Abstract
The mammalian gut harbours trillions of commensal bacteria that interact with their hosts through various bioactive molecules1,2. However, the mutualistic strategies that hosts evolve to benefit from these symbiotic relationships are largely unexplored. Here we report that mouse enterocytes secrete apolipoprotein L9a and b (APOL9a/b) in the presence of microbiota. By integrating flow cytometry sorting of APOL9-binding bacterial taxa with 16S ribosomal RNA gene sequencing (APOL9-seq), we identify that APOL9a/b, as well as their human equivalent APOL2, coat gut bacteria belonging to the order of Bacteroidales with a high degree of specificity through commensal ceramide-1-phosphate (Cer1P) lipids. Genetic abolition of ceramide-1-phosphate synthesis pathways in gut-dominant symbiote Bacteroides thetaiotaomicron significantly decreases the binding of APOL9a/b to the bacterium. Instead of lysing the bacterial cells, coating of APOL9a/b induces the production of outer membrane vesicles (OMVs) from the target bacteria. Subsequently, the Bacteroides-elicited outer membrane vesicles enhance the host’s interferon-γ signalling to promote major histocompatibility complex class II expression in the intestinal epithelial cells. In mice, the loss of Apol9a/b compromises the gut major histocompatibility complex class II-instructed immune barrier function, leading to early mortality from infection by intestinal pathogens. Our data show how a host-elicited factor benefits gut immunological homeostasis by selectively targeting commensal ceramide molecules.
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 51 print issues and online access
$199.00 per year
only $3.90 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 16S rRNA-seq, bulk RNA-seq and scRNA-seq data generated in this study are available in the NCBI database under BioProject IDs PRJNA1107192, PRJNA1107183, PRJNA1107279, PRJNA1108888 and PRJNA1198673. The public datasets used in this study are available in the NCBI database under the GEO accession numbers GSE92332 and GSE125970 (refs. 27,28). All other data supporting the findings of this study are included in this manuscript. Source data are provided with this paper.
Code availability
No custom code was created for this paper.
References
Donia, M. S. & Fischbach, M. A. Small molecules from the human microbiota. Science 349, 1254766 (2015).
Krautkramer, K. A., Fan, J. & Backhed, F. Gut microbial metabolites as multi-kingdom intermediates. Nat. Rev. Microbiol. 19, 77–94 (2021).
Arpaia, N. et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504, 451–455 (2013).
Smith, P. M. et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573 (2013).
Furusawa, Y. et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504, 446–450 (2013).
Hang, S. et al. Bile acid metabolites control TH17 and Treg cell differentiation. Nature 576, 143–148 (2019).
Song, X. et al. Microbial bile acid metabolites modulate gut RORγ+ regulatory T cell homeostasis. Nature 577, 410–415 (2020).
Campbell, C. et al. Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature 581, 475–479 (2020).
Cervantes-Barragan, L. et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 357, 806–810 (2017).
Song, X. et al. Gut microbial fatty acid isomerization modulates intraepithelial T cells. Nature 619, 837–843 (2023).
Mazmanian, S. K., Round, J. L. & Kasper, D. L. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453, 620–625 (2008).
Oh, S. F. et al. Host immunomodulatory lipids created by symbionts from dietary amino acids. Nature 600, 302–307 (2021).
Peterson, L. W. & Artis, D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat. Rev. Immunol. 14, 141–153 (2014).
Mukherjee, S. & Hooper, L. V. Antimicrobial defense of the intestine. Immunity 42, 28–39 (2015).
Wu, M. et al. Gut complement induced by the microbiota combats pathogens and spares commensals. Cell https://doi.org/10.1016/j.cell.2023.12.036 (2024).
Cash, H. L., Whitham, C. V., Behrendt, C. L. & Hooper, L. V. Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science 313, 1126–1130 (2006).
Vaishnava, S. et al. The antibacterial lectin RegIIIγ promotes the spatial segregation of microbiota and host in the intestine. Science 334, 255–258 (2011).
Bergstrom, J. H. et al. Gram-positive bacteria are held at a distance in the colon mucus by the lectin-like protein ZG16. Proc. Natl Acad. Sci. USA 113, 13833–13838 (2016).
McPherson, R. L. et al. Lectin-Seq: a method to profile lectin-microbe interactions in native communities. Sci. Adv. 9, eadd8766 (2023).
Palm, N. W. et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 158, 1000–1010 (2014).
Kau, A. L. et al. Functional characterization of IgA-targeted bacterial taxa from undernourished Malawian children that produce diet-dependent enteropathy. Sci. Transl. Med. 7, 276ra224 (2015).
Shapiro, J. M. et al. Immunoglobulin A targets a unique subset of the microbiota in inflammatory bowel disease. Cell Host Microbe 29, 83–93 e83 (2021).
Hooper, L. V. et al. Molecular analysis of commensal host-microbial relationships in the intestine. Science 291, 881–884 (2001).
Geva-Zatorsky, N. et al. Mining the human gut microbiota for immunomodulatory organisms. Cell 168, 928–943 e911 (2017).
Sun, L. et al. Type I interferons link viral infection to enhanced epithelial turnover and repair. Cell Host Microbe 17, 85–97 (2015).
Kreit, M., Vertommen, D., Gillet, L. & Michiels, T. The interferon-inducible mouse apolipoprotein L9 and prohibitins cooperate to restrict Theiler’s virus replication. PLoS ONE 10, e0133190 (2015).
Haber, A. L. et al. A single-cell survey of the small intestinal epithelium. Nature 551, 333–339 (2017).
Wang, Y. et al. Single-cell transcriptome analysis reveals differential nutrient absorption functions in human intestine. J. Exp. Med. https://doi.org/10.1084/jem.20191130 (2020).
Wexler, A. G. & Goodman, A. L. An insider’s perspective: Bacteroides as a window into the microbiome. Nat. Microbiol. 2, 17026 (2017).
Uzureau, S. et al. APOL1 C-terminal variants may trigger kidney disease through interference with APOL3 control of actomyosin. Cell Rep. 30, 3821–3836 e3813 (2020).
Brown, E. M. et al. Bacteroides-derived sphingolipids are critical for maintaining intestinal homeostasis and symbiosis. Cell Host Microbe 25, 668–680 e667 (2019).
Johnson, E. L. et al. Sphingolipids produced by gut bacteria enter host metabolic pathways impacting ceramide levels. Nat. Commun. 11, 2471 (2020).
Stankeviciute, G. et al. Convergent evolution of bacterial ceramide synthesis. Nat. Chem. Biol. 18, 305–312 (2022).
Dhakephalkar, T., Stukey, G. J., Guan, Z., Carman, G. M. & Klein, E. A. Characterization of an evolutionarily distinct bacterial ceramide kinase from Caulobacter crescentus. J. Biol. Chem. 299, 104894 (2023).
Heaver, S. L. et al. Characterization of inositol lipid metabolism in gut-associated Bacteroidetes. Nat. Microbiol. 7, 986–1000 (2022).
Lloyd-Price, J. et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569, 655–662 (2019).
Perez-Morga, D. et al. Apolipoprotein L-I promotes trypanosome lysis by forming pores in lysosomal membranes. Science 309, 469–472 (2005).
Thomson, R. & Finkelstein, A. Human trypanolytic factor APOL1 forms pH-gated cation-selective channels in planar lipid bilayers: relevance to trypanosome lysis. Proc. Natl Acad. Sci. USA 112, 2894–2899 (2015).
Gaudet, R. G. et al. A human apolipoprotein L with detergent-like activity kills intracellular pathogens. Science https://doi.org/10.1126/science.abf8113 (2021).
Stefan, K. L., Kim, M. V., Iwasaki, A. & Kasper, D. L. Commensal microbiota modulation of natural resistance to virus infection. Cell 183, 1312–1324 e1310 (2020).
Yang, D. et al. Dysregulated lung commensal bacteria drive interleukin-17B production to promote pulmonary fibrosis through their outer membrane vesicles. Immunity 50, 692–706 e697 (2019).
Shen, Y. et al. Outer membrane vesicles of a human commensal mediate immune regulation and disease protection. Cell Host Microbe 12, 509–520 (2012).
Heuberger, C., Pott, J. & Maloy, K. J. Why do intestinal epithelial cells express MHC class II? Immunology 162, 357–367 (2021).
Moon, S. et al. Niche-specific MHC II and PD-L1 regulate CD4+CD8αα+ intraepithelial lymphocyte differentiation. J. Exp. Med. https://doi.org/10.1084/jem.20201665 (2021).
Lockhart, A., Mucida, D. & Bilate, A. M. Intraepithelial lymphocytes of the intestine. Annu. Rev. Immunol. https://doi.org/10.1146/annurev-immunol-090222-100246 (2024).
Cullen, T. W. et al. Antimicrobial peptide resistance mediates resilience of prominent gut commensals during inflammation. Science 347, 170–175 (2015).
Garcia-Bayona, L. & Comstock, L. E. Streamlined genetic manipulation of diverse Bacteroides and Parabacteroides isolates from the human gut microbiota. mBio https://doi.org/10.1128/mBio.01762-19 (2019).
Sugiura, M. et al. Ceramide kinase, a novel lipid kinase. Molecular cloning and functional characterization. J. Biol. Chem. 277, 23294–23300 (2002).
Clarke, E. L. et al. Sunbeam: an extensible pipeline for analyzing metagenomic sequencing experiments. Microbiome 7, 46 (2019).
Kang, D. D. et al. MetaBAT 2: an adaptive binning algorithm for robust and efficient genome reconstruction from metagenome assemblies. PeerJ 7, e7359 (2019).
Parks, D. H., Imelfort, M., Skennerton, C. T., Hugenholtz, P. & Tyson, G. W. CheckM: assessing the quality of microbial genomes recovered from isolates, single cells, and metagenomes. Genome Res. 25, 1043–1055 (2015).
Lam, S. M. et al. A multi-omics investigation of the composition and function of extracellular vesicles along the temporal trajectory of COVID-19. Nat. Metab. 3, 909–922 (2021).
Yuan, X. et al. Identification of an endogenous ligand bound to a native orphan nuclear receptor. PLoS ONE 4, e5609 (2009).
Han, X., Yang, Y., Zhao, F., Zhang, T. & Yu, X. An improved protein lipid overlay assay for studying lipid-protein interactions. Plant Methods 16, 33 (2020).
Couter, C. J. & Surana, N. K. Isolation and flow cytometric characterization of murine small intestinal lymphocytes. J. Vis. Exp. https://doi.org/10.3791/54114 (2016).
Ivanov, I. I. et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139, 485–498 (2009).
Hsu, C. C., Okumura, R. & Takeda, K. Human LYPD8 protein inhibits motility of flagellated bacteria. Inflamm. Regen. 37, 23 (2017).
Wang, Y. C. et al. Intestinal cell type-specific communication networks underlie homeostasis and response to Western diet. J. Exp. Med. https://doi.org/10.1084/jem.20221437 (2023).
Gilmore, W. J. et al. Bacteroides fragilis outer membrane vesicles preferentially activate innate immune receptors compared to their parent bacteria. Front. Immunol. 13, 970725 (2022).
Bousbaine, D. et al. A conserved Bacteroidetes antigen induces anti-inflammatory intestinal T lymphocytes. Science 377, 660–666 (2022).
Jiang, L. et al. Salmonella typhimurium reprograms macrophage metabolism via T3SS effector SopE2 to promote intracellular replication and virulence. Nat. Commun. 12, 879 (2021).
Athman, R. et al. Shigella flexneri infection is dependent on villin in the mouse intestine and in primary cultures of intestinal epithelial cells. Cell. Microbiol. 7, 1109–1116 (2005).
Acknowledgements
The work received support from the National Key R&D Program of China grant no. 2020YFA0509100, and the NSF of China grant nos. 81830018 and 32030039 (to Y.Q.); the National Key R&D Program of China grant nos. 2022YFA0807300 and 2023YFA1800200, the Strategic Priority Research Program of the Chinese Academy of Sciences (grant no. XDB0990000), the International Partnership Program of the Chinese Academy of Sciences for Future Network (grant no. 318GJHZ2022030FN), the NSF of China grant nos. 32270945 and 92474105, and STCSM grant nos. 22ZR1468700 and 22140902400 (to X.S.); the NSF of China grant nos. 32200036 and 82341116 (to G.L.); and the Department of Defense grant no. HT9425-23-0226 (to D.L.K.). We thank L. Qiu, X. Miao, Y. Bu, Y. Zhai and J. Wu (Institutional Center for Shared Technologies and Facilities of SINH, CAS) for ultracentrifugation and FACS support. We thank Y. Qin, Z. Li and S. Yan (Institutional Center for Shared Technologies and Facilities of SINH, CAS) for technical assistance in microscopy. We appreciate the technical support for lipidomics or lipid structure analysis from LipidALL Technologies (Changzhou, China), Z. Li (Institutional Center for Shared Technologies and Facilities of SINH, CAS) and Z. Yang (Chemical Biology Core Facility of SIBCB, CAS). We also appreciate the technical support for proteomics analysis from PANOMIX Biomedical Tech Co. (Suzhou, China). We thank the staff of the Bioinformatics Core Facility of SIBCB for the scRNA-seq analysis, Y. Kong and L. Pan (Electron Microscopy Core Facilities of ION, CAS) for assistance with EM sample preparation and EM image analysis, and H. Jiang (Core Facility and Technical Service Center for SLSB of SJTU) for technical assistance with FACS analysis of intestinal bacteria. We also thank S. Lin for helping with STm infection experiments (Shanghai Institute of Immunity and Infection, CAS).
Author information
Authors and Affiliations
Contributions
Y.Q. and X.S. supervised the research. X.S., T.Y., D.L.K. and Y.Q. wrote the manuscript. T.Y. performed proteomics and lipidomics analysis, immunoblotting, immunofluorescence, H&E staining, ceramide kinase assays and bacterial killing assays. T.Y., X.H., F.C. and F.Y. performed flow cytometric sorting of APOL9-binding faecal bacterial taxa and flow cytometric analysis of APOL binding with human gut microbiota. X.H. performed TEM analysis of OMVs and in vitro DC–IEL–IEC co-culture experiments. T.Y. and X.H. performed all sequencing studies and STm infection experiments. F.C. cultured commensal bacterial strains and performed genetic manipulation of B. thet. F.Y., M.Z. and G.K. purified recombinant APOL proteins and B. thet ceramide kinase. B.N. assisted with immunofluorescence and H&E staining. T.Y., H.Z. and F.C. contributed to the flow cytometry of IELs and LPLs. G.L. and H.G. performed the phylogenetic analysis of the Cer1P biosynthesis genes. K.J., Y.L. and J.S. assisted in the analysis of scRNA-seq data. X.L. performed pathological scoring. T.Y. and M.Z. created all the graphical illustrations for the figures and Extended data figures using Adobe Illustrator CC 2018. D.L.K. and G.H. provided technical assistance and suggestions.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature thanks Kenya Honda and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data figures and tables
Extended Data Fig. 1 Microbiota-dependent regulation of ileal APOL9a/b.
a, Q-PCR analysis of Apol9a and Apol9b tissue expression in various organs of WT C57BL/6 J mice (n = 3). b, Q-PCR analysis of Apol9a and Apol9b expression in ileal ECs, IELs, and LPLs (n = 4). c and d, Q-PCR (c) or immunoblot (d) analysis of Apol9 expression in ileal ECs from GF, EX-GF (obtained by co-housing of 4-week-old GF mice with age- and gender-matched SPF mice), or SPF C57BL/6 J mice (n = 3). e, Immunoblot analysis of Apol9 expression in ileal ECs from water or antibiotics (ABX)-treated SPF C57BL/6 J mice for 4 weeks (n = 3). f and g, Q-PCR (f, n = 6) or immunoblot (g, n = 3) analysis of Apol9 expression in ileal ECs from Ifnar1+/+ or Ifnar1–/– mice. h and i, Q-PCR (h, n = 6) or immunoblot (i, n = 3) analysis of Apol9 expression in ileal ECs from Ifngr1+/+ or Ifngr1–/– mice. Data are representative of two (c,d,e) or three independent experiments (a,b,f,g,h,i). n represents biologically independent animals (a-i). Data are presented as mean ± SEM. Statistical analysis was performed using a two-tailed Student’s t-test (f,h) or one-way ANOVA followed by Bonferroni post hoc test (c). For gel source data, see Supplementary Fig. 2.
Extended Data Fig. 2 Generation of Apol9a/b–/– mice by CRISPR/Cas9 genomic targeting.
a, Schematic diagram of the Apol9a and Apol9b locus with the sgRNA target sequences depicted for generating the Apol9a and Apol9b double-deficient (Apol9a/b–/–) mice. b, Genotyping of Apol9a and Apol9b exon locus by DNA sequencing after CRISPR-CAS9 targeting. c, Schematic diagram showing the nonsense mutations in Apol9a and Apol9b locus that terminate the protein translation. d, Validation of knock-out efficiency of Apol9a and Apol9b in ileal ECs from Apol9a/b+/+ and Apol9a/b–/– mice by immunoblot analysis (n = 3). e, t-SNE plots showing Apol9a and Apol9b expression (left) in the indicated epithelial cell types (right) from a previously published single-cell RNA sequencing study on mouse small intestine (GSE92332). The heatmap represents the expression level. f, Immunostaining of ileal sections of WT SPF, WT GF, or Apol9a/b–/– mice. Blue, DAPI; green, UEA1 (mucus); red, APOL9. Scale bar, 50 μm. Data are representative of three independent experiments (d,f). n represents biologically independent animals (d). For gel source data, see Supplementary Fig. 2.
Extended Data Fig. 3 Purification of recombinant mouse APOL9a/b and human APOL1-APOL4 proteins.
a, Coomassie blue staining or immunoblot analysis of purified recombinant his-tagged APOL9a protein. b, Coomassie blue staining or immunoblot analysis of purified recombinant his-tagged APOL9b protein. c, Phylogenetic tree of mouse APOL9a and APOL9b with human APOL1-APOL6 proteins. d, Predicted domain structure of human APOL1, APOL2, APOL3, and APOL4. e, Coomassie blue staining or immunoblot analysis of purified recombinant flag-tagged human APOL1, APOL2, APOL3, and APOL4 proteins. f, t-SNE plots showing human APOL1, APOL2, APOL3, and APOL4 expression in the indicated epithelial cell types from a previously published single-cell RNA sequencing study on human small intestine (GSE125970). The Heatmap represents the expression level. Data are representative of two independent experiments (a,b,e). For gel source data, see Supplementary Fig. 2.
Extended Data Fig. 4 Phylogenetic analysis of Cer1P biosynthesis genes in human gut microbiota.
a and b, The distribution of key gene orthologues involved in Cer1P biosynthesis, including serine palmitoyltransferase (BT_0870), keto-reductase (BT_3075), ceramide synthase (BT_3032), and ceramide kinase (BT_0871), across fifty-two human commensal bacterial strains (a) used in this study or gut microbiota species (b) from the NIH Human Microbiome Project (HMP) database. c, Coomassie blue staining of purified recombinant his-tagged WT or kinase-dead B. thetaiotaomicron ceramide kinase (BT_0871) protein. Data are representative of two independent experiments (c). For gel source data, see Supplementary Fig. 2.
Extended Data Fig. 5 APOL9a and APOL2 induce bacterial membrane defense and biogenesis responses in B. thetaiotaomicron.
a, Different concentrations of rmAPOL9a or rmAPOL9b were added to ~104 CFU of log phase B. thetaiotaomicron for two hours, and surviving bacteria were enumerated by propidium iodide (PI) intake. BSA was used as a negative control (n = 3). b and c, rmAPOL9a (5 μg/mL) or rmAPOL9b (5 μg/mL) was added to log phase B. thetaiotaomicron under different pH conditions (c) or NaCl concentrations (d), and surviving bacteria were quantified by propidium iodide (PI) intake (n = 3). d, B. fragilis, B. vulgatus, P. distasonis, and P. goldsteinii were individually incubated with BSA, rmAPOL9a, or rhAPOL2 for three hours under anaerobic conditions, followed by transmission electron microscopy (TEM) observation. Representative images of a single bacteria producing OMVs are shown. Scale bar, 500 nm. e, E. coli was incubated with BSA or rmAPOL9a, rmAPOL9b, or rhAPOL2 for three hours under anaerobic conditions, followed by TEM observation. Representative TEM images of a single E. coli are shown. Scale bar, 500 nm. f, B. fragilis, B. vulgatus, P. distasonis, and P. goldsteinii were individually incubated with BSA, rmAPOL9a, or rhAPOL2 for six hours under anaerobic conditions. The OMVs produced in the bacterial culture supernatant were collected by ultracentrifuge and counted under TEM. The representative TEM images or OMV numbers per view (n = 8) are shown. Scale bar, 200 nm. g and h, Volcano plot of differentially expressed genes when WT or BT_0871-deficient B. thetaiotaomicron were exposed to rmAPOL9a (g), or rhAPOL2 (h) for four hours under anaerobic conditions, compared to an untreated control condition (|log2[FC] | > 2; FDR < 0.05) (n = 3). i and j, Gene ontology analysis of up-regulated differentially expressed genes when WT B. thetaiotaomicron were exposed to rmAPOL9a (i), or rhAPOL2 (j) for four hours under anaerobic conditions, compared to an untreated control condition (log2[FC] > 2; FDR < 0.05) (n = 3). Data are representative of two independent experiments (a-i). n represents biologically independent samples (a-c,f,g-j). Data are presented as mean ± SEM. Statistical analysis was performed using one-way ANOVA followed by Bonferroni post hoc test (a-c,f) or one-way ANOVA followed by False Discovery Rate (FDR) procedure (g,h).
Extended Data Fig. 6 Characterization of the size and composition of steady-state or APOL9a-induced B. thetaiotaomicron OMVs.
a, Size distribution of steady-state or APOL9a-induced B. thetaiotaomicron OMVs was determined using ZetaView Nanoparticle Tracking Analysis (NTA, n = 3). b, Quantification of OMV-associated protein, DNA, RNA, peptidoglycan, or LPS from per 109 steady-state or APOL9a-induced B. thetaiotaomicron OMVs (n = 3). c, Relative abundance of the most represented lipid species as a percentage of the total lipid composition for each fraction from pooled steady-state or APOL9a-induced B. thetaiotaomicron OMVs (n = 2). d and e, ELISA analysis of supernatant IFNγ production (d) or Q-PCR analysis of MHC-II gene expression in ileal ECs (e) in a Transwell coculture of DC-IELs under steady-state or APOL9a-induced B. thetaiotaomicron-derived OMVs incubation (n = 3). Data are representative of two independent experiments (a-e). n represents biologically independent samples (a-e). Data are presented as mean ± SEM. Statistical analysis was performed using one-way ANOVA followed by Bonferroni post hoc test (d,e) or two-tailed Student’s t-test (b).
Extended Data Fig. 7 Single-cell RNA sequencing on ileal intraepithelial CD45+ immune cells.
a, Dot plots showing the top DEGs for the populations depicted in ileal intraepithelial CD45+ immune cells in Fig. 5a. Color saturation indicates the strength of average gene expression, whereas the dot size reflects the percentage of each cell cluster expressing the gene. b, Violin plots showing the selected maker gene expression among all ileal intraepithelial CD45+ immune cells in Fig. 5a. c, Gene Set Enrichment Analysis (GSEA) analysis of lymphocyte differentiation, T cell activation, immune response regulation, positive regulation of cytokine production, or positive regulation of epithelial proliferation pathways in the downregulated genes of ileal CD4+CD8αα+ IELs from Apol9a/b–/– mice. d and e, The intraepithelial lymphocytes (n = 5) and lamina propria lymphocytes (n = 4) were isolated from ilea of Apol9a/b+/+ and Apol9a/b–/– littermates and were analyzed by flow cytometry. The percentages of ileal intraepithelial lymphocyte subsets (d) or the percentages of ileal lamina propria lymphocyte subsets (e) from Apol9a/b+/+ or Apol9a/b–/– mice are shown. f, Intraepithelial CD4+CD8αα+ cells in different small intestine segments (duodenum, jejunum, ileum) from Apol9a/b+/+ or Apol9a/b–/– mice (n = 6) were analyzed by flow cytometry. The representative flow cytometry plots and CD4+CD8αα+ cell percentages are shown. Data are representative of two independent experiments (d,e,f). n represents biologically independent animals (d,e,f). Data are presented as mean ± SEM. Statistical analysis was performed using a two-tailed Student’s t-test (d,e,f).
Extended Data Fig. 8 The ileal immune phenotypes of Apol9a/b-deficient mice are associated with Bacteroidales.
a, Schematic diagram of FMT of ABX-treated Apol9a/b+/+ or Apol9a/b–/– mice with ileal microbiota from a WT C57BL/6 strain largely devoid of gut Bacteroidales. After 4 weeks, indicated ileal tissues or cells were analyzed. b, 16S rRNA gene sequencing of the microbial composition of the ileal contents from a WT C57BL/6 strain largely devoid of gut Bacteroidales (n = 3). c-e, Q-PCR (c, ABX: n = 4, FMT: n = 6) and flow cytometry (d,e, ABX: n = 5, FMT: n = 6) analysis of representative MHC-II molecule expression in ileal ECs from mice as described in (a). f and g, Ileal intraepithelial CD4+CD8αα+ cells from mice as described in (a) were analyzed by flow cytometry. The representative flow cytometry plots (f) and CD4+CD8αα+ cell percentages (g, ABX: n = 5, FMT: n = 6) are shown. Data are representative from two independent experiments (a-g). n represents biologically independent samples (b) or biologically independent animals (c,e,g). Data are presented as mean ± SEM. Statistical analysis was performed using one-way ANOVA followed by Bonferroni post hoc test (c,e,g).
Extended Data Fig. 9 Ileal immunomodulatory functions of APOL9 require Bacteroides-APOL9 interaction in vivo.
a, Q-PCR analysis of the relative abundance of B. thetaiotaomicron in the feces from ABX-treated Apol9a/b+/+ or Apol9a/b–/– mice transferred with a gut Bacteroidales-minus ileal microbiota plus WT or BT_0871-deficient B. thetaiotaomicron strain (n = 8). b, ELISA analysis (n = 8) of ileal IFNγ production in the mice as described in (a). c-d, Q-PCR (c, n = 8), and flow cytometry (d, n = 8) analysis of representative MHC-II molecule expression in ileal ECs from mice as described in (a). e and f, Ileal intraepithelial CD4+CD8αα+ cells from mice as described in (a) were analyzed by flow cytometry. The representative flow cytometry plots (e) and CD4+CD8αα+ cell percentages (f, n = 8) are shown. Data are pooled from two independent experiments (a-f). n represents biologically independent animals (a-d,f). Data are presented as mean ± SEM. Statistical analysis was performed using one-way ANOVA followed by Bonferroni post hoc test (a-d,f).
Extended Data Fig. 10 APOL9a/b control mucosal infection in a microbiota context-dependent manner.
a and b, The representative flow cytometry plots (a) and binding efficiencies (b) of rmAPOL9a (5 μg/mL) or rmAPOL9b (5 μg/mL) bound with STm. Transferrin binding as a positive control (n = 3). c, Propidium iodide (PI) staining of STm after incubation with BSA (5 μg/mL), rmAPOL9a (5 μg/mL), or rmAPOL9b (5 μg/mL) for 3 h. EDTA (20 μM) plus Polymyxin B (25 μg/mL) treatment as a positive control (n = 3). d, The dynamics of CFU within the Apol9a/b+/+ or Apol9a/b–/– ileal ECs after STm in vitro infection (n = 3). e, STm quantification in the ileal lumen of Apol9a/b+/+ or Apol9a/b–/– mice after STm infection in the early or late time point (n = 6). f-h, FMT of ABX-treated Apol9a/b+/+ or Apol9a/b–/– mice with ileal microbiota from a WT C57BL/6 strain largely devoid of gut Bacteroidales, followed by STm infection. Survival curve (f, n = 8), ileal histology (day 5) (g), or bacteria quantitation (day 5) in the spleen and liver (h, n = 6) are shown. Scale bar, 10 μm. i and j, Apol9a/b+/+ or Apol9a/b–/– mice were infected intraperitoneally with STm. Survival curve (i, Apol9a/b+/+: n = 9; Apol9a/b–/–: n = 8) or bacteria quantitation (day 5) in the spleen and liver (j, n = 6) are shown. Data are representative of two independent experiments (a-j). n represents biologically independent samples (b-d) or biologically independent animals (e-j). Data are presented as mean ± SEM. Statistical analysis was performed using one-way ANOVA followed by Bonferroni post hoc test (b,c), two-tailed Student’s t-test (d,e,g,h,j), or log-rank (Mantel-Cox) test (f,i).
Supplementary information
Supplementary Fig. 1
Flow cytometry gating strategy.
Supplementary Fig. 2
Uncropped western blots.
Supplementary Tables
Supplementary Tables 1–9.
Source data
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Yang, T., Hu, X., Cao, F. et al. Targeting symbionts by apolipoprotein L proteins modulates gut immunity. Nature 643, 210–218 (2025). https://doi.org/10.1038/s41586-025-08990-4
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41586-025-08990-4
This article is cited by
-
APOL proteins tune gut immunity via commensal lipid recognition
Cell Research (2025)