Skip to main content
Top
Published in: Current Colorectal Cancer Reports 6/2017

01-12-2017 | Nutrition and Nutritional Interventions in Colorectal Cancer (K Wu, Section Editor)

Diet, Gut Microbiota, and Colorectal Cancer Prevention: a Review of Potential Mechanisms and Promising Targets for Future Research

Authors: Mingyang Song, Andrew T. Chan

Published in: Current Colorectal Cancer Reports | Issue 6/2017

Login to get access

Abstract

Diet plays an important role in the development of colorectal cancer. Emerging data have implicated the gut microbiota in colorectal cancer. Diet is a major determinant for the gut microbial structure and function. Therefore, it has been hypothesized that alterations in gut microbes and their metabolites may contribute to the influence of diet on the development of colorectal cancer. We review several major dietary factors that have been linked to gut microbiota and colorectal cancer, including major dietary patterns, fiber, red meat and sulfur, and obesity. Most of the epidemiologic evidence derives from cross-sectional or short-term, highly controlled feeding studies that are limited in size. Therefore, high-quality large-scale prospective studies with dietary data collected over the life course and comprehensive gut microbial composition and function assessed well prior to neoplastic occurrence are critically needed to identify microbiome-based interventions that may complement or optimize current diet-based strategies for colorectal cancer prevention and management.
Literature
1.
go back to reference Ferlay J, Soerjomataram I, Ervik M, et al. Cancer incidence and mortality worldwide: IARC CancerBase No. 11 [Internet]. GLOBOCAN 2012 v1.0. Lyon: International Agency for Research on Cancer; 2013. Ferlay J, Soerjomataram I, Ervik M, et al. Cancer incidence and mortality worldwide: IARC CancerBase No. 11 [Internet]. GLOBOCAN 2012 v1.0. Lyon: International Agency for Research on Cancer; 2013.
2.
go back to reference •• Song M, Garrett WS, Chan AT. Nutrients, foods, and colorectal cancer prevention. Gastroenterology. 2015;148:1244–1260 e16. A comprehensive review of epidemiologic and mechanistic evidence supporting the importance of nutritional factors in colorectal cancer prevention PubMedPubMedCentralCrossRef •• Song M, Garrett WS, Chan AT. Nutrients, foods, and colorectal cancer prevention. Gastroenterology. 2015;148:1244–1260 e16. A comprehensive review of epidemiologic and mechanistic evidence supporting the importance of nutritional factors in colorectal cancer prevention PubMedPubMedCentralCrossRef
3.
go back to reference Scanlan PD, Shanahan F, Clune Y, et al. Culture-independent analysis of the gut microbiota in colorectal cancer and polyposis. Environ Microbiol. 2008;10:789–98.PubMedCrossRef Scanlan PD, Shanahan F, Clune Y, et al. Culture-independent analysis of the gut microbiota in colorectal cancer and polyposis. Environ Microbiol. 2008;10:789–98.PubMedCrossRef
5.
go back to reference Wang T, Cai G, Qiu Y, et al. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 2012;6:320–9.PubMedCrossRef Wang T, Cai G, Qiu Y, et al. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 2012;6:320–9.PubMedCrossRef
7.
go back to reference Zackular JP, Rogers MA, Ruffin MT, et al. The human gut microbiome as a screening tool for colorectal cancer. Cancer Prev Res (Phila). 2014;7:1112–21.CrossRef Zackular JP, Rogers MA, Ruffin MT, et al. The human gut microbiome as a screening tool for colorectal cancer. Cancer Prev Res (Phila). 2014;7:1112–21.CrossRef
9.
go back to reference Feng Q, Liang S, Jia H, et al. Gut microbiome development along the colorectal adenoma-carcinoma sequence. Nat Commun. 2015;6:6528.PubMedCrossRef Feng Q, Liang S, Jia H, et al. Gut microbiome development along the colorectal adenoma-carcinoma sequence. Nat Commun. 2015;6:6528.PubMedCrossRef
10.
go back to reference Vogtmann E, Hua X, Zeller G, et al. Colorectal cancer and the human gut microbiome: reproducibility with whole-genome shotgun sequencing. PLoS One. 2016;11:e0155362.PubMedPubMedCentralCrossRef Vogtmann E, Hua X, Zeller G, et al. Colorectal cancer and the human gut microbiome: reproducibility with whole-genome shotgun sequencing. PLoS One. 2016;11:e0155362.PubMedPubMedCentralCrossRef
11.
go back to reference Yu J, Feng Q, Wong SH, et al. Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut. 2017;66:70–8.PubMedCrossRef Yu J, Feng Q, Wong SH, et al. Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut. 2017;66:70–8.PubMedCrossRef
12.
go back to reference Shah MS, DeSantis TZ, Weinmaier T, et al. Leveraging sequence-based faecal microbial community survey data to identify a composite biomarker for colorectal cancer. Gut. 2017. Shah MS, DeSantis TZ, Weinmaier T, et al. Leveraging sequence-based faecal microbial community survey data to identify a composite biomarker for colorectal cancer. Gut. 2017.
13.
go back to reference Liang Q, Chiu J, Chen Y, et al. Fecal bacteria act as novel biomarkers for noninvasive diagnosis of colorectal cancer. Clin Cancer Res. 2017;23:2061–70.PubMedCrossRef Liang Q, Chiu J, Chen Y, et al. Fecal bacteria act as novel biomarkers for noninvasive diagnosis of colorectal cancer. Clin Cancer Res. 2017;23:2061–70.PubMedCrossRef
14.
go back to reference Flemer B, Lynch DB, Brown JM, et al. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut. 2017;66:633–43.PubMedCrossRef Flemer B, Lynch DB, Brown JM, et al. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut. 2017;66:633–43.PubMedCrossRef
15.
go back to reference • Lasry A, Zinger A, Ben-Neriah Y. Inflammatory networks underlying colorectal cancer. Nat Immunol. 2016;17:230–40. An updated review of the inmportance of inflammatory components in colorectal cancer PubMedCrossRef • Lasry A, Zinger A, Ben-Neriah Y. Inflammatory networks underlying colorectal cancer. Nat Immunol. 2016;17:230–40. An updated review of the inmportance of inflammatory components in colorectal cancer PubMedCrossRef
16.
go back to reference Claesson MJ, Cusack S, O'Sullivan O, et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4586–91.PubMedCrossRef Claesson MJ, Cusack S, O'Sullivan O, et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4586–91.PubMedCrossRef
19.
20.
go back to reference Jalanka-Tuovinen J, Salonen A, Nikkila J, et al. Intestinal microbiota in healthy adults: temporal analysis reveals individual and common core and relation to intestinal symptoms. PLoS One. 2011;6:e23035.PubMedPubMedCentralCrossRef Jalanka-Tuovinen J, Salonen A, Nikkila J, et al. Intestinal microbiota in healthy adults: temporal analysis reveals individual and common core and relation to intestinal symptoms. PLoS One. 2011;6:e23035.PubMedPubMedCentralCrossRef
21.
go back to reference Rajilic-Stojanovic M, Heilig HG, Tims S, et al. Long-term monitoring of the human intestinal microbiota composition. Environ Microbiol. 2012; Rajilic-Stojanovic M, Heilig HG, Tims S, et al. Long-term monitoring of the human intestinal microbiota composition. Environ Microbiol. 2012;
22.
go back to reference • David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–63. A landmark study indicating that the gut microbiome can rapidly respond to altered diet PubMedCrossRef • David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–63. A landmark study indicating that the gut microbiome can rapidly respond to altered diet PubMedCrossRef
26.
go back to reference •• O'Keefe SJ, Li JV, Lahti L, et al. Fat, fibre and cancer risk in African Americans and rural Africans. Nat Commun. 2015;6:6342. The study provides strong eivdence for the role of the gut microbiome in mediating the relationship between dietary factors and cancer risk PubMedPubMedCentralCrossRef •• O'Keefe SJ, Li JV, Lahti L, et al. Fat, fibre and cancer risk in African Americans and rural Africans. Nat Commun. 2015;6:6342. The study provides strong eivdence for the role of the gut microbiome in mediating the relationship between dietary factors and cancer risk PubMedPubMedCentralCrossRef
27.
go back to reference •• Zeevi D, Korem T, Zmora N, et al. Personalized nutrition by prediction of glycemic responses. Cell. 2015;163:1079–94. The study suggests that the gut microbiome is an important determinant for the inter-individual variation in the metabolic response to dietary intervention PubMedCrossRef •• Zeevi D, Korem T, Zmora N, et al. Personalized nutrition by prediction of glycemic responses. Cell. 2015;163:1079–94. The study suggests that the gut microbiome is an important determinant for the inter-individual variation in the metabolic response to dietary intervention PubMedCrossRef
28.
go back to reference Zmora N, Zeevi D, Korem T, et al. Taking it personally: personalized utilization of the human microbiome in health and disease. Cell Host Microbe. 2016;19:12–20.PubMedCrossRef Zmora N, Zeevi D, Korem T, et al. Taking it personally: personalized utilization of the human microbiome in health and disease. Cell Host Microbe. 2016;19:12–20.PubMedCrossRef
29.
go back to reference Miller PE, Lesko SM, Muscat JE, et al. Dietary patterns and colorectal adenoma and cancer risk: a review of the epidemiological evidence. Nutr Cancer. 2010;62:413–24.PubMedCrossRef Miller PE, Lesko SM, Muscat JE, et al. Dietary patterns and colorectal adenoma and cancer risk: a review of the epidemiological evidence. Nutr Cancer. 2010;62:413–24.PubMedCrossRef
30.
go back to reference Magalhaes B, Peleteiro B, Lunet N. Dietary patterns and colorectal cancer: systematic review and meta-analysis. Eur J Cancer Prev. 2012;21:15–23.PubMedCrossRef Magalhaes B, Peleteiro B, Lunet N. Dietary patterns and colorectal cancer: systematic review and meta-analysis. Eur J Cancer Prev. 2012;21:15–23.PubMedCrossRef
31.
go back to reference Serino M, Luche E, Gres S, et al. Metabolic adaptation to a high-fat diet is associated with a change in the gut microbiota. Gut. 2012;61:543–53.PubMedCrossRef Serino M, Luche E, Gres S, et al. Metabolic adaptation to a high-fat diet is associated with a change in the gut microbiota. Gut. 2012;61:543–53.PubMedCrossRef
32.
go back to reference Martinez-Medina M, Denizot J, Dreux N, et al. Western diet induces dysbiosis with increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC colonisation. Gut. 2014;63:116–24.PubMedCrossRef Martinez-Medina M, Denizot J, Dreux N, et al. Western diet induces dysbiosis with increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC colonisation. Gut. 2014;63:116–24.PubMedCrossRef
33.
go back to reference Ley SH, Sun Q, Willett WC, et al. Associations between red meat intake and biomarkers of inflammation and glucose metabolism in women. Am J Clin Nutr. 2014;99:352–60.PubMedCrossRef Ley SH, Sun Q, Willett WC, et al. Associations between red meat intake and biomarkers of inflammation and glucose metabolism in women. Am J Clin Nutr. 2014;99:352–60.PubMedCrossRef
34.
go back to reference Schulze MB, Hoffmann K, Manson JE, et al. Dietary pattern, inflammation, and incidence of type 2 diabetes in women. Am J Clin Nutr. 2005;82:675–84. quiz 714-5PubMedPubMedCentral Schulze MB, Hoffmann K, Manson JE, et al. Dietary pattern, inflammation, and incidence of type 2 diabetes in women. Am J Clin Nutr. 2005;82:675–84. quiz 714-5PubMedPubMedCentral
35.
go back to reference Montonen J, Boeing H, Fritsche A, et al. Consumption of red meat and whole-grain bread in relation to biomarkers of obesity, inflammation, glucose metabolism and oxidative stress. Eur J Nutr. 2013;52:337–45.PubMedCrossRef Montonen J, Boeing H, Fritsche A, et al. Consumption of red meat and whole-grain bread in relation to biomarkers of obesity, inflammation, glucose metabolism and oxidative stress. Eur J Nutr. 2013;52:337–45.PubMedCrossRef
36.
go back to reference Esmaillzadeh A, Kimiagar M, Mehrabi Y, et al. Dietary patterns and markers of systemic inflammation among Iranian women. J Nutr. 2007;137:992–8.PubMed Esmaillzadeh A, Kimiagar M, Mehrabi Y, et al. Dietary patterns and markers of systemic inflammation among Iranian women. J Nutr. 2007;137:992–8.PubMed
37.
go back to reference Lopez-Garcia E, Schulze MB, Fung TT, et al. Major dietary patterns are related to plasma concentrations of markers of inflammation and endothelial dysfunction. Am J Clin Nutr. 2004;80:1029–35.PubMed Lopez-Garcia E, Schulze MB, Fung TT, et al. Major dietary patterns are related to plasma concentrations of markers of inflammation and endothelial dysfunction. Am J Clin Nutr. 2004;80:1029–35.PubMed
38.
go back to reference Brown K, DeCoffe D, Molcan E, et al. Diet-induced dysbiosis of the intestinal microbiota and the effects on immunity and disease. Nutrients. 2012;4:1095–119.PubMedPubMedCentralCrossRef Brown K, DeCoffe D, Molcan E, et al. Diet-induced dysbiosis of the intestinal microbiota and the effects on immunity and disease. Nutrients. 2012;4:1095–119.PubMedPubMedCentralCrossRef
40.
go back to reference Kramer CD, Weinberg EO, Gower AC, et al. Distinct gene signatures in aortic tissue from ApoE−/− mice exposed to pathogens or Western diet. BMC Genomics. 2014;15:1176.PubMedPubMedCentralCrossRef Kramer CD, Weinberg EO, Gower AC, et al. Distinct gene signatures in aortic tissue from ApoE−/− mice exposed to pathogens or Western diet. BMC Genomics. 2014;15:1176.PubMedPubMedCentralCrossRef
41.
go back to reference De Filippo C, Cavalieri D, Di Paola M, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010;107:14691–6.PubMedPubMedCentralCrossRef De Filippo C, Cavalieri D, Di Paola M, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010;107:14691–6.PubMedPubMedCentralCrossRef
42.
go back to reference Ou J, Carbonero F, Zoetendal EG, et al. Diet, microbiota, and microbial metabolites in colon cancer risk in rural Africans and African Americans. Am J Clin Nutr. 2013;98:111–20.PubMedPubMedCentralCrossRef Ou J, Carbonero F, Zoetendal EG, et al. Diet, microbiota, and microbial metabolites in colon cancer risk in rural Africans and African Americans. Am J Clin Nutr. 2013;98:111–20.PubMedPubMedCentralCrossRef
43.
go back to reference Mehta RS, Nishihara R, Cao Y, et al. Association of dietary patterns with risk of colorectal cancer subtypes classified by Fusobacterium nucleatum in tumor tissue. JAMA Oncol. 2017; Mehta RS, Nishihara R, Cao Y, et al. Association of dietary patterns with risk of colorectal cancer subtypes classified by Fusobacterium nucleatum in tumor tissue. JAMA Oncol. 2017;
44.
go back to reference Abed J, Emgard JE, Zamir G, et al. Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed gal-GalNAc. Cell Host Microbe. 2016;20:215–25.PubMedPubMedCentralCrossRef Abed J, Emgard JE, Zamir G, et al. Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed gal-GalNAc. Cell Host Microbe. 2016;20:215–25.PubMedPubMedCentralCrossRef
45.
go back to reference Castellarin M, Warren RL, Freeman JD, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22:299–306.PubMedPubMedCentralCrossRef Castellarin M, Warren RL, Freeman JD, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22:299–306.PubMedPubMedCentralCrossRef
46.
go back to reference Kostic AD, Gevers D, Pedamallu CS, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22:292–8.PubMedPubMedCentralCrossRef Kostic AD, Gevers D, Pedamallu CS, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22:292–8.PubMedPubMedCentralCrossRef
47.
48.
50.
go back to reference Allali I, Delgado S, Marron PI, et al. Gut microbiome compositional and functional differences between tumor and non-tumor adjacent tissues from cohorts from the US and Spain. Gut Microbes. 2015:0. Allali I, Delgado S, Marron PI, et al. Gut microbiome compositional and functional differences between tumor and non-tumor adjacent tissues from cohorts from the US and Spain. Gut Microbes. 2015:0.
52.
go back to reference • Mima K, Nishihara R, Qian ZR, et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut. 2016;65:1973–80. The study indicates that high abundance of Fusobacterium nucleatum in the tumor tissue is associated with worse survival of colorectal cancer, providing further support for the pro-colorectal cancer effect of this bacteria. PubMedCrossRef • Mima K, Nishihara R, Qian ZR, et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut. 2016;65:1973–80. The study indicates that high abundance of Fusobacterium nucleatum in the tumor tissue is associated with worse survival of colorectal cancer, providing further support for the pro-colorectal cancer effect of this bacteria. PubMedCrossRef
53.
go back to reference Kostic AD, Chun E, Robertson L, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14:207–15.PubMedPubMedCentralCrossRef Kostic AD, Chun E, Robertson L, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14:207–15.PubMedPubMedCentralCrossRef
54.
go back to reference Gur C, Ibrahim Y, Isaacson B, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42:344–55.PubMedPubMedCentralCrossRef Gur C, Ibrahim Y, Isaacson B, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42:344–55.PubMedPubMedCentralCrossRef
55.
go back to reference Rubinstein MR, Wang X, Liu W, et al. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14:195–206.PubMedPubMedCentralCrossRef Rubinstein MR, Wang X, Liu W, et al. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14:195–206.PubMedPubMedCentralCrossRef
57.
go back to reference Anderson JW, Baird P, Davis RH Jr, et al. Health benefits of dietary fiber. Nutr Rev. 2009;67:188–205.PubMedCrossRef Anderson JW, Baird P, Davis RH Jr, et al. Health benefits of dietary fiber. Nutr Rev. 2009;67:188–205.PubMedCrossRef
58.
go back to reference Pollak M. The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer. 2012;12:159–69.PubMed Pollak M. The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer. 2012;12:159–69.PubMed
59.
go back to reference Giovannucci E, Michaud D. The role of obesity and related metabolic disturbances in cancers of the colon, prostate, and pancreas. Gastroenterology. 2007;132:2208–25.PubMedCrossRef Giovannucci E, Michaud D. The role of obesity and related metabolic disturbances in cancers of the colon, prostate, and pancreas. Gastroenterology. 2007;132:2208–25.PubMedCrossRef
60.
go back to reference Giovannucci E, Harlan DM, Archer MC, et al. Diabetes and cancer: a consensus report. CA Cancer J Clin. 2010;60:207–21.PubMedCrossRef Giovannucci E, Harlan DM, Archer MC, et al. Diabetes and cancer: a consensus report. CA Cancer J Clin. 2010;60:207–21.PubMedCrossRef
62.
go back to reference Chen HM, Yu YN, Wang JL, et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr. 2013;97:1044–52.PubMedCrossRef Chen HM, Yu YN, Wang JL, et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr. 2013;97:1044–52.PubMedCrossRef
63.
go back to reference Donohoe DR, Garge N, Zhang X, et al. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011;13:517–26.PubMedPubMedCentralCrossRef Donohoe DR, Garge N, Zhang X, et al. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011;13:517–26.PubMedPubMedCentralCrossRef
64.
go back to reference Encarnacao JC, Abrantes AM, Pires AS, et al. Revisit dietary fiber on colorectal cancer: butyrate and its role on prevention and treatment. Cancer Metastasis Rev. 2015;34:465–78.PubMedCrossRef Encarnacao JC, Abrantes AM, Pires AS, et al. Revisit dietary fiber on colorectal cancer: butyrate and its role on prevention and treatment. Cancer Metastasis Rev. 2015;34:465–78.PubMedCrossRef
65.
go back to reference •• Donohoe DR, Holley D, Collins LB, et al. A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner. Cancer Discov. 2014;4:1387–97. The study suggests a model of mechanisms by which dietary fiber may protect against colorectal cancer in a gut microbiota- and butyrate-dependent manner, and provides a potential explanation for inconsistent findings about the relationship of fiber intake and colorectal cancer risk reported in epidemiologic studies. PubMedPubMedCentralCrossRef •• Donohoe DR, Holley D, Collins LB, et al. A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner. Cancer Discov. 2014;4:1387–97. The study suggests a model of mechanisms by which dietary fiber may protect against colorectal cancer in a gut microbiota- and butyrate-dependent manner, and provides a potential explanation for inconsistent findings about the relationship of fiber intake and colorectal cancer risk reported in epidemiologic studies. PubMedPubMedCentralCrossRef
66.
go back to reference Tang Y, Chen Y, Jiang H, et al. G-protein-coupled receptor for short-chain fatty acids suppresses colon cancer. Int J Cancer. 2011;128:847–56.PubMedCrossRef Tang Y, Chen Y, Jiang H, et al. G-protein-coupled receptor for short-chain fatty acids suppresses colon cancer. Int J Cancer. 2011;128:847–56.PubMedCrossRef
67.
go back to reference Singh N, Gurav A, Sivaprakasam S, et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014;40:128–39.PubMedPubMedCentralCrossRef Singh N, Gurav A, Sivaprakasam S, et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014;40:128–39.PubMedPubMedCentralCrossRef
68.
go back to reference Thangaraju M, Cresci GA, Liu K, et al. GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res. 2009;69:2826–32.PubMedPubMedCentralCrossRef Thangaraju M, Cresci GA, Liu K, et al. GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res. 2009;69:2826–32.PubMedPubMedCentralCrossRef
69.
go back to reference Arpaia N, Campbell C, Fan X, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:451–5.PubMedPubMedCentralCrossRef Arpaia N, Campbell C, Fan X, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:451–5.PubMedPubMedCentralCrossRef
70.
go back to reference Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504:446–50.PubMedCrossRef Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504:446–50.PubMedCrossRef
71.
go back to reference Smith PM, Howitt MR, Panikov N, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341:569–73.PubMedCrossRef Smith PM, Howitt MR, Panikov N, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341:569–73.PubMedCrossRef
72.
go back to reference Chang PV, Hao L, Offermanns S, et al. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A. 2014;111:2247–52.PubMedPubMedCentralCrossRef Chang PV, Hao L, Offermanns S, et al. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A. 2014;111:2247–52.PubMedPubMedCentralCrossRef
73.
go back to reference Hu Y, Le Leu RK, Christophersen CT, et al. Manipulation of the gut microbiota using resistant starch is associated with protection against colitis-associated colorectal cancer in rats. Carcinogenesis. 2016;37:366–75.PubMedCrossRef Hu Y, Le Leu RK, Christophersen CT, et al. Manipulation of the gut microbiota using resistant starch is associated with protection against colitis-associated colorectal cancer in rats. Carcinogenesis. 2016;37:366–75.PubMedCrossRef
74.
go back to reference Entin-Meer M, Rephaeli A, Yang X, et al. Butyric acid prodrugs are histone deacetylase inhibitors that show antineoplastic activity and radiosensitizing capacity in the treatment of malignant gliomas. Mol Cancer Ther. 2005;4:1952–61.PubMedCrossRef Entin-Meer M, Rephaeli A, Yang X, et al. Butyric acid prodrugs are histone deacetylase inhibitors that show antineoplastic activity and radiosensitizing capacity in the treatment of malignant gliomas. Mol Cancer Ther. 2005;4:1952–61.PubMedCrossRef
75.
go back to reference Kuefer R, Hofer MD, Altug V, et al. Sodium butyrate and tributyrin induce in vivo growth inhibition and apoptosis in human prostate cancer. Br J Cancer. 2004;90:535–41.PubMedPubMedCentralCrossRef Kuefer R, Hofer MD, Altug V, et al. Sodium butyrate and tributyrin induce in vivo growth inhibition and apoptosis in human prostate cancer. Br J Cancer. 2004;90:535–41.PubMedPubMedCentralCrossRef
76.
go back to reference Bras-Goncalves RA, Pocard M, Formento JL, et al. Synergistic efficacy of 3n-butyrate and 5-fluorouracil in human colorectal cancer xenografts via modulation of DNA synthesis. Gastroenterology. 2001;120:874–88.PubMedCrossRef Bras-Goncalves RA, Pocard M, Formento JL, et al. Synergistic efficacy of 3n-butyrate and 5-fluorouracil in human colorectal cancer xenografts via modulation of DNA synthesis. Gastroenterology. 2001;120:874–88.PubMedCrossRef
77.
go back to reference • Belcheva A, Irrazabal T, Robertson SJ, et al. Gut microbial metabolism drives transformation of Msh2-deficient colon epithelial cells. Cell. 2014;158:288–99. The study suggests that the effect of fiber on colorectal cancer depends on the host genetic background, with a procancer effect in the context of MSH2 −/− . PubMedCrossRef • Belcheva A, Irrazabal T, Robertson SJ, et al. Gut microbial metabolism drives transformation of Msh2-deficient colon epithelial cells. Cell. 2014;158:288–99. The study suggests that the effect of fiber on colorectal cancer depends on the host genetic background, with a procancer effect in the context of MSH2 −/− . PubMedCrossRef
78.
go back to reference Reitmair AH, Cai JC, Bjerknes M, et al. MSH2 deficiency contributes to accelerated APC-mediated intestinal tumorigenesis. Cancer Res. 1996;56:2922–6.PubMed Reitmair AH, Cai JC, Bjerknes M, et al. MSH2 deficiency contributes to accelerated APC-mediated intestinal tumorigenesis. Cancer Res. 1996;56:2922–6.PubMed
79.
go back to reference World Cancer Research Fund / American Institute for Cancer Research. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Washington DC: AICR; 2007. World Cancer Research Fund / American Institute for Cancer Research. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Washington DC: AICR; 2007.
80.
go back to reference Cross AJ, Ferrucci LM, Risch A, et al. A large prospective study of meat consumption and colorectal cancer risk: an investigation of potential mechanisms underlying this association. Cancer Res. 2010;70:2406–14.PubMedPubMedCentralCrossRef Cross AJ, Ferrucci LM, Risch A, et al. A large prospective study of meat consumption and colorectal cancer risk: an investigation of potential mechanisms underlying this association. Cancer Res. 2010;70:2406–14.PubMedPubMedCentralCrossRef
82.
go back to reference Magee EA, Richardson CJ, Hughes R, et al. Contribution of dietary protein to sulfide production in the large intestine: an in vitro and a controlled feeding study in humans. Am J Clin Nutr. 2000;72:1488–94.PubMed Magee EA, Richardson CJ, Hughes R, et al. Contribution of dietary protein to sulfide production in the large intestine: an in vitro and a controlled feeding study in humans. Am J Clin Nutr. 2000;72:1488–94.PubMed
84.
go back to reference Roediger WE, Moore J, Babidge W. Colonic sulfide in pathogenesis and treatment of ulcerative colitis. Dig Dis Sci. 1997;42:1571–9.PubMedCrossRef Roediger WE, Moore J, Babidge W. Colonic sulfide in pathogenesis and treatment of ulcerative colitis. Dig Dis Sci. 1997;42:1571–9.PubMedCrossRef
85.
go back to reference Rowan FE, Docherty NG, Coffey JC, et al. Sulphate-reducing bacteria and hydrogen sulphide in the aetiology of ulcerative colitis. Br J Surg. 2009;96:151–8.PubMedCrossRef Rowan FE, Docherty NG, Coffey JC, et al. Sulphate-reducing bacteria and hydrogen sulphide in the aetiology of ulcerative colitis. Br J Surg. 2009;96:151–8.PubMedCrossRef
86.
go back to reference Huycke MM, Gaskins HR. Commensal bacteria, redox stress, and colorectal cancer: mechanisms and models. Exp Biol Med. 2004;229:586–97.CrossRef Huycke MM, Gaskins HR. Commensal bacteria, redox stress, and colorectal cancer: mechanisms and models. Exp Biol Med. 2004;229:586–97.CrossRef
87.
go back to reference Deplancke B, Gaskins HR. Hydrogen sulfide induces serum-independent cell cycle entry in nontransformed rat intestinal epithelial cells. FASEB J. 2003;17:1310–2.PubMed Deplancke B, Gaskins HR. Hydrogen sulfide induces serum-independent cell cycle entry in nontransformed rat intestinal epithelial cells. FASEB J. 2003;17:1310–2.PubMed
88.
go back to reference Attene-Ramos MS, Wagner ED, Gaskins HR, et al. Hydrogen sulfide induces direct radical-associated DNA damage. Mol Cancer Res. 2007;5:455–9.PubMedCrossRef Attene-Ramos MS, Wagner ED, Gaskins HR, et al. Hydrogen sulfide induces direct radical-associated DNA damage. Mol Cancer Res. 2007;5:455–9.PubMedCrossRef
89.
go back to reference Ramasamy S, Singh S, Taniere P, et al. Sulfide-detoxifying enzymes in the human colon are decreased in cancer and upregulated in differentiation. Am J Physiol Gastrointest Liver Physiol. 2006;291:G288–96.PubMedCrossRef Ramasamy S, Singh S, Taniere P, et al. Sulfide-detoxifying enzymes in the human colon are decreased in cancer and upregulated in differentiation. Am J Physiol Gastrointest Liver Physiol. 2006;291:G288–96.PubMedCrossRef
90.
go back to reference Cai WJ, Wang MJ, Ju LH, et al. Hydrogen sulfide induces human colon cancer cell proliferation: role of Akt, ERK and p21. Cell Biol Int. 2010;34:565–72.PubMedCrossRef Cai WJ, Wang MJ, Ju LH, et al. Hydrogen sulfide induces human colon cancer cell proliferation: role of Akt, ERK and p21. Cell Biol Int. 2010;34:565–72.PubMedCrossRef
91.
go back to reference Carbonero F, Benefiel AC, Gaskins HR.Contributions of the microbial hydrogen economy to colonic homeostasis Nature reviews. Gastroenterol Hepatol 2012;9:504–518. Carbonero F, Benefiel AC, Gaskins HR.Contributions of the microbial hydrogen economy to colonic homeostasis Nature reviews. Gastroenterol Hepatol 2012;9:504–518.
92.
go back to reference Wu YC, Wang XJ, Yu L, et al. Hydrogen sulfide lowers proliferation and induces protective autophagy in colon epithelial cells. PLoS One. 2012;7:e37572.PubMedPubMedCentralCrossRef Wu YC, Wang XJ, Yu L, et al. Hydrogen sulfide lowers proliferation and induces protective autophagy in colon epithelial cells. PLoS One. 2012;7:e37572.PubMedPubMedCentralCrossRef
93.
go back to reference Roediger WE, Duncan A, Kapaniris O, et al. Reducing sulfur compounds of the colon impair colonocyte nutrition: implications for ulcerative colitis. Gastroenterology. 1993;104:802–9.PubMedCrossRef Roediger WE, Duncan A, Kapaniris O, et al. Reducing sulfur compounds of the colon impair colonocyte nutrition: implications for ulcerative colitis. Gastroenterology. 1993;104:802–9.PubMedCrossRef
94.
go back to reference Pitcher MC, Beatty ER, Cummings JH. The contribution of sulphate reducing bacteria and 5-aminosalicylic acid to faecal sulphide in patients with ulcerative colitis. Gut. 2000;46:64–72.PubMedPubMedCentralCrossRef Pitcher MC, Beatty ER, Cummings JH. The contribution of sulphate reducing bacteria and 5-aminosalicylic acid to faecal sulphide in patients with ulcerative colitis. Gut. 2000;46:64–72.PubMedPubMedCentralCrossRef
95.
go back to reference Vinolo MA, Rodrigues HG, Hatanaka E, et al. Short-chain fatty acids stimulate the migration of neutrophils to inflammatory sites. Clin Sci. 2009;117:331–8.PubMedCrossRef Vinolo MA, Rodrigues HG, Hatanaka E, et al. Short-chain fatty acids stimulate the migration of neutrophils to inflammatory sites. Clin Sci. 2009;117:331–8.PubMedCrossRef
96.
go back to reference Zeng H, Combs GF Jr. Selenium as an anticancer nutrient: roles in cell proliferation and tumor cell invasion. J Nutr Biochem. 2008;19:1–7.PubMedCrossRef Zeng H, Combs GF Jr. Selenium as an anticancer nutrient: roles in cell proliferation and tumor cell invasion. J Nutr Biochem. 2008;19:1–7.PubMedCrossRef
97.
go back to reference Miller TW, Wang EA, Gould S, et al. Hydrogen sulfide is an endogenous potentiator of T cell activation. J Biol Chem. 2012;287:4211–21.PubMedCrossRef Miller TW, Wang EA, Gould S, et al. Hydrogen sulfide is an endogenous potentiator of T cell activation. J Biol Chem. 2012;287:4211–21.PubMedCrossRef
98.
go back to reference O'Keefe SJ, Ou J, Aufreiter S, et al. Products of the colonic microbiota mediate the effects of diet on colon cancer risk. J Nutr. 2009;139:2044–8.PubMedCrossRef O'Keefe SJ, Ou J, Aufreiter S, et al. Products of the colonic microbiota mediate the effects of diet on colon cancer risk. J Nutr. 2009;139:2044–8.PubMedCrossRef
99.
100.
go back to reference Carbonero F, Benefiel AC, Alizadeh-Ghamsari AH, et al. Microbial pathways in colonic sulfur metabolism and links with health and disease. Front Physiol. 2012;3:448.PubMedPubMedCentralCrossRef Carbonero F, Benefiel AC, Alizadeh-Ghamsari AH, et al. Microbial pathways in colonic sulfur metabolism and links with health and disease. Front Physiol. 2012;3:448.PubMedPubMedCentralCrossRef
101.
go back to reference Larsson SC, Kumlin M, Ingelman-Sundberg M, et al. Dietary long-chain n-3 fatty acids for the prevention of cancer: a review of potential mechanisms. Am J Clin Nutr. 2004;79:935–45.PubMed Larsson SC, Kumlin M, Ingelman-Sundberg M, et al. Dietary long-chain n-3 fatty acids for the prevention of cancer: a review of potential mechanisms. Am J Clin Nutr. 2004;79:935–45.PubMed
102.
go back to reference Cockbain AJ, Toogood GJ, Hull MA. Omega-3 polyunsaturated fatty acids for the treatment and prevention of colorectal cancer. Gut. 2012;61:135–49.PubMedCrossRef Cockbain AJ, Toogood GJ, Hull MA. Omega-3 polyunsaturated fatty acids for the treatment and prevention of colorectal cancer. Gut. 2012;61:135–49.PubMedCrossRef
103.
104.
go back to reference •• Cockbain AJ, Volpato M, Race AD, et al. Anticolorectal cancer activity of the omega-3 polyunsaturated fatty acid eicosapentaenoic acid. Gut. 2014;63:1760–8. This randomized controlled study supports the chemopreventive effect of omega-3 fatty acid supplementation on colorectal cancer. PubMedCrossRef •• Cockbain AJ, Volpato M, Race AD, et al. Anticolorectal cancer activity of the omega-3 polyunsaturated fatty acid eicosapentaenoic acid. Gut. 2014;63:1760–8. This randomized controlled study supports the chemopreventive effect of omega-3 fatty acid supplementation on colorectal cancer. PubMedCrossRef
105.
go back to reference Clarke TC, Black LI, Stussman BJ, et al. Trends in the use of complementary health approaches among adults: United States, 2002–2012. Natl. Health Stat. Rep. 2015:1–16. Clarke TC, Black LI, Stussman BJ, et al. Trends in the use of complementary health approaches among adults: United States, 2002–2012. Natl. Health Stat. Rep. 2015:1–16.
106.
go back to reference West NJ, Clark SK, Phillips RK, et al. Eicosapentaenoic acid reduces rectal polyp number and size in familial adenomatous polyposis. Gut. 2010;59:918–25.PubMedCrossRef West NJ, Clark SK, Phillips RK, et al. Eicosapentaenoic acid reduces rectal polyp number and size in familial adenomatous polyposis. Gut. 2010;59:918–25.PubMedCrossRef
107.
go back to reference Calder PC. Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance. Biochim Biophys Acta. 1851;2015:469–84. Calder PC. Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance. Biochim Biophys Acta. 1851;2015:469–84.
108.
go back to reference Piazzi G, D'Argenio G, Prossomariti A, et al. Eicosapentaenoic acid free fatty acid prevents and suppresses colonic neoplasia in colitis-associated colorectal cancer acting on Notch signaling and gut microbiota. J Int Cancer. 2014;135:2004–13.CrossRef Piazzi G, D'Argenio G, Prossomariti A, et al. Eicosapentaenoic acid free fatty acid prevents and suppresses colonic neoplasia in colitis-associated colorectal cancer acting on Notch signaling and gut microbiota. J Int Cancer. 2014;135:2004–13.CrossRef
109.
go back to reference Jiang Y, Djuric Z, Sen A, et al. Biomarkers for personalizing omega-3 fatty acid dosing. Cancer Prev Res (Phila). 2014;7:1011–22.CrossRef Jiang Y, Djuric Z, Sen A, et al. Biomarkers for personalizing omega-3 fatty acid dosing. Cancer Prev Res (Phila). 2014;7:1011–22.CrossRef
110.
go back to reference Calviello G, Di Nicuolo F, Gragnoli S, et al. n-3 PUFAs reduce VEGF expression in human colon cancer cells modulating the COX-2/PGE2 induced ERK-1 and -2 and HIF-1alpha induction pathway. Carcinogenesis. 2004;25:2303–10.PubMedCrossRef Calviello G, Di Nicuolo F, Gragnoli S, et al. n-3 PUFAs reduce VEGF expression in human colon cancer cells modulating the COX-2/PGE2 induced ERK-1 and -2 and HIF-1alpha induction pathway. Carcinogenesis. 2004;25:2303–10.PubMedCrossRef
111.
go back to reference Bartram HP, Gostner A, Scheppach W, et al. Effects of fish oil on rectal cell proliferation, mucosal fatty acids, and prostaglandin E2 release in healthy subjects. Gastroenterology. 1993;105:1317–22.PubMedCrossRef Bartram HP, Gostner A, Scheppach W, et al. Effects of fish oil on rectal cell proliferation, mucosal fatty acids, and prostaglandin E2 release in healthy subjects. Gastroenterology. 1993;105:1317–22.PubMedCrossRef
112.
go back to reference Nowak J, Weylandt KH, Habbel P, et al. Colitis-associated colon tumorigenesis is suppressed in transgenic mice rich in endogenous n-3 fatty acids. Carcinogenesis. 2007;28:1991–5.PubMedCrossRef Nowak J, Weylandt KH, Habbel P, et al. Colitis-associated colon tumorigenesis is suppressed in transgenic mice rich in endogenous n-3 fatty acids. Carcinogenesis. 2007;28:1991–5.PubMedCrossRef
113.
go back to reference • Song M, Nishihara R, Cao Y, et al. Marine omega-3 polyunsaturated fatty acid intake and risk of colorectal cancer characterized by tumor-infiltrating T cells. JAMA Oncol. 2016;2:1197–206. This study suggests that the beneficial effect of high omega-3 fatty acid intake may be partly mediated by modulation of regulatory T cells in the tumor microenvironment. PubMedPubMedCentralCrossRef • Song M, Nishihara R, Cao Y, et al. Marine omega-3 polyunsaturated fatty acid intake and risk of colorectal cancer characterized by tumor-infiltrating T cells. JAMA Oncol. 2016;2:1197–206. This study suggests that the beneficial effect of high omega-3 fatty acid intake may be partly mediated by modulation of regulatory T cells in the tumor microenvironment. PubMedPubMedCentralCrossRef
114.
go back to reference Caesar R, Tremaroli V, Kovatcheva-Datchary P, et al. Crosstalk between gut microbiota and dietary lipids aggravates WAT inflammation through TLR signaling. Cell Metab. 2015;22:658–68.PubMedPubMedCentralCrossRef Caesar R, Tremaroli V, Kovatcheva-Datchary P, et al. Crosstalk between gut microbiota and dietary lipids aggravates WAT inflammation through TLR signaling. Cell Metab. 2015;22:658–68.PubMedPubMedCentralCrossRef
115.
go back to reference Devkota S, Wang Y, Musch MW, et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10 −/− mice. Nature. 2012;487:104–8.PubMedPubMedCentral Devkota S, Wang Y, Musch MW, et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10 −/− mice. Nature. 2012;487:104–8.PubMedPubMedCentral
116.
go back to reference Ghosh S, DeCoffe D, Brown K, et al. Fish oil attenuates omega-6 polyunsaturated fatty acid-induced dysbiosis and infectious colitis but impairs LPS dephosphorylation activity causing sepsis. PLoS One. 2013;8:e55468.PubMedPubMedCentralCrossRef Ghosh S, DeCoffe D, Brown K, et al. Fish oil attenuates omega-6 polyunsaturated fatty acid-induced dysbiosis and infectious colitis but impairs LPS dephosphorylation activity causing sepsis. PLoS One. 2013;8:e55468.PubMedPubMedCentralCrossRef
117.
go back to reference Patterson E, RM OD, Murphy EF, et al. Impact of dietary fatty acids on metabolic activity and host intestinal microbiota composition in C57BL/6J mice. Br J Nutr. 2014;1–13. Patterson E, RM OD, Murphy EF, et al. Impact of dietary fatty acids on metabolic activity and host intestinal microbiota composition in C57BL/6J mice. Br J Nutr. 2014;1–13.
118.
go back to reference Mujico JR, Baccan GC, Gheorghe A, et al. Changes in gut microbiota due to supplemented fatty acids in diet-induced obese mice. Br J Nutr. 2013;110:711–20.PubMedCrossRef Mujico JR, Baccan GC, Gheorghe A, et al. Changes in gut microbiota due to supplemented fatty acids in diet-induced obese mice. Br J Nutr. 2013;110:711–20.PubMedCrossRef
119.
go back to reference Shen W, Gaskins HR, McIntosh MK. Influence of dietary fat on intestinal microbes, inflammation, barrier function and metabolic outcomes. J Nutr Biochem. 2014;25:270–80.PubMedCrossRef Shen W, Gaskins HR, McIntosh MK. Influence of dietary fat on intestinal microbes, inflammation, barrier function and metabolic outcomes. J Nutr Biochem. 2014;25:270–80.PubMedCrossRef
120.
go back to reference Ghosh S, Molcan E, DeCoffe D, et al. Diets rich in n-6 PUFA induce intestinal microbial dysbiosis in aged mice. Br J Nutr. 2013;110:515–23.PubMedCrossRef Ghosh S, Molcan E, DeCoffe D, et al. Diets rich in n-6 PUFA induce intestinal microbial dysbiosis in aged mice. Br J Nutr. 2013;110:515–23.PubMedCrossRef
121.
go back to reference Kaliannan K, Wang B, Li XY, et al. Omega-3 fatty acids prevent early-life antibiotic exposure-induced gut microbiota dysbiosis and later-life obesity. Int J Obes (Lond). 2016. Kaliannan K, Wang B, Li XY, et al. Omega-3 fatty acids prevent early-life antibiotic exposure-induced gut microbiota dysbiosis and later-life obesity. Int J Obes (Lond). 2016.
122.
go back to reference Jenq RR, Ubeda C, Taur Y, et al. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J Exp Med. 2012;209:903–11.PubMedPubMedCentralCrossRef Jenq RR, Ubeda C, Taur Y, et al. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J Exp Med. 2012;209:903–11.PubMedPubMedCentralCrossRef
123.
go back to reference Peran L, Sierra S, Comalada M, et al. A comparative study of the preventative effects exerted by two probiotics, Lactobacillus reuteri and Lactobacillus fermentum, in the trinitrobenzenesulfonic acid model of rat colitis. Br J Nutr. 2007;97:96–103.PubMedCrossRef Peran L, Sierra S, Comalada M, et al. A comparative study of the preventative effects exerted by two probiotics, Lactobacillus reuteri and Lactobacillus fermentum, in the trinitrobenzenesulfonic acid model of rat colitis. Br J Nutr. 2007;97:96–103.PubMedCrossRef
124.
go back to reference Khazaie K, Zadeh M, Khan MW, et al. Abating colon cancer polyposis by Lactobacillus acidophilus deficient in lipoteichoic acid. Proc Natl Acad Sci U S A. 2012;109:10462–7.PubMedPubMedCentralCrossRef Khazaie K, Zadeh M, Khan MW, et al. Abating colon cancer polyposis by Lactobacillus acidophilus deficient in lipoteichoic acid. Proc Natl Acad Sci U S A. 2012;109:10462–7.PubMedPubMedCentralCrossRef
125.
go back to reference • Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–9. The study indicates that the efficacy of cancer immunotherapy may depend on the abundance of Bifidobacterium in the gut. PubMedPubMedCentralCrossRef • Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–9. The study indicates that the efficacy of cancer immunotherapy may depend on the abundance of Bifidobacterium in the gut. PubMedPubMedCentralCrossRef
126.
go back to reference Iida N, Dzutsev A, Stewart CA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342:967–70.PubMedCrossRef Iida N, Dzutsev A, Stewart CA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342:967–70.PubMedCrossRef
127.
go back to reference Kishino S, Takeuchi M, Park SB, et al. Polyunsaturated fatty acid saturation by gut lactic acid bacteria affecting host lipid composition. Proc Natl Acad Sci U S A. 2013;110:17808–13.PubMedPubMedCentralCrossRef Kishino S, Takeuchi M, Park SB, et al. Polyunsaturated fatty acid saturation by gut lactic acid bacteria affecting host lipid composition. Proc Natl Acad Sci U S A. 2013;110:17808–13.PubMedPubMedCentralCrossRef
128.
go back to reference Kishino S, Ogawa J, Yokozeki K, et al. Metabolic diversity in biohydrogenation of polyunsaturated fatty acids by lactic acid bacteria involving conjugated fatty acid production. Appl Microbiol Biotechnol. 2009;84:87–97.PubMedCrossRef Kishino S, Ogawa J, Yokozeki K, et al. Metabolic diversity in biohydrogenation of polyunsaturated fatty acids by lactic acid bacteria involving conjugated fatty acid production. Appl Microbiol Biotechnol. 2009;84:87–97.PubMedCrossRef
129.
go back to reference Hirata A, Kishino S, Park SB, et al. A novel unsaturated fatty acid hydratase toward C16 to C22 fatty acids from Lactobacillus acidophilus. J Lipid Res. 2015;56:1340–50.PubMedPubMedCentralCrossRef Hirata A, Kishino S, Park SB, et al. A novel unsaturated fatty acid hydratase toward C16 to C22 fatty acids from Lactobacillus acidophilus. J Lipid Res. 2015;56:1340–50.PubMedPubMedCentralCrossRef
130.
go back to reference Desbois AP, Smith VJ. Antibacterial free fatty acids: activities, mechanisms of action and biotechnological potential. Appl Microbiol Biotechnol. 2010;85:1629–42.PubMedCrossRef Desbois AP, Smith VJ. Antibacterial free fatty acids: activities, mechanisms of action and biotechnological potential. Appl Microbiol Biotechnol. 2010;85:1629–42.PubMedCrossRef
131.
132.
go back to reference Druart C, Bindels LB, Schmaltz R, et al. Ability of the gut microbiota to produce PUFA-derived bacterial metabolites: proof of concept in germ-free versus conventionalized mice. Mol Nutr Food Res. 2015;59:1603–13.PubMedPubMedCentralCrossRef Druart C, Bindels LB, Schmaltz R, et al. Ability of the gut microbiota to produce PUFA-derived bacterial metabolites: proof of concept in germ-free versus conventionalized mice. Mol Nutr Food Res. 2015;59:1603–13.PubMedPubMedCentralCrossRef
133.
go back to reference Druart C, Neyrinck AM, Vlaeminck B, et al. Role of the lower and upper intestine in the production and absorption of gut microbiota-derived PUFA metabolites. PLoS One. 2014;9:e87560.PubMedPubMedCentralCrossRef Druart C, Neyrinck AM, Vlaeminck B, et al. Role of the lower and upper intestine in the production and absorption of gut microbiota-derived PUFA metabolites. PLoS One. 2014;9:e87560.PubMedPubMedCentralCrossRef
134.
go back to reference Furumoto H, Nanthirudjanar T, Kume T, et al. 10-Oxo-trans-11-octadecenoic acid generated from linoleic acid by a gut lactic acid bacterium Lactobacillus plantarum is cytoprotective against oxidative stress. Toxicol Appl Pharmacol. 2016;296:1–9.PubMedCrossRef Furumoto H, Nanthirudjanar T, Kume T, et al. 10-Oxo-trans-11-octadecenoic acid generated from linoleic acid by a gut lactic acid bacterium Lactobacillus plantarum is cytoprotective against oxidative stress. Toxicol Appl Pharmacol. 2016;296:1–9.PubMedCrossRef
135.
go back to reference Miyamoto J, Mizukure T, Park SB, et al. A gut microbial metabolite of linoleic acid, 10-hydroxy-cis-12-octadecenoic acid, ameliorates intestinal epithelial barrier impairment partially via GPR40-MEK-ERK pathway. J Biol Chem. 2015;290:2902–18.PubMedCrossRef Miyamoto J, Mizukure T, Park SB, et al. A gut microbial metabolite of linoleic acid, 10-hydroxy-cis-12-octadecenoic acid, ameliorates intestinal epithelial barrier impairment partially via GPR40-MEK-ERK pathway. J Biol Chem. 2015;290:2902–18.PubMedCrossRef
136.
go back to reference Flint HJ, Duncan SH, Scott KP, et al. Links between diet, gut microbiota composition and gut metabolism. Proc Nutr Soc. 2015;74:13–22.PubMedCrossRef Flint HJ, Duncan SH, Scott KP, et al. Links between diet, gut microbiota composition and gut metabolism. Proc Nutr Soc. 2015;74:13–22.PubMedCrossRef
137.
go back to reference Belenguer A, Duncan SH, Calder AG, et al. Two routes of metabolic cross-feeding between Bifidobacterium adolescentis and butyrate-producing anaerobes from the human gut. Appl Environ Microbiol. 2006;72:3593–9.PubMedPubMedCentralCrossRef Belenguer A, Duncan SH, Calder AG, et al. Two routes of metabolic cross-feeding between Bifidobacterium adolescentis and butyrate-producing anaerobes from the human gut. Appl Environ Microbiol. 2006;72:3593–9.PubMedPubMedCentralCrossRef
138.
go back to reference Duncan SH, Louis P, Flint HJ. Lactate-utilizing bacteria, isolated from human feces, that produce butyrate as a major fermentation product. Appl Environ Microbiol. 2004;70:5810–7.PubMedPubMedCentralCrossRef Duncan SH, Louis P, Flint HJ. Lactate-utilizing bacteria, isolated from human feces, that produce butyrate as a major fermentation product. Appl Environ Microbiol. 2004;70:5810–7.PubMedPubMedCentralCrossRef
140.
go back to reference O'Keefe SJ. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol. 2016;13:691–706.PubMedCrossRef O'Keefe SJ. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol. 2016;13:691–706.PubMedCrossRef
141.
go back to reference Kong SY, Tran HQ, Gewirtz AT, et al. Serum endotoxins and flagellin and risk of colorectal cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC) Cohort. Cancer Epidemiol Biomark Prev. 2016;25:291–301.CrossRef Kong SY, Tran HQ, Gewirtz AT, et al. Serum endotoxins and flagellin and risk of colorectal cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC) Cohort. Cancer Epidemiol Biomark Prev. 2016;25:291–301.CrossRef
142.
go back to reference •• Kaliannan K, Wang B, Li XY, et al. A host-microbiome interaction mediates the opposing effects of omega-6 and omega-3 fatty acids on metabolic endotoxemia. Sci Rep. 2015;5:11276. The study proposes a model of mechanisms by which omega-3 fatty acid may influence the gut microbial composition through the influence on epithelial production of intestinal alkaline phosphatase. PubMedPubMedCentralCrossRef •• Kaliannan K, Wang B, Li XY, et al. A host-microbiome interaction mediates the opposing effects of omega-6 and omega-3 fatty acids on metabolic endotoxemia. Sci Rep. 2015;5:11276. The study proposes a model of mechanisms by which omega-3 fatty acid may influence the gut microbial composition through the influence on epithelial production of intestinal alkaline phosphatase. PubMedPubMedCentralCrossRef
143.
go back to reference Campbell EL, MacManus CF, Kominsky DJ, et al. Resolvin E1-induced intestinal alkaline phosphatase promotes resolution of inflammation through LPS detoxification. Proc Natl Acad Sci U S A. 2010;107:14298–303.PubMedPubMedCentralCrossRef Campbell EL, MacManus CF, Kominsky DJ, et al. Resolvin E1-induced intestinal alkaline phosphatase promotes resolution of inflammation through LPS detoxification. Proc Natl Acad Sci U S A. 2010;107:14298–303.PubMedPubMedCentralCrossRef
144.
go back to reference Polan CE, McNeill JJ, Tove SB. Biohydrogenation of unsaturated fatty acids by rumen bacteria. J Bacteriol. 1964;88:1056–64.PubMedPubMedCentral Polan CE, McNeill JJ, Tove SB. Biohydrogenation of unsaturated fatty acids by rumen bacteria. J Bacteriol. 1964;88:1056–64.PubMedPubMedCentral
145.
146.
go back to reference Finucane MM, Stevens GA, Cowan MJ, et al. National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants. Lancet. 2011;377:557–67.PubMedPubMedCentralCrossRef Finucane MM, Stevens GA, Cowan MJ, et al. National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants. Lancet. 2011;377:557–67.PubMedPubMedCentralCrossRef
147.
go back to reference Swinburn BA, Sacks G, Hall KD, et al. The global obesity pandemic: shaped by global drivers and local environments. Lancet. 2011;378:804–14.PubMedCrossRef Swinburn BA, Sacks G, Hall KD, et al. The global obesity pandemic: shaped by global drivers and local environments. Lancet. 2011;378:804–14.PubMedCrossRef
148.
go back to reference Malik VS, Willett WC, Hu FB. Global obesity: trends, risk factors and policy implications. Nat. Rev. Endocrinol. 2013;9:13–27.PubMedCrossRef Malik VS, Willett WC, Hu FB. Global obesity: trends, risk factors and policy implications. Nat. Rev. Endocrinol. 2013;9:13–27.PubMedCrossRef
149.
go back to reference Lauby-Secretan B, Scoccianti C, Loomis D, et al. Body fatness and cancer—viewpoint of the IARC Working Group. N Engl J Med. 2016;375:794–8.PubMedCrossRef Lauby-Secretan B, Scoccianti C, Loomis D, et al. Body fatness and cancer—viewpoint of the IARC Working Group. N Engl J Med. 2016;375:794–8.PubMedCrossRef
150.
go back to reference Doyle SL, Donohoe CL, Lysaght J, et al. Visceral obesity, metabolic syndrome, insulin resistance and cancer. Proc Nutr Soc. 2012;71:181–9.PubMedCrossRef Doyle SL, Donohoe CL, Lysaght J, et al. Visceral obesity, metabolic syndrome, insulin resistance and cancer. Proc Nutr Soc. 2012;71:181–9.PubMedCrossRef
151.
go back to reference Renehan AG, Zwahlen M, Egger M. Adiposity and cancer risk: new mechanistic insights from epidemiology. Nat Rev Cancer. 2015;15:484–98.PubMedCrossRef Renehan AG, Zwahlen M, Egger M. Adiposity and cancer risk: new mechanistic insights from epidemiology. Nat Rev Cancer. 2015;15:484–98.PubMedCrossRef
152.
go back to reference Ley RE, Turnbaugh PJ, Klein S, et al. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–3.PubMedCrossRef Ley RE, Turnbaugh PJ, Klein S, et al. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–3.PubMedCrossRef
153.
go back to reference Furet JP, Kong LC, Tap J, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes. 2010;59:3049–57.PubMedPubMedCentralCrossRef Furet JP, Kong LC, Tap J, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes. 2010;59:3049–57.PubMedPubMedCentralCrossRef
155.
go back to reference Duncan SH, Lobley GE, Holtrop G, et al. Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes. 2008;32:1720–4.CrossRef Duncan SH, Lobley GE, Holtrop G, et al. Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes. 2008;32:1720–4.CrossRef
156.
go back to reference Backhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A. 2004;101:15718–23.PubMedPubMedCentralCrossRef Backhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A. 2004;101:15718–23.PubMedPubMedCentralCrossRef
157.
go back to reference Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–31.PubMedCrossRef Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–31.PubMedCrossRef
158.
go back to reference Turnbaugh PJ, Backhed F, Fulton L, et al. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3:213–23.PubMedPubMedCentralCrossRef Turnbaugh PJ, Backhed F, Fulton L, et al. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3:213–23.PubMedPubMedCentralCrossRef
159.
160.
go back to reference • Cox LM, Yamanishi S, Sohn J, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell. 2014;158:705–21. The study suggests that disturbances of the gut microbiome in early life may contribute to subsequent development of obesity in later life. PubMedPubMedCentralCrossRef • Cox LM, Yamanishi S, Sohn J, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell. 2014;158:705–21. The study suggests that disturbances of the gut microbiome in early life may contribute to subsequent development of obesity in later life. PubMedPubMedCentralCrossRef
161.
go back to reference Cao Y, Wu K, Mehta R, et al. Long-term use of antibiotics and risk of colorectal adenoma. Gut. 2017. Cao Y, Wu K, Mehta R, et al. Long-term use of antibiotics and risk of colorectal adenoma. Gut. 2017.
162.
go back to reference Kilkkinen A, Rissanen H, Klaukka T, et al. Antibiotic use predicts an increased risk of cancer. Int J Cancer. 2008;123:2152–5.PubMedCrossRef Kilkkinen A, Rissanen H, Klaukka T, et al. Antibiotic use predicts an increased risk of cancer. Int J Cancer. 2008;123:2152–5.PubMedCrossRef
163.
go back to reference Boursi B, Haynes K, Mamtani R, et al. Impact of antibiotic exposure on the risk of colorectal cancer. Pharmacoepidemiol Drug Saf. 2015;24:534–42.PubMedCrossRef Boursi B, Haynes K, Mamtani R, et al. Impact of antibiotic exposure on the risk of colorectal cancer. Pharmacoepidemiol Drug Saf. 2015;24:534–42.PubMedCrossRef
164.
go back to reference Dik VK, van Oijen MG, Smeets HM, et al. Frequent use of antibiotics is associated with colorectal cancer risk: results of a nested case-control study. Dig Dis Sci. 2016;61:255–64.PubMedCrossRef Dik VK, van Oijen MG, Smeets HM, et al. Frequent use of antibiotics is associated with colorectal cancer risk: results of a nested case-control study. Dig Dis Sci. 2016;61:255–64.PubMedCrossRef
165.
166.
go back to reference Duca FA, Lam TK. Gut microbiota, nutrient sensing and energy balance. Diabetes Obes Metab. 2014;16(Suppl 1):68–76.PubMedCrossRef Duca FA, Lam TK. Gut microbiota, nutrient sensing and energy balance. Diabetes Obes Metab. 2014;16(Suppl 1):68–76.PubMedCrossRef
169.
go back to reference Yoshimoto S, Loo TM, Atarashi K, et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 2013;499:97–101.PubMedCrossRef Yoshimoto S, Loo TM, Atarashi K, et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 2013;499:97–101.PubMedCrossRef
170.
go back to reference Ohtani N, Yoshimoto S, Hara E. Obesity and cancer: a gut microbial connection. Cancer Res. 2014;74:1885–9.PubMedCrossRef Ohtani N, Yoshimoto S, Hara E. Obesity and cancer: a gut microbial connection. Cancer Res. 2014;74:1885–9.PubMedCrossRef
171.
go back to reference •• Loo TM, Kamachi F, Watanabe Y, et al. Gut microbiota promotes obesity-associated liver cancer through PGE2-mediated suppression of antitumor immunity. Cancer Discov. 2017. The study provides new evidence about how obesity may promote liver cancer through an influence on the functionality of the gut microbiota. •• Loo TM, Kamachi F, Watanabe Y, et al. Gut microbiota promotes obesity-associated liver cancer through PGE2-mediated suppression of antitumor immunity. Cancer Discov. 2017. The study provides new evidence about how obesity may promote liver cancer through an influence on the functionality of the gut microbiota.
173.
go back to reference Wang D, DuBois RN. PPARdelta and PGE2 signaling pathways communicate and connect inflammation to colorectal cancer. Inflamm Cell Signal. 2014;1. Wang D, DuBois RN. PPARdelta and PGE2 signaling pathways communicate and connect inflammation to colorectal cancer. Inflamm Cell Signal. 2014;1.
Metadata
Title
Diet, Gut Microbiota, and Colorectal Cancer Prevention: a Review of Potential Mechanisms and Promising Targets for Future Research
Authors
Mingyang Song
Andrew T. Chan
Publication date
01-12-2017
Publisher
Springer US
Published in
Current Colorectal Cancer Reports / Issue 6/2017
Print ISSN: 1556-3790
Electronic ISSN: 1556-3804
DOI
https://doi.org/10.1007/s11888-017-0389-y

Other articles of this Issue 6/2017

Current Colorectal Cancer Reports 6/2017 Go to the issue

Systemic Therapies in Colorectal Cancer (RD Kim, Section Editor)

Second-Line Therapy for Advanced Colorectal Cancer: EGFR vs. Continuation of VEGF Inhibition

Genetic Syndromes, Screening, and Surveillance in Colorectal Cancer (N Kubiliun, Section Editor)

Stool- and Blood-Based Molecular Tests in Screening for Colorectal Cancer: Ready for Prime Time?

Nutrition and Nutritional Interventions in Colorectal Cancer (K Wu, Section Editor)

Dietary Patterns and Colorectal Cancer Risk: a Review of 17 Years of Evidence (2000–2016)

Diagnostic and Interventional Radiology Innovations in Colorectal Cancer (G de Prisco, Section Editor)

Liver Imaging for Colorectal Cancer Metastases

Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine