Abstract
Recently, an unprecedented effort has been directed at understanding the interplay between chronic inflammation and development of cancer, with the case of inflammatory bowel disease (IBD)-associated colorectal cancer at the forefront of this research endeavor. The last decade has been particularly fertile, with the discovery of numerous innovative paradigms linking various inflammatory, proliferative, and innate and adaptive immune signaling pathways to the development of colorectal cancer. Because of the preponderant role of the intestinal microbiota in the initiation and progression of IBD, recent efforts have been directed at understanding the relationship between bacteria and colorectal cancer. The microbiota and its collective genome, the microbiome, form a diverse and complex ecological community that profoundly impacts intestinal homeostasis and disease states. This review will discuss the differential influence of the microbiota on the development of IBD-associated colorectal cancer and highlight the role of innate immune sensor-dependent as well as -independent mechanisms in this pathology.
Keywords: colorectal cancer, inflammatory bowel disease, intestinal microbiota
A significant risk of morbidity experienced by patients with inflammatory bowel disease (IBD) is the development of colitis-associated colorectal cancer. The risk of developing colitis-associated colorectal cancer increases with the duration and severity of inflammation and may confer a 10-fold greater risk than that observed in the healthy population. Thus, understanding the etiology of IBD represents a key step in elucidating the events leading to colitis-associated colorectal cancer and the elaboration of strategies aimed at preventing the pathology.
Genome-wide association studies and experimental models of IBD underscore the importance of the innate immune system and the intestinal microbiota in the pathogenesis of IBD.1-6 Numerous innate immune sensors and their associated signaling proteins have been linked to the maintenance of intestinal homeostasis through their effects on cellular proliferation, inflammation, and wound healing. An emerging body of literature highlights the impact of these innate immune sensors in protecting or promoting the development of colitis-associated colorectal cancer and much of the focus lies in the role of the complex intestinal microbiota. Indeed, the human gastrointestinal tract is colonized by thousands of bacterial species, amounting to more than 100 trillion bacteria. This complex microbial community carries a rich and diverse microbial genome, the microbiome, with a tremendous metabolic potential that influences intestinal homeostasis. Recent reports indicate that microbial composition may impact the status of various pathologic conditions including IBD and colorectal cancer. In this review we will focus on the relationship between innate sensors, the microbiota and development of colitis-associated colorectal cancer.
MICROBIAL SYMBIONTS OF THE MAMMALIAN INTESTINE
The human intestinal microbiota is a community of 1013 − 1014 microorganisms that reside in the intestine and normally participate in a symbiotic relationship with their eukaryotic host.7-9 While IBD has been recognized as a disease influenced by the intestinal microbiota, difficulty in culturing these complex microorganisms has precluded the formal identification of colitogenic bacteria. Moreover, IBD is likely not caused by a single microbial entity but rather through the action of a complex consortium of microorganisms that render identification of these bacteria even more challenging using classical microbiology culturing techniques. However, characterization of this complex ecological system has made an extensive leap forward through the use of newly developed molecular approaches focused on differentiating specific genetic signatures in ribosomal 16S genes.10,11 Unbiased metagenomics sequencing has revealed that the human distal intestinal microbiota comprises two predominant phyla, the Firmicutes and Bacteroidetes, with lesser contributions from Proteobacteria and Actinobacteria, and minor contributions from Fusobacteria, Verrucomicrobia, and Cyanobacteria.12,13 Importantly, the murine microbiota is remarkably similar14; thus studies performed in murine models will likely have translational implications relevant to human disease.
The microbiome contains a wealth of information, encompassing 150-fold as many genes as the human genome.15,16 Accordingly, a considerable worldwide effort is under way to identify both the constituents and unique traits of this complex microbial community. One such endeavor is the International Human Microbiome Consortium, which includes the Human Microbiome Project and the Metagenomics of the Human Intestinal Tract project (MetaHIT).17,18 A primary goal of these projects is to determine the presence, composition, and function of a putative core microbiome shared among all humans. Already these studies have hinted that a core human microbiome exists at the gene level, with a large number of microbial genes and pathways shared among individuals.13,18 Deviations from this core microbiome could potentially affect human health and promote disease. Thus, characterizing the vast intestinal microbial community present in healthy as well as pathologic conditions will likely revolutionize our conception of bacteria/host interactions and identify new physiological processes influenced by the microbiota. Already the microbiota has been linked to cardiac development,19 angiogenesis,20 innate and adaptive immunity,21-24 metabolism,25 nutrient acquisition,26 and gastrointestinal development and homeostasis.27 Furthermore, alterations in the microbial community are associated with multiple diseases, including obesity,13,14,28-30 fatty liver disease,31 type 1 and type 2 diabetes,32,33 kidney disease,34 arthritis,35 IBD,16,36 and colorectal cancer.37 In IBD, analyses of the fecal and mucosally adherent microbiota have revealed significant differences in microbial community structure between patients and healthy controls. For example, studies have consistently identified reductions in total gut microbial concentration, decreases in the richness and diversity of the microbiota, and changes in the proportion of the two dominant phyla, the Firmicutes and Bacteroidetes.16,38-40 At the moment, though, the relationship between alterations in the microbiota and development of IBD and colorectal cancer is unclear.
MICROBIOTA CAN CONFER HOST TRAITS
The major limitation of total microbial gene pool analysis in health and disease, through what is known as metagenomic studies, relates to the correlative nature of this experimental approach. In essence, do changes in the microbial composition and metagenome directly influence the development of disease? The ability to transfer host traits through microbiota transplants suggests that a dysbiotic microbial community plays an active, rather than passive role in disease process. Glimcher and colleagues41 have described a transferable trait in colitis-prone TRUC mice (Tbx21−/−; Rag2−/− ulcerative colitis). TRUC mice develop highly penetrable colitis that progresses to colorectal cancer.42 Housing either newborn or adult wildtype (WT) mice with TRUC mice induces aggressive colitis in this normally colitis-resistant strain.41 Although microbiome analyses were not performed in these mice, these observations suggest that the colitis phenotype is transferred through a dysbiotic microbiota present in TRUC mice. Analysis of the TRUC microbiome and microbiota transplants into germ-free mice may address this question and provide further insight into the transmissibility of such traits.
The impact of the microbial community on host traits is not limited to an intestinal phenotype. For example, obesity alters the composition of the gut microbiota and increases its metabolic potential to harvest energy from the host diet.13,14,28-30 Microbiota transplantation from obese mice into lean germ-free recipients induces weight gain and increases adiposity, demonstrating that these traits can be conferred through a microbial dependent mechanism.29,30 More recently, Tlr5-deficient mice were observed to become hyperphagic and develop metabolic syndrome comprising hyperlipidemia, hypertension, insulin resistance, and enhanced adiposity.43 In these mice, specific bacterial phylotypes were expanded and contracted compared to WT mice. Interestingly, hyperphagia and metabolic syndrome are transferable to WT mice upon fecal transplantation of microbiota obtained from Tlr5−/− mice.43 These fascinating experiments demonstrate that the microbiota has the potential to profoundly impact the host health status.
HOST RECOGNITION OF MICROBIAL SPECIES AFFECTS COLITIS AND COLORECTAL CANCER
Germ-free studies have revealed a key role for the microbiota in driving colorectal cancer. Hereditary colorectal cancer is commonly modeled with the ApcMin/+ (multiple intestinal neoplasia allele of the adenomatous polyposis coli gene) mouse, a model of human familial adenomatous polyposis. When raised in conventional or specific pathogen-free (SPF) conditions, a mutation in the tumor suppressor Apc causes this mouse strain to develop tens to hundreds of intestinal adenomas.44,45 However, mice raised in germ-free conditions exhibit ≈50% reduction in intestinal tumors compared to those housed in SPF conditions.46
More recently, the microbial composition has been found to influence the development of colitis-associated colorectal cancer. In the newly developed AOM/IL-10−/− model, intestinal inflammation occurs spontaneously from lack of immunosuppressive IL-10, and tumorigenesis is initiated with the colon-specific carcinogen azoxymethane (AOM).47 When raised in conventional or SPF conditions, these mice develop extensive intestinal inflammation and colonic adenomas. However, when mono-associated with Bacteroides vulgatus these mice exhibit fewer tumors. Remarkably, mice raised in germ-free conditions are devoid of intestinal inflammation and tumors.47 These findings highlight the critical role of specific members of the commensal microbiota in the development of colitis-associated colorectal cancer.47
Although the microbiota influences the development of colorectal cancer, colitis-associated colorectal cancer, and other forms of cancer (Table 1),48-97 the extent of host microbial recognition in this process is still unclear. Humans, mice, and other eukaryotes are equipped with an elegant repertoire of receptors, and each recognize specific conserved microbial patterns, such as components of bacterial cell walls or nucleic acids. These microbial sensors are termed pattern recognition receptors (PRR) and include retinoic acid inducible gene-I like RNA helicases (RLH), C-type lectin receptors (CLR), nucleotide-binding domain leucine-rich repeat proteins (NLR; also known as Nod-like receptors), and Toll-like receptors (TLR).98-101 Many of these receptors are expressed on intestinal epithelial cells (IEC) as well as on various mucosal immune cells. Upon engagement, these receptors initiate signaling cascades that activate numerous downstream effector systems including mitogen-activated protein kinases (MAPK), nuclear factor of kappa B (NF-κB), and interferon regulatory factors (IRFs) that then modulate apoptosis, proliferation, cell migration, and inflammation.102,103
TABLE 1.
Bacterial Associations with Carcinogenesis
Bacteria | Cancer | * | Evidence | Refs. |
---|---|---|---|---|
Helicobacter hepaticus | Colorectal | A | Augments AOM-induced, and spontaneous colorectal cancer in Smad3−/−, Rag2−/− and ApcMin/+ mice |
(48-50,179,180,213) |
H. hepaticus + H. bilis | Colorectal | A | Dual infection induces colorectal cancer in Mdr1a−/− mice |
(51,52) |
H. typhlonius + H. rodentium |
Colorectal | A | Dual infection in neonates induces colorectal cancer in Il10−/− mice |
(53,181) |
Streptococcus bovis | Colorectal | H |
S. bovis bacteremia and endocarditis associated with human colorectal cancer |
(54-58) |
Colorectal | A | Augments AOM-induced cancer in rats | (59) | |
Bacteroides fragilis | Colorectal | A | Enterotoxigenic B. fragilis augments spontaneous (ApcMin/+) colorectal cancer in mice |
(60) |
Colorectal | H | Increased prevalence of enterotoxigenic B. fragilis in human colorectal cancer |
(148) | |
B. vulgatus | Colorectal | A | Induces mild AOM-induced colorectal cancer in Il10−/− mice |
(47) |
Escherichia coli | Colorectal | H | Increased mucosa-associated E. coli in human Crohn’s and colorectal cancer |
(61) |
Citrobacter rodentium and C. freundii |
Colorectal | A | Etiologic agent of transmissible murine colonic hyperplasia |
(62) |
Colorectal | A | Augments spontaneous (ApcMin/+) and DMH-induced colorectal cancer in mice |
(63,64) | |
H. pylori | MALT lymphoma | H | Lymphoma regression after eradication of H. pylori in humans |
(65-68) |
H. mustelae | MALT lymphoma | A | Induces MALT-like lymphoma in ferrets | (69) |
H. pylori | Gastric | H | Causative agent of human peptic ulcer disease; Predisposes to gastric cancer |
(70,212) |
Gastric | A | Induces gastric cancer in gerbils | (71) | |
H. felis | Gastric | A | Induces gastric cancer in insulin-gastrin transgenic mice |
(72) |
S. anginosus | Oral and esophageal | H | Found in human oral and esophageal cancer tissue | (73-77) |
H. hepaticus | Hepatobiliary | A | Induces hepatitis and liver cancer in A/JCr mice | (78-80) |
Salmonella typhi | Hepatobiliary | H | Chronic infection increases risk of hepatobiliary cancer by up to 8-fold in humans |
(81-85) |
Chlamydia pneumoniae | Lung | H | Chronic infection raises lung cancer risk; renders epithelial cells resistant to apoptosis |
(86-91) |
H. hepaticus | Mammary | A | Promotes mammary cancer in ApcMin/+ and Rag2−/−; ApcMin/+ mice |
(48) |
Lawsonia intracellularis | Proliferative enteropathy |
A | Induces proliferative intestinal lesions in animals resembling human IBD lesions |
(92) |
Mycoplasma fermentans and M. penetrans |
General | C | Induce malignant transformation and independence from growth factors in vitro |
(93-95) |
A | Infected cells induce tumors with high H-ras and c-myc expression in mice |
(96) | ||
Bartonella sp. | General | A | Induces tumor-like structures and angiogenesis through VEGF |
(97) |
H, evidence in humans; A, evidence in animal models; C, cell lines.
The most well-characterized family of PRRs is the TLRs. With the exception of TLR3, these extracellular or endosomal PRRs signal through the adaptor protein myeloid differentiation factor 88 (MyD88).104,105 Accordingly, investigators have utilized Myd88−/− mice to link host innate signaling with the microbiota and development of colitis-associated colorectal cancer. Development of colitis and colorectal cancer is strongly reduced in Il10−/− mice deficient in MyD88 signaling.1,47,106 Importantly, deletion of Myd88 also diminishes development of spontaneous colorectal cancer in ApcMin/+ mice, reducing both intestinal tumor size and multiplicity.107 The cellular compartment responsible for MyD88-dependent proinflammatory and procarcinogenic signaling is not completely understood. However, MyD88 signaling in macrophages promotes colonic epithelial progenitor cell proliferation and cellular organization within colonic crypts,108,109 raising the possibility that disrupted MyD88 signaling may affect the balance between proliferation and apoptosis. Indeed, bone marrow transplant experiments in Tlr4−/− mice indicate that immune cells are critical in promoting the development of colitis-associated colorectal cancer.110,111 These data suggest that host recognition of microbial species is necessary for the development of colitis-associated colon cancer.112-115
Because host PRRs distinguish between a wide spectrum of microorganisms, it is important to establish their individual contribution to the development of colitis and colorectal cancer. Like TLR4,116 PRRs of the NLR family including NOD1, NOD2, and NLRP3 have been genetically linked to the development of IBD.112-115 However, in contrast to TLR4/MyD88, these intracellular sensors appear to play a protective role in gastrointestinal disease.117,118 Interestingly, Nod2-deficient mice harbored increased numbers of gut commensal bacteria and impaired bacterial killing and antimicrobial peptide secretion in the intestine,119 providing a potential mechanism by which Nod2 mutations may perturb intestinal homeostasis. Recent findings indicate a link between Nod2 polymorphisms and development of colorectal cancer.120,121 However, the lack of data generated from experimental models of colorectal cancer and colitis-associated colorectal cancer precludes a direct assessment of the function of NOD2 in the carcinogenic processes.
Similarly, NOD1, a close relative of NOD2, has been shown recently to play a protective role against development of colitis and colorectal cancer. Compared to WT mice, Nod1-deficient mice displayed exacerbated colitis in response to dextran sulfate sodium (DSS) exposure.122 Likewise, AOM/DSS-induced colitis-associated colorectal cancer and spontaneous colorectal cancer (ApcMin/+) is augmented in Nod1−/− mice, a phenomenon attenuated with broad-spectrum antibiotic treatment.122 These findings suggest a role for NOD1 and microbial signaling in the development of colorectal cancer. Furthermore, the related intracellular sensor NLRP3 also appears protective against colitis and colitis-associated colorectal cancer, a role attributed to its expression in immune cells.123 These findings suggest a protective role for NLRP3, NOD1, and microbial signaling in the development of colorectal cancer. The fact that TLR/MyD88 signaling promotes the development of colorectal cancer, whereas NLR signaling appears to prevent this pathology, highlights the complex interaction between the host and the microbiota. These differential responses may be dictated by the bacterial motif recognized by a particular PRR and/or the location of the PRR, i.e., intracellular versus extracellular. Since NOD1, NOD2, and NLRP3 reside in an intracellular location, perhaps bacterial invasion activates these microbial sensors, which then initiate a protective response to clear invading bacteria and restore intestinal homeostasis.
Although the above studies clearly indicate the importance of PRRs in modulating the development of colitis-associated colorectal cancer, the interplay between inflammation and colorectal cancer is not always clear. For example, development of colitis-associated colorectal is strongly reduced in AOM/DSS-treated Tlr4−/− mice compared to WT mice, whereas the inflammatory status is marginally affected in these mice.110 This incomplete relationship between inflammatory status and tumorigenesis is also observed in AOM/DSS-treated Il6−/− and Stat3IEC−/− mice.124 The IL-6/STAT3 signaling axis profoundly modulates intestinal homeostasis through pro-proliferative and antiapoptotic effects. Remarkably, while AOM/DSS-exposed Il6−/− or Stat3IEC−/− mice display increased intestinal inflammation and proinflammatory cytokine secretion, they exhibit significantly fewer intestinal tumors than WT mice. In addition, Enterococcus faecalis mono-associated Il10−/− mice develop inflammation to the same extent as conventionalized Il10−/− mice; however, tumors fail to develop in the mono-associated mice.125 These observations suggest that mechanisms other than inflammation can influence the development of colorectal cancer. Although this concept is at odds with epidemiologic data in humans showing a correlation between inflammatory status and colorectal cancer,126 an intriguing possibility is that microbial status could impact tumorigenesis without directly affecting the inflammatory status (Fig. 1). Indeed, up to 80% of patients with long-standing IBD (<30 years) do not develop colitis-associated colorectal cancer,127 suggesting that inflammation alone is not sufficient to promote colorectal cancer.
FIGURE 1.
The intestinal microbiota promotes inflammation and neoplasia in the colon. Conserved microbial signatures are recognized by pattern recognition receptors such as TLRs, which trigger downstream signaling pathways leading to the expression of various genes including growth factors and inflammatory mediators. Autocrine and paracrine signaling from these mediators amplifies inflammation and promotes neoplasia. Independent of inflammation, microbial enzymes of the metabolome process latent dietary procarcinogens to their biologically active form and elicit neoplastic changes.
MICROBIAL INFLUENCE ON CARCINOGENESIS
The impact of microorganisms on the development of colorectal cancer has been mostly studied from the point of view of PRR signaling and inflammatory responses. However, it has become apparent that the tremendous metabolic capacity offered by the microbiota (referred to as the metabolome) likely plays an important role during the carcinogenesis process, independently of overt inflammation (Table 2).
TABLE 2.
Bacterial Products Linked to Carcinogenesis
Bacterial Toxins | Prominent Producers | Cancer Relevance | Refs. |
---|---|---|---|
Bacillus fragilis enterotoxin |
Enterotoxic Bacillus fragilis | Induces Stat3 activation and Th17 cells that promote colorectal tumorigenesis; Increased prevalence in human colorectal cancer patients |
(60,128) |
Cytolethal distending toxin |
Helicobacter hepaticus | Induces progression of hepatitis to dysplasia in mice | (129) |
Escherichia coli, Campylobacter sp., Salmonella typhi, |
Acts as a DNase to create double-strand DNA breaks; arrests host cell cycle at G2/M transition through inactivation of Cdk1 |
(130-133) | |
Cycle inhibiting factor | Enteropathogenic E. coli | Disrupts cell cycle through stabilization of Cdk inhibitors p21 and p27, cells replicate DNA without dividing, results in hyperploidy |
(134-137) |
Cytotoxic necrotizing factor |
E. coli | Prevents apoptosis via Bcl-2 upregulation in epithelial cells; promotes motility in uroepithelial cells |
(138-140) |
Pasteurella multocida toxin |
Pasteurella multicida | Promotes anchorage-independent growth of enterocytes and fibroblasts |
(141,142) |
Epidermal differentiation inhibiting factor |
Staphylococcus aureus | Induces transient hyperplasia in the epidermis of mice | (143,144) |
Cytotoxin-associated antigen A |
H. pylori | Induces rapid progression through cell cycle and morphological changes that promote invasion; Augments the risk of human gastric cancer |
(145-148) |
Bacterial Enzymes | Enzymatic Action | Cancer Relevance | Refs. |
---|---|---|---|
β-glucuronidase | Hydrolyzes glucuronic acid conjugates in bile |
Converts AOM and heterocyclic amines from cooked meat to active carcinogens; induces colon cancer in rodents |
(149-150) |
High levels associated with a higher risk of human colorectal cancer |
(151) | ||
β-glucosidase | Hydrolyzes plant glycosides | Converts cyasin to its active carcinogen; induces colon cancer in rats |
(169) |
Nitroreductase | Reduces nitrates to nitrites, forming N-nitroso compounds |
Nitrates in red meat and processed foods linked to cancer |
(152,153) |
N-nitroso compounds act as DNA alkylating agents | (154) | ||
Azoreductase | Reduces azo compounds, may produce mutagenic amines |
Human intestinal microbiota can convert azo compounds in some food products to carcinogenic aromatic amines |
(155-156) |
Mucinase | Degrades protective mucins | Areas of colonic dysplasia located at mucin-depleted foci | (157-159) |
Catalase | Converts hydrogen peroxide to water and oxygen |
Produced by lactic acid bacteria; protective against DMH-induced colorectal cancer |
(160) |
Catalase attenuates E. faecalis-induced aneuploidy | (161) | ||
7-α/β dehydroxylation enzymes |
Catalyze the formation of secondary bile acids |
Levels of deoxycholic acid (DCA) correlate with risk of colorectal cancer; high DCA found in colon cancer patients |
(162-165) |
DCA activates beta-catenin, promotes proliferation and invasion; promotes carcinogenesis in animal models of colorectal cancer |
(166-168) |
The activation or detoxification of carcinogens that modulate the tumorigenic process is strongly influenced by the abundant enzymatic activities contributed by the intestinal microbiota.128-168 In the 1960s it was observed that germ-free rats did not develop intestinal tumors from exposure to the carcinogenic plant glycoside cyasin. However, intestinal tumors developed in germ-free rats directly administered methylazoxymethanol, the downstream active metabolite of cyasin.169 The generation of this metabolite is dependent on bacterial β-glucosidase enzyme activities,169 strongly implying that the microbiota influences the production of bioactive carcinogenic compounds. Further investigation has revealed that the intestinal microbiota can convert latent carcinogens into bioactive forms through the action of various enzymes such as β-glucuronidase, β-glucosidase, azoreductase, and nitroreductase.170 The most commonly used experimental colon carcinogen, AOM, is first hydrolyzed in the liver to methylazoxymethanol and conjugated with glucuronic acid before transport to the intestine through bile secretion.171 Further metabolism through bacterial β-glucuronidase converts the glucuronic acid-conjugated methylazoxymethanol to its carcinogenic form that spontaneously yields the highly reactive methyl carbonium ion.170,172,173 Interestingly, β-glucuronidase inhibition significantly reduces the ability of AOM to induce tumors in rats.149 In addition, the heterocyclic amines 2-amino-3-methylimidazo-[4,5-f]quinoline (IQ), 2-amino-3,8-dimethylimidazo-[4,5-f]quinoxaline (MeIQx), and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) that form when meat is cooked to high temperatures must be metabolically activated by the enzymatic activities of intestinal microbes to exert full mutagenic potential.150,174-177 These important observations have a profound implication for the field of colorectal cancer research since animal facilities at various institutions and even commercial vendors likely harbor different microbial communities.178 Presumably, their microbiota contain differential metabolic capacities that could influence their responses to procarcinogenic agents such as AOM. Likewise, phenotypic variability observed in Il10−/− and Smad3−/− mice appears to correlate with the presence of specific opportunistic microorganisms such as Helicobacter sp. and Citrobacter sp.179-182
Interestingly, probiotic bacteria such as Lactobacillus sp. and Bifidobacterium sp. exert anticarcinogenic effects, in part by inactivating microbial enzymes important for procarcinogen activation.183 For example, probiotic lactic acid bacteria including L. casei and L. acidophilus can decrease the activity of β-glucuronidase, azoreductase, and nitroreductase.184,185 In fact, B. longum reduces AOM-induced aberrant crypt formation, which correlates with a decrease in AOM-activating β-glucuronidase activity.186 In addition, other Lactobacillus sp. and Bifidobacterium sp. can inhibit DNA damage and tumorigenesis induced by N-methyl-N’-nitro-N-nitrosoguanidine (MNNG), 1,2-dimethylhydrazine (DMH), AOM, and heterocyclic amines IQ, MeIQx, and PhIP,187-191 although the extent to which inactivation of carcinogen-activating enzymes is involved in these processes remains unknown. This balance of activation and detoxification is reminiscent of the balance between host oncogenes and tumor suppressors and suggests that microbial community structure and its associated metabolomic capacity play a significant role in the initiating step of carcinogenesis (Fig. 1).
DNA damage and chromosomal instability are early genetic events involved in the development of colorectal cancer. Chromosomal instability, such as aneuploidy, is associated with long-standing IBD and frequently predicts the future development of colorectal cancer.192-194 A common commensal intestinal bacterium, E. faecalis, induces aneuploidy in colonic epithelial cells and aggressive colitis in mono-associated Il10−/− mice.161,195,196 Inhibitors of reactive oxygen and nitrogen species (RONS) prevent E. faecalis-induced aneuploidy,161,197 suggesting that the unique ability of this bacterium to induce RONS can lead to chromosomal instability in a susceptible host. Experimentation in mice deficient in the antioxidant enzymes glutathione peroxidase -1 and -2 (Gpx1/2−/−) further supports the role of the microbiota and RONS in carcinogenesis. Gpx1/2−/− mice spontaneously develop intestinal tumors with ≈25% penetrance when raised in conventional conditions, whereas this is reduced to <9% in SPF conditions and abolished in germ-free animals.198 These findings strongly indicate that the microbiota—and most likely specific members of the microbiota—induce RONS that promote carcinogenesis.
The protective role for the microbiota in intestinal homeostasis includes their ability to influence epithelial cell proliferation and apoptosis. A major mechanism by which this is accomplished involves microbial fermentation of dietary fibers to the short chain fatty acids (SCFA) acetate, propionate, and butyrate. These SCFAs, butyrate in particular, are avidly absorbed by colonocytes and used as a primary source of energy. In addition to their important antiinflammatory role,199,200 SCFAs support intestinal homeostasis and the resolution of intestinal injury by promoting cellular proliferation and differentiation in the normal nonneoplastic colon.200,201 Remarkably, SCFAs exert an opposite effect on cancerous cells. Butyrate in particular can induce apoptosis in colon cancer cell lines by a variety of mechanisms, most of which are associated with its role as a histone deacetylase inhibitor. This can involve hyperactivation of Wnt, sensitization to Fas-mediated apoptosis, and activation of the intrinsic/mitochondrial apoptosis pathway through upregulation of Bak, downregulation of Bcl-xL, cytochrome-c release, and caspase-9 activation.202-207 Nonetheless, butyrate may only provide protection during the early stages of tumorigenesis, as the two major receptors for butyrate, SLC5A8 and GPR109A, are frequently silenced in human cancers.208-211
Although colorectal cancer has not been linked to any specific microorganisms, some species have been identified as cancer-promoting bacteria (Table 1). For example, Helicobacter pylori infection in humans predisposes to gastric cancer.212 In addition, H. hepaticus augments both experimental colitis-associated colon cancer and spontaneous colorectal cancer in mice.179,213 Bacteroides fragilis is a common intestinal commensal, yet an enterotoxigenic variant induces spontaneous colonic tumorigenesis in ApcMin/+ mice.60 Therefore, exclusion of opportunistic pathogens by commensal bacteria may represent a natural defense against gastrointestinal diseases, including colorectal cancer. This is exemplified in the therapeutic use of probiotics, live bacteria that confer health benefits to the host. Probiotic bacteria engage the host to produce biofilms that prevent adhesion or invasion of pathogenic species, maintain gut barrier function by preventing redistribution of host tight junction proteins, induce host cytokines to modulate inflammation an immunity, and neutralize carcinogens and toxins.214-219 Commensal bacteria may also exert selective pressure against potential intruders as a mechanism to maintain a niche. For example, conditioned medium derived from a human intestinal microbiota directly inhibits synthesis of an enterohemorrhagic E. coli O157:H7 toxin.220 Communication with the commensal microbiota provokes antimicrobial responses from the host epithelium including the release of antibacterial lectins like RegIIIc, angiogenins, and α-defensins.221-223 Remarkably, these antibacterial products not only deplete subsets of potentially pathogenic bacteria, but also protect against subsequent aberrant immune responses. For example, the intestinal microbiota of mice expressing a human enteric α-defensin, DEFA5, is depleted of segmented filamentous bacteria.222 Intriguingly, this class of bacteria induces IL-17-producing T-helper (Th17) cells, which have been strongly linked to IBD and colorectal cancer.60,178,222,224,225 In fact, B. fragilis enterotoxin promotes colorectal carcinogenesis through IL-17,60 and B. fragilis prostate-specific antigen (PSA) exerts protection by suppressing Th17 differentiation.226
These numerous reports highlight the critical role of the intestinal microbiota in shaping intestinal health. Members of this microbial community can modulate immune and inflammatory responses, activate and detoxify carcinogens, promote DNA damage and chromosomal instability, shift the balance of proliferation and apoptosis, and thwart pathogen invasion.
FACTORS INFLUENCING MICROBIAL COMMUNITY STRUCTURE
The intestinal microbiota is a dynamic community strongly influenced by external forces including genetic factors, inflammation and infection, antibiotic treatment, and diet.16,29,30,227-231 Metagenomics studies have indicated greater similarity between the intestinal microbiota of family members versus unrelated individuals.14,229 Yet analyses of congenic mice bred from heterozygotes and raised in the same cage have revealed that deficiency of even one host gene can significantly change intestinal microbial community composition. For example, deficiency of the Crohn’s disease-associated Nod2 gene increases the total number of commensal bacteria and reduces the ability of these mice to clear H. hepaticus.119 NOD1-deficient mice harbor an increased total number of commensal bacteria, yet a decreased number of protective Lactobacillus species.232 MyD88-deficient NOD diabetic mice exhibit microbiota changes at the family level and in the proportion of the two prominent phyla, the Firmicutes and Bacteroidetes. Furthermore, deficiency of MyD88 allows bacterial penetration of the intestinal epithelial barrier and systemic dissemination.233,234 Taken together, these data suggest that host innate signaling influences the microbial community structure, which in turn shapes intestinal homeostasis.
Alterations in microbial community structure occur rapidly upon changes in dietary habits.29,30,235,236 Gordon and colleagues found that the intestinal microbial community structure of mice maintained on standard low-fat chow could be changed significantly within weeks of switching to a high-fat diet (HFD). Consumption of HFD was associated with a shift in the balance of the two dominant phyla, the Bacteroidetes and Firmicutes.29,30 In fact, the most prominent change was an expansion in one group of Firmicutes, the Mollicutes, which again contracted when mice were switched back to standard low-fat chow. The Mollicutes expansion was associated with an increased ability for energy harvest, and this trait could be transferred by microbiota transplant into lean recipients.29,30 Importantly, similar changes in the proportion of Firmicutes and Bacteroidetes have been identified in overweight and obese humans, genetically obese mice, and obesity-resistant mice fed HFD diet.13,14,28-30 As human colorectal cancer is tightly linked to diet,237,238 it will be interesting to see if diet-induced changes in the microbiota also influence the development of colorectal cancer.
CONCLUSIONS AND FUTURE DIRECTIONS
The events leading to the development of colorectal cancer are complex. Adding to the complexity of the disease is the growing evidence that the microbiota and its associated microbiome are active participants in the pathology. Within recent years, we have experienced exponential growth in our understanding of how the microbiota affects intestinal health and diseases. Gastrointestinal diseases including esophagitis/Barrett’s esophagus, gastric cancer, IBD, and colorectal cancer have been strongly linked to alterations in the composition of the gut microbiota.16,37,47,212,239 Although tantalizing, these observations have not moved beyond the descriptive phase and into functional and malleable territory. Consequently, numerous questions remain unanswered.
The most pressing question is if the shift in the microbiota directly alters the course of disease. Functional studies using dysbiotic microbiota obtained from germ-free animals and various disease states—inflammation, colorectal cancer, etc.—will help address this important question.
Another key question relates to the identity of the microorganisms promoting health and/or disease. Continuous progress in next-generation gene sequencing technology and more affordable cost associated with this high-throughput technique will likely yield important information regarding the composition of healthy and dysbiotic microbiota. This information may be sufficient to initiate functional experiments where a cause/effect relationship could be established using animal models. The fact that a “core microbiome” was recently identified in healthy individuals and that deviation from this core is associated with IBD16-18 indicate that a potential dysbiotic microbiota could be found in colorectal cancer patients and tested in experimental models.
Although identification of microbial consortia associated with particular pathological conditions represents an important milestone, this critical step is not sufficient to fully comprehend the role of the microbiota in health and disease. Indeed, the in-depth characterization of the metabolic capacity contributed by the microbiota in relation to the maintenance of intestinal homeostasis and development of disease states is undeniably a central piece of the metagenomic puzzle. With this knowledge will come the possibility to manipulate the human microbiota and its metabolic capacity as an innovative approach to treating and preventing IBD and colorectal cancer.
In summary, the microbiota can no longer be considered a bystander in the complex biological events regulating intestinal homeostasis. Both the composition of the microbiota and its associated metabolic capacity likely influence host susceptibility to developing various pathological conditions, including IBD and colorectal cancer (Fig. 2). Harnessing the power of the microbiota holds tremendous promise in medicine and could represent a novel means to alleviate these devastating diseases.
FIGURE 2.
Microbial dysbiosis is associated with a variety of diseases including IBD and colorectal cancer. In a healthy individual a balanced microbial community structure promotes the maintenance of intestinal homeostasis. The presence of a dysbiotic microbiota that is associated with loss of protective species and predominance of adherent/invasive species promotes inflammation, activation of innate and adaptive immunity, and loss of barrier function in a susceptible host. Longstanding IBD and persistence of microbial dysbiosis may encourage genetic changes, loss of cell cycle control, and immune dysfunction that promote neoplastic changes and development of colorectal cancer.
ACKNOWLEDGMENT
The authors thank Dr. John D. Lich for critical reading of the article.
Supported by National Institutes of Health (NIH) RO1 grants DK047700 and DK073338 to C. Jobin and by NIH T32 Fellowship DK007737 to J. Arthur.
REFERENCES
- 1.Rakoff-Nahoum S, Hao L, Medzhitov R. Role of Toll-like receptors in spontaneous commensal-dependent colitis. Immunity. 2006;25:319–329. doi: 10.1016/j.immuni.2006.06.010. [DOI] [PubMed] [Google Scholar]
- 2.Sellon RK, Tonkonogy S, Schultz M, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66:5224–5231. doi: 10.1128/iai.66.11.5224-5231.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Taurog JD, Richardson JA, Croft JT, et al. The germfree state prevents development of gut and joint inflammatory disease in HLA-B27 transgenic rats. J Exp Med. 1994;180:2359–2364. doi: 10.1084/jem.180.6.2359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Dianda L, Hanby AM, Wright NA, et al. T cell receptor-alpha beta-deficient mice fail to develop colitis in the absence of a microbial environment. Am J Pathol. 1997;150:91–97. [PMC free article] [PubMed] [Google Scholar]
- 5.Mathew CG. New links to the pathogenesis of Crohn disease provided by genome-wide association scans. Nat Rev Genet. 2008;9:9–14. doi: 10.1038/nrg2203. [DOI] [PubMed] [Google Scholar]
- 6.Cho JH. The genetics and immunopathogenesis of inflammatory bowel disease. Nat Rev Immunol. 2008;8:458–466. doi: 10.1038/nri2340. [DOI] [PubMed] [Google Scholar]
- 7.Neish AS. Microbes in gastrointestinal health and disease. Gastroenterology. 2009;136:65–80. doi: 10.1053/j.gastro.2008.10.080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hooper LV, Wong MH, Thelin A, et al. Molecular analysis of commensal host-microbial relationships in the intestine. Science. 2001;291:881–884. doi: 10.1126/science.291.5505.881. [DOI] [PubMed] [Google Scholar]
- 9.Hooper LV, Gordon JI. Commensal host-bacterial relationships in the gut. Science. 2001;292:1115–1118. doi: 10.1126/science.1058709. [DOI] [PubMed] [Google Scholar]
- 10.Hamady M, Knight R. Microbial community profiling for human microbiome projects: tools, techniques, and challenges. Genome Res. 2009;19:1141–1152. doi: 10.1101/gr.085464.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hamady M, Walker JJ, Harris JK, et al. Error-correcting barcoded primers for pyrosequencing hundreds of samples in multiplex. Nat Methods. 2008;5:235–237. doi: 10.1038/nmeth.1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–1638. doi: 10.1126/science.1110591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Turnbaugh PJ, Hamady M, Yatsunenko T, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480–484. doi: 10.1038/nature07540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ley RE, Backhed F, Turnbaugh P, et al. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102:11070–11075. doi: 10.1073/pnas.0504978102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Gill SR, Pop M, Deboy RT, et al. Metagenomic analysis of the human distal gut microbiome. Science. 2006;312:1355–1359. doi: 10.1126/science.1124234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Frank DN, St Amand AL, Feldman RA, et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A. 2007;104:13780–13785. doi: 10.1073/pnas.0706625104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Turnbaugh PJ, Ley RE, Hamady M, et al. The human microbiome project. Nature. 2007;449:804–810. doi: 10.1038/nature06244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59–65. doi: 10.1038/nature08821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Crawford PA, Crowley JR, Sambandam N, et al. Regulation of myocardial ketone body metabolism by the gut microbiota during nutrient deprivation. Proc Natl Acad Sci U S A. 2009;106:11276–11281. doi: 10.1073/pnas.0902366106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Stappenbeck TS, Hooper LV, Gordon JI. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proc Natl Acad Sci U S A. 2002;99:15451–15455. doi: 10.1073/pnas.202604299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Clarke TB, Davis KM, Lysenko ES, et al. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nat Med. 2010;16:228–231. doi: 10.1038/nm.2087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Strober W. The multifaceted influence of the mucosal microflora on mucosal dendritic cell responses. Immunity. 2009;31:377–388. doi: 10.1016/j.immuni.2009.09.001. [DOI] [PubMed] [Google Scholar]
- 23.Chervonsky AV. Influence of microbial environment on autoimmunity. Nat Immunol. 2010;11:28–35. doi: 10.1038/ni.1801. [DOI] [PubMed] [Google Scholar]
- 24.Eberl G, Lochner M. The development of intestinal lymphoid tissues at the interface of self and microbiota. Mucosal Immunol. 2009;2:478–485. doi: 10.1038/mi.2009.114. [DOI] [PubMed] [Google Scholar]
- 25.Cani PD, Delzenne NM. Interplay between obesity and associated metabolic disorders: new insights into the gut microbiota. Curr Opin Pharmacol. 2009;9:737–743. doi: 10.1016/j.coph.2009.06.016. [DOI] [PubMed] [Google Scholar]
- 26.Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient environment of the mammalian intestine. Annu Rev Nutr. 2002;22:283–307. doi: 10.1146/annurev.nutr.22.011602.092259. [DOI] [PubMed] [Google Scholar]
- 27.Hooper LV. Bacterial contributions to mammalian gut development. Trends Microbiol. 2004;12:129–134. doi: 10.1016/j.tim.2004.01.001. [DOI] [PubMed] [Google Scholar]
- 28.Ley RE, Turnbaugh PJ, Klein S, et al. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–1023. doi: 10.1038/4441022a. [DOI] [PubMed] [Google Scholar]
- 29.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;3:213–223. doi: 10.1016/j.chom.2008.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–1031. doi: 10.1038/nature05414. [DOI] [PubMed] [Google Scholar]
- 31.Dumas ME, Barton RH, Toye A, et al. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc Natl Acad Sci U S A. 2006;103:12511–12516. doi: 10.1073/pnas.0601056103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wen L, Ley RE, Volchkov PY, et al. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature. 2008;455:1109–1113. doi: 10.1038/nature07336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Larsen N, Vogensen FK, van den Berg FW, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;5:e9085. doi: 10.1371/journal.pone.0009085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Sidhu H, Allison MJ, Chow JM, et al. Rapid reversal of hyperoxaluria in a rat model after probiotic administration of Oxalobacter formigenes. J Urol. 2001;166:1487–1491. [PubMed] [Google Scholar]
- 35.Abdollahi-Roodsaz S, Joosten LA, Koenders MI, et al. Stimulation of TLR2 and TLR4 differentially skews the balance of T cells in a mouse model of arthritis. J Clin Invest. 2008;118:205–216. doi: 10.1172/JCI32639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tannock GW. Molecular analysis of the intestinal microflora in IBD. Mucosal Immunol. 2008;1(suppl 1):S15–18. doi: 10.1038/mi.2008.54. [DOI] [PubMed] [Google Scholar]
- 37.Uronis JM, Jobin C. Microbes and colorectal cancer: is there a relationship? Curr Oncol. 2009;16:22–24. doi: 10.3747/co.v16i4.472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ott SJ, Musfeldt M, Wenderoth DF, et al. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004;53:685–693. doi: 10.1136/gut.2003.025403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ott SJ, Plamondon S, Hart A, et al. Dynamics of the mucosa-associated flora in ulcerative colitis patients during remission and clinical relapse. J Clin Microbiol. 2008;46:3510–3513. doi: 10.1128/JCM.01512-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Manichanh C, Rigottier-Gois L, Bonnaud E, et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut. 2006;55:205–211. doi: 10.1136/gut.2005.073817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Garrett WS, Lord GM, Punit S, et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell. 2007;131:33–45. doi: 10.1016/j.cell.2007.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Garrett WS, Punit S, Gallini CA, et al. Colitis-associated colorectal cancer driven by T-bet deficiency in dendritic cells. Cancer Cell. 2009;16:208–219. doi: 10.1016/j.ccr.2009.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Vijay-Kumar M, Aitken JD, Carvalho FA, et al. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science. 2010;328:228–231. doi: 10.1126/science.1179721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Moser AR, Pitot HC, Dove WF. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science. 1990;247:322–324. doi: 10.1126/science.2296722. [DOI] [PubMed] [Google Scholar]
- 45.Su LK, Kinzler KW, Vogelstein B, et al. Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science. 1992;256:668–670. doi: 10.1126/science.1350108. [DOI] [PubMed] [Google Scholar]
- 46.Dove WF, Clipson L, Gould KA, et al. Intestinal neoplasia in the ApcMin mouse: independence from the microbial and natural killer (beige locus) status. Cancer Res. 1997;57:812–814. [PubMed] [Google Scholar]
- 47.Uronis JM, Muhlbauer M, Herfarth HH, et al. Modulation of the intestinal microbiota alters colitis-associated colorectal cancer susceptibility. PLoS One. 2009;4:e6026. doi: 10.1371/journal.pone.0006026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Rao VP, Poutahidis T, Ge Z, et al. Innate immune inflammatory response against enteric bacteria Helicobacter hepaticus induces mammary adenocarcinoma in mice. Cancer Res. 2006;66:7395–7400. doi: 10.1158/0008-5472.CAN-06-0558. [DOI] [PubMed] [Google Scholar]
- 49.Erdman SE, Poutahidis T, Tomczak M, et al. CD4+ CD25+ regulatory T lymphocytes inhibit microbially induced colon cancer in Rag2-deficient mice. Am J Pathol. 2003;162:691–702. doi: 10.1016/S0002-9440(10)63863-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Erdman SE, Rao VP, Poutahidis T, et al. CD4(+)CD25(+) regulatory lymphocytes require interleukin 10 to interrupt colon carcinogenesis in mice. Cancer Res. 2003;63:6042–6050. [PubMed] [Google Scholar]
- 51.Maggio-Price L, Bielefeldt-Ohmann H, Treuting P, et al. Dual infection with Helicobacter bilis and Helicobacter hepaticus in p-glycoprotein-deficient mdr1a−/− mice results in colitis that progresses to dysplasia. Am J Pathol. 2005;166:1793–1806. doi: 10.1016/S0002-9440(10)62489-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Maggio-Price L, Shows D, Waggie K, et al. Helicobacter bilis infection accelerates and H. hepaticus infection delays the development of colitis in multiple drug resistance-deficient (mdr1a−/−) mice. Am J Pathol. 2002;160:739–751. doi: 10.1016/S0002-9440(10)64894-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Chichlowski M, Sharp JM, Vanderford DA, et al. Helicobacter typhlonius and Helicobacter rodentium differentially affect the severity of colon inflammation and inflammation-associated neoplasia in IL10-deficient mice. Comp Med. 2008;58:534–541. [PMC free article] [PubMed] [Google Scholar]
- 54.Waisberg J, Matheus Cde O, Pimenta J. Infectious endocarditis from Streptococcus bovis associated with colonic carcinoma: case report and literature review. Arq Gastroenterol. 2002;39:177–180. doi: 10.1590/s0004-28032002000300008. [DOI] [PubMed] [Google Scholar]
- 55.Gold JS, Bayar S, Salem RR. Association of Streptococcus bovis bacteremia with colonic neoplasia and extracolonic malignancy. Arch Surg. 2004;139:760–765. doi: 10.1001/archsurg.139.7.760. [DOI] [PubMed] [Google Scholar]
- 56.Klein RS, Recco RA, Catalano MT, et al. Association of Streptococcus bovis with carcinoma of the colon. N Engl J Med. 1977;297:800–802. doi: 10.1056/NEJM197710132971503. [DOI] [PubMed] [Google Scholar]
- 57.Zarkin BA, Lillemoe KD, Cameron JL, et al. The triad of Streptococcus bovis bacteremia, colonic pathology, and liver disease. Ann Surg. 1990;211:786–791. doi: 10.1097/00000658-199006000-00019. discussion 791-782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ruoff KL, Miller SI, Garner CV, et al. Bacteremia with Streptococcus bovis and Streptococcus salivarius: clinical correlates of more accurate identification of isolates. J Clin Microbiol. 1989;27:305–308. doi: 10.1128/jcm.27.2.305-308.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ellmerich S, Scholler M, Duranton B, et al. Promotion of intestinal carcinogenesis by Streptococcus bovis. Carcinogenesis. 2000;21:753–756. doi: 10.1093/carcin/21.4.753. [DOI] [PubMed] [Google Scholar]
- 60.Wu S, Rhee KJ, Albesiano E, et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med. 2009;15:1016–1022. doi: 10.1038/nm.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Martin HM, Campbell BJ, Hart CA, et al. Enhanced Escherichia coli adherence and invasion in Crohn’s disease and colon cancer. Gastroenterology. 2004;127:80–93. doi: 10.1053/j.gastro.2004.03.054. [DOI] [PubMed] [Google Scholar]
- 62.Luperchio SA, Schauer DB. Molecular pathogenesis of Citrobacter rodentium and transmissible murine colonic hyperplasia. Microbes Infect. 2001;3:333–340. doi: 10.1016/s1286-4579(01)01387-9. [DOI] [PubMed] [Google Scholar]
- 63.Barthold SW, Jonas AM. Morphogenesis of early 1, 2-dimethylhydrazine-induced lesions and latent period reduction of colon carcinogenesis in mice by a variant of Citrobacter freundii. Cancer Res. 1977;37:4352–4360. [PubMed] [Google Scholar]
- 64.Newman JV, Kosaka T, Sheppard BJ, et al. Bacterial infection promotes colon tumorigenesis in ApcMin/+ mice. J Infect Dis. 2001;184:227–230. doi: 10.1086/321998. [DOI] [PubMed] [Google Scholar]
- 65.Wotherspoon AC, Doglioni C, Diss TC, et al. Regression of primary low-grade B-cell gastric lymphoma of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori. Lancet. 1993;342:575–577. doi: 10.1016/0140-6736(93)91409-f. [DOI] [PubMed] [Google Scholar]
- 66.Wotherspoon AC, Ortiz-Hidalgo C, Falzon MR, et al. Helicobacter pylori-associated gastritis and primary B-cell gastric lymphoma. Lancet. 1991;338:1175–1176. doi: 10.1016/0140-6736(91)92035-z. [DOI] [PubMed] [Google Scholar]
- 67.Weber DM, Dimopoulos MA, Anandu DP, et al. Regression of gastric lymphoma of mucosa-associated lymphoid tissue with antibiotic therapy for Helicobacter pylori. Gastroenterology. 1994;107:1835–1838. doi: 10.1016/0016-5085(94)90828-1. [DOI] [PubMed] [Google Scholar]
- 68.Montalban C, Santon A, Boixeda D, et al. Regression of gastric high grade mucosa associated lymphoid tissue (MALT) lymphoma after Helicobacter pylori eradication. Gut. 2001;49:584–587. doi: 10.1136/gut.49.4.584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Erdman SE, Correa P, Coleman LA, et al. Helicobacter mustelae-associated gastric MALT lymphoma in ferrets. Am J Pathol. 1997;151:273–280. [PMC free article] [PubMed] [Google Scholar]
- 70.Marshall BJ, Warren JR. Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet. 1984;1:1311–1315. doi: 10.1016/s0140-6736(84)91816-6. [DOI] [PubMed] [Google Scholar]
- 71.Watanabe T, Tada M, Nagai H, et al. Helicobacter pylori infection induces gastric cancer in mongolian gerbils. Gastroenterology. 1998;115:642–648. doi: 10.1016/s0016-5085(98)70143-x. [DOI] [PubMed] [Google Scholar]
- 72.Kobayashi M, Lee H, Schaffer L, et al. A distinctive set of genes is upregulated during the inflammation-carcinoma sequence in mouse stomach infected by Helicobacter felis. J Histochem Cytochem. 2007;55:263–274. doi: 10.1369/jhc.6A7097.2006. [DOI] [PubMed] [Google Scholar]
- 73.SJ Hooper, Crean SJ, Lewis MA, et al. Viable bacteria present within oral squamous cell carcinoma tissue. J Clin Microbiol. 2006;44:1719–1725. doi: 10.1128/JCM.44.5.1719-1725.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Narikiyo M, Tanabe C, Yamada Y, et al. Frequent and preferential infection of Treponema denticola, Streptococcus mitis, and Streptococcus anginosus in esophageal cancers. Cancer Sci. 2004;95:569–574. doi: 10.1111/j.1349-7006.2004.tb02488.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Morita E, Narikiyo M, Yano A, et al. Different frequencies of Streptococcus anginosus infection in oral cancer and esophageal cancer. Cancer Sci. 2003;94:492–496. doi: 10.1111/j.1349-7006.2003.tb01471.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Sasaki H, Ishizuka T, Muto M, et al. Presence of Streptococcus anginosus DNA in esophageal cancer, dysplasia of esophagus, and gastric cancer. Cancer Res. 1998;58:2991–2995. [PubMed] [Google Scholar]
- 77.Sasaki M, Yamaura C, Ohara-Nemoto Y, et al. Streptococcus anginosus infection in oral cancer and its infection route. Oral Dis. 2005;11:151–156. doi: 10.1111/j.1601-0825.2005.01051.x. [DOI] [PubMed] [Google Scholar]
- 78.Ward JM, Fox JG, Anver MR, et al. Chronic active hepatitis and associated liver tumors in mice caused by a persistent bacterial infection with a novel Helicobacter species. J Natl Cancer Inst. 1994;86:1222–1227. doi: 10.1093/jnci/86.16.1222. [DOI] [PubMed] [Google Scholar]
- 79.Theve EJ, Feng Y, Taghizadeh K, et al. Sex hormone influence on hepatitis in young male A/JCr mice infected with Helicobacter hepaticus. Infect Immun. 2008;76:4071–4078. doi: 10.1128/IAI.00401-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sipowicz MA, Weghorst CM, Shiao YH, et al. Lack of p53 and ras mutations in Helicobacter hepaticus-induced liver tumors in A/JCr mice. Carcinogenesis. 1997;18:233–236. doi: 10.1093/carcin/18.1.233. [DOI] [PubMed] [Google Scholar]
- 81.Lazcano-Ponce EC, Miquel JF, Munoz N, et al. Epidemiology and molecular pathology of gallbladder cancer. CA Cancer J Clin. 2001;51:349–364. doi: 10.3322/canjclin.51.6.349. [DOI] [PubMed] [Google Scholar]
- 82.Welton JC, Marr JS, Friedman SM. Association between hepatobiliary cancer and typhoid carrier state. Lancet. 1979;1:791–794. doi: 10.1016/s0140-6736(79)91315-1. [DOI] [PubMed] [Google Scholar]
- 83.Caygill CP, Hill MJ, Braddick M, et al. Cancer mortality in chronic typhoid and paratyphoid carriers. Lancet. 1994;343:83–84. doi: 10.1016/s0140-6736(94)90816-8. [DOI] [PubMed] [Google Scholar]
- 84.Dutta U, Garg PK, Kumar R, et al. Typhoid carriers among patients with gallstones are at increased risk for carcinoma of the gallbladder. Am J Gastroenterol. 2000;95:784–787. doi: 10.1111/j.1572-0241.2000.01860.x. [DOI] [PubMed] [Google Scholar]
- 85.Shukla VK, Singh H, Pandey M, et al. Carcinoma of the gallbladder—is it a sequel of typhoid? Dig Dis Sci. 2000;45:900–903. doi: 10.1023/a:1005564822630. [DOI] [PubMed] [Google Scholar]
- 86.Littman AJ, White E, Jackson LA, et al. Chlamydia pneumoniae infection and risk of lung cancer. Cancer Epidemiol Biomarkers Prev. 2004;13:1624–1630. [PubMed] [Google Scholar]
- 87.Koyi H, Branden E, Gnarpe J, et al. An association between chronic infection with Chlamydia pneumoniae and lung cancer. A prospective 2-year study. APMIS. 2001;109:572–580. doi: 10.1034/j.1600-0463.2001.d01-177.x. [DOI] [PubMed] [Google Scholar]
- 88.Kocazeybek B. Chronic Chlamydophila pneumoniae infection in lung cancer, a risk factor: a case-control study. J Med Microbiol. 2003;52:721–726. doi: 10.1099/jmm.0.04845-0. [DOI] [PubMed] [Google Scholar]
- 89.Verbeke P, Welter-Stahl L, Ying S, et al. Recruitment of BAD by the Chlamydia trachomatis vacuole correlates with host-cell survival. PLoS Pathog. 2006;2:e45. doi: 10.1371/journal.ppat.0020045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Fischer SF, Harlander T, Vier J, et al. Protection against CD95-induced apoptosis by chlamydial infection at a mitochondrial step. Infect Immun. 2004;72:1107–1115. doi: 10.1128/IAI.72.2.1107-1115.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Rajalingam K, Al-Younes H, Muller A, et al. Epithelial cells infected with Chlamydophila pneumoniae (Chlamydia pneumoniae) are resistant to apoptosis. Infect Immun. 2001;69:7880–7888. doi: 10.1128/IAI.69.12.7880-7888.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Smith DG, Lawson GH. Lawsonia intracellularis: getting inside the pathogenesis of proliferative enteropathy. Vet Microbiol. 2001;82:331–345. doi: 10.1016/s0378-1135(01)00397-2. [DOI] [PubMed] [Google Scholar]
- 93.Tsai S, Wear DJ, Shih JW, et al. Mycoplasmas and oncogenesis: persistent infection and multistage malignant transformation. Proc Natl Acad Sci U S A. 1995;92:10197–10201. doi: 10.1073/pnas.92.22.10197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Feng SH, Tsai S, Rodriguez J, et al. Mycoplasmal infections prevent apoptosis and induce malignant transformation of interleukin-3-dependent 32D hematopoietic cells. Mol Cell Biol. 1999;19:7995–8002. doi: 10.1128/mcb.19.12.7995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Gerlic M, Horowitz J, Horowitz S. Mycoplasma fermentans inhibits tumor necrosis factor alpha-induced apoptosis in the human myelomonocytic U937 cell line. Cell Death Differ. 2004;11:1204–1212. doi: 10.1038/sj.cdd.4401482. [DOI] [PubMed] [Google Scholar]
- 96.Zhang B, Shih JW, Wear DJ, et al. High-level expression of H-ras and c-myc oncogenes in mycoplasma-mediated malignant cell transformation. Proc Soc Exp Biol Med. 1997;214:359–366. doi: 10.3181/00379727-214-44104. [DOI] [PubMed] [Google Scholar]
- 97.Dehio C, Sander A. Bartonella as emerging pathogens. Trends Microbiol. 1999;7:226–228. doi: 10.1016/s0966-842x(99)01523-1. [DOI] [PubMed] [Google Scholar]
- 98.Geijtenbeek TB, van Vliet SJ, Engering A, et al. Self- and nonself-recognition by C-type lectins on dendritic cells. Annu Rev Immunol. 2004;22:33–54. doi: 10.1146/annurev.immunol.22.012703.104558. [DOI] [PubMed] [Google Scholar]
- 99.Abreu MT, Fukata M, Arditi M. TLR signaling in the gut in health and disease. J Immunol. 2005;174:4453–4460. doi: 10.4049/jimmunol.174.8.4453. [DOI] [PubMed] [Google Scholar]
- 100.Ishii KJ, Akira S. Potential link between the immune system and metabolism of nucleic acids. Curr Opin Immunol. 2008;20:524–529. doi: 10.1016/j.coi.2008.07.002. [DOI] [PubMed] [Google Scholar]
- 101.Rakoff-Nahoum S, Medzhitov R. Innate immune recognition of the indigenous microbial flora. Mucosal Immunol. 2008;1(suppl 1):S10–14. doi: 10.1038/mi.2008.49. [DOI] [PubMed] [Google Scholar]
- 102.Carpenter S, O’Neill LA. How important are Toll-like receptors for antimicrobial responses? Cell Microbiol. 2007;9:1891–1901. doi: 10.1111/j.1462-5822.2007.00965.x. [DOI] [PubMed] [Google Scholar]
- 103.Le Bourhis L, Benko S, Girardin SE. Nod1 and Nod2 in innate immunity and human inflammatory disorders. Biochem Soc Trans. 2007;35:1479–1484. doi: 10.1042/BST0351479. [DOI] [PubMed] [Google Scholar]
- 104.Kopp E, Medzhitov R. Recognition of microbial infection by Toll-like receptors. Curr Opin Immunol. 2003;15:396–401. doi: 10.1016/s0952-7915(03)00080-3. [DOI] [PubMed] [Google Scholar]
- 105.Medzhitov R, Preston-Hurlburt P, Kopp E, et al. MyD88 is an adaptor protein in the hToll/IL-1 receptor family signaling pathways. Mol Cell. 1998;2:253–258. doi: 10.1016/s1097-2765(00)80136-7. [DOI] [PubMed] [Google Scholar]
- 106.Karrasch T, Kim JS, Muhlbauer M, et al. Gnotobiotic IL-10−/−;NF-kappa BEGFP mice reveal the critical role of TLR/NF-kappa B signaling in commensal bacteria-induced colitis. J Immunol. 2007;178:6522–6532. doi: 10.4049/jimmunol.178.10.6522. [DOI] [PubMed] [Google Scholar]
- 107.Rakoff-Nahoum S, Medzhitov R. Regulation of spontaneous intestinal tumorigenesis through the adaptor protein MyD88. Science. 2007;317:124–127. doi: 10.1126/science.1140488. [DOI] [PubMed] [Google Scholar]
- 108.Brown SL, Riehl TE, Walker MR, et al. Myd88-dependent positioning of Ptgs2-expressing stromal cells maintains colonic epithelial proliferation during injury. J Clin Invest. 2007;117:258–269. doi: 10.1172/JCI29159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Pull SL, Doherty JM, Mills JC, et al. Activated macrophages are an adaptive element of the colonic epithelial progenitor niche necessary for regenerative responses to injury. Proc Natl Acad Sci U S A. 2005;102:99–104. doi: 10.1073/pnas.0405979102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Fukata M, Chen A, Vamadevan AS, et al. Toll-like receptor-4 promotes the development of colitis-associated colorectal tumors. Gastroenterology. 2007;133:1869–1881. doi: 10.1053/j.gastro.2007.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Fukata M, Hernandez Y, Conduah D, et al. Innate immune signaling by Toll-like receptor-4 (TLR4) shapes the inflammatory microenvironment in colitis-associated tumors. Inflamm Bowel Dis. 2009;15:997–1006. doi: 10.1002/ibd.20880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Villani AC, Lemire M, Fortin G, et al. Common variants in the NLRP3 region contribute to Crohn’s disease susceptibility. Nat Genet. 2009;41:71–76. doi: 10.1038/ng285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Hugot JP, Chamaillard M, Zouali H, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature. 2001;411:599–603. doi: 10.1038/35079107. [DOI] [PubMed] [Google Scholar]
- 114.Ogura Y, Bonen DK, Inohara N, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature. 2001;411:603–606. doi: 10.1038/35079114. [DOI] [PubMed] [Google Scholar]
- 115.McGovern DP, Hysi P, Ahmad T, et al. Association between a complex insertion/deletion polymorphism in NOD1 (CARD4) and susceptibility to inflammatory bowel disease. Hum Mol Genet. 2005;14:1245–1250. doi: 10.1093/hmg/ddi135. [DOI] [PubMed] [Google Scholar]
- 116.De Jager PL, Franchimont D, Waliszewska A, et al. The role of the Toll receptor pathway in susceptibility to inflammatory bowel diseases. Genes Immun. 2007;8:387–397. doi: 10.1038/sj.gene.6364398. [DOI] [PubMed] [Google Scholar]
- 117.Watanabe T, Asano N, Murray PJ, et al. Muramyl dipeptide activation of nucleotide-binding oligomerization domain 2 protects mice from experimental colitis. J Clin Invest. 2008;118:545–559. doi: 10.1172/JCI33145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Yang Z, Fuss IJ, Watanabe T, et al. NOD2 transgenic mice exhibit enhanced MDP-mediated down-regulation of TLR2 responses and resistance to colitis induction. Gastroenterology. 2007;133:1510–1521. doi: 10.1053/j.gastro.2007.07.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Petnicki-Ocwieja T, Hrncir T, Liu YJ, et al. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci U S A. 2009;106:15813–15818. doi: 10.1073/pnas.0907722106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Tian Y, Li Y, Hu Z, et al. Differential effects of NOD2 polymorphisms on colorectal cancer risk: a meta-analysis. Int J Colorectal Dis. 2010;25:161–168. doi: 10.1007/s00384-009-0809-9. [DOI] [PubMed] [Google Scholar]
- 121.Mockelmann N, von Schonfels W, Buch S, et al. Investigation of innate immunity genes CARD4, CARD8 and CARD15 as germline susceptibility factors for colorectal cancer. BMC Gastroenterol. 2009;9:79. doi: 10.1186/1471-230X-9-79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Chen GY, Shaw MH, Redondo G, et al. The innate immune receptor Nod1 protects the intestine from inflammation-induced tumorigenesis. Cancer Res. 2008;68:10060–10067. doi: 10.1158/0008-5472.CAN-08-2061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Allen IC, McElvania Tekippe E, Woodford R-MT, et al. The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis associated cancer. J Exp Med. 2010 doi: 10.1084/jem.20100050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Grivennikov S, Karin E, Terzic J, et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell. 2009;15:103–113. doi: 10.1016/j.ccr.2009.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Uronis JM, Muehlbauer M, Keku TO, et al. Intestinal inflammation is not sufficient to promote the development of colitis associated colorectal cancer: role of the enteric microbiota. Digestive Diseases Week; New Orleans, LA: 2010. [Google Scholar]
- 126.Rutter MD, Saunders BP, Wilkinson KH, et al. Cancer surveillance in longstanding ulcerative colitis: endoscopic appearances help predict cancer risk. Gut. 2004;53:1813–1816. doi: 10.1136/gut.2003.038505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut. 2001;48:526–535. doi: 10.1136/gut.48.4.526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Toprak NU, Yagci A, Gulluoglu BM, et al. A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer. Clin Microbiol Infect. 2006;12:782–786. doi: 10.1111/j.1469-0691.2006.01494.x. [DOI] [PubMed] [Google Scholar]
- 129.Ge Z, Rogers AB, Feng Y, et al. Bacterial cytolethal distending toxin promotes the development of dysplasia in a model of microbially induced hepatocarcinogenesis. Cell Microbiol. 2007;9:2070–2080. doi: 10.1111/j.1462-5822.2007.00939.x. [DOI] [PubMed] [Google Scholar]
- 130.Haghjoo E, Galan JE. Salmonella typhi encodes a functional cytolethal distending toxin that is delivered into host cells by a bacterial-internalization pathway. Proc Natl Acad Sci U S A. 2004;101:4614–4619. doi: 10.1073/pnas.0400932101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Bielaszewska M, Sinha B, Kuczius T, et al. Cytolethal distending toxin from Shiga toxin-producing Escherichia coli O157 causes irreversible G2/M arrest, inhibition of proliferation, and death of human endothelial cells. Infect Immun. 2005;73:552–562. doi: 10.1128/IAI.73.1.552-562.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Comayras C, Tasca C, Peres SY, et al. Escherichia coli cytolethal distending toxin blocks the HeLa cell cycle at the G2/M transition by preventing cdc2 protein kinase dephosphorylation and activation. Infect Immun. 1997;65:5088–5095. doi: 10.1128/iai.65.12.5088-5095.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Sert V, Cans C, Tasca C, et al. The bacterial cytolethal distending toxin (CDT) triggers a G2 cell cycle checkpoint in mammalian cells without preliminary induction of DNA strand breaks. Oncogene. 1999;18:6296–6304. doi: 10.1038/sj.onc.1203007. [DOI] [PubMed] [Google Scholar]
- 134.Marches O, Ledger TN, Boury M, et al. Enteropathogenic and enterohaemorrhagic Escherichia coli deliver a novel effector called Cif, which blocks cell cycle G2/M transition. Mol Microbiol. 2003;50:1553–1567. doi: 10.1046/j.1365-2958.2003.03821.x. [DOI] [PubMed] [Google Scholar]
- 135.Samba-Louaka A, Nougayrede JP, Watrin C, et al. The enteropathogenic Escherichia coli effector Cif induces delayed apoptosis in epithelial cells. Infect Immun. 2009;77:5471–5477. doi: 10.1128/IAI.00860-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Jubelin G, Chavez CV, Taieb F, et al. Cycle inhibiting factors (CIFs) are a growing family of functional cyclomodulins present in invertebrate and mammal bacterial pathogens. PLoS One. 2009;4:e4855. doi: 10.1371/journal.pone.0004855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Nougayrede JP, Boury M, Tasca C, et al. Type III secretion-dependent cell cycle block caused in HeLa cells by enteropathogenic Escherichia coli O103. Infect Immun. 2001;69:6785–6795. doi: 10.1128/IAI.69.11.6785-6795.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Fiorentini C, Matarrese P, Straface E, et al. Toxin-induced activation of Rho GTP-binding protein increases Bcl-2 expression and influences mitochondrial homeostasis. Exp Cell Res. 1998;242:341–350. doi: 10.1006/excr.1998.4057. [DOI] [PubMed] [Google Scholar]
- 139.Miraglia AG, Travaglione S, Meschini S, et al. Cytotoxic necrotizing factor 1 prevents apoptosis via the Akt/IkappaB kinase pathway: role of nuclear factor-kappaB and Bcl-2. Mol Biol Cell. 2007;18:2735–2744. doi: 10.1091/mbc.E06-10-0910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Doye A, Mettouchi A, Bossis G, et al. CNF1 exploits the ubiquitin-proteasome machinery to restrict Rho GTPase activation for bacterial host cell invasion. Cell. 2002;111:553–564. doi: 10.1016/s0092-8674(02)01132-7. [DOI] [PubMed] [Google Scholar]
- 141.Higgins TE, Murphy AC, Staddon JM, et al. Pasteurella multocida toxin is a potent inducer of anchorage-independent cell growth. Proc Natl Acad Sci U S A. 1992;89:4240–4244. doi: 10.1073/pnas.89.10.4240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Lax AJ, Grigoriadis AE. Pasteurella multocida toxin: the mitogenic toxin that stimulates signalling cascades to regulate growth and differentiation. Int J Med Microbiol. 2001;291:261–268. doi: 10.1078/1438-4221-00129. [DOI] [PubMed] [Google Scholar]
- 143.Sugai M, Chen CH, Wu HC. Bacterial ADP-ribosyltransferase with a substrate specificity of the rho protein disassembles the Golgi apparatus in Vero cells and mimics the action of brefeldin A. Proc Natl Acad Sci U S A. 1992;89:8903–8907. doi: 10.1073/pnas.89.19.8903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Sugai M, Hashimoto K, Kikuchi A, et al. Epidermal cell differentiation inhibitor ADP-ribosylates small GTP-binding proteins and induces hyperplasia of epidermis. J Biol Chem. 1992;267:2600–2604. [PubMed] [Google Scholar]
- 145.Bagnoli F, Buti L, Tompkins L, et al. Helicobacter pylori CagA induces a transition from polarized to invasive phenotypes in MDCK cells. Proc Natl Acad Sci U S A. 2005;102:16339–16344. doi: 10.1073/pnas.0502598102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Segal ED, Cha J, Lo J, et al. Altered states: involvement of phosphorylated CagA in the induction of host cellular growth changes by Helicobacter pylori. Proc Natl Acad Sci U S A. 1999;96:14559–14564. doi: 10.1073/pnas.96.25.14559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Peek RM, Jr, Vaezi MF, Falk GW, et al. Role of Helicobacter pylori cagA(+) strains and specific host immune responses on the development of premalignant and malignant lesions in the gastric cardia. Int J Cancer. 1999;82:520–524. doi: 10.1002/(sici)1097-0215(19990812)82:4<520::aid-ijc9>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
- 148.Huang JQ, Zheng GF, Sumanac K, et al. Meta-analysis of the relationship between cagA seropositivity and gastric cancer. Gastroenterology. 2003;125:1636–1644. doi: 10.1053/j.gastro.2003.08.033. [DOI] [PubMed] [Google Scholar]
- 149.Takada H, Hirooka T, Hiramatsu Y, et al. Effect of beta-glucuronidase inhibitor on azoxymethane-induced colonic carcinogenesis in rats. Cancer Res. 1982;42:331–334. [PubMed] [Google Scholar]
- 150.Humblot C, Murkovic M, Rigottier-Gois L, et al. Beta-glucuronidase in human intestinal microbiota is necessary for the colonic genotoxicity of the food-borne carcinogen 2-amino-3-methylimidazo[4,5-f]quinoline in rats. Carcinogenesis. 2007;28:2419–2425. doi: 10.1093/carcin/bgm170. [DOI] [PubMed] [Google Scholar]
- 151.Kim DH, Jin YH. Intestinal bacterial beta-glucuronidase activity of patients with colon cancer. Arch Pharm Res. 2001;24:564–567. doi: 10.1007/BF02975166. [DOI] [PubMed] [Google Scholar]
- 152.Mirvish SS, Haorah J, Zhou L, et al. Total N-nitroso compounds and their precursors in hot dogs and in the gastrointestinal tract and feces of rats and mice: possible etiologic agents for colon cancer. J Nutr. 2002;132:3526S–3529S. doi: 10.1093/jn/132.11.3526S. [DOI] [PubMed] [Google Scholar]
- 153.Bingham SA, Pignatelli B, Pollock JR, et al. Does increased endogenous formation of N-nitroso compounds in the human colon explain the association between red meat and colon cancer? Carcinogenesis. 1996;17:515–523. doi: 10.1093/carcin/17.3.515. [DOI] [PubMed] [Google Scholar]
- 154.Archer MC. Mechanisms of action of N-nitroso compounds. Cancer Surv. 1989;8:241–250. [PubMed] [Google Scholar]
- 155.Xu H, Heinze TM, Chen S, et al. Anaerobic metabolism of 1-amino-2-naphthol-based azo dyes (Sudan dyes) by human intestinal microflora. Appl Environ Microbiol. 2007;73:7759–7762. doi: 10.1128/AEM.01410-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Chung KT, Stevens SE, Jr, Cerniglia CE. The reduction of azo dyes by the intestinal microflora. Crit Rev Microbiol. 1992;18:175–190. doi: 10.3109/10408419209114557. [DOI] [PubMed] [Google Scholar]
- 157.Caderni G, Femia AP, Giannini A, et al. Identification of mucin-depleted foci in the unsectioned colon of azoxymethane-treated rats: correlation with carcinogenesis. Cancer Res. 2003;63:2388–2392. [PubMed] [Google Scholar]
- 158.Femia AP, Dolara P, Caderni G. Mucin-depleted foci (MDF) in the colon of rats treated with azoxymethane (AOM) are useful biomarkers for colon carcinogenesis. Carcinogenesis. 2004;25:277–281. doi: 10.1093/carcin/bgh005. [DOI] [PubMed] [Google Scholar]
- 159.Femia AP, Dolara P, Giannini A, et al. Frequent mutation of Apc gene in rat colon tumors and mucin-depleted foci, preneoplastic lesions in experimental colon carcinogenesis. Cancer Res. 2007;67:445–449. doi: 10.1158/0008-5472.CAN-06-3861. [DOI] [PubMed] [Google Scholar]
- 160.de Moreno de LeBlanc A, LeBlanc JG, Perdigon G, et al. Oral administration of a catalase-producing Lactococcus lactis can prevent a chemically induced colon cancer in mice. J Med Microbiol. 2008;57:100–105. doi: 10.1099/jmm.0.47403-0. [DOI] [PubMed] [Google Scholar]
- 161.Wang X, Allen TD, May RJ, et al. Enterococcus faecalis induces aneuploidy and tetraploidy in colonic epithelial cells through a bystander effect. Cancer Res. 2008;68:9909–9917. doi: 10.1158/0008-5472.CAN-08-1551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.McGarr SE, Ridlon JM, Hylemon PB. Diet, anaerobic bacterial metabolism, and colon cancer: a review of the literature. J Clin Gastroenterol. 2005;39:98–109. [PubMed] [Google Scholar]
- 163.Bayerdorffer E, Mannes GA, Ochsenkuhn T, et al. Unconjugated secondary bile acids in the serum of patients with colorectal adenomas. Gut. 1995;36:268–273. doi: 10.1136/gut.36.2.268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Bayerdorffer E, Mannes GA, Richter WO, et al. Increased serum deoxycholic acid levels in men with colorectal adenomas. Gastroenterology. 1993;104:145–151. doi: 10.1016/0016-5085(93)90846-5. [DOI] [PubMed] [Google Scholar]
- 165.Reddy BS, Watanabe K, Weisburger JH, et al. Promoting effect of bile acids in colon carcinogenesis in germ-free and conventional F344 rats. Cancer Res. 1977;37:3238–3242. [PubMed] [Google Scholar]
- 166.Pai R, Tarnawski AS, Tran T. Deoxycholic acid activates beta-catenin signaling pathway and increases colon cell cancer growth and invasiveness. Mol Biol Cell. 2004;15:2156–2163. doi: 10.1091/mbc.E03-12-0894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Reddy BS, Wynder EL. Metabolic epidemiology of colon cancer. Fecal bile acids and neutral sterols in colon cancer patients and patients with adenomatous polyps. Cancer. 1977;39:2533–2539. doi: 10.1002/1097-0142(197706)39:6<2533::aid-cncr2820390634>3.0.co;2-x. [DOI] [PubMed] [Google Scholar]
- 168.Flynn C, Montrose DC, Swank DL, et al. Deoxycholic acid promotes the growth of colonic aberrant crypt foci. Mol Carcinog. 2007;46:60–70. doi: 10.1002/mc.20253. [DOI] [PubMed] [Google Scholar]
- 169.Laqueur GL, McDaniel EG, Matsumoto H. Tumor induction in germfree rats with methylazoxymethanol (MAM) and synthetic MAM acetate. J Natl Cancer Inst. 1967;39:355–371. [PubMed] [Google Scholar]
- 170.Rowland IR. The role of the gastrointestinal microbiota in colorectal cancer. Curr Pharm Des. 2009;15:1524–1527. doi: 10.2174/138161209788168191. [DOI] [PubMed] [Google Scholar]
- 171.Fiala ES. Investigations into the metabolism and mode of action of the colon carcinogens 1,2-dimethylhydrazine and azoxymethane. Cancer. 1977;40:2436–2445. doi: 10.1002/1097-0142(197711)40:5+<2436::aid-cncr2820400908>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
- 172.Weisburger JH. Colon carcinogens: their metabolism and mode of action. Cancer. 1971;28:60–70. doi: 10.1002/1097-0142(197107)28:1<60::aid-cncr2820280113>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
- 173.Neufert C, Becker C, Neurath MF. An inducible mouse model of colon carcinogenesis for the analysis of sporadic and inflammation-driven tumor progression. Nat Protoc. 2007;2:1998–2004. doi: 10.1038/nprot.2007.279. [DOI] [PubMed] [Google Scholar]
- 174.Van Tassell RL, Kingston DG, Wilkins TD. Metabolism of dietary genotoxins by the human colonic microflora; the fecapentaenes and heterocyclic amines. Mutat Res. 1990;238:209–221. doi: 10.1016/0165-1110(90)90013-2. [DOI] [PubMed] [Google Scholar]
- 175.Vanhaecke L, Vercruysse F, Boon N, et al. Isolation and characterization of human intestinal bacteria capable of transforming the dietary carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine. Appl Environ Microbiol. 2008;74:1469–1477. doi: 10.1128/AEM.02064-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Kassie F, Rabot S, Kundi M, et al. Intestinal microflora plays a crucial role in the genotoxicity of the cooked food mutagen 2-amino-3-methylimidazo [4,5-f]quinoline. Carcinogenesis. 2001;22:1721–1725. doi: 10.1093/carcin/22.10.1721. [DOI] [PubMed] [Google Scholar]
- 177.Knasmuller S, Steinkellner H, Hirschl AM, et al. Impact of bacteria in dairy products and of the intestinal microflora on the genotoxic and carcinogenic effects of heterocyclic aromatic amines. Mutat Res. 2001;480-481:129–138. doi: 10.1016/s0027-5107(01)00176-2. [DOI] [PubMed] [Google Scholar]
- 178.Ivanov II, Frutos Rde L, Manel N, et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe. 2008;4:337–349. doi: 10.1016/j.chom.2008.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Nagamine CM, Rogers AB, Fox JG, et al. Helicobacter hepaticus promotes azoxymethane-initiated colon tumorigenesis in BALB/c-IL10-deficient mice. Int J Cancer. 2008;122:832–838. doi: 10.1002/ijc.23175. [DOI] [PubMed] [Google Scholar]
- 180.Maggio-Price L, Treuting P, Zeng W, et al. Helicobacter infection is required for inflammation and colon cancer in SMAD3-deficient mice. Cancer Res. 2006;66:828–838. doi: 10.1158/0008-5472.CAN-05-2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Hale LP, Perera D, Gottfried MR, et al. Neonatal co-infection with helicobacter species markedly accelerates the development of inflammation-associated colonic neoplasia in IL-10−/− mice. Helicobacter. 2007;12:598–604. doi: 10.1111/j.1523-5378.2007.00552.x. [DOI] [PubMed] [Google Scholar]
- 182.Chichlowski M, Hale LP. Effects of Helicobacter infection on research: the case for eradication of Helicobacter from rodent research colonies. Comp Med. 2009;59:10–17. [PMC free article] [PubMed] [Google Scholar]
- 183.Geier MS, Butler RN, Howarth GS. Probiotics, prebiotics and synbiotics: a role in chemoprevention for colorectal cancer? Cancer Biol Ther. 2006;5:1265–1269. doi: 10.4161/cbt.5.10.3296. [DOI] [PubMed] [Google Scholar]
- 184.Goldin BR, Gorbach SL. Alterations of the intestinal microflora by diet, oral antibiotics, and Lactobacillus: decreased production of free amines from aromatic nitro compounds, azo dyes, and glucuronides. J Natl Cancer Inst. 1984;73:689–695. [PubMed] [Google Scholar]
- 185.Goldin BR, Swenson L, Dwyer J, et al. Effect of diet and Lactobacillus acidophilus supplements on human fecal bacterial enzymes. J Natl Cancer Inst. 1980;64:255–261. doi: 10.1093/jnci/64.2.255. [DOI] [PubMed] [Google Scholar]
- 186.Rowland IR, Rumney CJ, Coutts JT, et al. Effect of Bifidobacterium longum and inulin on gut bacterial metabolism and carcinogen-induced aberrant crypt foci in rats. Carcinogenesis. 1998;19:281–285. doi: 10.1093/carcin/19.2.281. [DOI] [PubMed] [Google Scholar]
- 187.Nowak A, Libudzisz Z. Ability of probiotic Lactobacillus casei DN 114001 to bind or/and metabolise heterocyclic aromatic amines in vitro. Eur J Nutr. 2009;48:419–427. doi: 10.1007/s00394-009-0030-1. [DOI] [PubMed] [Google Scholar]
- 188.Zsivkovits M, Fekadu K, Sontag G, et al. Prevention of heterocyclic amine-induced DNA damage in colon and liver of rats by different lactobacillus strains. Carcinogenesis. 2003;24:1913–1918. doi: 10.1093/carcin/bgg167. [DOI] [PubMed] [Google Scholar]
- 189.Pool-Zobel BL, Bertram B, Knoll M, et al. Antigenotoxic properties of lactic acid bacteria in vivo in the gastrointestinal tract of rats. Nutr Cancer. 1993;20:271–281. doi: 10.1080/01635589309514295. [DOI] [PubMed] [Google Scholar]
- 190.Reddy BS, Rivenson A. Inhibitory effect of Bifidobacterium longum on colon, mammary, and liver carcinogenesis induced by 2-amino-3-methylimidazo[4,5-f]quinoline, a food mutagen. Cancer Res. 1993;53:3914–3918. [PubMed] [Google Scholar]
- 191.Pool-Zobel BL, Neudecker C, Domizlaff I, et al. Lactobacillus- and bifidobacterium-mediated antigenotoxicity in the colon of rats. Nutr Cancer. 1996;26:365–380. doi: 10.1080/01635589609514492. [DOI] [PubMed] [Google Scholar]
- 192.Porschen R, Robin U, Schumacher A, et al. DNA aneuploidy in Crohn’s disease and ulcerative colitis: results of a comparative flow cytometric study. Gut. 1992;33:663–667. doi: 10.1136/gut.33.5.663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Sjoqvist U, Befrits R, Soderlund S, et al. Colorectal cancer in colonic Crohn’s disease—high frequency of DNA-aneuploidy. Anticancer Res. 2005;25:4393–4397. [PubMed] [Google Scholar]
- 194.Rubin CE, Haggitt RC, Burmer GC, et al. DNA aneuploidy in colonic biopsies predicts future development of dysplasia in ulcerative colitis. Gastroenterology. 1992;103:1611–1620. doi: 10.1016/0016-5085(92)91185-7. [DOI] [PubMed] [Google Scholar]
- 195.Balish E, Warner T. Enterococcus faecalis induces inflammatory bowel disease in interleukin-10 knockout mice. Am J Pathol. 2002;160:2253–2257. doi: 10.1016/S0002-9440(10)61172-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Kim SC, Tonkonogy SL, Albright CA, et al. Variable phenotypes of enterocolitis in interleukin 10-deficient mice monoassociated with two different commensal bacteria. Gastroenterology. 2005;128:891–906. doi: 10.1053/j.gastro.2005.02.009. [DOI] [PubMed] [Google Scholar]
- 197.Wang X, Huycke MM. Extracellular superoxide production by Enterococcus faecalis promotes chromosomal instability in mammalian cells. Gastroenterology. 2007;132:551–561. doi: 10.1053/j.gastro.2006.11.040. [DOI] [PubMed] [Google Scholar]
- 198.Chu FF, Esworthy RS, Chu PG, et al. Bacteria-induced intestinal cancer in mice with disrupted Gpx1 and Gpx2 genes. Cancer Res. 2004;64:962–968. doi: 10.1158/0008-5472.can-03-2272. [DOI] [PubMed] [Google Scholar]
- 199.Segain JP, Raingeard de la Bletiere D, Bourreille A, et al. Butyrate inhibits inflammatory responses through NFkappaB inhibition: implications for Crohn’s disease. Gut. 2000;47:397–403. doi: 10.1136/gut.47.3.397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Maslowski KM, Vieira AT, Ng A, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282–1286. doi: 10.1038/nature08530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Scheppach W. Effects of short chain fatty acids on gut morphology and function. Gut. 1994;35:S35–38. doi: 10.1136/gut.35.1_suppl.s35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Heerdt BG, Houston MA, Augenlicht LH. Short-chain fatty acid-initiated cell cycle arrest and apoptosis of colonic epithelial cells is linked to mitochondrial function. Cell Growth Differ. 1997;8:523–532. [PubMed] [Google Scholar]
- 203.Hague A, Elder DJ, Hicks DJ, et al. Apoptosis in colorectal tumour cells: induction by the short chain fatty acids butyrate, propionate and acetate and by the bile salt deoxycholate. Int J Cancer. 1995;60:400–406. doi: 10.1002/ijc.2910600322. [DOI] [PubMed] [Google Scholar]
- 204.Bordonaro M, Lazarova DL, Sartorelli AC. Hyperinduction of Wnt activity: a new paradigm for the treatment of colorectal cancer? Oncol Res. 2008;317:1–9. doi: 10.3727/096504008784046108. [DOI] [PubMed] [Google Scholar]
- 205.Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 2006;5:769–784. doi: 10.1038/nrd2133. [DOI] [PubMed] [Google Scholar]
- 206.Ruemmele FM, Schwartz S, Seidman EG, et al. Butyrate induced Caco-2 cell apoptosis is mediated via the mitochondrial pathway. Gut. 2003;52:94–100. doi: 10.1136/gut.52.1.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Bonnotte B, Favre N, Reveneau S, et al. Cancer cell sensitization to fas-mediated apoptosis by sodium butyrate. Cell Death Differ. 1998;5:480–487. doi: 10.1038/sj.cdd.4400371. [DOI] [PubMed] [Google Scholar]
- 208.Park JY, Helm JF, Zheng W, et al. Silencing of the candidate tumor suppressor gene solute carrier family 5 member 8 (SLC5A8) in human pancreatic cancer. Pancreas. 2008;36:e32–39. doi: 10.1097/MPA.0b013e3181630ffe. [DOI] [PubMed] [Google Scholar]
- 209.Bennett KL, Karpenko M, Lin MT, et al. Frequently methylated tumor suppressor genes in head and neck squamous cell carcinoma. Cancer Res. 2008;68:4494–4499. doi: 10.1158/0008-5472.CAN-07-6509. [DOI] [PubMed] [Google Scholar]
- 210.Whitman SP, Hackanson B, Liyanarachchi S, et al. DNA hypermethylation and epigenetic silencing of the tumor suppressor gene, SLC5A8, in acute myeloid leukemia with the MLL partial tandem duplication. Blood. 2008;112:2013–2016. doi: 10.1182/blood-2008-01-128595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.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;69:2826–2832. doi: 10.1158/0008-5472.CAN-08-4466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Marshall BJ. The 1995 Albert Lasker Medical Research Award. Helicobacter pylori. The etiologic agent for peptic ulcer. JAMA. 1995;274:1064–1066. doi: 10.1001/jama.274.13.1064. [DOI] [PubMed] [Google Scholar]
- 213.Nagamine CM, Sohn JJ, Rickman BH, et al. Helicobacter hepaticus infection promotes colon tumorigenesis in the BALB/c-Rag2−/− ApcMin/+ mouse. Infect Immun. 2008;76:2758–2766. doi: 10.1128/IAI.01604-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Jones SE, Versalovic J. Probiotic Lactobacillus reuteri biofilms produce antimicrobial and anti-inflammatory factors. BMC Microbiol. 2009;9:35. doi: 10.1186/1471-2180-9-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Qin H, Zhang Z, Hang X, et al. L. plantarum prevents enteroinvasive Escherichia coli-induced tight junction proteins changes in intestinal epithelial cells. BMC Microbiol. 2009;9:63. doi: 10.1186/1471-2180-9-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Lievin-Le Moal V, Amsellem R, Servin AL, et al. Lactobacillus acidophilus (strain LB) from the resident adult human gastrointestinal microflora exerts activity against brush border damage promoted by a diarrhoeagenic Escherichia coli in human enterocyte-like cells. Gut. 2002;50:803–811. doi: 10.1136/gut.50.6.803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Resta-Lenert S, Barrett KE. Live probiotics protect intestinal epithelial cells from the effects of infection with enteroinvasive Escherichia coli (EIEC) Gut. 2003;52:988–997. doi: 10.1136/gut.52.7.988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Pagnini C, Saeed R, Bamias G, et al. Probiotics promote gut health through stimulation of epithelial innate immunity. Proc Natl Acad Sci U S A. 2010;107:454–459. doi: 10.1073/pnas.0910307107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Sartor RB. Probiotic therapy of intestinal inflammation and infections. Curr Opin Gastroenterol. 2005;21:44–50. [PubMed] [Google Scholar]
- 220.de Sablet T, Chassard C, Bernalier-Donadille A, et al. Human microbiota-secreted factors inhibit shiga toxin synthesis by enterohemorrhagic Escherichia coli O157:H7. Infect Immun. 2009;77:783–790. doi: 10.1128/IAI.01048-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Cash HL, Whitham CV, Behrendt CL, et al. Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science. 2006;313:1126–1130. doi: 10.1126/science.1127119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Salzman NH, Hung K, Haribhai D, et al. Enteric defensins are essential regulators of intestinal microbial ecology. Nat Immunol. 2010;11:76–83. doi: 10.1038/ni.1825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Hooper LV, Stappenbeck TS, Hong CV, et al. Angiogenins: a new class of microbicidal proteins involved in innate immunity. Nat Immunol. 2003;4:269–273. doi: 10.1038/ni888. [DOI] [PubMed] [Google Scholar]
- 224.Abraham C, Cho J. Interleukin-23/Th17 pathways and inflammatory bowel disease. Inflamm Bowel Dis. 2009;15:1090–1100. doi: 10.1002/ibd.20894. [DOI] [PubMed] [Google Scholar]
- 225.Ivanov II, Atarashi K, Manel N, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139:485–498. doi: 10.1016/j.cell.2009.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453:620–625. doi: 10.1038/nature07008. [DOI] [PubMed] [Google Scholar]
- 227.Preidis GA, Versalovic J. Targeting the human microbiome with antibiotics, probiotics, and prebiotics: gastroenterology enters the metagenomics era. Gastroenterology. 2009;136:2015–2031. doi: 10.1053/j.gastro.2009.01.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Wlodarska M, Finlay BB. Host immune response to antibiotic perturbation of the microbiota. Mucosal Immunol. 2010;3:100–103. doi: 10.1038/mi.2009.135. [DOI] [PubMed] [Google Scholar]
- 229.Turnbaugh PJ, Gordon JI. The core gut microbiome, energy balance and obesity. J Physiol. 2009;587:4153–4158. doi: 10.1113/jphysiol.2009.174136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Hoffmann C, Hill DA, Minkah N, et al. Community-wide response of the gut microbiota to enteropathogenic Citrobacter rodentium infection revealed by deep sequencing. Infect Immun. 2009;77:4668–4678. doi: 10.1128/IAI.00493-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Lupp C, Robertson ML, Wickham ME, et al. Host-mediated inflammation disrupts the intestinal microbiota and promotes the overgrowth of Enterobacteriaceae. Cell Host Microbe. 2007;2:204. doi: 10.1016/j.chom.2007.08.002. [DOI] [PubMed] [Google Scholar]
- 232.Bouskra D, Brezillon C, Berard M, et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature. 2008;456:507–510. doi: 10.1038/nature07450. [DOI] [PubMed] [Google Scholar]
- 233.Vaishnava S, Behrendt CL, Ismail AS, et al. Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface. Proc Natl Acad Sci U S A. 2008;105:20858–20863. doi: 10.1073/pnas.0808723105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Slack E, Hapfelmeier S, Stecher B, et al. Innate and adaptive immunity cooperate flexibly to maintain host-microbiota mutualism. Science. 2009;325:617–620. doi: 10.1126/science.1172747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Zhang C, Zhang M, Wang S, et al. Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J. 2010;4:232–241. doi: 10.1038/ismej.2009.112. [DOI] [PubMed] [Google Scholar]
- 236.aCani PD, Delzenne NM. The role of the gut microbiota in energy metabolism and metabolic disease. Curr Pharm Des. 2009;15:1546–1558. doi: 10.2174/138161209788168164. [DOI] [PubMed] [Google Scholar]
- 237.ACS What are the risk factors for colorectal cancer? 2010 Available at: http://www.cancer.org/docroot/CRI/content/CRI_2_4_2X_What_are_the_risk_factors_for_colon_and_rectum_cancer.asp?mav=cri.
- 238.Bingham S, Riboli E. Diet and cancer—the European Prospective Investigation into Cancer and Nutrition. Nat Rev Cancer. 2004;4:206–215. doi: 10.1038/nrc1298. [DOI] [PubMed] [Google Scholar]
- 239.Yang L, Lu X, Nossa CW, et al. Inflammation and intestinal metaplasia of the distal esophagus are associated with alterations in the microbiome. Gastroenterology. 2009;137:588–597. doi: 10.1053/j.gastro.2009.04.046. [DOI] [PMC free article] [PubMed] [Google Scholar]