Abstract
Diarrhoeal diseases are the second leading cause of death in children worldwide. Epidemiological studies show that co-infection with the protozoan parasite Giardia intestinalis decreases diarrhoeal severity. Here we show a high incidence of asymptomatic Giardia infection in school-aged children from Nigeria. In a mouse model, Giardia induced a Type 2 mucosal immune response, characterized by antigen-specific Th2 cells, IL-25, Type 2 cytokines, and goblet cell hyperplasia. Single-cell RNA sequencing and multiparameter flow cytometry revealed expansion of IL-10-producing Th2 cells, which promoted parasite persistence and protected against Toxoplasma gondii-induced ileitis and dextran sulfate sodium-induced colitis. This protective effect was STAT6 dependent, as IL-4R blockade or STAT6 deficiency impaired IL-10+ Th2 responses, resulting in Th1/Th17-driven tissue damage, inflammation and clearance of Giardia infection. Our findings demonstrate that Giardia reshapes mucosal immunity toward a Type 2 response, facilitating parasitism and conferring mutualistic protection from inflammatory pathologies, highlighting a key role for protists in mucosal defence regulation.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout






Similar content being viewed by others
Data availability
The data supporting the findings of this study are available in the Article, Extended Data and Source Data. The raw single-cell RNA-seq data generated in this study have been deposited in the NCBI SRA database under BioProject ID PRJNA1082359, available at https://www.ncbi.nlm.nih.gov/bioproject/PRJNA1082359. The processed Seurat objects and the associated R code are available at https://doi.org/10.5281/zenodo.15390320 (ref. 72). Source data are provided with this paper.
References
Sardinha-Silva, A., Alves-Ferreira, E. V. C. & Grigg, M. E. Intestinal immune responses to commensal and pathogenic protozoa. Front. Immunol. 13, 963723 (2022).
McGhee, J. R. & Fujihashi, K. Inside the mucosal immune system. PLoS Biol. 10, e1001397 (2012).
GBD 2016 Diarrhoeal Disease Collaborators. Estimates of the global, regional, and national morbidity, mortality, and aetiologies of diarrhoea in 195 countries: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Infect. Dis. 18, 1211–1228 (2018).
Key Facts (WHO, 2017).
Hajare, S. T., Chekol, Y. & Chauhan, N. M. Assessment of prevalence of Giardia lamblia infection and its associated factors among government elementary school children from Sidama zone, SNNPR, Ethiopia. PLoS ONE 17, e0264812 (2022).
Esch, K. J. & Petersen, C. A. Transmission and epidemiology of zoonotic protozoal diseases of companion animals. Clin. Microbiol. Rev. 26, 58–85 (2013).
Dixon, B. R. Giardia duodenalis in humans and animals – transmission and disease. Res. Vet. Sci. 135, 283–289 (2021).
Adam, E. A., Yoder, J. S., Gould, L. H., Hlavsa, M. C. & Gargano, J. W. Giardiasis outbreaks in the United States, 1971–2011. Epidemiol. Infect. 144, 2790–2801 (2016).
Muhsen, K., Cohen, D. & Levine, M. M. Can Giardia lamblia infection lower the risk of acute diarrhea among preschool children? J. Trop. Pediatr. 60, 99–103 (2014).
Veenemans, J. et al. Protection against diarrhea associated with Giardia intestinalis is lost with multi-nutrient supplementation: a study in Tanzanian children. PLoS Negl. Trop. Dis. 5, e1158 (2011).
Cotton, J. A. et al. Giardia duodenalis infection reduces granulocyte infiltration in an in vivo model of bacterial toxin-induced colitis and attenuates inflammation in human intestinal tissue. PLoS ONE 9, e109087 (2014).
Oberhelman, R. A. et al. Asymptomatic salmonellosis among children in day-care centers in Merida, Yucatan, Mexico. Pediatr. Infect. Dis. J. 20, 792–797 (2001).
Wang, L. et al. Concurrent infections of Giardia duodenalis, Enterocytozoon bieneusi, and Clostridium difficile in children during a cryptosporidiosis outbreak in a pediatric hospital in China. PLoS Negl. Trop. Dis. 7, e2437 (2013).
Bhavnani, D., Goldstick, J. E., Cevallos, W., Trueba, G. & Eisenberg, J. N. Synergistic effects between rotavirus and coinfecting pathogens on diarrheal disease: evidence from a community-based study in northwestern Ecuador. Am. J. Epidemiol. 176, 387–395 (2012).
Siwila, J., Phiri, I. G., Enemark, H. L., Nchito, M. & Olsen, A. Intestinal helminths and protozoa in children in pre-schools in Kafue district, Zambia. Trans. R. Soc. Trop. Med. Hyg. 104, 122–128 (2010).
Fraser, D. et al. Natural history of Giardia lamblia and Cryptosporidium infections in a cohort of Israeli Bedouin infants: a study of a population in transition. Am. J. Trop. Med. Hyg. 57, 544–549 (1997).
Ish-Horowicz, M. et al. Asymptomatic giardiasis in children. Pediatr. Infect. Dis. J. 8, 773–779 (1989).
Waldram, A., Vivancos, R., Hartley, C. & Lamden, K. Prevalence of Giardia infection in households of Giardia cases and risk factors for household transmission. BMC Infect. Dis. 17, 486 (2017).
Das, R. et al. Symptomatic and asymptomatic enteric protozoan parasitic infection and their association with subsequent growth parameters in under five children in South Asia and sub-Saharan Africa. PLoS Negl. Trop. Dis. 17, e0011687 (2023).
Rogawski, E. T. et al. Determinants and impact of Giardia infection in the first 2 years of life in the MAL-ED birth cohort. J. Pediatr. Infect. Dis. Soc. 6, 153–160 (2017).
Dann, S. M. et al. IL-17A promotes protective IgA responses and expression of other potential effectors against the lumen-dwelling enteric parasite Giardia. Exp. Parasitol. 156, 68–78 (2015).
Dreesen, L. et al. Giardia muris infection in mice is associated with a protective interleukin 17A response and induction of peroxisome proliferator-activated receptor alpha. Infect. Immun. 82, 3333–3340 (2014).
Munoz-Cruz, S. et al. Giardia lamblia: identification of molecules that contribute to direct mast cell activation. Parasitol. Res. 117, 2555–2567 (2018).
Singer, S. M., Fink, M. Y. & Angelova, V. V. Recent insights into innate and adaptive immune responses to Giardia. Adv. Parasitol. 106, 171–208 (2019).
Solaymani-Mohammadi, S. & Singer, S. M. Host immunity and pathogen strain contribute to intestinal disaccharidase impairment following gut infection. J. Immunol. 187, 3769–3775 (2011).
Adam, R. D. Giardia duodenalis: biology and pathogenesis. Clin. Microbiol. Rev. 34, e0002419 (2021).
Saghaug, C. S. et al. Human memory CD4+ T cell immune responses against Giardia lamblia. Clin. Vaccin. Immunol. 23, 11–18 (2016).
Yordanova, I. A. et al. RORγt+ Treg to Th17 ratios correlate with susceptibility to Giardia infection. Sci. Rep. 9, 20328 (2019).
Bartelt, L. A. et al. Persistent G. lamblia impairs growth in a murine malnutrition model. J. Clin. Invest. 123, 2672–2684 (2013).
Bartelt, L. A. et al. Cross-modulation of pathogen-specific pathways enhances malnutrition during enteric coinfection with Giardia lamblia and enteroaggregative Escherichia coli. PLoS Pathog. 13, e1006471 (2017).
Dann, S. M., Le, C. H. Y., Hanson, E. M., Ross, M. C. & Eckmann, L. Giardia infection of the small intestine induces chronic colitis in genetically susceptible hosts. J. Immunol. 201, 548–559 (2018).
Oberhuber, G., Mesteri, I., Kopf, W. & Muller, H. Demonstration of trophozoites of G. lamblia in ileal mucosal biopsy specimens may reveal giardiasis in patients with significantly inflamed parasite-free duodenal mucosa. Am. J. Surg. Pathol. 40, 1280–1285 (2016).
Fink, M. Y., Shapiro, D. & Singer, S. M. Giardia lamblia: laboratory maintenance, lifecycle induction, and infection of murine models. Curr. Protoc. Microbiol. 57, e102 (2020).
Rogawski, E. T. et al. Use of quantitative molecular diagnostic methods to investigate the effect of enteropathogen infections on linear growth in children in low-resource settings: longitudinal analysis of results from the MAL-ED cohort study. Lancet Glob. Health 6, e1319–e1328 (2018).
Kiner, E. et al. Gut CD4+ T cell phenotypes are a continuum molded by microbes, not by T(H) archetypes. Nat. Immunol. 22, 216–228 (2021).
Liesenfeld, O., Kosek, J., Remington, J. S. & Suzuki, Y. Association of CD4+ T cell-dependent, interferon-gamma-mediated necrosis of the small intestine with genetic susceptibility of mice to peroral infection with Toxoplasma gondii. J. Exp. Med. 184, 597–607 (1996).
Liesenfeld, O. Oral infection of C57BL/6 mice with Toxoplasma gondii: a new model of inflammatory bowel disease? J. Infect. Dis. 185, S96–S101 (2002).
Oldenhove, G. et al. Decrease of Foxp3+ Treg cell number and acquisition of effector cell phenotype during lethal infection. Immunity 31, 772–786 (2009).
Singer, S. M. & Nash, T. E. T-cell-dependent control of acute Giardia lamblia infections in mice. Infect. Immun. 68, 170–175 (2000).
Gazzinelli-Guimaraes, P. H. et al. Eosinophil trafficking in allergen-mediated pulmonary inflammation relies on IL-13-driven CCL-11 and CCL-24 production by tissue fibroblasts and myeloid cells. J. Allergy Clin. Immunol. Glob. 2, 100131 (2023).
Metenou, S. et al. At homeostasis filarial infections have expanded adaptive T regulatory but not classical Th2 cells. J. Immunol. 184, 5375–5382 (2010).
Jimenez, J. C. et al. Antibody and cytokine responses to Giardia excretory/secretory proteins in Giardia intestinalis-infected BALB/c mice. Parasitol. Res. 113, 2709–2718 (2014).
Jimenez, J. C., Fontaine, J., Grzych, J. M., Capron, M. & Dei-Cas, E. Antibody and cytokine responses in BALB/c mice immunized with the excreted/secreted proteins of Giardia intestinalis: the role of cysteine proteases. Ann. Trop. Med. Parasitol. 103, 693–703 (2009).
Al-Mekhlafi, M. S. et al. Giardiasis as a predictor of childhood malnutrition in Orang Asli children in Malaysia. Trans. R. Soc. Trop. Med. Hyg. 99, 686–691 (2005).
Botero-Garces, J. H., Garcia-Montoya, G. M., Grisales-Patino, D., Aguirre-Acevedo, D. C. & Alvarez-Uribe, M. C. Giardia intestinalis and nutritional status in children participating in the complementary nutrition program, Antioquia, Colombia, May to October 2006. Rev. Inst. Med. Trop. Sao Paulo 51, 155–162 (2009).
Nematian, J., Gholamrezanezhad, A. & Nematian, E. Giardiasis and other intestinal parasitic infections in relation to anthropometric indicators of malnutrition: a large, population-based survey of schoolchildren in Tehran. Ann. Trop. Med. Parasitol. 102, 209–214 (2008).
Shaima, S. N. et al. Anthropometric indices of Giardia-infected under-five children presenting with moderate-to-severe diarrhea and their healthy community controls: data from the Global Enteric Multicenter Study. Children https://doi.org/10.3390/children8121186 (2021).
Barash, N. R., Maloney, J. G., Singer, S. M. & Dawson, S. C. Giardia alters commensal microbial diversity throughout the murine gut. Infect. Immun. 85, e00948-16 (2017).
Fekete, E., Allain, T., Siddiq, A., Sosnowski, O. & Buret, A. G. Giardia spp. and the gut microbiota: dangerous liaisons. Front. Microbiol. 11, 618106 (2020).
Gazzinelli-Guimaraes, P. H. & Nutman, T. B. Helminth parasites and immune regulation. F1000Res. https://doi.org/10.12688/f1000research.15596.1 (2018).
Cancado, G. G. et al. Hookworm products ameliorate dextran sodium sulfate-induced colitis in BALB/c mice. Inflamm. Bowel Dis. 17, 2275–2286 (2011).
Ferreira, I. et al. Hookworm excretory/secretory products induce interleukin-4 (IL-4)+ IL-10+ CD4+ T cell responses and suppress pathology in a mouse model of colitis. Infect. Immun. 81, 2104–2111 (2013).
Yang, X. et al. Excretory/secretory products from Trichinella spiralis adult worms ameliorate DSS-induced colitis in mice. PLoS ONE 9, e96454 (2014).
Eichenberger, R. M. et al. Hookworm secreted extracellular vesicles interact with host cells and prevent inducible colitis in mice. Front. Immunol. 9, 850 (2018).
Kamda, J. D. & Singer, S. M. Phosphoinositide 3-kinase-dependent inhibition of dendritic cell interleukin-12 production by Giardia lamblia. Infect. Immun. 77, 685–693 (2009).
Banik, S. et al. Giardia duodenalis arginine deiminase modulates the phenotype and cytokine secretion of human dendritic cells by depletion of arginine and formation of ammonia. Infect. Immun. 81, 2309–2317 (2013).
Steinfelder, S. et al. The major component in schistosome eggs responsible for conditioning dendritic cells for Th2 polarization is a T2 ribonuclease (omega-1). J. Exp. Med. 206, 1681–1690 (2009).
Johnston, C. J. C. et al. A structurally distinct TGF-β mimic from an intestinal helminth parasite potently induces regulatory T cells. Nat. Commun. 8, 1741 (2017).
Smyth, D. J. et al. Oral delivery of a functional algal-expressed TGF-β mimic halts colitis in a murine DSS model. J. Biotechnol. 340, 1–12 (2021).
Smyth, D. J. et al. Protection from T cell-dependent colitis by the helminth-derived immunomodulatory mimic of transforming growth factor-β, Hp-TGM. Discov. Immunol. 2, kyad001 (2023).
Howitt, M. R. et al. Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut. Science 351, 1329–1333 (2016).
Lei, W. et al. Activation of intestinal tuft cell-expressed Sucnr1 triggers type 2 immunity in the mouse small intestine. Proc. Natl Acad. Sci. USA 115, 5552–5557 (2018).
Nadjsombati, M. S. et al. Detection of succinate by intestinal tuft cells triggers a type 2 innate immune circuit. Immunity 49, 33–41.e7 (2018).
Schneider, C. et al. A metabolite-triggered tuft cell-ILC2 circuit drives small intestinal remodeling. Cell 174, 271–284.e14 (2018).
Chudnovskiy, A. et al. Host–protozoan interactions protect from mucosal infections through activation of the inflammasome. Cell 167, 444–456.e14 (2016).
Escalante, N. K. et al. The common mouse protozoa Tritrichomonas muris alters mucosal T cell homeostasis and colitis susceptibility. J. Exp. Med. 213, 2841–2850 (2016).
Verweij, J. J. et al. Simultaneous detection of Entamoeba histolytica, Giardia lamblia, and Cryptosporidium parvum in fecal samples by using multiplex real-time PCR. J. Clin. Microbiol. 42, 1220–1223 (2004).
Wirtz, S. et al. Chemically induced mouse models of acute and chronic intestinal inflammation. Nat. Protoc. 12, 1295–1309 (2017).
Kim, E., Tran, M., Sun, Y. & Huh, J. R. Isolation and analyses of lamina propria lymphocytes from mouse intestines. STAR Protoc. 3, 101366 (2022).
Oliveira, F. M. et al. Susceptibility to Entamoeba histolytica intestinal infection is related to reduction in natural killer T-lymphocytes in C57BL/6 mice. Infect. Dis. Rep. 4, e27 (2012).
Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).
Sardinha-Silva, A. et al. scRNA-seq for article: Giardia intestinalis-induced Type 2 mucosal immunity attenuates bystander intestinal inflammation - filtered and annotated data (2.1). Zenodo https://doi.org/10.5281/zenodo.15390320 (2025).
Acknowledgements
We thank B. Joshua, D. Samuel, M. Ali, N. Victor, A. Onekutu and E. Effanga who coordinated the samples collection in Nigeria, as well as administered the questionnaire used to establish whether infection was associated with symptomatic disease. We also thank C. de Oliveira Silva Souza and P. Loke from the Type 2 Immunity Section, LPD, NIAID, for providing us the STAT6−/− mice, and S. Ganesan from the Biological Imaging Section, RTB, NIAID, for assistance with histology image scanning. This work was supported by the Division of Intramural Research of the National Institute of Allergy and Infectious Diseases (NIAID) at the National Institutes of Health, and NIH extramural grant AI109591 to S.M.S.
Author information
Authors and Affiliations
Contributions
A.S.-S., C.H.C., S.M.S. and M.E.G. conceptualized the project. A.S.-S., P.H.G.-G., C.H.C. and T.R.F. designed the methodology. A.S.-S., P.H.G.G., O.G.A., F.M.S.O., T.R.F., E.V.C.A.-F., E.T.T., B.G. and M.Y.F. conducted investigation. A.S.-S. and P.H.G.G. wrote the original manuscript draft. A.S.-S., P.H.G.-G., S.M.S. and M.E.G. reviewed and edited the manuscript. S.M.S. and M.E.G. acquired funding and resources. M.E.G. supervised the project.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Microbiology thanks Peter Cook and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 Nigeria map showing sampled states.
1. Benue (latitude: 7° 19’ 60.00” N and longitude: 8° 44’ 59.99” E); 2. Cross River (latitude: 4° 34’ 59.99” N and longitude: 8° 24’ 59.99” E); 3. Enugu (latitude: 6° 27’ 9.60” N and longitude: 7° 30’ 37.20” E); 4. Jigawa (latitudes 11.00°N to 13.00°N and longitudes 8.00°E to 10.15°E); 5. Kano (latitude: 12° 00’ 0.43” N and longitude: 8° 31’ 0.19” E); 6. Plateau (latitudes 8°24’ N to 10°30’ N and longitudes 8°32’ E to 10°38’).
Extended Data Fig. 2 Cytokine production kinetics across the three segments from the small intestine in Giardia-infected mice.
Levels of IFN-γ (a), IL-4 (b), IL-13 (c), IL-17A (d), and IL-10 (e) in the three different sections of the small intestine tissue homogenate (normalized by 100 mg of tissue) from naïve (n = 3) or Giardia-infected. (n-5) mice at 5-, 7-, 9-, 12-, and 21-days post-infection (measured by Luminex). Data are represented as mean ± SEM for each time point and significance was calculated with one-way ANOVA test followed by Sidak’s multiple comparisons test. *p≤0.05, **p≤0.01, ***p≤0.001.
Extended Data Fig. 3 Gating strategy for immunophenotypic analysis by Flow Cytometry.
(a) The gating strategy 1 was used for the overall immunophenotypic analysis of the experiments reported in Figs. 1b, 1g, 2d, 2g, 4b, 4c, 4i, 5f, 5k, 6f, 6l, and Extended Data Fig. 6b. Briefly, singlets (a) were gated followed by subsequent lamina propria cell gating by FSC-A vs SSC-A (b). (c) Live hematopoietic cells were gated as CD45/Alexa Fluor700+ and Live/Dead/UV496−. (d) B cells and myeloid cells were then excluded by gating as CD19/PE-Cy5− and CD11b/BV605−. (e) CD4+ T cells were further gated as TCRβ/BUV737+ and CD4/BV510+. Subsets of CD4+ T cells were further subdivided into either the single or co-expression of FoxP3/BV421 in combination with single or co-expression of Tbet/PE-Cy7 (f), GATA3/APC (h) or RORγt/PE-CF594 (k). Finally, IFN-γ/BV650 (g), IL-10/BV711 (i), IL-13/PE (j), and IL-17A/PerCP-Cy5.5 (l) were analyzed into the populations of Tbet+ Th1 cells, GATA3+ Th2 cells, and RORγt+ Th17 cells, respectively. (b) The gating strategy 2 was design specifically for the identification of the major sources of IL-10 in the lamina propria of either naïve- and Giardia-infected IL-10 GFP reporter mice (Fig. 2f). For this, live CD45/Alex Fluor 700+ lamina propria cells (a, b) were gated for the IL-10 GFP expression (d), using a WT mouse as a control for a negative gate (c). (e) IL-10 producing hematopoietic cells were further characterized as either TCRβ/BUV737+ T cells, or CD11b/BV605+ cells or TCRβ−CD11b− cells. (f) T cells were subdivided as CD4/BV510+ cells or CD8β/APC+ cells. (g) CD64/BV421+ macrophages were identified within the CD11b+ cells; and (h) CD64/BV421−, CD11c+/PE-CF596 + , MHCII+/BV711+ cells were classified as Dendritic Cells (DCs). (i) Non-T and non-myeloid cells were further gated as CD19/PE-Cy5+ cells or TCRγδ/PerCP.Cy5.5 T cells. (j) Finally, IL-10 producing CD45+TCRβ−CD11b−CD19−TCRγδ−CD90/BV785+ cells were classified as innate lymphoid cells (ILCs).
Extended Data Fig. 4 Molecular characterization of IL-10 producing CD4+ T cells in the small intestine lamina propria.
(a) UMAP plot showing the clustering analysis of the merged dataset of all sorted IL-10 producing CD4+ T subsets in the small intestine lamina propria from both naïve and Giardia-infected IL-10-GFP reporter mice. The plot distinctly identifies five key clusters, corresponding to Th1, Th2, Th17, Treg, and Th2 Treg cells subset. (b) Dot plot graph highlighting the top10 highly expressed genes within each cluster of IL-10 producing CD4+ T cells in the small intestine lamina propria from naïve and Giardia-infected mice. (c) UMAP plot of Giardia-induced IL-10-producing CD4+ T cells five clusters mapped against to public available scRNA-seq data set for CD4+ T cells isolated from lamina propria of mice infected with different pathogens (Kiner et al. 2021). (d) dot plot graph highlighting the top20 highly expressed genes in the Th2 cells induced by Heligmosomoides polygyrus (Kiner et al., 2021) within each cluster of IL-10 producing CD4+ T cells in the small intestine lamina propria from naïve and Giardia-infected mice.
Extended Data Fig. 5 Giardia induces expansion of ILC2s in the small intestine in a STAT6-dependent manner.
(a) Gating strategy for immunophenotypic analysis of ILCs by flow cytometry. (b-d) Scatter plot graphs indicating the frequency of ILC1 (CD90.2+IFN-γ+), ILC2 (CD90.2+IL-13+), and ILC3 (CD90.2+IL-17A+) cells in the small intestine lamina propria of naïve (n = 4) Giardia-infected (n = 4) mice (7 d.p.i.). Gated on Live CD45+CD19−CD11b−TCRβ−CD4−. Data are represented as mean ± SEM for each time point and significance was calculated with one-way ANOVA test followed by Sidak’s multiple comparisons test. *p≤0.05, **p≤0.01, ***p≤0.001.
Extended Data Fig. 6 The effect of Giardia chronicity in the co-infection with T. gondii.
(a) Bioluminescent detection in photons/sec/cm2 shows Toxoplasma burden in vivo (7 d.p.T.i.) in mice co-infected (n = 18) or not (n = 18) with Giardia GS/M strain three days before. (b) Scatter plot graphs indicating the frequency of Th1 (FoxP3−Tbet+), total Treg (FoxP3+), GATA3+ Treg (GATA3+FoxP3+), Th2 (GATA3−Foxp3+), and IL-10+ Th2 (FoxP3−GATA3+IL-10+) cells in the small intestine lamina propria of naïve (n = 4), Giardia- (three days before, d-3) (n = 4), Toxoplasma- (n = 4), or Giardia (d-3)+Toxoplasma-infected (n = 4) mice 4 days post-Toxoplasma infection (4 d.p.T.i.). Gated on Live CD45+TCRβ+CD4+. (c) IL-10 levels in the proximal small intestine of naïve (n = 4) and Giardia-chronically infected (n = 5 per group) mice (4 weeks post-infection, 4 wks). (d) Representative image of H&E staining of the proximal small intestine from naïve, Toxoplasma-, Giardia (d-3)+Toxoplasma-, or Giardia (4 wks)+Toxoplasma-infected mice (8 days-post Toxoplasma infection). Scale bars represent 100 μm. Data are represented as mean ± SEM for each time point and significance was calculated with one-way ANOVA test followed by Sidak’s multiple comparisons test. *p≤0.05, **p≤0.01, ***p≤0.001. Data are representative of two independent (a-b) and one (c-d) experiment.
Extended Data Fig. 7 IL-4R blockage reduces Giardia-mediated protection against bystander intestinal inflammation.
(a) Experimental schematic. Female WT mice (C57BL/6 background) were perorally infected with 1×106 Giardia GS/M strain trophozoites and three days later mice were perorally co-infected with 10 cysts of Toxoplasma gondii, followed by intraperitoneal injection of anti-IL-4R (50 μg) or anti-IL10R (100 μg) for five consecutive days. Euthanasia was performed 8 days post-Toxoplasma infection (8 d.p.T.i.). Created with BioRender. (b) Body weight loss of WT mice co-infected with Giardia and Toxoplasma followed treatment with isotype control (n = 5), anti-IL-4R (n = 5) or anti-IL-10R (n = 5) were monitored daily. (c) Representative image of H&E staining of the jejunum from WT mice co-infected with Giardia and Toxoplasma treated with isotype control (n = 5), anti-IL-4R (n = 5) or anti-IL-10R (n = 5) and scatter plot graph showing the histopathology score (8 d.p.T.i.). Scale bars represent 500 μm (top) and 300 μm (bottom). (d) Experimental schematic. Female WT mice (C57BL/6 background) were perorally infected with 1×106 Giardia GS/M strain trophozoites and three days later mice were administered 2% DSS drinking water for 7 consecutive days, followed by intraperitoneal injection of anti-IL-4R (50 μg) or anti-IL10R (100 μg) for five consecutive days. Euthanasia was performed on day 8 (8 d.p.t.). Created with BioRender. (e) Body weight loss of WT Giardia-infected and DSS-treated mice followed isotype control (n = 4), anti-IL-4R (n = 5) or anti-IL-10R (n = 5) injection. (f) Representative image of H&E staining of the colon from WT mice Giardia-infected and DSS-treated mice followed isotype control (n = 4), anti-IL-4R (n = 5) or anti-IL-10R (n = 5) injection and scatter plot graph showing the histopathology score (8 d.p.t.). Scale bars represent 500 μm (top) and 300 μm (bottom). Figure symbols in c and f: [*] inflammatory infiltrate in the mucosa; [$] inflammatory infiltrate in the submucosa; [#] inflammatory infiltrate in the muscle layer; [arrow] mucosal ulceration; [short arrow] mucosal desquamation. Data are represented as mean ± SEM and significance was calculated with one-way ANOVA test followed by Sidak’s multiple comparisons test (c,f). *p≤0.05, **p≤0.01, ***p≤0.001, ****p≤0.0001. Data are representative of one independent experiment (n = 5).
Supplementary information
Supplementary Tables 1–3
Supplementary Table 1. Giardia intestinalis prevalence in children ≤10 years old in Nigeria. Table 2. Asymptomatic vs Symptomatic infected children ≤10 years old in Nigeria. Table 3. Scoring criteria for semi-quantitative histopathological analysis.
Source data
Source Data Figs. 1–6, Extended Data Figs. 1–7 and Supplementary Tables 1–3
Numeric source data.
Rights and permissions
About this article
Cite this article
Sardinha-Silva, A., Gazzinelli-Guimaraes, P.H., Ajakaye, O.G. et al. Giardia-induced Type 2 mucosal immunity attenuates intestinal inflammation caused by co-infection or colitis in mice. Nat Microbiol 10, 1886–1901 (2025). https://doi.org/10.1038/s41564-025-02051-2
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41564-025-02051-2
This article is cited by
-
Giardia helps the immune system pick its battles
Nature Immunology (2025)