Microbiota, Inflammation and Colorectal Cancer
Abstract
:1. Introduction
2. Determinant Factors of Colorectal Cancer (CRC)
3. Intestinal Microbiota and Gut Homeostasis
3.1. Nutrient Metabolism
3.2. Intestinal Barrier Maintenance
3.3. Modulation of Immune System
3.3.1. Intestinal Innate Immune Cells
3.3.2. Intestinal Adaptive Immune Cells
3.4. Protection against Pathogens
4. Intestinal Microbiota and CRC
5. Possible Mechanisms of Action of the Intestinal Microbiota in Colorectal Carcinogenesis
5.1. Enterococcus faecalis
5.2. Bacteroides fragilis
5.3. Fusobacterium nucleatum
5.4. Streptococcus bovis/gallolyticus
5.5. Clostridium septicum
5.6. Helicobacter pylori
5.7. Escherichia coli
6. Conclusions and Future Directions/Clinical Application
Acknowledgments
Conflicts of Interest
Abbreviations
AIEC | Adherent-invasive Escherichia coli |
AKT | Protein kinase B |
AMPs | Antimicrobial peptides |
AOM | Azoxymethane |
Apc | Adenomatous polyposis coli |
BAX | Bcl-2-associated X |
BCL-2 | B-cell lymphoma 2 |
BfPAI | Bacteroides fragilis pathogenicity island |
CDT | Cytolethal distending toxin |
CIF | Cycle-inhibiting factor |
CIMP | CpG island methylator phenotype |
CIN | Chromosomal instability |
CLRs | C-type lectin receptors |
CNF | Cytotoxic necrotizing factor |
COX-2 | Cyclo-oxygénase-2 |
CRC | Colorectal cancer |
CTLA4 | Cytotoxic T lymphocyte-associated antigen 4 |
DAEC | Diffusely adherent Escherichia coli |
DSS | Dextran sodium sulfate |
EAEC | Enteroaggregative Escherichia coli |
EHEC | Enterohemorrhagic Escherichia coli |
EIEC | Enteroinvasive Escherichia coli |
EPEC | Enteropathogenic Escherichia coli |
ETBF | Enterotoxigenic Bacteroides fragilis |
ETEC | Enterotoxigenic Escherichia coli |
FAP | Familial adenomatous polyposis |
GM-CSF | Granulocyte macrophage colony-stimulating factor |
HP-NAP | Helicobacter pylori neutrophil-activating protein |
IBD | Inflammatory bowel diseases |
IECs | Intestinal epithelial cells |
IgA | Immunoglobulin-A |
IL | Interleukin |
ILC3 | Innate lymphoid cells |
IFN-γ | Interferon gamma |
M cells | Microfold cells |
MALT | Mucosa-associated lymphoid tissue |
MAMPs | Microbe-associated molecular patterns |
MAPK | Mitogen-activated protein kinase |
MLH1 | MutL homolog 1 |
MSI | Microsatellite instability |
mTOR | Mammalian target of rapamycin |
MyD88 | Myeloid differentiation primary response gene 88 |
NF-κB | Nuclear factor-kappa B |
NLRs | NOD-like receptors |
NOD | Nucleotide-binding oligomerization domain |
NTBF | Nontoxigenic Bacteroides fragilis |
PAMPs | Pathogen-associated molecular patterns |
PGE2 | Prostaglandin E2 |
PI3K | Phosphoinositide 3-kinase |
pks | Polyketide synthase |
PRRs | Pattern recognition receptors |
ROS | Reactive oxygen species |
RNS | Reactive nitrogen species |
SCFAs | Short-chain fatty acids |
SFB | Segmented filamentous bacteria |
SNP | Single nucleotide polymorphism |
STAT3 | Signal transducer and activator of transcription 3 |
STEAEC | Shiga toxin-producing enteroaggregative Escherichia coli |
TCF | T-cell factor |
Th17 | T helper 17 |
TLRs | Toll-like receptors |
TNF-α | Tumor necrosis factor-α |
References
- Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef] [PubMed]
- Terzić, J.; Grivennikov, S.; Karin, E.; Karin, M. Inflammation and colon cancer. Gastroenterology 2010, 138, 2101–2114. [Google Scholar] [CrossRef] [PubMed]
- Brenner, H.; Kloor, M.; Pox, C.P. Colorectal cancer. Lancet 2014, 383, 1490–1502. [Google Scholar] [CrossRef]
- Tjalsma, H.; Boleij, A.; Marchesi, J.R.; Dutilh, B.E. A bacterial driver–passenger model for colorectal cancer: Beyond the usual suspects. Nat. Rev. Microbiol. 2012, 10, 575–582. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.; Liu, F.; Ling, Z.; Tong, X.; Xiang, C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS ONE 2012, 7, e39743. [Google Scholar] [CrossRef] [PubMed]
- Lu, Y.; Chen, J.; Zheng, J.; Hu, G.; Wang, J.; Huang, C.; Lou, L.; Wang, X.; Zeng, Y. Mucosal adherent bacterial dysbiosis in patients with colorectal adenomas. Sci. Rep. 2016, 6, 26337. [Google Scholar] [CrossRef] [PubMed]
- Gao, Z.; Guo, B.; Gao, R.; Zhu, Q.; Qin, H. Microbiota disbiosis is associated with colorectal cancer. Front. Microbiol. 2015. [Google Scholar] [CrossRef] [PubMed]
- Schwabe, R.F.; Jobin, C. The microbiome and cancer. Nat. Rev. Cancer 2013, 13, 800–812. [Google Scholar] [CrossRef] [PubMed]
- Van der Beek, C.M.; Dejong, C.H.C.; Troost, F.J.; Masclee, A.A.M.; Lenaerts, K. Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutr. Rev. 2017, 75, 286–305. [Google Scholar] [CrossRef] [PubMed]
- Cougnoux, A.; Delmas, J.; Gibold, L.; Faïs, T.; Romagnoli, C.; Robin, F.; Cuevas-Ramos, G.; Oswald, E.; Darfeuille-Michaud, A.; Prati, F.; et al. Small-molecule inhibitors prevent the genotoxic and protumoural effects induced by colibactin-producing bacteria. Gut 2016, 65, 278–285. [Google Scholar] [CrossRef] [PubMed]
- Qamar, T.R.; Syed, F.; Nasir, M.; Rehman, H.; Zahid, M.N.; Liu, R.H.; Iqbal, S. Novel Combination of Prebiotics Galacto-Oligosaccharides and Inulin-Inhibited Aberrant Crypt Foci Formation and Biomarkers of Colon Cancer in Wistar Rats. Nutrients 2016, 8, 465. [Google Scholar] [CrossRef] [PubMed]
- Zsivkovits, M.; Fekadu, K.; Sontag, G.; Nabinger, U.; Huber, W.W.; Kundi, M.; Chakraborty, A.; Foissy, H.; Knasmüller, S. Prevention of heterocyclic amine-induced DNA damage in colon and liver of rats by different lactobacillus strains. Carcinogenesis 2003, 24, 1913–1918. [Google Scholar] [CrossRef] [PubMed]
- Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.-L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [PubMed]
- Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.M.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
- Johns, L.E.; Houlston, R.S. A systematic review and meta-analysis of familial colorectal cancer risk. Am. J. Gastroenterol. 2001, 96, 2992–3003. [Google Scholar] [CrossRef] [PubMed]
- Jasperson, K.W.; Tuohy, T.M.; Neklason, D.W.; Burt, R.W. Hereditary and familial colon cancer. Gastroenterology 2010, 138, 2044–2058. [Google Scholar] [CrossRef] [PubMed]
- Doubeni, C.A.; Laiyemo, A.O.; Major, J.M.; Schootman, M.; Lian, M.; Park, Y.; Graubard, B.I.; Hollenbeck, A.R.; Sinha, R. Socioeconomic status and the risk of colorectal cancer: An analysis of over one-half million adults in the NIH-AARP Diet and Health Study. Cancer 2012, 118, 3636–3644. [Google Scholar] [CrossRef] [PubMed]
- Ryan-Harshman, M.; Aldoori, W. Diet and colorectal cancer. Can. Fam. Physician 2007, 53, 1913–1920. [Google Scholar] [PubMed]
- Boffetta, P.; Hashibe, M. Alcohol and cancer. Lancet Oncol. 2006, 7, 149–156. [Google Scholar] [CrossRef]
- Cho, E.; Smith-Warner, S.A.; Ritz, J.; van den Brandt, P.A.; Colditz, G.A.; Folsom, A.R.; Freudenheim, J.L.; Giovannucci, E.; Goldbohm, R.A.; Graham, S.; et al. Alcohol intake and colorectal cancer: A pooled analysis of 8 cohort studies. Ann. Intern. Med. 2004, 140, 603–613. [Google Scholar] [CrossRef] [PubMed]
- Botteri, E.; Iodice, S.; Bagnardi, V.; Raimondi, S.; Lowenfels, A.B.; Maisonneuve, P. Smoking and colorectal cancer: A meta-analysis. JAMA 2008, 300, 2765–2778. [Google Scholar] [CrossRef] [PubMed]
- Berger, N.A. Obesity and cancer pathogenesis. Ann. N. Y. Acad. Sci. 2014, 1311, 57–76. [Google Scholar] [CrossRef] [PubMed]
- Ma, Y.; Yang, Y.; Wang, F.; Zhang, P.; Shi, C.; Zou, Y.; Qin, H. Obesity and Risk of Colorectal Cancer: A Systematic Review of Prospective Studies. PLoS ONE 2013, 8, e53916. [Google Scholar] [CrossRef] [PubMed]
- Lin, J.E.; Colon-Gonzalez, F.; Blomain, E.; Kim, G.W.; Aing, A.; Stoecker, B.; Rock, J.; Snook, A.E.; Zhan, T.; Hyslop, T.M.; et al. Obesity-induced colorectal cancer is driven by caloric silencing of the guanylin-GUCY2C paracrine signaling axis. Cancer Res. 2016, 76, 339–346. [Google Scholar] [CrossRef] [PubMed]
- Koda, M.; Sulkowska, M.; Kanczuga-Koda, L.; Surmacz, E.; Sulkowski, S. Overexpression of the obesity hormone leptin in human colorectal cancer. J. Clin. Pathol. 2007, 60, 902–906. [Google Scholar] [CrossRef] [PubMed]
- Wang, D.; Chen, J.; Chen, H.; Duan, Z.; Xu, Q.; Wei, M.; Wang, L.; Zhong, M. Leptin regulates proliferation and apoptosis of colorectal carcinoma through PI3K/Akt/mTOR signalling pathway. J. Biosci. 2012, 37, 91–101. [Google Scholar] [CrossRef] [PubMed]
- Walter, V.; Jansen, L.; Knebel, P.; Chang-Claude, J.; Hoffmeister, M.; Brenner, H. Physical activity and survival of colorectal cancer patients: Population-based study from Germany. Int. J. Cancer 2017, 140, 1985–1997. [Google Scholar] [CrossRef] [PubMed]
- Golshiri, P.; Rasooli, S.; Emami, M.; Najimi, A. Effects of Physical Activity on Risk of Colorectal Cancer: A Case-control Study. Int. J. Prev. Med. 2016, 7, 32. [Google Scholar] [PubMed]
- Ghafari, M.; Mohammadian, M.; Valipour, A.A.; Mohammadian-Hafshejani, A. Physical Activity and Colorectal Cancer. Iran. J. Public Health 2016, 45, 1673–1674. [Google Scholar] [PubMed]
- Kruk, J.; Czerniak, U. Physical activity and its relation to cancer risk: Updating the evidence. Asian Pac. J. Cancer Prev. 2013, 14, 3993–4003. [Google Scholar] [CrossRef] [PubMed]
- Jess, T.; Rungoe, C.; Peyrin-Biroulet, L. Risk of colorectal cancer in patients with ulcerative colitis: A meta-analysis of population-based cohort studies. Clin. Gastroenterol. Hepatol. 2012, 10, 639–645. [Google Scholar] [CrossRef] [PubMed]
- Farraye, F.A.; Odze, R.D.; Eaden, J.; Itzkowitz, S.H. AGA Medical Position Statement on the Diagnosis and Management of Colorectal Neoplasia in Inflammatory Bowel Disease. Gastroenterology 2010, 138, 738–745. [Google Scholar] [CrossRef] [PubMed]
- Haslam, A.; Robb, S.W.; Hébert, J.R.; Huang, H.; Wirth, M.D.; Shivappa, N.; Ebell, M.H. The association between Dietary Inflammatory Index scores and the prevalence of colorectal adenoma. Public Health Nutr. 2017. [Google Scholar] [CrossRef] [PubMed]
- Gupta, R.A.; Dubois, R.N. Colorectal cancer prevention and treatment by inhibition of cyclooxygenase-2. Nat. Rev. Cancer 2001, 1, 11–21. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, T.; Kohno, H.; Suzuki, R.; Hata, K.; Sugie, S.; Niho, N.; Sakano, K.; Takahashi, M.; Wakabayashi, K. Dextran sodium sulfate strongly promotes colorectal carcinogenesis in ApcMin/+ mice: Inflammatory stimuli by dextran sodium sulfate results in development of multiple colonic neoplasms. Int. J. Cancer 2006, 118, 25–34. [Google Scholar] [CrossRef] [PubMed]
- Neufert, C.; Becker, C.; Neurath, M.F. An inducible mouse model of colon carcinogenesis for the analysis of sporadic and inflammation-driven tumor progression. Nat. Protoc. 2007, 2, 1998–2004. [Google Scholar] [CrossRef] [PubMed]
- Bayarsaihan, D. Epigenetic Mechanisms in Inflammation. J. Dent. Res. 2011, 90, 9–17. [Google Scholar] [CrossRef] [PubMed]
- Meira, L.B.; Bugni, J.M.; Green, S.L.; Lee, C.-W.; Pang, B.; Borenshtein, D.; Rickman, B.H.; Rogers, A.B.; Moroski-Erkul, C.A.; McFaline, J.L.; et al. DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice. J. Clin. Investig. 2008, 118, 2516–2525. [Google Scholar] [CrossRef] [PubMed]
- Francescone, R.; Hou, V.; Grivennikov, S.I. Cytokines, IBD and colitis-associated cancer. Inflamm. Bowel Dis. 2015, 21, 409–418. [Google Scholar] [CrossRef] [PubMed]
- Basavaraju, U.; Shebl, F.M.; Palmer, A.J.; Berry, S.; Hold, G.L.; El-Omar, E.M.; Rabkin, C.S. Cytokine gene polymorphisms, cytokine levels and the risk of colorectal neoplasia in a screened population of Northeast Scotland. Eur. J. Cancer Prev. 2015, 24, 296–304. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Keku, T.O.; Martin, C.; Galanko, J.; Woosley, J.T.; Schroeder, J.C.; Satia, J.A.; Halabi, S.; Sandler, R.S. Circulating levels of inflammatory cytokines and risk of colorectal adenomas. Cancer Res. 2008, 68, 323–328. [Google Scholar] [CrossRef] [PubMed]
- Song, M.; Mehta, R.S.; Wu, K.; Fuchs, C.S.; Ogino, S.; Giovannucci, E.L.; Chan, A.T. Plasma Inflammatory Markers and Risk of Advanced Colorectal Adenoma in Women. Cancer Prev. Res. 2016, 9, 27–34. [Google Scholar] [CrossRef] [PubMed]
- Knüpfer, H.; Preiss, R. Serum interleukin-6 levels in colorectal cancer patients—A summary of published results. Int. J. Colorectal Dis. 2010, 25, 135–140. [Google Scholar] [CrossRef] [PubMed]
- Hsu, C.-P.; Chung, Y.-C. Influence of interleukin-6 on the invasiveness of human colorectal carcinoma. Anticancer Res. 2006, 26, 4607–4614. [Google Scholar] [PubMed]
- Grivennikov, S.; Karin, E.; Terzic, J.; Mucida, D.; Yu, G.-Y.; Vallabhapurapu, S.; Scheller, J.; Rose-John, S.; Cheroutre, H.; Eckmann, L.; et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell 2009, 15, 103–113. [Google Scholar] [CrossRef] [PubMed]
- Kühn, R.; Löhler, J.; Rennick, D.; Rajewsky, K.; Müller, W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 1993, 75, 263–274. [Google Scholar] [CrossRef]
- Uronis, J.M.; Mühlbauer, M.; Herfarth, H.H.; Rubinas, T.C.; Jones, G.S.; Jobin, C. Modulation of the Intestinal Microbiota Alters Colitis-Associated Colorectal Cancer Susceptibility. PLoS ONE 2009. [Google Scholar] [CrossRef] [PubMed]
- Tomkovich, S.; Yang, Y.; Winglee, K.; Gauthier, J.; Mühlbauer, M.; Sun, X.; Mohamadzadeh, M.; Liu, X.; Martin, P.; Wang, G.P.; et al. Locoregional Effects of Microbiota in a Preclinical Model of Colon Carcinogenesis. Cancer Res. 2017, 77, 2620–2632. [Google Scholar] [CrossRef] [PubMed]
- Arthur, J.C.; Perez-Chanona, E.; Mühlbauer, M.; Tomkovich, S.; Uronis, J.M.; Fan, T.-J.; Campbell, B.J.; Abujamel, T.; Dogan, B.; Rogers, A.B.; et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 2012, 338, 120–123. [Google Scholar] [CrossRef] [PubMed]
- Medvedev, A.E. Toll-like receptor polymorphisms, inflammatory and infectious diseases, allergies, and cancer. J. Interferon Cytokine Res. 2013, 33, 467–484. [Google Scholar] [CrossRef] [PubMed]
- Weber, A.N.; Försti, A. Toll-like receptor genetic variants and colorectal cancer. Oncoimmunology. 2014, 3, e27763. [Google Scholar] [CrossRef] [PubMed]
- Semlali, A.; Parine, N.R.; Al Amri, A.; Azzi, A.; Arafah, M.; Kohailan, M.; Shaik, J.P.; Almadi, M.A.; Aljebreen, A.M.; Alharbi, O.; et al. Association between TLR-9 polymorphisms and colon cancer susceptibility in Saudi Arabian female patients. OncoTargets Ther. 2016, 10, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Sipos, F.; Fűri, I.; Constantinovits, M.; Tulassay, Z.; Műzes, G. Contribution of TLR signaling to the pathogenesis of colitis-associated cancer in inflammatory bowel disease. World J. Gastroenterol. 2014, 20, 12713–12721. [Google Scholar] [CrossRef] [PubMed]
- Peng, G.; Guo, Z.; Kiniwa, Y.; Voo, K.S.; Peng, W.; Fu, T.; Wang, D.Y.; Li, Y.; Wang, H.Y.; Wang, R.-F. Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science 2005, 309, 1380–1384. [Google Scholar] [CrossRef] [PubMed]
- Hoon Rhee, S.; Im, E.; Pothoulakis, C. Toll-like receptor 5 engagement modulates tumor development and growth in a mouse xenograft model of human colon cancer. Gastroenterology 2008, 135, 518–528. [Google Scholar] [CrossRef] [PubMed]
- Heckelsmiller, K.; Beck, S.; Rall, K.; Sipos, B.; Schlamp, A.; Tuma, E.; Rothenfusser, S.; Endres, S.; Hartmann, G. Combined dendritic cell- and CpG oligonucleotide-based immune therapy cures large murine tumors that resist chemotherapy. Eur. J. Immunol. 2002, 32, 3235–3245. [Google Scholar] [CrossRef]
- Lowe, E.L.; Crother, T.R.; Rabizadeh, S.; Hu, B.; Wang, H.; Chen, S.; Shimada, K.; Wong, M.H.; Michelsen, K.S.; Arditi, M. Toll-Like Receptor 2 Signaling Protects Mice from Tumor Development in a Mouse Model of Colitis-Induced Cancer. PLoS ONE 2010, 5, e13027. [Google Scholar] [CrossRef] [PubMed]
- Huang, B.; Zhao, J.; Li, H.; He, K.-L.; Chen, Y.; Mayer, L.; Unkeless, J.C.; Xiong, H. Toll-Like Receptors on Tumor Cells Facilitate Evasion of Immune Surveillance. Cancer Res. 2005, 65, 5009–5014. [Google Scholar] [CrossRef] [PubMed]
- Fukata, M.; Hernandez, Y.; Conduah, D.; Cohen, J.; Chen, A.; Breglio, K.; Goo, T.; Hsu, D.; Xu, R.; Abreu, M.T. Innate immune signaling by Toll-like receptor-4 (TLR4) shapes the inflammatory microenvironment in colitis-associated tumors. Inflamm. Bowel Dis. 2009, 15, 997–1006. [Google Scholar] [CrossRef] [PubMed]
- Rakoff-Nahoum, S.; Medzhitov, R. Regulation of spontaneous intestinal tumorigenesis through the adaptor protein MyD88. Science 2007, 317, 124–127. [Google Scholar] [CrossRef] [PubMed]
- Kurzawski, G.; Suchy, J.; Kładny, J.; Grabowska, E.; Mierzejewski, M.; Jakubowska, A.; Debniak, T.; Cybulski, C.; Kowalska, E.; Szych, Z.; et al. The NOD2 3020insC mutation and the risk of colorectal cancer. Cancer Res. 2004, 64, 1604–1606. [Google Scholar] [CrossRef] [PubMed]
- Philpott, D.J.; Sorbara, M.T.; Robertson, S.J.; Croitoru, K.; Girardin, S.E. NOD proteins: Regulators of inflammation in health and disease. Nat. Rev. Immunol. 2014, 14, 9–23. [Google Scholar] [CrossRef] [PubMed]
- Zhan, Y.; Seregin, S.S.; Chen, J.; Chen, G.Y. Nod1 Limits Colitis-Associated Tumorigenesis by Regulating IFN-γ Production. J. Immunol. 2016, 196, 5121–5129. [Google Scholar] [CrossRef] [PubMed]
- Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [PubMed]
- Dominguez-Bello, M.G.; Blaser, M.J.; Ley, R.E.; Knight, R. Development of the human gastrointestinal microbiota and insights from high-throughput sequencing. Gastroenterology 2011, 140, 1713–1719. [Google Scholar] [CrossRef] [PubMed]
- Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Reddy, D.N. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; O’Riordan, M.X.D. Regulation of Bacterial Pathogenesis by Intestinal Short-Chain Fatty Acids. Adv. Appl. Microbiol. 2013, 85, 93–118. [Google Scholar] [PubMed]
- Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2017. [Google Scholar] [CrossRef] [PubMed]
- Woting, A.; Blaut, M. The Intestinal Microbiota in Metabolic Disease. Nutrients 2016, 8, 202. [Google Scholar] [CrossRef] [PubMed]
- Vernocchi, P.; Del Chierico, F.; Putignani, L. Gut Microbiota Profiling: Metabolomics Based Approach to Unravel Compounds Affecting Human Health. Front. Microbiol. 2016. [Google Scholar] [CrossRef] [PubMed]
- Fukiya, S.; Arata, M.; Kawashima, H.; Yoshida, D.; Kaneko, M.; Minamida, K.; Watanabe, J.; Ogura, Y.; Uchida, K.; Itoh, K.; et al. Conversion of cholic acid and chenodeoxycholic acid into their 7-oxo derivatives by Bacteroides intestinalis AM-1 isolated from human feces. FEMS Microbiol. Lett. 2009, 293, 263–270. [Google Scholar] [CrossRef] [PubMed]
- Ajouz, H.; Mukherji, D.; Shamseddine, A. Secondary bile acids: An underrecognized cause of colon cancer. World J. Surg. Oncol. 2014, 12, 164. [Google Scholar] [CrossRef] [PubMed]
- Neis, E.P.; Dejong, C.H.; Rensen, S.S. The Role of Microbial Amino Acid Metabolism in Host Metabolism. Nutrients 2015, 7, 2930–2946. [Google Scholar] [CrossRef] [PubMed]
- Hill, M.J. Intestinal flora and endogenous vitamin synthesis. Eur. J. Cancer Prev. 1997, 6, S43–S45. [Google Scholar] [CrossRef] [PubMed]
- Frick, P.G.; Riedler, G.; Brögli, H. Dose response and minimal daily requirement for vitamin K in man. J. Appl. Physiol. 1967, 23, 387–389. [Google Scholar] [PubMed]
- Gustafsson, B.E.; Daft, F.S.; McDaniel, E.G.; Smith, J.C.; Fitzgerald, R.J. Effects of vitamin K-active compounds and intestinal microorganisms in vitamin K-deficient germfree rats. J. Nutr. 1962, 78, 461–468. [Google Scholar] [PubMed]
- Johansson, M.E.V.; Phillipson, M.; Petersson, J.; Velcich, A.; Holm, L.; Hansson, G.C. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc. Natl. Acad. Sci. USA 2008, 105, 15064–15069. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.-B.; Wang, P.-Y.; Wang, X.; Wan, Y.-L.; Liu, Y.-C. Butyrate Enhances Intestinal Epithelial Barrier Function via Up-Regulation of Tight Junction Protein Claudin-1 Transcription. Dig. Dis. Sci. 2012, 57, 3126–3135. [Google Scholar] [CrossRef] [PubMed]
- Gaudier, E.; Jarry, A.; Blottière, H.M.; de Coppet, P.; Buisine, M.P.; Aubert, J.P.; Laboisse, C.; Cherbut, C.; Hoebler, C. Butyrate specifically modulates MUC gene expression in intestinal epithelial goblet cells deprived of glucose. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 287, G1168–G1174. [Google Scholar] [CrossRef] [PubMed]
- Hernández-Chirlaque, C.; Aranda, C.J.; Ocón, B.; Capitán-Cañadas, F.; Ortega-González, M.; Carrero, J.J.; Suárez, M.D.; Zarzuelo, A.; Sánchez de Medina, F.; Martínez-Augustin, O. Germ-free and Antibiotic-treated Mice are Highly Susceptible to Epithelial Injury in DSS Colitis. J. Crohns. Colitis 2016, 10, 1324–1335. [Google Scholar] [CrossRef] [PubMed]
- Bain, C.C.; Mowat, A.M. Macrophages in intestinal homeostasis and inflammation. Immunol. Rev. 2014, 260, 102–117. [Google Scholar] [CrossRef] [PubMed]
- Zindl, C.L.; Lai, J.-F.; Lee, Y.K.; Maynard, C.L.; Harbour, S.N.; Ouyang, W.; Chaplin, D.D.; Weaver, C.T. IL-22-producing neutrophils contribute to antimicrobial defense and restitution of colonic epithelial integrity during colitis. Proc. Natl. Acad. Sci. USA 2013, 110, 12768–12773. [Google Scholar] [CrossRef] [PubMed]
- Chu, V.T.; Beller, A.; Rausch, S.; Strandmark, J.; Zänker, M.; Arbach, O.; Kruglov, A.; Berek, C. Eosinophils promote generation and maintenance of immunoglobulin-A-expressing plasma cells and contribute to gut immune homeostasis. Immunity 2014, 40, 582–593. [Google Scholar] [CrossRef] [PubMed]
- Sonnenberg, G.F.; Artis, D. Innate lymphoid cells in the initiation, regulation and resolution of inflammation. Nat. Med. 2015, 21, 698–708. [Google Scholar] [CrossRef] [PubMed]
- Mortha, A.; Chudnovskiy, A.; Hashimoto, D.; Bogunovic, M.; Spencer, S.P.; Belkaid, Y.; Merad, M. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 2014, 343, 1249288. [Google Scholar] [CrossRef] [PubMed]
- Hepworth, M.R.; Monticelli, L.A.; Fung, T.C.; Ziegler, C.G.K.; Grunberg, S.; Sinha, R.; Mantegazza, A.R.; Ma, H.-L.; Crawford, A.; Angelosanto, J.M.; et al. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature 2013, 498, 113–117. [Google Scholar] [CrossRef] [PubMed]
- Bekiaris, V.; Persson, E.K.; Agace, W.W. Intestinal dendritic cells in the regulation of mucosal immunity. Immunol. Rev. 2014, 260, 86–101. [Google Scholar] [CrossRef] [PubMed]
- Slack, E.; Balmer, M.L.; Macpherson, A.J. B cells as a critical node in the microbiota-host immune system network. Immunol. Rev. 2014, 260, 50–66. [Google Scholar] [CrossRef] [PubMed]
- Yamanaka, T.; Helgeland, L.; Farstad, I.N.; Fukushima, H.; Midtvedt, T.; Brandtzaeg, P. Microbial colonization drives lymphocyte accumulation and differentiation in the follicle-associated epithelium of Peyer’s patches. J. Immunol. 2003, 170, 816–822. [Google Scholar] [CrossRef] [PubMed]
- Macpherson, A.J.; Harris, N.L. Interactions between commensal intestinal bacteria and the immune system. Nat. Rev. Immunol. 2004, 4, 478–485. [Google Scholar] [CrossRef] [PubMed]
- Ivanov, I.I.; Atarashi, K.; Manel, N.; Brodie, E.L.; Shima, T.; Karaoz, U.; Wei, D.; Goldfarb, K.C.; Santee, C.A.; Lynch, S.V.; et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009, 139, 485–498. [Google Scholar] [CrossRef] [PubMed]
- Cao, A.T.; Yao, S.; Gong, B.; Elson, C.O.; Cong, Y. Th17 cells upregulate polymeric Ig receptor and intestinal IgA and contribute to intestinal homeostasis. J. Immunol. 2012, 189, 4666–4673. [Google Scholar] [CrossRef] [PubMed]
- Cerf-Bensussan, N.; Gaboriau-Routhiau, V. The immune system and the gut microbiota: Friends or foes? Nat. Rev. Immunol. 2010, 10, 735–744. [Google Scholar] [CrossRef] [PubMed]
- Schnupf, P.; Gaboriau-Routhiau, V.; Gros, M.; Friedman, R.; Moya-Nilges, M.; Nigro, G.; Cerf-Bensussan, N.; Sansonetti, P.J. Growth and host interaction of mouse segmented filamentous bacteria in vitro. Nature 2015, 520, 99–103. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Macpherson, A.J.; Smith, K. Mesenteric lymph nodes at the center of immune anatomy. J. Exp. Med. 2006, 203, 497–500. [Google Scholar] [CrossRef] [PubMed]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [PubMed]
- Burgess, S.L.; Buonomo, E.; Carey, M.; Cowardin, C.; Naylor, C.; Noor, Z.; Wills-Karp, M.; Petri, W.A. Bone marrow dendritic cells from mice with an altered microbiota provide interleukin 17A-dependent protection against Entamoeba histolytica colitis. MBio 2014. [Google Scholar] [CrossRef] [PubMed]
- Bammann, L.L.; Clark, W.B.; Gibbons, R.J. Impaired colonization of gnotobiotic and conventional rats by streptomycin-resistant strains of Streptococcus mutans. Infect. Immun. 1978, 22, 721–726. [Google Scholar] [PubMed]
- Rupnik, M.; Wilcox, M.H.; Gerding, D.N. Clostridium difficile infection: New developments in epidemiology and pathogenesis. Nat. Rev. Microbiol. 2009, 7, 526–536. [Google Scholar] [CrossRef] [PubMed]
- Boleij, A.; Tjalsma, H. Gut bacteria in health and disease: A survey on the interface between intestinal microbiology and colorectal cancer. Biol. Rev. Camb. Philos. Soc. 2012, 87, 701–730. [Google Scholar] [CrossRef] [PubMed]
- Schamberger, G.P.; Diez-Gonzalez, F. Selection of recently isolated colicinogenic Escherichia coli strains inhibitory to Escherichia coli O157:H7. J. Food Prot. 2002, 65, 1381–1387. [Google Scholar] [CrossRef] [PubMed]
- Fukuda, S.; Toh, H.; Hase, K.; Oshima, K.; Nakanishi, Y.; Yoshimura, K.; Tobe, T.; Clarke, J.M.; Topping, D.L.; Suzuki, T.; et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 2011, 469, 543–547. [Google Scholar] [CrossRef] [PubMed]
- Gantois, I.; Ducatelle, R.; Pasmans, F.; Haesebrouck, F.; Hautefort, I.; Thompson, A.; Hinton, J.C.; Van Immerseel, F. Butyrate Specifically Down-Regulates Salmonella Pathogenicity Island 1 Gene Expression. Appl. Environ. Microbiol. 2006, 72, 946–949. [Google Scholar] [CrossRef] [PubMed]
- Momose, Y.; Hirayama, K.; Itoh, K. Competition for proline between indigenous Escherichia coli and E. coli O157:H7 in gnotobiotic mice associated with infant intestinal microbiota and its contribution to the colonization resistance against E. coli O157:H7. Antonie Van Leeuwenhoek 2008, 94, 165–171. [Google Scholar] [CrossRef] [PubMed]
- Marteyn, B.; West, N.; Browning, D.; Cole, J.; Shaw, J.; Palm, F.; Mounier, J.; Prévost, M.-C.; Sansonetti, P.; Tang, C. Modulation of Shigella virulence in response to available oxygen in vivo. Nature 2010, 465, 355–358. [Google Scholar] [CrossRef] [PubMed]
- Takeuchi, O.; Akira, S. Pattern recognition receptors and inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [PubMed]
- Salzman, N.H.; Underwood, M.A.; Bevins, C.L. Paneth cells, defensins, and the commensal microbiota: A hypothesis on intimate interplay at the intestinal mucosa. Semin. Immunol. 2007, 19, 70–83. [Google Scholar] [CrossRef] [PubMed]
- Vaishnava, S.; Behrendt, C.L.; Ismail, A.S.; Eckmann, L.; Hooper, L.V. Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface. Proc. Natl. Acad. Sci. USA 2008, 105, 20858–20863. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, K.S.; Chamaillard, M.; Ogura, Y.; Henegariu, O.; Inohara, N.; Nuñez, G.; Flavell, R.A. Nod2-Dependent Regulation of Innate and Adaptive Immunity in the Intestinal Tract. Science 2005, 307, 731–734. [Google Scholar] [CrossRef] [PubMed]
- Frantz, A.L.; Rogier, E.W.; Weber, C.R.; Shen, L.; Cohen, D.A.; Fenton, L.A.; Bruno, M.E. C.; Kaetzel, C.S. Targeted deletion of MyD88 in intestinal epithelial cells results in compromised antibacterial immunity associated with downregulation of polymeric immunoglobulin receptor, mucin-2, and antibacterial peptides. Mucosal Immunol. 2012, 5, 501–512. [Google Scholar] [CrossRef] [PubMed]
- Cash, H.L.; Whitham, C.V.; Behrendt, C.L.; Hooper, L.V. Symbiotic Bacteria Direct Expression of an Intestinal Bactericidal Lectin. Science 2006, 313, 1126–1130. [Google Scholar] [CrossRef] [PubMed]
- Giel, J.L.; Sorg, J.A.; Sonenshein, A.L.; Zhu, J. Metabolism of Bile Salts in Mice Influences Spore Germination in Clostridium difficile. PLoS ONE 2010. [Google Scholar] [CrossRef] [PubMed]
- Plummer, M.; de Martel, C.; Vignat, J.; Ferlay, J.; Bray, F.; Franceschi, S. Global burden of cancers attributable to infections in 2012: A synthetic analysis. Lancet Glob. Health 2016, 4, e609–e616. [Google Scholar] [CrossRef]
- Schottenfeld, D.; Beebe-Dimmer, J. The cancer burden attributable to biologic agents. Ann. Epidemiol. 2015, 25, 183–187. [Google Scholar] [CrossRef] [PubMed]
- Moore, W.E.; Moore, L.H. Intestinal floras of populations that have a high risk of colon cancer. Appl. Environ. Microbiol. 1995, 61, 3202–3207. [Google Scholar] [PubMed]
- Wang, T.; Cai, G.; Qiu, Y.; Fei, N.; Zhang, M.; Pang, X.; Jia, W.; Cai, S.; Zhao, L. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 2012, 6, 320–329. [Google Scholar] [CrossRef] [PubMed]
- Sobhani, I.; Tap, J.; Roudot-Thoraval, F.; Roperch, J.P.; Letulle, S.; Langella, P.; Corthier, G.; Tran Van Nhieu, J.; Furet, J.P. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS ONE 2011. [Google Scholar] [CrossRef] [PubMed]
- Shen, X.J.; Rawls, J.F.; Randall, T.; Burcal, L.; Mpande, C.N.; Jenkins, N.; Jovov, B.; Abdo, Z.; Sandler, R.S.; Keku, T.O. Molecular characterization of mucosal adherent bacteria and associations with colorectal adenomas. Gut Microbes 2010, 1, 138–147. [Google Scholar] [CrossRef] [PubMed]
- McCoy, A.N.; Araújo-Pérez, F.; Azcárate-Peril, A.; Yeh, J.J.; Sandler, R.S.; Keku, T.O. Fusobacterium Is Associated with Colorectal Adenomas. PLoS ONE 2013. [Google Scholar] [CrossRef] [PubMed]
- Wu, N.; Yang, X.; Zhang, R.; Li, J.; Xiao, X.; Hu, Y.; Chen, Y.; Yang, F.; Lu, N.; Wang, Z.; et al. Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb. Ecol. 2013, 66, 462–470. [Google Scholar] [CrossRef] [PubMed]
- Viljoen, K.S.; Dakshinamurthy, A.; Goldberg, P.; Blackburn, J.M. Quantitative profiling of colorectal cancer-associated bacteria reveals associations between fusobacterium spp., enterotoxigenic Bacteroides fragilis (ETBF) and clinicopathological features of colorectal cancer. PLoS ONE 2015. [Google Scholar] [CrossRef] [PubMed]
- Zackular, J.P.; Baxter, N.T.; Iverson, K.D.; Sadler, W.D.; Petrosino, J.F.; Chen, G.Y.; Schloss, P.D. The gut microbiome modulates colon tumorigenesis. MBio 2013. [Google Scholar] [CrossRef] [PubMed]
- Morgan, X.C.; Tickle, T.L.; Sokol, H.; Gevers, D.; Devaney, K.L.; Ward, D.V.; Reyes, J.A.; Shah, S.A.; LeLeiko, N.; Snapper, S.B.; et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Q.; Jin, Z.; Wu, W.; Gao, R.; Guo, B.; Gao, Z.; Yang, Y.; Qin, H. Analysis of the Intestinal Lumen Microbiota in an Animal Model of Colorectal Cancer. PLoS ONE 2014. [Google Scholar] [CrossRef] [PubMed]
- Strofilas, A.; Lagoudianakis, E.E.; Seretis, C.; Pappas, A.; Koronakis, N.; Keramidaris, D.; Koukoutsis, I.; Chrysikos, I.; Manouras, I.; Manouras, A. Association of Helicobacter Pylori Infection and Colon Cancer. J. Clin. Med. Res. 2012, 4, 172–176. [Google Scholar] [CrossRef] [PubMed]
- Balamurugan, R.; Rajendiran, E.; George, S.; Samuel, G.V.; Ramakrishna, B.S. Real-time polymerase chain reaction quantification of specific butyrate-producing bacteria, Desulfovibrio and Enterococcus faecalis in the feces of patients with colorectal cancer. J. Gastroenterol. Hepatol. 2008, 23, 1298–1303. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; He, H.; Xu, H.; Li, Y.; Li, Z.; Du, Y.; He, J.; Zhou, Y.; Wang, H.; Nie, Y.; et al. Association of oncogenic bacteria with colorectal cancer in South China. Oncotarget 2016, 7, 80794–80802. [Google Scholar] [CrossRef] [PubMed]
- Amarnani, R.; Rapose, A. Colon cancer and enterococcus bacteremia co-affection: A dangerous alliance. J. Infect. Public Health 2017. [Google Scholar] [CrossRef] [PubMed]
- Balish, E.; Warner, T. Enterococcus faecalis induces inflammatory bowel disease in interleukin-10 knockout mice. Am. J. Pathol. 2002, 160, 2253–2257. [Google Scholar] [CrossRef]
- Ruiz, P.A.; Shkoda, A.; Kim, S.C.; Sartor, R.B.; Haller, D. IL-10 gene-deficient mice lack TGF-beta/Smad signaling and fail to inhibit proinflammatory gene expression in intestinal epithelial cells after the colonization with colitogenic Enterococcus faecalis. J. Immunol. 2005, 174, 2990–2999. [Google Scholar] [CrossRef] [PubMed]
- Huycke, M.M.; Joyce, W.; Wack, M.F. Augmented production of extracellular superoxide by blood isolates of Enterococcus faecalis. J. Infect. Dis. 1996, 173, 743–746. [Google Scholar] [CrossRef] [PubMed]
- Huycke, M.M.; Abrams, V.; Moore, D.R. Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis 2002, 23, 529–536. [Google Scholar] [CrossRef] [PubMed]
- Limoli, C.L.; Giedzinski, E. Induction of Chromosomal Instability by Chronic Oxidative Stress. Neoplasia 2003, 5, 339–346. [Google Scholar] [CrossRef]
- Wang, X.; Huycke, M.M. Extracellular superoxide production by Enterococcus faecalis promotes chromosomal instability in mammalian cells. Gastroenterology 2007, 132, 551–561. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Yang, Y.; Huycke, M.M. Commensal bacteria drive endogenous transformation and tumour stem cell marker expression through a bystander effect. Gut 2015, 64, 459–468. [Google Scholar] [CrossRef] [PubMed]
- Zhang, G.; Svenungsson, B.; Kärnell, A.; Weintraub, A. Prevalence of enterotoxigenic Bacteroides fragilis in adult patients with diarrhea and healthy controls. Clin. Infect. Dis. 1999, 29, 590–594. [Google Scholar] [CrossRef] [PubMed]
- Sears, C.L. The toxins of Bacteroides fragilis. Toxicon 2001, 39, 1737–1746. [Google Scholar] [CrossRef]
- Toprak, N.U.; Yagci, A.; Gulluoglu, B.M.; Akin, M.L.; Demirkalem, P.; Celenk, T.; Soyletir, G. A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer. Clin. Microbiol. Infect. 2006, 12, 782–786. [Google Scholar] [CrossRef] [PubMed]
- Boleij, A.; Hechenbleikner, E.M.; Goodwin, A.C.; Badani, R.; Stein, E.M.; Lazarev, M.G.; Ellis, B.; Carroll, K.C.; Albesiano, E.; Wick, E.C.; et al. The Bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients. Clin. Infect. Dis. 2015, 60, 208–215. [Google Scholar] [CrossRef] [PubMed]
- Purcell, R.V.; Pearson, J.; Aitchison, A.; Dixon, L.; Frizelle, F.A.; Keenan, J.I. Colonization with enterotoxigenic Bacteroides fragilis is associated with early-stage colorectal neoplasia. PLoS ONE 2017. [Google Scholar] [CrossRef] [PubMed]
- Obiso, R.J.; Azghani, A.O.; Wilkins, T.D. The Bacteroides fragilis toxin fragilysin disrupts the paracellular barrier of epithelial cells. Infect. Immun. 1997, 65, 1431–1439. [Google Scholar] [PubMed]
- Wells, C.; van de Westerlo, E.; Jechorek, R.; Feltis, B.; Wilkins, T.; Erlandsen, S. Bacteroides fragilis enterotoxin modulates epithelial permeability and bacterial internalization by HT-29 enterocytes. Gastroenterology 1996, 110, 1429–1437. [Google Scholar] [CrossRef] [PubMed]
- Riegler, M.; Lotz, M.; Sears, C.; Pothoulakis, C.; Castagliuolo, I.; Wang, C.C.; Sedivy, R.; Sogukoglu, T.; Cosentini, E.; Bischof, G.; et al. Bacteroides fragilis toxin 2 damages human colonic mucosa in vitro. Gut 1999, 44, 504–510. [Google Scholar] [CrossRef] [PubMed]
- Wu, S.; Morin, P.J.; Maouyo, D.; Sears, C.L. Bacteroides fragilis enterotoxin induces c-Myc expression and cellular proliferation. Gastroenterology 2003, 124, 392–400. [Google Scholar] [CrossRef] [PubMed]
- Wu, S.; Rhee, K.-J.; Albesiano, E.; Rabizadeh, S.; Wu, X.; Yen, H.-R.; Huso, D.L.; Brancati, F.L.; Wick, E.; McAllister, F.; et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat. Med. 2009, 15, 1016–1022. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.W. Fusobacterium nucleatum: A commensal-turned pathogen. Curr. Opin. Microbiol. 2015, 23, 141–147. [Google Scholar] [CrossRef] [PubMed]
- Castellarin, M.; Warren, R.L.; Freeman, J.D.; Dreolini, L.; Krzywinski, M.; Strauss, J.; Barnes, R.; Watson, P.; Allen-Vercoe, E.; Moore, R.A.; et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012, 22, 299–306. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.-Y.; Ge, Q.-X.; Cao, J.; Zhou, Y.-J.; Du, Y.-L.; Shen, B.; Wan, Y.-J.Y.; Nie, Y.-Q. Association of Fusobacterium nucleatum infection with colorectal cancer in Chinese patients. World J. Gastroenterol. 2016, 22, 3227–3233. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Weng, W.; Peng, J.; Hong, L.; Yang, L.; Toiyama, Y.; Gao, R.; Liu, M.; Yin, M.; Pan, C.; et al. Fusobacterium nucleatum Increases Proliferation of Colorectal Cancer Cells and Tumor Development in Mice by Activating Toll-Like Receptor 4 Signaling to Nuclear Factor-κB, and Up-regulating Expression of MicroRNA-21. Gastroenterology 2017, 152, 851–866. [Google Scholar] [CrossRef] [PubMed]
- Medina, P.P.; Nolde, M.; Slack, F.J. OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature 2010, 467, 86–90. [Google Scholar] [CrossRef] [PubMed]
- Kostic, A.D.; Chun, E.; Robertson, L.; Glickman, J.N.; Gallini, C.A.; Michaud, M.; Clancy, T.E.; Chung, D.C.; Lochhead, P.; Hold, G.L.; et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor immune microenvironment. Cell Host Microbe 2013, 14, 207–215. [Google Scholar] [CrossRef] [PubMed]
- Rubinstein, M.R.; Wang, X.; Liu, W.; Hao, Y.; Cai, G.; Han, Y.W. Fusobacterium nucleatum Promotes Colorectal Carcinogenesis by Modulating E-Cadherin/β-Catenin Signaling via its FadA Adhesin. Cell Host Microbe 2013, 14, 195–206. [Google Scholar] [CrossRef] [PubMed]
- McCOY, W.C.; Mason, J.M. Enterococcal endocarditis associated with carcinoma of the sigmoid; report of a case. J. Med. Assoc. State Ala. 1951, 21, 162–166. [Google Scholar] [PubMed]
- Klein, R.S.; Recco, R.A.; Catalano, M.T.; Edberg, S.C.; Casey, J.I.; Steigbigel, N.H. Association of Streptococcus bovis with carcinoma of the colon. N. Engl. J. Med. 1977, 297, 800–802. [Google Scholar] [CrossRef] [PubMed]
- Boleij, A.; van Gelder, M.M.; Swinkels, D.W.; Tjalsma, H. Clinical Importance of Streptococcus gallolyticus infection among colorectal cancer patients: Systematic review and meta-analysis. Clin. Infect. Dis. 2011, 53, 870–878. [Google Scholar] [CrossRef] [PubMed]
- Corredoira-Sánchez, J.; García-Garrote, F.; Rabuñal, R.; López-Roses, L.; García-País, M.J.; Castro, E.; González-Soler, R.; Coira, A.; Pita, J.; López-Álvarez, M.J.; et al. Association Between Bacteremia Due to Streptococcus gallolyticus subsp. gallolyticus (Streptococcus bovis I) and Colorectal Neoplasia: A Case-Control Study. Clin. Infect. Dis. 2012, 55, 491–496. [Google Scholar]
- Biarc, J.; Nguyen, I.S.; Pini, A.; Gossé, F.; Richert, S.; Thiersé, D.; Van Dorsselaer, A.; Leize-Wagner, E.; Raul, F.; Klein, J.-P.; et al. Carcinogenic properties of proteins with pro-inflammatory activity from Streptococcus infantarius (formerly S.bovis). Carcinogenesis 2004, 25, 1477–1484. [Google Scholar] [CrossRef] [PubMed]
- Ellmerich, S.; Schöller, M.; Duranton, B.; Gossé, F.; Galluser, M.; Klein, J.P.; Raul, F. Promotion of intestinal carcinogenesis by Streptococcus bovis. Carcinogenesis 2000, 21, 753–756. [Google Scholar] [CrossRef] [PubMed]
- Abdulamir, A.S.; Hafidh, R.R.; Bakar, F.A. Molecular detection, quantification, and isolation of Streptococcus gallolyticus bacteria colonizing colorectal tumors: Inflammation-driven potential of carcinogenesis via IL-1, COX-2, and IL-8. Mol. Cancer 2010, 9, 249. [Google Scholar] [CrossRef] [PubMed]
- Ballard, J.; Bryant, A.; Stevens, D.; Tweten, R.K. Purification and characterization of the lethal toxin (alpha-toxin) of Clostridium septicum. Infect. Immun. 1992, 60, 784–790. [Google Scholar] [PubMed]
- Kennedy, C.L.; Krejany, E.O.; Young, L.F.; O’Connor, J.R.; Awad, M.M.; Boyd, R.L.; Emmins, J.J.; Lyras, D.; Rood, J.I. The alpha-toxin of Clostridium septicum is essential for virulence. Mol. Microbiol. 2005, 57, 1357–1366. [Google Scholar] [CrossRef] [PubMed]
- Chew, S.S.; Lubowski, D.Z. Clostridium septicum and malignancy. ANZ J. Surg. 2001, 71, 647–649. [Google Scholar] [CrossRef] [PubMed]
- Corredoira, J.; Grau, I.; Garcia-Rodriguez, J.F.; García-País, M.J.; Rabuñal, R.; Ardanuy, C.; García-Garrote, F.; Coira, A.; Alonso, M.P.; Boleij, A.; et al. Colorectal neoplasm in cases of Clostridium septicum and Streptococcus gallolyticus subsp. gallolyticus bacteraemia. Eur. J. Intern. Med. 2017, 41, 68–73. [Google Scholar] [CrossRef] [PubMed]
- Mirza, N.N.; McCloud, J.M.; Cheetham, M.J. Clostridium septicum sepsis and colorectal cancer—A reminder. World J. Surg. Oncol. 2009, 7, 73. [Google Scholar] [CrossRef] [PubMed]
- Chakravorty, A.; Awad, M.M.; Cheung, J.K.; Hiscox, T.J.; Lyras, D.; Rood, J.I. The Pore-Forming α-Toxin from Clostridium septicum Activates the MAPK Pathway in a Ras-c-Raf-Dependent and Independent Manner. Toxins 2015, 7, 516–534. [Google Scholar] [CrossRef] [PubMed]
- Suerbaum, S.; Michetti, P. Helicobacter pylori infection. N. Engl. J. Med. 2002, 347, 1175–1186. [Google Scholar] [CrossRef] [PubMed]
- Testerman, T.L.; Morris, J. Beyond the stomach: An updated view of Helicobacter pylori pathogenesis, diagnosis, and treatment. World J. Gastroenterol. 2014, 20, 12781–12808. [Google Scholar] [CrossRef] [PubMed]
- Meucci, G.; Tatarella, M.; Vecchi, M.; Ranzi, M.L.; Biguzzi, E.; Beccari, G.; Clerici, E.; de Franchis, R. High prevalence of Helicobacter pylori infection in patients with colonic adenomas and carcinomas. J. Clin. Gastroenterol. 1997, 25, 605–607. [Google Scholar] [CrossRef] [PubMed]
- Fujimori, S.; Kishida, T.; Kobayashi, T.; Sekita, Y.; Seo, T.; Nagata, K.; Tatsuguchi, A.; Gudis, K.; Yokoi, K.; Tanaka, N.; et al. Helicobacter pylori infection increases the risk of colorectal adenoma and adenocarcinoma, especially in women. J. Gastroenterol. 2005, 40, 887–893. [Google Scholar] [CrossRef] [PubMed]
- Hong, S.N.; Lee, S.M.; Kim, J.H.; Lee, T.Y.; Kim, J.H.; Choe, W.H.; Lee, S.-Y.; Cheon, Y.K.; Sung, I.K.; Park, H.S.; et al. Helicobacter pylori infection increases the risk of colorectal adenomas: Cross-sectional study and meta-analysis. Dig. Dis. Sci. 2012, 57, 2184–2194. [Google Scholar] [CrossRef] [PubMed]
- Inoue, I.; Mukoubayashi, C.; Yoshimura, N.; Niwa, T.; Deguchi, H.; Watanabe, M.; Enomoto, S.; Maekita, T.; Ueda, K.; Iguchi, M.; et al. Elevated risk of colorectal adenoma with Helicobacter pylori-related chronic gastritis: A population-based case-control study. Int. J. Cancer 2011, 129, 2704–2711. [Google Scholar] [CrossRef] [PubMed]
- Siddheshwar, R.K.; Muhammad, K.B.; Gray, J.C.; Kelly, S.B. Seroprevalence of helicobacter pylori in patients with colorectal polyps and colorectal carcinoma. Am. J. Gastroenterol. 2001, 96, 84–88. [Google Scholar] [CrossRef] [PubMed]
- Bae, R.C.; Jeon, S.W.; Cho, H.J.; Jung, M.K.; Kweon, Y.O.; Kim, S.K. Gastric dysplasia may be an independent risk factor of an advanced colorectal neoplasm. World J. Gastroenterol. 2009, 15, 5722–5726. [Google Scholar] [CrossRef] [PubMed]
- Moss, S.F.; Neugut, A.I.; Garbowski, G.C.; Wang, S.; Treat, M.R.; Forde, K.A. Helicobacter pylori seroprevalence and colorectal neoplasia: Evidence against an association. J. Natl. Cancer Inst. 1995, 87, 762–763. [Google Scholar] [CrossRef] [PubMed]
- Kim, T.J.; Kim, E.R.; Chang, D.K.; Kim, Y.-H.; Baek, S.-Y.; Kim, K.; Hong, S.N. Helicobacter pylori infection is an independent risk factor of early and advanced colorectal neoplasm. Helicobacter 2017. [Google Scholar] [CrossRef] [PubMed]
- Nam, J.H.; Hong, C.W.; Kim, B.C.; Shin, A.; Ryu, K.H.; Park, B.J.; Kim, B.; Sohn, D.K.; Han, K.S.; Kim, J.; et al. Helicobacter pylori infection is an independent risk factor for colonic adenomatous neoplasms. Cancer Causes Control. 2017, 28, 107–115. [Google Scholar] [CrossRef] [PubMed]
- Yan, Y.; Chen, Y.-N.; Zhao, Q.; Chen, C.; Lin, C.-J.; Jin, Y.; Pan, S.; Wu, J.-S. Helicobacter pylori infection with intestinal metaplasia: An independent risk factor for colorectal adenomas. World J. Gastroenterol. 2017, 23, 1443–1449. [Google Scholar] [CrossRef] [PubMed]
- Hartwich, A.; Konturek, S.; Pierzchalski, P.; Zuchowicz, M.; Labza, H.; Konturek, P.; Karczewska, E.; Bielanski, W.; Marlicz, K.; Starzynska, T.; et al. Helicobacter pylori infection, gastrin, cyclooxygenase-2, and apoptosis in colorectal cancer. Int. J. Colorectal Dis. 2001, 16, 202–210. [Google Scholar] [CrossRef] [PubMed]
- Koh, T.J.; Dockray, G.J.; Varro, A.; Cahill, R.J.; Dangler, C.A.; Fox, J.G.; Wang, T.C. Overexpression of glycine-extended gastrin in transgenic mice results in increased colonic proliferation. J. Clin. Investig. 1999, 103, 1119–1126. [Google Scholar] [CrossRef] [PubMed]
- Tatishchev, S.F.; VanBeek, C.; Wang, H.L. Helicobacter pylori infection and colorectal carcinoma: Is there a causal association? J. Gastrointest. Oncol. 2012, 3, 380–385. [Google Scholar] [PubMed]
- Handa, O.; Naito, Y.; Yoshikawa, T. Helicobacter pylori: A ROS-inducing bacterial species in the stomach. Inflamm. Res. 2010, 59, 997–1003. [Google Scholar] [CrossRef] [PubMed]
- Shmuely, H.; Passaro, D.; Figer, A.; Niv, Y.; Pitlik, S.; Samra, Z.; Koren, R.; Yahav, J. Relationship between Helicobacter pylori CagA status and colorectal cancer. Am. J. Gastroenterol. 2001, 96, 3406–3410. [Google Scholar] [CrossRef] [PubMed]
- Cover, T.L.; Blanke, S.R. Helicobacter pylori VacA, a paradigm for toxin multifunctionality. Nat. Rev. Microbiol. 2005, 3, 320–332. [Google Scholar] [CrossRef] [PubMed]
- Evans, D.J.; Evans, D.G.; Takemura, T.; Nakano, H.; Lampert, H.C.; Graham, D.Y.; Granger, D.N.; Kvietys, P.R. Characterization of a Helicobacter pylori neutrophil-activating protein. Infect. Immun. 1995, 63, 2213–2220. [Google Scholar] [PubMed]
- Wessler, S.; Krisch, L.M.; Elmer, D.P.; Aberger, F. From inflammation to gastric cancer—The importance of Hedgehog/GLI signaling in Helicobacte r pylori-induced chronic inflammatory and neoplastic diseases. Cell Commun. Signal. 2017. [Google Scholar] [CrossRef] [PubMed]
- Sousa, C.P. The versatile strategies of Escherichia coli pathotypes: A mini review. J. Venom. Anim. Toxins Trop. Dis. 2006, 12, 363–373. [Google Scholar] [CrossRef]
- Escobar-Páramo, P.; Grenet, K.; Le Menac’h, A.; Rode, L.; Salgado, E.; Amorin, C.; Gouriou, S.; Picard, B.; Rahimy, M.C.; Andremont, A.; et al. Large-scale population structure of human commensal Escherichia coli isolates. Appl. Environ. Microbiol. 2004, 70, 5698–5700. [Google Scholar] [CrossRef] [PubMed]
- Darfeuille-Michaud, A.; Neut, C.; Barnich, N.; Lederman, E.; Di Martino, P.; Desreumaux, P.; Gambiez, L.; Joly, B.; Cortot, A.; Colombel, J.F. Presence of adherent Escherichia coli strains in ileal mucosa of patients with Crohn’s disease. Gastroenterology 1998, 115, 1405–1413. [Google Scholar] [CrossRef]
- Darfeuille-Michaud, A.; Boudeau, J.; Bulois, P.; Neut, C.; Glasser, A.-L.; Barnich, N.; Bringer, M.-A.; Swidsinski, A.; Beaugerie, L.; Colombel, J.-F. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology 2004, 127, 412–421. [Google Scholar] [CrossRef] [PubMed]
- Swidsinski, A.; Khilkin, M.; Kerjaschki, D.; Schreiber, S.; Ortner, M.; Weber, J.; Lochs, H. Association between intraepithelial Escherichia coli and colorectal cancer. Gastroenterology 1998, 115, 281–286. [Google Scholar] [CrossRef]
- Martin, H.M.; Campbell, B.J.; Hart, C.A.; Mpofu, C.; Nayar, M.; Singh, R.; Englyst, H.; Williams, H.F.; Rhodes, J.M. Enhanced Escherichia coli adherence and invasion in Crohn’s disease and colon cancer. Gastroenterology 2004, 127, 80–93. [Google Scholar] [CrossRef] [PubMed]
- Maddocks, O.D.K.; Short, A.J.; Donnenberg, M.S.; Bader, S.; Harrison, D.J. Attaching and effacing Escherichia coli downregulate DNA mismatch repair protein in vitro and are associated with colorectal adenocarcinomas in humans. PLoS ONE 2009. [Google Scholar] [CrossRef] [PubMed]
- Buc, E.; Dubois, D.; Sauvanet, P.; Raisch, J.; Delmas, J.; Darfeuille-Michaud, A.; Pezet, D.; Bonnet, R. High Prevalence of Mucosa-Associated E. coli Producing Cyclomodulin and Genotoxin in Colon Cancer. PLoS ONE 2013. [Google Scholar] [CrossRef] [PubMed]
- Prorok-Hamon, M.; Friswell, M.K.; Alswied, A.; Roberts, C.L.; Song, F.; Flanagan, P.K.; Knight, P.; Codling, C.; Marchesi, J.R.; Winstanley, C.; et al. Colonic mucosa-associated diffusely adherent afaC+ Escherichia coli expressing lpfA and pks are increased in inflammatory bowel disease and colon cancer. Gut 2014, 63, 761–770. [Google Scholar] [CrossRef] [PubMed]
- Bonnet, M.; Buc, E.; Sauvanet, P.; Darcha, C.; Dubois, D.; Pereira, B.; Déchelotte, P.; Bonnet, R.; Pezet, D.; Darfeuille-Michaud, A. Colonization of the human gut by E. coli and colorectal cancer risk. Clin. Cancer Res. 2014, 20, 859–867. [Google Scholar] [CrossRef] [PubMed]
- Raisch, J.; Buc, E.; Bonnet, M.; Sauvanet, P.; Vazeille, E.; de Vallée, A.; Déchelotte, P.; Darcha, C.; Pezet, D.; Bonnet, R.; et al. Colon cancer-associated B2 Escherichia coli colonize gut mucosa and promote cell proliferation. World J. Gastroenterol. 2014, 20, 6560–6572. [Google Scholar] [CrossRef] [PubMed]
- Marchès, O.; Ledger, T.N.; Boury, M.; Ohara, M.; Tu, X.; Goffaux, F.; Mainil, J.; Rosenshine, I.; Sugai, M.; De Rycke, J.; et al. Enteropathogenic and enterohaemorrhagic Escherichia coli deliver a novel effector called Cif, which blocks cell cycle G2/M transition. Mol. Microbiol. 2003, 50, 1553–1567. [Google Scholar] [CrossRef] [PubMed]
- Flatau, G.; Lemichez, E.; Gauthier, M.; Chardin, P.; Paris, S.; Fiorentini, C.; Boquet, P. Toxin-induced activation of the G protein p21 Rho by deamidation of glutamine. Nature 1997, 387, 729–733. [Google Scholar] [PubMed]
- Taieb, F.; Petit, C.; Nougayrède, J.-P.; Oswald, E. The Enterobacterial Genotoxins: Cytolethal Distending Toxin and Colibactin. EcoSal Plus 2016. [Google Scholar] [CrossRef]
- Nougayrède, J.-P.; Homburg, S.; Taieb, F.; Boury, M.; Brzuszkiewicz, E.; Gottschalk, G.; Buchrieser, C.; Hacker, J.; Dobrindt, U.; Oswald, E. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science 2006, 313, 848–851. [Google Scholar] [CrossRef] [PubMed]
- Cuevas-Ramos, G.; Petit, C.R.; Marcq, I.; Boury, M.; Oswald, E.; Nougayrède, J.-P. Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc. Natl. Acad. Sci. USA 2010, 107, 11537–11542. [Google Scholar] [CrossRef] [PubMed]
- Vizcaino, M.I.; Crawford, J.M. The colibactin warhead crosslinks DNA. Nat. Chem. 2015, 7, 411–417. [Google Scholar] [CrossRef] [PubMed]
- Secher, T.; Samba-Louaka, A.; Oswald, E.; Nougayrède, J.-P. Escherichia coli producing colibactin triggers premature and transmissible senescence in mammalian cells. PLoS ONE 2013. [Google Scholar] [CrossRef] [PubMed]
- Cougnoux, A.; Dalmasso, G.; Martinez, R.; Buc, E.; Delmas, J.; Gibold, L.; Sauvanet, P.; Darcha, C.; Déchelotte, P.; Bonnet, M.; et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut 2014, 63, 1932–1942. [Google Scholar] [CrossRef] [PubMed]
- Raisch, J.; Rolhion, N.; Dubois, A.; Darfeuille-Michaud, A.; Bringer, M.-A. Intracellular colon cancer-associated Escherichia coli promote protumoral activities of human macrophages by inducing sustained COX-2 expression. Lab. Investig. 2015, 95, 296–307. [Google Scholar] [CrossRef] [PubMed]
- Gagnière, J.; Bonnin, V.; Jarrousse, A.-S.; Cardamone, E.; Agus, A.; Uhrhammer, N.; Sauvanet, P.; Déchelotte, P.; Barnich, N.; Bonnet, R.; et al. Interactions between microsatellite instability and human gut colonization by Escherichia coli in colorectal cancer. Clin. Sci. 1979 2017, 131, 471–485. [Google Scholar] [CrossRef] [PubMed]
- Tian, Y.; Wang, K.; Ji, G. P112 Short-chain fatty acids administration is protective in colitis-associated colorectal cancer development. J. Crohns Colitis 2017. [Google Scholar] [CrossRef]
- Goodwin, A.C.; Destefano Shields, C.E.; Wu, S.; Huso, D.L.; Wu, X.; Murray-Stewart, T.R.; Hacker-Prietz, A.; Rabizadeh, S.; Woster, P.M.; Sears, C.L.; et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc. Natl. Acad. Sci. USA 2011, 108, 15354–15359. [Google Scholar] [CrossRef] [PubMed]
- Mauro, M.O.; Monreal, M.T.; Silva, M.T.; Pesarini, J.R.; Mantovani, M.S.; Ribeiro, L.R.; Dichi, J.B.; Carreira, C.M.; Oliveira, R.J. Evaluation of the antimutagenic and anticarcinogenic effects of inulin in vivo. Genet. Mol. Res. 2013, 12, 2281–2293. [Google Scholar] [CrossRef] [PubMed]
- Chen, Z.-Y.; Hsieh, Y.-M.; Huang, C.-C.; Tsai, C.-C. Inhibitory Effects of Probiotic Lactobacillus on the Growth of Human Colonic Carcinoma Cell Line HT-29. Molecules 2017, 22, 107. [Google Scholar] [CrossRef] [PubMed]
- Del Carmen, S.; de Moreno de LeBlanc, A.; Levit, R.; Azevedo, V.; Langella, P.; Bermúdez-Humarán, L.G.; LeBlanc, J.G. Anti-cancer effect of lactic acid bacteria expressing antioxidant enzymes or IL-10 in a colorectal cancer mouse model. Int. Immunopharmacol. 2017, 42, 122–129. [Google Scholar] [CrossRef] [PubMed]
- Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970. [Google Scholar] [CrossRef] [PubMed]
- West, N.R.; Powrie, F. Immunotherapy Not Working? Check Your Microbiota. Cancer Cell 2015, 28, 687–689. [Google Scholar] [CrossRef] [PubMed]
- Li, T.-T.; Ogino, S.; Qian, Z.R. Toll-like receptor signaling in colorectal cancer: Carcinogenesis to cancer therapy. World J. Gastroenterol. 2014, 20, 17699–17708. [Google Scholar] [PubMed]
- Fukata, M.; Shang, L.; Santaolalla, R.; Sotolongo, J.; Pastorini, C.; España, C.; Ungaro, R.; Harpaz, N.; Cooper, H.S.; Elson, G.; et al. Constitutive activation of epithelial TLR4 augments inflammatory responses to mucosal injury and drives colitis-associated tumorigenesis. Inflamm. Bowel Dis. 2011, 17, 1464–1473. [Google Scholar] [CrossRef] [PubMed]
© 2017 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Lucas, C.; Barnich, N.; Nguyen, H.T.T. Microbiota, Inflammation and Colorectal Cancer. Int. J. Mol. Sci. 2017, 18, 1310. https://doi.org/10.3390/ijms18061310
Lucas C, Barnich N, Nguyen HTT. Microbiota, Inflammation and Colorectal Cancer. International Journal of Molecular Sciences. 2017; 18(6):1310. https://doi.org/10.3390/ijms18061310
Chicago/Turabian StyleLucas, Cécily, Nicolas Barnich, and Hang Thi Thu Nguyen. 2017. "Microbiota, Inflammation and Colorectal Cancer" International Journal of Molecular Sciences 18, no. 6: 1310. https://doi.org/10.3390/ijms18061310
APA StyleLucas, C., Barnich, N., & Nguyen, H. T. T. (2017). Microbiota, Inflammation and Colorectal Cancer. International Journal of Molecular Sciences, 18(6), 1310. https://doi.org/10.3390/ijms18061310