The gut microbiome modulates colon tumorigenesis

Joseph P Zackular, Nielson T Baxter, Kathryn D Iverson, William D Sadler, Joseph F Petrosino, Grace Y Chen, Patrick D Schloss, Joseph P Zackular, Nielson T Baxter, Kathryn D Iverson, William D Sadler, Joseph F Petrosino, Grace Y Chen, Patrick D Schloss

Abstract

Recent studies have shown that individuals with colorectal cancer have an altered gut microbiome compared to healthy controls. It remains unclear whether these differences are a response to tumorigenesis or actively drive tumorigenesis. To determine the role of the gut microbiome in the development of colorectal cancer, we characterized the gut microbiome in a murine model of inflammation-associated colorectal cancer that mirrors what is seen in humans. We followed the development of an abnormal microbial community structure associated with inflammation and tumorigenesis in the colon. Tumor-bearing mice showed enrichment in operational taxonomic units (OTUs) affiliated with members of the Bacteroides, Odoribacter, and Akkermansia genera and decreases in OTUs affiliated with members of the Prevotellaceae and Porphyromonadaceae families. Conventionalization of germfree mice with microbiota from tumor-bearing mice significantly increased tumorigenesis in the colon compared to that for animals colonized with a healthy gut microbiome from untreated mice. Furthermore, at the end of the model, germfree mice colonized with microbiota from tumor-bearing mice harbored a higher relative abundance of populations associated with tumor formation in conventional animals. Manipulation of the gut microbiome with antibiotics resulted in a dramatic decrease in both the number and size of tumors. Our results demonstrate that changes in the gut microbiome associated with inflammation and tumorigenesis directly contribute to tumorigenesis and suggest that interventions affecting the composition of the microbiome may be a strategy to prevent the development of colon cancer.

Importance: The trillions of bacteria that live in the gut, known collectively as the gut microbiome, are important for normal functioning of the intestine. There is now growing evidence that disruptive changes in the gut microbiome are strongly associated with the development colorectal cancer. However, how the gut microbiome changes with time during tumorigenesis and whether these changes directly contribute to disease have not been determined. We demonstrate using a mouse model of inflammation-driven colon cancer that there are dramatic, continual alterations in the microbiome during the development of tumors, which are directly responsible for tumor development. Our results suggest that interventions that target these changes in the microbiome may be an effective strategy for preventing the development of colorectal cancer.

Figures

FIG 1
FIG 1
Inflammation-induced tumorigenesis is commensal dependent. (A) Mice were injected with azoxymethane (AOM) on day 1, followed by 3 subsequent rounds of water-administered 2% DSS. Colons were harvested 73 days after AOM, and tumors were grossly counted. Black wedges indicate fecal samples used for gut microbiome analysis (n = 12). (B) Representative mice were euthanized following each round of DSS to identify macroscopic tumors (n = 5 for each time point). An antibiotic cocktail of metronidazole, streptomycin, and vancomycin was administered in the drinking water of a separate cohort of mice for the duration of the model (n = 9). Statistical analysis was performed using a two-tailed Student’s t test. *, P < 0.01. (C) Representative images of tumors in the distal colon of conventional mice treated with AOM/DSS (n = 12) and mice treated with an antibiotic cocktail and AOM/DSS (n = 9). Error bars represent ±SEM.
FIG 2
FIG 2
Development of a dysbiotic gut microbiome during colon tumorigenesis. Microbiome analysis was performed with fecal samples from 10 representative mice; color coding is as indicated in Fig. 1A. (A) Inverse Simpson’s diversity index. (B) Observed community richness estimate. Statistical analysis was performed using repeated-measures paired group analysis of variance. (C) Nonmetric multidimensional scaling (NMDS) ordination based on θyc distances for all 10 mice during the AOM/DSS model. (D) Average θYC distance within (black) and between (gray) phases of the model. Error bars represent ±SEM.
FIG 3
FIG 3
Heat map of OTUs with relative abundances that are significantly different from their relative abundances at the time of AOM administration. The average OTU abundance between mice for each OTU was calculated for each time point. The timeline is colored for the following groups: baseline samples (prior to AOM), black; following the first round of DSS, blue; following the second round of DSS, green; following the third round of DSS, red. The OTU number and taxonomic group based on RDP classification are represented for each row. Repeated-measures paired group analysis of variance was used to identify significantly altered OTUs.
FIG 4
FIG 4
Tumor-associated gut microbiome alterations exacerbate tumorigenesis in germfree mice. (A) Number of tumors observed at the end of the model when germfree mice were colonized using bedding from healthy mice (Healthy community) or mice with tumors (Dysbiotic community). (B) Representative images of tumors in the distal colon of mice conventionalized with a healthy microbiome (n = 10) or the microbiome of tumor-bearing mice (n = 9). (C) NMDS ordination based on θyc distances for all 19 mice following conventionalization with a healthy microbiome (Healthy community) or the microbiome of tumor-bearing mice (Dysbiotic community). Error bars represent ±SEM.

References

    1. Parkin DM, Bray F, Ferlay J, Pisani P. 2005. Global cancer statistics, 2002. CA Cancer J. Clin. 55:74–108
    1. Chambers WM, Warren BF, Jewell DP, Mortensen NJ. 2005. Cancer surveillance in ulcerative colitis. Br. J. Surg. 92:928–936
    1. Huxley RR, Ansary-Moghaddam A, Clifton P, Czernichow S, Parr CL, Woodward M. 2009. The impact of dietary and lifestyle risk factors on risk of colorectal cancer: a quantitative overview of the epidemiological evidence. Int. J. Cancer 125:171–180
    1. Larsson SC, Rafter J, Holmberg L, Bergkvist L, Wolk A. 2005. Red meat consumption and risk of cancers of the proximal colon, distal colon and rectum: the Swedish Mammography Cohort. Int. J. Cancer 113:829–834
    1. Slattery ML. 2000. Diet, lifestyle, and colon cancer. Semin. Gastrointest. Dis. 11:142–146
    1. Cheesman SE, Neal JT, Mittge E, Seredick BM, Guillemin K. 2011. Epithelial cell proliferation in the developing zebrafish intestine is regulated by the Wnt pathway and microbial signaling via Myd88. Proc. Natl. Acad. Sci. U. S. A. 108:4570–4577
    1. Dolara P, Caderni G, Salvadori M, Morozzi G, Fabiani R, Cresci A, Orpianesi C, Trallori G, Russo A, Palli D. 2002. Fecal levels of short-chain fatty acids and bile acids as determinants of colonic mucosal cell proliferation in humans. Nutr. Cancer 42:186–190
    1. Stappenbeck TS, Hooper LV, Gordon JI. 2002. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proc. Natl. Acad. Sci. U. S. A. 99:15451–15455
    1. Rakoff-Nahoum S, Medzhitov R. 2007. Regulation of spontaneous intestinal tumorigenesis through the adaptor protein MyD88. Science 317:124–127
    1. Chen HM, Yu YN, Wang JL, Lin YW, Kong X, Yang CQ, Yang L, Liu ZJ, Yuan YZ, Liu F, Wu JX, Zhong L, Fang DC, Zou W, Fang JY. 2013. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am. J. Clin. Nutr. 97:1044–1052
    1. Chen W, Liu F, Ling Z, Tong X, Xiang C. 2012. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS One 7:e39743.10.1371/journal.pone.0039743
    1. Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, Ojesina AI, Jung J, Bass AJ, Tabernero J, Baselga J, Liu C, Shivdasani RA, Ogino S, Birren BW, Huttenhower C, Garrett WS, Meyerson M. 2012. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 22:292–298
    1. Geng J, Fan H, Tang X, Zhai H, Zhang Z. 2013. Diversified pattern of the human colorectal cancer microbiome. Gut Pathog. 5:2.10.1186/1757-4749-5-2
    1. Shen XJ, Rawls JF, Randall T, Burcal L, Mpande CN, Jenkins N, Jovov B, Abdo Z, Sandler RS, Keku TO. 2010. Molecular characterization of mucosal adherent bacteria and associations with colorectal adenomas. Gut Microbes 1:138–147
    1. Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, Corthier G, Tran Van Nhieu J, Furet JP. 2011. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS One 6:e16393.10.1371/journal.pone.0016393
    1. Wang T, Cai G, Qiu Y, Fei N, Zhang M, Pang X, Jia W, Cai S, Zhao L. 2012. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 6:320–329
    1. Arthur JC, Perez-Chanona E, Mühlbauer M, Tomkovich S, Uronis JM, Fan TJ, Campbell BJ, Abujamel T, Dogan B, Rogers AB, Rhodes JM, Stintzi A, Simpson KW, Hansen JJ, Keku TO, Fodor AA, Jobin C. 2012. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 338:120–123
    1. Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X, Yen HR, Huso DL, Brancati FL, Wick E, McAllister F, Housseau F, Pardoll DM, Sears CL. 2009. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat. Med. 15:1016–1022
    1. Hague A, Elder DJ, Hicks DJ, Paraskeva C. 1995. Apoptosis in colorectal tumor-cells—induction by the short-chain fatty-acids butyrate, propionate and acetate and by the bile-salt deoxycholate. Int. J. Cancer 60:400–406
    1. Ruemmele FM, Schwartz S, Seidman EG, Dionne S, Levy E, Lentze MJ. 2003. Butyrate induced Caco-2 cell apoptosis is mediated via the mitochondrial pathway. Gut 52:94–100
    1. Ullman TA, Itzkowitz SH. 2011. Intestinal inflammation and cancer. Gastroenterology 140:1807–1816
    1. Eaden J, Abrams K, Ekbom A, Jackson E, Mayberry J. 2000. Colorectal cancer prevention in ulcerative colitis: a case-control study. Aliment. Pharmacol. Ther. 14:145–153
    1. Arthur JC, Jobin C. 2013. The complex interplay between inflammation, the microbiota and colorectal cancer. Gut Microbes 4:253–258
    1. Lin WW, Karin M. 2007. A cytokine-mediated link between innate immunity, inflammation, and cancer. J. Clin. Invest. 117:1175–1183
    1. De Robertis M, Massi E, Poeta ML, Carotti S, Morini S, Cecchetelli L, Signori E, Fazio VM. 2011. The AOM/DSS murine model for the study of colon carcinogenesis: from pathways to diagnosis and therapy studies. J. Carcinog. 10:9.10.4103/1477-3163.78279
    1. Tanaka T, Kohno H, Suzuki R, Yamada Y, Sugie S, Mori H. 2003. A novel inflammation-related mouse colon carcinogenesis model induced by azoxymethane and dextran sodium sulfate. Cancer Sci. 94:965–973
    1. Chen GY, Shaw MH, Redondo G, Núñez G. 2008. The innate immune receptor Nod1 protects the intestine from inflammation-induced tumorigenesis. Cancer Res. 68:10060–10067
    1. Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. 2009. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci. Transl. Med. 1:6ra14.10.1126/scitranslmed.3000322
    1. Smith MI, Yatsunenko T, Manary MJ, Trehan I, Mkakosya R, Cheng J, Kau AL, Rich SS, Concannon P, Mychaleckyj JC, Liu J, Houpt E, Li JV, Holmes E, Nicholson J, Knights D, Ursell LK, Knight R, Gordon JI. 2013. Gut microbiomes of Malawian twin pairs discordant for Kwashiorkor. Science 339:548–554
    1. Couturier-Maillard A, Secher T, Rehman A, Normand S, De Arcangelis A, Haesler R, Huot L, Grandjean T, Bressenot A, Delanoye-Crespin A, Gaillot O, Schreiber S, Lemoine Y, Ryffel B, Hot D, Nùñez G, Chen G, Rosenstiel P, Chamaillard M. 2013. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J. Clin. Invest. 123:700–711
    1. Sears CL, Islam S, Saha A, Arjumand M, Alam NH, Faruque AS, Salam MA, Shin J, Hecht D, Weintraub A, Sack RB, Qadri F. 2008. Association of enterotoxigenic Bacteroides fragilis infection with inflammatory diarrhea. Clin. Infect. Dis. 47:797–803
    1. Segain JP, de la Bletiere DR, Bourreille A, Leray V, Gervois N, Rosales C, Ferrier L, Bonnet C, Blottière HM, Galmiche JP. 2000. Butyrate inhibits inflammatory responses through NF kappaB inhibition: implications for Crohn’s disease. Gut 47:397–403
    1. Aune D, Chan DS, Lau R, Vieira R, Greenwood DC, Kampman E, Norat T. 2011. Dietary fibre, whole grains, and risk of colorectal cancer: systematic review and dose-response meta-analysis of prospective studies. BMJ 343:d6617.10.1136/bmj.d6617
    1. O’Keefe SJ, Ou J, Aufreiter S, O’Connor D, Sharma S, Sepulveda J, Fukuwatari T, Shibata K, Mawhinney T. 2009. Products of the colonic microbiota mediate the effects of diet on colon cancer risk. J. Nutr. 139:2044–2048
    1. Schloss PD, Schubert AM, Zackular JP, Iverson KD, Young VB, Petrosino JF. 2012. Stabilization of the murine gut microbiome following weaning. Gut Microbes 3:383–393
    1. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, Lesniewski RA, Oakley BB, Parks DH, Robinson CJ, Sahl JW, Stres B, Thallinger GG, Van Horn DJ, Weber CF. 2009. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 75:7537–7541
    1. Schloss PD, Gevers D, Westcott SL. 2011. Reducing the effects of PCR amplification and sequencing artifacts on 16S rRNA-based studies. PLoS One 6:e27310.10.1371/journal.pone.0027310
    1. Quince C, Lanzén A, Curtis TP, Davenport RJ, Hall N, Head IM, Read LF, Sloan WT. 2009. Accurate determination of microbial diversity from 454 pyrosequencing data. Nat. Methods 6:639–641
    1. Pruesse E, Quast C, Knittel K, Fuchs BM, Ludwig W, Peplies J, Glöckner FO. 2007. SILVA: a comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Res. 35:7188–7196
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. 2007. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl. Environ. Microbiol. 73:5261–5267
    1. Magurran AE. 1988. Ecological diversity and its measurement. Princeton University Press, Princeton, NJ.
    1. Yue JC, Clayton MK. 2005. A similarity measure based on species proportions. Commun. Statist. Theory Methods 34:2123–2131
    1. Lozupone C, Knight R. 2005. UniFrac: a new phylogenetic method for comparing microbial communities. Appl. Environ. Microbiol. 71:8228–8235
    1. Sheneman L, Evans J, Foster JA. 2006. Clearcut: a fast implementation of relaxed neighbor joining. Bioinformatics 22:2823–2824
    1. Martin AP. 2002. Phylogenetic approaches for describing and comparing the diversity of microbial communities. Appl. Environ. Microbiol. 68:3673–3682
    1. Benjamini Y, Hochberg Y. 1995. Controlling the false discovery rate—a practical and powerful approach to multiple testing. J. R. Stat. Soc. B Stat. Methodol. 57:289–300
    1. Liaw A, Wiener M. 2002. Classification and regression by randomForest. R News 2:18–22
    1. Chao A, Chazdon RL, Colwell RK, Shen TJ. 2006. Abundance-based similarity indices and their estimation when there are unseen species in samples. Biometrics 62:361–371
    1. Vaishnava S, Yamamoto M, Severson KM, Ruhn KA, Yu X, Koren O, Ley R, Wakeland EK, Hooper LV. 2011. The antibacterial lectin RegIIIgamma promotes the spatial segregation of microbiota and host in the intestine. Science 334:255–258
    1. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. 2011. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics 27:2194–2200

Source: PubMed

3
S'abonner