Abstract _ Full Text (HTML) _ Full Text (PDF) 

Review

Cancer Research Frontiers. 2017; 3(1): 1-28. doi: 10.17980/2017.1

Interplay between the gut microbiota and epithelial innate signaling in colitis-associated colon carcinogenesis

Linda Chia-Hui Yu1, Shu-Chen Wei2, and Yen-Hsuan Ni3*

1Graduate Institute of Physiology, National Taiwan University College of Medicine (e-mail: );

2Department of Internal Medicine, National Taiwan University Hospital (e-mail: ), and

3Department of Pediatrics, National Taiwan University Hospital, Taipei 100, Taiwan (e-mail: )

 

*Corresponding author: Yen-Hsuan Ni, Professor, Department of Pediatrics, National Taiwan University College of Medicine and Hospital, 7 Chung-Shan South Road, Taipei, Taiwan. E-mail: ; Tel: 886-2-23123456 ext. 71516; Fax: 886-2-23938871.

Citation: Linda Chia-Hui Yu, et al. Interplay between the gut microbiota and epithelial innate signaling in colitis-associated colon carcinogenesis. Cancer Research Frontiers. 2017; 3(1): 1-28. doi: 10.17980/2017.1

Copyright: @ 2017 Linda Chia-Hui Yu, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Competing Interests: The authors declare no competing financial interests.

Received Dec 2, 2016; Revised Mar 9, 2017; Accepted Mar 13, 2017. Published Mar 25, 2017

 

 

Abstract

Intestinal microbiota is involved in the maintenance of gut homeostasis as well as the regulation of colitis-associated colorectal tumorigenesis. The aberrant host immune signaling and the presence of opportunistic commensals with potential pathogenic characteristics (pathobionts) have been suggested to be incorporated into the genetic paradigm of colon carcinogenesis. The reciprocal relationship between innate immune response and microbial composition in tumorigenesis is highlighted in this article. A two-hit theory is proposed here that dysregulated host epithelial signaling and dysbiotic microbiota are synergistic factors to drive malignant transformation. Innate immune receptors such as Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors are involved in colitis-associated carcinogenesis through the regulation of epithelial cell death and proliferation, as well as the shaping of microbial community. From the microbial side, Escherichia coli, Fusobacterium nucleatum, enterotoxigenic Bacteroides fragilis, are identified as pro-tumorigenic pathobionts in colitis-associated tumor models. Probiotics such as Lactobacillus, Bifidobacterium, and butyrate-producing bacteria displayed tumor-suppressing effects. The Gram-negative characteristics of the mucosa-associated pathobionts indicate the involvement of lipopolysaccharide–dependent epithelial CD14/TLR4 signaling in cancer development. Virulence factors of the pathobionts were also identified in causing epithelial genotoxicity and signaling. The mechanistic insights of the interplay between host innate immunity and bacterial composition, and the understanding of how the dysfunction of one impacts on the other, will shed light to the development of novel strategies for the clinical management of inflammatory bowel disease and colon cancers.

Keywords: commensal bacteria, tumor biology, epithelial cells, Toll-like receptors, NOD-like receptors, virulence factors

 

 

INTRODUCTION

The gastrointestinal tract is a unique internal organ with a densely populated microbial ecosystem, in contrast to other aseptic viscera, in the human body (1, 2). Adult human intestine is inhabited by approximately 1014 bacterial cells, with the highest amount in the colon (3, 4). Over 1000 bacterial species mainly belonging to four phyla were identified in a cohort study with each individual harboring at least 160 species (5). A large inter-individual diversity was found and about 30-40 species are shared among individuals (5-8). The number of bacterial genes are estimated to be 100-fold higher than those of human genes (3, 5, 9). Besides the commensal bacteria, virus and fungi also exist in the intestine and are collectively defined as the gut microbiome (6, 10).

In the post-human genome era, much attention is now focused on this complex gut ecosystem. Advances in DNA sequencing and bioinformatics have fostered progress in human microbiome research. The explosion of knowledge in environment-diet-microbe-host interactions has greatly re-shaped our view of human physiology (5, 6, 11). Currently, enteric dysbiosis (a term that describes the condition of having microbial compositional, spatial, or number change within the body) is regarded not only as a key component of gastrointestinal diseases but also of extraintestinal and systemic disorders.

Dysbiosis has been reported in inflammatory bowel disease (IBD), colorectal cancer (CRC), atherosclerosis, obesity, type II diabetes, non-alcoholic liver diseases, multiple sclerosis, and chronic fatigue syndrome (12-15). A reduction of fecal bacterial diversity is found in patients with Crohn’s disease and ulcerative colitis (16-18) and colonic carcinoma (19, 20), which indicates that even fewer species could be making up the majority of a disease-associated microbial population. Recently, bacteria with colitogenic and pro-tumorigenic characteristics are suggested to play critical roles in the pathogenesis of colitis-associated CRC (21-23).

Patients with Crohn’s disease and ulcerative colitis are at higher risk of CRC (24, 25). A link between inflammation and cancer were also observed in gastritis-associated gastric carcinoma, hepatitis-associated hepatocellular carcinoma, and cholangitis-associated bile duct cancer (26, 27). Besides genetic instability mediated by inflammatory free radicals (26, 28, 29), the presence of disease-associated bacteria with virulence factors (21-23) and the aberrant innate immune responses to gut microbial products (30-32) are also involved in the multifaceted pathogenesis of CRC.

Taken into account the juxtaposition of bacteria and mucosa, microbial dysbiosis and dysregulated innate signals derived from intestinal epithelium are the focus of this review (33-36). A two-hit theory was proposed that aberrant host epithelial signaling and dysbiotic microbiota are co-existing factors that synergistically drive colitis-associated carcinogenesis (Figure 1). In this article, we aimed to highlight bidirectional evidence of epithelial innate signaling affecting the microbes and vice versa, and to discuss how aberrant interaction between bacteria and epithelium may contribute to tumor development and progression.

 

 

1675 FIG1

Figure 1. Schema of a two-hit theory of synergistic host and microbial factors involved in colon carcinogenesis. Aberrant innate signaling by host intestinal epithelium and immune cells to bacterial products has been demonstrated to alter the susceptibility of tumorigenesis. Enteric dysbiosis including microbiota changes and virulence increase also play crucial roles in the development of colorectal tumors. Modulation of host immune factors by antagonists or bacterial factors by antibiotics and virulence deletion are potential therapeutic strategies to reduce tumor growth.

 

HOST-MICROBE CROSSTALK AND EPITHELIAL INNATE IMMUNITY

Innate immune signaling are actively involved in microbial recognition and colitis-associated CRC development. A long line of evidence for IBD-associated polymorphisms in Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) (37-40) implicate that aberrant innate responses to their own microbial products is involved in disease pathogenesis. For information on the adaptive aspect of immunopathogenesis of IBD, other reviews are recommended (40, 41). Gene polymorphisms in lipopolysaccharide (LPS) receptors CD14 and TLR4 are observed in patients with Crohn’s disease and ulcerative colitis, and the polymorphisms are correlated with a higher risk of CRC (42-45). Gene polymorphisms in TLR2 are also linked with susceptibility of IBD and higher risks of CRC (46-49). NOD2 was the first gene to be identified with Crohn’s susceptibility; variants of NOD2 was found in a subset of patients with fibrostenosing Crohn’s disease (50-54). Although NOD2 mutation was used as a predictor for aggressive diseases in Crohn’s patients (55, 56), no correlation was found with CRC development (39, 57).

 These innate receptors were originally identified in monocytic cells for induction of proinflammatory responses following binding to bacterial products. The TLRs are known to be expressed on the cell surface, whereas NLRs are mostly recognized in the cytosols of immune cells (58, 59). The LPS receptor CD14/TLR4 activates a myeloid differentiation factor (MyD88)-dependent proinflammatory signaling (e.g. mitogen-activated protein kinases (MAPKs) and nuclear factor-kappa B (NFκB)) for production of proinflammatory cytokines in monocytes (Figure 2A) (60, 61). TLR2 (a bacterial peptidoglycan and lipoteichoic acid receptor) was shown to complex with TLR1, TLR6, or CD14, and to induce MyD88-dependent signals (62, 63). Moreover, NOD2 after binding to a peptidoglycan component, i.e. muramyl dipeptide (MDP), activates inflammasome pathways and NFkB signaling in immune cells (58, 59).

 

 

1675 FIG2

Figure 2. Bacterial LPS receptor subunits (CD14 and TLR4) are involved in proinflammatory signaling and regulation of cell death and proliferation in intestinal epithelium. (A) Binding of LPS to the CD14/TLR4 complex on lipid raft triggers myeloid differentiation factor (MyD88)-dependent and -independent signaling for proinflammatory cytokine production in intestinal epithelial cells. Early studies show that TLR4 activates MyD88-dependent pathways, including inhibitor of κB kinase (IKK)/inhibitor of κB (IκB)/nuclear factor-kappa B (NFκB), phosphatidylinositide- 3 kinase (PI3K)/Akt, and mitogen-activated protein kinases (MAPK) such as JNK, ERK, and p38. The TLR4-mediated MyD88-indepent pathway includes interferon regulatory factor 3 (IRF3). Nuclear translocation of NFκB subunits (p65 and p50), AP-1, or IRF3 cause the transcription of proinflammatory cytokines. (B) Recent findings indicate that LPS/CD14 binding on lipid raft triggers a number of lipid messengers to induce epithelial cell apoptosis which is counteracted by upregulation of TLR4. The cascade of lipid signaling involves conversion of membranous phophatidylcholine (PC) to diacylglycerol (DAG) by PC-specific phospholipase (PC-PLC), activation of sphingomyelinase (SMase) for sphingolipid metabolism and ceramide production, and phosphorylation of protein kinase C ζ (PKCζ). In absence of TLR4, the activation of PKCζ leads to caspase-dependent cell apoptosis in intestinal epithelial cells. However, upregulation of TLR4 serves as a antagonizing signal to inhibit epithelial cell apoptosis following PKCζ-dependent recruitment of TLR4 onto raft domains, acts as hyperproliferative signals through IKK/NFκB and PI3K/Akt molecules, and is involved in tumorigenesis via macrophage-associated cyclooxygenase-2 (COX-2) and epidermal growth factor receptor (EGFR)-dependent pathways.

 

A weak expression of TLR2/4 (64-67) and NOD2 (68, 69) was found in the intestinal epithelium of healthy individuals, and was traditionally considered a means to tolerate gut commensals. Constitutive CD14 expression was noted in the cell surface of normal intestinal epithelium (34, 64). In IBD and CRC patients, increased expression of CD14 and TLR4 was reported in the mucosal tissues and epithelial layers (Table 1) (64-66). Overexpression of TLR2 and NOD2 was also found in the epithelial cells of inflamed colon in Crohn’s disease (68, 70, 71). In addition, membrane recruitment of wild type NOD2 in contrast to the cytosolic presence of mutant NOD2 (R702W and G908R) have been reported in human intestinal epithelial cell lines (72, 73). Accumulating evidence indicate that the aberrant innate receptor expression and signaling on intestinal epithelium are involved in tumor progression.

The cause-and-effect relationship between innate responses and carcinogenesis was first implicated by the observation of diminished tumor formation in APC(Min/+) mice when TLRs or MyD88 signaling was ablated (32, 74). Spontaneous intestinal tumor development is seen in the multiple intestinal neoplasia (Min) mice, which carry a heterozygous mutation in the adenomatous polyposis coli (Apc) gene (75). Further evidence was shown in experimental models that epithelia-specific or systemic knockout of TLR4 display reduced colon tumor numbers and sizes (33, 76, 77), whereas mice with genetic deficiency in NLRs, such as NOD1, NOD2, NLRC4, NLRP3, and NLRP6, demonstrated higher susceptibility to CRC (78-83); inconsistent data were observed for the role of TLR2 in colon tumorigenesis (84-86). So far, TLR4 is the best characterized innate receptor expressed on intestinal epithelium for promoting colon tumorigenesis (33, 76, 77).

The opposite effects of TLR4 and NLRs on regulation of CRC growth are striking cause both types of innate receptors are known to activate NFκB signals (58, 59). A number of features have been proposed to explain the discrepancy between the two receptors in colitis-associated tumor formation. One of the possible reasons is that dysregulated epithelial cell death and proliferation mediated by imbalances in epithelial CD14/TLR4 signaling (uncoupled to proinflammatory responses) serves as a key mechanism in LPS-induced CRC progression (33-35, 87). On the other hand, inflammasome-dependent autophagy pathways (80, 88) and shaping of the microbiome (79, 89) are involved in the mechanisms of NLR-dependent suppression of tumorigenesis. The role of TLR4 in CRC development are described in this section, whereas the role of NLRs are discussed in relation to dysbiotic microbiota in the next section.

 

Table 1. Expression of LPS receptors in primary human intestinal epithelial cells.

1675 table1

Note: CRC, colorectal carcinoma; IBD, inflammatory bowel disease; Flow, flow cytometry; IF, immunofluorescent staining; IHC, immunohistochemistry; qPCR, real time quantitative polymerase chain reaction; RT-PCR, reverse transcription polymerase chain reaction.

 

Previous studies using bone marrow chimera have demonstrated that epithelial TLR4 overexpression plays a more dominant role than the receptor expression on myeloid cells in driving colon tumor growth (33, 76, 87). Intestinal epithelial cell studies had shown that downstream signals of TLR4 such as NFκB and Akt were involved in the epithelial hyperproliferative responses in vitro (36, 90, 91). Moreover, upregulation of TLR4/MyD88 promotes colon carcinogenesis via cyclooxygase-2 (COX-2) and epidermal growth factor receptor (EGFR)-dependent pathways in mouse models of colitis-associated CRC (Figure 2B) (74, 76, 92, 93). Our recent studies showed that TLR4 played an antagonistic role against its co-receptor CD14 in regulation of epithelial cell survival. Normal colonocytes respond to bacterial LPS through the constitutively expressed CD14 (34, 35). The intestinal epithelial cells undergo apoptosis following LPS/CD14 activation via lipid messengers and protein kinase C ζ (PKCζ) signals in the absence of TLR4, whereas upregulation of TLR4 expression inhibited CD14-mediated epithelial cell death and promoted tumor development (Figure 2B) (34, 35). Use of eritoran, which is a LPS mimicking molecule that acts as a CD14 agonist and TLR4 antagonist, caused an increase of cell death and a decrease of cell proliferation in tumor cells and significantly reduced tumor burden in a mouse CRC models (34, 35). Overall, functional antagonism between CD14 and TLR4 was identified in the bacterial regulation of epithelial apoptosis and proliferation, and the imbalance between the receptor subunits on epithelial cells plays a critical role in promoting tumorigenesis (34, 35, 94).

In keeping with early studies showing that commensal bacterial products are actively involved in the regulation of epithelial turnover and restitution (30, 95-97), our studies support the concept that aberrant epithelial signaling tips the balance toward malignancy. These findings provided novel information on the bacteria-regulated malignant transformation through innate signaling; however, the question remains as to what roles enteric dysbiosis play.

 

ENTERIC DYSBIOSIS

Alterations in the intestinal microbiota (such as changes in bacterial population or distribution) were documented in patients with IBD (98, 99), and CRC (100-102). Reduced microbial diversity was found in mucosal biopsies of patients with Crohn’s disease (16, 17) and ulcerative colitis (18). Lower bacterial diversity was also reported in biopsy tissues and stool samples of carcinoma patients compared to controls (19, 20). However, colonic adenoma biopsies showed higher diversity and greater numbers of bacteria compared to healthy individuals (103, 104). It is noteworthy that live bacteria reside in gut mucosa of IBD and CRC patients, in contrast to the mostly lumen-dwelling commensals in normal subjects (105, 106).

A role of bacteria in intestinal carcinogenesis was first suggested in 1978 by Reddy et al (107), based upon the observation of a lower incidence of chemically induced duodenal and colonic tumors in germ-free rats than in conventional rats. Reduced severity and delayed onset of chemically or genetically induced colitis was later reported in germ-free mice (108-110). Nevertheless, a causative role of bacteria was challenged by the notion of the lack of immune maturation and/or tolerance (which is dependent on commensal colonization) in germ-free intestine (111-113). Additional studies showed that bacterial depletion by antibiotics significantly reduced tumor burden in the mutagen-induced wild type mice (114, 115), as well as in the tumor-susceptible NOD1(-/-), NOD2(-/-) and NLRP6(-/-) mice (78, 79, 82), providing direct evidence of bacterial involvement in tumorigenesis.

The existence of pathobionts (opportunistic pathogenic bacteria converted from commensals) with colitogenic and pro-tumorigenic abilities was not confirmed until clear evidence of ‘transmissible’ colitis and CRC was demonstrated through fecal microbial transplantation and co-housing experiments (79, 82, 114). Previous studies have shown that the severe colitis and high tumor susceptibility in NOD2(-/-) or NLRP6(-/-) mice are ‘communicable’ through fecal transplantation to the recipients of wild type mice (79, 82). Indeed, the fecal microbial composition was altered in NOD2(-/-) and NLRP6(-/-) mice compared to their wild type counterparts through imbalance in the inflammasome-mediated regulation in antimicrobial peptide (AMP) profiles (89, 116-118). The findings indicate the emergence of dysbiotic microbiota as a result of the host genetic deficiency in NLRs. In addition, changes in mucosal defensin levels were also documented after TLR4 signaling (119-121), implicating AMP-dependent modulation in microbial ecology by TLR4. The increase of tumor burden in NLR-deficient mice strongly supported that dysbiotic microbiota plays an active role in colitis and carcinogenesis. Clinical observation showed beneficial effects of antibiotic therapy in the induction of remission in IBD patients (122-124). Overall, these studies suggested that host NLRs were essential in the shaping of gut microbiota, and the lack of NOD1, NOD2, or NLRP6 might alter the microbial community to a disease-associated profile.

Although the presence of pathobionts (such as colitogenic bacteria and infectious carcinogens) was confirmed in rodent models by mouse to mouse fecal transplantation (79, 82), a recent study transferring stool samples from CRC and healthy patients to germ-free mice before mutagen exposure had shown surprising results (125). Mice receiving human CRC-associated bacteria developed fewer tumors than those given bacteria from tumor-free subjects (125). The authors concluded that the initial microbiome structure developed by the recipient mice following fecal transplantation, but not the cancer status of the human donors, was the main factor determining tumor incidence in the recipient mice. They also found that Gram-negative bacteria such as Bacteroides are positively correlated with increased tumor burden, whereas Gram-positive bacteria such as Clostridiales are negatively associated with tumor growth (125). In spite of the unexpected results, a crucial role of gut bacteria in the regulation of cancer formation is supported by this study. More importantly, the exact strains and composition of gut microbiota with beneficial or detrimental effects on CRC development remain to be elucidated.

 

PATHOBIONTS AND PROBIOTICS

The presence of pathobionts and/or a shortage of probiotics (beneficial bacteria to the host) both serve as key factors for disease development. Clinically, bactericidal antibiotic treatment is recommended for the management of ulcerative colitis if infectious complications are suspected (126). Moreover, antibiotics that increase the abundance of beneficial bacteria, the so-called ‘eubiotics’, are emerging as a new treatment option (127). The good and the evil of gut microbiota are equivocal aspects in deciphering the bacterial strains for regulation of colitis-associated CRC.

Particular bacterial strains were characterized in germ-free and antibiotic-depleted mice by monoassociation experiments. These studies offered pivotal evidence supporting the presence of bacteria with pro-tumorigenic or colitogenic ability. Nevertheless, it should be kept in mind that the majority of monoassociation studies have utilized cancer-prone or immune-compromised mice with genetic defects to identify the bacterial strains (79, 82, 128, 129). The findings therefore support the idea that disease progression in patient subsets with genetic predispositions is partly attributable to pathobionts. However, this remains uncertain for individuals in the general heterogeneous population who have chronic inflammation and sporadic cancers but lack particular genetic traits. The pathobionts (Table 2) and probiotics with either direct or causal links to colitis and CRC are summarized below.

 

Table 2. Potential pathobionts involved in colon carcinogenesis.

1675 table2

Note: pks, polyketide synthase gene complex; afa, afimbrial adhesin; lpf, long polar fimbriae; fim, fimbrial adhesin or type-1 pili.

 

Escherichia coli

Enrichment of mucosa-associated or internalized Enterobacteriaceae family or Escherichia coli was long observed in biopsy samples of IBD and CRC patients (18, 130-133). Increased Escherichia genus was also identified in fecal bacterial population in Crohn’s disease, ulcerative colitis, and CRC patients compared to healthy individuals (101, 134). The levels of E. coli colonization appear to correlate positively with the proliferation index of colorectal tumor cells (130).

Diffusely adherent E. coli found in IBD and CRC patients possess a number of virulence genes such as afimbrial adhesin (afa), long polar fimbriae (lpf), fimbrial adhesin or type-1 pili (fim) and polyketide synthase gene complex (pks) (131, 132, 135). A subpopulation of E. coli originally identified in the ileal mucosa of Crohn’s disease patients, termed adherent-invasive E. coli (AIEC), is well-characterized for its mucosal attachment and ability to survive intracellularly in epithelial cells and macrophages (136, 137). Although the adherent-invasive ability was observed in these types of E. coli, they were not categorized as pathogens according to the classical definition due to their lack of known genetic invasive or toxigenic determinants (138, 139).

Recent studies indicated that pks-positive E. coli encoding a genotoxin (colibactin) increased the susceptibility to colorectal cancer in mutagen-induced IL-10(-/-) mice (128, 140). DNA damage and cell cycle arrest were noted in epithelial cells and mouse crypts after exposure to these pks-positive E. coli, implicating a pro-tumorigenic mechanism (128, 140). Colibactin-producing E. coli also indirectly enhance tumor growth by inducing the emergence of senescent cells that secrete hepatocyte growth factors in models of mutagen-induced IL-10(-/-) and wild-type mice (141).

In addition, colitogenic and pro-tumorigenic characteristics of AIEC were observed in transgenic mice expressing the human-specific carcinoembryonic antigen-related cell adhesion molecules 6 (CEACAM6) receptors on epithelial cells (142, 143). The epithelial CEACAM6 allowed bacterial colonization via type 1 pili (fimbriae)(142, 143). The lipid A moiety of Gram-negative bacteria also plays a role in preventing epithelial CEACAM shedding and in facilitating mucosal colonization by bacteria (144, 145). AIEC owes its pathogenicity to its active invasion, which is associated with sustained macrophage-derived cyclooxygenase-2 production, which promotes mucosal inflammation and epithelial proliferation (135, 146). Other studies using in vivo and in vitro models have shown that AIEC colonization increased mucosal permeability and tight junction disruption, implicating a direct role of bacteria in triggering gut leakiness, which might be another factor leading to chronic inflammation (138, 147-149). Further investigation of AIEC-induced epithelial innate signaling in barrier regulation and cell proliferation is warranted. In sum, virulence factors in E. coli conferring mucosal adherence/invasion and genotoxicity properties are relevant to disease progression in colitis and CRC.

 

Fusobacterium nucleatum

Fusobacterium species are commonly found in the oral cavity but rarely in the intestinal tract of healthy individuals (150, 151). However, abundance of Fusobacterium spp. and F. nucleatum were reported not only in fecal samples but also in inflamed mucosa of Crohn’s disease patients and in tumor specimens of CRC patients (19, 102, 152-157). The presence of mucosa-associated Fusobacterium in biopsy specimens of IBD and CRC has sparked interest in the emergence of possible invasive strains (99, 154, 158). Fusobacterium recovered from inflamed tissues of IBD patients displayed higher invasive ability to human carcinoma Caco-2 cell lines, compared to strains isolated from healthy tissues or control patients (158, 159). Moreover, the amount of Fusobacterium DNA in tumor tissues was found to be positively associated with poor prognosis in cancer patients (160).

In animal studies, eight weeks of daily feeding of human isolates of F. nucleatum accelerated the onset of cancer formation, increased tumor multiplicity, and selectively recruited tumor-infiltrating myeloid cells in APC(Min/+) mice (161). This observation of increased tumor burden by F. nucleatum is not associated with the exacerbation of colitis in APC(Min/+) mice (161). Further experiments were conducted in colitogenic mouse strains such as IL-10(-/-) and T-bet(-/-)/Rag2(-/-) mice to elaborate on the dissociation between colitis and tumors, and it was demonstrated that inoculation of F. nucleatum did not aggravate intestinal inflammation nor induce tumors in these colitic mice (161). These elegant studies suggested that F. nucleatum, albeit with protumorigenic potential under conditions of oncogenic mutation, did not possess colitogenic characteristics or the ability to trigger cancer in a colitis background. Moreover, in vitro studies had shown that F. nucleatum increased cell hyperproliferation in adenocarcinoma cell lines with APC mutation (e.g. HT29, DLD1, and SW480) or with β-catenin mutation (e.g. HCT116), but not in noncancerous HEK293 cells (162). Taken together, the findings suggested that pre-existing oncogenic mutation precede the F. nucleatum-driven tumorigenesis.

Several virulence factors of F. nucleatum have been implicated in colon tumorigenesis. A recent study demonstrated that FadA adhesin via binding to E-cadherin induced nuclear translocation of β-catenin for oncogene transcription in human adenocarcinoma cell lines (162). Indirect evidence of a role of FadA in promoting tumor growth was demonstrated by xenograft studies (162). Moreover, FadA also binds to vascular endothelial cadherin and helps F. nucleatum to adhere and breach endothelial cells to enhance the penetration of E. coli in in vitro transwell assays (163). A report has identified a novel lectin-like outermembrane protein Fap2 expressed on F. nucleatum that binds to a polysaccharide, Gal-GalNAc, on mouse CRC, suggesting another potential pro-tumorigenic virulence factor for epithelial anchoring and signaling (164). In addition, it was shown that bacterial surface protein Fap2 and RadD facilitated the adherence of F. nucleatum to lymphocytes for contact-dependent immune cell apoptosis (165). However, the association of these F. nucleatum proteins with in vivo orthotopic CRC has not yet been documented.

 

Bacteroides fragilis

A subclass of the human commensal Bacteroides species, enterotoxigenic Bacteroides fragilis (ETBF), was associated with acute inflammatory diarrheal disease and CRC in patients (155, 166, 167). Presence of B. fragilis and ETBF was found in the stool and biopsy specimens of both normal and CRC patients, but the amount of bacteria and toxin was significantly higher in late-stage CRC samples (101, 102, 155, 167, 168). One report has shown inconsistent data of decreased abundance of Bacteroides genus in stool specimens of CRC patients compared to healthy volunteers (101).

 Experimental models of colitis were used for the assessment of proinflammatory and pro-tumorigenic ability of ETBF. Orogastric administration of ETBF following antibiotic disruption of normal flora to promote colonization had caused acute colitis that persisted up to one year in wild-type mice (169-171). Moreover, ETBF worsened the severity of colitis induced by dextran sodium sulfate (169). Recent reports demonstrated that colonization by ETBF but not its non-toxigenic counterpart induced colitis and promoted colon tumorigenesis in APC(Min/+) mice (129, 172). This observation is different from the solely pro-oncogenic role of F. nucleatum (161). Several mechanisms have been proposed of its virulence factor, B. fragilis enterotoxin (also known as fragilysin) which acts as a metalloprotease. The pathogenic mechanisms include a direct cytotoxic effect by causing oxidative DNA damage, induction of epithelial E-cadherin cleavage for increased mucosal permeability and cell proliferation, and activation of Stat3 with a Th17 immune response (170-172). These studies indicated that adaptive T cell responses, beyond innate signaling in epithelial and immune cells, may also contribute to infection-induced carcinogenesis by ETBF.

 

Helicobacter species

Helicobacter pylori is classified as a class I carcinogen by the World Health Organization for its role in gastric cancer. Two of the extensively studied virulence factors cytotoxin-associated gene A (CagA) and vaculating cytotoxin A (VacA) have been associated with precancerous gastric lesions, through activation of epithelial proinflammatory and hyperproliferative signaling, and disruption of epithelial barrier (for a complete review, please see other articles (173, 174)). Although a relationship between H. pylori and CRC has been proposed, the evidence falls short of a definitive causal link due to conflicting results (175-180).

Infection with H. hepaticus has been shown to promote intestinal inflammation and CRC development in immunocompromised, colitogenic, or tumor-prone mouse models, such as Rag2(-/-), Rag2(-/-)APC(Min/+), mutagen-induced IL-10(-/-) mice (181-184). Although chronic life-long infection of H. hepaticus in the liver and colonic crypts are seen in immunodeficient mice, the bacteria do not colonize well nor cause disease in immunocompetent wild type animals (185, 186). In addition, there is no evidence of H. hepaticus infection on colorectal tumor samples in patient studies. The relevance of helicobacter species as a pathogen or pathobiont in promoting human CRC needs further investigation.

 

Lactobacillus and Bifidobacterium

Probiotics as dietary supplements have been investigated for their anti-inflammatory and anti-tumorigenic effects in experimental models. Numerous studies have demonstrated that a single species or mixtures of probiotics (such as Lactobacillus and Bifidobacterium) prevent intestinal inflammation in chemical-induced colitis models or in IL-10(-/-) mice (187-191). Other reports had shown that administration of Lactobacillus and Bifidobacterium spp. suppressed tumor formation in mutagen-induced CRC and APC(Min/+) mice models (84, 192-195). The beneficial effects of probiotics in the prevention of colitis have been generally attributed to their immunoregulatory and barrier-fortifying actions (188, 189). However, the anti-cancer mechanisms of Lactobacillus and Bifidobacterium are suspected to be either related to their anti-inflammatory effects or to the modulation of epithelial turnover. In vitro data have shown direct inhibition of proliferation and induction of apoptosis, and strengthening of barrier integrity, in intestinal epithelial cell lines by multiple strains of Lactobacillus (e.g. L. acidophilus, L. fermentum, L. reuteri, L. caseri, L. rhamnosus, and L. gasseri) (196-201) and Bifidobacterium (e.g. B. lactis and B. bifidum (202, 203)). A complete review of the beneficial effects of probiotics against cancer can be found in the literature (204, 205).

Nevertheless, there are conflicting data regarding the abundance of Lactobacillus and Bifidobacterium in inflamed and non-inflamed mucosa in patients with IBD (206-209). In addition, there is no evidence to support the efficacy of probiotics in CD patients, while improvement in disease activity is observed only in subsets of UC patients (210). In cancer patients, one report has revealed lower counts of Bifidobacterium in mucosal samples (208), whereas another report found no difference in Lactobacillus and Bifidobacterium abundance compared to normal individuals (100).

 

Butyrate-producing bacteria

Short-chain fatty acids, including acetate, propionate, and butyrate, are important fermentative metabolites produced from dietary fibers by anaerobic commensals in the colon. Butyrate is utilized by normal colonocytes as the primary energy source through mitochondrial oxidation, where its consumption is greater than that of glucose or glutamine (211-215). Moreover, butyrate is well known for its inhibitory actions on histone deacetylase (HDAC) (216, 217). Butyrate treatment induces histone hyperacetylation and transcriptional activation of pro-apoptotic genes such as Fas and the cell-cycle regulator p21(Waf1/Cip1), thereby stimulating cell death and arresting the cell cycle (216, 217).

Mounting evidence indicates that butyrate-producing bacterial genera such as Faecalibacterium, Eubacterium, and Roseburia are significantly less abundant and the amount of butyrate is decreased in fecal samples of IBD and CRC patients (101, 152, 218-220). A reduction in other butyrate-producing bacteria, such as Lachnospiraceae and Ruminococcaceae at the family levels, were also found in biopsy and surgical specimens of IBD patients (18, 221).

A tumor-suppressing effect of butyrate-producing bacteria was observed in early nutritional studies in mouse models by repeated oral administration of Butyrivibrio fibrisolvens one week before and during chemical induction of CRC (222). B. fibrisolvens is a ruminant bacterium, which also resides in human intestine in low numbers. Other reports have shown that a high fiber diet, which causes a large amount of butyrate production, decreases the rate of aberrant crypt foci formation in rats (223). A recent study using a gnotobiotic mouse model polyassociated with four commensal bacteria demonstrated that supplementation with high dietary fiber and B. fibrisolvens significantly decreased tumor growth (224). These findings support a role for butyrate-producing bacteria in the prevention of tumorigenesis and provide novel insights into the differential usage of butyrate between normal and tumor cells. Butyrate, a preferential energy fuel for normal colonocytes (212, 214, 225), is less utilized in tumor cells which perform aerobic glycolysis (so called the “Warburg effect”) (224, 226). The colonization of the mouse intestine by butyrate-producing bacteria prior to the chemical induction of CRC caused intracellular accumulation of butyrate and lowered HDAC activity, leading to increased histone acetylation and the expression of specific tumor-suppressor genes in cancer cells (224). Further investigation into the therapeutic effect of butyrate-producing bacteria in tumor-bearing mice is needed to verify the potential of butyrate as a treatment for CRC.

 

CHARACTERISTICS OF GRAM STAINING AND AEROTOLERANCE OF BACTERIA

The aforementioned pathobionts including E. coli, F. nucleatum, and B. fragilis are Gram-negative rod-shaped cells. Among these bacteria, E. coli is a well-known facultative anaerobe, and numerous studies showed that E. coli is capable of adhering and invading into intestinal epithelial cells and macrophages in oxygenated conditions for triggering innate signaling (138, 227, 228). Although F. nucleatum and B. fragilis are reported obligate anaerobes, a few reports have shown that F. nucleatum may grow as monoculture and even support other strict anaerobic bacteria in co-cultures in aerated environments (229, 230). Invasive strains of Fusobacterium were found in gut mucosa biopsies of IBD and CRC patients, and were able to survive and activate signals in epithelial cell lines (99, 154, 158, 159). Moreover, clinical strains of B. fragilis isolated from intestinal, blood and peritoneal specimens can be grown in oxygenated conditions (231, 232), and are capable of activating innate signaling (233, 234).

In contrast, the probiotic families such as Lactobacillaceae, Bifidobacteriaceae, Clostridiaceae (e.g. Faecalibacterium genus), Lachnospiraceae (e.g. Lachnospira, Roseburia and Butyrivibrio genus), and Ruminococcaceae are all Gram-positive rod-shaped cells. These probiotic bacterial strains are known as obligate anaerobes, except Lactobacillus being a facultative anaerobe.

A dichotomy seems to exist that Gram-negative bacteria plays a detrimental role in tumorigenesis whereas Gram-positive bacteria appears to be beneficial. It would be plausible to suspect that in addition to specific virulence factors, the outer lipid membranous product LPS of Gram-negative bacteria might be partly involved in its pro-tumorigenic properties through activation of epithelial and monocytic TLR4 signaling. On the other hand, MDP (a constituent of the peptidoglycan wall) which is in large quantities in Gram-positive bacteria and a lesser content in Gram-negative bacteria may be associated with NOD2 signaling for tumor-suppressive effects by probiotics. Together with the evidence of microbiome shaping by innate immune receptors (89, 116-118), a proposed model of the reciprocal relationship between innate immune responses and bacterial composition in carcinogenesis is depicted in Figure 3.

Other bacterial characteristics such as the ability to survive in close proximity to the oxygenated mucosa should also be considered as an advantage to increase its chance to cause pathology. However, solid tumor core is known to be relatively hypoxic (226, 235) and anaerobic bacteria may survive in oxygenated milieu with surrounding oxygen-consuming bacteria or in a biofilm (229, 236). Therefore, there is insufficient evidence to claim the necessity of oxygen-tolerance or -intolerence to be a basic requirement for bacteria to act as opportunistic pathogens.

 

 

1675 FIG3

Figure 3. A proposed model of the reciprocal relationship between innate immunity and microbiota in the pathogenesis of colon cancer. During the transition from physiological intestinal epithelium to pathological colorectal carcinoma, two co-existing factors (i.e. innate immune response and microbial composition) synergistically determine the fate of malignant transformation on top of host genetic predisposition. Under a healthy diet and lifestyle, nucleotide-binding oligomerization domain 2 (NOD2) shapes a healthy microbiome via inflammasome-mediated regulation of antimicrobial peptides (AMP). The healthy microbiota contains Gram-positive (G(+)) bacteria as probiotics that produces butyrate and large quantities of muramyl dipeptide (MDP). MDP binding to NOD2 induces inflammasome-dependent autophagy pathways which are involved in the maintenance of epithelial homeostasis. However, with an unhealthy diet and after exposure to colitogenic and carcinogenic agents, overexpression of Toll-like receptor 4 (TLR4) instigates proinflammatory, hyperproliferative and anti-apoptotic signals in colonic epithelial cells after lipopolysaccharide (LPS) binding, and also alters the mucosal defensin level and causes dysbiosis. The dysbiotic microbiota contains Gram-negative ((G(-)) pathobionts with virulence factors and outer lipid membranous product LPS. Binding to LPS further increases TLR4 expression on cells, leading to a viscous cycle of activation. The list of virulence factors (*) with genotoxic, adherence, and invasive properties in the pathobionts is shown in Table 2. The bidirectional aggravation between pathobiont LPS and epithelial TLR4 further contributes to colon tumor development.

 

UNANSWERED QUESTIONS, EXISTING CHALLENGES, AND FUTURE DIRECTIONS

The pathobionts were generally identified by their dominance on inflamed mucosa and cancerous tissues, and their pro-tumorigenic roles supported by evidence of increased tumor burden after inoculating large numbers of bacteria in chemically induced or genetically prone animal models of CRC. On the other hand, previously identified or widely ingested dietary probiotics are tested for their beneficial role in preventing cancer in animal models, but with limited evidence in patient fecal studies. While the search of individual bacterial species with essential roles in intestinal carcinogenesis could be a start for teasing out this complex host-microbe interplay, several fundamental questions remain unanswered.

First, the immunocompromised status or genetic mutation (either engineered or chemically induced) of the host seems to be a prerequisite for the suspected pathobiont to aggravate tumor development. The transplantation of dysbiotic microbiota or the inoculation of pathobionts only exacerbated diseases under pre-existing stimuli (e.g. colitogenic and carcinogenic agents, or genetic abnormality), but did not initiate lesions in untreated wild type conditions. It is noted that E. coli and B. fragilis are commonly seen in normal gut microbiota, while F. nucleatum mainly resides in the oral cavity of healthy individuals. Therefore, is host immune defect or early malignancy driving the emergence of disease-associated bacteria that further fuels the tumor growth? This hypothesis is in agreement with our proposed two-hit theory of host and bug, and further suggests that host abnormality may come first but may not act alone in tumorigenesis (as in germ-free condition). Although some may argue that increase of tumor susceptibility in wild type mice following fecal transplantation of the dysbiotic bacteria from NOD2(-/-) and NLRP6(-/-) mice (79, 82) is sufficient evidence for the existence of pathogenic bacteria uncoupled to host genetics, it should be kept in mind that the pro-tumorigenic stool bacteria were harvested from genetic deficient mice. Whether the pathobionts colonized preferentially in fecal contents and malignant niches are clonally developed or are orally acquired are still unknown. Considering that the presence of virus and bacteriophage is common in the gut and multiplying bacteria react to environmental cues rapidly, clonal lineages of gut commensals are perhaps more likely to make an opportunistic pathobiont in the stressed intestine.

Second, the majority of studies use fecal material for microbiome analysis whereas tissue biopsy data are mostly for identifying particular mucosa-associated bacteria. Inconsistent data regarding the abundance of Bacteroidetes phylum in fecal and mucosal samples of CRC patients were reported (103, 152), leading to the question of whether changes in stool or mucosal microbiota represent the disease-associated pattern. So far, a positive correlation between the abundance of fecal and mucosa-associated bacteria in terms of CRC risk are seen with the aforementioned pathobionts, i.e. Escherichia (101, 130-133) and Fusobacterium (19, 152-156), and ETBF (155, 167, 168). Whether fecal bacterial population simply reflects the counterbalance of space and nutrient demand between the mucosa-docking and free-floating bacteria remains to be determined. Since E. coli (138, 227, 228) and F. nucleatum (99, 154, 158, 159) adhere to or intracellularly survive in epithelial and tumor tissues, and aerotolerant B. fragilis are isolated from clinical peritoneal samples suggesting bacterial translocation (231, 232), the mucosal anchoring of bacteria could potentially increase its percentage in the fecal population. We believe that the mucosa-associated bacteria populated in vast numbers adjacent to epithelium and immune cells would be more relevant to disease progression, by which the virulence factors facilitating bacterial adherence and survival might be recognized by pattern recognition receptors for consequences of inflammation or tumorigenicity. Another line of evidence is that the amount of disease-associated bacteria in stool samples of CRC patients (e.g. Escherichia spp., Fusobacterium spp., and B. fragilis), although increased compared to healthy individuals, are still a minor component (<3%) of fecal microbiota in disease states (101, 102, 152). In contrast, when tumor tissues were used for microbiome analysis, the abundance of Fusobacterium genus jumps up to ~10% (154, 157). Therefore, the stratification of bacteria by radial locations rather than by numbers in fecal contents may be more important in terms of host interaction. Furthermore, mucosal bacterial taxa derived from pyrosequencing should be cross-validated by the culturing of viable internalized bacteria to rule out passive uptake of dead bacterial residues which might confound the microfloral data.

Third, whether the wax and wane of particular bacterial species are influencing the viability of other microbes or even the whole microbiota population to impact on tumorigenesis is unclear. A bacterial driver-passenger model was proposed by Tjalsma et al indicating that particular species may play an active role (by initiating or aggravating lesions) or a passive role (as a bystander) in tumorigenesis, and the concept was suggested to be incorporated into the genetic paradigm of cancer progression (237). To answer this question, in vitro testing of a single bacterial strain to modulate proliferative and death response in epithelial cells would support a driver role of the microbe on epithelial-derived cancers. However, the possibility of this particular strain of bacteria acting on other members of the microbiota (as an assistant in altering tumorigenesis) is not mutually exclusive from its direct role and cannot be ruled out in in vivo settings. It is well known that maximized mutual fitness and bacteriocin-mediated competition co-exists among related species of Escherichia (238, 239). Uni- or bi-directional enhancement of bacterial growth with Fusobacterium, Porphyromonas, and Bacteroides species has been reported in in vivo subcutaneous abscess models and in vitro microbial co-cultures (229, 240). Moreover, probiotic mixture and eubiotic/antibiotic studies have shown that interaction among bacteria plays a key role in shifting the microbial community for suppression of cancer growth (115, 241, 242). Furthermore, the use of a combination of bacterial operational taxonomy units as a screening tool was shown to improve the probability of identifying adenoma and the prognosis of aggressive malignant transformation (156, 243, 244). These observations suggest that a consortium of bacterial complex instead of one particular species is in control of tumor progression. Employing large numbers of the suspected bacteria for a long-term repeated inoculation might overrule the necessity of supportive microbes for nutrient sharing and species competition, or mask the need of prebiotics and dietary metabolism that are otherwise important in microbiota shaping in normal conditions. The dynamics between particular bacteria and related species in the microbial community could be investigated through biofilm or mixed infection studies to provide a more holistic view of this complex interaction. We believe that although the presence of some bacterial taxa may not seem to be crucial in tumor-prone or tumor-inducing experimental settings, they may play regulatory roles in shaping a “healthy” intestinal microbiota and in maintaining epithelial and immune homeostasis to suppress the transition to malignancy. To date, no common species can be conclusively ruled out as having roles in intestinal cancer.

Fourth, the time-dependent change of genetic signatures of a particular bacteria clone throughout the course of tumorigenesis, or the temporal profile of genetic diversity of intestinal microbiota with the development of pathological conditions may clarify the ‘snapshot’ observation in cancer which is widely used in current studies and may help differentiate the driver or passenger role of bacteria. Moreover, employing antibiotic mixtures for bacterial depletion at various times to modulate tumor growth would be an efficient reductionist strategy. The finding of a critical period for bacteria-regulated tumorigenesis may justify the application of large scale metagenomics to decipher key molecules in host-microbial interaction and to speed up the search of potential therapeutic targets.

Fifth, the effect of diet either directly or indirectly on bacterial composition adds another element of complexity in the quest of determining disease-associated bacteria. Dietary substances may indirectly modulate the bacterial community through endocrine and immune regulation. Other than fibers being the fermentative source of bacteria-derived butyrate with a clear tumor-suppressing role, high fat diet is known to increase the level of bacteria-derived secondary bile acid (e.g. deoxycholic acid) which shows a positive correlation to tumor growth (245, 246). Further information on diet in relation to dysbiosis and CRC risk could be found in recent articles (247-249), and will not be discussed in details here. While a direct link between diet and bacterial composition is irreputable, it should be kept in mind that dietary metabolites by affecting gut-brain-liver axis for glucose, lipid, and energy homeostasis (250, 251) have direct effects on the host systemically, with or without the involvement of secondary bacterial factors.

 

CONCLUDING REMARKS

Harnessing the aberrant signaling of the host epithelium and correcting the virulence of pathobionts as a two-hit intervention could be an effective strategy for the treatment of colitis-associated CRC. The appropriate timing, dosage, duration, and combination of therapeutic antibiotics or eubiotics to abort disease progression has yet to be determined. The impact of antibiotics on host innate signaling which might further modulate the course of colitis and tumorigenesis needs to be clarified. Binding of LPS and MDP has been previously shown to elicit positive or negative feedbacks for surface and vesicle expression of CD14, TLR4, or NOD2 (252-255). The phenomena of cross-tolerance or costimulation of TLRs and NLRs by agoinsts have also been documented (256-258). Hence, the effect of antibiotic treatment on mucosal levels of TLRs and NLRs will provide additional information on the microbial regulation of tumor growth. Bacterial engineering would be another approach to manipulate cancer progression either by directly killing pathobionts, by colonization of targeted bacteria to outgrow their parent strains, or by improvement of probiotics with higher synthesis of beneficial metabolites and more stable colonization (259-263). In summary, manipulation of the gut microbiota to alter the epithelial response or vice versa is considered new therapeutic strategies for cancer treatment beyond gene-related therapy. The understanding of host and microbial interplay would benefit the development of novel strategies for disease intervention in patients with IBD and CRC.

 

Acknowledgement

This paper has been supported by the National Health Research Institute, Taiwan (NHRI-EX105-10520BI, NHRI-EX106-10520BI, NHRI-EX107-10520BI) and Ministry of Science and Technology (MoST 105-2320-B-002-063).

 

Abbreviations:

afa, afimbrial adhesin;

AIEC, adherent-invasive Escherichia coli;

AMP, antimicrobial peptide;

APC, adenomatous polyposis coli;

CagA, cytotoxin-associated gene A;

CEACAM6, carcinoembryonic antigen-related cell adhesion molecules 6;

COX, cyclooxygenase-2;

CRC, colorectal cancer;

DAG, diacylglycerol;

EGFR, epidermal growth factor receptor;

ETBF, enterotoxigenic Bacteroides fragilis;

fim, fimbrial adhesin;

HDAC, histone deacetylase;

IBD, inflammatory bowel disease;

IKK, IκB kinase;

IRF3, interferon regulatory factor 3;

IκB, Inhibitor of κB;

lpf, long polar fimbriae;

LPS, lipopolysaccharide (LPS);

MAPK, mitogen-activated protein kinase;

MDP, muramyl dipeptide;

Min, multiple intestinal neoplasia;

MyD88, myeloid differentiation factor;

NFκB, nuclear factor-kappa B;

NLR, nucleotide-binding oligomerization domain-like receptor;

NOD, nucleotide-binding oligomerization domain;

PC, phophatidylcholine;

PC-PLC, PC-specific phospholipase;

PI3K, phosphatidylinositide-3 kinase;

PKCζ, protein kinase C ζ;

pks, polyketide synthase gene complex;

SMase, sphingomyelinase.

TLR, Toll-like receptors;

VacA, vaculating cytotoxin A;

 

 

REFERENCES

  1. Yu LC, Wang JT, Wei SC, Ni YH. Host-microbial interactions and regulation of intestinal epithelial barrier function: From physiology to pathology. World J Gastrointest Pathophysiol. 2012 Feb 15;3(1):27-43. DOI: 10.4291/wjgp.v3.i1.27.
  2. Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006 Feb 24;124(4):837-48. DOI: 10.1016/j.cell.2006.02.017.
  3. Sender R, Fuchs S, Milo R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016 Aug;14(8):e1002533. DOI: 10.1371/journal.pbio.1002533.
  4. Sender R, Fuchs S, Milo R. Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell. 2016 Jan 28;164(3):337-40. DOI: 10.1016/j.cell.2016.01.013.
  5. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010 Mar 04;464(7285):59-65. DOI: 10.1038/nature08821.
  6. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007 Oct 18;449(7164):804-10. DOI: 10.1038/nature06244.
  7. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006 Dec 21;444(7122):1022-3. DOI: 10.1038/4441022a.
  8. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, et al. Diversity of the human intestinal microbial flora. Science. 2005 Jun 10;308(5728):1635-8. DOI: 10.1126/science.1110591.
  9. Southan C. Has the yo-yo stopped? An assessment of human protein-coding gene number. Proteomics. 2004;4(6):1712-26.
  10. Yu YB, Zuo XL, Zhao QJ, Chen FX, Yang J, Dong YY, et al. Brain-derived neurotrophic factor contributes to abdominal pain in irritable bowel syndrome. Gut. 2012 May;61(5):685-94. DOI: 10.1136/gutjnl-2011-300265.
  11. Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science. 2016 Apr 29;352(6285):565-9. DOI: 10.1126/science.aad3369.
  12. Smits LP, Bouter KE, de Vos WM, Borody TJ, Nieuwdorp M. Therapeutic potential of fecal microbiota transplantation. Gastroenterology. 2013 Nov;145(5):946-53. DOI: 10.1053/j.gastro.2013.08.058.
  13. Zhang X, Zheng X, Yuan Y. Treatment of insulin resistance: straight from the gut. Drug Discov Today. 2016 Aug;21(8):1284-90. DOI: 10.1016/j.drudis.2016.06.016.
  14. Saad MJ, Santos A, Prada PO. Linking Gut Microbiota and Inflammation to Obesity and Insulin Resistance. Physiology (Bethesda). 2016 Jul;31(4):283-93. DOI: 10.1152/physiol.00041.2015.
  15. Guinane CM, Cotter PD. Role of the gut microbiota in health and chronic gastrointestinal disease: understanding a hidden metabolic organ. Therap Adv Gastroenterol. 2013 Jul;6(4):295-308. DOI: 10.1177/1756283X13482996.
  16. Walker AW, Sanderson JD, Churcher C, Parkes GC, Hudspith BN, Rayment N, et al. High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease. BMC Microbiol. 2011 Jan 10;11:7. DOI: 10.1186/1471-2180-11-7.
  17. Hansen R, Russell RK, Reiff C, Louis P, McIntosh F, Berry SH, et al. Microbiota of de-novo pediatric IBD: increased Faecalibacterium prausnitzii and reduced bacterial diversity in Crohn’s but not in ulcerative colitis. Am J Gastroenterol. 2012 Dec;107(12):1913-22. DOI: 10.1038/ajg.2012.335.
  18. Lepage P, Hasler R, Spehlmann ME, Rehman A, Zvirbliene A, Begun A, et al. Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology. 2011 Jul;141(1):227-36. DOI: 10.1053/j.gastro.2011.04.011.
  19. Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, Shi J, et al. Human gut microbiome and risk for colorectal cancer. J Natl Cancer Inst. 2013 Dec 18;105(24):1907-11. DOI: 10.1093/jnci/djt300.
  20. Huipeng W, Lifeng G, Chuang G, Jiaying Z, Yuankun C. The differences in colonic mucosal microbiota between normal individual and colon cancer patients by polymerase chain reaction-denaturing gradient gel electrophoresis. J Clin Gastroenterol. 2014 Feb;48(2):138-44. DOI: 10.1097/MCG.0b013e3182a26719.
  21. Owyang C, Wu GD. The gut microbiome in health and disease. Gastroenterology. 2014 May;146(6):1433-6. DOI: 10.1053/j.gastro.2014.03.032.
  22. Wlodarska M, Kostic AD, Xavier RJ. An integrative view of microbiome-host interactions in inflammatory bowel diseases. Cell Host Microbe. 2015 May 13;17(5):577-91. DOI: 10.1016/j.chom.2015.04.008.
  23. Sears CL, Garrett WS. Microbes, microbiota, and colon cancer. Cell Host Microbe. 2014 Mar 12;15(3):317-28. DOI: 10.1016/j.chom.2014.02.007.
  24. Munkholm P. Review article: the incidence and prevalence of colorectal cancer in inflammatory bowel disease. Aliment Pharmacol Ther. 2003 Sep;18 Suppl 2:1-5. DOI: 1697 [pii].
  25. Brackmann S, Andersen SN, Aamodt G, Langmark F, Clausen OP, Aadland E, et al. Relationship between clinical parameters and the colitis-colorectal cancer interval in a cohort of patients with colorectal cancer in inflammatory bowel disease. Scand J Gastroenterol. 2009;44(1):46-55. DOI: 10.1080/00365520801977568.
  26. Shimizu T, Marusawa H, Endo Y, Chiba T. Inflammation-mediated genomic instability: roles of activation-induced cytidine deaminase in carcinogenesis. Cancer Sci. 2012 Jul;103(7):1201-6. DOI: 10.1111/j.1349-7006.2012.02293.x.
  27. Tozun N, Vardareli E. Gut Microbiome and Gastrointestinal Cancer: Les liaisons Dangereuses. J Clin Gastroenterol. 2016 Nov/Dec;50 Suppl 2, Proceedings from the 8th Probiotics, Prebiotics & New Foods for Microbiota and Human Health meeting held in Rome, Italy on September 13-15, 2015:S191-S6. DOI: 10.1097/MCG.0000000000000714.
  28. Farinati F, Piciocchi M, Lavezzo E, Bortolami M, Cardin R. Oxidative stress and inducible nitric oxide synthase induction in carcinogenesis. Dig Dis. 2010;28(4-5):579-84. DOI: 10.1159/000320052.
  29. Tahara T, Arisawa T. Potential usefulness of DNA methylation as a risk marker for digestive cancer associated with inflammation. Expert Rev Mol Diagn. 2012 Jun;12(5):489-97. DOI: 10.1586/erm.12.38.
  30. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118(2):229-41.
  31. Rakoff-Nahoum S, Hao L, Medzhitov R. Role of toll-like receptors in spontaneous commensal-dependent colitis. Immunity. 2006 Aug;25(2):319-29. DOI: 10.1016/j.immuni.2006.06.010.
  32. Rakoff-Nahoum S, Medzhitov R. Role of toll-like receptors in tissue repair and tumorigenesis. Biochemistry (Mosc). 2008 May;73(5):555-61.
  33. Fukata M, Shang L, Santaolalla R, Sotolongo J, Pastorini C, Espana C, et al. Constitutive activation of epithelial TLR4 augments inflammatory responses to mucosal injury and drives colitis-associated tumorigenesis. Inflamm Bowel Dis. 2011 Jul;17(7):1464-73. DOI: 10.1002/ibd.21527.
  34. Kuo WT, Lee TC, Yang HY, Chen CY, Au YC, Lu YZ, et al. LPS receptor subunits have antagonistic roles in epithelial apoptosis and colonic carcinogenesis. Cell Death Differ. 2015 Oct;22(10):1590-604. DOI: 10.1038/cdd.2014.240.
  35. Kuo WT, Lee TC, Yu LC. Eritoran Suppresses Colon Cancer by Altering a Functional Balance in Toll-like Receptors That Bind Lipopolysaccharide. Cancer Res. 2016 Aug 15;76(16):4684-95. DOI: 10.1158/0008-5472.CAN-16-0172.
  36. Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, et al. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004 Aug 06;118(3):285-96. DOI: 10.1016/j.cell.2004.07.013.
  37. Kosovac K, Brenmoehl J, Holler E, Falk W, Schoelmerich J, Hausmann M, et al. Association of the NOD2 genotype with bacterial translocation via altered cell-cell contacts in Crohn’s disease patients. Inflamm Bowel Dis. 2010 Aug;16(8):1311-21. DOI: 10.1002/ibd.21223.
  38. Baumgart DC, Buning C, Geerdts L, Schmidt HH, Genschel J, Fiedler T, et al. The c.1-260C>T promoter variant of CD14 but not the c.896A>G (p.D299G) variant of toll-like receptor 4 (TLR4) genes is associated with inflammatory bowel disease. Digestion. 2007;76(3-4):196-202.
  39. Freire P, Portela F, Donato MM, Figueiredo P, Ferreira M, Amaro P, et al. CARD15 mutations and colorectal cancer in a South European country. Int J Colorectal Dis. 2010 Oct;25(10):1211-9. DOI: 10.1007/s00384-010-1028-0.
  40. de Souza HS, Fiocchi C. Immunopathogenesis of IBD: current state of the art. Nat Rev Gastroenterol Hepatol. 2016 Jan;13(1):13-27. DOI: 10.1038/nrgastro.2015.186.
  41. Park JH, Peyrin-Biroulet L, Eisenhut M, Shin JI. IBD immunopathogenesis: A comprehensive review of inflammatory molecules. Autoimmun Rev. 2017 Feb 14. DOI: 10.1016/j.autrev.2017.02.013.
  42. Araki Y, Sugihara H, Hattori T. In vitro effects of dextran sulfate sodium on a Caco-2 cell line and plausible mechanisms for dextran sulfate sodium-induced colitis. Oncol Rep. 2006 Dec;16(6):1357-62.
  43. Chen R, Luo FK, Wang YL, Tang JL, Liu YS. LBP and CD14 polymorphisms correlate with increased colorectal carcinoma risk in Han Chinese. World J Gastroenterol. 2011 May 14;17(18):2326-31. DOI: 10.3748/wjg.v17.i18.2326.
  44. Eyking A, Ey B, Runzi M, Roig AI, Reis H, Schmid KW, et al. Toll-like receptor 4 variant D299G induces features of neoplastic progression in Caco-2 intestinal cells and is associated with advanced human colon cancer. Gastroenterology. 2011 Dec;141(6):2154-65. DOI: 10.1053/j.gastro.2011.08.043.
  45. Franchimont D, Vermeire S, El HH, Pierik M, Van SK, Gustot T, et al. Deficient host-bacteria interactions in inflammatory bowel disease? The toll-like receptor (TLR)-4 Asp299gly polymorphism is associated with Crohn’s disease and ulcerative colitis. Gut. 2004;53(7):987-92.
  46. Pimentel-Nunes P, Teixeira AL, Pereira C, Gomes M, Brandao C, Rodrigues C, et al. Functional polymorphisms of Toll-like receptors 2 and 4 alter the risk for colorectal carcinoma in Europeans. Dig Liver Dis. 2013 Jan;45(1):63-9. DOI: 10.1016/j.dld.2012.08.006.
  47. Slattery ML, Herrick JS, Bondurant KL, Wolff RK. Toll-like receptor genes and their association with colon and rectal cancer development and prognosis. Int J Cancer. 2012 Jun 15;130(12):2974-80. DOI: 10.1002/ijc.26314.
  48. Bank S, Skytt Andersen P, Burisch J, Pedersen N, Roug S, Galsgaard J, et al. Polymorphisms in the inflammatory pathway genes TLR2, TLR4, TLR9, LY96, NFKBIA, NFKB1, TNFA, TNFRSF1A, IL6R, IL10, IL23R, PTPN22, and PPARG are associated with susceptibility of inflammatory bowel disease in a Danish cohort. PLoS One. 2014;9(6):e98815. DOI: 10.1371/journal.pone.0098815.
  49. Cheng Y, Zhu Y, Huang X, Zhang W, Han Z, Liu S. Association between TLR2 and TLR4 Gene Polymorphisms and the Susceptibility to Inflammatory Bowel Disease: A Meta-Analysis. PLoS One. 2015;10(5):e0126803. DOI: 10.1371/journal.pone.0126803.
  50. Hampe J, Cuthbert A, Croucher PJ, Mirza MM, Mascheretti S, Fisher S, et al. Association between insertion mutation in NOD2 gene and Crohn’s disease in German and British populations. Lancet. 2001 Jun 16;357(9272):1925-8. DOI: 10.1016/S0140-6736(00)05063-7.
  51. Hugot JP, Chamaillard M, Zouali H, Lesage S, Cezard JP, Belaiche J, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature. 2001 May 31;411(6837):599-603. DOI: 10.1038/35079107.
  52. Ogura Y, Saab L, Chen FF, Benito A, Inohara N, Nunez G. Genetic variation and activity of mouse Nod2, a susceptibility gene for Crohn’s disease. Genomics. 2003 Apr;81(4):369-77.
  53. Inoue N, Tamura K, Kinouchi Y, Fukuda Y, Takahashi S, Ogura Y, et al. Lack of common NOD2 variants in Japanese patients with Crohn’s disease. Gastroenterology. 2002 Jul;123(1):86-91.
  54. Leong RW, Armuzzi A, Ahmad T, Wong ML, Tse P, Jewell DP, et al. NOD2/CARD15 gene polymorphisms and Crohn’s disease in the Chinese population. Aliment Pharmacol Ther. 2003 Jun 15;17(12):1465-70.
  55. Yarur AJ, Strobel SG, Deshpande AR, Abreu MT. Predictors of aggressive inflammatory bowel disease. Gastroenterol Hepatol (N Y). 2011 Oct;7(10):652-9.
  56. Liverani E, Scaioli E, Digby RJ, Bellanova M, Belluzzi A. How to predict clinical relapse in inflammatory bowel disease patients. World J Gastroenterol. 2016 Jan 21;22(3):1017-33. DOI: 10.3748/wjg.v22.i3.1017.
  57. Lakatos PL, Hitre E, Szalay F, Zinober K, Fuszek P, Lakatos L, et al. Common NOD2/CARD15 variants are not associated with susceptibility or the clinicopathologic characteristics of sporadic colorectal cancer in Hungarian patients. BMC Cancer. 2007 Mar 27;7:54. DOI: 10.1186/1471-2407-7-54.
  58. Claes AK, Zhou JY, Philpott DJ. NOD-Like Receptors: Guardians of Intestinal Mucosal Barriers. Physiology (Bethesda). 2015 May;30(3):241-50. DOI: 10.1152/physiol.00025.2014.
  59. Saxena M, Yeretssian G. NOD-Like Receptors: Master Regulators of Inflammation and Cancer. Front Immunol. 2014;5:327. DOI: 10.3389/fimmu.2014.00327.
  60. da Silva CJ, Soldau K, Christen U, Tobias PS, Ulevitch RJ. Lipopolysaccharide is in close proximity to each of the proteins in its membrane receptor complex. transfer from CD14 to TLR4 and MD-2. JBiolChem. 2001;276(24):21129-35.
  61. Dunzendorfer S, Lee HK, Soldau K, Tobias PS. TLR4 is the signaling but not the lipopolysaccharide uptake receptor. JImmunol. 2004;173(2):1166-70.
  62. Buchholz BM, Bauer AJ. Membrane TLR signaling mechanisms in the gastrointestinal tract during sepsis. Neurogastroenterol Motil. 2010 Mar;22(3):232-45. DOI: 10.1111/j.1365-2982.2009.01464.x.
  63. Schroder NW, Morath S, Alexander C, Hamann L, Hartung T, Zahringer U, et al. Lipoteichoic acid (LTA) of Streptococcus pneumoniae and Staphylococcus aureus activates immune cells via Toll-like receptor (TLR)-2, lipopolysaccharide-binding protein (LBP), and CD14, whereas TLR-4 and MD-2 are not involved. J Biol Chem. 2003 May 02;278(18):15587-94. DOI: 10.1074/jbc.M212829200.
  64. Martin-Villa JM, Ferre-Lopez S, Lopez-Suarez JC, Corell A, Perez-Blas M, Arnaiz-Villena A. Cell surface phenotype and ultramicroscopic analysis of purified human enterocytes: a possible antigen-presenting cell in the intestine. Tissue Antigens. 1997 Dec;50(6):586-92.
  65. Belmonte L, Beutheu Youmba S, Bertiaux-Vandaele N, Antonietti M, Lecleire S, Zalar A, et al. Role of toll like receptors in irritable bowel syndrome: differential mucosal immune activation according to the disease subtype. PLoS One. 2012;7(8):e42777. DOI: 10.1371/journal.pone.0042777.
  66. Wang EL, Qian ZR, Nakasono M, Tanahashi T, Yoshimoto K, Bando Y, et al. High expression of Toll-like receptor 4/myeloid differentiation factor 88 signals correlates with poor prognosis in colorectal cancer. Br J Cancer. 2010 Mar 02;102(5):908-15. DOI: 10.1038/sj.bjc.6605558.
  67. Wolfs TG, Derikx JP, Hodin CM, Vanderlocht J, Driessen A, de Bruine AP, et al. Localization of the lipopolysaccharide recognition complex in the human healthy and inflamed premature and adult gut. Inflamm Bowel Dis. 2010 Jan;16(1):68-75. DOI: 10.1002/ibd.20995.
  68. Berrebi D, Maudinas R, Hugot JP, Chamaillard M, Chareyre F, De Lagausie P, et al. Card15 gene overexpression in mononuclear and epithelial cells of the inflamed Crohn’s disease colon. Gut. 2003 Jun;52(6):840-6.
  69. Hisamatsu T, Suzuki M, Reinecker HC, Nadeau WJ, McCormick BA, Podolsky DK. CARD15/NOD2 functions as an antibacterial factor in human intestinal epithelial cells. Gastroenterology. 2003;124(4):993-1000.
  70. Szebeni B, Veres G, Dezsofi A, Rusai K, Vannay A, Mraz M, et al. Increased expression of Toll-like receptor (TLR) 2 and TLR4 in the colonic mucosa of children with inflammatory bowel disease. Clin Exp Immunol. 2008 Jan;151(1):34-41. DOI: 10.1111/j.1365-2249.2007.03531.x.
  71. Begue B, Dumant C, Bambou JC, Beaulieu JF, Chamaillard M, Hugot JP, et al. Microbial induction of CARD15 expression in intestinal epithelial cells via toll-like receptor 5 triggers an antibacterial response loop. J Cell Physiol. 2006 Nov;209(2):241-52. DOI: 10.1002/jcp.20739.
  72. Barnich N, Aguirre JE, Reinecker HC, Xavier R, Podolsky DK. Membrane recruitment of NOD2 in intestinal epithelial cells is essential for nuclear factor-{kappa}B activation in muramyl dipeptide recognition. J Cell Biol. 2005 Jul 04;170(1):21-6. DOI: 10.1083/jcb.200502153.
  73. McDonald C, Chen FF, Ollendorff V, Ogura Y, Marchetto S, Lecine P, et al. A role for Erbin in the regulation of Nod2-dependent NF-kappaB signaling. J Biol Chem. 2005 Dec 02;280(48):40301-9. DOI: 10.1074/jbc.M508538200.
  74. Rakoff-Nahoum S, Medzhitov R. Regulation of spontaneous intestinal tumorigenesis through the adaptor protein MyD88. Science. 2007 Jul 06;317(5834):124-7. DOI: 10.1126/science.1140488.
  75. Oshima M, Oshima H, Kitagawa K, Kobayashi M, Itakura C, Taketo M. Loss of Apc heterozygosity and abnormal tissue building in nascent intestinal polyps in mice carrying a truncated Apc gene. Proc Natl Acad Sci U S A. 1995 May 09;92(10):4482-6.
  76. Fukata M, Chen A, Vamadevan AS, Cohen J, Breglio K, Krishnareddy S, et al. Toll-like receptor-4 promotes the development of colitis-associated colorectal tumors. Gastroenterology. 2007 Dec;133(6):1869-81. DOI: 10.1053/j.gastro.2007.09.008.
  77. Hernandez Y, Sotolongo J, Breglio K, Conduah D, Chen A, Xu R, et al. The role of prostaglandin E2 (PGE 2) in toll-like receptor 4 (TLR4)-mediated colitis-associated neoplasia. BMC Gastroenterol. 2010 Jul 16;10:82. DOI: 10.1186/1471-230X-10-82.
  78. Chen GY, Shaw MH, Redondo G, Nunez G. The innate immune receptor Nod1 protects the intestine from inflammation-induced tumorigenesis. Cancer Res. 2008 Dec 15;68(24):10060-7. DOI: 10.1158/0008-5472.CAN-08-2061.
  79. Couturier-Maillard A, Secher T, Rehman A, Normand S, De Arcangelis A, Haesler R, et al. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J Clin Invest. 2013 Feb;123(2):700-11. DOI: 10.1172/JCI62236.
  80. Hu B, Elinav E, Huber S, Booth CJ, Strowig T, Jin C, et al. Inflammation-induced tumorigenesis in the colon is regulated by caspase-1 and NLRC4. Proc Natl Acad Sci U S A. 2010 Dec 14;107(50):21635-40. DOI: 10.1073/pnas.1016814108.
  81. Allen IC, TeKippe EM, Woodford RM, Uronis JM, Holl EK, Rogers AB, et al. The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med. 2010 May 10;207(5):1045-56. DOI: 10.1084/jem.20100050.
  82. Hu B, Elinav E, Huber S, Strowig T, Hao L, Hafemann A, et al. Microbiota-induced activation of epithelial IL-6 signaling links inflammasome-driven inflammation with transmissible cancer. Proc Natl Acad Sci U S A. 2013 Jun 11;110(24):9862-7. DOI: 10.1073/pnas.1307575110.
  83. Normand S, Delanoye-Crespin A, Bressenot A, Huot L, Grandjean T, Peyrin-Biroulet L, et al. Nod-like receptor pyrin domain-containing protein 6 (NLRP6) controls epithelial self-renewal and colorectal carcinogenesis upon injury. Proc Natl Acad Sci U S A. 2011 Jun 07;108(23):9601-6. DOI: 10.1073/pnas.1100981108.
  84. Kuugbee ED, Shang X, Gamallat Y, Bamba D, Awadasseid A, Suliman MA, et al. Structural Change in Microbiota by a Probiotic Cocktail Enhances the Gut Barrier and Reduces Cancer via TLR2 Signaling in a Rat Model of Colon Cancer. Dig Dis Sci. 2016 Oct;61(10):2908-20. DOI: 10.1007/s10620-016-4238-7.
  85. Lowe EL, Crother TR, Rabizadeh S, Hu B, Wang H, Chen S, et al. Toll-like receptor 2 signaling protects mice from tumor development in a mouse model of colitis-induced cancer. PLoS One. 2010 Sep 27;5(9):e13027. DOI: 10.1371/journal.pone.0013027.
  86. Maeda S, Hikiba Y, Sakamoto K, Nakagawa H, Hirata Y, Hayakawa Y, et al. Colon cancer-derived factors activate NF-kappaB in myeloid cells via TLR2 to link inflammation and tumorigenesis. Mol Med Rep. 2011 Nov-Dec;4(6):1083-8. DOI: 10.3892/mmr.2011.545.
  87. Fukata M, Hernandez Y, Conduah D, Cohen J, Chen A, Breglio K, et al. Innate immune signaling by Toll-like receptor-4 (TLR4) shapes the inflammatory microenvironment in colitis-associated tumors. Inflamm Bowel Dis. 2009 Jul;15(7):997-1006. DOI: 10.1002/ibd.20880.
  88. Zaki MH, Vogel P, Body-Malapel M, Lamkanfi M, Kanneganti TD. IL-18 production downstream of the Nlrp3 inflammasome confers protection against colorectal tumor formation. J Immunol. 2010 Oct 15;185(8):4912-20. DOI: 10.4049/jimmunol.1002046.
  89. Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 2011 May 27;145(5):745-57. DOI: 10.1016/j.cell.2011.04.022.
  90. Nenci A, Becker C, Wullaert A, Gareus R, van Loo G, Danese S, et al. Epithelial NEMO links innate immunity to chronic intestinal inflammation. Nature. 2007 Mar 29;446(7135):557-61. DOI: 10.1038/nature05698.
  91. Edelblum KL, Washington MK, Koyama T, Robine S, Baccarini M, Polk DB. Raf protects against colitis by promoting mouse colon epithelial cell survival through NF-kappaB. Gastroenterology. 2008;135(2):539-51.
  92. Fukata M, Chen A, Klepper A, Krishnareddy S, Vamadevan AS, Thomas LS, et al. Cox-2 is regulated by Toll-like receptor-4 (TLR4) signaling: Role in proliferation and apoptosis in the intestine. Gastroenterology. 2006;131(3):862-77.
  93. Santaolalla R, Sussman DA, Ruiz JR, Davies JM, Pastorini C, Espana CL, et al. TLR4 activates the beta-catenin pathway to cause intestinal neoplasia. PLoS One. 2013;8(5):e63298. DOI: 10.1371/journal.pone.0063298.
  94. Kuo WT, Lee TC, Yu LC. Janus-faced bacterial regulation of epithelial cell death and survival: Association with colon carcinogenesis. Mol Cell Oncol. 2016 Jan;3(1):e1029064. DOI: 10.1080/23723556.2015.1029064.
  95. Shirkey TW, Siggers RH, Goldade BG, Marshall JK, Drew MD, Laarveld B, et al. Effects of commensal bacteria on intestinal morphology and expression of proinflammatory cytokines in the gnotobiotic pig. Exp Biol Med (Maywood). 2006 Sep;231(8):1333-45.
  96. Willing BP, Van Kessel AG. Enterocyte proliferation and apoptosis in the caudal small intestine is influenced by the composition of colonizing commensal bacteria in the neonatal gnotobiotic pig. J Anim Sci. 2007;85(12):3256-66.
  97. Fukata M, Michelsen KS, Eri R, Thomas LS, Hu B, Lukasek K, et al. Toll-like receptor-4 is required for intestinal response to epithelial injury and limiting bacterial translocation in a murine model of acute colitis. Am J Physiol GastrointestLiver Physiol. 2005;288(5):G1055-G65.
  98. Prosberg M, Bendtsen F, Vind I, Petersen AM, Gluud LL. The association between the gut microbiota and the inflammatory bowel disease activity: a systematic review and meta-analysis. Scand J Gastroenterol. 2016 Dec;51(12):1407-15. DOI: 10.1080/00365521.2016.1216587.
  99. Forbes JD, Van Domselaar G, Bernstein CN. Microbiome Survey of the Inflamed and Noninflamed Gut at Different Compartments Within the Gastrointestinal Tract of Inflammatory Bowel Disease Patients. Inflamm Bowel Dis. 2016 Apr;22(4):817-25. DOI: 10.1097/MIB.0000000000000684.
  100. Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, et al. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS One. 2011 Jan 27;6(1):e16393. DOI: 10.1371/journal.pone.0016393.
  101. Wang T, Cai G, Qiu Y, Fei N, Zhang M, Pang X, et al. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 2012 Feb;6(2):320-9. DOI: 10.1038/ismej.2011.109.
  102. Kasai C, Sugimoto K, Moritani I, Tanaka J, Oya Y, Inoue H, et al. Comparison of human gut microbiota in control subjects and patients with colorectal carcinoma in adenoma: Terminal restriction fragment length polymorphism and next-generation sequencing analyses. Oncol Rep. 2016 Jan;35(1):325-33. DOI: 10.3892/or.2015.4398.
  103. Shen XJ, Rawls JF, Randall T, Burcal L, Mpande CN, Jenkins N, et al. Molecular characterization of mucosal adherent bacteria and associations with colorectal adenomas. Gut Microbes. 2010 May-Jun;1(3):138-47. DOI: 10.4161/gmic.1.3.12360.
  104. Sanapareddy N, Legge RM, Jovov B, McCoy A, Burcal L, Araujo-Perez F, et al. Increased rectal microbial richness is associated with the presence of colorectal adenomas in humans. ISME J. 2012 Oct;6(10):1858-68. DOI: 10.1038/ismej.2012.43.
  105. Swidsinski A, Ladhoff A, Pernthaler A, Swidsinski S, Loening-Baucke V, Ortner M, et al. Mucosal flora in inflammatory bowel disease. Gastroenterology. 2002 Jan;122(1):44-54.
  106. Swidsinski A, Khilkin M, Kerjaschki D, Schreiber S, Ortner M, Weber J, et al. Association between intraepithelial Escherichia coli and colorectal cancer. Gastroenterology. 1998 Aug;115(2):281-6.
  107. Reddy BS, Weisburger JH, Narisawa T, Wynder EL. Colon carcinogenesis in germ-free rats with 1,2-dimethylhydrazine and N-methyl-n’-nitro-N-nitrosoguanidine. Cancer Res. 1974 Sep;34(9):2368-72.
  108. Hudcovic T, Stepankova R, Cebra J, Tlaskalova-Hogenova H. The role of microflora in the development of intestinal inflammation: acute and chronic colitis induced by dextran sulfate in germ-free and conventionally reared immunocompetent and immunodeficient mice. Folia Microbiol (Praha). 2001;46(6):565-72.
  109. Kim SC, Tonkonogy SL, Albright CA, Tsang J, Balish EJ, Braun J, et al. Variable phenotypes of enterocolitis in interleukin 10-deficient mice monoassociated with two different commensal bacteria. Gastroenterology. 2005 Apr;128(4):891-906.
  110. Bohn E, Bechtold O, Zahir N, Frick JS, Reimann J, Jilge B, et al. Host gene expression in the colon of gnotobiotic interleukin-2-deficient mice colonized with commensal colitogenic or noncolitogenic bacterial strains: common patterns and bacteria strain specific signatures. Inflamm Bowel Dis. 2006 Sep;12(9):853-62. DOI: 10.1097/01.mib.0000231574.73559.75.
  111. Chung H, Pamp SJ, Hill JA, Surana NK, Edelman SM, Troy EB, et al. Gut immune maturation depends on colonization with a host-specific microbiota. Cell. 2012 Jun 22;149(7):1578-93. DOI: 10.1016/j.cell.2012.04.037.
  112. Vannucci L, Stepankova R, Kozakova H, Fiserova A, Rossmann P, Tlaskalova-Hogenova H. Colorectal carcinogenesis in germ-free and conventionally reared rats: different intestinal environments affect the systemic immunity. Int J Oncol. 2008 Mar;32(3):609-17.
  113. Rhee KJ, Sethupathi P, Driks A, Lanning DK, Knight KL. Role of commensal bacteria in development of gut-associated lymphoid tissues and preimmune antibody repertoire. J Immunol. 2004 Jan 15;172(2):1118-24.
  114. Zackular JP, Baxter NT, Iverson KD, Sadler WD, Petrosino JF, Chen GY, et al. The gut microbiome modulates colon tumorigenesis. MBio. 2013 Nov 05;4(6):e00692-13. DOI: 10.1128/mBio.00692-13.
  115. Zackular JP, Baxter NT, Chen GY, Schloss PD. Manipulation of the Gut Microbiota Reveals Role in Colon Tumorigenesis. mSphere. 2016 Jan-Feb;1(1). DOI: 10.1128/mSphere.00001-15.
  116. Rehman A, Sina C, Gavrilova O, Hasler R, Ott S, Baines JF, et al. Nod2 is essential for temporal development of intestinal microbial communities. Gut. 2011;60(10):1354-62.
  117. Levy M, Thaiss CA, Zeevi D, Dohnalova L, Zilberman-Schapira G, Mahdi JA, et al. Microbiota-Modulated Metabolites Shape the Intestinal Microenvironment by Regulating NLRP6 Inflammasome Signaling. Cell. 2015 Dec 03;163(6):1428-43. DOI: 10.1016/j.cell.2015.10.048.
  118. Wehkamp J, Harder J, Weichenthal M, Schwab M, Schaffeler E, Schlee M, et al. NOD2 (CARD15) mutations in Crohn’s disease are associated with diminished mucosal alpha-defensin expression. Gut. 2004 Nov;53(11):1658-64. DOI: 10.1136/gut.2003.032805.
  119. Scarpa M, Grillo A, Scarpa M, Brun P, Castoro C, Pozza A, et al. Innate immune environment in ileal pouch mucosa: alpha5 defensin up-regulation as predictor of chronic/relapsing pouchitis. J Gastrointest Surg. 2012 Jan;16(1):188-201; discussion -2. DOI: 10.1007/s11605-011-1720-6.
  120. Menendez A, Willing BP, Montero M, Wlodarska M, So CC, Bhinder G, et al. Bacterial stimulation of the TLR-MyD88 pathway modulates the homeostatic expression of ileal Paneth cell alpha-defensins. J Innate Immun. 2013;5(1):39-49. DOI: 10.1159/000341630.
  121. Ayala-Sumuano JT, Tellez-Lopez VM, Dominguez-Robles Mdel C, Shibayama-Salas M, Meza I. Toll-like receptor signaling activation by Entamoeba histolytica induces beta defensin 2 in human colonic epithelial cells: its possible role as an element of the innate immune response. PLoS Negl Trop Dis. 2013;7(2):e2083. DOI: 10.1371/journal.pntd.0002083.
  122. Rahimi R, Nikfar S, Abdollahi M. Meta-analysis technique confirms the effectiveness of anti-TNF-alpha in the management of active ulcerative colitis when administered in combination with corticosteroids. Med Sci Monit. 2007 Jul;13(7):PI13-8.
  123. Khan KJ, Ullman TA, Ford AC, Abreu MT, Abadir A, Marshall JK, et al. Antibiotic therapy in inflammatory bowel disease: a systematic review and meta-analysis. Am J Gastroenterol. 2011 Apr;106(4):661-73. DOI: 10.1038/ajg.2011.72.
  124. Wang SL, Wang ZR, Yang CQ. Meta-analysis of broad-spectrum antibiotic therapy in patients with active inflammatory bowel disease. Exp Ther Med. 2012 Dec;4(6):1051-6. DOI: 10.3892/etm.2012.718.
  125. Baxter NT, Zackular JP, Chen GY, Schloss PD. Structure of the gut microbiome following colonization with human feces determines colonic tumor burden. Microbiome. 2014;2:20. DOI: 10.1186/2049-2618-2-20.
  126. Dignass A, Van Assche G, Lindsay JO, Lemann M, Soderholm J, Colombel JF, et al. The second European evidence-based Consensus on the diagnosis and management of Crohn’s disease: Current management. J Crohns Colitis. 2010 Feb;4(1):28-62. DOI: 10.1016/j.crohns.2009.12.002.
  127. Ianiro G, Tilg H, Gasbarrini A. Antibiotics as deep modulators of gut microbiota: between good and evil. Gut. 2016 Nov;65(11):1906-15. DOI: 10.1136/gutjnl-2016-312297.
  128. Arthur JC, Perez-Chanona E, Muhlbauer M, Tomkovich S, Uronis JM, Fan TJ, et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 2012 Oct 05;338(6103):120-3. DOI: 10.1126/science.1224820.
  129. Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X, Yen HR, et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med. 2009 Sep;15(9):1016-22. DOI: 10.1038/nm.2015.
  130. Bonnet M, Buc E, Sauvanet P, Darcha C, Dubois D, Pereira B, et al. Colonization of the human gut by E. coli and colorectal cancer risk. Clin Cancer Res. 2014 Feb 15;20(4):859-67. DOI: 10.1158/1078-0432.CCR-13-1343.
  131. Prorok-Hamon M, Friswell MK, Alswied A, Roberts CL, Song F, Flanagan PK, et al. Colonic mucosa-associated diffusely adherent afaC+ Escherichia coli expressing lpfA and pks are increased in inflammatory bowel disease and colon cancer. Gut. 2014 May;63(5):761-70. DOI: 10.1136/gutjnl-2013-304739.
  132. Chassaing B, Rolhion N, de Vallee A, Salim SY, Prorok-Hamon M, Neut C, et al. Crohn disease–associated adherent-invasive E. coli bacteria target mouse and human Peyer’s patches via long polar fimbriae. J Clin Invest. 2011 Mar;121(3):966-75. DOI: 10.1172/JCI44632.
  133. Martin HM, Campbell BJ, Hart CA, Mpofu C, Nayar M, Singh R, et al. Enhanced Escherichia coli adherence and invasion in Crohn’s disease and colon cancer. Gastroenterology. 2004 Jul;127(1):80-93.
  134. Chen L, Wang W, Zhou R, Ng SC, Li J, Huang M, et al. Characteristics of fecal and mucosa-associated microbiota in Chinese patients with inflammatory bowel disease. Medicine (Baltimore). 2014 Aug;93(8):e51. DOI: 10.1097/MD.0000000000000051.
  135. Raisch J, Buc E, Bonnet M, Sauvanet P, Vazeille E, de Vallee A, et al. Colon cancer-associated B2 Escherichia coli colonize gut mucosa and promote cell proliferation. World J Gastroenterol. 2014 Jun 07;20(21):6560-72. DOI: 10.3748/wjg.v20.i21.6560.
  136. Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, Glasser AL, Barnich N, et al. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology. 2004 Aug;127(2):412-21. DOI: S0016508504007711 [pii].
  137. Martinez-Medina M, Aldeguer X, Lopez-Siles M, Gonzalez-Huix F, Lopez-Oliu C, Dahbi G, et al. Molecular diversity of Escherichia coli in the human gut: new ecological evidence supporting the role of adherent-invasive E. coli (AIEC) in Crohn’s disease. Inflamm Bowel Dis. 2009 Jun;15(6):872-82. DOI: 10.1002/ibd.20860.
  138. Denizot J, Sivignon A, Barreau F, Darcha C, Chan HF, Stanners CP, et al. Adherent-invasive Escherichia coli induce claudin-2 expression and barrier defect in CEABAC10 mice and Crohn’s disease patients. Inflamm Bowel Dis. 2012 Feb;18(2):294-304. DOI: 10.1002/ibd.21787.
  139. Mimouna S, Goncalves D, Barnich N, Darfeuille-Michaud A, Hofman P, Vouret-Craviari V. Crohn disease-associated Escherichia coli promote gastrointestinal inflammatory disorders by activation of HIF-dependent responses. Gut Microbes. 2011 Nov-Dec;2(6):335-46. DOI: 10.4161/gmic.18771.
  140. Arthur JC, Gharaibeh RZ, Muhlbauer M, Perez-Chanona E, Uronis JM, McCafferty J, et al. Microbial genomic analysis reveals the essential role of inflammation in bacteria-induced colorectal cancer. Nat Commun. 2014 Sep 03;5:4724. DOI: 10.1038/ncomms5724.
  141. Cougnoux A, Dalmasso G, Martinez R, Buc E, Delmas J, Gibold L, et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut. 2014 Dec;63(12):1932-42. DOI: 10.1136/gutjnl-2013-305257.
  142. Chan CH, Cook D, Stanners CP. Increased colon tumor susceptibility in azoxymethane treated CEABAC transgenic mice. Carcinogenesis. 2006 Sep;27(9):1909-16. DOI: 10.1093/carcin/bgl040.
  143. Carvalho FA, Barnich N, Sivignon A, Darcha C, Chan CH, Stanners CP, et al. Crohn’s disease adherent-invasive Escherichia coli colonize and induce strong gut inflammation in transgenic mice expressing human CEACAM. J Exp Med. 2009 Sep 28;206(10):2179-89. DOI: 10.1084/jem.20090741.
  144. Naghibalhossaini F, Sayadi K, Jaberie H, Bazargani A, Eftekhar E, Hosseinzadeh M. Inhibition of CEA release from epithelial cells by lipid A of Gram-negative bacteria. Cell Mol Biol Lett. 2015 Sep;20(3):374-84. DOI: 10.1515/cmble-2015-0022.
  145. Pakdel A, Naghibalhossaini F, Mokarram P, Jaberipour M, Hosseini A. Regulation of carcinoembryonic antigen release from colorectal cancer cells. Mol Biol Rep. 2012 Apr;39(4):3695-704. DOI: 10.1007/s11033-011-1144-0.
  146. Raisch J, Rolhion N, Dubois A, Darfeuille-Michaud A, Bringer MA. Intracellular colon cancer-associated Escherichia coli promote protumoral activities of human macrophages by inducing sustained COX-2 expression. Lab Invest. 2015 Mar;95(3):296-307. DOI: 10.1038/labinvest.2014.161.
  147. Martinez-Medina M, Denizot J, Dreux N, Robin F, Billard E, Bonnet R, et al. Western diet induces dysbiosis with increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC colonisation. Gut. 2014 Jan;63(1):116-24. DOI: 10.1136/gutjnl-2012-304119.
  148. Wine E, Ossa JC, Gray-Owen SD, Sherman PM. Adherent-invasive Escherichia coli, strain LF82 disrupts apical junctional complexes in polarized epithelia. BMC Microbiol. 2009 Aug 26;9:180. DOI: 10.1186/1471-2180-9-180.
  149. Sasaki M, Sitaraman SV, Babbin BA, Gerner-Smidt P, Ribot EM, Garrett N, et al. Invasive Escherichia coli are a feature of Crohn’s disease. Lab Invest. 2007 Oct;87(10):1042-54. DOI: 10.1038/labinvest.3700661.
  150. Bashir A, Miskeen AY, Bhat A, Fazili KM, Ganai BA. Fusobacterium nucleatum: an emerging bug in colorectal tumorigenesis. Eur J Cancer Prev. 2015 Sep;24(5):373-85. DOI: 10.1097/CEJ.0000000000000116.
  151. Bashir A, Miskeen AY, Hazari YM, Asrafuzzaman S, Fazili KM. Fusobacterium nucleatum, inflammation, and immunity: the fire within human gut. Tumour Biol. 2016 Mar;37(3):2805-10. DOI: 10.1007/s13277-015-4724-0.
  152. Wu N, Yang X, Zhang R, Li J, Xiao X, Hu Y, et al. Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb Ecol. 2013 Aug;66(2):462-70. DOI: 10.1007/s00248-013-0245-9.
  153. Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012 Feb;22(2):292-8. DOI: 10.1101/gr.126573.111.
  154. Burns MB, Lynch J, Starr TK, Knights D, Blekhman R. Virulence genes are a signature of the microbiome in the colorectal tumor microenvironment. Genome Med. 2015;7(1):55. DOI: 10.1186/s13073-015-0177-8.
  155. Viljoen KS, Dakshinamurthy A, Goldberg P, Blackburn JM. Quantitative profiling of colorectal cancer-associated bacteria reveals associations between fusobacterium spp., enterotoxigenic Bacteroides fragilis (ETBF) and clinicopathological features of colorectal cancer. PLoS One. 2015;10(3):e0119462. DOI: 10.1371/journal.pone.0119462.
  156. Yu J, Feng Q, Wong SH, Zhang D, Liang QY, Qin Y, et al. Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut. 2017 Jan;66(1):70-8. DOI: 10.1136/gutjnl-2015-309800.
  157. Gao Z, Guo B, Gao R, Zhu Q, Qin H. Microbiota disbiosis is associated with colorectal cancer. Front Microbiol. 2015;6:20. DOI: 10.3389/fmicb.2015.00020.
  158. Dharmani P, Strauss J, Ambrose C, Allen-Vercoe E, Chadee K. Fusobacterium nucleatum infection of colonic cells stimulates MUC2 mucin and tumor necrosis factor alpha. Infect Immun. 2011 Jul;79(7):2597-607. DOI: 10.1128/IAI.05118-11.
  159. Strauss J, Kaplan GG, Beck PL, Rioux K, Panaccione R, Devinney R, et al. Invasive potential of gut mucosa-derived Fusobacterium nucleatum positively correlates with IBD status of the host. Inflamm Bowel Dis. 2011 Sep;17(9):1971-8. DOI: 10.1002/ibd.21606.
  160. Mima K, Nishihara R, Qian ZR, Cao Y, Sukawa Y, Nowak JA, et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut. 2016 Dec;65(12):1973-80. DOI: 10.1136/gutjnl-2015-310101.
  161. Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013 Aug 14;14(2):207-15. DOI: 10.1016/j.chom.2013.07.007.
  162. Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013 Aug 14;14(2):195-206. DOI: 10.1016/j.chom.2013.07.012.
  163. Fardini Y, Wang X, Temoin S, Nithianantham S, Lee D, Shoham M, et al. Fusobacterium nucleatum adhesin FadA binds vascular endothelial cadherin and alters endothelial integrity. Mol Microbiol. 2011 Dec;82(6):1468-80. DOI: 10.1111/j.1365-2958.2011.07905.x.
  164. Abed J, Emgard JE, Zamir G, Faroja M, Almogy G, Grenov A, et al. Fap2 Mediates Fusobacterium nucleatum Colorectal Adenocarcinoma Enrichment by Binding to Tumor-Expressed Gal-GalNAc. Cell Host Microbe. 2016 Aug 10;20(2):215-25. DOI: 10.1016/j.chom.2016.07.006.
  165. Kaplan CW, Ma X, Paranjpe A, Jewett A, Lux R, Kinder-Haake S, et al. Fusobacterium nucleatum outer membrane proteins Fap2 and RadD induce cell death in human lymphocytes. Infect Immun. 2010 Nov;78(11):4773-8. DOI: 10.1128/IAI.00567-10.
  166. Sears CL, Islam S, Saha A, Arjumand M, Alam NH, Faruque AS, et al. Association of enterotoxigenic Bacteroides fragilis infection with inflammatory diarrhea. Clin Infect Dis. 2008 Sep 15;47(6):797-803. DOI: 10.1086/591130.
  167. Toprak NU, Yagci A, Gulluoglu BM, Akin ML, Demirkalem P, Celenk T, et al. A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer. Clin Microbiol Infect. 2006 Aug;12(8):782-6. DOI: 10.1111/j.1469-0691.2006.01494.x.
  168. Boleij A, Hechenbleikner EM, Goodwin AC, Badani R, Stein EM, Lazarev MG, et al. The Bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients. Clin Infect Dis. 2015 Jan 15;60(2):208-15. DOI: 10.1093/cid/ciu787.
  169. Rabizadeh S, Rhee KJ, Wu S, Huso D, Gan CM, Golub JE, et al. Enterotoxigenic bacteroides fragilis: a potential instigator of colitis. Inflamm Bowel Dis. 2007 Dec;13(12):1475-83. DOI: 10.1002/ibd.20265.
  170. Rhee KJ, Wu S, Wu X, Huso DL, Karim B, Franco AA, et al. Induction of persistent colitis by a human commensal, enterotoxigenic Bacteroides fragilis, in wild-type C57BL/6 mice. Infect Immun. 2009 Apr;77(4):1708-18. DOI: 10.1128/IAI.00814-08.
  171. Wick EC, Rabizadeh S, Albesiano E, Wu X, Wu S, Chan J, et al. Stat3 activation in murine colitis induced by enterotoxigenic Bacteroides fragilis. Inflamm Bowel Dis. 2014 May;20(5):821-34. DOI: 10.1097/MIB.0000000000000019.
  172. Goodwin AC, Destefano Shields CE, Wu S, Huso DL, Wu X, Murray-Stewart TR, et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc Natl Acad Sci U S A. 2011 Sep 13;108(37):15354-9. DOI: 10.1073/pnas.1010203108.
  173. Ishaq S, Nunn L. Helicobacter pylori and gastric cancer: a state of the art review. Gastroenterol Hepatol Bed Bench. 2015 Spring;8(Suppl 1):S6-S14.
  174. Naumann M, Sokolova O, Tegtmeyer N, Backert S. Helicobacter pylori: A Paradigm Pathogen for Subverting Host Cell Signal Transmission. Trends Microbiol. 2017 Apr;25(4):316-28. DOI: 10.1016/j.tim.2016.12.004.
  175. Shmuely H, Passaro D, Figer A, Niv Y, Pitlik S, Samra Z, et al. Relationship between Helicobacter pylori CagA status and colorectal cancer. Am J Gastroenterol. 2001 Dec;96(12):3406-10. DOI: 10.1111/j.1572-0241.2001.05342.x.
  176. Inoue I, Mukoubayashi C, Yoshimura N, Niwa T, Deguchi H, Watanabe M, et al. Elevated risk of colorectal adenoma with Helicobacter pylori-related chronic gastritis: a population-based case-control study. Int J Cancer. 2011 Dec 01;129(11):2704-11. DOI: 10.1002/ijc.25931.
  177. Nam JH, Hong CW, Kim BC, Shin A, Ryu KH, Park BJ, et al. Helicobacter pylori infection is an independent risk factor for colonic adenomatous neoplasms. Cancer Causes Control. 2017 Feb;28(2):107-15. DOI: 10.1007/s10552-016-0839-x.
  178. Machida-Montani A, Sasazuki S, Inoue M, Natsukawa S, Shaura K, Koizumi Y, et al. Atrophic gastritis, Helicobacter pylori, and colorectal cancer risk: a case-control study. Helicobacter. 2007 Aug;12(4):328-32. DOI: 10.1111/j.1523-5378.2007.00513.x.
  179. Siddheshwar RK, Muhammad KB, Gray JC, Kelly SB. Seroprevalence of Helicobacter pylori in patients with colorectal polyps and colorectal carcinoma. Am J Gastroenterol. 2001 Jan;96(1):84-8. DOI: 10.1111/j.1572-0241.2001.03355.x.
  180. Moss SF, Neugut AI, Garbowski GC, Wang S, Treat MR, Forde KA. Helicobacter pylori seroprevalence and colorectal neoplasia: evidence against an association. J Natl Cancer Inst. 1995 May 17;87(10):762-3.
  181. Boulard O, Kirchberger S, Royston DJ, Maloy KJ, Powrie FM. Identification of a genetic locus controlling bacteria-driven colitis and associated cancer through effects on innate inflammation. J Exp Med. 2012 Jul 02;209(7):1309-24. DOI: 10.1084/jem.20120239.
  182. Mangerich A, Knutson CG, Parry NM, Muthupalani S, Ye W, Prestwich E, et al. Infection-induced colitis in mice causes dynamic and tissue-specific changes in stress response and DNA damage leading to colon cancer. Proc Natl Acad Sci U S A. 2012 Jul 03;109(27):E1820-9. DOI: 10.1073/pnas.1207829109.
  183. Nagamine CM, Sohn JJ, Rickman BH, Rogers AB, Fox JG, Schauer DB. Helicobacter hepaticus infection promotes colon tumorigenesis in the BALB/c-Rag2(-/-) Apc(Min/+) mouse. Infect Immun. 2008 Jun;76(6):2758-66. DOI: 10.1128/IAI.01604-07.
  184. Nagamine CM, Rogers AB, Fox JG, Schauer DB. Helicobacter hepaticus promotes azoxymethane-initiated colon tumorigenesis in BALB/c-IL10-deficient mice. Int J Cancer. 2008 Feb 15;122(4):832-8. DOI: 10.1002/ijc.23175.
  185. Ward JM, Anver MR, Haines DC, Melhorn JM, Gorelick P, Yan L, et al. Inflammatory large bowel disease in immunodeficient mice naturally infected with Helicobacter hepaticus. Lab Anim Sci. 1996 Feb;46(1):15-20.
  186. Li X, Fox JG, Whary MT, Yan L, Shames B, Zhao Z. SCID/NCr mice naturally infected with Helicobacter hepaticus develop progressive hepatitis, proliferative typhlitis, and colitis. Infect Immun. 1998 Nov;66(11):5477-84.
  187. Nanda Kumar NS, Balamurugan R, Jayakanthan K, Pulimood A, Pugazhendhi S, Ramakrishna BS. Probiotic administration alters the gut flora and attenuates colitis in mice administered dextran sodium sulfate. J Gastroenterol Hepatol. 2008 Dec;23(12):1834-9. DOI: 10.1111/j.1440-1746.2008.05723.x.
  188. Mennigen R, Nolte K, Rijcken E, Utech M, Loeffler B, Senninger N, et al. Probiotic mixture VSL#3 protects the epithelial barrier by maintaining tight junction protein expression and preventing apoptosis in a murine model of colitis. Am J Physiol Gastrointest Liver Physiol. 2009 May;296(5):G1140-9. DOI: 10.1152/ajpgi.90534.2008.
  189. Bassaganya-Riera J, Viladomiu M, Pedragosa M, De Simone C, Hontecillas R. Immunoregulatory mechanisms underlying prevention of colitis-associated colorectal cancer by probiotic bacteria. PLoS One. 2012;7(4):e34676. DOI: 10.1371/journal.pone.0034676.
  190. O’Mahony L, Feeney M, O’Halloran S, Murphy L, Kiely B, Fitzgibbon J, et al. Probiotic impact on microbial flora, inflammation and tumour development in IL-10 knockout mice. Aliment Pharmacol Ther. 2001 Aug;15(8):1219-25.
  191. Madsen KL, Doyle JS, Jewell LD, Tavernini MM, Fedorak RN. Lactobacillus species prevents colitis in interleukin 10 gene-deficient mice. Gastroenterology. 1999 May;116(5):1107-14.
  192. Chen CC, Lin WC, Kong MS, Shi HN, Walker WA, Lin CY, et al. Oral inoculation of probiotics Lactobacillus acidophilus NCFM suppresses tumour growth both in segmental orthotopic colon cancer and extra-intestinal tissue. Br J Nutr. 2012 Jun;107(11):1623-34. DOI: 10.1017/S0007114511004934.
  193. Talero E, Bolivar S, Avila-Roman J, Alcaide A, Fiorucci S, Motilva V. Inhibition of chronic ulcerative colitis-associated adenocarcinoma development in mice by VSL#3. Inflamm Bowel Dis. 2015 May;21(5):1027-37. DOI: 10.1097/MIB.0000000000000346.
  194. Appleyard CB, Cruz ML, Isidro AA, Arthur JC, Jobin C, De Simone C. Pretreatment with the probiotic VSL#3 delays transition from inflammation to dysplasia in a rat model of colitis-associated cancer. Am J Physiol Gastrointest Liver Physiol. 2011 Dec;301(6):G1004-13. DOI: 10.1152/ajpgi.00167.2011.
  195. Kahouli I, Malhotra M, Westfall S, Alaoui-Jamali MA, Prakash S. Design and validation of an orally administrated active L. fermentum-L. acidophilus probiotic formulation using colorectal cancer Apc Min/+ mouse model. Appl Microbiol Biotechnol. 2017 Mar;101(5):1999-2019. DOI: 10.1007/s00253-016-7885-x.
  196. Tiptiri-Kourpeti A, Spyridopoulou K, Santarmaki V, Aindelis G, Tompoulidou E, Lamprianidou EE, et al. Lactobacillus casei Exerts Anti-Proliferative Effects Accompanied by Apoptotic Cell Death and Up-Regulation of TRAIL in Colon Carcinoma Cells. PLoS One. 2016;11(2):e0147960. DOI: 10.1371/journal.pone.0147960.
  197. Soltan Dallal MM, Mojarrad M, Baghbani F, Raoofian R, Mardaneh J, Salehipour Z. Effects of probiotic Lactobacillus acidophilus and Lactobacillus casei on colorectal tumor cells activity (CaCo-2). Arch Iran Med. 2015 Mar;18(3):167-72. DOI: 0151803/AIM.006.
  198. Di Luccia B, Manzo N, Baccigalupi L, Calabro V, Crescenzi E, Ricca E, et al. Lactobacillus gasseri SF1183 affects intestinal epithelial cell survival and growth. PLoS One. 2013;8(7):e69102. DOI: 10.1371/journal.pone.0069102.
  199. Anderson RC, Cookson AL, McNabb WC, Kelly WJ, Roy NC. Lactobacillus plantarum DSM 2648 is a potential probiotic that enhances intestinal barrier function. FEMS MicrobiolLett. 2010;309(2):184-92.
  200. Anderson RC, Cookson AL, McNabb WC, Park Z, McCann MJ, Kelly WJ, et al. Lactobacillus plantarum MB452 enhances the function of the intestinal barrier by increasing the expression levels of genes involved in tight junction formation. BMC Microbiol. 2010 Dec 09;10:316. DOI: 10.1186/1471-2180-10-316.
  201. Orlando A, Linsalata M, Notarnicola M, Tutino V, Russo F. Lactobacillus GG restoration of the gliadin induced epithelial barrier disruption: the role of cellular polyamines. BMC Microbiol. 2014 Jan 31;14:19. DOI: 10.1186/1471-2180-14-19.
  202. Putaala H, Salusjarvi T, Nordstrom M, Saarinen M, Ouwehand AC, Bech Hansen E, et al. Effect of four probiotic strains and Escherichia coli O157:H7 on tight junction integrity and cyclo-oxygenase expression. Res Microbiol. 2008 Nov-Dec;159(9-10):692-8. DOI: 10.1016/j.resmic.2008.08.002.
  203. Hsieh CY, Osaka T, Moriyama E, Date Y, Kikuchi J, Tsuneda S. Strengthening of the intestinal epithelial tight junction by Bifidobacterium bifidum. Physiol Rep. 2015 Mar;3(3). DOI: 10.14814/phy2.12327.
  204. So SS, Wan ML, El-Nezami H. Probiotics-mediated suppression of cancer. Curr Opin Oncol. 2017 Jan;29(1):62-72. DOI: 10.1097/CCO.0000000000000342.
  205. Kumar M, Nagpal R, Verma V, Kumar A, Kaur N, Hemalatha R, et al. Probiotic metabolites as epigenetic targets in the prevention of colon cancer. Nutr Rev. 2013 Jan;71(1):23-34. DOI: 10.1111/j.1753-4887.2012.00542.x.
  206. Fyderek K, Strus M, Kowalska-Duplaga K, Gosiewski T, Wedrychowicz A, Jedynak-Wasowicz U, et al. Mucosal bacterial microflora and mucus layer thickness in adolescents with inflammatory bowel disease. World J Gastroenterol. 2009 Nov 14;15(42):5287-94.
  207. Mylonaki M, Rayment NB, Rampton DS, Hudspith BN, Brostoff J. Molecular characterization of rectal mucosa-associated bacterial flora in inflammatory bowel disease. Inflamm Bowel Dis. 2005 May;11(5):481-7.
  208. Gueimonde M, Ouwehand A, Huhtinen H, Salminen E, Salminen S. Qualitative and quantitative analyses of the bifidobacterial microbiota in the colonic mucosa of patients with colorectal cancer, diverticulitis and inflammatory bowel disease. World J Gastroenterol. 2007 Aug 07;13(29):3985-9.
  209. Wang W, Chen L, Zhou R, Wang X, Song L, Huang S, et al. Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease. J Clin Microbiol. 2014 Feb;52(2):398-406. DOI: 10.1128/JCM.01500-13.
  210. Durchschein F, Petritsch W, Hammer HF. Diet therapy for inflammatory bowel diseases: The established and the new. World J Gastroenterol. 2016 Feb 21;22(7):2179-94. DOI: 10.3748/wjg.v22.i7.2179.
  211. Roediger WE. Role of anaerobic bacteria in the metabolic welfare of the colonic mucosa in man. Gut. 1980 Sep;21(9):793-8.
  212. Roediger WE. Utilization of nutrients by isolated epithelial cells of the rat colon. Gastroenterology. 1982 Aug;83(2):424-9.
  213. Donohoe DR, Garge N, Zhang X, Sun W, O’Connell TM, Bunger MK, et al. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011 May 04;13(5):517-26. DOI: 10.1016/j.cmet.2011.02.018.
  214. Huang CY, Pai YC, Yu LC. Glucose-mediated cytoprotection in the gut epithelium under ischemic and hypoxic stress. Histol Histopathol. 2016 Nov 08:11839. DOI: 10.14670/HH-11-839.
  215. Huang CY, Kuo WT, Huang CY, Lee TC, Chen CT, Peng WH, et al. Distinct cytoprotective roles of pyruvate and ATP by glucose metabolism on epithelial necroptosis and crypt proliferation in ischaemic gut. J Physiol. 2017 Jan 15;595(2):505-21. DOI: 10.1113/JP272208.
  216. Davie JR. Inhibition of histone deacetylase activity by butyrate. J Nutr. 2003 Jul;133(7 Suppl):2485S-93S.
  217. Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost FJ, Brummer RJ. Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther. 2008 Jan 15;27(2):104-19. DOI: 10.1111/j.1365-2036.2007.03562.x.
  218. Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2014 Aug;63(8):1275-83. DOI: 10.1136/gutjnl-2013-304833.
  219. Balamurugan R, Rajendiran E, George S, Samuel GV, Ramakrishna BS. Real-time polymerase chain reaction quantification of specific butyrate-producing bacteria, Desulfovibrio and Enterococcus faecalis in the feces of patients with colorectal cancer. J Gastroenterol Hepatol. 2008 Aug;23(8 Pt 1):1298-303. DOI: 10.1111/j.1440-1746.2008.05490.x.
  220. Weir TL, Manter DK, Sheflin AM, Barnett BA, Heuberger AL, Ryan EP. Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults. PLoS One. 2013;8(8):e70803. DOI: 10.1371/journal.pone.0070803.
  221. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A. 2007 Aug 21;104(34):13780-5. DOI: 10.1073/pnas.0706625104.
  222. Ohkawara S, Furuya H, Nagashima K, Asanuma N, Hino T. Oral administration of butyrivibrio fibrisolvens, a butyrate-producing bacterium, decreases the formation of aberrant crypt foci in the colon and rectum of mice. J Nutr. 2005 Dec;135(12):2878-83.
  223. Perrin P, Pierre F, Patry Y, Champ M, Berreur M, Pradal G, et al. Only fibres promoting a stable butyrate producing colonic ecosystem decrease the rate of aberrant crypt foci in rats. Gut. 2001 Jan;48(1):53-61.
  224. Donohoe DR, Holley D, Collins LB, Montgomery SA, Whitmore AC, Hillhouse A, et al. A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner. Cancer Discov. 2014 Dec;4(12):1387-97. DOI: 10.1158/2159-8290.CD-14-0501.
  225. Wong JM, de Souza R, Kendall CW, Emam A, Jenkins DJ. Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol. 2006 Mar;40(3):235-43.
  226. Huang CY, Kuo WT, Huang YC, Lee TC, Yu LC. Resistance to hypoxia-induced necroptosis is conferred by glycolytic pyruvate scavenging of mitochondrial superoxide in colorectal cancer cells. Cell Death Dis. 2013 May 02;4:e622. DOI: 10.1038/cddis.2013.149.
  227. Bringer MA, Barnich N, Glasser AL, Bardot O, Darfeuille-Michaud A. HtrA stress protein is involved in intramacrophagic replication of adherent and invasive Escherichia coli strain LF82 isolated from a patient with Crohn’s disease. Infect Immun. 2005 Feb;73(2):712-21. DOI: 10.1128/IAI.73.2.712-721.2005.
  228. Dreux N, Denizot J, Martinez-Medina M, Mellmann A, Billig M, Kisiela D, et al. Point mutations in FimH adhesin of Crohn’s disease-associated adherent-invasive Escherichia coli enhance intestinal inflammatory response. PLoS Pathog. 2013 Jan;9(1):e1003141. DOI: 10.1371/journal.ppat.1003141.
  229. Diaz PI, Zilm PS, Rogers AH. Fusobacterium nucleatum supports the growth of Porphyromonas gingivalis in oxygenated and carbon-dioxide-depleted environments. Microbiology. 2002 Feb;148(Pt 2):467-72. DOI: 10.1099/00221287-148-2-467.
  230. Diaz PI, Zilm PS, Rogers AH. The response to oxidative stress of Fusobacterium nucleatum grown in continuous culture. FEMS Microbiol Lett. 2000 Jun 01;187(1):31-4.
  231. Park Y, Choi JY, Yong D, Lee K, Kim JM. Clinical features and prognostic factors of anaerobic infections: a 7-year retrospective study. Korean J Intern Med. 2009 Mar;24(1):13-8. DOI: 10.3904/kjim.2009.24.1.13.
  232. Rocha ER, Smith CJ. Ferritin-like family proteins in the anaerobe Bacteroides fragilis: when an oxygen storm is coming, take your iron to the shelter. Biometals. 2013 Aug;26(4):577-91. DOI: 10.1007/s10534-013-9650-2.
  233. Chu H, Khosravi A, Kusumawardhani IP, Kwon AH, Vasconcelos AC, Cunha LD, et al. Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science. 2016 May 27;352(6289):1116-20. DOI: 10.1126/science.aad9948.
  234. Shen Y, Giardino Torchia ML, Lawson GW, Karp CL, Ashwell JD, Mazmanian SK. Outer membrane vesicles of a human commensal mediate immune regulation and disease protection. Cell Host Microbe. 2012 Oct 18;12(4):509-20. DOI: 10.1016/j.chom.2012.08.004.
  235. Huang CY, Yu LC. Pathophysiological mechanisms of death resistance in colorectal carcinoma. World J Gastroenterol. 2015 Nov 07;21(41):11777-92. DOI: 10.3748/wjg.v21.i41.11777.
  236. Hassett DJ, Sutton MD, Schurr MJ, Herr AB, Caldwell CC, Matu JO. Pseudomonas aeruginosa hypoxic or anaerobic biofilm infections within cystic fibrosis airways. Trends Microbiol. 2009 Mar;17(3):130-8. DOI: 10.1016/j.tim.2008.12.003.
  237. Tjalsma H, Boleij A, Marchesi JR, Dutilh BE. A bacterial driver-passenger model for colorectal cancer: beyond the usual suspects. Nat Rev Microbiol. 2012 Jun 25;10(8):575-82. DOI: 10.1038/nrmicro2819.
  238. Keymer JE, Galajda P, Lambert G, Liao D, Austin RH. Computation of mutual fitness by competing bacteria. Proc Natl Acad Sci U S A. 2008 Dec 23;105(51):20269-73. DOI: 10.1073/pnas.0810792105.
  239. Majeed H, Gillor O, Kerr B, Riley MA. Competitive interactions in Escherichia coli populations: the role of bacteriocins. ISME J. 2011 Jan;5(1):71-81. DOI: 10.1038/ismej.2010.90.
  240. Brook I, Walker RI. The relationship between Fusobacterium species and other flora in mixed infection. J Med Microbiol. 1986 Mar;21(2):93-100. DOI: 10.1099/00222615-21-2-93.
  241. Li J, Sung CY, Lee N, Ni Y, Pihlajamaki J, Panagiotou G, et al. Probiotics modulated gut microbiota suppresses hepatocellular carcinoma growth in mice. Proc Natl Acad Sci U S A. 2016 Mar 01;113(9):E1306-15. DOI: 10.1073/pnas.1518189113.
  242. Horie H, Zeisig M, Hirayama K, Midtvedt T, Moller L, Rafter J. Probiotic mixture decreases DNA adduct formation in colonic epithelium induced by the food mutagen 2-amino-9H-pyrido[2,3-b]indole in a human-flora associated mouse model. Eur J Cancer Prev. 2003 Apr;12(2):101-7. DOI: 10.1097/01.cej.0000063505.05852.8a.
  243. Hale VL, Chen J, Johnson S, Harrington SC, Yab TC, Smyrk TC, et al. Shifts in the Fecal Microbiota Associated with Adenomatous Polyps. Cancer Epidemiol Biomarkers Prev. 2017 Jan;26(1):85-94. DOI: 10.1158/1055-9965.EPI-16-0337.
  244. Zackular JP, Rogers MA, Ruffin MTt, Schloss PD. The human gut microbiome as a screening tool for colorectal cancer. Cancer Prev Res (Phila). 2014 Nov;7(11):1112-21. DOI: 10.1158/1940-6207.CAPR-14-0129.
  245. Hullar MA, Burnett-Hartman AN, Lampe JW. Gut microbes, diet, and cancer. Cancer Treat Res. 2014;159:377-99. DOI: 10.1007/978-3-642-38007-5_22.
  246. Bernstein C, Holubec H, Bhattacharyya AK, Nguyen H, Payne CM, Zaitlin B, et al. Carcinogenicity of deoxycholate, a secondary bile acid. Arch Toxicol. 2011 Aug;85(8):863-71. DOI: 10.1007/s00204-011-0648-7.
  247. Hagland HR, Soreide K. Cellular metabolism in colorectal carcinogenesis: Influence of lifestyle, gut microbiome and metabolic pathways. Cancer Lett. 2015 Jan 28;356(2 Pt A):273-80. DOI: 10.1016/j.canlet.2014.02.026.
  248. O’Keefe SJ. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol. 2016 Dec;13(12):691-706. DOI: 10.1038/nrgastro.2016.165.
  249. Candela M, Biagi E, Maccaferri S, Turroni S, Brigidi P. Intestinal microbiota is a plastic factor responding to environmental changes. Trends Microbiol. 2012 Aug;20(8):385-91. DOI: 10.1016/j.tim.2012.05.003.
  250. Rasmussen BA, Breen DM, Lam TK. Lipid sensing in the gut, brain and liver. Trends Endocrinol Metab. 2012 Feb;23(2):49-55. DOI: 10.1016/j.tem.2011.11.001.
  251. Cheung GW, Kokorovic A, Lam TK. Upper intestinal lipids regulate energy and glucose homeostasis. Cell Mol Life Sci. 2009 Sep;66(18):3023-7. DOI: 10.1007/s00018-009-0062-y.
  252. Hornef MW, Normark BH, Vandewalle A, Normark S. Intracellular recognition of lipopolysaccharide by toll-like receptor 4 in intestinal epithelial cells. J Exp Med. 2003 Oct 20;198(8):1225-35. DOI: 10.1084/jem.20022194.
  253. Mollen KP, Gribar SC, Anand RJ, Kaczorowski DJ, Kohler JW, Branca MF, et al. Increased expression and internalization of the endotoxin coreceptor CD14 in enterocytes occur as an early event in the development of experimental necrotizing enterocolitis. J Pediatr Surg. 2008;43(6):1175-81.
  254. Yu M, Shao D, Yang J, Feng S, Xu J. Ketamine suppresses intestinal TLR4 expression and NF-kappaB activity in lipopolysaccharide-treated rats. Croat Med J. 2006 Dec;47(6):825-31.
  255. Wu J, Zhang Y, Xin Z, Wu X. The crosstalk between TLR2 and NOD2 in Aspergillus fumigatus keratitis. Mol Immunol. 2015 Apr;64(2):235-43. DOI: 10.1016/j.molimm.2014.11.021.
  256. Otte JM, Cario E, Podolsky DK. Mechanisms of cross hyporesponsiveness to Toll-like receptor bacterial ligands in intestinal epithelial cells. Gastroenterology. 2004 Apr;126(4):1054-70.
  257. Schaffler H, Demircioglu DD, Kuhner D, Menz S, Bender A, Autenrieth IB, et al. NOD2 stimulation by Staphylococcus aureus-derived peptidoglycan is boosted by Toll-like receptor 2 costimulation with lipoproteins in dendritic cells. Infect Immun. 2014 Nov;82(11):4681-8. DOI: 10.1128/IAI.02043-14.
  258. Sun J, Ding Y. NOD2 agonist promotes the production of inflammatory cytokines in VSMC in synergy with TLR2 and TLR4 agonists. ScientificWorldJournal. 2012;2012:607157. DOI: 10.1100/2012/607157.
  259. Khazaie K, Zadeh M, Khan MW, Bere P, Gounari F, Dennis K, et al. Abating colon cancer polyposis by Lactobacillus acidophilus deficient in lipoteichoic acid. Proc Natl Acad Sci U S A. 2012 Jun 26;109(26):10462-7. DOI: 10.1073/pnas.1207230109.
  260. Motta JP, Bermudez-Humaran LG, Deraison C, Martin L, Rolland C, Rousset P, et al. Food-grade bacteria expressing elafin protect against inflammation and restore colon homeostasis. Sci Transl Med. 2012 Oct 31;4(158):158ra44. DOI: 10.1126/scitranslmed.3004212.
  261. Carroll IM, Andrus JM, Bruno-Barcena JM, Klaenhammer TR, Hassan HM, Threadgill DS. Anti-inflammatory properties of Lactobacillus gasseri expressing manganese superoxide dismutase using the interleukin 10-deficient mouse model of colitis. Am J Physiol Gastrointest Liver Physiol. 2007 Oct;293(4):G729-38. DOI: 10.1152/ajpgi.00132.2007.
  262. Coakley M, Johnson MC, McGrath E, Rahman S, Ross RP, Fitzgerald GF, et al. Intestinal bifidobacteria that produce trans-9, trans-11 conjugated linoleic acid: a fatty acid with antiproliferative activity against human colon SW480 and HT-29 cancer cells. Nutr Cancer. 2006;56(1):95-102. DOI: 10.1207/s15327914nc5601_13.
  263. DeWeerdt S. Microbiome: Microbial mystery. Nature. 2015 May 14;521(7551):S10-1. DOI: 10.1038/521S10a.
  264. Doan HQ, Bowen KA, Jackson LA, Evers BM. Toll-like receptor 4 activation increases Akt phosphorylation in colon cancer cells. Anticancer Res. 2009 Jul;29(7):2473-8.
  265. Frolova L, Drastich P, Rossmann P, Klimesova K, Tlaskalova-Hogenova H. Expression of Toll-like receptor 2 (TLR2), TLR4, and CD14 in biopsy samples of patients with inflammatory bowel diseases: upregulated expression of TLR2 in terminal ileum of patients with ulcerative colitis. J Histochem Cytochem. 2008 Mar;56(3):267-74. DOI: 10.1369/jhc.7A7303.2007.
  266. Cario E, Podolsky DK. Differential alteration in intestinal epithelial cell expression of toll-like receptor 3 (TLR3) and TLR4 in inflammatory bowel disease. Infect Immun. 2000 Dec;68(12):7010-7.
  267. Abreu MT, Arnold ET, Thomas LS, Gonsky R, Zhou Y, Hu B, et al. TLR4 and MD-2 expression is regulated by immune-mediated signals in human intestinal epithelial cells. J Biol Chem. 2002;277(23):20431-7.

 

 

 

Multiselect Ultimate Query Plugin by InoPlugs Web Design Vienna | Webdesign Wien and Juwelier SchönmannMultiselect Ultimate Query Plugin by InoPlugs Web Design Vienna | Webdesign Wien and Juwelier Schönmann