett’s correction (D) p-values are shown.

Based on early data, we selected 2 clones, RG1 and RG2, for analysis of antigen specificity as they were found in high frequencies in Teff subsets (Figures 4BC, S4A). RG1 and RG2 TCRα clones were retrovirally transduced into hybridoma cells expressing the fixed TCRβ chain and an NFAT-GFP reporter for T cell activation (Chai et al., 2017; Lathrop et al., 2011). We found that both RG1 and RG2 were activated by Flt3L-induced DCs presenting SI epithelial cell preparations from Ct-STL infected mice as well as Cp oocysts (Figure 4D). The antigens recognized by these TCRs likely differ as RG1 preferentially reacted to the SI epithelial preparation, whereas RG2 exhibited stronger reactivity to the Cp oocysts, consistent with differences in the CDR3 amino acid composition of the TCRs tested (Figures 4CD). As controls, we observed that SI epithelial preparations from uninfected mice did not activate RG1 or RG2. Also, Ct-STL or Cp antigen preparations did not activate a Helicobacter-reactive CT6 hybridoma, arguing against antigen non-specific mechanisms of T cell activation. Later, we also discovered that RG3 was reactive to Cp oocysts and confirmed that all 3 TCRs recognized antigen via conventional MHC Class II presentation (Figure S4B). Taken together, these data demonstrate that 3 common Teff TCRs that expanded with Ct-STL infection in mice treated with anti-IFNγ are specific to Cryptosporidium antigens.

We next asked whether these TCRs react to Ct-STL in WT colonized mice at homeostasis. We retrovirally transduced RG1 or RG2 into TCliαβ T cells (Lathrop et al., 2011), which expressed the original TCRβ chain and an αβ-TCR reactive to an irrelevant foreign peptide (human CLIP). RG1 or RG2 transduced T cells transferred in vivo were barely recoverable from control mice but were readily detected in Ct-STL colonized mice and underwent extensive cell division based on Cell Trace Violet (CTV) dilution 1 week after transfer (Figures 4E and S4C). These data demonstrate that Ct-STL antigens are constitutively acquired by antigen presenting cells (APCs) and presented to CD4+ T cells during homeostatic colonization of WT mice.

We also asked if RG1 and RG2 T cells could develop into peripheral Treg (pTreg) cells in vivo under homeostatic conditions, akin to the adaptive T cell response to pathobionts of the Helicobacter genus (Chai et al., 2017; Xu et al., 2018). Although the process of retroviral transduction requires in vitro activation, we are able to maintain the ability of the transferred cells to undergo pTreg cell differentiation (Chai et al., 2017). We observed that neither RG1 nor RG2 differentiated into Treg cells in 1 week when transferred early or late after Ct-STL colonization, and instead increased expression of CXCR3 in the MLN, which is suggestive of Th1 development (Groom, 2019) (Figures 4FG and S4D). We confirmed that RG1 and RG2 T cells increased expression of Tbet and had minimal FoxP3 expression using intracellular staining (Figures 4H and S4EF). In summary, these data suggest that RG1 and RG2 TCRs undergo Th1 development to Ct-STL in vivo under homeostatic conditions.

Ct-STL colonization alters the small intestine TCR repertoire at homeostasis

As the above T cell studies utilized retroviral transduction or anti-IFNγ treatment, the results may not mimic naïve T cell differentiation and miss pTreg cell selection to Ct-STL. We therefore analyzed the TCRα repertoire at homeostasis using triple reporter IFNγYFP IL17AGFP Foxp3IRES-Thy1.1 mice. IL17AGFP+ TCR repertoires were not analyzed due to variably low cell numbers, as Ct-STL induces a loss of these cells (Figure 3F). NMDS of Bray-Curtis distances revealed that SILP Teff subsets, including Th1, and IFNγYFP− IL17AGFP− double negative (Teff-DN) cells, underwent shifts as a result of Ct-STL colonization (PERMANOVA p-values: SILP Th1=0.006, SILP Teff DN=0.106, SILP Treg=0.593, MLN Th1=0.149, MLN Teff-DN=0.078, MLN Treg=0.435) (Figures 4A, 5A). Treg TCR repertoires were less affected by Ct-STL (Figure S5A).

Figure 5. Ct-STL colonization alters the small intestine TCR repertoire at homeostasis.

Figure 5.

(A-D) CD4+ T cell TCR repertoire analysis of the SILP and MLN. Fixed TCRβ IFNγYFP IL17GFP Foxp3IRES-Thy1.1 Tcra+/− mice were colonized with Ct-STL at weaning for 3 weeks before sorting CD4+ CD62Llo CD44hi Th1 (Foxp3 IFNγYFP+ IL17GFP−), Teff-DN (Foxp3 IFNγYFP− IL17GFP−) and Treg (Foxp3+) populations (n=5–6, expt.=3 for SILP and MLN DN and Treg; n=4, expt.=2 for MLN Th1). (A) Bray-Curtis NMDS analysis of Teff TCRα sequences. Each dot indicates a T cell subset from an individual mouse. (B) Comparison of non-Cryptosporidium dependent TCR frequencies in control and Ct-STL colonized mice. Mean frequency of TCRs found in both control and Ct-STL data sets are plotted (see Methods). Each dot represents the mean frequency of an individual TCR that was found in at least 1 control mouse and at least one Ct-STL colonized mouse. (C) Identification of TCRs that distinguish between Ct-STL and control by machine learning. GBM was used to classify control vs Ct-STL using the data from Th1 and Teff-DN cells from the MLN, SILP, and colon (see Methods). Data shown are the average prediction values for each sample (left) and the average relative influence for each TCR. (D) Analysis of SILP top 10 Th1 and Treg TCRs by heatmap (left, average frequency) and frequency bar plot (right). RG3-like TCR with CDR3 amino acid difference is indicated by superscript. Each dot represents data from an individual host (A, D right) or individual TCR (B-C). Violin plot with median and quartiles (C left) or mean ± S.E.M (D right) are shown. Paired t-test (B) and Mann-Whitney U (C left) p-values are shown. N.D. = not detected.

To further assess the changes induced by Ct-STL colonization on specific T cell subsets, we compared the frequencies of TCR clones that were present in both control and Ct-STL colonized mice. SILP Th1, but not Teff-DN or Treg TCR, clones underwent substantial decreases in frequency in Ct-STL-colonized mice (Figure 5B). We interpret this to mean that the antigenic landscape for Teff-DN or Treg T cells are not as dramatically altered by Ct-STL, whereas the relative frequencies of Th1 cells that recognize non-Ct-STL antigens are diminished either by Ct-STL specific Th1 expansion or via alteration of cDC presentation of those antigens.

As another approach for identifying TCRs associated with Ct-STL infection, we used gradient boosted models (GBM), a tree based machine learning algorithm related to random forest. Using the effector TCR datasets (Teff-DN and Th1) from the SILP, MLN and colon lamina propria (cLP) at homeostasis, we optimized the parameters to achieve an AUC of 0.99 (Figure 5C). The TCRs with highest relative influence (Figure 5C) concur with the assessment that RG1, RG2, and RG3 are common TCRs that distinguish the homeostatic effector TCR repertoire during Ct-STL colonization.

We also looked at the top TCR clones by average frequency induced by Ct-STL colonization. The top 9 out of 10 Ct-STL SILP Th1 TCR clones, including RG1 and RG2, were absent in control mice and in Treg cells (Figures 5D and S5B, Table S3). By contrast, the majority of the top SILP Treg TCR clones in Ct-STL were also present in control mice (Figure 5D). Many of the top SILP Th1 TCRs were also found in the Teff-DN subset (Figures S5BC), suggesting that only a portion of T cells of the same specificity are actively synthesizing IFNγ, or that these TCRs are present in other effector T cell phenotypes. The top MLN Th1 and Teff-DN TCRs during homeostasis were not as uniformly absent in control mice (Figure S5D), indicating a more limited impact of Ct-STL in the MLN consistent with the NMDS (Figure 5A). TCR repertoire analysis of individual TCRs using DESeq2 also showed that RG1, RG2, and RG3 were mostly increased in the Th1 and Teff-DN but not Treg cell populations in Ct-STL-colonized versus control mice (padj < 0.05) (Figure S5E). Thus, Ct-STL colonization during homeostatic conditions elicits primarily a Th1-type effector, and not Treg, cell response, suggesting that pTreg cell selection is not an important mechanism by which host:commensal tolerance is achieved for Ct-STL.

cDC1s are important for Ct-STL control and Th1 differentiation

To confirm that the fulminant cryptosporidiosis observed in cDC1-deficient mice was due to Ct-STL, we rederived hLangDTA Irf8 +32 5’−/− mice to be Cryptosporidium-free via in vitro fertilization. cDC1-deficient mice colonized with Ct-STL at weaning shed increased oocysts (Figure 6A). Loss of DP cDC2s in hLangDTA mice did not impact oocyst shedding (Figure 6A). Thus, cDC1s, but not DP cDC2s, are required for control of Ct-STL.

Figure 6. cDC1s are important for Ct-STL control and Th1 differentiation.

Figure 6.

(A-C) Littermate cDC subset-deficient hLangDTA Irf8 +32 5’ mice were colonized at weaning with Ct-STL (expt.=4) and tracked for (A) oocyst shedding using flow cytometry, and (B) weight change. “X” symbols in (B, right) indicate mice that died between 7–12 weeks after Ct-STL colonization. (C) Histologic analysis of jejunums 13 weeks after Ct-STL colonization. (D-F) Analysis of polyclonal CD4+ T cells and Ct-STL reactive RG1-expressing T cells in the SILP of cDC subset-deficient mice (expt.=2). 105 retrovirally-transduced RG1 T cells were transferred 3 days after Ct-STL colonization, and assessed after 1 week for transcription factor expression by intracellular staining. FACS plots are representative of high FoxP3 and RORγt mice. Each dot represents data from an individual host. Geometric mean ± geometric S.D. (A) or mean ± S.E.M. are shown. Repeated measures mixed effects analysis (A, B left) or ANOVA with Dunnett’s correction (B right, C-F) p-values are shown.

We wondered whether colonization of rederived cDC1-deficient mice would recapitulate the disease we observed in cDC1-deficient mice in our colony, partially fulfilling Koch’s postulates. Over the course of 12 weeks after Ct-STL colonization at weaning, 2/20 (10%) Irf8 +32 5’−/− mice vs 0/15 Irf8 +32 5’+/− mice perished. Weight was maintained in the majority of cDC1-deficient mice, suggesting that fulminant cryptosporidiosis was a stochastic event – consistent with our initial observations in Irf8 +32 5’−/− mice (Figures 1 and 6B). Rederived cDC1-deficient mice that were not colonized with Ct-STL showed no signs of illness and did not die prematurely (data not shown). Histologic analysis of surviving cDC-deficient mice showed cDC1-deficient mice had significantly more Ct-STL organisms per villus and higher epithelial scores due to villus epithelial dysplasia but otherwise comparable villus and crypt properties (Figures 6C and S6A). From these observations, we conclude that the morbidity and mortality observed in a subset of our original cDC1-deficient mice was due to fulminant cryptosporidiosis from Ct-STL.

Altered T cell development in cDC1-deficient mice is one explanation for the severity of Ct-STL-mediated disease observed in Irf8 +32 5’−/− mice relative to Rag1−/− mice. For example, genetic regulation of Th1 vs Th2 responses results in dramatically different outcomes to Leishmania major infection (Sadick et al., 1990; Scott et al., 1988). We therefore analyzed RG1 after transfer into Ct-STL colonized, cDC-deficient mice. RG1 recovery was increased in the SILP of cDC1-deficient mice, consistent with increased Ct-STL antigen burden (Figure S6B). Notably, the frequency of RG1 FoxP3+ Treg cells was increased in 4/10 cDC1-deficient mice (Figure 6D and S6C). Additionally, Tbet+ Th1 generation of RG1 was diminished in cDC1-deficient mice, which was associated with skewing toward other Teff subsets: RORγt+ Th17 and GATA3+ Th2 cells (Figures 6EF and S7DE). With the exception of Treg cells in the MLN, RG1 differentiation mirrored the changes in polyclonal CD4+ T cell phenotype in cDC-deficient mice (Figure 6DF and Figure S6CE), suggesting that RG1 may be representative of the Ct-STL specific response. These shifts in Ct-STL-specific T cell differentiation from Th1 to Treg, Th2, and Th17 cells in cDC1-deficient mice may promote loss of immune control to Ct-STL.

In contrast, no changes in RG1 or polyclonal T cell development were observed in DP cDC2-deficient mice (Figures 6DF and S7CE). RG1 recovery was decreased in the MLN of hLangDTA mice lacking DP cDC2s, indicating a role for this cDC subset in antigen presentation (Figure S6B). Deficiency of both cDC1s and DP cDC2s marginally increased the differentiation of RG1 into FoxP3+ Treg and RORγt+ Th17 cells versus cDC1 deficiency alone, but decreased the differentiation of GATA3+ Th2 cells (Figure 6DF and Figure S6CE). Thus, DP cDC2s contribute to the presentation of Ct-STL antigens and in the absence of cDC1s, are observed to promote Th2, but not Th1, differentiation.

Homeostatic Ct-STL induces colon Th1 responses and facilitates immunity to pathogens

In humans, Cp infection predominantly affects the jejunum and ileum (Leitch and He, 2012). To establish the extent to which Ct-STL colonizes different segments of the gastrointestinal tract, we performed qPCR of Ct-STL Gapdh on the intestinal epithelial cell preparations of the duodenum, jejunum, ileum, and colon of Ct-STL colonized mice. We found that while Ct-STL DNA was highest in the ileum followed by the jejunum, Ct-STL DNA was detectable in duodenum epithelial preparations (Figure 7A). While the presence of DNA does not necessarily indicate productive infection, these data indicate the Ct-STL can be found throughout the SI.

Figure 7. Homeostatic Ct-STL induces colon Th1 responses and facilitates immunity to pathogens.

Figure 7.

(A-D) Analysis of mice 3 weeks after colonization with Ct-STL at weaning. (A) Relative quantification of Ct-STL Gapdh to mouse Trac DNA in different segments of intestinal epithelium in mice 3 weeks after Ct-STL colonization. Ct Gapdh fold change is normalized to the average of all samples. (expt.=2). (B-C) IFNγ expression in cLP polyclonal CD4+ T cells by (B) flow cytometry of IFNγYFP IL17GFP Foxp3Thy1.1 mice (expt.= 3) or (C) qPCR of sorted CD4+ T cells for Ifng and Tbx21 expression (expt.=2). (D) Cytokine analysis of colons by multiplex Luminex assay. PCA using RANTES, IL12p40, and Figure S7G cytokines is displayed (expt.=2). (E) Comparison of the top 10 colon Th1 TCRs vs SILP TCRs in Ct-STL colonized mice (see also Figure 5, expt.=5). TCRs tested are indicated (n=5 for Ct-STL cLP, n=3 for control cLP, n=5–6 for SILP). (F) In vitro reactivity of Ct-STL-colonized colon Th1 TCRs shown in (E) to Cp oocyst antigens as per Figure 4D (expt.=2). (G) Cumulative frequencies of known Ct-STL-reactive TCRs in the MLN, SILP, and cLP Th1 TCR repertoires at homeostasis. (H) 2×105 retroviral RG1-expressing IFNγYFP TCli-β T cells were transferred into 3.5-week-old CR WT mice 3 days after Ct-STL colonization, and analyzed for CTV dilution and IFNγYFP expression after 2 weeks in the pLN (axillary, inguinal, and brachial), spleen, MLN, SILP, and cLP. (expt.=2). (I) C. rodentium infection of 2 strains of C3H from 2 different vendors 3 weeks after colonization at 3.5 weeks of age. Survival is based on euthanasia at 20% weight loss (n=10 per group, expt.=2 per C3H strain). C. rodentium CFUs in stool are shown (right, expt.=3). (J) Colon injury in DSS and anti-IL10R-treated, Ct-STL-colonized and control mice. 3.5 week old CR WT mice were colonized with Ct-STL as in (I) and then treated with 1 mg of anti-IL10R one time plus 1% DSS in drinking water for 7 days. Survival, weight loss, and colon length at end of experiment or euthanasia threshold (20% weight loss or morbidity) were assessed (n=16–19 per group, expt.=3). Each dot represents data from an individual host (A-D, G-H, I right, J right) or individual hybrid experiment well (F). Mean ± S.E.M. error bars are shown. Student’s t-test (B-C, H bottom, J right), Kaplan-Meier (I, J), one-way ANOVA with Dunnet’s (F) or Sidak’s (H) correction, Mann-Whitney test (I right), or repeated measures mixed effects analysis (J middle) p-values are shown. Multiple t-tests with FDR-correction (Benjamini, Krieger, and Yekutieli) q-values are shown in (D).

As Ct-STL colonization alters T cell responses in SI, we wondered whether T cell tolerance to food antigens is altered. pTreg cell generation to oral OVA is blocked by SI infections (Bouziat et al., 2017; Esterhazy et al., 2019). In Ct-STL colonized mice, OT-II pTreg cell differentiation was unaffected (Figures S7AB). Furthermore, Ct-STL colonization did not lead to a difference in inflammation with subsequent subcutaneous OVA challenge (Figure S7C). Although OVA is absorbed along the entire length of the SI (Esterhazy et al., 2019; Kulkarni et al., 2020), OT-II pTreg differentiation is highest in the duodenal-draining LNs (Esterhazy et al., 2019). Thus, lower Ct-STL in the duodenum may be one reason that pTreg cell generation to a food antigen is spared (Figure 7A). Alternatively, Ct-STL colonization may not affect SI T cell differentiation to other antigens due to cDC microenvironment restricted T cell development.

Cryptosporidium primarily infects the small, but not large, intestine in humans (Current and Reese, 1986). Consistent with this, qPCR of Ct-STL from mouse colon epithelial cells was at or below the limit of detection (>150-fold lower than ileal epithelial cells) and histologic analysis of Ct-STL-colonized hLangDTA Irf8 +32 5’−/− mice showed Ct-STL parasites in SI but not colon epithelial cells (Figures 7A and S7D). Furthermore, tracking of RG1 localization using T cells that retrovirally express Akaluc luciferase (Iwano et al., 2018) transferred into Rag1−/− mice showed they preferentially localized to the SI, in contrast to colon Helicobacter-specific CT2 cells, which localized to the cecum and colon (Figure S7E) (Chai et al., 2017; Nutsch et al., 2016). Nevertheless, analysis of colon polyclonal T cells after Ct-STL colonization revealed that IFNγYFP+ CD4+ T cells were also increased in the cLP with Ct-STL (Figure 7B), which was confirmed by qRT-PCR of Ifng and Tbx21 (Figure 7C). Ct-STL colonization also resulted in slightly decreased frequencies of Treg and Th17 cells in the cLP and distal colon-draining MLN (dMLN) (Figures 7B and S7F). Thus, these data suggest that Ct-STL also affects the colon T cell population and skews it toward a Th1 response.

To determine which other colon cytokines are affected by Ct-STL, we performed multiplex cytokine analysis of whole colons after Ct-STL colonization. We found that both IL12p40 and RANTES (CCL5) were increased in Ct-STL-colonized mice (Figures 7D and S7G). RANTES can be induced by IFNγ (Liu et al., 2005) to recruit monocytes and activated T cells (Schall et al., 1990), and RANTES expression in the colon is strongly associated with inflammation, including exacerbation of DSS-colitis (Ajuebor et al., 2001; Elinav et al., 2011). Overall, these data suggest that Ct-STL induces pro-inflammatory cytokine mediators of both innate and adaptive immunity in the colon.

Colon Ct-STL-induced effects may be due to changes in the colon microbiome. We assessed changes in bacterial communities using 16S rRNA sequencing and analysis of amplicon sequence variants (ASVs) (Callahan et al., 2016). Colonization with Ct-STL minimally decreased species diversity (Shannon index) but shifted bacterial composition as assessed by PERMANOVA (p = 0.001) or NMDS (Figure S7HI). Ct-STL colonization induced an increase in the frequency of Prevotellaceae at the expense of Lachnospiraceae (Figure S7JK), the latter of which are producers of short chain fatty acids that are associated with the expansion of Treg cells (Arpaia et al., 2013; Furusawa et al., 2013; Smith et al., 2013). Additionally, increases in Prevotellaceae induce RANTES expression in colon epithelial cells (Elinav et al., 2011), possibly by stimulation of TLRs and NOD1/2 (Berube et al., 2009; Vaughan et al., 1991). However, study of the interaction of Ct-STL and microbiota is complicated, as treatment with broad-spectrum antibiotics increased Ct-STL oocyst shedding by 100–1000-fold (Figure S7L), consistent with previous Cryptosporidium studies (Charania et al., 2020). These data demonstrate that Ct-STL colonization can both alter and be altered by the composition of the colon microbiome in WT mice.

The alterations in gut bacteria by Ct-STL suggest that increased colon Th1 cells could be generated to commensal antigens. Analysis of the top cLP Th1 TCR clones revealed overlap with SILP Th1 TCR sequences (Figure 7E, Table S4), and included the Ct-STL specific TCRs RG1, RG2, and RG3 (Figures 4CD and S4B). We tested 4 additional cLP Th1 TCRs (LA1–4) in vitro for reactivity to Cryptosporidium as well as Ct-STL-negative colon stool contents. Although LA1–4 did not react to stool antigens, this experiment does not exclude TCR recognition of luminal antigens that are in large particles or found at low concentration. However, we did discover that LA3 was reactive to Cp oocysts (Figure 7F). Together, the Ct-STL specific TCRs RG1, RG2, RG3, and LA3 made up a substantial fraction of the Th1 TCR repertoire in SILP and cLP tissues (Figure 7G). Additionally, RG1 T cells transferred into Ct-STL colonized mice expressed IFNγYFP in the cLP at similar frequencies as RG1 cells recovered from the SILP, albeit with lower MFI (Figure 7H). In contrast, RG1 cells recovered from the spleen, peripheral (p)LNs, and MLN did not express IFNγYFP (Figure 7H). RG1 cells recovered from the SILP and cLP fully diluted CTV, while RG1 recovered from the pLN, spleen, and MLN, some of which may represent recirculating cells, underwent fewer cell divisions (Figure 7H). Thus, these data indicate that the increase in colon Th1 cells with Ct-STL is driven, in large part, by the presence of Ct-STL specific T cells that are activated by Ct-STL antigens that are presented in the colon.

Finally, we asked whether Ct-STL affects colon immune function. We first assessed whether Ct-STL protects against Citrobacter (C.) rodentium, which infects the cecum and colon. For these experiments, we used strains of C3H (HeNCrl and HeOuJ) mice, which are more susceptible to C. rodentium than C57BL/6 mice (Vallance et al., 2003). Consistent with the role for type I immunity in C. rodentium infection (Bry et al., 2006; Shiomi et al., 2010; Spahn et al., 2008), we observed that colonization with Ct-STL decreased lethality (Figures 7I). Analysis of C. rodentium colony forming units (CFUs) from stool at early timepoints revealed that a subset of Ct-STL colonized mice had decreased CFUs at day 4 compared to control mice (Figure 7I). We also tested whether Ct-STL colonization modulated colon inflammation after treatment with DSS drinking water plus anti-IL10R antibody (Chai et al., 2017). As would be predicted from other studies where increased RANTES expression exacerbates DSS-colitis (Elinav et al., 2011), Ct-STL colonization was associated with increased lethality and colon injury in this model (Figures 7J). Thus, these data all suggest that although Ct-STL colonization primarily occurs in the SI, Ct-STL induces clear functional changes in the colon immune system that may be due to a combination of IFNγ-expressing Ct-STL specific colon T cells, increased innate cytokines, and changes to the colon microbiome.

Discussion

The serendipitous discovery of a Cryptosporidium commensal pathobiont in our colony (Ct-STL) was revealed by spontaneous illness in cDC1-deficient mice. Ct-STL may have a very different infection profile compared to other Ct strains isolated from “wild” mice. These “wild” Ct strains induce robust immunity in laboratory mice, which stop shedding oocysts 2–3 weeks after infection (Ren et al., 2012; Sateriale et al., 2019), whereas WT C57BL/6 mice shed Ct-STL oocysts for months. Although a direct functional and genomic comparison between these strains is required to establish the pathogenic vs commensal “nature” of Ct strains and exclude variables such as Ct dose and host microbiota, we suspect that Ct-STL has low pathogenicity and is adapted to be a commensal in C57BL/6 mice. It is also important to note that although Ct-STL acts as a commensal in our mouse colony, it remains possible that malnutrition, exposure to other pathogens, or alterations in background microbiota may result in morbidity from commensal Cryptosporidium. Despite the great amount of work focused on the bacterial microbiota, parasites such as Ct-STL and Tritrichomonas (Chudnovskiy et al., 2016) may play a much greater role in dictating host:commensal immunity than previously appreciated, contributing to different results between labs and even between strains within a lab.

Our observation that Ct-STL elicits strong changes in the CD4+ T cell compartment is consistent with existing knowledge of Cryptosporidium infections. First, cryptosporidiosis in humans is associated with low CD4+ T cell counts in AIDS patients (Current et al., 1983; Petersen, 1992). Second, infection with “wild” Ct leads to persistent oocyst shedding over 6 months in Rag1−/− and athymic Foxn1nu mice (Sateriale et al., 2019). Finally, mice that lack MHC Class II, but not Class I, are susceptible to Cp infection (Aguirre et al., 1994). Although other immune cells likely contribute, CD4+ T cells appear to be the most important immune cell subset for the control of Cryptosporidium infection.

Nonetheless, we were surprised to observe that Ct-STL, as a single commensal species, could cause such a dramatic induction of antigen-specific Th1 responses. The first reason is that a Th1 response to commensals has not been described, although other responses are reported. For example, segmented filamentous bacteria induce non-pathogenic Th17 cells (Ivanov et al., 2009), Helicobacter–pTreg cells (Chai et al., 2017; Xu et al., 2018), and Akkermansia–a mixed Th1, Th17, Treg, and Tfh cell response (Ansaldo et al., 2019). A colon Th1 and Th17 response is observed with Tritrichomonas parasites, although antigen specificity has not been examined (Chudnovskiy et al., 2016; Escalante et al., 2016). A second reason is that Th1 responses are thought to portend inflammation and tissue destruction and not maintenance of homeostasis. One hypothesis is that Ct-STL may induce IL-10 producing Tr1 cells, which are also seen during Toxoplasma infection (Jankovic et al., 2007), and may be more important than Treg cells for maintaining tolerance in the SI (Roncarolo et al., 2018).

Homeostasis during Ct-STL colonization is dependent on cDC1-mediated type 1 immunity, consistent with previous reports. cDC1s are an important source of IL-12 for Th1 development in Toxoplasma gondii infection (Mashayekhi et al., 2011), which we also observed for Ct-STL. In addition, Batf3-mutant mice deficient in cDC1s (Hildner et al., 2008) are more susceptible to Cp infection (Potiron et al., 2019). However, adult Batf3−/− mice clear Cp within a week, which may reflect differences between Cryptosporidium species or residual mucosal cDC1s in the gut of Batf3−/− mice that can be expanded by infection (Mashayekhi et al., 2011; Tussiwand et al., 2012). Although all cDC1-deficient mice had increased shedding of Ct-STL oocysts, not all cDC1-deficient mice succumbed to cryptosporidiosis. The variables that influence infection severity are still unclear. Additionally, the specific mechanisms by which cDC1s, and not cDC2s, recognize Cryptosporidium and induce IL-12 remain to be elucidated.

Interestingly, we were still able to recover RG1 T cells from CD103+ cDC-deficient mice, suggesting that other APCs are capable of presenting Ct-STL antigens. One unexpected effect of cDC1-deficiency was that a fraction of Ct-STL specific RG1 T cells underwent pTreg cell differentiation. We hypothesize that cDC1s encountering Ct-STL provide a dominant signal such as IL-12 that prevents pTreg cell generation by other cDC subsets. Nonetheless, it remains unknown why cDC2s would facilitate pTreg cell selection to Ct-STL with the loss of cDC1s, and whether the ratio of pTreg to Th1 cell selection to Ct-STL could precipitate fulminant cryptosporidiosis.

Another unexpected finding is that Ct-STL’s geographical impact extends well past the site of infection in the SI and also induces a Th1 response in the colon. There are several potential mechanisms for increased colon Th1 cells, including Ct-STL effects on the microbiota, colon epithelium, or innate immune milieu. However, our data showing that a substantial fraction of colon Th1 cells are Ct-STL specific and express IFNγ suggest that these T cells reencounter antigen in the colon acquired from either low level infection of colon epithelial cells or colon sampling of antigens present in the fecal stream. Increased Th1 cells in the colon was associated with increased resistance to colon infection by Citrobacter and susceptibility to injury by DSS plus anti-IL-10R treatment. Increased IFNγ may “pre-arm” innate cells such as macrophages to resist infection, akin to that seen with latent gamma-herpes virus-mediated protection against Listeria (Barton et al., 2007). Alternatively, IFNγ may induce the release of antimicrobial defenses from colon epithelium, as is seen for SI Paneth cells (Farin et al., 2014), or act directly on colon epithelial cells to alter homeostasis (Kominsky et al., 2014; Nava et al., 2010).

Study of the immune response to commensal Ct-STL may be relevant to human disease. In the United States, Cryptosporidium is typically thought of as a pathogen of periodic small outbreaks, but it is often endemic in the developing world (Checkley et al., 2015), where malnourished children have increased Cryptosporidium infection associated with more severe diarrheal illness (Mondal et al., 2012). However, there are likely commensal or pathobiont strains that infect humans, as different strains of Cp and Cryptosporidium hominis produce varying clinical manifestations from severe diarrhea to asymptomatic infection (Cama et al., 2008). In addition, the prevalence of chronic asymptomatic Cryptosporidium colonization of humans may be underappreciated, as typical methods of detection are not sensitive and qPCR methods are not routinely utilized (Heine et al., 1984; Kuhls et al., 1992; Mead et al., 1991). Even in the United States, it is estimated that less than 1% of Cryptosporidium infections are diagnosed (Scallan et al., 2011). Finally, recent reports from Lebanon and Poland suggest that the presence of Cryptosporidium convey a 3 to 11-fold increased risk for colon adenocarcinoma (Osman et al., 2017a; Sulzyc-Bielicka et al., 2018). This is intriguing in light of our observation that Ct-STL increases susceptibility to colon injury, which can facilitate tumorigenesis (Terzic et al., 2010). Thus, Ct-STL may provide a useful murine model for understanding asymptomatic Cryptosporidium infection in human disease and the role of type 1 immunity in host:commensal tolerance.

Limitations of the Study

This study describes a strain of Cryptosporidium with characteristics of a commensal organism in mice. However, comparative studies using other mouse strains as well as studies in humans will be required to generalize these findings. We found that cDC1s are important for preventing Th17 and pTreg cell development to Cryptosporidium, but it remains unproven whether skewed T cell development and reduced IL-12 or IFNγ production in cDC1-deficient mice causes fulminant cryptosporidiosis. Finally, the notion that Cryptosporidium can markedly affect adaptive immunity in the colon will require future studies with other organisms to determine if this is a general phenomenon of small intestinal infections.

STAR METHODS

RESOURCE AVAILABILITY

Lead contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Chyi-Song Hsieh (chsieh@wustl.edu).

Materials availability

Ct-STL oocysts and TCR expression plasmids are available upon request.

Data and code availability

TCR and 16S rRNA sequencing data have been deposited at European Nucleotide Archive (ENA) and are publicly available as of the date of publication. Accession numbers are listed in the key resources table. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

KEY RESOURCES TABLE.
REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
anti-IFNγ Clone H22 Leinco Cat#I-438
PIP Leinco Cat#P-376
anti-Thy1.2 clone 30H12 BioXCell Cat#BE0066
LTF-2 BioXCell Cat#BE0090
anti-NK1.1 clone PK136 BioXCell Cat##BE0036
C1.18.4 BioXCell Cat#BE0085
anti-IL10R BioXCell Cat#BE0050
anti-CD3ε BioXCell Cat#BE0001-1
anti-MHCII BioXCell Cat#BE010
anti-CD28 BioXCell Cat#BE0015-1
anti-TGFβ BioXCell Cat#BE0057
anti-IL-12 BioXCell Cat#BE0052
anti-IFNγ BioXCell Cat#BE0054
anti-IL-4 BioXCell Cat#BE0045
Anti-mouse CD3ε (clone#145-2C11) FITC, PE, and BV421 BioLegend Cat#100305/07/35
Anti-mouse/human B220 (clone#RA3-6B2) APC/Cy7, A700, and PE/Cy7 BioLegend Cat#103223/31/22
Anti-mouse CD19 (clone#6D5) APC/Cy7 BioLegend Cat#115529
Anti-mouse I-Ab (clone#AF6-120.1) APC and PerCP/Cy5.5 BioLegend Cat#116417/15
Anti-mouse CD11c (clone#N418) PE/Cy7 and BV605 BioLegend Cat#117317/33
Anti-mouse/human CD11b (clone#M1/70) BV711 BioLegend Cat#101241
Anti-mouse CD103 (clone#2E7) BV421 BioLegend Cat#121421
Anti-mouse F4/80 (clone#BM8) PE/Cy7 BioLegend Cat#123113
Anti-mouse CD4 (clone#RM4-5) BV711, PE, PB, and BV605 BioLegend Cat#100549/11/34/48
Anti-mouse CD25 (clone#PC61) APC, BV605, PerCP/Cy5.5, and PE/Cy7 BioLegend Cat#102011/35/29/15
Anti-mouse/human CD44 (clone#IM7) APC/Cy7 and BV605 BioLegend Cat#103027/47
Anti-mouse CD62L (clone#MEL-14) APC/Cy7 and BV605 BioLegend Cat#104427/37
Anti-mouse FOXP3 (clone#FJK-16s) FITC ThermoFisher Cat#11-5773-82
Anti-mouse GATA3 (clone#16E10A23) PE BioLegend Cat#653803
Anti-mouse RORγt (clone#B2D) APC ThermoFisher Cat#17-6981-80
Anti-mouse TBET (clone#4B10) PE/Cy7 BioLegend Cat#644823
Anti-mouse CXCR3 (clone#CXCR3-173) BV421, PE, and APC BioLegend Cat#126521/06/12
Anti-mouse Thy1.1 (clone#OX-7) BV711, PE/Cy7, and PerCP/Cy5.5 BioLegend Cat#202539/18/16
Anti-mouse CD80 (clone#16-10A1) APC BioLegend Cat#104713
Anti-mouse CD45.1 (clone#A20) PerCP/Cy5.5, PE, and A700 BioLegend Cat#110728/08/24
Anti-mouse CD45.2 (clone#104) APC, A700, and PE BioLegend Cat#109814/22/08
Anti-mouse VB6 (clone#RR4-7) PE and APC BioLegend Cat#140003/5
Anti-mouse VB5 (clone#MR9-4) PE BioLegend Cat#139503
Anti-mouse Va2 (clone#B20.1) APC/Cy7 and PerCP/Cy5.5 BioLegend Cat#127818/14
Bacterial and virus strains
Citrobacter rodentium Gift from M. Colonna (Wash. U.)
Biological samples
Cryptosporidium tyzzeri (Ct-STL) Our mouse colony N/A
Chemicals, peptides, and recombinant proteins
FITC-Dextran Sigma Cat#FD4
Ovalbumin (Grade V) Sigma Cat#A5503
Incomplete Freund’s Adjuvant Sigma Cat#F5506
Ovalbumin (Grade VI) Sigma Cat#A2512
Dextran Sodium Sulfate TdB Labs Cat#9011-18-1
Critical commercial assays
Quick-DNA Fecal/Soil Microbe Prep Kit Zymo Cat#D6010
Luminaris Color HiGreen qPCR Master Mix ThermoFisher Cat#K0391
CountBright Absolute Counting Beads ThermoFisher Cat#C36950
Crypt-a-Glo Waterborne Cat#A400FLR
TransIT-293 ThermoFisher Cat#MIR2700
CellTrace Violet (CTV) ThermoFisher Cat#C34571
SuperScript III Reverse Transcriptase ThermoFisher Cat#18080093
Accuzyme DNA Polymerase Bioline Cat#BIO-21052
Foxp3/Transcription Factor Staining Buffer Set ThermoFisher Cat#00-5523-00
Bio-Plex Mouse Cytokine 23-plex Assay BioRad Cat#M60009RDPD
Deposited data
TCR Sequencing This paper ENA: accession #PRJEB46540
16S rRNA This paper ENA: accession #PRJEB46540
Experimental models: Cell lines
NFAT-GFP T cell hybridoma Gift from K. Murphy (Wash. U.) (Ise et al., 2010) N/A
B16 flt3L melanoma Gift from G. Dranoff (Mach et al., 2000) N/A
Experimental models: Organisms/strains
Mouse: C57BL/6-Ly5.1 Charles River Cat#494
Mouse: C57BL/6NCrl (Ly5.2) Charles River Cat#027
Mouse: C57BL/6J JAX Cat#000664
Mouse: Rag1−/− JAX Cat#002216
Mouse: hLangDTA Gift from D. Kaplan (U. Pitt.) (Kaplan et al., 2005) N/A
Mouse: Irf8 +32 5’−/− Gift from K. Murphy (Wash. U.) (Durai et al., 2019) N/A
Mouse: Foxp3IRES-GFP JAX Cat#006772
Mouse: Foxp3IRES-Thy1.1 Gift from A. Rudensky (MSKCC) (Liston et al., 2008) N/A
Mouse: IL-17AIRES-GFP JAX Cat#18472
Mouse: IL12p40IRES-YFP JAX Cat#015864
Mouse: Ifngr1−/− JAX Cat#003288
Mouse: C3H/HeOuJ JAX Cat#000635
Mouse: IFNγIRES-YFP Gift from R. Locksley (UCSF) (Reinhardt et al., 2009) N/A
Mouse: TCli TCRβ Transgenic Gift from A. Rudensky (MSKCC) (Wong et al., 2000) N/A
Mouse: TCli TCRαβ Transgenic Gift from A. Rudensky (MSKCC) (Wong et al., 2000) N/A
Mouse: Tcra−/− Jax Cat#002116
Mouse: C3H/HeNCrl Charles River Cat#025
Mouse: OT-II Transgenic JAX Cat#004194
Oligonucleotides
Primers for TCR-seq, see Table S1 N/A N/A
Other primers, see Table S5 N/A N/A
Recombinant DNA
(RG1/RG2/RG3/LA1/LA2/LA3/LA4/CT6)-IRES-Thy1.1 This paper and (Hsieh et al., 2004; Lathrop et al., 2011) N/A
RG1-P2A-Akaluc This paper and (Iwano et al., 2018) N/A
CT2-P2A-Akaluc This paper and (Iwano et al., 2018) N/A
Software and algorithms
Prism v9 https://www.graphpad.com N/A
FlowJo v10 https://www.flowjo.com N/A
ImageJ v1.53a https://imagej.nih.gov/ij/download.html N/A
MegAlign v16 https://www.dnastar.com N/A
R https://www.r-project.org/ N/A
dada2 v1.14 https://www.bioconductor.org/packages/release/bioc/html/dada2.html N/A
Vegan v2.4 https://cran.r-project.org/web/packages/vegan/index.html N/A
Phyloseq v1.32 https://www.bioconductor.org/packages/release/bioc/html/phyloseq.html N/A
DESeq2 v1.28 https://bioconductor.org/packages/release/bioc/html/DESeq2.html N/A
Gbm v2.1 https://cran.r-project.org/web/packages/gbm/index.html N/A

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Animal breeding and experiments were performed in a specific pathogen-free animal facility using protocols approved by the Washington University Institutional Animal Care and Use Committee (protocol #20–0036). Mice were maintained negative for the following agents: mouse hepatitis virus, Sendai virus, Theiler’s murine encephalomyelitis virus, reovirus, ectromelia virus, pneumonia virus of mice, rotavirus, lymphocytic choriomeningitis virus, minute virus of mice, k virus, polyoma virus, mouse cytomegalovirus, mouse adenovirus, mouse parvovirus, Mycoplasma pulmonis, Clostridium piliforme, Streptococcus pneumoniae, Bordetella bronchiseptica, Streptobacillus moniliformis, Corynebacterium kutscheri, Salmonella species, Citrobacter rodentium, fur mites, and pinworms. Mice were housed up to 5 per cage in autoclaved individually ventilated caging (NexGen, Allentown, NJ), with corncob bedding (The Andersons, Maumee, OH). Mice were fed ad lib irradiated rodent chow (LabDiet 5053WU, PMI Nutrition, St Louis, MO) and provided autoclaved municipal water in bottles. The mice were maintained on a 12:12 hour light cycle. Room temperature was maintained between 20–23° C and humidity between 30–70%. Littermates were used for all comparisons of genetic strains and Ct-STL colonization experiments unless noted otherwise. Irf8 +32 5’−/− (149 bp deletion) (Durai et al., 2019) mice were a gift from K. Murphy (Wash.U.). Human Langerin-DTA mice (hLangDTA) mice were a gift from D. Kaplan from U.Minn. (Kaplan et al., 2005). Rag1−/− (#002216), OT-II (#004194), Foxp3IRES-GFP (#006772), IL-17AIRES-GFP (#18472), p40IRES-YFP (#015864), Ifngr1−/− (#003288), and C3H/HeOuJ (#000635) mice were obtained from The Jackson Laboratory (JAX). IFNγIRES-YFP “Great” mice (Reinhardt et al., 2009) were a gift from R. Locksley (University of California, San Francisco). TCli TCRβ TCRα+/− and TCli TCRαβ mice were previously described (Solomon and Hsieh, 2016; Wong et al., 2000). Foxp3IRES-Thy1.1 mice were a gift from A. Rudensky (Memorial Sloan Kettering Cancer Center). C3H/HeNCrl (#025) were obtained from Charles River Laboratories. Ly5.1 and Ly5.2 mice from Charles River Laboratories were analyzed as wild-type mice. Both male and female mice were used for experiments with the exception of WT CR Ly5.1, Ly5.2, and C3H/HeNCrl mice, which were all male.

METHOD DETAILS

MLN and intestine lymphocyte isolation

For cDC analyses, dMLN or pMLN were dissociated in RPMI-Enzyme, containing 5% bovine calf serum, penicillin and streptomycin, 1 mM sodium pyruvate, non-essential amino acids, 50 μM beta-mercaptoethanol, 65.8 μg/ml collagenase VIII (Sigma # C2139), and 0.2 U/ml dispase (Thermo Fisher # CB-40235), for 45 minutes at 37°C with continuous stirring. Small intestine and colon lamina propria cells were isolated by performing 4, 12-minute washes in 5 mM EDTA in HBSS, shaking at 140 rpm at 37°C. Intestines were cut into small pieces and incubated in RPMI-Enzyme for 2 hours, shaking at 140 rpm at 37°C. Cells were filtered through a 100 μm filter before staining for FACS analysis with an FACSAria IIu (Becton Dickinson).

Intestine histology

Luminal contents were removed, and intestines flushed with 10% (v/v) formalin and fixed in ‘swiss rolls’ in cassettes in 10% (v/v) formalin overnight at room temperature (RT). Cassettes were washed and left in 70% ethanol at 4°C until paraffin embedding, cutting, and staining with H&E by Histology Consultation Services, Inc. (http://www.histocs.com). 5 continuous villus and crypt pairs in the same plane of section from the middle of the small intestine were imaged per slide using a ThermoFisher EVOS FL Auto Imaging System. Villus height and width, crypt depth and width, goblet cells per villus and crypt pair, mitoses per crypt, Ct-STL organisms per villus, and epithelial were quantified in a blinded fashion using ImageJ. For epithelial score (max 14), points were assigned for villus epithelial separation or sloughing (0=none, 1=cell separation from villus tip of 1–2 out of 5 villi evaluated, 2=cell separation from villus tip of 3–5 out of 5 villi evaluated, 3=denuded villus tips, 4=denuded villus tips with fibrin and/or inflammatory exudate), villus epithelial dysplasia (0=none, cells well aligned in monolayer, 1=cytoplasmic basophilia with high nuclear:cytoplasmic ratio, 2=mild, multifocal epithelial disorganization, 3=regional moderate epithelial malalignment, 4=marked regional malalignment with cell piling), villus epithelial attenuation (0=none, epithelium are uniformly tall and columnar-shaped, 1=multifocal cuboidal- shaped epithelium, 2=regional-multiregional cuboidal shaped-epithelium, 3=flattened villus epithelium), and degree of crypt branching (0=none, 1=1–2 branches observed for 1–2 out of 5 crypts evaluated, 2=1–2 branches observed for 3–5 out of 5 crypts evaluated, 3=greater than 2 branches within 1 or more crypts out of 5 crypts evaluated) (Sateriale et al., 2019). Quantifications were averaged for each slide.

qPCR and sequencing of Cryptosporidium DNA

Quick-DNA Fecal/Soil Microbe Prep Kit (Zymo) was used to isolate DNA from terminal pellets. qPCR was carried out with Luminaris Color HiGreen qPCR Master Mix (ThermoFisher K0391) using a LightCycler 480 (Roche). For qPCR, Cryptosporidium-specific Gapdh was amplified with the primer pairs: F 5’-AAGGACTGGAGAGCAGGAAG −3’ and R 5’-CCATACCTGTGAGCTTTCCA-3’ (Wilke et al., 2018). Ct values were compared to Cp DNA corresponding to a known quantity of oocysts to calculate fecal Ct-STL oocyst equivalents. For Sanger sequencing of Gapdh, forward 5’-CGGTTTCGGACGTATTGGT-3’, 5’-GACCCAGCAGAAATCCCATG;−3’ and reverse 5’-TCCCTTCATTGGACCATTGGA-3’, 5’-TGAAGCAACATAAACGGCGA-3’ primers were used for Ct-STL and Cp-AUCP-1. For Sanger sequencing of Gp60, forward 5’-ATAGTCTCCGCTGTATTC-3’ (AL3531) and reverse 5’-GGAAGGAACGATGTATCT-3’ (AL3535) primers were used (Stensvold et al., 2014). Reference sequences were obtained from NCBI and CryptoDB and were aligned using MUSCLE in MegAlign (v16, DNASTAR) with Gap Penalty 25, Tamura-Nei distance, and pairwise gap removal with other options at the default setting; and plotted using ggtree (v2.1) in R. Quantification of Ct-STL DNA in intestinal epithelial cell preparations was performed from 6-week-old CR B6 mice, 3 weeks after gavage with 4×103 Ct-STL oocysts. Murine Trac qPCR was used to control for input DNA quantity. intestines were washed 4 times in PBS to remove stool. Duodenum, jejunum, and ileum were isolated based on the first 25%, middle 50%, and last 25% of SI (Esterhazy et al., 2019). Colon included the cecum. Intestines were incubated for 20-minutes in 5 mM EDTA in HBSS with 5% bovine calf serum, shaking at 140 rpm at 37°C. Intestines were removed, and the epithelial cells in suspension were filtered through a 100 μm filter and pelleted. DNA was isolated using DirectPCR Lysis Reagent (Viagen #301-C) and qPCR for Ct-STL Gapdh was performed as described above. ΔCt values were calculated using Trac qPCR to quantify epithelial cell input for each sample (primers: forward 5’-TTGAGCCAGGAGGAGACTTC-3’ and reverse 5’-ATCAATGTGCCGAAAACCAT-3’).

Cryptosporidium oocyst flow cytometry

Terminal pellets were resuspended at 50 mg/ml in 5 mM EDTA in PBS. Pellets were vortexed for 20 minutes at 4°C, sonicated for 4 minutes, and passed through a 70 μM filter. CountBright Absolute Counting Beads (2.5 μl ThermoFisher #C36950) were added to 25 μl of filtrate, incubated for 20 minutes in 200 μl 5 mM N-acetyl-cysteine (Sigma) on ice. Samples were washed twice with PBS and blocked >20 minutes to overnight in 20% FBS in PBS on ice, stained with DAPI and 1:1000 and 20x Crypt-a-Glo (Waterborne #A400FLR) for 30 minutes, washed, and analyzed by flow cytometry. Limit of specific detection was determined by pooling flow cytometry data for 82 known Cryptosporidium-free samples and calculating the 95% probability by negative binomial distribution that <2 oocyst events are detected in the average number of CountBright beads analyzed per sample.

Ct-STL repository

Colon contents were harvested from Rag1−/− mice one week after gavage with stool from Ct-STL vertically infected Irf8 +32 5’−/− mice. Rag1−/− mice were also treated with 1 mg/ml ampicillin, 1 mg/ml neomycin, and 0.5 mg/ml vancomycin in drinking water and 250 μg anti-IFNγ blocking antibody clone H22 (Leinco #I-438) for 2 days before infection. Feces were resuspended in cold sterile PBS by vortexing. Large debris was pelleted by slow spin at 200 rpm for 5 minutes at 4°C and the supernatant was transferred to a new tube. 100% bleach was added to the supernatant to a final concentration of 40%, incubated on ice for 10 minutes, and then washed 3 times with cold sterile PBS by centrifugation at 15,000 RPM. Oocysts in the sample were quantified by FACS analysis, stored at 4°C, and checked to ensure zero bacterial growth on aerobic LB plates at 37°C. Unless noted otherwise, for Ct-STL colonization experiments, mice were gavaged at weaning or 3.5 weeks of age with 200 μl PBS or 4×103 Ct-STL oocysts in 200 μl PBS.

Analysis of fecal water weight, SI transit velocity, and FITC uptake

Terminal fecal pellets were immediately collected in a pre-weighed Eppendorf tube, weighed again, and dried overnight at 80°C in open tubes. Pellets and tubes were re-weighed. The percentage of water in feces was obtained from the ratio of dry/pre-baked to fresh pellet weights. For SI transit velocity, mice were gavaged with red dye and the velocity of gastrointestinal transit was calculated by measuring the distance traveled in the small intestine over 30 minutes. The total GI transit time was determined by the amount of time after which the red dye was excreted in stool pellets. FITC-dextran 44mg/100g weight (Sigma #FD4) intestinal uptake in the serum was assessed as described at 4 hours (Gupta and Nebreda, 2014).

TCR sequencing and repertoire analysis

MLN and SILP CXCR3+/− Teff cells (CD4+ Vβ6+ CD44hi CD62Llo Foxp3) and Treg cells (CD4+ Vβ6+CD44hi CD62LloFoxp3+) were sorted from TCli TCRβ Foxp3IRES-GFP TCRα+/− or TCli TCRβ Foxp3Thy1.1 IFNγYFP IL-17AGFP Tcra+/− mice using a FACSAria IIu. TCRα DNA synthesis from sorted cells was performed as described (Chai et al., 2017). Amplification was performed using a multiplex PCR as described in Table S1. MiSeq TCR sequencing data of T cell populations from individual mice were analyzed via dada2 (v1.14) (Callahan et al., 2016) to identify amplicon sequence variants and reduce noise due to sequencing errors. TCR sequences were parsed as before to identify the TRAV and CDR3 amino acid sequence (Chai et al., 2017), which together are used to designate a unique TCR. TCR frequencies were calculated by dividing read counts for an individual TCR by the total reads in a sample. TCR repertoires were analyzed in R using phyloseq (v1.32), DESeq2 (v.1.28), and GBM (v2.1). Each GBM iteration was trained with a random 75% of the samples and tested against the remaining 25%, optimizing the variables n.min, interaction depth, and bag fraction for AUC. The final data represents the average of 400 iterations for relative influence of each TCR, and 100 iterations of each sample for prediction (25% of time as test subset). Heatmaps are representative of TCR frequency averages from individual mice and TCRs selected for inclusion in heatmap analyses were present in at least 2 mice in the population being analyzed.

T cell hybridoma stimulation

T cell hybridoma cells expressing GFP under a minimal NFAT promoter (Ise et al., 2010) were retrovirally transduced with TCRα chains of interest as previously described (Chai et al., 2017; Lathrop et al., 2011). Hybridoma cells (1.5×104) were cultured with splenic CD11c+ DCs (5×104) expanded in vivo with Flt3-ligand B16 melanoma cells in 200 μl D10 (200 μl complete DMEM (Thermo Fisher #SH3008101) with 10% FBS, Glutamax, 50 μM beta-mercaptoethanol, 1 mM sodium pyruvate, 10 mM HEPES, non-essential amino acids, and penicillin and streptomycin) in 96-well flat bottom plates with indicated concentrations of antigens for 40 hours at 37°C. TCRβ+ cells were analyzed for GFP expression by FACS. Where noted, 50 μg/ml soluble anti-CD3ε (BioXCell BE0001–1) or 20 μg/ml MHC Class II blocking antibody (BioXCell BE010). was added to cultures.

Cryptosporidium and other antigen preparations

Cp oocyst antigen was prepared as previously described for vaccination (Wilke et al., 2019). For small intestine epithelial antigen preparations, Charles River mice were treated to enhance infection with 250 μg anti-IFNγ blocking antibody clone H22 (Leinco #I-438) and placed on 1 mg/ml ampicillin, 1 mg/ml neomycin, and 0.5 mg/ml vancomycin drinking water for one week. Mice were either gavaged with control PBS or 4×103 Ct-STL repository oocysts. Small intestine epithelial cells were isolated by a 12-minute wash in 5 mM EDTA in HBSS, shaking at 140 rpm at 37°C, filtered through a 40 μm filter, and then washed with sterile PBS. Stool antigens were prepared by collecting total colon luminal contents from Ct-STL negative TCli TCRβ Foxp3Thy1.1 IFNγYFP IL-17AGFP mice. Antigen samples were resuspended in sterile PBS, autoclaved, and filtered through a 37 μM filter. Stool antigens were also spun at 2000 rpm for 2 minutes to remove large particles; supernatant was used for antigen stimulation. Protein concentration was quantified by Bradford Assay.

Transduction and analysis of Ct-STL-specific TCRs in vivo

24 well plates were coated with 10 μg/ml anti-CD3ε (BioXCell BE0001–1) overnight at 4°C. Plates were washed out with PBS, and plated with 5×105 to 1.5×106 naïve (CD4+Foxp3CD25CD44loCD62Lhi) T cells FACS sorted from TCli TCRαβ Foxp3IRES-GFP Rag1+/− Tg or TCli TCRβ Foxp3Thy1.1 IFNγYFP IL-17AGFP Tcra+/− mice in D10 in Th0 conditions (1 μg/ml anti-CD28 (BioXcell #BE0015–1), 10 μg/ml anti-TGFβ (BioXcell #BE0057), 5 μg/ml anti-IL-12 (BioXCell #BE0052), 5 μg/ml anti-IFNγ (BioXCell #BE0054), 5 mg/ml anti-IL4 (BioXCell #BE0045)). TCRα chain IRES-Thy1.1 retroviral transduction was performed as described (Hsieh et al., 2004) using TransIT-293 (Thermo Fisher #MIR2700). Transduced cultures were left to rest in Th0 media for ~66 hours. Foxp3 TCRα-transduced Thy1.1+ cells were sorted by FACS and 2×104 sorted cells were retro-orbitally injected into each host for in vivo analysis unless otherwise specified. Cells were stained with CTV (Thermo Fisher #C34571) in some experiments. Transferred cells were identified as CD4+ Vβ6+ Thy1.1+. For transcription factor staining experiments, 105 cells were injected per mouse to increase recovery. Transcription factors were stained using the Foxp3/Transcription Factor Staining Buffer Set (Thermo Fisher #00–5523-00).

Bioluminescence Imaging

TCli TCRαβ Foxp3IRES-GFP Rag1+/− Tg mice were transduced with RG1 and CT2 TCRα chain P2A-Akaluc (Iwano et al., 2018)-expressing retroviruses in Th0 conditions as described above. 106 FACS-sorted RG1- or CT2-expressing cells were retro-orbitally-injected into Rag1−/− mice that had been gavaged with either 4×103 Ct-STL oocysts or Helicobacter typhlonius-containing stool 3 weeks prior. In vivo bioluminescence imaging was performed ten days later on an IVIS 50 (PerkinElmer, Waltham, MA; Living Image 4.3.1, 1 minute exposures, 4 bin, FOV12cm, f/stop1, open filter). Mice were injected intraperitoneally with AkaLumine-HCl (5mg in PBS;Sigma, St. Louis, MO) and imaged 5 minutes later using isoflurane anesthesia (2% vaporized in O2). Total photon flux (photons/sec) was measured from the abdomen using Living Image 2.6.

16S rRNA Sequencing

Fecal DNA was purified via column (Zymogenetics), and used in triplicate PCR of the bacterial V4 hypervariable region of 16S rRNA using barcoded primers described previously (Caporaso et al., 2011). PCR products were sequenced using the Illumina MiSeq platform (2×250-bp paired-end reads), and ASVs and taxonomy including species designations if possible (silva 1.32) determined by dada2 (Callahan et al., 2016). ASV data were analyzed by phyloseq (v1.32), vegan (v2.4), and DESeq2 (v.1.28) in R (3.6)

In vivo Antibody Treatment, Oral Tolerance, Colon Cytokine Analysis, Citrobacter rodentium Infection, DSS + anti-IL10R Treatment, and Antibiotic Treatment

Where indicated, mice were treated IP with weekly 250 μg anti-IFNγ blocking antibody clone H22 (Leinco #I-438), 250 μg H22 isotype control clone PIP (Leinco #P-376), 500 μg anti-Thy1.2 clone 30H12 (BioXCell #BE0066), 500 μg 30H12 IgG2b isotype control clone LTF-2 (BioXCell #BE0090), 200 μg anti-NK1.1 clone PK136 (BioXCell #BE0036), or 200 μg PK136 isotype control IgG2a clone C1.18.4 (BioXCell #BE0085). Mice were colonized with 4×103 Ct-STL or gavaged with PBS at 3.5 weeks of age. Functional studies were performed 3 weeks later. Colon polyclonal CD4+ T cells were analyzed by qPCR using the primers Ifng-forward 5’-TCCACATCTATGCCACTTGAG-3’, Ifng-reverse 5’-CTGAGACAATGAACGCTACACA-3’, Tbx21-forward 5’-CAACAACCCCTTTGCCAAAG-3’, and Tbx21-reverse 5’-TCCCCCAAGCAGTTGACAGT-3’. For oral tolerance, mice were gavaged with 50 mg of OVA (Sigma #A5503) on day 1 and 2. Naïve CD4+ CD25 Foxp3 CD44lo CD62Lhi OT-II cells were sorted and 5×104 cells were injected per mouse on day 2. OT-II cells were analyzed from MLNs on day 8. For delayed type hypersensitivity (DTH) response, mice were immunized subcutaneously 2 and 4 weeks after oral OVA with 100 μg OVA in incomplete Freund’s Adjuvant (Sigma #F5506). 3 weeks after the last immunization, mice were challenged with injection of 20 μg OVA (Sigma #A2512) into the footpad. Footpad thickness was measured blinded with micrometer calipers before footpad challenge and 24 hours after. For colon cytokine analysis, whole colons were washed in PBS and homogenized at 300 mg/ml in PBS plus protease inhibitor (Thermo Fisher #A32953). The homogenates were centrifuged repeatedly and supernatants isolated until no pellets were visible. Supernatants were analyzed at 1:4 dilutions using the Bio-Plex Mouse Cytokine 23-plex Assay (BioRad #M60009RDPD) on the BioRad Bio-PlexTM 200 System with Bio-Plex Manager software. Limits of detection were defined using the standard curves’ lowest concentrations of detectable cytokines. Cytokines that had values out of range for >2 samples were excluded from analysis. For C. rodentium infection, mice were infected with 5×107 CFU C. rodentium by gavage. C. rodentium CFUs were calculated by resuspending terminal pellets at 50 mg/ml in sterile PBS and after 24 hours of growth of serial dilutions on Macconkey at 37°C. For DSS + anti-IL10R treatment, mice were injected IP with anti-IL10R blocking antibody (BioXCell BE0050) and put on sterile 1% (w/v) DSS drinking water (TdB Labs 9011–18-1) before being switched to regular water after 1 week. For VAN antibiotic treatment, mice were put on control sterile drinking water or sterile drinking water containing 0.5 mg/ml vancomycin, 1 mg/ml ampicillin, and 1 mg/ml neomycin for one week.

QUANTIFICATION AND STATISTICAL ANALYSIS

Graphpad Prism v9 was used for used for statistical and graphical analysis unless noted. For in vivo experiments, each dot represents data from an individual host. In figures representing experiment means, n noted in the figure legend. Independent experimental replicates are indicated in the figure legends. Statistical tests utilized for individual experiments are noted in the figure legends. FDR, Sidak’s or Dunnet’s correction for multiple comparisons are used where noted. Significance was defined by *p< 0.05, **p< 0.01, ***p<0.001, ***p<0.0001.

Supplementary Material

replacement_suppl_file

Highlights.

  • Cryptosporidium tyzzeri in our colony (Ct-STL) is a commensal pathobiont

  • Ct-STL elicits a dominant antigen-specific Th1 response

  • cDC1s are required for efficient Th1 induction and blockade of Th17/Treg generation

  • Ct-STL markedly impacts the immune system of both the small and large intestine

Acknowledgements:

We would like to thank Nicole Santacruz and Patricia Hsieh for technical assistance (Wash.U.). C.-S.H. is supported by the grants U01 AI131349, R01 AI136515, R01 AI079187, BWF, and the Wolff Professorship. L.D.S. is supported by NIAID AI145496. E.R.G. is supported by NIH grants F30 DK111071 and T32 GM007200-42. Additional acknowledgements are provided in the supplement.

Footnotes

Declaration of Interests: The authors declare no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Aguirre SA, Mason PH, and Perryman LE (1994). Susceptibility of major histocompatibility complex (MHC) class I- and MHC class II-deficient mice to Cryptosporidium parvum infection. Infect Immun 62, 697–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ajuebor MN, Hogaboam CM, Kunkel SL, Proudfoot AE, and Wallace JL (2001). The chemokine RANTES is a crucial mediator of the progression from acute to chronic colitis in the rat. J Immunol 166, 552–558. [DOI] [PubMed] [Google Scholar]
  3. Ansaldo E, Slayden LC, Ching KL, Koch MA, Wolf NK, Plichta DR, Brown EM, Graham DB, Xavier RJ, Moon JJ, and Barton GM (2019). Akkermansia muciniphila induces intestinal adaptive immune responses during homeostasis. Science 364, 1179–1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, Deroos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, and Rudensky AY (2013). Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Barbosa JM, Costa-de-Oliveira S, Rodrigues AG, Hanscheid T, Shapiro H, and Pina-Vaz C (2008). A flow cytometric protocol for detection of Cryptosporidium spp. Cytometry A 73, 44–47. [DOI] [PubMed] [Google Scholar]
  6. Barton ES, White DW, Cathelyn JS, Brett-McClellan KA, Engle M, Diamond MS, Miller VL, and Virgin H.W.t. (2007). Herpesvirus latency confers symbiotic protection from bacterial infection. Nature 447, 326–329. [DOI] [PubMed] [Google Scholar]
  7. Berube J, Bourdon C, Yao Y, and Rousseau S (2009). Distinct intracellular signaling pathways control the synthesis of IL-8 and RANTES in TLR1/TLR2, TLR3 or NOD1 activated human airway epithelial cells. Cell Signal 21, 448–456. [DOI] [PubMed] [Google Scholar]
  8. Bouziat R, Hinterleitner R, Brown JJ, Stencel-Baerenwald JE, Ikizler M, Mayassi T, Meisel M, Kim SM, Discepolo V, Pruijssers AJ, et al. (2017). Reovirus infection triggers inflammatory responses to dietary antigens and development of celiac disease. Science 356, 44–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bry L, Brigl M, and Brenner MB (2006). CD4+-T-cell effector functions and costimulatory requirements essential for surviving mucosal infection with Citrobacter rodentium. Infect Immun 74, 673–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJ, and Holmes SP (2016). DADA2: High-resolution sample inference from Illumina amplicon data. Nature methods 13, 581–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Cama VA, Bern C, Roberts J, Cabrera L, Sterling CR, Ortega Y, Gilman RH, and Xiao L (2008). Cryptosporidium species and subtypes and clinical manifestations in children, Peru. Emerg Infect Dis 14, 1567–1574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, Fierer N, and Knight R (2011). Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proceedings of the National Academy of Sciences of the United States of America 108 Suppl 1, 4516–4522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Chai JN, Peng Y, Rengarajan S, Solomon BD, Ai TL, Shen Z, Perry JSA, Knoop KA, Tanoue T, Narushima S, et al. (2017). Helicobacter species are potent drivers of colonic T cell responses in homeostasis and inflammation. Sci Immunol 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Charania R, Wade BE, McNair NN, and Mead JR (2020). Changes in the Microbiome of Cryptosporidium-Infected Mice Correlate to Differences in Susceptibility and Infection Levels. Microorganisms 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Checkley W, White AC Jr., Jaganath D, Arrowood MJ, Chalmers RM, Chen XM, Fayer R, Griffiths JK, Guerrant RL, Hedstrom L, et al. (2015). A review of the global burden, novel diagnostics, therapeutics, and vaccine targets for cryptosporidium. Lancet Infect Dis 15, 85–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Chen W, Harp JA, and Harmsen AG (1993a). Requirements for CD4+ cells and gamma interferon in resolution of established Cryptosporidium parvum infection in mice. Infect Immun 61, 3928–3932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Chen W, Harp JA, Harmsen AG, and Havell EA (1993b). Gamma interferon functions in resistance to Cryptosporidium parvum infection in severe combined immunodeficient mice. Infect Immun 61, 3548–3551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chudnovskiy A, Mortha A, Kana V, Kennard A, Ramirez JD, Rahman A, Remark R, Mogno I, Ng R, Gnjatic S, et al. (2016). Host-Protozoan Interactions Protect from Mucosal Infections through Activation of the Inflammasome. Cell 167, 444–456 e414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Collaborators, G.B.D.D.D. (2017). Estimates of global, regional, and national morbidity, mortality, and aetiologies of diarrhoeal diseases: a systematic analysis for the Global Burden of Disease Study 2015. Lancet Infect Dis 17, 909–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Condlova S, Horcickova M, Havrdova N, Sak B, Hlaskova L, Perec-Matysiak A, Kicia M, McEvoy J, and Kvac M (2019). Diversity of Cryptosporidium spp. in Apodemus spp. in Europe. Eur J Protistol 69, 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Cooper AM, and Khader SA (2007). IL-12p40: an inherently agonistic cytokine. Trends in immunology 28, 33–38. [DOI] [PubMed] [Google Scholar]
  22. Cortez VS, and Colonna M (2016). Diversity and function of group 1 innate lymphoid cells. Immunology letters 179, 19–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Current WL, and Reese NC (1986). A comparison of endogenous development of three isolates of Cryptosporidium in suckling mice. J Protozool 33, 98–108. [DOI] [PubMed] [Google Scholar]
  24. Current WL, Reese NC, Ernst JV, Bailey WS, Heyman MB, and Weinstein WM (1983). Human cryptosporidiosis in immunocompetent and immunodeficient persons. Studies of an outbreak and experimental transmission. The New England journal of medicine 308, 1252–1257. [DOI] [PubMed] [Google Scholar]
  25. Durai V, Bagadia P, Granja JM, Satpathy AT, Kulkarni DH, Davidson J.T.t., Wu R, Patel SJ, Iwata A, Liu TT, et al. (2019). Cryptic activation of an Irf8 enhancer governs cDC1 fate specification. Nat Immunol 20, 1161–1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, Peaper DR, Bertin J, Eisenbarth SC, Gordon JI, and Flavell RA (2011). NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 145, 745–757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Escalante NK, Lemire P, Cruz Tleugabulova M, Prescott D, Mortha A, Streutker CJ, Girardin SE, Philpott DJ, and Mallevaey T (2016). The common mouse protozoa Tritrichomonas muris alters mucosal T cell homeostasis and colitis susceptibility. J Exp Med 213, 2841–2850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Esterhazy D, Canesso MCC, Mesin L, Muller PA, de Castro TBR, Lockhart A, ElJalby M, Faria AMC, and Mucida D (2019). Compartmentalized gut lymph node drainage dictates adaptive immune responses. Nature 569, 126–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Farin HF, Karthaus WR, Kujala P, Rakhshandehroo M, Schwank G, Vries RG, Kalkhoven E, Nieuwenhuis EE, and Clevers H (2014). Paneth cell extrusion and release of antimicrobial products is directly controlled by immune cell-derived IFN-gamma. J Exp Med 211, 1393–1405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Fayer R, Santin M, and Macarisin D (2010). Cryptosporidium ubiquitum n. sp. in animals and humans. Vet Parasitol 172, 23–32. [DOI] [PubMed] [Google Scholar]
  31. Flanigan TP, and Soave R (1993). Cryptosporidiosis. Prog Clin Parasitol 3, 1–20. [DOI] [PubMed] [Google Scholar]
  32. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, et al. (2013). Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. [DOI] [PubMed] [Google Scholar]
  33. Gharpure R, Perez A, Miller AD, Wikswo ME, Silver R, and Hlavsa MC (2019). Cryptosporidiosis Outbreaks - United States, 2009–2017. MMWR Morb Mortal Wkly Rep 68, 568–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Groom JR (2019). Regulators of T-cell fate: Integration of cell migration, differentiation and function. Immunol Rev 289, 101–114. [DOI] [PubMed] [Google Scholar]
  35. Gupta J, and Nebreda AR (2014). Analysis of Intestinal Permeability in Mice. Bio-protocol 4, e1289. [Google Scholar]
  36. Hand TW, Dos Santos LM, Bouladoux N, Molloy MJ, Pagan AJ, Pepper M, Maynard CL, Elson CO 3rd, and Belkaid Y (2012). Acute gastrointestinal infection induces long-lived microbiota-specific T cell responses. Science 337, 1553–1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Heine J, Moon HW, and Woodmansee DB (1984). Persistent Cryptosporidium infection in congenitally athymic (nude) mice. Infect Immun 43, 856–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Hildner K, Edelson BT, Purtha WE, Diamond M, Matsushita H, Kohyama M, Calderon B, Schraml BU, Unanue ER, Diamond MS, et al. (2008). Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Hsieh C-S, Liang Y, Tyznik AJ, Self SG, Liggitt D, and Rudensky AY (2004). Recognition of the peripheral self by naturally arising CD25+ CD4+ T cell receptors. Immunity 21, 267–277. [DOI] [PubMed] [Google Scholar]
  40. Hsieh CS, Macatonia SE, Tripp CS, Wolf SF, O’Garra A, and Murphy KM (1993). Development of TH1 CD4+ T cells through IL-12 produced by Listeria-induced macrophages. Science 260, 547–549. [DOI] [PubMed] [Google Scholar]
  41. Huang J, Mullapudi N, Lancto CA, Scott M, Abrahamsen MS, and Kissinger JC (2004). Phylogenomic evidence supports past endosymbiosis, intracellular and horizontal gene transfer in Cryptosporidium parvum. Genome Biol 5, R88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Ise W, Kohyama M, Nutsch KM, Lee HM, Suri A, Unanue ER, Murphy TL, and Murphy KM (2010). CTLA-4 suppresses the pathogenicity of self antigen-specific T cells by cell-intrinsic and cell-extrinsic mechanisms. Nat Immunol 11, 129–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al. (2009). Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139, 485–498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Iwano S, Sugiyama M, Hama H, Watakabe A, Hasegawa N, Kuchimaru T, Tanaka KZ, Takahashi M, Ishida Y, Hata J, et al. (2018). Single-cell bioluminescence imaging of deep tissue in freely moving animals. Science 359, 935–939. [DOI] [PubMed] [Google Scholar]
  45. Jankovic D, Kullberg MC, Feng CG, Goldszmid RS, Collazo CM, Wilson M, Wynn TA, Kamanaka M, Flavell RA, and Sher A (2007). Conventional T-bet(+)Foxp3(−) Th1 cells are the major source of host-protective regulatory IL-10 during intracellular protozoan infection. J Exp Med 204, 273–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Kaplan DH, Jenison MC, Saeland S, Shlomchik WD, and Shlomchik MJ (2005). Epidermal langerhans cell-deficient mice develop enhanced contact hypersensitivity. Immunity 23, 611–620. [DOI] [PubMed] [Google Scholar]
  47. Kominsky DJ, Campbell EL, Ehrentraut SF, Wilson KE, Kelly CJ, Glover LE, Collins CB, Bayless AJ, Saeedi B, Dobrinskikh E, et al. (2014). IFN-gamma-mediated induction of an apical IL-10 receptor on polarized intestinal epithelia. J Immunol 192, 1267–1276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Kotloff KL, Nasrin D, Blackwelder WC, Wu Y, Farag T, Panchalingham S, Sow SO, Sur D, Zaidi AKM, Faruque ASG, et al. (2019). The incidence, aetiology, and adverse clinical consequences of less severe diarrhoeal episodes among infants and children residing in low-income and middle-income countries: a 12-month case-control study as a follow-on to the Global Enteric Multicenter Study (GEMS). Lancet Glob Health 7, e568–e584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Kuhls TL, Greenfield RA, Mosier DA, Crawford DL, and Joyce WA (1992). Cryptosporidiosis in adult and neonatal mice with severe combined immunodeficiency. J Comp Pathol 106, 399–410. [DOI] [PubMed] [Google Scholar]
  50. Kulkarni DH, Gustafsson JK, Knoop KA, McDonald KG, Bidani SS, Davis JE, Floyd AN, Hogan SP, Hsieh CS, and Newberry RD (2020). Goblet cell associated antigen passages support the induction and maintenance of oral tolerance. Mucosal Immunol 13, 271–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, Peterson DA, Stappenbeck TS, and Hsieh CS (2011). Peripheral education of the immune system by colonic commensal microbiota. Nature 478, 250–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Leitch GJ, and He Q (2012). Cryptosporidiosis-an overview. J Biomed Res 25, 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Liu J, Guan X, and Ma X (2005). Interferon regulatory factor 1 is an essential and direct transcriptional activator for interferon {gamma}-induced RANTES/CCl5 expression in macrophages. The Journal of biological chemistry 280, 24347–24355. [DOI] [PubMed] [Google Scholar]
  54. Lv C, Zhang L, Wang R, Jian F, Zhang S, Ning C, Wang H, Feng C, Wang X, Ren X, et al. (2009). Cryptosporidium spp. in wild, laboratory, and pet rodents in china: prevalence and molecular characterization. Appl Environ Microbiol 75, 7692–7699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Mashayekhi M, Sandau MM, Dunay IR, Frickel EM, Khan A, Goldszmid RS, Sher A, Ploegh HL, Murphy TL, Sibley LD, and Murphy KM (2011). CD8alpha(+) dendritic cells are the critical source of interleukin-12 that controls acute infection by Toxoplasma gondii tachyzoites. Immunity 35, 249–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. McDonald V, Robinson HA, Kelly JP, and Bancroft GJ (1994). Cryptosporidium muris in adult mice: adoptive transfer of immunity and protective roles of CD4 versus CD8 cells. Infect Immun 62, 2289–2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Mead JR, Arrowood MJ, Sidwell RW, and Healey MC (1991). Chronic Cryptosporidium parvum infections in congenitally immunodeficient SCID and nude mice. J Infect Dis 163, 1297–1304. [DOI] [PubMed] [Google Scholar]
  58. Mondal D, Minak J, Alam M, Liu Y, Dai J, Korpe P, Liu L, Haque R, and Petri WA Jr. (2012). Contribution of enteric infection, altered intestinal barrier function, and maternal malnutrition to infant malnutrition in Bangladesh. Clin Infect Dis 54, 185–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Morgan-Ryan UM, Fall A, Ward LA, Hijjawi N, Sulaiman I, Fayer R, Thompson RC, Olson M, Lal A, and Xiao L (2002). Cryptosporidium hominis n. sp. (Apicomplexa: Cryptosporidiidae) from Homo sapiens. J Eukaryot Microbiol 49, 433–440. [DOI] [PubMed] [Google Scholar]
  60. Morgan UM, Sturdee AP, Singleton G, Gomez MS, Gracenea M, Torres J, Hamilton SG, Woodside DP, and Thompson RC (1999). The Cryptosporidium “mouse” genotype is conserved across geographic areas. J Clin Microbiol 37, 1302–1305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Nava P, Koch S, Laukoetter MG, Lee WY, Kolegraff K, Capaldo CT, Beeman N, Addis C, Gerner-Smidt K, Neumaier I, et al. (2010). Interferon-gamma regulates intestinal epithelial homeostasis through converging beta-catenin signaling pathways. Immunity 32, 392–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Nutsch K, Chai JN, Ai TL, Russler-Germain E, Feehley T, Nagler CR, and Hsieh CS (2016). Rapid and Efficient Generation of Regulatory T Cells to Commensal Antigens in the Periphery. Cell Rep 17, 206–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Osman M, Benamrouz S, Guyot K, Baydoun M, Frealle E, Chabe M, Gantois N, Delaire B, Goffard A, Aoun A, et al. (2017a). High association of Cryptosporidium spp. infection with colon adenocarcinoma in Lebanese patients. PLoS One 12, e0189422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Osman M, El Safadi D, Benamrouz-Vanneste S, Cian A, Moriniere R, Gantois N, Delgado-Viscogliosi P, Guyot K, Bosc S, Chabe M, et al. (2017b). Prevalence, transmission, and host specificity of Cryptosporidium spp. in various animal groups from two French zoos. Parasitol Res 116, 3419–3422. [DOI] [PubMed] [Google Scholar]
  65. Petersen C (1992). Cryptosporidiosis in patients infected with the human immunodeficiency virus. Clin Infect Dis 15, 903–909. [DOI] [PubMed] [Google Scholar]
  66. Pollok RC, Farthing MJ, Bajaj-Elliott M, Sanderson IR, and McDonald V (2001). Interferon gamma induces enterocyte resistance against infection by the intracellular pathogen Cryptosporidium parvum. Gastroenterology 120, 99–107. [DOI] [PubMed] [Google Scholar]
  67. Potiron L, Lacroix-Lamande S, Marquis M, Levern Y, Fort G, Franceschini I, and Laurent F (2019). Batf3-Dependent Intestinal Dendritic Cells Play a Critical Role in the Control of Cryptosporidium parvum Infection. J Infect Dis 219, 925–935. [DOI] [PubMed] [Google Scholar]
  68. Reinhardt RL, Liang HE, and Locksley RM (2009). Cytokine-secreting follicular T cells shape the antibody repertoire. Nat Immunol 10, 385–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Ren X, Zhao J, Zhang L, Ning C, Jian F, Wang R, Lv C, Wang Q, Arrowood MJ, and Xiao L (2012). Cryptosporidium tyzzeri n. sp. (Apicomplexa: Cryptosporidiidae) in domestic mice (Mus musculus). Exp Parasitol 130, 274–281. [DOI] [PubMed] [Google Scholar]
  70. Roncarolo MG, Gregori S, Bacchetta R, Battaglia M, and Gagliani N (2018). The Biology of T Regulatory Type 1 Cells and Their Therapeutic Application in Immune-Mediated Diseases. Immunity 49, 1004–1019. [DOI] [PubMed] [Google Scholar]
  71. Sadick MD, Heinzel FP, Holaday BJ, Pu RT, Dawkins RS, and Locksley RM (1990). Cure of murine leishmaniasis with anti-interleukin 4 monoclonal antibody. Evidence for a T cell-dependent, interferon gamma-independent mechanism. J Exp Med 171, 115–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Sateriale A, Slapeta J, Baptista R, Engiles JB, Gullicksrud JA, Herbert GT, Brooks CF, Kugler EM, Kissinger JC, Hunter CA, and Striepen B (2019). A Genetically Tractable, Natural Mouse Model of Cryptosporidiosis Offers Insights into Host Protective Immunity. Cell Host Microbe 26, 135–146 e135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Scallan E, Hoekstra RM, Angulo FJ, Tauxe RV, Widdowson MA, Roy SL, Jones JL, and Griffin PM (2011). Foodborne illness acquired in the United States--major pathogens. Emerg Infect Dis 17, 7–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Schall TJ, Bacon K, Toy KJ, and Goeddel DV (1990). Selective attraction of monocytes and T lymphocytes of the memory phenotype by cytokine RANTES. Nature 347, 669–671. [DOI] [PubMed] [Google Scholar]
  75. Schoenborn JR, and Wilson CB (2007). Regulation of interferon-gamma during innate and adaptive immune responses. Adv Immunol 96, 41–101. [DOI] [PubMed] [Google Scholar]
  76. Scott P, Natovitz P, Coffman RL, Pearce E, and Sher A (1988). Immunoregulation of cutaneous leishmaniasis. T cell lines that transfer protective immunity or exacerbation belong to different T helper subsets and respond to distinct parasite antigens. J Exp Med 168, 1675–1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Shiomi H, Masuda A, Nishiumi S, Nishida M, Takagawa T, Shiomi Y, Kutsumi H, Blumberg RS, Azuma T, and Yoshida M (2010). Gamma interferon produced by antigen-specific CD4+ T cells regulates the mucosal immune responses to Citrobacter rodentium infection. Infect Immun 78, 2653–2666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN, and Garrett WS (2013). The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Solomon BD, and Hsieh CS (2016). Antigen-Specific Development of Mucosal Foxp3+RORγt+ T Cells from Regulatory T Cell Precursors. J Immunol 197, 3512–3519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Spahn TW, Ross M, von Eiff C, Maaser C, Spieker T, Kannengiesser K, Domschke W, and Kucharzik T (2008). CD4+ T cells transfer resistance against Citrobacter rodentium-induced infectious colitis by induction of Th 1 immunity. Scandinavian journal of immunology 67, 238–244. [DOI] [PubMed] [Google Scholar]
  81. Stensvold CR, Beser J, Axen C, and Lebbad M (2014). High applicability of a novel method for gp60-based subtyping of Cryptosporidium meleagridis. J Clin Microbiol 52, 2311–2319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Sulzyc-Bielicka V, Kolodziejczyk L, Jaczewska S, Bielicki D, Safranow K, Bielicki P, Kladny J, and Rogowski W (2018). Colorectal cancer and Cryptosporidium spp. infection. PLoS One 13, e0195834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Tandel J, English ED, Sateriale A, Gullicksrud JA, Beiting DP, Sullivan MC, Pinkston B, and Striepen B (2019). Life cycle progression and sexual development of the apicomplexan parasite Cryptosporidium parvum. Nat Microbiol 4, 2226–2236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Terzic J, Grivennikov S, Karin E, and Karin M (2010). Inflammation and colon cancer. Gastroenterology 138, 2101–2114 e2105. [DOI] [PubMed] [Google Scholar]
  85. Tessema TS, Schwamb B, Lochner M, Forster I, Jakobi V, and Petry F (2009). Dynamics of gut mucosal and systemic Th1/Th2 cytokine responses in interferon-gamma and interleukin-12p40 knock out mice during primary and challenge Cryptosporidium parvum infection. Immunobiology 214, 454–466. [DOI] [PubMed] [Google Scholar]
  86. Theodos CM, Sullivan KL, Griffiths JK, and Tzipori S (1997). Profiles of healing and nonhealing Cryptosporidium parvum infection in C57BL/6 mice with functional B and T lymphocytes: the extent of gamma interferon modulation determines the outcome of infection. Infect Immun 65, 4761–4769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Tussiwand R, Lee WL, Murphy TL, Mashayekhi M, Kc W, Albring JC, Satpathy AT, Rotondo JA, Edelson BT, Kretzer NM, et al. (2012). Compensatory dendritic cell development mediated by BATF-IRF interactions. Nature 490, 502–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Upton SJ, and Current WL (1985). The species of Cryptosporidium (Apicomplexa: Cryptosporidiidae) infecting mammals. J Parasitol 71, 625–629. [PubMed] [Google Scholar]
  89. Urban JF Jr., Fayer R, Chen SJ, Gause WC, Gately MK, and Finkelman FD (1996). IL-12 protects immunocompetent and immunodeficient neonatal mice against infection with Cryptosporidium parvum. J Immunol 156, 263–268. [PubMed] [Google Scholar]
  90. Vallance BA, Deng W, Jacobson K, and Finlay BB (2003). Host susceptibility to the attaching and effacing bacterial pathogen Citrobacter rodentium. Infect Immun 71, 3443–3453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Vaughan JA, Noden BH, and Beier JC (1991). Concentrations of human erythrocytes by anopheline mosquitoes (Diptera: Culicidae) during feeding. J Med Entomol 28, 780–786. [DOI] [PubMed] [Google Scholar]
  92. Welty NE, Staley C, Ghilardi N, Sadowsky MJ, Igyarto BZ, and Kaplan DH (2013). Intestinal lamina propria dendritic cells maintain T cell homeostasis but do not affect commensalism. J Exp Med 210, 2011–2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Wilke G, Funkhouser-Jones LJ, Wang Y, Ravindran S, Wang Q, Beatty WL, Baldridge MT, VanDussen KL, Shen B, Kuhlenschmidt MS, et al. (2019). A Stem-Cell-Derived Platform Enables Complete Cryptosporidium Development In Vitro and Genetic Tractability. Cell Host Microbe 26, 123–134 e128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Wilke G, Ravindran S, Funkhouser-Jones L, Barks J, Wang Q, VanDussen KL, Stappenbeck TS, Kuhlenschmidt TB, Kuhlenschmidt MS, and Sibley LD (2018). Monoclonal Antibodies to Intracellular Stages of Cryptosporidium parvum Define Life Cycle Progression In Vitro. mSphere 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Wong P, Goldrath AW, and Rudensky AY (2000). Competition for specific intrathymic ligands limits positive selection in a TCR transgenic model of CD4+ T cell development. J Immunol 164, 6252–6259. [DOI] [PubMed] [Google Scholar]
  96. Xiao L (2010). Molecular epidemiology of cryptosporidiosis: an update. Exp Parasitol 124, 80–89. [DOI] [PubMed] [Google Scholar]
  97. Xu M, Pokrovskii M, Ding Y, Yi R, Au C, Harrison OJ, Galan C, Belkaid Y, Bonneau R, and Littman DR (2018). c-MAF-dependent regulatory T cells mediate immunological tolerance to a gut pathobiont. Nature 554, 373–377. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

replacement_suppl_file

Data Availability Statement

TCR and 16S rRNA sequencing data have been deposited at European Nucleotide Archive (ENA) and are publicly available as of the date of publication. Accession numbers are listed in the key resources table. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

KEY RESOURCES TABLE.

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
anti-IFNγ Clone H22 Leinco Cat#I-438
PIP Leinco Cat#P-376
anti-Thy1.2 clone 30H12 BioXCell Cat#BE0066
LTF-2 BioXCell Cat#BE0090
anti-NK1.1 clone PK136 BioXCell Cat##BE0036
C1.18.4 BioXCell Cat#BE0085
anti-IL10R BioXCell Cat#BE0050
anti-CD3ε BioXCell Cat#BE0001-1
anti-MHCII BioXCell Cat#BE010
anti-CD28 BioXCell Cat#BE0015-1
anti-TGFβ BioXCell Cat#BE0057
anti-IL-12 BioXCell Cat#BE0052
anti-IFNγ BioXCell Cat#BE0054
anti-IL-4 BioXCell Cat#BE0045
Anti-mouse CD3ε (clone#145-2C11) FITC, PE, and BV421 BioLegend Cat#100305/07/35
Anti-mouse/human B220 (clone#RA3-6B2) APC/Cy7, A700, and PE/Cy7 BioLegend Cat#103223/31/22
Anti-mouse CD19 (clone#6D5) APC/Cy7 BioLegend Cat#115529
Anti-mouse I-Ab (clone#AF6-120.1) APC and PerCP/Cy5.5 BioLegend Cat#116417/15
Anti-mouse CD11c (clone#N418) PE/Cy7 and BV605 BioLegend Cat#117317/33
Anti-mouse/human CD11b (clone#M1/70) BV711 BioLegend Cat#101241
Anti-mouse CD103 (clone#2E7) BV421 BioLegend Cat#121421
Anti-mouse F4/80 (clone#BM8) PE/Cy7 BioLegend Cat#123113
Anti-mouse CD4 (clone#RM4-5) BV711, PE, PB, and BV605 BioLegend Cat#100549/11/34/48
Anti-mouse CD25 (clone#PC61) APC, BV605, PerCP/Cy5.5, and PE/Cy7 BioLegend Cat#102011/35/29/15
Anti-mouse/human CD44 (clone#IM7) APC/Cy7 and BV605 BioLegend Cat#103027/47
Anti-mouse CD62L (clone#MEL-14) APC/Cy7 and BV605 BioLegend Cat#104427/37
Anti-mouse FOXP3 (clone#FJK-16s) FITC ThermoFisher Cat#11-5773-82
Anti-mouse GATA3 (clone#16E10A23) PE BioLegend Cat#653803
Anti-mouse RORγt (clone#B2D) APC ThermoFisher Cat#17-6981-80
Anti-mouse TBET (clone#4B10) PE/Cy7 BioLegend Cat#644823
Anti-mouse CXCR3 (clone#CXCR3-173) BV421, PE, and APC BioLegend Cat#126521/06/12
Anti-mouse Thy1.1 (clone#OX-7) BV711, PE/Cy7, and PerCP/Cy5.5 BioLegend Cat#202539/18/16
Anti-mouse CD80 (clone#16-10A1) APC BioLegend Cat#104713
Anti-mouse CD45.1 (clone#A20) PerCP/Cy5.5, PE, and A700 BioLegend Cat#110728/08/24
Anti-mouse CD45.2 (clone#104) APC, A700, and PE BioLegend Cat#109814/22/08
Anti-mouse VB6 (clone#RR4-7) PE and APC BioLegend Cat#140003/5
Anti-mouse VB5 (clone#MR9-4) PE BioLegend Cat#139503
Anti-mouse Va2 (clone#B20.1) APC/Cy7 and PerCP/Cy5.5 BioLegend Cat#127818/14
Bacterial and virus strains
Citrobacter rodentium Gift from M. Colonna (Wash. U.)
Biological samples
Cryptosporidium tyzzeri (Ct-STL) Our mouse colony N/A
Chemicals, peptides, and recombinant proteins
FITC-Dextran Sigma Cat#FD4
Ovalbumin (Grade V) Sigma Cat#A5503
Incomplete Freund’s Adjuvant Sigma Cat#F5506
Ovalbumin (Grade VI) Sigma Cat#A2512
Dextran Sodium Sulfate TdB Labs Cat#9011-18-1
Critical commercial assays
Quick-DNA Fecal/Soil Microbe Prep Kit Zymo Cat#D6010
Luminaris Color HiGreen qPCR Master Mix ThermoFisher Cat#K0391
CountBright Absolute Counting Beads ThermoFisher Cat#C36950
Crypt-a-Glo Waterborne Cat#A400FLR
TransIT-293 ThermoFisher Cat#MIR2700
CellTrace Violet (CTV) ThermoFisher Cat#C34571
SuperScript III Reverse Transcriptase ThermoFisher Cat#18080093
Accuzyme DNA Polymerase Bioline Cat#BIO-21052
Foxp3/Transcription Factor Staining Buffer Set ThermoFisher Cat#00-5523-00
Bio-Plex Mouse Cytokine 23-plex Assay BioRad Cat#M60009RDPD
Deposited data
TCR Sequencing This paper ENA: accession #PRJEB46540
16S rRNA This paper ENA: accession #PRJEB46540
Experimental models: Cell lines
NFAT-GFP T cell hybridoma Gift from K. Murphy (Wash. U.) (Ise et al., 2010) N/A
B16 flt3L melanoma Gift from G. Dranoff (Mach et al., 2000) N/A
Experimental models: Organisms/strains
Mouse: C57BL/6-Ly5.1 Charles River Cat#494
Mouse: C57BL/6NCrl (Ly5.2) Charles River Cat#027
Mouse: C57BL/6J JAX Cat#000664
Mouse: Rag1−/− JAX Cat#002216
Mouse: hLangDTA Gift from D. Kaplan (U. Pitt.) (Kaplan et al., 2005) N/A
Mouse: Irf8 +32 5’−/− Gift from K. Murphy (Wash. U.) (Durai et al., 2019) N/A
Mouse: Foxp3IRES-GFP JAX Cat#006772
Mouse: Foxp3IRES-Thy1.1 Gift from A. Rudensky (MSKCC) (Liston et al., 2008) N/A
Mouse: IL-17AIRES-GFP JAX Cat#18472
Mouse: IL12p40IRES-YFP JAX Cat#015864
Mouse: Ifngr1−/− JAX Cat#003288
Mouse: C3H/HeOuJ JAX Cat#000635
Mouse: IFNγIRES-YFP Gift from R. Locksley (UCSF) (Reinhardt et al., 2009) N/A
Mouse: TCli TCRβ Transgenic Gift from A. Rudensky (MSKCC) (Wong et al., 2000) N/A
Mouse: TCli TCRαβ Transgenic Gift from A. Rudensky (MSKCC) (Wong et al., 2000) N/A
Mouse: Tcra−/− Jax Cat#002116
Mouse: C3H/HeNCrl Charles River Cat#025
Mouse: OT-II Transgenic JAX Cat#004194
Oligonucleotides
Primers for TCR-seq, see Table S1 N/A N/A
Other primers, see Table S5 N/A N/A
Recombinant DNA
(RG1/RG2/RG3/LA1/LA2/LA3/LA4/CT6)-IRES-Thy1.1 This paper and (Hsieh et al., 2004; Lathrop et al., 2011) N/A
RG1-P2A-Akaluc This paper and (Iwano et al., 2018) N/A
CT2-P2A-Akaluc This paper and (Iwano et al., 2018) N/A
Software and algorithms
Prism v9 https://www.graphpad.com N/A
FlowJo v10 https://www.flowjo.com N/A
ImageJ v1.53a https://imagej.nih.gov/ij/download.html N/A
MegAlign v16 https://www.dnastar.com N/A
R https://www.r-project.org/ N/A
dada2 v1.14 https://www.bioconductor.org/packages/release/bioc/html/dada2.html N/A
Vegan v2.4 https://cran.r-project.org/web/packages/vegan/index.html N/A
Phyloseq v1.32 https://www.bioconductor.org/packages/release/bioc/html/phyloseq.html N/A
DESeq2 v1.28 https://bioconductor.org/packages/release/bioc/html/DESeq2.html N/A
Gbm v2.1 https://cran.r-project.org/web/packages/gbm/index.html N/A

RESOURCES