Gut immune maturation depends on colonization with a host-specific microbiota

Hachung Chung, Sünje J Pamp, Jonathan A Hill, Neeraj K Surana, Sanna M Edelman, Erin B Troy, Nicola C Reading, Eduardo J Villablanca, Sen Wang, Jorge R Mora, Yoshinori Umesaki, Diane Mathis, Christophe Benoist, David A Relman, Dennis L Kasper, Hachung Chung, Sünje J Pamp, Jonathan A Hill, Neeraj K Surana, Sanna M Edelman, Erin B Troy, Nicola C Reading, Eduardo J Villablanca, Sen Wang, Jorge R Mora, Yoshinori Umesaki, Diane Mathis, Christophe Benoist, David A Relman, Dennis L Kasper

Abstract

Gut microbial induction of host immune maturation exemplifies host-microbe mutualism. We colonized germ-free (GF) mice with mouse microbiota (MMb) or human microbiota (HMb) to determine whether small intestinal immune maturation depends on a coevolved host-specific microbiota. Gut bacterial numbers and phylum abundance were similar in MMb and HMb mice, but bacterial species differed, especially the Firmicutes. HMb mouse intestines had low levels of CD4(+) and CD8(+) T cells, few proliferating T cells, few dendritic cells, and low antimicrobial peptide expression--all characteristics of GF mice. Rat microbiota also failed to fully expand intestinal T cell numbers in mice. Colonizing GF or HMb mice with mouse-segmented filamentous bacteria (SFB) partially restored T cell numbers, suggesting that SFB and other MMb organisms are required for full immune maturation in mice. Importantly, MMb conferred better protection against Salmonella infection than HMb. A host-specific microbiota appears to be critical for a healthy immune system.

Copyright © 2012 Elsevier Inc. All rights reserved.

Figures

Figure 1. MMb and HMb Mouse Gut…
Figure 1. MMb and HMb Mouse Gut Microbiotas Are Similar in Major Bacterial Phyla Abundance with Differences at the OTU Level
(A) Schematic of colonization model (see text for details) is illustrated. Blue and red arrowheads indicate fecal pellet collection for bacterial 16S rDNA sequencing. Offspring were sacrificed for immune system analysis. (B) Relative abundance of major bacterial phyla in the gut microbiota from MMb and HMb mice is shown. P0, parents; F1, first-generation offspring; F2, secondgeneration offspring. Each bar represents an individual mouse. Apparent differences in the Firmicutes-to-Bacteroidetes ratio between inoculum samples and recipients may have resulted from the observed differential DNA extraction performances of fecal suspensions (high water content) and fecal pellets (low water content). (C–E) Detailed relative abundance of bacterial taxa in the three most abundant major phyla is presented. (F) Number (percentage) of shared OTUs in each major bacterial phylum in MMb and HMb fecal pellets is demonstrated. See also Figures S1D and S1E. (G and H) Gut microbial communities from individual mice, clustered according to principal coordinates analysis of unweighted UniFrac distances, is illustrated. Percentages of variation explained by plotted principal coordinates P1 and P2 are indicated on the x and y axes, respectively.
Figure 2. MMb Mice Have More Small…
Figure 2. MMb Mice Have More Small Intestinal T Cells Than Do HMb Mice
(A and B) IELs were extracted from the small intestine; the remaining LP tissue was digested. Absolute numbers of CD3+CD103+TCRβ+ among IELs (B) and CD3+CD4+ and CD3+CD8+ cells from LP (A) were quantitated by flow cytometry and normalized to small intestine length. SPF and GF SW mice were age matched. See also Figures S2A and S2B. *p < 0.05, **p < 0.01, ***p < 0.001. NS, not significant. (C) Sections of small intestine were stained with FITC-conjugated antibody to CD3 (green) and counterstained with DAPI (blue).
Figure 3. The MMb, but Not the…
Figure 3. The MMb, but Not the HMb or RMb, Expands T Cell Populations in Small Intestinal Tissue and Secondary Gut Lymphoid Organs
(A–C) PP number (A) and average PP size (B) per small intestine were compared. PPs were mashed, stained for CD3, and subjected to flow cytometry (C). See also Figures S3A and S3B. (D and E) Total T cell numbers in MLNs (D) and spleen (E) are shown. See also Figures S3C–S3G. (F) GF mice (3–4 weeks old) were orally gavaged with the original mouse (M) or human (H) inoculum or with feces pooled from ten additional human donors (H10 inoculum). T cell numbers were measured after 4 weeks of colonization. (G) GF mice were orally gavaged with Sprague-Dawley rat feces and bred in vinyl isolators to obtain RMb offspring. T cell numbers in age-matched MMb, HMb, and RMb offspring were compared. *p < 0.05, **p < 0.01, ***p < 0.001. NS, not significant.
Figure 4. Host-Specific Gut Microbiota Induction of…
Figure 4. Host-Specific Gut Microbiota Induction of T Cell Proliferation in Secondary Gut Lymphoid Organs Leads to Expansion of Small Intestinal T Cells
(A) Representative flow cytometry plots of CD44hiCD62Llo (effector/memory) and CD44loCD62Lhi (naive) expression on CD3+CD4+ T cells in PPs of MMb and HMb offspring are presented. Numbers indicate cell percentages in the quadrant. (B) Combined data for PP CD3+CD4+ and CD3+CD8+ cells (n = 7) are illustrated. (C) Mice injected with BrdU were sacrificed 2 hr later. CD3+ T cells were stained with FITC-conjugated antibody to BrdU for detection of proliferating cells. See also Figures S4A–S4C. *p < 0.05, **p < 0.01, ***p < 0.001. NS, not significant.
Figure 5. Distinct Gene Expression Profile in…
Figure 5. Distinct Gene Expression Profile in Small Intestinal T Cells from HMb Mice
(A) Microarray analysis comparing CD4+ T cell gene expression in GF mice with that in HMb mice (left) and MMb mice (right) is demonstrated. CD4+ T cells were sorted from spleen (SPL), MLNs, and small intestinal LP. Data are mean values from three to five independent experiments. Numbers indicate genes showing a ≥2-fold difference in expression between groups; red numbers indicate overexpression and blue numbers underexpression. (B) Fold change versus fold-change analysis compares MMb mice with GF mice in terms of gene expression in CD4+ T cells sorted from MLNs (y axis) and spleen (x axis) (left). A heatmap (right) shows differentially expressed genes in CD4+ T cells sorted from the MLN. Some genes (Hspa1a, Socs3) were detected with multiple probes. Genes with the highest and lowest transcript levels are red and blue, respectively. See also Figure S5A. (C) Fold change versus fold-change analysis compares gene expression in CD4+ T cells sorted from small intestinal LP of GF mice versus MMb mice (y axis) or HMb mice (x axis). (D) Heatmap shows differential cytokine expression in CD4+ T cells from small intestinal LP. Data are from three independent experiments. See also Figures S5B–S5F. *p < 0.05, MMb versus GF; **p < 0.05, HMb versus GF; ***p < 0.05, HMb versus MMb.
Figure 6. SFB Play a Role in…
Figure 6. SFB Play a Role in Rescuing Intestinal T Cell Numbers and Exhibit Host Specificity
(A) Abundance of SFB in MMb, HMb, and SFB-monocolonized mice, measured as SFB-specific 16S rDNA copy numbers by qPCR analysis of fecal pellets, is shown. Inset values indicate number of SFB 16S rDNA copies/ml in inocula. ND, not detected. (B and C) Absolute T cell numbers in IEL (CD3+CD103+TCRb+) and PP (CD3+CD4+ and CD3+CD8+) compartments of MMb, SFB-monocolonized, and GF mice (B) and HMb mice cohoused with MMb or SFB-monocolonized mice for 4 weeks (C) are presented. In (C), as a negative control, HMb mice were cohoused with HMb mice. See also Figures S6A–S6E.
Figure 7. MMb Confers Better Protection against…
Figure 7. MMb Confers Better Protection against Salmonella enterica Serovar Typhimurium Than HMb
(A–C) Mice colonized with different microbiotas were orally gavaged with ~1 × 107 salmonellae; the Salmonella load in fecal pellets was measured daily (A). Mice were sacrificed on day 4 after infection, and the Salmonella load in the spleen was measured (B). Cecal sections were stained with hematoxylin and eosin, and disease was scored (C). ***p < 0.001. (D and E) Number of CD3+CD4+ T cells in PPs expressing RORgt+ (D) and Foxp3+ (E), as derived by intracellular staining and flow cytometry, is illustrated. See alsoFigures S6F–S6G. (F) Abundance of SFB in fecal samples from Sprague-Dawley rats and RMb-colonized mice, measured as SFB-specific 16S rDNA copy numbers by qPCR, is shown. Fecal pellets from RMb parents were collected on postgavage days 3 (RMb P 3d) and 29 (RMb P 29d); those from RMb offspring were collected at 6 weeks of age (RMb F1 6w). ND, not detected. (G) Gram-stained Sprague-Dawley rat fecal pellets resuspended in PBS are illustrated. Blue arrows indicate bacteria with long filamentous structures representative of SFB. *p < 0.05, **p < 0.01, ***p < 0.001. NS, not significant.

References

    1. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, Fernandes GR, Tap J, Bruls T, Batto JM, et al. MetaHIT Consortium Enterotypes of the human gut microbiome. Nature. 2011;473:174–180.
    1. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, Cheng G, Yamasaki S, Saito T, Ohba Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–341.
    1. Bandeira A, Mota-Santos T, Itohara S, Degermann S, Heusser C, Tonegawa S, Coutinho A. Localization of gamma/delta T cells to the intestinal epithelium is independent of normal microbial colonization. J. Exp. Med. 1990;172:239–244.
    1. Chassin C, Kocur M, Pott J, Duerr CU, Gütle D, Lotz M, Hornef MW. miR-146a mediates protective innate immune tolerance in the neonate intestine. Cell Host Microbe. 2010;8:358–368.
    1. Chung H, Kasper DL. Microbiota-stimulated immune mechanisms to maintain gut homeostasis. Curr. Opin. Immunol. 2010;22:455–460.
    1. Croswell A, Amir E, Teggatz P, Barman M, Salzman NH. Prolonged impact of antibiotics on intestinal microbial ecology and susceptibility to enteric Salmonella infection. Infect. Immun. 2009;77:2741–2753.
    1. Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc. Natl. Acad. Sci. USA. 2011;108(Suppl 1):4554–4561.
    1. Dethlefsen L, McFall-Ngai M, Relman DA. An ecological and evolutionary perspective on human-microbe mutualism and disease. Nature. 2007;449:811–818.
    1. Duan J, Chung H, Troy E, Kasper DL. Microbial colonization drives expansion of IL-1 receptor 1-expressing and IL-17-producing gamma/delta T cells. Cell Host Microbe. 2010;7:140–150.
    1. Eberl G, Boneca IG. Bacteria and MAMP-induced morphogenesis of the immune system. Curr. Opin. Immunol. 2010;22:448–454.
    1. Eberl G, Marmon S, Sunshine MJ, Rennert PD, Choi Y, Littman DR. An essential function for the nuclear receptor RORgamma(t) in the generation of fetal lymphoid tissue inducer cells. Nat. Immunol. 2004;5:64–73.
    1. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–1638.
    1. Ferreira RB, Gill N, Willing BP, Antunes LC, Russell SL, Croxen MA, Finlay BB. The intestinal microbiota plays a role in Salmonella-induced colitis independent of pathogen colonization. PLoS One. 2011;6:e20338.
    1. Feuerer M, Hill JA, Kretschmer K, von Boehmer H, Mathis D, Benoist C. Genomic definition of multiple ex vivo regulatory T cell subphenotypes. Proc. Natl. Acad. Sci. USA. 2010;107:5919–5924.
    1. Gaboriau-Routhiau V, Rakotobe S, Lécuyer E, Mulder I, Lan A, Bridonneau C, Rochet V, Pisi A, De Paepe M, Brandi G, et al. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity. 2009;31:677–689.
    1. Glover JR, Lindquist S. Hsp104, Hsp70, and Hsp40: a novel chaperone system that rescues previously aggregated proteins. Cell. 1998;94:73–82.
    1. Hand T, Belkaid Y. Microbial control of regulatory and effector T cell responses in the gut. Curr. Opin. Immunol. 2010;22:63–72.
    1. Heczko U, Abe A, Finlay BB. Segmented filamentous bacteria prevent colonization of enteropathogenic Escherichia coli O103 in rabbits. J. Infect. Dis. 2000;181:1027–1033.
    1. Imaoka A, Setoyama H, Takagi A, Matsumoto S, Umesaki Y. Improvement of human faecal flora-associated mouse model for evaluation of the functional foods. J. Appl. Microbiol. 2004;96:656–663.
    1. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126:1121–1133.
    1. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139:485–498.
    1. Johansson ME, Hansson GC. Microbiology. Keeping bacteria at a distance. Science. 2011;334:182–183.
    1. Kelsall BL, Rescigno M. Mucosal dendritic cells in immunity and inflammation. Nat. Immunol. 2004;5:1091–1095.
    1. Klaasen HL, Koopman JP, Van den Brink ME, Bakker MH, Poelma FG, Beynen AC. Intestinal, segmented, filamentous bacteria in a wide range of vertebrate species. Lab. Anim. 1993;27:141–150.
    1. Koropatnick TA, Engle JT, Apicella MA, Stabb EV, Goldman WE, McFall-Ngai MJ. Microbial factor-mediated development in a host-bacterial mutualism. Science. 2004;306:1186–1188.
    1. Kunkel EJ, Campbell DJ, Butcher EC. Chemokines in lymphocyte trafficking and intestinal immunity. Microcirculation. 2003;10:313–323.
    1. Leishman AJ, Gapin L, Capone M, Palmer E, MacDonald HR, Kronenberg M, Cheroutre H. Precursors of functional MHC class I- or class II-restricted CD8alphaalpha(+) T cells are positively selected in the thymus by agonist self-peptides. Immunity. 2002;16:355–364.
    1. Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA. 2005;102:11070–11075.
    1. Ley RE, Hamady M, Lozupone C, Turnbaugh PJ, Ramey RR, Bircher JS, Schlegel ML, Tucker TA, Schrenzel MD, Knight R, Gordon JI. Evolution of mammals and their gut microbes. Science. 2008a;320:1647–1651.
    1. Ley RE, Lozupone CA, Hamady M, Knight R, Gordon JI. Worlds within worlds: evolution of the vertebrate gut microbiota. Nat. Rev. Microbiol. 2008b;6:776–788.
    1. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122:107–118.
    1. Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453:620–625.
    1. Mora JR, Bono MR, Manjunath N, Weninger W, Cavanagh LL, Rosemblatt M, Von Andrian UH. Selective imprinting of gut-homing T cells by Peyer’s patch dendritic cells. Nature. 2003;424:88–93.
    1. Mowat AM. Anatomical basis of tolerance and immunity to intestinal antigens. Nat. Rev. Immunol. 2003;3:331–341.
    1. Obata T, Goto Y, Kunisawa J, Sato S, Sakamoto M, Setoyama H, Matsuki T, Nonaka K, Shibata N, Gohda M, et al. Indigenous opportunistic bacteria inhabit mammalian gut-associated lymphoid tissues and share a mucosal antibody-mediated symbiosis. Proc. Natl. Acad. Sci. USA. 2010;107:7419–7424.
    1. Ochman H, Worobey M, Kuo CH, Ndjango JB, Peeters M, Hahn BH, Hugenholtz P. Evolutionary relationships of wild hominids recapitulated by gut microbial communities. PLoS Biol. 2010;8:e1000546.
    1. Olszak T, An D, Zeissig S, Vera MP, Richter J, Franke A, Glickman JN, Siebert R, Baron RM, Kasper DL, et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science. 2012;336:489–493.
    1. Pais R, Lohs C, Wu Y, Wang J, Aksoy S. The obligate mutualist Wigglesworthia glossinidia influences reproduction, digestion, and immunity processes of its host, the tsetse fly. Appl. Environ. Microbiol. 2008;74:5965–5974.
    1. Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO. Development of the human infant intestinal microbiota. PLoS Biol. 2007;5:e177.
    1. Prakash T, Oshima K, Morita H, Fukuda S, Imaoka A, Kumar N, Sharma VK, Kim SW, Takahashi M, Saitou N, et al. Complete genome sequences of rat and mouse segmented filamentous bacteria, a potent inducer of th17 cell differentiation. Cell Host Microbe. 2011;10:273–284.
    1. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. MetaHIT Consortium A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59–65.
    1. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118:229–241.
    1. Rawls JF, Mahowald MA, Ley RE, Gordon JI. Reciprocal gut microbiota transplants from zebrafish and mice to germ-free recipients reveal host habitat selection. Cell. 2006;127:423–433.
    1. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat. Rev. Immunol. 2009;9:313–323.
    1. Salazar-Gonzalez RM, Niess JH, Zammit DJ, Ravindran R, Srinivasan A, Maxwell JR, Stoklasek T, Yadav R, Williams IR, Gu X, et al. CCR6-mediated dendritic cell activation of pathogen-specific T cells in Peyer’s patches. Immunity. 2006;24:623–632.
    1. Sczesnak A, Segata N, Qin X, Gevers D, Petrosino JF, Huttenhower C, Littman DR, Ivanov II. The genome of th17 cell-inducing segmented filamentous bacteria reveals extensive auxotrophy and adaptations to the intestinal environment. Cell Host Microbe. 2011;10:260–272.
    1. Smith K, McCoy KD, Macpherson AJ. Use of axenic animals in studying the adaptation of mammals to their commensal intestinal microbiota. Semin. Immunol. 2007;19:59–69.
    1. Snel J, Heinen PP, Blok HJ, Carman RJ, Duncan AJ, Allen PC, Collins MD. Comparison of 16S rRNA sequences of segmented filamentous bacteria isolated from mice, rats, and chickens and proposal of “Candidatus Arthromitus”. Int. J. Syst. Bacteriol. 1995;45:780–782.
    1. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007;449:804–810.
    1. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, et al. A core gut microbiome in obese and lean twins. Nature. 2009a;457:480–484.
    1. Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci. Transl. Med. 2009b;1:6ra14.
    1. Umesaki Y, Setoyama H, Matsumoto S, Imaoka A, Itoh K. Differential roles of segmented filamentous bacteria and clostridia in development of the intestinal immune system. Infect. Immun. 1999;67:3504–3511.
    1. Vaishnava S, Behrendt CL, Ismail AS, Eckmann L, Hooper LV. Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface. Proc. Natl. Acad. Sci. USA. 2008;105:20858–20863.
    1. Vaishnava S, Yamamoto M, Severson KM, Ruhn KA, Yu X, Koren O, Ley R, Wakeland EK, Hooper LV. The antibacterial lectin RegIIIgamma promotes the spatial segregation of microbiota and host in the intestine. Science. 2011;334:255–258.
    1. Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, Stonebraker AC, Hu C, Wong FS, Szot GL, Bluestone JA, et al. Innate immunity and intestinal microbiota in the development of type 1 diabetes. Nature. 2008;455:1109–1113.
    1. Wills-Karp M, Santeliz J, Karp CL. The germless theory of allergic disease: revisiting the hygiene hypothesis. Nat. Rev. Immunol. 2001;1:69–75.
    1. Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, Littman DR, Benoist C, Mathis D. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity. 2010;32:815–827.
    1. Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, Chung Y, Ma L, Shah B, Panopoulos AD, Schluns KS, et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity. 2008;28:29–39.

Source: PubMed

3
Iratkozz fel