Identifying species of symbiont bacteria from the human gut that, alone, can induce intestinal Th17 cells in mice

Tze Guan Tan, Esen Sefik, Naama Geva-Zatorsky, Lindsay Kua, Debdut Naskar, Fei Teng, Lesley Pasman, Adriana Ortiz-Lopez, Ray Jupp, Hsin-Jung Joyce Wu, Dennis L Kasper, Christophe Benoist, Diane Mathis, Tze Guan Tan, Esen Sefik, Naama Geva-Zatorsky, Lindsay Kua, Debdut Naskar, Fei Teng, Lesley Pasman, Adriana Ortiz-Lopez, Ray Jupp, Hsin-Jung Joyce Wu, Dennis L Kasper, Christophe Benoist, Diane Mathis

Abstract

Th17 cells accrue in the intestine in response to particular microbes. In rodents, segmented filamentous bacteria (SFB) induce intestinal Th17 cells, but analogously functioning microbes in humans remain undefined. Here, we identified human symbiont bacterial species, in particular Bifidobacterium adolescentis, that could, alone, induce Th17 cells in the murine intestine. Similar to SFB, B. adolescentis was closely associated with the gut epithelium and engendered cognate Th17 cells without attendant inflammation. However, B. adolescentis elicited a transcriptional program clearly distinct from that of SFB, suggesting an alternative mechanism of promoting Th17 cell accumulation. Inoculation of mice with B. adolescentis exacerbated autoimmune arthritis in the K/BxN mouse model. Several off-the-shelf probiotic preparations that include Bifidobacterium strains also drove intestinal Th17 cell accumulation.

Keywords: Th17 cells; intestine; microbiota; mucosal immunology; probiotic.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Fig. 1.
Fig. 1.
B. adolescentis (BA) induces a robust intestinal Th17 population. (A) Frequency of SI-LP Th17 cells in GF mice monocolonized with individual symbiont bacteria as described in Materials and Methods. White and gray symbols represent GF and SFB-monocolonized mice, respectively. Arrow indicates BA-monocolonized mice. (B) Inflammatory cytokine production by SI-LP CD4+ T cells in mice colonized as indicated. (Left) Representative flow cytometric dot plot. (Right) Summary data. (C) Frequencies of (Upper) Th17 and (Lower) Th1 cells in various tissues of mice colonized as indicated. (D) Frequencies of (Left) Th17 and (Right) Th1 cells in the SI-LP of SPF mice gavaged as described in Materials and Methods with the indicated microbes. SFB+ SPF mice were bred at Harvard Medical School and naturally colonized with SFB. (E) Induction of intestinal RORγt expressers. (Left) Representative flow cytometric dot plot of SI-LP CD4+ T cells; summary data for frequencies of (Center) RORγt+ Foxp3− cells and (Right) RORγt+ Helios− Treg cells. Numbers in B and E refer to the fractions of cells in the identical gates. (B–E) Mean ± SEM pooled from two to four independent experiments. BF, B. fragilis; Ce, cecum; CH, C. histolyticum; Co, colon; IEL, intraepithelial lymphocyte layer; ILN, inguinal lymph node; MLN, mesenteric lymph nodes; PP, Peyer’s patches; SI, small intestine. *P < 0.05 (Mann–Whitney u test); **P < 0.01 (Mann–Whitney u test); ***P < 0.001 (Mann–Whitney u test).
Fig. S1.
Fig. S1.
Effects of BA monocolonization on other intestinal immunocyte populations. (A) No correlation between induction of intestinal Th17 and RORγt+ Treg cells after monocolonization with the panel of bacteria detailed in Fig. 1A. Lines represent linear regressions performed on the two immunocyte populations in the respective tissues. Pearson correlations are found to be not significant. (B) Data in A plotted as SI-LP Th17 vs. colonic RORγt+ Treg frequencies. Linear regression lines and correlations were calculated as in A (not significant). (C) Frequencies of IL-17A+ cells within the RORγt+ CD4+ T-cell population in various intestinal tissues in BA-monocolonized mice. (D) Frequencies of intestinal Foxp3+ CD4+ T cells in mice colonized as indicated. (E–G) Frequencies of (E) IL-17A–producing γδ T cells, (F) IL-22–producing Thy1+ RORγt+ type 3 innate lymphoid cells (ILC3s), and (G) IL-17A–producing ILC3s in various tissues of mice colonized as indicated. (H) Frequencies of GC B cells in the Peyer’s patches (PPs) of mice colonized as indicated. (I) Frequencies of IgA+ B220− plasma cells in the SI-LP of mice colonized as indicated. SPF mice from Jax were SFB− or colonized with SFB. Each symbol represents one mouse, and data were pooled from at least two independent experiments. Mean ± SEM. Ce, cecum; CH, Clostridium histolyticum; Co, colon; Duo, duodenum; Il, ileum; ILN, inguinal lymph node; Jej, jejunum; MLN, mesenteric lymph nodes; SI, small intestine. *P < 0.05 (Mann–Whitney u test); **P < 0.01 (Mann–Whitney u test); ***P < 0.001 (Mann–Whitney u test).
Fig. 2.
Fig. 2.
B. adolescentis (BA) does not provoke intestinal inflammation. (A–D) Myeloid cells. Frequencies of the indicated myeloid cell populations in the intestines of mice colonized as indicated. Each symbol represents one mouse. Mean ± SEM. Data for GF and BA are pooled from at least two independent experiments. P value was not significant for all comparisons (Kruskal–Wallis test and Dunn’s multiple comparisons test). (E and F) Histopathology. H&E staining of representative sections of (E) the small intestine (SI) and (F) the colon. (Scale bar: 50 μm.) (G) Th17 cell phenotype. Fold change (FC)/FC plots comparing transcripts induced by BA vs. SFB in SI-LP CD4+ T cells. Red indicates transcripts up-regulated in (Upper) pathogenic Th17 cells or (Lower) canonical Th17 cells. Example genes induced for each Th17 phenotype are indicated (n = 3–4 per group). BF, B. fragilis; CH, C. histolyticum.
Fig. S2.
Fig. S2.
BA does not provoke intestinal inflammation. (A) Numbers of CD45+ cells and (B) frequencies of CD11b+F4/80+CD103− macrophages in the intestines of mice colonized as indicated. Each symbol represents one mouse. Data for GF and BA pooled from at least two independent experiments. *P < 0.05 (Kruskal–Wallis test and Dunn’s multiple comparisons test). (C) Pathology scores of ileum and colon sections from GF and BA-monocolonized mice. Scoring method is detailed in SI Materials and Methods. Each symbol represents one mouse, and data were pooled from two independent experiments. BF, Bacteroides fragilis; CH, Clostridium histolyticum; SI, small intestine.
Fig. 3.
Fig. 3.
B. adolescentis (BA)-driven intestinal Th17 responses are cognate. (A and B) BA-specific Th17 cells. Frequencies of SI-LP CD4+ T cells producing (A) IL-17A or (B) IFN-γ on overnight stimulation by splenic dendritic cells incubated with media alone or lysates prepared from the indicated bacterial species. +Anti–MHC-II (the blocking antibody M5/114.15.2) was added. (C) Frequency of SI-LP CD4+ T cells producing IL-17A on stimulation with media alone or BA lysate when T cells were isolated from mice colonized as indicated. (D) The degree of TCRVβ14 enrichment in SI-LP CD4+ T cells from SPF mice colonized as indicated, calculated as (percent of Vβ14+ cells in Th17 fraction)/(percent of TCRVβ14+ cells in non-Th17 fraction). Each symbol represents one mouse. Data are pooled from two to four independent experiments. Mean ± SEM. BF, B. fragilis; P + I, PMA and ionomycin. *P < 0.05 (Kruskal–Wallis test and Dunn’s multiple comparisons test); **P < 0.01 (Kruskal–Wallis test and Dunn’s multiple comparisons test); ***P < 0.001 (Kruskal–Wallis test and Dunn’s multiple comparisons test).
Fig. 4.
Fig. 4.
B. adolescentis (BA) colonizes the entire length of the intestines, closely associating with the ileal epithelium. (A) Titers of bacteria associated with various segments of the intestinal mucosa or shed into the corresponding lumen or the stool (St). Normalized to tissue or St weight or the volume of luminal wash. Each symbol represents one mouse. Data are pooled from three to four independent experiments. Mean ± SEM. (B) Frequencies of Th17 cells along the length of the intestinal LP in mice colonized as indicated. Each line represents frequencies from one mouse. Data are pooled from three independent experiments. (C) FISH quantification. Normalized bacterial fluorescence vs. distance from the epithelial surface of the terminal ileum (Il) from mice monocolonized as indicated. Data are plotted as the average fluorescence from six to eight total images from two to three fields of view per section from two to four mice per microbe. (D) Representative SEM photograph of the ileal surface in mice colonized as indicated. Arrows indicate sites of bacterial association with the intestinal epithelium. Ce, cecum; CH, C. histolyticum; Co, colon; Duo, duodenum; Jej, jejunum.
Fig. 5.
Fig. 5.
B. adolescentis (BA) and SFB both induce B-cell transcripts, but otherwise, they trigger distinct transcriptional programs. (A and B) Whole-tissue transcriptomes. Fold change (FC)/FC plots comparing ileum tissue transcripts induced by (A) BA vs. SFB (Th17 inducing) or (B) BA vs. C. histolyticum (CH; Th17 noninducing). In A, blue indicates genes up-regulated primarily by SFB, black indicates genes up-regulated primarily by BA, and red indicates genes up-regulated by both bacteria. Certain transcripts previously associated with SFB-mediated induction of Th17 cells or encoding Igs are indicated. In B, the genes highlighted in A are again highlighted in the same colors. (C) Statistics for A and B. FC distribution of (Upper) SFB-induced Ig or (Lower) non-Ig RNAs in the ileal transcriptomes of mice colonized as indicated. Only transcripts increased by SFB by at least ≥1.6-fold with a P value of <0.05 were plotted. ***P < 0.001 (Kolmogorov–Smirnov test). (D) Pathways (from the two databases as indicated) enriched in transcripts specifically up-regulated in the ileum by BA relative to GF mice were determined using Enrichr. P values are indicated to the right. (E and F) S-IEC transcriptomes. As per A and B, except that transcripts from isolated S-IECs were examined. In E, orange indicates genes up-regulated primarily by SFB, cyan indicates genes up-regulated primarily by BA, and purple indicates genes up-regulated by both bacteria. In F, the genes highlighted in E are again highlighted in the same colors. (G) Statistics for E and F. Calculated as per C. n = 3 (AD) or 2–4 (E and F) per group. ***P < 0.001.
Fig. 6.
Fig. 6.
B. adolescentis (BA) exacerbates K/BxN arthritis. K/BxN mice were pretreated with antibiotics from 22 to 32 d of age and subsequently gavaged with PBS, C. histolyticum (CH), or BA over 12–13 d. Arrows indicate gavage time points. (A) Ankle-thickening values over the course of bacterial gavage. (B, Left) Representative flow cytometric plot of IL-17A production from SI-LP CD4+ T cells 11–13 d after the initial bacterial gavage. Summary data of (B, Center) frequencies and (B, Right) numbers of SI-LP Th17 cells at the same time point. (C) Antiglucose-6-phosphate isomerase titers 11–13 d after initial bacterial gavage. Each symbol represents one mouse [n = 3 (PBS), 8 (CH), 11 (BA), and 2–5 (SFB)]. Data for CH and BA are pooled from two to three independent experiments. Mean ± SEM. GPI, glucose-6-phosphate isomerase. *P < 0.05 (Kruskal–Wallis test with Dunn’s multiple comparisons test); **P < 0.01 (Kruskal–Wallis test with Dunn’s multiple comparisons test); ***P < 0.001; (Kruskal–Wallis test with Dunn’s multiple comparisons test).
Fig. 7.
Fig. 7.
Some probiotic formulations containing bifidobacterial species also elicit Th17 populations in the SI-LP. (A) Frequencies of SI-LP (Left) Th17 or (Right) Th1 cells in GF mice colonized with the indicated probiotic mixes. (B) Th17 cell phenotype. Fold change (FC)/FC plots comparing transcripts induced by B. adolescentis (BA) vs. the probiotic mix, Nexabiotic, in SI-LP CD4+ T cells. Symbols in red and labels are as per Fig. 2G (n = 2–3 per group). (C) Frequencies of SI-LP (Left) Th17 and (Right) Th1 cells in SPF (SFB−) mice gavaged with PBS or one of three other probiotic preparations in A. Full names and corresponding abbreviations of probiotics can be found in SI Materials and Methods. Each symbol represents one mouse. Data are pooled from two to three independent experiments. Mean ± SEM. *P < 0.05 (Mann–Whitney u test); **P < 0.01 (Mann–Whitney u test).
Fig. S3.
Fig. S3.
BA and related species are enriched in the microbiotas of IBD patients. Comparison of the microbiotas of 85 healthy vs. 39 IBD subjects in the MetaHIT database using LEfSe as outlined in SI Materials and Methods. Microbial clades with the largest effect sizes (>3.5) with a false discovery rate q < 0.05 (Kruskal–Wallis test with Benjamini–Hochberg correction) are shown. (Left) Negative and (Right) positive values correspond to healthy controls (HCs) and IBD patients, respectively. BA and other bifidobacterial species are represented as white bars.

References

    1. Kamada N, Chen GY, Inohara N, Núñez G. Control of pathogens and pathobionts by the gut microbiota. Nat Immunol. 2013;14(7):685–690.
    1. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome and the immune system. Nature. 2011;474(7351):327–336.
    1. Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature. 2016;535(7610):75–84.
    1. Smith PM, Garrett WS. The gut microbiota and mucosal T cells. Front Microbiol. 2011;2:111.
    1. Atarashi K, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331(6015):337–341.
    1. Atarashi K, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500(7461):232–236.
    1. Faith JJ, Ahern PP, Ridaura VK, Cheng J, Gordon JI. Identifying gut microbe-host phenotype relationships using combinatorial communities in gnotobiotic mice. Sci Transl Med. 2014;6(220):220ra11.
    1. Sefik E, et al. MUCOSAL IMMUNOLOGY. Individual intestinal symbionts induce a distinct population of RORγ⁺ regulatory T cells. Science. 2015;349(6251):993–997.
    1. Gaboriau-Routhiau V, et al. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity. 2009;31(4):677–689.
    1. Ivanov II, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(3):485–498.
    1. Atarashi K, et al. Th17 cell induction by adhesion of microbes to intestinal epithelial cells. Cell. 2015;163(2):367–380.
    1. Ivanov II, et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe. 2008;4(4):337–349.
    1. Kleinschek MA, et al. Circulating and gut-resident human Th17 cells express CD161 and promote intestinal inflammation. J Exp Med. 2009;206(3):525–534.
    1. Pène J, et al. Chronically inflamed human tissues are infiltrated by highly differentiated Th17 lymphocytes. J Immunol. 2008;180(11):7423–7430.
    1. Kumar P, et al. Intestinal interleukin-17 receptor signaling mediates reciprocal control of the gut microbiota and autoimmune inflammation. Immunity. 2016;44(3):659–671.
    1. Maxwell JR, et al. Differential roles for interleukin-23 and interleukin-17 in intestinal immunoregulation. Immunity. 2015;43(4):739–750.
    1. Lee JS, et al. Interleukin-23-independent IL-17 production regulates intestinal epithelial permeability. Immunity. 2015;43(4):727–738.
    1. Hirota K, Ahlfors H, Duarte JH, Stockinger B. Regulation and function of innate and adaptive interleukin-17-producing cells. EMBO Rep. 2012;13(2):113–120.
    1. Gaffen SL, Jain R, Garg AV, Cua DJ. The IL-23-IL-17 immune axis: From mechanisms to therapeutic testing. Nat Rev Immunol. 2014;14(9):585–600.
    1. Kanai T, Mikami Y, Sujino T, Hisamatsu T, Hibi T. RORγt-dependent IL-17A-producing cells in the pathogenesis of intestinal inflammation. Mucosal Immunol. 2012;5(3):240–247.
    1. Scher JU, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. eLife. 2013;2:e01202.
    1. Gevers D, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014;15(3):382–392.
    1. Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4615–4622.
    1. Wu HJ, et al. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity. 2010;32(6):815–827.
    1. Ohnmacht C, et al. MUCOSAL IMMUNOLOGY. The microbiota regulates type 2 immunity through RORγt⁺ T cells. Science. 2015;349(6251):989–993.
    1. Sano T, et al. An IL-23R/IL-22 circuit regulates epithelial serum amyloid A to promote local effector Th17 responses. Cell. 2015;163(2):381–393.
    1. Cua DJ, Tato CM. Innate IL-17-producing cells: The sentinels of the immune system. Nat Rev Immunol. 2010;10(7):479–489.
    1. Bunker JJ, et al. Innate and adaptive humoral responses coat distinct commensal bacteria with immunoglobulin A. Immunity. 2015;43(3):541–553.
    1. Lécuyer E, et al. Segmented filamentous bacterium uses secondary and tertiary lymphoid tissues to induce gut IgA and specific T helper 17 cell responses. Immunity. 2014;40(4):608–620.
    1. Hirota K, et al. Plasticity of Th17 cells in Peyer’s patches is responsible for the induction of T cell-dependent IgA responses. Nat Immunol. 2013;14(4):372–379.
    1. Zigmond E, et al. Ly6C hi monocytes in the inflamed colon give rise to proinflammatory effector cells and migratory antigen-presenting cells. Immunity. 2012;37(6):1076–1090.
    1. Lee Y, et al. Induction and molecular signature of pathogenic TH17 cells. Nat Immunol. 2012;13(10):991–999.
    1. Nurieva RI, et al. Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages. Immunity. 2008;29(1):138–149.
    1. Goto Y, et al. Segmented filamentous bacteria antigens presented by intestinal dendritic cells drive mucosal Th17 cell differentiation. Immunity. 2014;40(4):594–607.
    1. Yang Y, et al. Focused specificity of intestinal TH17 cells towards commensal bacterial antigens. Nature. 2014;510(7503):152–156.
    1. Klaasen HL, Koopman JP, Poelma FG, Beynen AC. Intestinal, segmented, filamentous bacteria. FEMS Microbiol Rev. 1992;8(3-4):165–180.
    1. Farkas AM, et al. Induction of Th17 cells by segmented filamentous bacteria in the murine intestine. J Immunol Methods. 2015;421:104–111.
    1. Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nat Rev Immunol. 2014;14(10):667–685.
    1. Talham GL, Jiang HQ, Bos NA, Cebra JJ. Segmented filamentous bacteria are potent stimuli of a physiologically normal state of the murine gut mucosal immune system. Infect Immun. 1999;67(4):1992–2000.
    1. Umesaki Y, Okada Y, Matsumoto S, Imaoka A, Setoyama H. Segmented filamentous bacteria are indigenous intestinal bacteria that activate intraepithelial lymphocytes and induce MHC class II molecules and fucosyl asialo GM1 glycolipids on the small intestinal epithelial cells in the ex-germ-free mouse. Microbiol Immunol. 1995;39(8):555–562.
    1. Teng F, et al. Gut microbiota drive autoimmune arthritis by promoting differentiation and migration of Peyer’s Patch T follicular helper cells. Immunity. 2016;44(4):875–888.
    1. Chung H, et al. Gut immune maturation depends on colonization with a host-specific microbiota. Cell. 2012;149(7):1578–1593.
    1. Klaasen HL, et al. Intestinal, segmented, filamentous bacteria in a wide range of vertebrate species. Lab Anim. 1993;27(2):141–150.
    1. Yin Y, et al. Comparative analysis of the distribution of segmented filamentous bacteria in humans, mice and chickens. ISME J. 2013;7(3):615–621.
    1. Caselli M, Tosini D, Gafà R, Gasbarrini A, Lanza G. Segmented filamentous bacteria-like organisms in histological slides of ileo-cecal valves in patients with ulcerative colitis. Am J Gastroenterol. 2013;108(5):860–861.
    1. Rodriguez-Palacios A, et al. Stereomicroscopic 3D-pattern profiling of murine and human intestinal inflammation reveals unique structural phenotypes. Nat Commun. 2015;6:7577.
    1. Yatsunenko T, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–227.
    1. Bäckhed F, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. 2015;17(5):690–703.
    1. Bottacini F, Ventura M, van Sinderen D, O’Connell Motherway M. Diversity, ecology and intestinal function of bifidobacteria. Microb Cell Fact. 2014;13(Suppl 1):S4.
    1. Qin J, et al. MetaHIT Consortium A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65.
    1. Shapiro H, Thaiss CA, Levy M, Elinav E. The cross talk between microbiota and the immune system: Metabolites take center stage. Curr Opin Immunol. 2014;30:54–62.
    1. Cerovic V, Bain CC, Mowat AM, Milling SW. Intestinal macrophages and dendritic cells: What’s the difference? Trends Immunol. 2014;35(6):270–277.
    1. Raffatellu M, et al. Simian immunodeficiency virus-induced mucosal interleukin-17 deficiency promotes Salmonella dissemination from the gut. Nat Med. 2008;14(4):421–428.
    1. Ryan ES, et al. Loss of function of intestinal IL-17 and IL-22 producing cells contributes to inflammation and viral persistence in SIV-infected rhesus macaques. PLoS Pathog. 2016;12(2):e1005412.
    1. Stieh DJ, et al. Th17 cells are preferentially infected very early after vaginal transmission of SIV in Mmcaques. Cell Host Microbe. 2016;19(4):529–540.
    1. Chabaud M, et al. Human interleukin-17: A T cell-derived proinflammatory cytokine produced by the rheumatoid synovium. Arthritis Rheum. 1999;42(5):963–970.
    1. Kotake S, et al. IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis. J Clin Invest. 1999;103(9):1345–1352.
    1. Duerr RH, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006;314(5804):1461–1463.
    1. Stahl EA, et al. BIRAC Consortium; YEAR Consortium Genome-wide association study meta-analysis identifies seven new rheumatoid arthritis risk loci. Nat Genet. 2010;42(6):508–514.
    1. Liu Y, et al. A genome-wide association study of psoriasis and psoriatic arthritis identifies new disease loci. PLoS Genet. 2008;4(3):e1000041.
    1. Tanabe S. The effect of probiotics and gut microbiota on Th17 cells. Int Rev Immunol. 2013;32(5-6):511–525.
    1. Ritchie ML, Romanuk TN. A meta-analysis of probiotic efficacy for gastrointestinal diseases. PLoS One. 2012;7(4):e34938.
    1. Vaishnava S, et al. The antibacterial lectin RegIIIgamma promotes the spatial segregation of microbiota and host in the intestine. Science. 2011;334(6053):255–258.
    1. Howitt MR, et al. Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut. Science. 2016;351(6279):1329–1333.
    1. Earle KA, et al. Quantitative Imaging of Gut Microbiota Spatial Organization. Cell Host Microbe. 2015;18(4):478–488.
    1. Segata N, et al. Metagenomic microbial community profiling using unique clade-specific marker genes. Nat Methods. 2012;9(8):811–814.
    1. Segata N, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12(6):R60.
    1. Cipolletta D, et al. PPAR-γ is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature. 2012;486(7404):549–553.
    1. Kuleshov MV, et al. Enrichr: A comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44(W1):W90–W97.

Source: PubMed

3
Iratkozz fel