Abstract
The human gut is home to a complex and diverse microbiota that contributes to the overall homeostasis of the host. Increasingly, the intestinal microbiota is recognized as an important player in human illness such as colorectal cancer (CRC), inflammatory bowel diseases, and obesity. CRC in itself is one of the major causes of cancer mortality in the Western world. The mechanisms by which bacteria contribute to CRC are complex and not fully understood, but increasing evidence suggests a link between the intestinal microbiota and CRC as well as diet and inflammation, which are believed to play a role in carcinogenesis. It is thought that the gut microbiota interact with dietary factors to promote chronic inflammation and CRC through direct influence on host cell physiology, cellular homeostasis, energy regulation, and/or metabolism of xenobiotics. This review provides an overview on the role of commensal gut microbiota in the development of human CRC and explores its association with diet and inflammation.
Keywords: colorectal adenoma, diet, inflammation, bacterial metabolites
colorectal cancer (CRC) has the third highest cancer incidence in the world. In 2012 CRC accounted for ∼1,361,000 new cases (9.7% of total cancer incidence) and ∼694,000 deaths (8.5% of total cancer deaths) globally (45). In the United States, CRC is the second leading cause of cancer mortality and the third most commonly diagnosed cancer. It is estimated to account for roughly 50,310 deaths and about 136,830 new cases in 2014 (142).
Despite extensive research, the precise etiology for CRC is still unknown, but genetic, epigenetic, and environmental factors such as diet have been implicated. CRC arises as part of a multistep process associated with the accumulation of a series of genetic and epigenetic alterations (49) that influence fitness and clonal expansion of altered cells in the transformation of normal colonic epithelium to adenomas and cancer.
The first model depicting key genetic alterations in CRC included the activation of K-ras oncogene (44, 84, 163) and the inactivation of tumor suppressor genes such as adenomatous polyposis coli (104, 133) and p53 (encoded by TP53 gene in human) (164, 169). Following this initial model, a number of acquired genetic mutations including PIK3CA, FBXW7, SMAD4, TCF7L2, NRAS, FAM123B, CTNNB1, SMAD2, alterations in pathways of chromosomal and microsatellite instability, mismatch repair (23, 25), and epigenetic CpG island methylation (3, 128), have also been observed to contribute to colorectal carcinogenesis.
In addition to genetic factors, an environmental component to CRC is also strongly implicated. Studies of immigrants show that cancer rates for migrants quickly match those of their current country of residence, even when they originated from a country with a lower baseline cancer rate (46, 102). Because genes do not change over such a short period, diet, a component that changes with migration, is thought to be an important contributor to CRC development. However, studies of dietary elements, such as fiber and CRC, to date are largely inconsistent with some studies reporting protective effects of fiber and others showing no effects (6, 118). This is likely because of an overlooked component of the large bowel, the gut microbiome. There is mounting evidence that the intestinal gut bacteria (microbiota) play an important role in colorectal carcinogenesis (37, 69, 126, 157, 174).
Intestinal Microbiota
The human large bowel is home to complex and diverse communities of microbiota that play important roles in health and disease. Several studies suggest that, depending on the route of delivery, the human gastrointestinal tract is colonized at birth, at least with limited microbes (73, 134) that gradually become diverse to reach more than 1014 microbes comprising 1,000 or more heterogeneous species of bacteria, viruses, archaea, and fungi (135). It is estimated that the number of bacterial cells of the human gut exceeds the number of human cells by 10-fold (48). The collective bacterial genome, referred to as the gut microbiota, harbors ∼150-fold more genes than the human genome (120, 171).
Within the large bowel, bacteria are present in two compartments: a luminal compartment and a mucosal adherent compartment. The microbiota in the luminal compartment, while reflective of the bacterial communities in the colonic lumen, may not reflect the composition and localization of epithelial and cryptal communities (135). The colonic mucosa harbors dense cohesive communities of bacteria that adhere to surface-associated polysaccharide matrices and resist hydrodynamic shear forces (146). Some of these bacteria establish themselves as entrenched residents of the mucosa (146) despite the rapid turnover and propulsion of debris and water through the gut. These adherent resident bacteria interact with the mucosal immune system and as such may be more relevant to diseases such as inflammatory bowel diseases (IBD) and CRC.
Although the gut bacteria have long been considered commensal residents, it is now recognized that they serve diverse and important functions (18) some of which may contribute to CRC etiology. The intestinal bacteria are essential in digestion and absorption of indigestible carbohydrates (fibers), production of vitamins B and K, metabolism of endogenous and exogenous compounds, immune potentiation, are actively involved in innate and cell-mediated immunity, help to maintain intestinal barrier function, and assist with an appropriate immune response against pathogenic microbes (9, 74). Their symbiotic relationship (“normobiosis”) with the host is critical to maintaining a balance (homeostasis) in the gut. A shift in this balance (“dysbiosis”) under abnormal conditions can lead to detrimental consequences for the host. For instance, dysbiosis of the normal microbiota is associated with overgrowth of opportunistic pathogens that are normally inhibited by commensal bacteria (12). Moreover, microbial dysbiosis has been observed in IBD (32, 61, 80, 152), obesity (11, 20, 140, 154, 159), colorectal adenomas, and cancer (26, 37, 92, 99, 109, 116, 130, 138, 144, 145, 171). One cause of dysbiosis may be antibiotic treatment, which has been linked to development of Clostridium difficile colitis (59, 64, 139, 170).
The composition of intestinal microbiota is determined by various factors, including host genetics (159), environment (16, 21), diet (26, 43, 103, 160, 176), and hygiene (7). Recent findings demonstrated that paneth cell defensins, innate antimicrobial peptides that contribute to mucosal host defense, can regulate the composition of the intestinal bacteria (129). Although host genetics may have a significant impact on the microbiota composition, discovery of specific genetic factors that interact with the gut microbiota is in the beginning stages. α1,2-Fucosyltransferases (FUT2) secretor genotype is correlated with the abundance of bifidobacteria in the colon (165). FUT2 gene is responsible for the histo-blood group antigens, and polymorphisms in this gene have been associated with IBD and CRC (56, 76).
Intestinal Microbiota and CRC
Several human studies have demonstrated a link between the gut bacteria and CRC. One of the earliest studies to correlate gut bacteria with colorectal neoplasm was conducted by Moore and Moore (101) who assessed fecal samples from polyp patients using culture methods. They observed that the abundance of Bacteroides and bifidobacteria was associated with increased risk of colon polyps, whereas Lactobacillus and Eubacterium aerofaciens were protective. Early studies by Swidsinski et al. (150) also reported an association between the abundance of Escherichia coli and colorectal adenomas and cancer. O'Keefe et al. (113) observed that high abundance of hydrogen sulfide (H2S)- and bile salt-producing bacteria was associated with increased risk of colon cancer. The majority of bacteria are not culturable, therefore these early studies evaluated only bacteria that are culturable. However, the advances in molecular biology and sequencing technology have revolutionized the microbiome field such that it is now possible to characterize bacteria without culturing. Human studies using high-throughput molecular sequencing methods reveal changes in fecal microbiota composition in CRC subjects compared with healthy controls (145, 166). Although the ease of collection facilitates the evaluation of fecal samples, the luminal contents may not accurately represent the adherent microbiome, since they contain many transient organisms. Several studies have used high-throughput molecular 16S-based methods to examine either the fecal or mucosal microbiome in relation to colorectal adenomas and cancer. Compared with control subjects without adenomas, case subjects with adenomas had significantly increased diversity and richness of bacterial species (130, 141). Chen et al. (26) observed reduced abundance of Clostridium, Roseburia, Eubacteria spp., and other butyrate-producing bacteria in fecal samples of adenoma subjects compared with healthy controls. Marchesi et al. (92) profiled the microbiota in colon tumors and matching normal colon tissues and observed very different microbial patterns and signatures between the two sites. Specifically, they observed an overabundance of Fusobacterium on the tumor compared with matching normal tissue. Sobhani et al. (145) observed that altered fecal bacterial profile was linked with elevated IL-17 in CRC patients compared with healthy controls. A summary of the findings from human studies on gut bacteria and CRC is presented in Table 1. Together, these studies suggest that alterations that favor increased abundance of potentially pathogenic bacteria and reduction of beneficial bacteria are associated with colorectal adenomas and cancer. Although bacterial dysbiosis is associated with CRC, there is limited information on the contribution of specific bacteria. This is currently an area of intense investigation.
Table 1.
Human studies of gut bacteria associated with adenoma and CRC
| Study (author, year, and reference no.) | Sampling Materials and Site | Disease | Findings |
|---|---|---|---|
| Nugent et al. 2014 (109) | Rectal mucosa | Adenoma | Bacterial dysbiosis, altered metabolome, and microbiota in the rectal mucosal tissue of colorectal adenomas compared with nonadenoma controls. |
| Brim et al. 2013 (19a) | Feces | Adenoma | Alterations of microbiota at the subgenus level in colon adenoma compared with normal tissue; however, overall genome existed unaltered. |
| Chen et al. 2013 (26) | Feces | Adenoma | Lower SCFAs production in adenoma groups relative to healthy control groups, reduced butyrate/butyrate-producing bacteria and lower prevalence of Clostridium, Roseburia, and Eubacteria in adenoma cases compared with healthy controls. |
| McCoy et al. 2013 (95a) | Rectal mucosa | Adenoma | Higher relative abundance of Fusobacterium in adenoma subjects. |
| Sanapareddy et al. 2012 (130) | Rectal mucosa | Adenoma | Bacterial dysbiosis, altered diversity, and increased richness. |
| Pagnini et al. 2011 (116) | Biopsies from polyps | Adenoma | Microbial dysbiosis in colonic adenoma, hyper production of α-defensins in adenoma compared with normal tissues |
| Shen et al. 2010 (141) | Colonic mucosa of adenoma/nonadenoma | Adenoma | Bacterial dysbiosis, altered diversity, higher relative. abundance of Proteobacteria and lower relative abundance of Bacteroides in adenoma cases relative to nonadenoma controls. |
| Geng et al. 2014 (46a) | Biopsy samples | Adenoma and CRC | Members of Enterobacteriaceae (7 genera such as Enterobacter, Pseudomonadaceae, Neisseriaceae) as potential bacterial drivers and 12 genera such as Streptococcaceae, Streptophyta, Microbacter, Methylobacter, and Staphylococcus as possible proinflammatory passenger bacteria in adenoma and CRC suggesting bacterial driver-passenger model for CRC. |
| Mira-Pascual et al. In press (99) | Mucosa and feces | Adenoma and CRC | Bacterial dysbiosis, higher abundance of Fusobacterium nucleatum and Enterobacteriaceae in CRC, altered microbial composition in adenoma. |
| Zackular et al. 2014 (175) | Feces | Adenoma and CRC | Microbial dysbiosis, enrichment of pathogenic bacteria in adenoma and CRC, higher relative abundances of Fusobacterium, Porphyromonas, Lachnospiraceae, and Enterobacteriaceae and lower relative abundances of Bacteroides, Lachnospiraceae, Clostridiales, and Clostridium in adenoma and CRC groups compared with healthy groups. |
| Ohigashi et al. 2013 (111a) | Fecal samples from CRC/adenoma/nonadenoma | Adenoma and CRC | Drastic alterations in intestinal environment; altered microbiota (particular decrease in obligate anaerobes), decreased SCFAs, and elevated pH in CRC. |
| Scanlan et al. 2008 (135) | Feces | Adenoma (Polyps) and CRC | Lower temporal stability and altered intestinal microbial diversity and metabolites in polyps/CRC compared with control. Higher relative diversity of the Clostridium leptum and C. coccoides in polyps/CRC relative to healthy controls. |
| Swidsinski et al. 1998 (150) | Biopsy specimens | Adenoma and CRC | Marked abundance of E. coli and coli-like bacteria in the colonic mucosa of patients with colon adenoma and CRC but not detected in normal colonic mucosa. |
| Kubota 1990 (80c) | Feces | Adenoma and CRC | A decrease in Bifidobacterium and Clostridium in colonic adenoma/CRC. |
| Tahara et al. 2014 (151a) | CRC tissues and matching adjacent normal mucosae | CRC | Significant enrichment (250-fold) of F. nucleatum and Pan-fusobacterium in CRC tissues compared with adjacent normal mucosae. Significant abundance of F. nucleatum in genetical susceptible groups such as CIMP, hMLH1, MSI, and CHD7/8. |
| Ahn et al. 2013 (1) | Feces | CRC | Reduced bacterial diversity in CRC cases. |
| Chen et al. 2013 (26) | Feces | CRC | Distinct differences in fecal microbiota communities, Clostridium, Roseburia, and Eubacterium significantly less prevalent, whereas Enterococcus and Streptococcus more prevalent in the CRA group compared with healthy controls. |
| Geng et al. 2013 (46b) | Tumor/matching normal tissue of Chinese CRC patients | CRC | Overabundance of Fusobacterium spp., Roseburia in tumor tissues, and overrepresentation of Microbacterium, Anoxybacillus bacteria away from tumor site. |
| Ohigashi et al. 2013 (111b) | Fecal samples before/after surgery | CRC | Marked decrease of obligate anaerobes, increased pathogenic bacteria, and reduction of SCFA detected after surgery for CRC. |
| Warren et al. 2013 (166a) | CRC/matching normal tissues | CRC | Enriched Fusobacterium, Leptotrichia, and Campylobacter in tumor tissues compared with normal tissues. Bacteria detected in tumors are mostly gram-negative anaerobes. Host proinflammatory genes such as IL-8 overexpressed in tumor tissues having enriched polymicrobial signature. |
| Weir et al. 2013 (166b) | Feces | CRC | Decreased butyrate-producing bacteria in CRC. |
| Wu et al. 2013 (171) | Feces | CRC | Bacterial dysbiosis, altered diversity, enriched Bacteroides, overabundance of Fusobacterium and Campylobacter, and decreased butyrate-producing bacteria in CRC cases compared with healthy controls. |
| Castellarin et al. 2012 (24a) | Tumor/matching normal tissues | CRC | Overabundance of F. nucleatum sequences in tumor tissues relative to matching normal tissues. |
| Kostic et al. 2012 (80a) | Tumor/matching normal tissues | CRC | Altered microbiota, higher relative abundance of Fusobacterium sequences, and lower Bacteroides and Firmicutes sequences in tumors compared with matching normal tissues. |
| Marchesi et al. 2011 (92) | Tumor/matching normal tissues | CRC | Bacterial dysbiosis, high relative abundance of Fusobacterium in tumors relative to normal matching tissues. |
| Sobhani et al. 2011 (145) | Feces | CRC | Bacterial dysbiosis linked with elevated IL-17 in CRC patients. |
SCFA, short-chain fatty acids; CRC, colorectal cancer; CIMP, CpG island methylation; hMLH1, MutL homolog 1; MSI, microsatellite instability; CHD7/8, chromodomain-helicase-DNA-binding protein 7/8; IL, interleukin.
To fully understand the role of gut bacteria in CRC, mechanistic studies in animal models are critical. Vital studies of genetically engineered rodents raised in germ-free environments support the role of bacteria in CRC. For example, IL-10 or transforming growth factor (TGF)-β/Rag2 knockout mice maintained under germ-free conditions do not develop tumors and have significantly lower levels of inflammation (29, 41, 137, 172). Findings from studies in murine models of chemically induced colon cancer indicate that intestinal bacteria promote colon carcinogenesis by increasing proliferation and formation of aberrant crypt foci (40, 115). Horie et al. (65) found that the incidence of tumors in germ-free mice monoassociated with bacteria ranged from 30 to 68% depending on the colonizing bacteria and thus supports the notion that the intestinal microflora has potent effects on cancer development. Another study showed that infection with Helicobacter spp. promoted colon cancer in SMAD3-deficient mice, suggesting that gut bacteria in combination with genetic alterations in the TGF-β pathway may contribute to colon carcinogenesis (90). More recent studies in animal models suggest that the bacterial dysbiosis phenotype is transferable. Transplantation of feces from tumor-bearing mice to conventionalized germ-free mice resulted in an increased colon inflammation and tumorigenesis (175). Similarly, fecal transplants from human CRC patients into germ-free mice resulted in increased tumor burden in mice (14). A summary of the findings from animal studies on gut bacteria and CRC is presented in Table 2. Although these studies in animal models provide compelling evidence to support the contribution of intestinal microbiota to CRC, the relationship is likely to be more complex and may involve interactions between the gut bacteria, chronic inflammation, mutations in oncogenic pathways, and diet to promote colorectal carcinogenesis (63, 69, 108).
Table 2.
Animal studies of gut bacteria associated with CRC
| Study (author, year, and reference no.) | Animal Background and Model | Experimental Design | Findings |
|---|---|---|---|
| Baxter et al. 2014 (14) | GF C57BL/6 mice, AOM/DSS | Fecal microbiota from CRC patients'/control healthy human transplated into GF mice | Gram-negative Bacteroides, Parabacteroides, Alistipes, Akkermansia and mucin-degrading bacteria increase tumor burden, however, butyrate-producing bacteria (Clostridium group XIVa) protects against tumor. |
| Lu et al. 2014 (88a) | SPF C57BL/6 mice, AOM/DSS induced CRC | Salmonella typhimirium AvrA- inflammation-associated CRC | Salmonella-expressing AvrA activates β-catenin, significantly increased CRC. |
| Zhu et al. 2014 (178) | Wister rats, 1,2-dimethylhydrazine (DMH)-induced CRC | Microbial composition in lumen | Altered microbial composition of intestinal lumen in tumor groups, significant difference in intestinal bacterial flora in CRC rats vs. healthy controls, reduction of butyrate-producing bacteria (Roseburia and Eubacterium), significant increase in Desulfovibrio, Erysipelotrichaceae, and Fusobacterium and decrease in probiotic species (Ruminococcus and Lactobacillus) in tumor groups. |
| Kostic et al. 2013 (80b) | C57BL/6-ApcMin/+, BALB/c Il-10−/−, and BALB/c T-bet−/− × Rag2−/− mice | Fusobacterium nucleatum, Streptococcus anginosus, S. parasanguinis, and S. sanguinis | ApcMin/+ mice colonized with F. nucleatum developed a significantly higher number of colonic tumors (but no colitis) compared with ApcMin/+ mice colonized with Streptococcus spp. Higher expression of proinflammatory genes detected in mouse/human tumors with higher abundance of F. nucleatum. |
| Zackular et al. 2013 (174) | SPF/GF C57BL/6-AOM/DSS induced colon tumorigenesis | Microbiome roles in the development of CRC, using fecal transplants from tumor-bearing mice to conven-tionalized GF mice | Abnormal microbial community structure in colonic tumorigenesis in mice. Enriched Bacteroides, Odoribacter, and Akkermansia genera and decreased Prevotellaceae and Porphyromonadaceae families in the colon of tumor-bearing mice. Microbiota transplant from tumor-bearing mice to germ-free mice significantly increased colon tumorigenesis compared with mice colonized with a healthy gut microbiome. Changes in the gut microbiome associated with inflammation directly contribute to tumorigenesis in mice. |
| Arthur et al. 2012 (4) | GF IL10−/− and WT129/SvEv mice, AOM-induced CRC | Genotoxic island polyketide synthase (pks) containing Escherichia coli and pks deleteion E. coli mutant, Enterococcus faecalis | No tumor detected in pks + E. coli-associated WT mice, E. faecalis monoassociated, AOM-treated IL10−/− mice developed severe colitis but rarely adenocarcinoma, pks/colibactin containing E. coli NC101 induced severe inflammation-associated colitis and invasive mucinous adenocarcinoma in AOM-treated IL10−/− mice. |
| Uronis et al. 2009 (161a) | SPF and conventionalized IL10−/−; Myd88−/−, and control IL10−/− C57BL/6, 129ScEv mice, AOM | Bacteroides vulgatus | B. vulgatis (a weak inflammation inducer) colonized with IL10−/− GF WT mice displayed a significantly lower tumor multiplicity compared with their conventionalized counterparts. Bacterial-induced inflammation is essential for the development of colitis-associated colorectal cancer dependent on TLR/MyD88 pathway signaling. |
| Wu et al. 2009 (171a) | Multiple intestinal neoplasia (Min) mice | Enterotoxigenic B. fragilis (ETBF) and Nontoxigenic B. fragilis (NTBF) | Only ETBF triggers inflammatory colitis via BFT and promotes infection-induced colon carcinogenesis. |
| Nagamine et al. 2008 (103a) | BALB/c-Rag2−/−ApcMin/+ mice | Helicobacter hepaticusinfection | Significant increase in colon tumor incidence in H. hepaticus-infected BALB-RagMin mice compared with uninfected BALB-RagMin mice. |
| Nagamine et al. 2008 (103b) | BALB/c-IL10, AOM-induced colon tumorgenesis | H. hepaticus | Significant increase in AOM-induced, exophytic adenomas and colon tumors in IL10 KO mice. |
| Newman et al. 2001 (106a) | ApcMin (Min) C57BL/6J mice, DMH | Citrobacter rodentium infection | C. rodentium infection promotes adenoma formation and enhances carcinogenesis in the colon of genetically susceptible Min mice. |
| Ellmerich et al. 2000 (40) | Wister rats, AOM-induced CRC | S. bovis and S. bovis cell wall-extracted antigens | S. bovis and its cell wall proteins act as promotors for the progression of preneoplastic lesions in colonic mucosa. |
GF, germ free; AvrA, antivirulence gene; SPF, specific pathogen free; WT, wild type; TLR, Toll-like receptor; MyD88, myeloid differentiation primary response gene 88; BFT, Bacteroides fragilis toxin; KO, knockout; DMH, dimethylhydrazine.
Although the exact role of the microbiota and the mechanisms by which it promotes CRC has yet to be fully elucidated, potential mechanisms have been described as follows.
Cell wall antigens and bacterial colicins.
Klein et al. and Huycke et al. (67, 78) showed that Streptococcus gallolyticus (formerly S. bovis) antigens promote premalignant lesions through aberrant crypt formation. Colibactin produced by E. coli strains have been shown to induce DNA double-strand breaks in intestinal cells and trigger chromosomal instability, gene mutations, and cell transformation (4, 14, 26, 108). Cougnoux et al. (33) reported that colibactin-producing E. coli enhanced colon tumor growth in both xenograft and azoxymethane/dextran sodium sulfate IL-10−/− mouse models by inducing a senescence-associated secretory phenotype (4).
Inflammation.
The long-standing presence of infection with potential pathogenic bacteria can induce chronic inflammation. Chronic inflammation in the colorectal mucosa has been linked to CRC development (77, 79, 95, 161).
Production of toxic metabolic byproducts from dietary carbohydrates, protein, bile, and mutagenic precursors.
H2S produced by bacteria in the gut are related to CRC etiology. H2S and reactive oxygen radicals are toxic to the epithelium (5a, 35, 50, 68, 112, 122). Phenolic compounds such as amines, N-nitroso compounds from meat consumption, can also be toxic to the host (91, 143).
This review will focus on inflammation and diet as potential mechanisms, since these risk factors for CRC are well established in the literature.
Inflammation, Intestinal Microbiota, and CRC
The normal colon maintains a continuous state of low-grade inflammation in response to the presence of endogenous bacteria and dietary antigens. Under normal physiological conditions, this low-grade inflammation is maintained by components of the innate and adaptive immune systems that contribute to the homeostasis between proinflammatory mediators [e.g., IL-1B, interferon-γ (IFNγ), IL-8, TNF-α, IL-23, IL-12 IL-17, and IL-6] (24, 100, 111) and anti-inflammatory mediators (e.g., IL-10 and TGF-β). The specific components of the innate immune system that regulate this state of homeostasis include monocytes, macrophages, dendritic cells, and natural killer cells, whereas the adaptive immune system lymphocytes (B cells, T cells, and helper cells) and regulatory cells serve a similar role. Disruption of such inflammatory mediator homeostasis may contribute to chronic inflammation and increased risk of colorectal adenomas and cancer (28, 97, 168).
There is evidence to suggest that chronic inflammation increases colon cancer risk (52, 106, 155). Evidence comes from studies in IBD (34, 86) and colitis in animal models (17, 123). Studies in animal models support the importance of IL-10 in the suppression of chronic inflammation and CRC. In addition, anti-inflammatory agents, such as aspirin and nonsteroidal anti-inflammatory drugs, have been repeatedly shown to decrease the risk of colorectal adenomas and cancer (10, 13, 123).
An important aspect to consider is the resident gut microbiota and their role in inflammation and cancer. Under normal conditions, commensal gut bacteria maintain a symbiotic relationship with the mucosal immune system and are thought to be in a state of tolerance. The presence of commensal bacteria is vital to the development of the gut immune system. For instance, the immune systems of conventionalized animals are highly developed compared with germ-free animals (17, 30, 57, 72). Disruption of the symbiotic relationship between the intestinal microbiota and the host is thought to trigger chronic inflammation. For example, the gut microbiota is a source of antigens for the inflammatory processes associated with IBD (18, 151) and the pathogenesis of enterocolitis and colon cancer in IL-10 knockout mice (114). Recent studies have shown reduced inflammation in IBD through manipulation of the gut microbiota with probiotics (2, 15, 114).
Alterations in the ratio of “harmful” and “beneficial” commensal bacteria contribute to increased mucosal permeability, bacterial translocation, and increased activation of components of the innate and adaptive immune system to promote chronic inflammation (71, 94, 132). Activation of the innate immune system by commensal bacteria (Fig. 1) leads to increased production of proinflammatory cytokines such as IL-12, IL-23, TNF-α, and IFNγ by macrophages, dendritic cells, and natural killer cells, with subsequent induction of the acquired immune system that includes lymphocytes such as T cells, B cells, and various inflammatory mediators. Bacteria may also be responsible in regulating Th17 cells and IL-17 in the lamina propria (71). For example, the presence of segmented filamentous bacteria was shown to promote the accumulation of Th17 cells and increase expression of inflammatory genes in the intestinal mucosa (5, 70). A major consequence of the inflammatory response to commensal bacteria is the activation of key prosurvival and proproliferative signaling pathways by transcription factors such as NF-κB and signal transducer and activator of transcription 3 in epithelial cells (51, 53, 62, 75, 87, 156) as well as generation of reactive oxygen or nitrogen species that leads to oxidative stress, DNA damage, aberrant proliferation, and, ultimately, development of colorectal adenomas and cancer (Fig. 1). Colonization of germ-free animals with Enterococcus faecalis and Bacteroides vulgatus has been observed to induce NF-κB signaling (75, 127) in epithelial cells. Thus, studies to date suggest that disruption of the normal homeostasis between the host and its commensal bacteria community is crucial to inducing inflammation and further downstream changes that promote colon carcinogenesis.
Fig. 1.
Mechanisms of gut microbiome to chronic inflammation, adenoma, and colorectal cancer (CRC). The gut microbiome involves several mechanisms to modulate colorectal carcinogenesis. Bacterial dysbiosis weakens the host defense mechanism of barrier function disruption of epithelial layer tight junctions and the mucus film covering. The action causes increased permeability of the intestinal epithelium and favors bacterial translocation through mucosal epithelium (173). Macrophages on recognition of microorganism-associated molecular patterns (MAMPs) expressed by commensal microbiota, through molecular pattern-recognition receptors (TLR), trigger various signal pathways to produce inflammatory responses (55, 96). Activation of nuclear factor-kB (NF-kB) pathways stimulates the transcription of proinflammatory genes, resulting in increased production of proinflammatory cytokines (TNF, IL-1, IL-23). Proinflammatory cytokines induce signal transducer and activation of transcription 3 (STAT3) and NF-κB signaling, which leads to the suppression of apoptosis and the promotion of cell cycle progression that progress to chronic inflammation and carcinogenic activity (52, 83, 105, 155). During chronic inflammation, there is an overall imbalance in the gut due to production of toxic chemicals or procarcinogens. The production of such chemicals as reactive oxygen species (ROS)/reactive nitrogen species (RNS) from inflammatory cells such as macrophages, as well as bacterial genotoxins (colibactin) and hydrogen sulfide (H2S) from the bacteria, results in oxidative stress which in the target tissue exhorts DNA damage and upshoots DNA repair (46, 167). TLR, Toll-like receptor; TH17, T helper 17; Th, T helper. STAT3, signal transducer and activator of transcription 3.
Intestinal Microbiota and Diet
Diet has been extensively studied in relation to colorectal adenomas and cancer. Diet has a major impact on the composition and activity of intestinal bacteria (38, 119). As such, the link between diet and CRC may be explained, in part, by the activities of the intestinal microbiota. The principal components of dietary intake, carbohydrates, protein, and fat, undergo bacterial digestion to generate byproducts, some of which have potential carcinogenic activity (Fig. 2A). Humans lack the enzymes to digest fiber, but bacteria have glycoside hydrolases and polysaccharide lysases that can ferment plant cell wall polysaccharides (147). One of the major functions of bacteria in the colon is fermentation of nondigestible dietary fiber residues and carbohydrates such as resistant starches, pectins, gums, and cellulose. Fermentation leads to generation of short-chain fatty acids (SCFA) such as butyrate, which serves as a source of energy for colonocytes (121) and demonstrates a protective effect against the development of CRC (22, 36, 124, 125, 148). Butyrate promotes large bowel functions such as modulation of colonic motility, enhanced visceral blood flow, and prevention of potential pathogen overgrowth (19, 54, 85, 89, 158). Butyrate has also been shown to reduce colonic inflammation, induce apoptosis, inhibit tumor cell progression, and protect against development of CRC (19, 54, 60, 85, 117, 136, 177). Increased production of SCFA significantly lowers the intestinal pH, which promotes colonic fermentation and reduced absorption of carcinogens (89), all of which have a potential to reduce CRC risk (88, 153). Diets rich in fiber and complex carbohydrates are thought to have preventive effects against CRC because of the effect of polyphenol metabolites produced by colonic microbiota on cyclooxygenase-2 and glutathione S-transferase θ2 expression in colon cells (98), promotion of colonocyte homeostasis, and DNA repair. The chemopreventive effects of butyrate are mediated through induction of p21 (66) as shown in Fig. 2B.
Fig. 2.
A: roles of various components of diet and bacteria in CRC. The principal components of diet intake are carbohydrates, protein, and fat, which undergo colonic bacterial digestion to generate byproducts that are shown to involve in CRC via various mechanism. Some gut bacteria such as Clostridium spp. convert primary bile acids (intermediate product of fat) into a secondary bile acid such as deoxycholic acid (DCA). DCA is widely considered as a carcinogen that is associated with DNA damage via the production of free radicals or ROS and further induce chronic inflammation and CRC (138). B: metabolism of complex carbohydrates and fibers by colonic bacteria produces the metabolites for the involvement of CRC inhibition. A wide variety of fermentable fibers in the diet include nonstarch polysaccharides (cellulose, pectins, gums, arabinoxylans, mucilages, insulin, galacto-oligosaccharides), carbohydrate fibers (resistant starches, dextrins), and lignans (waxes, tannins). Fermentation of these complex carbohydrates and fibers by gut bacteria results in the production of short-chain fatty acids (SCFAs) such as formic acid, acetic acid, propionic acid, butyric acid, and valeric acid. SCFAs promote large-bowel functions modulating colonic motility, promoting visceral blood flow, and preventing potential pathogen overgrowth (153, 158). Butyric acid or butyrate promotes colonic fermentation, lowers the absorption of procarcinogens, protects against inflammation, and reduces CRC risk (162). Butyrate can also act as a histone deactylase (HDAC), inhibiting the function of HDAC thereby favoring an acetylated state of histone in the cell (60). HDAC inhibition via SCFAs results in upregulation of p21, a key regulatory molecule of cell cycle arrest that is also involved in cell proliferation, differentiation, and apoptosis (27, 66). SCFAs enhance the barrier functions promoting epithelial cell attachment to the basement while suppressing type IV collagen activity. All these mechanisms involve maintaining gut homeostasis and CRC inhibition. C: breakdown of protein by colonic bacteria to potential carcinogenic metabolites and CRC. (modified from Ref. 89). The breakdown of proteins and peptides by colonic microorganisms also generate end products that are considered as procarcinogenic, mutagenic, and genotoxic besides health-benefiting SCFAs and energy. A variety of branched-chain fatty acids (BCFAs), such as isobutyrate, 2-methylbutyrate, and isovalerate, are usually one carbon shorter than their respective amino acid precursors valine, isoleucine, and leucine. The BCFAs are mainly produced in the colon via digestion by various gut bacteria; the major contributor is Clostridia (89). BCFAs are not considered to be risk factors for CRC. Ammonia, phenol, and H2S produced during amino acid fermentation are putrefactive toxic substances. Amines (end product of amino acids via deamination) act as mutagen-precursors, and phenols and indoles act as procarcinogens (89). Thioles and H2S, also numerously reported as genetoxic, can release ROS, damage host DNA, and induce CRC.
With increased protein ingestion, the colonic residue contains more sulfur, nitrates, ammonia, amines, branched-chain amino acids, and H2S. Residues from protein digestion can stimulate growth of sulfur-reducing bacteria such as Desulfovibrio and Desulfomonas spp. (110). H2S, an end product of protein metabolism, is proinflammatory and genotoxic (50). Many of the end products of protein catabolism are mutagens and genotoxic and are associated with development of CRC via genetic mutations summarized in Fig. 2C. Increased intake of red or processed meats has generally been shown to be a risk factor for development of colorectal adenomas and cancer (81, 107, 131). Bacteria metabolize meat proteins to produce nitrosamines, known promoters of colon tumors in animal models (31).
Bile acids are byproducts of fat metabolism. Secondary bile acids such as deoxycholic acid (DCA) contribute to increased reactive oxygen species (ROS), DNA damage, genomic instability, and tumor growth (39, 47) (Fig. 2A). Bacteria such as Clostridium spp. convert bile acids into secondary bile acids such as DCA to generate free radicals and induce chronic inflammation, ROS, and CRC (8). The high-fat, low-fiber (low intake of fruits and vegetables) diet in Western countries is thought to increase the risk of colon cancer possibly through increased secondary bile acids and ROS, which induces DNA damage and genomic instability. Consumers of a Western diet rich in fat and protein have a higher proportion of inflammatory 7-α-dehydroxylating bacteria and sulfur-reducing bacteria producing H2S and secondary bile acids, respectively (50).
Together, studies suggest that byproducts of carbohydrate protein and fat metabolism may contribute to colorectal carcinogenesis through a variety of mechanisms. Additional studies in humans and animal models are needed to fully understand the mechanisms underlying the interactions of diet, microbiota, and colorectal adenomas and cancer. In particular, studies are needed to characterize the functional aspects of the microbiome. In a recent study Faith et al. (42), employed this approach to evaluate the interrelationship between diet and human gut bacteria. They evaluated germ-free mice colonized with a model bacterial community (10 defined human bacteria phylotypes) and four different diets. They found that certain factors in the diet influenced bacterial community membership. This type of approach will not only help to define underlying mechanisms but also identify metabolic functions, host-microbe, and microbe-microbe interactions as well as develop ways to manipulate the gut microbiota to prevent or treat CRC.
Summary and Future Directions
Until recently, much of our understanding of the complexity and diversity of the microorganisms in the gastrointestinal tract relied on observations from microbiological culture. However, the development of culture-independent molecular methods based on the highly conserved bacterial 16S ribosomal RNA gene and advances in high-throughput sequencing technology have allowed more in-depth probing of the human gut microbiota (58, 82, 93, 149). Several studies have demonstrated an association of bacterial dysbiosis with development of colorectal adenomas and cancer. However, specific causative bacteria have yet to be identified. Microbial signatures of colorectal adenomas and cancer identified in human studies will need to be verified in animal studies using humanized and gnotobiotic mice. Similarly, mouse models can be employed to investigate the potential protective role of specific microbial groups on CRC onset.
A better understanding of the complex interactions between the microbiome and development of CRC will require system-based approaches that incorporate metagenomics, metabolomics, proteomics, and transcriptomics studies. Such an approach will facilitate the identification of possible interactions between the intestinal microbiota, procarcinogenic factors, and CRC.
Challenges in CRC include identifying at-risk individuals and early detection. While no specific bacteria have been identified as a causative agent for CRC, ascertaining a bacterial signature for development of adenomas and cancer holds great promise for providing novel preventive and therapeutic strategies. It is conceivable that an intestinal bacterial profile might 1) predict an individual's risk of developing adenomas or CRC and 2) allow manipulation of the microbiota for prevention or treatment. Modulation of the human gut microbiota will likely be accomplished with pro- and prebiotics, although other agents may also be beneficial. Large-scale clinical trials will be required to introduce such therapeutic or preventive interventions.
To understand the relationship between intestinal microbiota and development of CRC in humans, epidemiological studies are needed to verify the findings from animal models in humans. Ideally, these studies should be conducted in various populations, with a sufficient sample size that would allow meaningful examination of the contribution of the microbiota to disease after taking into account confounders (e.g., age and gender) and interaction effects with diet. The studies should be prospective to provide information on temporal sequence. Moreover, these studies will require interdisciplinary collaborations with experts in biostatistics, bioinformatics, microbiology, molecular biology epidemiology, and others to analyze and interpret the massive microbial data generated by the high-throughput sequencing technologies.
Conclusions
The human gut microbiome and its role in CRC and other diseases is an active field of research. While initial correlative studies have informed us on the question of: “Who is there?” they also raise questions concerning the functional aspects of the gut microbiota and the exact means by which they influence human disease. These initial studies are likely to give rise to mechanistic studies that will explore host-microbe interactions, microbe-microbe interactions, as well as studies on modulation of the gut microbiome to prevent disease. Continued funding in microbiome research could potentially increase the number of significant discoveries in the microbiome field and provide much needed mechanistic insights that could contribute to disease prevention and treatment.
GRANTS
This work was supported by funding from the National Institutes of Health: P50-CA-106991, R01-CA-136887, R01-CA-44684, and P30-DK-034987.
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
Author contributions: T.O.K., B.J., and X.H. conception and design of research; T.O.K., S.D., and A.D. prepared figures; T.O.K., S.D., A.D., B.J., and X.H. edited and revised manuscript; T.O.K., S.D., A.D., B.J., and X.H. approved final version of manuscript; A.D., B.J., and X.H. drafted manuscript.
ACKNOWLEDGMENTS
We thank Drs. Robert Sandler, Gary Asher, and Andrea Azcarate-Peril for helpful suggestions. We also thank Amber McCoy for editorial assistance.
REFERENCES
- 1.Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, Shi J, Goedert JJ, Hayes RB, Yang L. Human gut microbiome and risk for colorectal cancer. J Natl Cancer Inst 105: 1907–1911, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Araki Y, Andoh A, Takizawa J, Takizawa W, Fujiyama Y. Clostridium butyricum, a probiotic derivative, suppresses dextran sulfate sodium-induced experimental colitis in rats. Int J Mol Med 13: 577–580, 2004. [PubMed] [Google Scholar]
- 3.Arends MJ. Pathways of colorectal carcinogenesis. Appl Immunohistochem Mol Mmorphol 21: 97–102, 2013. [DOI] [PubMed] [Google Scholar]
- 4.Arthur JC, Perez-Chanona E, Muhlbauer M, Tomkovich S, Uronis JM, Fan TJ, Campbell BJ, Abujamel T, Dogan B, Rogers AB, Rhodes JM, Stintzi A, Simpson KW, Hansen JJ, Keku TO, Fodor AA, Jobin C. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 338: 120–123, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, Cheng G, Yamasaki S, Saito T, Ohba Y, Taniguchi T, Takeda K, Hori S, Ivanov II, Umesaki Y, Itoh K, Honda K. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331: 337–341, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5a.Attene-Ramos MS, Wagner ED, Plewa MJ, Gaskins HR. Evidence that hydrogen sulfide is a genotoxic agent. Mol Cancer Res 4: 9–14, 2006. [DOI] [PubMed] [Google Scholar]
- 6.Aune D, Chan DS, Lau R, Vieira R, Greenwood DC, Kampman E, Norat T. Dietary fibre, whole grains, and risk of colorectal cancer: systematic review and dose-response meta-analysis of prospective studies. BMJ 343: d6617, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Azad MB, Konya T, Maughan H, Guttman DS, Field CJ, Sears MR, Becker AB, Scott JA, Kozyrskyj AL. Infant gut microbiota and the hygiene hypothesis of allergic disease: impact of household pets and siblings on microbiota composition and diversity (Abstract). Allergy Asthma Clin Immunol 9: 15, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Azcarate-Peril MA, Sikes M, Bruno-Barcena JM. The intestinal microbiota, gastrointestinal environment and colorectal cancer: a putative role for probiotics in prevention of colorectal cancer? Am J Physiol Gastrointest Liver Physiol 301: G401–G424, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science 307: 1915–1920, 2005. [DOI] [PubMed] [Google Scholar]
- 10.Backlund MG, Mann JR, Dubois RN. Mechanisms for the prevention of gastrointestinal cancer: the role of prostaglandin E2. Oncology 69, Suppl 1: 28–32, 2005. [DOI] [PubMed] [Google Scholar]
- 11.Bajzer M, Seeley RJ. Physiology: obesity and gut flora. Nature 444: 1009–1010, 2006. [DOI] [PubMed] [Google Scholar]
- 12.Barman M, Unold D, Shifley K, Amir E, Hung K, Bos N, Salzman N. Enteric salmonellosis disrupts the microbial ecology of the murine gastrointestinal tract. Infect Immun 76: 907–915, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Baron JA, Cole BF, Sandler RS, Haile RW, Ahnen D, Bresalier R, McKeown-Eyssen G, Summers RW, Rothstein R, Burke CA, Snover DC, Church TR, Allen JI, Beach M, Beck GJ, Bond JH, Byers T, Greenberg ER, Mandel JS, Marcon N, Mott LA, Pearson L, Saibil F, van Stolk RU. A randomized trial of aspirin to prevent colorectal adenomas. N Engl J Med 348: 891–899, 2003. [DOI] [PubMed] [Google Scholar]
- 14.Baxter NT, Zackular JP, Chen GY, Schloss PD. Structure of the gut microbiome following colonization with human feces determines colonic tumor burden (Abstract). Microbiome 2: 20, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bengmark S. Bioecological control of inflammatory bowel disease. Clin Nutr 26: 169–181, 2007. [DOI] [PubMed] [Google Scholar]
- 16.Benson AK, Kelly SA, Legge R, Ma F, Low SJ, Kim J, Zhang M, Oh PL, Nehrenberg D, Hua K, Kachman SD, Moriyama EN, Walter J, Peterson DA, Pomp D. Individuality in gut microbiota composition is a complex polygenic trait shaped by multiple environmental and host genetic factors. Proc Natl Acad Sci USA 107: 18933–18938, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Berg DJ, Davidson N, Kuhn R, Muller W, Menon S, Holland G, Thompson-Snipes L, Leach MW, Rennick D. Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4(+) TH1-like responses. J Clin Invest 98: 1010–1020, 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Bibiloni R, Mangold M, Madsen KL, Fedorak RN, Tannock GW. The bacteriology of biopsies differs between newly diagnosed, untreated, Crohn's disease and ulcerative colitis patients. J Med Microbiol 55: 1141–1149, 2006. [DOI] [PubMed] [Google Scholar]
- 19.Blottiere HM, Buecher B, Galmiche JP, Cherbut C. Molecular analysis of the effect of short-chain fatty acids on intestinal cell proliferation. Proc Nutr Soc 62: 101–106, 2003. [DOI] [PubMed] [Google Scholar]
- 19a.Brim H, Yooseph S, Zoetendal EG, Lee E, Torralbo M, Laiyemo AO, Shokrani B, Nelson K, Ashktorab H. Microbiome analysis of stool samples from african americans with colon polyps. PloS One 8: e81352, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Brown K, DeCoffe D, Molcan E, Gibson DL. Diet-induced dysbiosis of the intestinal microbiota and the effects on immunity and disease. Nutrients 4: 1095–1119, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Campbell JH, Foster CM, Vishnivetskaya T, Campbell AG, Yang ZK, Wymore A, Palumbo AV, Chesler EJ, Podar M. Host genetic and environmental effects on mouse intestinal microbiota. ISME J 6: 2033–2044, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Canani RB, Costanzo MD, Leone L, Pedata M, Meli R, Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol 17: 1519–1528, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Carethers JM. DNA testing and molecular screening for colon cancer. Clin Gastroenterol Hepatol 12: 377–381, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Carol M, Lambrechts A, Van Gossum A, Libin M, Goldman M, Mascart-Lemone F. Spontaneous secretion of interferon gamma and interleukin 4 by human intraepithelial and lamina propria gut lymphocytes. Gut 42: 643–649, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24a.Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, Barnes R, Watson P, Allen-Vercoe E, Moore RA, Holt RA. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res 22: 299–306, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chang CL, Marra G, Chauhan DP, Ha HT, Chang DK, Ricciardiello L, Randolph A, Carethers JM, Boland CR. Oxidative stress inactivates the human DNA mismatch repair system. Am J Physiol Cell Physiol 283: C148–C154, 2002. [DOI] [PubMed] [Google Scholar]
- 26.Chen HM, Yu YN, Wang JL, Lin YW, Kong X, Yang CQ, Yang L, Liu ZJ, Yuan YZ, Liu F, Wu JX, Zhong L, Fang DC, Zou W, Fang JY. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr 97: 1044–1052, 2013. [DOI] [PubMed] [Google Scholar]
- 27.Chen YX, Fang JY, Lu J, Qiu DK. Regulation of histone acetylation on the expression of cell cycle-associated genes in human colon cancer cell lines. Zhonghua yi xue za zhi 84: 312–317, 2004. [PubMed] [Google Scholar]
- 28.Chow J, Mazmanian SK. A pathobiont of the microbiota balances host colonization and intestinal inflammation. Cell Host Microbe 7: 265–276, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chu FF, Esworthy RS, Chu PG, Longmate JA, Huycke MM, Wilczynski S, Doroshow JH. Bacteria-induced intestinal cancer in mice with disrupted Gpx1 and Gpx2 genes. Cancer Res 64: 962–968, 2004. [DOI] [PubMed] [Google Scholar]
- 30.Chung H, Pamp SJ, Hill JA, Surana NK, Edelman SM, Troy EB, Reading NC, Villablanca EJ, Wang S, Mora JR, Umesaki Y, Mathis D, Benoist C, Relman DA, Kasper DL. Gut immune maturation depends on colonization with a host-specific microbiota. Cell 149: 1578–1593, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Clinton SK, Bostwick DG, Olson LM, Mangian HJ, Visek WJ. Effects of ammonium acetate and sodium cholate on N-methyl-N′-nitro-N-nitrosoguanidine-induced colon carcinogenesis of rats. Cancer Res 48: 3035–3039, 1988. [PubMed] [Google Scholar]
- 32.Collins SM. A role for the gut microbiota in IBS. Nat Rev Gastroenterol Hepatol 11: 497–505, 2014. [DOI] [PubMed] [Google Scholar]
- 33.Cougnoux A, Dalmasso G, Martinez R, Buc E, Delmas J, Gibold L, Sauvanet P, Darcha C, Dechelotte P, Bonnet M, Pezet D, Wodrich H, Darfeuille-Michaud A, Bonnet R. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut 63: 1932–1942, 2014. [DOI] [PubMed] [Google Scholar]
- 34.Coussens LM, Werb Z. Inflammation and cancer. Nature 420: 860–867, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Deplancke B, Gaskins HR. Hydrogen sulfide induces serum-independent cell cycle entry in nontransformed rat intestinal epithelial cells. FASEB J 17: 1310–1312, 2003. [DOI] [PubMed] [Google Scholar]
- 36.Di Sabatino A, Morera R, Ciccocioppo R, Cazzola P, Gotti S, Tinozzi FP, Tinozzi S, Corazza GR. Oral butyrate for mildly to moderately active Crohn's disease. Aliment Pharmacol Ther 22: 789–794, 2005. [DOI] [PubMed] [Google Scholar]
- 37.Dulal S, Keku TO. Gut microbiome and colorectal adenomas. Cancer J 20: 225–231, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Duncan SH, Belenguer A, Holtrop G, Johnstone AM, Flint HJ, Lobley GE. Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces. Appl Environ Microbiol 73: 1073–1078, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Dvorak K, Payne CM, Chavarria M, Ramsey L, Dvorakova B, Bernstein H, Holubec H, Sampliner RE, Guy N, Condon A, Bernstein C, Green SB, Prasad A, Garewal HS. Bile acids in combination with low pH induce oxidative stress and oxidative DNA damage: relevance to the pathogenesis of Barrett's oesophagus. Gut 56: 763–771, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ellmerich S, Scholler M, Duranton B, Gosse F, Galluser M, Klein JP, Raul F. Promotion of intestinal carcinogenesis by Streptococcus bovis. Carcinogenesis 21: 753–756, 2000. [DOI] [PubMed] [Google Scholar]
- 41.Engle SJ, Ormsby I, Pawlowski S, Boivin GP, Croft J, Balish E, Doetschman T. Elimination of colon cancer in germ-free transforming growth factor beta 1-deficient mice. Cancer Res 62: 6362–6366, 2002. [PubMed] [Google Scholar]
- 42.Faith JJ, McNulty NP, Rey FE, Gordon JI. Predicting a human gut microbiota's response to diet in gnotobiotic mice. Science 333: 101–104, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Fan W, Huo G, Li X, Yang L, Duan C. Impact of diet in shaping gut microbiota revealed by a comparative study in infants during the six months of life. J Microbiol Biotechnol 24: 133–143, 2014. [DOI] [PubMed] [Google Scholar]
- 44.Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 61: 759–767, 1990. [DOI] [PubMed] [Google Scholar]
- 45.Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer Incidence and Mortality Worldwide: IARC Cancer Base No. 11 (Internet) Lyon, France: Int Agency Res Cancer; Available from: http://globocan.iarc.fr, 2013. [Google Scholar]
- 46.Flood DM, Weiss NS, Cook LS, Emerson JC, Schwartz SM, Potter JD. Colorectal cancer incidence in Asian migrants to the United States and their descendants. Cancer Causes Control 11: 403–411, 2000. [DOI] [PubMed] [Google Scholar]
- 46a.Geng J, Song Q, Tang X, Liang X, Fan H, Peng H, Guo Q, Zhang Z. Co-occurrence of driver and passenger bacteria in human colorectal cancer (Abstract). Gut Pathogens 6: 26, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46b.Geng J, Fan H, Tang X, Zhai H, Zhang Z. Diversified pattern of the human colorectal cancer microbiome (Abstract). Gut Pathogens 5: 2, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Gill CI, Rowland IR. Diet and cancer: assessing the risk. Br J Nutr 88, Suppl 1: S73–S87, 2002. [DOI] [PubMed] [Google Scholar]
- 48.Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, Gordon JI, Relman DA, Fraser-Liggett CM, Nelson KE. Metagenomic analysis of the human distal gut microbiome. Science 312: 1355–1359, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Grady WM, Carethers JM. Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology 135: 1079–1099, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Greer JB, O'Keefe SJ. Microbial induction of immunity, inflammation, and cancer (Abstract). Front Physiol 1: 168, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, Kagnoff MF, Karin M. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 118: 285–296, 2004. [DOI] [PubMed] [Google Scholar]
- 52.Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, cancer. Cell 140: 883–899, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Guarner F. Enteric flora in health and disease. Digestion 73, Suppl 1: 5–12, 2006. [DOI] [PubMed] [Google Scholar]
- 54.Guilloteau P, Martin L, Eeckhaut V, Ducatelle R, Zabielski R, Van Immerseel F. From the gut to the peripheral tissues: the multiple effects of butyrate. Nutr Res Rev 23: 366–384, 2010. [DOI] [PubMed] [Google Scholar]
- 55.Hajishengallis G, Lambris JD. Microbial manipulation of receptor crosstalk in innate immunity. Nat Rev Immunol 11: 187–200, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Hallouin F, Goupille C, Bureau V, Meflah K, Le Pendu J. Increased tumorigenicity of rat colon carcinoma cells after alpha1,2-fucosyltransferase FTA anti-sense cDNA transfection. Int J Cancer 80: 606–611, 1999. [DOI] [PubMed] [Google Scholar]
- 57.Hansen CH, Nielsen DS, Kverka M, Zakostelska Z, Klimesova K, Hudcovic T, Tlaskalova-Hogenova H, Hansen AK. Patterns of early gut colonization shape future immune responses of the host. PLos One 7: e34043, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hayashi H, Sakamoto M, Benno Y. Fecal microbial diversity in a strict vegetarian as determined by molecular analysis and cultivation. Microbiol Immunol 46: 819–831, 2002. [DOI] [PubMed] [Google Scholar]
- 59.Hill DA, Hoffmann C, Abt MC, Du Y, Kobuley D, Kirn TJ, Bushman FD, Artis D. Metagenomic analyses reveal antibiotic-induced temporal and spatial changes in intestinal microbiota with associated alterations in immune cell homeostasis. Mucosal Immunol 3: 148–158, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hinnebusch BF, Meng S, Wu JT, Archer SY, Hodin RA. The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation. J Nutr 132: 1012–1017, 2002. [DOI] [PubMed] [Google Scholar]
- 61.Hold GL, Smith M, Grange C, Watt ER, El-Omar EM, Mukhopadhya I. Role of the gut microbiota in inflammatory bowel disease pathogenesis: what have we learnt in the past 10 years? World J Gastroenterol 20: 1192–1210, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, Gordon JI. Molecular analysis of commensal host-microbial relationships in the intestine. Science 291: 881–884, 2001. [DOI] [PubMed] [Google Scholar]
- 63.Hope ME, Hold GL, Kain R, El-Omar EM. Sporadic colorectal cancer–role of the commensal microbiota. FEMS Microbiol Lett 244: 1–7, 2005. [DOI] [PubMed] [Google Scholar]
- 64.Hopkins MJ, Macfarlane GT. Changes in predominant bacterial populations in human faeces with age and with Clostridium difficile infection. J Med Microbiol 51: 448–454, 2002. [DOI] [PubMed] [Google Scholar]
- 65.Horie H, Kanazawa K, Okada M, Narushima S, Itoh K, Terada A. Effects of intestinal bacteria on the development of colonic neoplasm: an experimental study. Eur J Cancer Prev 8: 237–245, 1999. [DOI] [PubMed] [Google Scholar]
- 66.Hu S, Dong TS, Dalal SR, Wu F, Bissonnette M, Kwon JH, Chang EB. The microbe-derived short chain fatty acid butyrate targets miRNA-dependent p21 gene expression in human colon cancer. PLos One 6: e16221, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Huycke MM, Abrams V, Moore DR. Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis 23: 529–536, 2002. [DOI] [PubMed] [Google Scholar]
- 68.Huycke MM, Gaskins HR. Commensal bacteria, redox stress, and colorectal cancer: mechanisms and models. Exp Biol Med (Maywood) 229: 586–597, 2004. [DOI] [PubMed] [Google Scholar]
- 69.Irrazabal T, Belcheva A, Girardin SE, Martin A, Philpott DJ. The multifaceted role of the intestinal microbiota in colon cancer. Mol Cell 54: 309–320, 2014. [DOI] [PubMed] [Google Scholar]
- 70.Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, Tanoue T, Imaoka A, Itoh K, Takeda K, Umesaki Y, Honda K, Littman DR. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139: 485–498, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ivanov II, Frutos Rde L, Manel N, Yoshinaga K, Rifkin DB, Sartor RB, Finlay BB, Littman DR. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 4: 337–349, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ivanov II, Zhou L, Littman DR. Transcriptional regulation of Th17 cell differentiation. Semin Immunol 19: 409–417, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Jimenez E, Fernandez L, Marin ML, Martin R, Odriozola JM, Nueno-Palop C, Narbad A, Olivares M, Xaus J, Rodriguez JM. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Curr Microbiol 51: 270–274, 2005. [DOI] [PubMed] [Google Scholar]
- 74.Jones MLMC, Ganopolsky JG, Labbé A, Prakash S. The human microbiome and bile acid metabolism: dysbiosis, dysmetabolism, disease and intervention. Expert Opin Biol Ther 14: 467–482, 2014. [DOI] [PubMed] [Google Scholar]
- 75.Karrasch T, Kim JS, Muhlbauer M, Magness ST, Jobin C. Gnotobiotic IL-10−/−;NF-kappa B(EGFP) mice reveal the critical role of TLR/NF-kappa B signaling in commensal bacteria-induced colitis. J Immunol 178: 6522–6532, 2007. [DOI] [PubMed] [Google Scholar]
- 76.Kelly RJ, Rouquier S, Giorgi D, Lennon GG, Lowe JB. Sequence and expression of a candidate for the human Secretor blood group alpha(1,2)fucosyltransferase gene (FUT2). Homozygosity for an enzyme-inactivating nonsense mutation commonly correlates with the non-secretor phenotype. J Biol Chem 270: 4640–4649, 1995. [DOI] [PubMed] [Google Scholar]
- 77.Kim S, Keku TO, Martin C, Galanko J, Woosley JT, Schroeder JC, Satia JA, Halabi S, Sandler RS. Circulating levels of inflammatory cytokines and risk of colorectal adenomas. Cancer Res 68: 323–328, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Klein RS, Recco RA, Catalano MT, Edberg SC, Casey JI, Steigbigel NH. Association of Streptococcus bovis with carcinoma of the colon. N Engl J Med 297: 800–802, 1977. [DOI] [PubMed] [Google Scholar]
- 79.Kosa P, Szabo R, Molinolo AA, Bugge TH. Suppression of tumorigenicity-14, encoding matriptase, is a critical suppressor of colitis and colitis-associated colon carcinogenesis. Oncogene 31: 3679–3695, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 146: 1489–1499, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80a.Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, Ojesina AI, Jung J, Bass AJ, Tabernero J, Baselga J, Shivdasani RA, Ogino S, Birren BW, Huttenhower C, Garrett WS, Meyerson M. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res 22: 292–298, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80b.Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, Clancy TE, Chung DC, Lochhead P, Hold GL, El-Omar EM, Brenner D, Fuchs CS, Meyerson M, Garrett WS. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 14: 207–215, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80c.Kubota Y. Fecal intestinal flora in patients with colon adenoma and colon cancer. Japn J Gastroenterol 87: 771–779, 1990. [PubMed] [Google Scholar]
- 81.Larsson SC, Wolk A. Meat consumption and risk of colorectal cancer: a meta-analysis of prospective studies. Int J Cancer 119: 2657–2664, 2006. [DOI] [PubMed] [Google Scholar]
- 82.Lawson PA. Taxonomic and systematics of predominant gut anaerobes. Dordrcht. New York, NY: Kluwer, 1999, p. 149–166. [Google Scholar]
- 83.Lee H, Herrmann A, Deng JH, Kujawski M, Niu G, Li Z, Forman S, Jove R, Pardoll DM, Yu H. Persistently activated Stat3 maintains constitutive NF-kappaB activity in tumors. Cancer Cell 15: 283–293, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in human cancers. Nature 396: 643–649, 1998. [DOI] [PubMed] [Google Scholar]
- 85.Leonel AJ, Alvarez-Leite JI. Butyrate: implications for intestinal function. Curr Opin Clin Nutr Metab Care 15: 474–479, 2012. [DOI] [PubMed] [Google Scholar]
- 86.Levin B. Ulcerative colitis and colon cancer: biology and surveillance. J Cell Biochem 16G: 47–50, 1992. [DOI] [PubMed] [Google Scholar]
- 87.Li Y, Kundu P, Seow SW, de Matos CT, Aronsson L, Chin KC, Karre K, Pettersson S, Greicius G. Gut microbiota accelerate tumor growth via c-jun and STAT3 phosphorylation in APCMin/+ mice. Carcinogenesis 33: 1231–1238, 2012. [DOI] [PubMed] [Google Scholar]
- 88.Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev 12: 661–672, 2014. [DOI] [PubMed] [Google Scholar]
- 88a.Lu R, Wu S, Zhang YG, Xia Y, Liu X, Zheng Y, Chen H, Schaefer KL, Zhou Z, Bissonnette M, Li L, Sun J. Enteric bacterial protein AvrA promotes colonic tumorigenesis and activates colonic beta-catenin signaling pathway (Abstract). Oncogenesis 3: e105, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Macfarlane GT, Macfarlane S. Bacteria, colonic fermentation, and gastrointestinal health. J AOAC Int 95: 50–60, 2012. [DOI] [PubMed] [Google Scholar]
- 90.Maggio-Price L, Treuting P, Zeng W, Tsang M, Bielefeldt-Ohmann H, Iritani BM. Helicobacter infection is required for inflammation and colon cancer in SMAD3-deficient mice. Cancer Res 66: 828–838, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Manning TS, Gibson GR. Microbial-gut interactions in health and disease. Prebiotics Best Pract Res 18: 287–298, 2004. [DOI] [PubMed] [Google Scholar]
- 92.Marchesi JR, Dutilh BE, Hall N, Peters WH, Roelofs R, Boleij A, Tjalsma H. Towards the human colorectal cancer microbiome. PLos One 6: e20447, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Marsh TL, Saxman P, Cole J, Tiedje J. Terminal restriction fragment length polymorphism analysis program, a web-based research tool for microbial community analysis. Appl Environ Microbiol 66: 3616–3620, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Marteau P, Seksik P, Shanahan F. Manipulation of the bacterial flora in inflammatory bowel disease. Best Pract Res 17: 47–61, 2003. [DOI] [PubMed] [Google Scholar]
- 95.McClellan JL, Davis JM, Steiner JL, Day SD, Steck SE, Carmichael MD, Murphy EA. Intestinal inflammatory cytokine response in relation to tumorigenesis in the Apc(Min/+) mouse. Cytokine 57: 113–119, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95a.McCoy AN, Araujo-Perez F, Azcarate-Peril A, Yeh JJ, Sandler RS, Keku TO. Fusobacterium is associated with colorectal adenomas. PloS One 8: e53653, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Medzhitov R, Janeway CA Jr. Decoding the patterns of self and nonself by the innate immune system. Science 296: 298–300, 2002. [DOI] [PubMed] [Google Scholar]
- 97.Meira LB, Bugni JM, Green SL, Lee CW, Pang B, Borenshtein D, Rickman BH, Rogers AB, Moroski-Erkul CA, McFaline JL, Schauer DB, Dedon PC, Fox JG, Samson LD. DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice. J Clin Invest 118: 2516–2525, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Miene C, Weise A, Glei M. Impact of polyphenol metabolites produced by colonic microbiota on expression of COX-2 and GSTT2 in human colon cells (LT97). Nutr Cancer 63: 653–662, 2011. [DOI] [PubMed] [Google Scholar]
- 99.Mira-Pascual L, Cabrera-Rubio R, Ocon S, Costales P, Parra A, Suarez A, Moris F, Rodrigo L, Mira A, Collado MC. Microbial mucosal colonic shifts associated with the development of colorectal cancer reveal the presence of different bacterial and archaeal biomarkers. J Gastroenterol In press. [DOI] [PubMed] [Google Scholar]
- 100.Monteleone I, Vavassori P, Biancone L, Monteleone G, Pallone F. Immunoregulation in the gut: success and failures in human disease. Gut 50, Suppl 3: III60–III64, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Moore WE, Moore LH. Intestinal floras of populations that have a high risk of colon cancer. Appl Environ Microbiol 61: 3202–3207, 1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Moradi T, Delfino RJ, Bergstrom SR, Yu ES, Adami HO, Yuen J. Cancer risk among Scandinavian immigrants in the US and Scandinavian residents compared with US whites, 1973–89. Eur J Cancer Prev 7: 117–125, 1998. [PubMed] [Google Scholar]
- 103.Moschen AR, Wieser V, Tilg H. Dietary factors: major regulators of the gut's microbiota. Gut Liver 6: 411–416, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103a.Nagamine CM, Sohn JJ, Rickman BH, Rogers AB, Fox JG, Schauer DB. Helicobacter hepaticus infection promotes colon tumorigenesis in the BALB/c-Rag2(−/−) Apc(Min/+) mouse. Infect Immun 76: 2758–2766, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103b.Nagamine CM, Rogers AB, Fox JG, Schauer DB. Helicobacter hepaticus promotes azoxymethane-initiated colon tumorigenesis in BALB/c-IL10-deficient mice. Int J Cancer 122: 832–838, 2008. [DOI] [PubMed] [Google Scholar]
- 104.Nakamura Y, Nishisho I, Kinzler KW, Vogelstein B, Miyoshi Y, Miki Y, Ando H, Horii A, Nagase H. Mutations of the adenomatous polyposis coli gene in familial polyposis coli patients and sporadic colorectal tumors. Princess Takamatsu Symp 22: 285–292, 1991. [PubMed] [Google Scholar]
- 105.Naugler WE, Karin M. NF-kappaB and cancer-identifying targets and mechanisms. Curr Opin Genet Dev 18: 19–26, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Naugler WE, Karin M. The wolf in sheep's clothing: the role of interleukin-6 in immunity, inflammation and cancer. Trends Mol Med 14: 109–119, 2008. [DOI] [PubMed] [Google Scholar]
- 106a.Newman JV, Kosaka T, Sheppard BJ, Fox JG, Schauer DB. Bacterial infection promotes colon tumorigenesis in Apc(Min/+) mice. J Infect Dis 184: 227–230, 2001. [DOI] [PubMed] [Google Scholar]
- 107.Norat T, Lukanova A, Ferrari P, Riboli E. Meat consumption and colorectal cancer risk: dose-response meta-analysis of epidemiological studies. Int J Cancer 98: 241–256, 2002. [DOI] [PubMed] [Google Scholar]
- 108.Nougayrede JP, Oswald E. Microbiota and colorectal cancer: genotoxic bacteria in the intestinal tract. Bull l'Acad Natl Med 195: 1295–1304, 2011. [PubMed] [Google Scholar]
- 109.Nugent JL, McCoy AN, Addamo CJ, Jia W, Sandler RS, Keku TO. Altered tissue metabolites correlate with microbial dysbiosis in colorectal adenomas. J Proteome Res 13: 1921–1929, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Nyangale EP, Mottram DS, Gibson GR. Gut microbial activity, implications for health and disease: the potential role of metabolite analysis. J Proteome Res 11: 5573–5585, 2012. [DOI] [PubMed] [Google Scholar]
- 111.O'Farrelly C. Just how inflamed is the normal gut? Gut 42: 603–604, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111a.Ohigashi S, Sudo K, Kobayashi D, Takahashi O, Takahashi T, Asahara T, Nomoto K, Onodera H. Changes of the intestinal microbiota, short chain fatty acids, and fecal pH in patients with colorectal cancer. Dig Dis Sci 58: 1717–1726, 2013. [DOI] [PubMed] [Google Scholar]
- 111b.Ohigashi S, Sudo K, Kobayashi D, Takahashi T, Nomoto K, Onodera H. Significant changes in the intestinal environment after surgery in patients with colorectal cancer. J Gastrointest Surg 17: 1657–1664, 2013. [DOI] [PubMed] [Google Scholar]
- 112.O'Keefe SJ. Nutrition and colonic health: the critical role of the microbiota. Curr Opin Gastroenterol 24: 51–58, 2008. [DOI] [PubMed] [Google Scholar]
- 113.O'Keefe SJ, Chung D, Mahmoud N, Sepulveda AR, Manafe M, Arch J, Adada H, van der Merwe T. Why do African Americans get more colon cancer than Native Africans? J Nutr 137: 175S–182S, 2007. [DOI] [PubMed] [Google Scholar]
- 114.O'Mahony L, Feeney M, O'Halloran S, Murphy L, Kiely B, Fitzgibbon J, Lee G, O'Sullivan G, Shanahan F, Collins JK. Probiotic impact on microbial flora, inflammation and tumour development in IL-10 knockout mice. Aliment Pharmacol Ther 15: 1219–1225, 2001. [DOI] [PubMed] [Google Scholar]
- 115.Onoue M, Kado S, Sakaitani Y, Uchida K, Morotomi M. Specific species of intestinal bacteria influence the induction of aberrant crypt foci by 1,2-dimethylhydrazine in rats. Cancer Lett 113: 179–186, 1997. [DOI] [PubMed] [Google Scholar]
- 116.Pagnini C, Corleto VD, Mangoni ML, Pilozzi E, Torre MS, Marchese R, Carnuccio A, Giulio ED, Delle Fave G. Alteration of local microflora and alpha-defensins hyper-production in colonic adenoma mucosa J Clin Gastroenterol 45: 602–610, 2011. [DOI] [PubMed] [Google Scholar]
- 117.Pajak B, Orzechowski A. Sodium butyrate-dependent sensitization of human colon adenocarcinoma COLO 205 cells to TNF-alpha-induced apoptosis. J Physiol Pharmacol 58, Suppl 3: 163–176, 2007. [PubMed] [Google Scholar]
- 118.Park Y, Hunter DJ, Spiegelman D, Bergkvist L, Berrino F, van den Brandt PA, Buring JE, Colditz GA, Freudenheim JL, Fuchs CS, Giovannucci E, Goldbohm RA, Graham S, Harnack L, Hartman AM, Jacobs DR Jr, Kato I, Krogh V, Leitzmann MF, McCullough ML, Miller AB, Pietinen P, Rohan TE, Schatzkin A, Willett WC, Wolk A, Zeleniuch-Jacquotte A, Zhang SM, Smith-Warner SA. Dietary fiber intake and risk of colorectal cancer: a pooled analysis of prospective cohort studies. J Am Med Assoc 294: 2849–2857, 2005. [DOI] [PubMed] [Google Scholar]
- 119.Parker TJNS, Riordan AM, Hunter JO. Management of patients with food intolerance in IBS: the development and use of an exclusion diet. J Hum Nutr Diet 8: 159–166, 1995. [Google Scholar]
- 120.Proctor LM. The Human Microbiome Project in 2011 and beyond. Cell Host Microbe 10: 287–291, 2011. [DOI] [PubMed] [Google Scholar]
- 121.Pryde SE, Duncan SH, Hold GL, Stewart CS, Flint HJ. The microbiology of butyrate formation in the human colon. FEMS Microbiol Lett 217: 133–139, 2002. [DOI] [PubMed] [Google Scholar]
- 122.Ramasamy S, Singh S, Taniere P, Langman MJ, Eggo MC. Sulfide-detoxifying enzymes in the human colon are decreased in cancer and upregulated in differentiation. Am J Physiol Gastrointest Liver Physiol 291: G288–G296, 2006. [DOI] [PubMed] [Google Scholar]
- 123.Rhodes JM, Campbell BJ. Inflammation and colorectal cancer: IBD-associated and sporadic cancer compared. Trends Mol Med 8: 10–16, 2002. [DOI] [PubMed] [Google Scholar]
- 124.Rickard KL, Gibson PR, Wilson NJ, Mariadason JM, Phillips WA. Short-chain fatty acids reduce expression of specific protein kinase C isoforms in human colonic epithelial cells. J Cell Physiol 182: 222–231, 2000. [DOI] [PubMed] [Google Scholar]
- 125.Rombeau JL, Kripke SA. Metabolic and intestinal effects of short-chain fatty acids. Jpen 14: 181S–185S, 1990. [DOI] [PubMed] [Google Scholar]
- 126.Rowland IR. The role of the gastrointestinal microbiota in colorectal cancer. Curr Pharm Des 15: 1524–1527, 2009. [DOI] [PubMed] [Google Scholar]
- 127.Ruiz PA, Shkoda A, Kim SC, Sartor RB, Haller D. IL-10 gene-deficient mice lack TGF-beta/Smad-mediated TLR2 degradation and fail to inhibit proinflammatory gene expression in intestinal epithelial cells under conditions of chronic inflammation. Ann NY Acad Sci 1072: 389–394, 2006. [DOI] [PubMed] [Google Scholar]
- 128.Sakai E, Nakajima A, Kaneda A. Accumulation of aberrant DNA methylation during colorectal cancer development. World J Gastroenterol 20: 978–987, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Salzman NH, Hung K, Haribhai D, Chu H, Karlsson-Sjoberg J, Amir E, Teggatz P, Barman M, Hayward M, Eastwood D, Stoel M, Zhou Y, Sodergren E, Weinstock GM, Bevins CL, Williams CB, Bos NA. Enteric defensins are essential regulators of intestinal microbial ecology. Nat Immunol 11: 76–83, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Sanapareddy N, Legge RM, Jovov B, McCoy A, Burcal L, Araujo-Perez F, Randall TA, Galanko J, Benson A, Sandler RS, Rawls JF, Abdo Z, Fodor AA, Keku TO. Increased rectal microbial richness is associated with the presence of colorectal adenomas in humans. ISME J 6: 1858–1868, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Sandhu MS, White IR, McPherson K. Systematic review of the prospective cohort studies on meat consumption and colorectal cancer risk: a meta-analytical approach. Cancer Epidemiol Biomarkers Prev 10: 439–446, 2001. [PubMed] [Google Scholar]
- 132.Sartor RB. Microbial influences in inflammatory bowel diseases. Gastroenterology 134: 577–594, 2008. [DOI] [PubMed] [Google Scholar]
- 133.Sasaki M, Okamoto M, Sato C, Sugio K, Soejima J, Iwama T, Ikeuchi T, Tonomura A, Miyaki M, Sasazuki T. Loss of constitutional heterozygosity in colorectal tumors from patients with familial polyposis coli and those with nonpolyposis colorectal carcinoma. Cancer Res 49: 4402–4406, 1989. [PubMed] [Google Scholar]
- 134.Satokari R, Gronroos T, Laitinen K, Salminen S, Isolauri E. Bifidobacterium and Lactobacillus DNA in the human placenta. Lett Appl Microbiol 48: 8–12, 2009. [DOI] [PubMed] [Google Scholar]
- 135.Savage DC. Microbial ecology of the gastrointestinal tract. Ann Rev Microbiol 31: 107–133, 1977. [DOI] [PubMed] [Google Scholar]
- 135a.Scanlan PD, Shanahan F, Clune Y, Collins JK, O'Sullivan GC, O'Riordan M, Holmes E, Wang Y, Marchesi JR. Culture-independent analysis of the gut microbiota in colorectal cancer and polyposis. Environ Microbiol 10: 789–798, 2008. [DOI] [PubMed] [Google Scholar]
- 136.Scharlau D, Borowicki A, Habermann N, Hofmann T, Klenow S, Miene C, Munjal U, Stein K, Glei M. Mechanisms of primary cancer prevention by butyrate and other products formed during gut flora-mediated fermentation of dietary fibre. Mutat Res 682: 39–53, 2009. [DOI] [PubMed] [Google Scholar]
- 137.Schultz M, Tonkonogy SL, Sellon RK, Veltkamp C, Godfrey VL, Kwon J, Grenther WB, Balish E, Horak I, Sartor RB. IL-2-deficient mice raised under germfree conditions develop delayed mild focal intestinal inflammation. Am J Physiol Gastrointest Liver Physiol 276: G1461–G1472, 1999. [DOI] [PubMed] [Google Scholar]
- 138.Schwabe RF, Jobin C. The microbiome and cancer. Nat Rev Cancer 13: 800–812, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol Rev 90: 859–904, 2010. [DOI] [PubMed] [Google Scholar]
- 140.Shen J, Obin MS, Zhao L. The gut microbiota, obesity and insulin resistance. Mol Aspects Med 34: 39–58, 2013. [DOI] [PubMed] [Google Scholar]
- 141.Shen XJ, Rawls JF, Randall T, Burcal L, Mpande CN, Jenkins N, Jovov B, Abdo Z, Sandler RS, Keku TO. Molecular characterization of mucosal adherent bacteria and associations with colorectal adenomas. Gut Microbes 1: 138–147, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Siegel R, Ma J, Zou Z, Jemal CA. Cancer statistics A, 2014. Cancer J Clinicians 64: 9–29, 2014. [DOI] [PubMed] [Google Scholar]
- 143.Simmering R, Pforte H, Jacobasch G, Blaut M. The growth of the flavonoid-degrading intestinal bacterium, Eubacterium ramulus, is stimulated by dietary flavonoids in vivo. FEMS Microbiol Ecol 40: 243–248, 2002. [DOI] [PubMed] [Google Scholar]
- 144.Sobhani I, Amiot A, Le Baleur Y, Levy M, Auriault ML, Van Nhieu JT, Delchier JC. Microbial dysbiosis and colon carcinogenesis: could colon cancer be considered a bacteria-related disease? Ther Advance Gastroenterol 6: 215–229, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, Corthier G, Tran Van Nhieu J, Furet JP. Microbial dysbiosis in colorectal cancer (CRC) patients. PLos One 6: e16393, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Sonnenburg JL, Angenent LT, Gordon JI. Getting a grip on things: how do communities of bacterial symbionts become established in our intestine? Nat Immunol 5: 569–573, 2004. [DOI] [PubMed] [Google Scholar]
- 147.Sonnenburg JL, Xu J, Leip DD, Chen CH, Westover BP, Weatherford J, Buhler JD, Gordon JI. Glycan foraging in vivo by an intestine-adapted bacterial symbiont. Science 307: 1955–1959, 2005. [DOI] [PubMed] [Google Scholar]
- 148.Soret R, Chevalier J, De Coppet P, Poupeau G, Derkinderen P, Segain JP, Neunlist M. Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. Gastroenterology 138: 1772–1782, 2010. [DOI] [PubMed] [Google Scholar]
- 149.Suau A, Bonnet R, Sutren M, Godon JJ, Gibson GR, Collins MD, Dore J. Direct analysis of genes encoding 16S rRNA from complex communities reveals many novel molecular species within the human gut. Appl Environ Microbiol 65: 4799–4807, 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Swidsinski A, Khilkin M, Kerjaschki D, Schreiber S, Ortner M, Weber J, Lochs H. Association between intraepithelial Escherichia coli and colorectal cancer. Gastroenterology 115: 281–286, 1998. [DOI] [PubMed] [Google Scholar]
- 151.Swidsinski A, Ladhoff A, Pernthaler A, Swidsinski S, Loening-Baucke V, Ortner M, Weber J, Hoffmann U, Schreiber S, Dietel M, Lochs H. Mucosal flora in inflammatory bowel disease. Gastroenterology 122: 44–54, 2002. [DOI] [PubMed] [Google Scholar]
- 151a.Tahara T, Yamamoto E, Suzuki H, Maruyama R, Chung W, Garriga J, Jelinek J, Yamano HO, Sugai T, An B, Shureiqi I, Toyota M, Knodo Y, Estecio MRH, Issa JPJ. Fusobacterium in colonic flora and molecular features of colorectal carcinoma. Cancer Res 74: 1311–1318, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Tamboli CP, Neut C, Desreumaux P, Colombel JF. Dysbiosis in inflammatory bowel disease. Gut 53: 1–4, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L. The role of short-chain fatty acids in health and disease. Adv Immunol 121: 91–119, 2014. [DOI] [PubMed] [Google Scholar]
- 154.Teixeira TF, Collado MC, Ferreira CL, Bressan J, Peluzio Mdo C. Potential mechanisms for the emerging link between obesity and increased intestinal permeability. Nutr Res 32: 637–647, 2012. [DOI] [PubMed] [Google Scholar]
- 155.Terzic J, Grivennikov S, Karin E, Karin M. Inflammation and colon cancer. Gastroenterology 138: 2101–2114, 2010. [DOI] [PubMed] [Google Scholar]
- 156.Tian B, Brasier AR. Identification of a nuclear factor kappa B-dependent gene network. Recent Prog Horm Res 58: 95–130, 2003. [DOI] [PubMed] [Google Scholar]
- 157.Tlaskalova-Hogenova H, Vannucci L, Klimesova K, Stepankova R, Krizan J, Kverka M. Microbiome and colorectal carcinoma: insights from germ-free and conventional animal models. Cancer J 20: 217–224, 2014. [DOI] [PubMed] [Google Scholar]
- 158.Topping DL, Clifton PM. Short-chain fatty acids and human colonic function: roles of resistant starch and nonstarch polysaccharides. Physiol Rev 81: 1031–1064, 2001. [DOI] [PubMed] [Google Scholar]
- 159.Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, Egholm M, Henrissat B, Heath AC, Knight R, Gordon JI. A core gut microbiome in obese and lean twins. Nature 457: 480–484, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Science Trans Med 1: 6–14, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Ullman TA, Itzkowitz SH. Intestinal inflammation and cancer. Gastroenterology 140: 1807–1816, 2011. [DOI] [PubMed] [Google Scholar]
- 161a.Uronis JM, Muhlbauer M, Herfarth HH, Rubinas TC, Jones GS, Jobin C. Modulation of the intestinal microbiota alters colitis-associated colorectal cancer susceptibility. PloS One 4: e6026, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Vinolo MA, Rodrigues HG, Nachbar RT, Curi R. Regulation of inflammation by short chain fatty acids. Nutrients 3: 858–876, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, Nakamura Y, White R, Smits AM, Bos JL. Genetic alterations during colorectal-tumor development. N Engl J Med 319: 525–532, 1988. [DOI] [PubMed] [Google Scholar]
- 164.Vogelstein B, Kinzler KW. Through the glass lightly (Abstract). Science 267: 1613, 1995. [DOI] [PubMed] [Google Scholar]
- 165.Wacklin P, Makivuokko H, Alakulppi N, Nikkila J, Tenkanen H, Rabina J, Partanen J, Aranko K, Matto J. Secretor genotype (FUT2 gene) is strongly associated with the composition of Bifidobacteria in the human intestine. PLos One 6: e20113, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Wang TCG, Qui 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. Int Soc Microbial Ecol 2011August18. doi: 10.1038/ismej.2011.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166a.Warren RL, Freeman DJ, Pleasance S, Watson P, Moore RA, Cochrane K, Allen-Vercoe E, Holt RA. Co-occurrence of anaerobic bacteria in colorectal carcinomas (Abstract). Microbiome 1: 16, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166b.Weir TL, Manter DK, Sheflin AM, Barnett BA, Heuberger AL, Ryan EP. Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults. PLoS One 8: e70803, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Westbrook AM, Wei B, Braun J, Schiestl RH. Intestinal mucosal inflammation leads to systemic genotoxicity in mice. Cancer Res 69: 4827–4834, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Westbrook AM, Wei B, Braun J, Schiestl RH. More damaging than we think: systemic effects of intestinal inflammation. Cell Cycle 8: 2482–2483, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.White RL. Tumor suppressing pathways. Cell 92: 591–592, 1998. [DOI] [PubMed] [Google Scholar]
- 170.Willing BP, Russell SL, Finlay BB. Shifting the balance: antibiotic effects on host-microbiota mutualism. Nat Rev 9: 233–243, 2011. [DOI] [PubMed] [Google Scholar]
- 171.Wu N, Yang X, Zhang R, Li J, Xiao X, Hu Y, Chen Y, Yang F, Lu N, Wang Z, Luan C, Liu Y, Wang B, Xiang C, Wang Y, Zhao F, Gao GF, Wang S, Li L, Zhang H, Zhu B. Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb Ecol 66: 462–470, 2013. [DOI] [PubMed] [Google Scholar]
- 171a.Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X, Yen HR, Huso DL, Brancati FL, Wick E, McAllister F, Housseau F, Pardoll DM, Sears CL. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med 15: 1016–1022, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Yang L, Pei Z. Bacteria, inflammation, and colon cancer. World J Gastroenterol 12: 6741–6746, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Yu LC, Wang JT, Wei SC, Ni YH. Host-microbial interactions and regulation of intestinal epithelial barrier function: from physiology to pathology. World J Gastroenterol 3: 27–43, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Zackular JP, Baxter NT, Iverson KD, Sadler WD, Petrosino JF, Chen GY, Schloss PD. The gut microbiome modulates colon tumorigenesis. mBio 4: e00692–e00613, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Zackular JP, Rogers MA, Ruffin MTt, Schloss PD. The human gut microbiome as a screening tool for colorectal cancer. Cancer Prevention Res 7: 1112–1121, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Zhang J, Guo Z, Lim AA, Zheng Y, Koh EY, Ho D, Qiao J, Huo D, Hou Q, Huang W, Wang L, Javzandulam C, Narangerel C, Jirimutu Menghebilige Lee YK, Zhang H. Mongolians core gut microbiota and its correlation with seasonal dietary changes (Abstract). Scientific Rep 4: 5001, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Zhang Y, Zhou L, Bao YL, Wu Y, Yu CL, Huang YX, Sun Y, Zheng LH, Li YX. Butyrate induces cell apoptosis through activation of JNK MAP kinase pathway in human colon cancer RKO cells. Chemico-biol Int 185: 174–181, 2010. [DOI] [PubMed] [Google Scholar]
- 178.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 9: e90849, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]


