Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Advertisers
    • Editorial Board
  • Other Publications
    • Anticancer Research
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • In Vivo
    • Anticancer Research
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
In Vivo
  • Other Publications
    • In Vivo
    • Anticancer Research
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
In Vivo

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Advertisers
    • Editorial Board
  • Other Publications
    • Anticancer Research
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Visit iiar on Facebook
  • Follow us on Linkedin
Review ArticleR

Mouse Models of Inflammatory Bowel Disease - Insights into the Mechanisms of Inflammation-associated Colorectal Cancer

DESSISLAVA MLADENOVA and MAIJA R. J. KOHONEN-CORISH
In Vivo July 2012, 26 (4) 627-646;
DESSISLAVA MLADENOVA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MAIJA R. J. KOHONEN-CORISH
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: m.corish@garvan.org.au
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

The association between chronic inflammation and cancer has been noted for at least a century but the exact molecular mechanisms of cancer initiation and promotion by such inflammation are still poorly understood. The gastrointestinal tract is a unique organ where maintaining a balance between the colonic epithelial cells, the immune system and a fine-tuned response to the resident microflora is crucial for preserving the gut homeostasis. A breakdown of the tight interdependent regulation of the epithelium–immunity–microbiota triangle leads to inflammatory bowel disorders and may promote cancer. This review focuses on inflammation-associated colorectal cancer in mouse models of the disease and highlights emerging research trends.

  • Inflammatory bowel disease
  • mouse models inflammation
  • colorectal cancer
  • review

Inflammation-associated Colorectal Cancer

Inflammatory bowel disease (IBD) is the collective name for a group of gastrointestinal chronic inflammatory disorders with two major types of clinical presentations: ulcerative colitis (UC) and Crohn's disease (CD). IBD is an idiopathic disorder but it is now accepted that both environmental and genetic factors contribute to the pathogenesis of IBD. The molecular events leading to a breakdown in intestinal homeostasis and driving IBD pathogenesis include dysregulation of the innate and adaptive immune responses, loss of immune tolerance to the commensal microflora and a disruption of the intestinal epithelial integrity (1, 2). There is regional heterogeneity in the incidence of UC and CD worldwide and IBD disorders affect an estimated 2.4 million people in Europe alone and 1.3 million people in the USA (3). Colorectal cancer (CRC) is the cause of approximately 15% of all deaths in patients suffering from IBD (4).

UC affects the colon in a continuous fashion and always involves the rectum in adults, while CD can affect any part of the gastrointestinal tract, as intermittent lesions, but most commonly the terminal ileum or the perianal region (5, 6). Generally in UC the inflammation is superficial and restricted to the mucosa, whereas in CD the inflammation is often transmural and the mucosa appears thickened. In addition, granulomas, abscesses, fistulas and strictures are common features of CD. The disease location is relatively stable in patients with CD but the phenotype changes from non-stricturing and non-penetrating to either stricturing or penetrating over the course of the disease (7).

Patients with IBD have an increased risk of developing CRC, most of which is thought to be due to the persistent inflammatory response rather than a genetic predisposition (8). The risk of developing CRC increases with disease duration and extent, early onset, presence of primary sclerosing cholangitis (chronic inflammation of the bile ducts) and a family history of sporadic CRC (9-12). Although there is a clear association between chronic inflammation and cancer risk, the exact molecular mechanisms responsible for this increased risk are still poorly understood.

Molecular Alterations in Inflammation-associated Colorectal Cancer

In sporadic CRC the progression from normal epithelium to cancer involves the development of an adenomatous polyp (adenoma) precursor, which can be removed by endoscopic polypectomy. In contrast, in IBD-associated cancer, the precursor of neoplasia is usually a flat dysplastic lesion, and therefore its detection and removal is difficult (8). Inflammation-associated and sporadic CRC share similar molecular features: chromosomal instability (CIN), microsatellite instability (MSI) and CpG island methylator phenotype (CIMP), but their timing and frequency may differ. For example, in contrast to sporadic CRC, p53 inactivation is an early event in IBD-associated cancer, while mutations of v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) and adenomatous polyposis coli (APC) are infrequent and occur later (13-15). High-level MSI (MSI-H), resulting from deficient mismatch repair, is found in 15% of neoplastic lesions in IBD, which is similar to sporadic CRC. However, low-level MSI (MSI-L) may be more frequent in IBD-associated neoplastic and non-neoplastic tissues due to chronic inflammation (16-18).

Methylation silencing of tumour suppressor genes is an important feature of sporadic CRC. CIMP is characterized by concordant methylation of cancer-specific genes and long-range epigenetic silencing involves large genomic regions, such as the entire 4-Mb band of chromosome 2q.14.2 (19, 20). In addition to CpG islands and gene promoters, methylation is found at sequences up to 2 kb distant from CpG islands, termed CpG island shores (21). Methylation also occurs in IBD-related cancer and a high degree of age-related methylation has been found in the dysplastic and inflamed mucosa of patients with UC, indicating that methylation of CpG islands precedes dysplasia (22). Frequent and early promoter hypermethylation has been found in UC dysplasia and neoplasias for a number of genes including genes frequently altered in sporadic CRC such as p14ARF and p16INK4A. However, not all reports support an association between increased CpG island promoter methylation and IBD-related cancer (23-26).

Sporadic colorectal tumours arising in the proximal or distal colon may differ significantly in their molecular, clinical and histopathological characteristics, and the progression model (27, 28). CIN tumours are more frequently located in the distal colon and rectum, whereas, MSI tumours are predominantly located in the proximal colon and are associated with mucinous histology, poor differentiation and lymphocytic infiltration (29-31). CIMP tumours are characterized by a distinct set of features such as proximal colon location, v-raf murine sarcoma viral oncogene homolog B1 (BRAF) mutations and MSI-H (32-34). There is limited information about the tumour location prevalence of inflammation-associated CRC. One study examined the spatial distribution of CRC in 3,124 patients with CD and 3,093 patients with UC (35). The majority of carcinomas developed after 8 years of disease (CD 75%; UC 90%) in the areas of macroscopic disease. The anatomical distribution of cancer in the two diseases was significantly different. Tumours were located in the proximal colon in 49% of patients with CD and in 36% of patients with UC. Remarkably, the clinicopathological features of the tumours developing in CD and in UC were strikingly similar, including proportions of mucinous and signet ring carcinomas (35).

Mouse models of IBD have given some insights into the key factors and processes that contribute to colorectal tumour initiation and growth in the context of chronic inflammation. Here we review how the main mouse models of IBD and cancer (Table I) highlight the complex interactions between colonic epithelial cells, the immune system and the colonic microbiota, which are dysregulated in carcinogenesis.

The Role of the Colonic Epithelial Barrier in Maintaining Gut Health

Disruption of the epithelial barrier function can lead to impaired mucosal integrity, chronic inflammation and carcinogenesis, which has been exploited in many mouse models of IBD and cancer. In the healthy intestine there is a close spatial localization and a well-orchestrated crosstalk between cells of the mucosal epithelial barrier and cells of the mucosal innate and adaptive immune system. In addition, gut epithelial and dendritic cells (DCs) are in constant communication with the intestinal microbiota and more importantly, sensing of the commensal bacteria in normal conditions is essential for the maintenance of mucosal integrity and homeostasis (36). Because of this close spatio-functional relationship with the resident microbiota, the intestinal immune system functions in a tolerogenic state, which maintains tolerance towards the commensal microflora and harmless food components. For example, intestinal mucosal DCs preferentially induce differentiation of T-cells towards T-helper type 2 (Th2) and T-regulatory (Treg) subsets with tolerogenic properties (37, 38).

The colonic epithelial barrier consists of a monolayer of cells with intercellular tight junctions, and biochemical adaptations, which serves as an additional level of defence against the luminal microbiota. One of these adaptations is the glycosylated mucin-rich layer, forming a thick, impermeable sheet on the apical surface of intestinal epithelial cells (IECs). IECs also secrete a broad range of peptides with antimicrobial properties, such as defensins, cathelicidins and calprotectins (39, 40). Increasing evidence suggests that the epithelial layer not only serves as a passive physical barrier between the host and intestinal microflora, but is a dynamic, active participant in sensing external or endogenous ‘danger’ signals and effectively mounting immune responses (41).

IECs can recognise ‘danger’ signals. Microbial molecules, cytokines, and pro-inflammatory lipids signal infection and trigger an inflammatory response. An inflammatory response can also be initiated by tissue injury signals (heat-shock proteins, neuropeptides, mitochondrial peptides) (42). Stimulation of human colonic epithelial cells with pathogenic bacteria and cytokines results in up-regulation of a distinct array of pro-inflammatory cytokines such as tumour necrosis factor alpha (TNFα), interleukin-8 (IL-8), monocyte chemotactic protein-1 (MCP-1), and granulocyte macrophage colony-stimulating factor (GM-CSF) (43). Once the inflammatory response has been initiated, efficient resolution of inflammation is important in order to minimize tissue injury and restore the integrity of the mucosal barrier. IECs play an active role in the resolution phase of inflammation by secreting anti-inflammatory mediators that generally inhibit neutrophil function. For example, anti-inflammatory lipoxins, derived from IECs inhibit neutrophil migration and stimulate neutrophil apoptosis and macrophage-mediated scavenging of neutrophils (44). Importantly, lipoxins inhibit the production of IEC-derived IL-8, a potent neutrophil chemoattractant (45).

Deficiency in the core 3-derived O-glycans, a major component of the intestinal mucus, results in susceptibility to dextran sodium sulfate (DSS)-induced colitis and tumourigenesis in mice (46). Altered O-glycans are a feature of UC and are detected in more than 90% of UC-associated CRC (47, 48). Compromised integrity of the epithelial barrier allows direct contact of the intestinal mucosa with the resident colonic bacteria and plays a crucial role in the development of inflammation. Other defects that lead to colitis due to increased intestinal permeability include altered intercellular adhesion (N-cadherin transgenic mice), and deficiency in mucosal structural and protective proteins such as keratin-8 and MUC2 (49-52). MUC2 is the main secretory mucin in the mucus layer. Mice deficient in MUC2 exhibit spontaneous colitis, epithelial hyperproliferation, loss of crypt architecture and develop invasive adenocarcinomas in the small bowel, as well as in the rectum. Similarly, chemically-induced disruption of the colonic epithelium integrity with DSS leads to mucosal ulceration and dysplasia even in wild-type mice (53, 54).

Mice with Dysregulation of the Immunosuppressive IL-10 or Transforming Growth Factor beta (TGF-β) Pathways

Under normal conditions, IECs serve as communicators between the commensal bacteria and the host immune system and such cross-talk is essential for the maintenance of epithelial integrity and for the normal development and function of the host immune cells (2, 41). IECs secrete thymic stromal lymphopoietin (TSLP), TGF-β and IL-10, which modify the cytokines produced by DCs and macrophages, allowing the expansion and survival of Treg cells with immunosuppressive functions. In addition to TSLP, TGF-β and IL-10, other IEC-derived factors, secretory leukocyte peptidase inhibitor (SLPI), B-cell-activating factor (BAFF), a proliferation-inducing ligand (APRIL) and prostaglandin E2 (PGE2), regulate the function of lymphocytes in the intestine (41).

IL-10 is also produced by B- and T-cells, as well as myeloid cells. IL-10 is critical for Treg cell function (55). Treg cells can suppress T-cell-mediated and innate immune pathologies (55-58). Myeloid cell production of IL-10 is important for maintaining forkhead box P3 (Foxp3) expression on Treg cells, which mediates immunosuppressive functions in the colon (59). Polymorphisms found in the IL-10 receptor genes IL10RA and IL10RB are associated with severe early-onset IBD and polymorphisms in IL-10 itself are also associated with susceptibility to IBD (5,60).

Mice deficient in IL-10 develop spontaneous colitis and colitis-associated cancer, located predominantly in the proximal colon (61). The clinicopathological features of this model resemble CD and are driven by aberrant Th1 cytokine response, dependent on interferon-gamma (IFNγ) and IL-12 (61-63). The disease is exacerbated by Helicobacter hepaticus infection and treatment of these mice with IL-12p40 monoclonal antibody ameliorates inflammation (64). Similarly, depletion of local macrophages in the intestine reduces inflammation in Il10−/− mice (65). IL-10-secreting macrophages play an important role in immune tolerance and in clearance of apoptotic cells, whereas a deficiency in the latter function may result in exacerbation of inflammation and an increased susceptibility to autoimmune reactions (59, 66). Both epigenetic modifications and pathogenic microflora have been implicated in colitis-associated carcinogenesis in Il10−/− mice. Histone deacetylase (HDAC) inhibitors reduce tumour number and size, while treatment of mice with probiotics reduces both inflammation and carcinogenesis in Il10−/− mice (67-69). Infection of Il-10-deficient mice with pathogenic bacteria such as Helicobacter spp. increases tumour incidence (69). Paradoxically, the non-steroidal anti-inflammatory drugs (NSAIDs) celecoxib, rofecoxib and indomethacin exacerbate colitis and increase dysplasia in Il10−/− mice, suggesting that in the absence of the immunoregulatory functions of IL-10, NSAIDs contribute to further dysregulation of the immune response (70).

MUC1 is an epithelial cell surface-associated glycoprotein that exhibits altered glycosylation and is overexpressed in the majority of human adenocarcinomas and their precursor lesions (71). Mice transgenic for human MUC1 (MUC1-Tg) crossed with Il10−/− mice demonstrate exacerbation of the colon inflammation, accelerated tumour development compared to Il10−/− mice (8-12 weeks), a significant increase of tumours in the proximal colon and profound neutrophilic infiltration (72). These results suggest that aberrant expression of Muc1 in Il10−/− mice may promote tumourigenesis by a number of signalling pathways, such as interaction with β-catenin. No mutations of genes frequently involved in CRC have yet been identified in the carcinomas of IL-10-deficient mice (73).

The crucial role of TGF-β in modulating the immune response is now well-established. The main function of TGF-β is to maintain T-cell tolerance to self or harmless antigens through its regulatory effects on effector and regulatory T-cells (74). T-Cells from patients with IBD showed unresponsiveness to TGF-β through up-regulation of SMAD family member 7 (SMAD7), a negative regulator of TGF-β signalling (75). TGF-β receptor type II (TGFBR2) gene is mutated through mismatch repair deficiency in a small subset (6%) of UC-associated neoplasms (76). TGF-β1 deficiency in mice results in multiorgan inflammation and early death (77). When crossed with immunodeficient recombination activating gene 2 (Rag2)−/− mice, the double-knockout mice survive longer and develop inflammation-associated colonic hyperplasia and carcinoma. The model establishes the protective role of TGF-β1 in early cancer promotion in immunocompromised animals by maintaining epithelial tissue organization and suppressing the transformation from hyperplasia to dysplasia (78). Carcinogenesis in this model is dependent on infection with pathogens and the genetic background of the mouse (79). TGF-β signalling is mediated by the SMAD family of intracellular proteins, including SMAD2, SMAD3 and SMAD4 (80). Smad3ex2/ex2-deficient mice also develop chronic intestinal inflammation and frequent caecal and colonic adenocarcinomas (81). However, in another report, Smad3ex8/ex8-deficient mice showed infrequent colon adenomas at the age of 6 months (82). This discrepancy may be due to the different pathogen status of the animals in the two studies. Helicobacter-infected but not Helicobacter-free Smad3−/− mice have been found to develop colon cancer (83).

View this table:
  • View inline
  • View popup
Table I.

Mouse models of Inflammatory bowel disease (IBD)-associated colorectal cancer.

Mice with Defects in the Adaptive Immunity

Rag2−/− mice are characterised by a lack of mature B- and T-cells. When infected with H. hepaticus these mice develop colon inflammation, hyperplasia, dysplasia and carcinoma. Both inflammation and malignant changes can be reduced by transfer of Treg cells (characterized by CD4+CD45RBloCD25+ expression) but not by Treg cells from IL-10-deficient donors. This establishes the crucial role of IL-10 competent Treg cells in the restoration of epithelial integrity and prevention of pathogen-induced inflammation and cancer (84-86). In addition, neutralization of IL-6 reversed the invasive mucinous adenocarcinoma phenotype in this model. Mice receiving IL-10–Ig fusion protein exhibited down-regulation of IL-6 in response to H. hepaticus infection (86). Thus, the study establishes a model in which IL-10 suppresses pathogen-induced elevation of IL-6 and carcinogenesis.

Mice deficient in T-cell receptor β chain lack mature T-cells and develop colitis resembling UC. When crossed with p53-deficient mice, the double-knockout mice develop a high frequency of adenocarcinoma. Both the inflammatory response and cancer development are dependent on the intestinal microflora as these mice do not develop tumours in microbe-free conditions (87).

IL-2 is an important regulatory cytokine for T-cell growth and expansion and plays a central role in the differentiation of naive CD4+ T-cells into Th2 cells (88). Furthermore IL-2 is essential for Treg cell growth and for sustaining the Treg cell population in order to mediate immune homeostasis and tolerance to self (89). Young (3 to 4-week-old) IL-2-deficient mice have normal immune responses with no apparent intestinal or hepatic inflammation; however, older mice develop colitis mediated by CD4+ T-cells (90-92). The initiation of the disease is dependent on thymus-derived T-cells that infiltrate the colon and bone marrow, inducing colitis, lymphoadenopathy, splenomegaly, loss of B-cells and anemia (93). Strikingly, microbe-free Il2−/− mice do not develop colitis in contrast to specific pathogen-free (SPF) Il2−/− mice, while the extraintestinal pathology is independent of the microbial flora. However microbe-free Il2−/− mice exhibit abnormalities in the generation and maintenance of T-cell receptor gamma delta (TCRγδ+) subsets of intestinal intraepithelial lymphocytes (94), which are known to accumulate in the inflamed tissue in CD and UC (95). Despite the severe colitis, 6-month-old Il2−/− mice do not develop gastrointestinal cancer. However, mice deficient in both IL-2 and β2-microglobulin (a component of MHC class I molecules) develop colitis with faster onset compared to Il2−/− mice (96). These mice lack CD8+ T-cells and live longer due to less pronounced systemic disease. By 6 months of age they develop proximally located adenocarcinoma, characterized by mutations in the Apc and p53 genes, and MSI (97, 98). Interestingly, mutations in the β2-microglobulin gene have been detected in colorectal tumours and therefore these cells have reduced MHC class I expression and are not recognized by CD8+ T-cells. An increased density of CD8+ cytotoxic lymphocytes in colonic tumours is associated with a good prognosis, suggesting that an escape of immune recognition by cytotoxic CD8+ T-cells may be a mechanism for tumour progression in inflammation-associated CRC (99, 100).

Mice with Defects in Signal Transduction and Transcription Factors

G-Proteins are heterotrimeric guanine nucleotide binding proteins that regulate signal transduction through adenylyl cyclases, phospholipase C and ion channels (101). Mice deficient in the G-protein subunit αi2 have defects in T-cell maturation and function, such as defective chemotactic migration of thymic and colonic T-cells, and UC-like inflammation with mucinous adenocarcinoma predominantly located in the proximal colon (102). These tumours exhibit MSI and epigenetic silencing of the Mlh1 promoter that is mediated by the transcriptional repressor DEC-1 and is dependent on inflammatory hypoxia (103).

Suppressor of cytokine signalling-1 (SOCS1) is an intracellular protein that inhibits janus kinase (JAK)-mediated cytokine signalling. SOCS-1 deficient mice die shortly after birth but mice with restoration of SOCS1 specifically in T- and B-cells (SOCS1−/−Tg) survive for more than one year. These mice develop spontaneous colitis at 3 months of age and cancer by 6 months of age, characterized by IFNγ up-regulation and hyperactivation of signal transducer and activator of transcription 1 (STAT1) signalling. Constitutive activation of STAT3 and NF-κB signalling pathways were also observed in SOCS1−/−Tg mice, but their activity was not a contributing factor to tumourigenesis (104). In contrast, inactivation of IFNγ suppressed STAT1 hyperactivation and tumourigenesis in this model, implicating SOCS1 activity as a tumour suppressor and an important regulator of IFNγ signalling (104).

STAT3 inactivation specifically in haematopoietic cells (mainly in macrophages, with partial deletion in other myeloid and lymphoid cells) results in colitis and inflammation-associated colonic cancer in mice (105). Since STAT3 functions as a major mediator of IL-10 signalling and IL-10 has potent immune suppression functions, it is possible that chronic inflammation in this model is induced due to unregulated activation of both myeloid and lymphoid cells. Microbiota are required for both the development of inflammation and tumourigenesis in this model. The chronic inflammatory environment leads to disruption of the epithelial barrier and to epithelial hyperproliferation, associated with up-regulation of mammalian target of rapamycin (mTOR) and STAT3 activity in colonic epithelial cells. Furthermore, epithelial hyperproliferation was mTOR-dependent and aberrant upregulation of mTOR-STAT3 crosstalk was observed in the mucosa of patients with IBD (105).

In summary, immunoregulatory functions of T-cells mediated by IL-10 and TGF-β signalling contribute to mucosal homeostasis and prevent abnormal immune responses to the intestinal microflora. In addition, disruption of signal transduction pathways regulating T-cell maturation and function, as well as uncontrolled cytokine signalling, contribute to the onset of colitis. The sustained inflammatory response creates a landscape associated with an increase in pro-oncogenic cytokines, such as IL-6, and hypoxic conditions that may promote silencing of DNA repair genes and induce genetic instability (86, 103). The introduction of additional defects such as p53 and β2-microglobulin deficiency accelerate tumourigenesis in the context of defective adaptive immunity and chronic inflammation (87, 97, 98).

The Role of DCs in Inflammation and Inflammation-associated Colorectal Carcinogenesis

DCs act as ‘sentinels’ for the presence of pathogenic bacteria and play a key role in regulating immune homeostasis and tolerance in the colon. DCs can be seen as a regulatory hub that integrates different environmental signals and subsequently ‘issues’ appropriate directions to the adaptive immunity, thereby shaping the immune response. Therefore aberrant DC function may directly contribute to the pathogenesis of IBD and IBD-associated cancer (106). DCs extend dendrites into the colonic lumen to sample and evaluate commensal and pathogenic bacteria (107-109). DCs also receive signals from the IECs, which secrete factors such as TSLP and retinoic acid that ‘condition’ DCs to promote the development of Th2 cells and Treg cell subsets with tolerogenic functions (110, 111).

T-bet (encoded by the Tbx21 gene) is a T-box transcription factor family member that regulates type 1 inflammatory immune response in both adaptive and innate immunity (112). T-bet is required for the optimal production of IFNγ by DCs (113). Mice deficient in Rag2 and T-bet transcription factor [T-bet−/− x Rag2−/− UC (TRUC) mice] develop inflammation resembling UC and colonic adenocarcinoma (114, 115). Malignant changes in TRUC mice are driven by T-bet deficiency in DCs and exhibit molecular alterations that are characteristic of cancer in patients with UC, such as p53 loss of function, overexpression of cyclooxygenase 2 (Cox-2) and aberrant expression of β-catenin. Carcinogenesis is driven by dysregulated expression of TNFα and is dependent on commensal bacteria. Targeted re-expression of T-bet in DCs ameliorated colitis and carcinogenesis and reduced the levels of TNFα, suggesting that T-bet may act as a repressor of TNFα in DCs. The severity of the colitis, as well as dysplasia and adenocarcinoma, in the TRUC model is dependent on the murine strain background and is controlled by the cytokine deficiency-induced colitis susceptibility-1 (Cdcs1) locus on chromosome 3 through the innate immune cells (116). This highlights the importance of T-bet dysfunction in DCs and strain-specific genetic modifiers in the promotion of inflammation-associated cancer.

Mouse Models with Chemically-induced Colitis and Cancer

DSS model. Chronic inflammation resembling UC can be induced by oral administration of DSS, which triggers inflammation by damaging the gut-epithelial barrier (53, 117). In the DSS model, inflammation is accompanied by generation of reactive oxygen species (ROS) and a decrease in the antioxidant defense of the inflamed mucosa (118, 119) similar to IBD (120, 121). Repeated cycles of DSS are required to induce carcinogenesis in a subset of mice. However, genetic defects, such as mutations in key tumour suppressor genes, accelerate tumour promotion in the context of DSS-induced inflammation. For example, 60% of mice deficient for the DNA mismatch repair gene Msh2 developed colonic dysplasia or adenocarcinoma with the DSS treatment, compared to 29% of their wild type siblings. Untreated Msh2−/− mice develop spontaneous tumours in the small intestine but not in the colon (122). Thus, defects in the DNA mismatch repair genes accelerate tumourigenesis in the context of chronic inflammation and can shift the gastrointestinal location of tumour formation. Although repeated cycles of DSS induce proximal and distal colon inflammation, the inflammatory response is more severe in the distal colon and this is associated with the majority of cases of dysplasia and cancer (123). p53 deficiency also increases the number of flat cancer lesions in the DSS model (124).

Azoxymethane (AOM)/DSS model of colitis-associated cancer. Inflammation-associated cancer induced by DSS in mice requires prolonged time and exposure to repeated cycles of DSS. Injection of the carcinogen AOM prior to exposure to DSS reduces the time required for cancer development (125). The AOM/DSS model of inflammation-associated cancer has been widely utilized to study the molecular factors that trigger malignant transformation and promote tumour growth. This model is frequently referred to as colitis-associated cancer model (CAC).

Insights into the Mechanism of Carcinogenesis from the CAC Model

Pro-tumorigeneic role of NF-κB. NF-κB, a family of transcription factors, is a master regulator of gene expression with key roles in orchestrating pro- and anti-inflammatory responses, as well as cell survival and differentiation programs. Increase in NF-κB activity has been found in human colorectal cancer samples and in colon cancer cell lines (126, 127). In patients with Crohn's disease, there is an increase in NF-κB in epithelial cells as well as in infiltrating macrophages (128), indicating that NF-κB activity is involved in colon inflammatory pathogenesis. Similarly, NF-κB activity is up-regulated in the Il10−/− mouse model of chronic colon inflammation, and administration of anti-sense oligonucleotide to the RelA subunit of NF-κB alleviates the symptoms of the disease (129). NF-κB target genes include cytokines and chemokines, which participate in tumour promotion in the context of chronic inflammation. NF-κB is involved in the up-regulation of genes that are associated with tumourigenesis such as TNFα, IL-1β and IL-8 (15). NF-κB also controls the expression of anti-apoptotic genes such as B-cell lymphoma 2 (Bcl-2) and B-cell lymphoma-extra large (Bcl-xL) (130). One of the most convincing pieces of evidence that NF-κB activity is implicated in cancer comes from studies of the CAC model. Enterocyte-specific ablation of IKKβ, an essential kinase in NF-κB signalling, resulted in a decrease in colitis-associated tumour incidence (131). In this context, epithelial NF-κB functions as a pro-tumourigenic factor due to induction of anti-apoptotic genes such as Bcl-xL.

NF-κB signalling may have different or even opposing roles depending on the tissue context or grade of inflammation. NF-κB loss in epithelial cells during acute DSS-induced inflammation hinders mucosal healing and prevents recruitment of inflammatory cells that produce cytoprotective factors such as IL-11 and IL-22. In contrast, inactivation of epithelial NF-κB signalling has no effect in the Il10−/− mouse model of chronic inflammation but NF-κB ablation in myeloid cells ameliorates inflammation in the same model (132). NF-κB signalling promotes cancer in the AOM/DSS model but through different mechanisms in the colonic epithelial and myeloid cell components. Conditional ablation of IKKβ in enterocytes reduces tumour incidence by 80% in this model, possibly by increasing apoptosis of premalignant cells, whereas deletion of IKKβ in myeloid cells (macrophages, neutrophils, DCs) results in a significant reduction of tumour size. These elegant studies demonstrate that NF-κB signalling in myeloid cells results in up-regulation of pro-inflammatory factors that function as tumour growth factors (131). One of the cytokines implicated in this process is IL-6, and the IL-6–STAT3 signalling axis has been shown to regulate proliferation and survival of tumour initiating IECs (133).

IL-6, IL-21 and TNFα are major tumour-promoting cytokines. IL-6 was identified as a critical NF-κB-dependent cytokine that is produced by myeloid cells of the lamina propria (mainly DCs and macrophages). IL-6 stimulates survival and proliferation of IECs through STAT3 activation downstream of the gp130 receptor and promoted tumourigenesis in the AOM/DSS model (133). This study identified the importance of trans-signalling cross-talk between the immune compartment and the IECs in response to acute colitis and the critical role of myeloid-produced factors in tumour promotion. IL-6 and pSTAT3 overexpression have been detected in the epithelial cells of patients with active UC (134). Similarly, mice with IEC-specific deficiency in SOCS3, a negative regulator of receptor mediated activation of STAT3, exhibit increased tumourigenesis in the AOM/DSS model (135). IL-6 also plays an important role in intestinal homeostasis and epithelial regeneration, and can activate three signalling cascades: SH2-containing phosphotyrosine phosphatase (SHP2)–Ras–extracellular signal-regulated kinase (ERK), JAK1/2–STAT3 and phosphoinositide-3-kinase (PI3K)–v-akt murine thymoma viral oncogene homolog 1 (AKT)–mTOR (136-138). However, IL-6 can further propagate inflammatory signals through the regulation of pathogenic T-cells. IL-6 inhibits the generation of the anti-inflammatory Foxp3+ Treg cells and instead, in concert with TGF-β, promotes generation of pathogenic Th17 from naïve T-cells (139). Recently it has been demonstrated that in addition to driving gp130-mediated STAT3 activation, IL-6 drives the overexpression of vascular endothelial growth factor receptor 2 (VEGFR2) in IECs in the CAC model, thereby enhancing STAT3 activation through VEGF signalling, which further promotes epithelial cell proliferation and tumour growth (140).

IL-21 is expressed by CD4+ cells, including Th1 and Th17 cells, and has been shown to promote Th17 responses (141). IL-21 is overexpressed in the mucosa and tumours of patients with UC (142). IL-21 deficiency in AOM/DSS-treated mice results in a reduction in tumour load, paralleled by a decrease in IL-6 and IL-17A expression (142). IL-21 is a negative regulator of CD4+Foxp3+ Treg cells and these cells are increased in the colon of IL-21-deficient mice, providing yet another affirmation of the cancer suppressor role of Treg cells. As IL-21 is produced by CD4+ T-cells and IL-21R is broadly expressed by T- and myeloid cells, it is likely that IL-21 promotes tumour growth through paracrine/autocrine mechanisms inducing the production of the pro-carcinogenic IL-6 (predominantly produced by myeloid cells) and IL-17A (produced by T-cells).

Although TNFα was initially associated with tumour necrosis, mounting evidence suggested that TNFα plays a role in tumour growth and progression (143). TNFα signalling contributes to mutagenic ROS generation, and induction of metalloproteases and genes involved in inflammation, tissue repair and angiogenesis as well as recruitment of activated leukocytes to the site of inflammation (144-148). Anti-TNFα therapy is successful in a subset of patients with IBD (2). Suppression of TNFα signalling in the mouse model of CAC reduces tumourigenesis, inflammation and infiltration of macrophages and neutrophils (149). Similarly, TNFα neutralization prevents cancer formation in other mouse models of inflammation-associated cancer.

Dysregulation of pattern recognition receptor signalling promotes tumourigenesis. Pattern recognition receptors (PRRs) sense microbial agents and play a crucial role in the mucosal homeostasis. Aberrant activation of PRRs is a major contributor to IBD in humans and in mouse models. The innate immune system has several classes of PRRs. Toll-like receptors (TLRs) sense microbes on the cell surface and endosomes, while nucleotide-binding oligomerization domain containing (NOD)-like receptors (NLRs), such as NOD1 and NOD2, and retinoid-inducible gene 1 (RIG-1)-like receptors (RLRs) recognize cytosolic microbial components.

Signalling through these receptors triggers several downstream kinases, some of which are involved in up-regulation of cytokines and chemokines (150,151). TLR signalling activates NF-κB and AP-1 transcription factors through the TLR adaptor MyD88. However, mice deficient in MyD88 exhibit susceptibility to DSS-induced colitis and an increased mortality due to defects in epithelial regeneration and tissue repair (36). MyD88-deficient mice also demonstrate dramatically increased tumourigenesis in the CAC model (152). Remarkably, these mice do not demonstrate increased epithelial cell proliferation observed in other models of inflammation-associated cancer, but instead, have alterations in the inflammatory microenvironment associated with an increased expression in wound healing factors such as IL-6, IL-11, EGFR ligands, COX-2 and hypoxia-inducible factor 1 alpha (HIF-1α) (152). These mice also express phosphorylated STAT3 in the nuclei of IECs, suggesting activation of the pro-tumorigenic IL-6/IL-11–STAT3 cascade (136).

Dysregulated epithelial cell–microbiota interaction through uncontrolled TLR signalling can also contribute to colitis. Mice deficient in the single immunoglobulin IL-1 receptor-related molecule (SIGIRR), a negative regulator for Toll–IL-1R signalling, exhibit commensal bacteria-dependent defects in epithelial cell homeostasis, constitutive expression of pro-inflammatory mediators, hyperactivation of NF-κB and STAT3, susceptibility to DSS-induced colitis and increased AOM/DSS-induced tumourigenesis (153).

TLR4 is overexpressed in the malignant tissues of patients with UC as well as in AOM/DSS-induced cancer in mice (154). Unlike MyD88−/− mice, TLR4-deficient mice are protected from AOM/DSS-induced tumourigenesis. The acute phase of inflammation is ameliorated in TLR4-deficient mice and this is associated with a decrease in NF-κB signalling, as well as a down-regulation of the pro-tumorigenic COX2/PGE2. PGE2, produced downstream of TLR4 activation, can act in a paracrine or autocrine manner to stimulate the expression and secretion of the epidermal growth factor receptor (EGFR) ligand amphiregulin. Amphiregulin activation of EGFR results in an increased proliferation of colonocytes that may contribute to oncogenic transformation (154). PGE2 deficiency confers significant protection against AOM-induced colonic tumours (155). Remarkably, loss of PGE2 reduces the expansion of CD4+Foxp3+ Treg cells with immunosuppressive functions in the colon draining mesenteric lymph nodes in mice, unveiling a potential mechanism of how loss of PGE2 modifies the tumour inflammatory microenvironment and promotes antitumour immunity (155).

The inflammasomes and IL-18 activity suppress CAC. A set of NLRs result in the activation of caspase-1 through an assembly of large protein complexes termed inflammasomes (156). Caspase-1 activity has a critical function in the secretion and maturation of IL-1β and IL-18 (157, 158). Inflammasomes are categorized into subgroups according to the major components NLRP1, NLRP3, NLRC4, NLRP6 and absent in melanoma 2 (AIM2) (159). The role of inflammasomes and NLR in inflammatory disease has currently taken central stage (2). Genome-wide association studies (GWAS) have identified IBD-associated polymorphisms on NLRP3 and IL18RAP (160, 161). Levels of IL-18 and IL-1β are increased in patients with IBD (162).

Oncosuppressive functions have been attributed to inflammasomes. Inflammasomes can eliminate malignant cells through induction of apoptosis (159). For example, Nlrc4−/− and Casp1−/−-deficient mice exhibit resistance to apoptosis and increased epithelial cell proliferation in response to injury (163). Similarly, deficiency of NLRP6 in mouse IECs results in reduction of IL-18 levels, accompanied by quantitative and qualitative changes in numerous bacterial taxa (dysbiosis). The dysbiosis in NLRP6 inflammasome-deficient mice mediates transmissible autoinflammation, spontaneous hyperplasia and exacerbation of DSS-induced colitis (164).

An antitumour function has been assigned to NLRP3. Mice deficient in Casp1 and Nlrp3 exhibit increased susceptibility to DSS-induced colitis and reduced tumour load due to down-regulation of IL-18 expression. The role of IL-18, in this context, appears paradoxical because IL-18 is important for maintenance of the epithelial integrity and proliferation in response to injury in the acute phase of inflammation. However, in the chronic inflammatory environment, IL-18 acts as a tumour suppressor by inhibiting epithelial cell proliferation, a function that may be mediated in part by IFNγ antitumor activities through STAT1-dependent signalling (165, 166). Furthermore, IL-18 promotes IFNγ production in activated T- and natural killer (NK) cells, thus promoting Th1 cell polarization that may contribute to its antitumourigenic role (167, 168). In contrast to NLRC4, which exerts its antitumorigenic activity through its effects on IECs, NLRP3 suppresses tumourigenesis through its activity in the haematopoietic/myeloid compartment (169). Deficiency in another PRR involved in maintaining the epithelial integrity, NOD1, results in an increased susceptibility to DSS-induced acute injury associated with cytokine production and an increase in epithelial cell proliferation. Nod1−/− mice exhibit increased colitis-associated tumourigenesis where tumour development is dependent on the presence of intestinal microbiota (170).

Is HIF an Unappreciated Target of Inflammation-associated CRC?

The gastrointestinal tract functions in a state of low-grade inflammation and in rapid, drastic changes in tissue oxygen availability (171). Hypoxia-inducible factor (HIF) plays a central role in maintaining the epithelial barrier and in promoting cell survival under hypoxic conditions [reviewed in (171)]. HIF is rapidly degraded under normoxia and is stabilized in hypoxia. HIF is a heterodimeric transcription factor and consists of a constitutively expressed HIF-1β subunit and a highly regulated HIF-1α, or the related HIF-2α and HIF-3α subunits (172). HIF regulates the expression of genes mainly involved in adaptation to hypoxic microenvironments, such as genes coding enzymes from the anaerobic glycolysis pathway and genes implicated in angiogenesis. HIF-1 expression has been documented in cells of the innate and adaptive immune system, as immune cell function necessitates rapid adaptation to varying oxygen tissue tension. Highly proliferating cells, such as cells of the immune system, use glycolysis as their primary energy production pathway (173-176). As most of the enzymes of the glycolytic pathway are target genes of HIF-1, HIF-1 activity is essential for the function of the immune system.

HIF-1α appears to have different functions and importance in different cell types of the immune system. HIF-1α protects from activation-induced cell death in peripheral T-cells under hypoxic conditions (177). However, HIF-1 activation in thymocytes leads to caspase-8-mediated apoptosis (178). HIF-1α deficiency is embryonically lethal but was bypassed by the generation of Hif1α−/− → Rag2−/− chimeric mice. In these mice, HIF-1α deficiency in B- and T-cells leads to an autoimmune reaction and abnormal B-lymphocyte development (179). HIF-1α in myeloid cells is essential for their invasiveness, motility, aggregation and bacterial killing (176). A range of cytokines with pro-tumourigenic role can affect HIF-1 regulation. Some of them include TGF-β1, TNFα, IL-1β (180, 181). Conversely, HIF-1α can also modulate the expression of cytokines, such as IL-1, IL-4, IL-6 and IL-12 in macrophages (182). In addition, there is substantial evidence for a bi-directional relationship between HIF-1 and NF-κB, each enhancing the activity of the other (183,184).

Apart from inflammatory cells, HIF is also expressed in epithelial cells. HIF overexpression in colonic epithelial cells increased inflammatory infiltration and induced colonic oedema under normal conditions and resulted in exacerbation of DSS-induced colitis (185). Therefore, aberrant HIF-1 activation in epithelial cells may disturb the precise regulation of the inflammatory response and result in exacerbation of pathological conditions. Different strategies to inhibit HIF-1 signalling have been proposed for the treatment of inflammatory disorders (186). On the other hand, an increase in HIF-1 through inhibition or deficiency of prolyl hydrohylases (PHDs), the main hydroxylases promoting HIF-1 degradation, has been found to be protective in murine models of colitis (187-189). Therefore HIF-1-related strategies have been proposed for the treatment of chronic intestinal disorders. These strategies aim to stabilize HIF-1 expression and HIF-mediated maintenance of epithelial barrier function and induction of barrier protective factors (186, 190-192). However, PHD inhibitors are not specific to HIF-1 protein level stabilization. PHD inhibition also leads to an increase in NF-κB activity (193). Therefore, it is unknown whether the protective effect of PHD can be attributed mainly to increased levels of HIF-1 or to an increase in NF-κB activity.

HIF-1α overexpression has been observed in numerous types of cancer, including colon, breast, gastric, lung, skin, ovarian, pancreatic, prostate and renal tumours (194). However HIF-1 has not been well studied in relation to colon inflammation-induced oncogenesis. Recently, overexpression of microRNA-31 (miR-31) was found in IBD-related neoplasia. miR-31 increases HIF-1α expression through targeting its inhibitor factor, inhibiting HIF-1 (FIH-1). In this study miR-31 expression was a unique feature of IBD-associated cancer and was not found in sporadic CRC. Furthermore, miR-31 increased significantly during the transformation from normal epithelium to dysplasia and from dysplasia to cancer (195). Thus, this study suggests a role for HIF-1 in the malignant transformation in the context of IBD.

Polymorphisms in genes involved in the Th17 signalling pathway are associated with IBD (2, 196). Th17 cells are key players in the anti-inflammatory response to pathogens in the intestine, however, Th17 signalling can also drive autoimmune disorders, IBD, and inflammation-associated colon cancer (197, 198). HIF-1α was recently found to play a pivotal role in the development of Th17 cells and in the regulation of the Th17:Treg cell balance (199). Thus HIF-1 expression may participate in a vicious circle of propagating inflammation through promoting Th17 differentiation and IL-17 signalling, which in turn can promote sustained HIF-1 action (199).

HIF-1α overexpression in IECs was found in a mouse model of NSAID-induced inflammation-associated colon cancer (200). Although the tumour-promoting role of HIF-1α in this model requires further study, IEC-specific ablation of HIF-1α ameliorated the inflammatory response induced by the NSAID sulindac. In this model, oral administration of sulindac induced inflammatory lesions with serrated neoplasia which were most pronounced on the mucosal folds of the proximal colon. The lesions displayed mild to moderate acute and chronic inflammation, progressing to serrated neoplasia and mucinous adenocarcinoma in genetically-modified mice. In an experiment comparing Msh2 and p53-deficient mice with their wild-type siblings, adenocarcinoma was observed in up to 25% of the knockout mice on the C57Bl6J background. The wild-type siblings also developed inflammatory lesions on the sulindac diet but adenocarcinoma was rare. Sulindac-treated mice in this experiment had only a few lesions intercepted by macroscopically normal appearing mucosa, and few or no external symptoms of IBD. Microscopic analysis revealed areas of early surface erosion and rare ulceration in the non-neoplastic mucosa, suggesting that damage to the mucosal barrier may play a role in carcinogenesis in this model (200).

Both malignant and premalignant lesions were characterized by a marked overexpression of the pro-tumourigenic factors MIP-2 (the murine IL-8 homologue), IL-1β and COX-2, as well as HIF-1α (200). IL-8 plays a key role in cancer by initiation of tumour-associated inflammation, angiogenesis, proliferation and survival of endothelial and cancer cells (201, 202). Remarkably, in the distal colon of mice in the same model (200), sulindac prevented AOM-induced tumours, consistent with its role as a chemopreventive agent. This model may be useful for the study of proximal colon carcinogenesis and the serrated neoplasia pathway, which is characterized by proximal location and HIF-1α expression (203). Further analysis is required to determine how the sulindac model (200) compares with other mouse models of inflammation-associated colon cancer discussed in this review.

The Role of the Microbiota in Inflammation-associated CRC

While the relationship of microbiota with colitis and inflammation-associated CRC is one of the most rapidly expanding fields, it is not the focus of this review, however, a few points will be made here.

One of the most interesting recent advances in our understanding of the gut-colonizing bacteria is that their functions extend far beyond aiding digestion. The intestinal bacteria actively shape and influence not only the gut epithelial and immune homeostasis but also extraintestinal systems such as the cardiovascular and nervous systems (204-206). A ‘Western-style’ diet alters the microbiotic composition and induces adiposity in humanized gnotobiotic mice (adult human fecal microbial communities transplanted into microbe-free mice) and this trait is transmissible to non-obese mice through microbiotic transplantation (207). Diet is not the only factor that controls bacterial populations. Defects in innate immunity have a marked influence on microbiotic composition and the shift of microbial communities can induce communicable (transmissible) pathologies, such as colitis and metabolic syndrome in mice (208, 209). A UC twin study recently found a distinct microbial species composition and decreased diversity in the mucosa of patients with UC compared to their twin healthy siblings. The presence of certain bacterial genera correlated with host mucosal gene expression, indicating the interdependency of microbiotic metabolism and the host transcriptome (210).

How do microbiota contribute to inflammation-associated cancer? As discussed in this review, a large proportion of mouse IBD-related carcinomas require interaction with commensal or pathogenic bacterial species, suggesting that colitis is required but is not sufficient to induce cancer in some models (79, 197, 211, 212). The main mechanisms of bacterial-induced carcinogenesis are either through microbiota-mediated uncontrolled pro-inflammatory signalling or through exerting cytotoxic effects on mucosal cells (direct or mediated through microbial metabolites/activated host cells) (204). For example, the best known cancer-causing pathogen H. pylori has been proposed to induce carcinogenesis through persistent T-cell-mediated immune response and production of DNA-damaging ROS (204, 213-215). Other bacterial species have different mechanisms of promoting CRC (15). Mice deficient in the antioxidant enzymes glutathione peroxidase 1 and 2 develop microflora-associated intestinal inflammation and tumourigenesis, establishing the crucial role of antioxidant defense against bacteria-induced oxidative stress and cancer (212).

The microbiotic composition has a spatial variation along the axis of the large intestine (216). Despite the fact that the intestinal epithelial barrier is separated from the microflora by a thick mucus layer, bacteria interact directly with the intestinal crypts and significantly more crypts of the proximal colon are in contact with bacteria compared to crypts in the middle and distal colon (216). In Smad3−/− mice, Helicobacter bacteria preferentially colonized the caecum, and to a less extent, the proximal colon compared to the distal colon, reflecting the predominant site of tumourigenesis in these mice (83).

Perspective and Conclusion

Based on animal models, the traditionally proposed mechanisms of cancer induction by chronic inflammation include tissue damage and subsequent regenerative hyperproliferation of epithelial cells. The chronic inflammatory microenvironment is rich in macrophages that participate in the generation of ROS (217). ROS are mutagenic agents and combined with increased cellular proliferation at the site of inflammation, ROS can trigger oncogenic transformation (217, 218). As the inflammatory microenvironment is rich in both mutagenic ROS and secreted cytokines and growth factors, which can stimulate cell proliferation, cells with DNA damage may be allowed to go through the cell cycle before efficient DNA repair has occurred. Thus, potentially oncogenic mutations can be transmitted to progeny cells. In chemically-induced liver carcinogenesis, for example, compensatory proliferation causes initiated hepatocytes to enter the cell cycle and pass mutations onto the daughter cells (219, 220). However, mutations in classic tumour suppressor genes are not always found in inflammation-associated colon cancer in the mouse. For example, in the IL-10-deficient mouse model of colitis-associated cancer, no mutations in p53, K-ras and Apc have been found yet, indicating that IBD-related cancer may exploit additional/alternative oncogenic pathways or epigenetic mechanisms (15, 62, 73).

The increased rate of epithelial proliferation in response to mucosal barrier damage may accelerate the normal age-related epigenetic changes occurring in the colon mucosa. This in turn may leads to silencing of key tumour suppressor or DNA damage repair genes (67, 103). One of the key features of most mouse models of inflammation-associated CRC is that they require prolonged exposure to the inflammatory microenvironment, the introduction of additional an genetic defects ‘precipitates’ cancer initiation. These additional defects can be caused by treatment with carcinogens or by using mice with tumour suppressor gene mutations. Therefore chronic inflammation by itself may not be sufficient to induce oncogenic transformation. Even in the same model, some mouse strains are more susceptible to inflammation-associated cancers than others, implying that genetic variation also plays a key role. Another important point is that the inflammatory response affects the whole colon in many mouse models of colitis, however, cancer develops preferentially in the caecum or the proximal and distal ends of the colon. As discussed above, this may be due to regional differences in bacterial colonisation or perhaps intrinsic molecular differences of colonocytes or specialised mucosal immunity in different colonic segments. A pitfall of the introduced animal models is that they rarely involve the ileum, which is usually affected in CD (197). It can be speculated that disturbances in mucosal homeostasis regardless of the initiating factor lead to dysregulated, sustained and hyperactivated host inflammatory response to microbiota and drive carcinogenesis. Alternatively it can be proposed that the prolonged host immune dysregulation re-structures microbial communities in the gut, allowing the thriving of pathogenic species, which promote carcinogenesis.

  • Received May 25, 2012.
  • Revision received June 12, 2012.
  • Accepted June 12, 2012.
  • Copyright © 2012 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved

References

  1. ↵
    1. Kaser A,
    2. Zeissig S,
    3. Blumberg RS
    : Inflammatory bowel disease. Annu Rev Immunol 28: 573-621, 2010.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Maloy KJ,
    2. Powrie F
    : Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature 474(7351): 298-306, 2011.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Cosnes J,
    2. Gower-Rousseau C,
    3. Seksik P,
    4. Cortot A
    : Epidemiology and natural history of inflammatory bowel diseases. Gastroenterology 140(6): 1785-1794, 2011.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Munkholm P
    : Review article: the incidence and prevalence of colorectal cancer in inflammatory bowel disease. Aliment Pharmacol Ther 18(Suppl 2): 1-5, 2003.
    OpenUrlPubMed
  5. ↵
    1. Khor B,
    2. Gardet A,
    3. Xavier RJ
    : Genetics and pathogenesis of inflammatory bowel disease. Nature 474(7351): 307-317, 2011.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Baumgart DC,
    2. Sandborn WJ
    : Inflammatory bowel disease: clinical aspects and established and evolving therapies. Lancet 369(9573): 1641-1657, 2007.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Louis E,
    2. Collard A,
    3. Oger AF,
    4. Degroote E,
    5. Aboul Nasr El Yafi FA,
    6. Belaiche J
    : Behaviour of Crohn's disease according to the Vienna classification: changing pattern over the course of the disease. Gut 49(6): 777-782, 2001.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    1. Ullman TA,
    2. Itzkowitz SH
    : Intestinal inflammation and cancer. Gastroenterology 140(6): 1807-1816, 2011.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Ekbom A,
    2. Helmick C,
    3. Zack M,
    4. Adami HO
    : Increased risk of large-bowel cancer in Crohn's disease with colonic involvement. Lancet 336(8711): 357-359, 1990.
    OpenUrlCrossRefPubMed
    1. Nuako KW,
    2. Ahlquist DA,
    3. Mahoney DW,
    4. Schaid DJ,
    5. Siems DM,
    6. Lindor NM
    : Familial predisposition for colorectal cancer in chronic ulcerative colitis: a case–control study. Gastroenterology 115(5): 1079-1083, 1998.
    OpenUrlPubMed
    1. Askling J,
    2. Dickman PW,
    3. Karlen P,
    4. Brostrom O,
    5. Lapidus A,
    6. Lofberg R,
    7. Ekbom A
    : Colorectal cancer rates among first-degree relatives of patients with inflammatory bowel disease: a population-based cohort study. Lancet 357(9252): 262-266, 2001.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Kornfeld D,
    2. Ekbom A,
    3. Ihre T
    : Is there an excess risk for colorectal cancer in patients with ulcerative colitis and concomitant primary sclerosing cholangitis? A population based study. Gut 41(4): 522-525, 1997.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Hussain SP,
    2. Amstad P,
    3. Raja K,
    4. Ambs S,
    5. Nagashima M,
    6. Bennett WP,
    7. Shields PG,
    8. Ham AJ,
    9. Swenberg JA,
    10. Marrogi AJ,
    11. Harris CC
    : Increased p53 mutation load in noncancerous colon tissue from ulcerative colitis: a cancer-prone chronic inflammatory disease. Cancer Res 60(13): 3333-3337, 2000.
    OpenUrlAbstract/FREE Full Text
    1. Lyda MH,
    2. Noffsinger A,
    3. Belli J,
    4. Fenoglio-Preiser CM
    : Microsatellite instability and K-ras mutations in patients with ulcerative colitis. Hum Pathol 31(6): 665-671, 2000.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Terzic J,
    2. Grivennikov S,
    3. Karin E,
    4. Karin M
    : Inflammation and colon cancer. Gastroenterology 138(6): 2101-2114 e2105, 2010.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Boland CR,
    2. Goel A
    : Microsatellite instability in colorectal cancer. Gastroenterology 138(6): 2073-2087 e2073, 2010.
    OpenUrlCrossRefPubMed
    1. Cawkwell L,
    2. Sutherland F,
    3. Murgatroyd H,
    4. Jarvis P,
    5. Gray S,
    6. Cross D,
    7. Shepherd N,
    8. Day D,
    9. Quirke P
    : Defective hMSH2/hMLH1 protein expression is seen infrequently in ulcerative colitis-associated colorectal cancers. Gut 46(3): 367-369, 2000.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Schulmann K,
    2. Mori Y,
    3. Croog V,
    4. Yin J,
    5. Olaru A,
    6. Sterian A,
    7. Sato F,
    8. Wang S,
    9. Xu Y,
    10. Deacu E,
    11. Berki AT,
    12. Hamilton JP,
    13. Kan T,
    14. Abraham JM,
    15. Schmiegel W,
    16. Harpaz N,
    17. Meltzer SJ
    : Molecular phenotype of inflammatory bowel disease-associated neoplasms with microsatellite instability. Gastroenterology 129(1): 74-85, 2005.
    OpenUrlCrossRefPubMed
  15. ↵
    1. Frigola J,
    2. Song J,
    3. Stirzaker C,
    4. Hinshelwood RA,
    5. Peinado MA,
    6. Clark SJ
    : Epigenetic remodeling in colorectal cancer results in coordinate gene suppression across an entire chromosome band. Nat Genet 38(5): 540-549, 2006.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Lao VV,
    2. Grady WM
    : Epigenetics and colorectal cancer. Nat Rev Gastroenterol Hepatol 8(12): 686-700, 2011.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Irizarry RA,
    2. Ladd-Acosta C,
    3. Wen B,
    4. Wu Z,
    5. Montano C,
    6. Onyango P,
    7. Cui H,
    8. Gabo K,
    9. Rongione M,
    10. Webster M,
    11. Ji H,
    12. Potash JB,
    13. Sabunciyan S,
    14. Feinberg AP
    : The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet 41(2): 178-186, 2009.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Issa JP,
    2. Ahuja N,
    3. Toyota M,
    4. Bronner MP,
    5. Brentnall TA
    : Accelerated age-related CpG island methylation in ulcerative colitis. Cancer Res 61(9): 3573-3577, 2001.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Fleisher AS,
    2. Esteller M,
    3. Harpaz N,
    4. Leytin A,
    5. Rashid A,
    6. Xu Y,
    7. Liang J,
    8. Stine OC,
    9. Yin J,
    10. Zou TT,
    11. Abraham JM,
    12. Kong D,
    13. Wilson KT,
    14. James SP,
    15. Herman JG,
    16. Meltzer SJ
    : Microsatellite instability in inflammatory bowel disease-associated neoplastic lesions is associated with hypermethylation and diminished expression of the DNA mismatch repair gene, hMLH1. Cancer Res 60(17): 4864-4868, 2000.
    OpenUrlAbstract/FREE Full Text
    1. Hsieh CJ,
    2. Klump B,
    3. Holzmann K,
    4. Borchard F,
    5. Gregor M,
    6. Porschen R
    : Hypermethylation of the p16INK4a promoter in colectomy specimens of patients with long-standing and extensive ulcerative colitis. Cancer Res 58(17): 3942-3945, 1998.
    OpenUrlAbstract/FREE Full Text
    1. Moriyama T,
    2. Matsumoto T,
    3. Nakamura S,
    4. Jo Y,
    5. Mibu R,
    6. Yao T,
    7. Iida M
    : Hypermethylation of p14 (ARF) may be predictive of colitic cancer in patients with ulcerative colitis. Dis Colon Rectum 50(9): 1384-1392, 2007.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Konishi K,
    2. Shen L,
    3. Wang S,
    4. Meltzer SJ,
    5. Harpaz N,
    6. Issa JP
    : Rare CpG island methylator phenotype in ulcerative colitis-associated neoplasias. Gastroenterology 132(4): 1254-1260, 2007.
    OpenUrlCrossRefPubMed
  21. ↵
    1. Pocard M,
    2. Salmon RJ,
    3. Muleris M,
    4. Remvikos Y,
    5. Bara J,
    6. Dutrillaux B,
    7. Poupon MF
    : Two colons–two cancers? Proximal or distal adenocarcinoma: arguments for a different carcinogenesis. Bull Cancer 82(1): 10-21, 1995 (in French).
    OpenUrlPubMed
  22. ↵
    1. Distler P,
    2. Holt PR
    : Are right- and left-sided colon neoplasms distinct tumors? Dig Dis 15(4-5): 302-311, 1997.
    OpenUrlCrossRefPubMed
  23. ↵
    1. Delattre O,
    2. Olschwang S,
    3. Law DJ,
    4. Melot T,
    5. Remvikos Y,
    6. Salmon RJ,
    7. Sastre X,
    8. Validire P,
    9. Feinberg AP,
    10. Thomas G
    : Multiple genetic alterations in distal and proximal colorectal cancer. Lancet 2(8659): 353-356, 1989.
    OpenUrlCrossRefPubMed
    1. Jernvall P,
    2. Makinen M,
    3. Karttunen T,
    4. Makela J,
    5. Vihko P
    : Conserved region mutations of the p53 gene are concentrated in distal colorectal cancers. Int J Cancer 74(1): 97-101, 1997.
    OpenUrlCrossRefPubMed
  24. ↵
    1. Gervaz P,
    2. Bucher P,
    3. Morel P
    : Two colons–two cancers: paradigm shift and clinical implications. J Surg Oncol 88(4): 261-266, 2004.
    OpenUrlCrossRefPubMed
  25. ↵
    1. Issa JP
    : CpG island methylator phenotype in cancer. Nat Rev Cancer 4(12): 988-993, 2004.
    OpenUrlCrossRefPubMed
    1. Toyota M,
    2. Ohe-Toyota M,
    3. Ahuja N,
    4. Issa JP
    : Distinct genetic profiles in colorectal tumors with or without the CpG island methylator phenotype. Proc Natl Acad Sci USA 97(2): 710-715, 2000.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Hawkins N,
    2. Norrie M,
    3. Cheong K,
    4. Mokany E,
    5. Ku SL,
    6. Meagher A,
    7. O'Connor T,
    8. Ward R
    : CpG island methylation in sporadic colorectal cancers and its relationship to microsatellite instability. Gastroenterology 122(5): 1376-1387, 2002.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Choi PM,
    2. Zelig MP
    : Similarity of colorectal cancer in Crohn's disease and ulcerative colitis: implications for carcinogenesis and prevention. Gut 35(7): 950-954, 1994.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    1. Rakoff-Nahoum S,
    2. Paglino J,
    3. Eslami-Varzaneh F,
    4. Edberg S,
    5. Medzhitov R
    : Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 118(2): 229-241, 2004.
    OpenUrlCrossRefPubMed
  29. ↵
    1. Iwasaki A,
    2. Kelsall BL
    : Freshly isolated Peyer's patch, but not spleen, dendritic cells produce interleukin 10 and induce the differentiation of T-helper type 2 cells. J Exp Med 190(2): 229-239, 1999.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Kelsall BL,
    2. Leon F
    : Involvement of intestinal dendritic cells in oral tolerance, immunity to pathogens, and inflammatory bowel disease. Immunol Rev 206: 132-148, 2005.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Turner JR
    : Intestinal mucosal barrier function in health and disease. Nat Rev Immunol 9(11): 799-809, 2009.
    OpenUrlCrossRefPubMed
  32. ↵
    1. Ganz T
    : Defensins: antimicrobial peptides of innate immunity. Nat Rev Immunol 3(9): 710-720, 2003.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Artis D
    : Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nat Rev Immunol 8(6): 411-420, 2008.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Blikslager AT,
    2. Moeser AJ,
    3. Gookin JL,
    4. Jones SL,
    5. Odle J
    : Restoration of barrier function in injured intestinal mucosa. Physiol Rev 87(2): 545-564, 2007.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Jung HC,
    2. Eckmann L,
    3. Yang SK,
    4. Panja A,
    5. Fierer J,
    6. Morzycka-Wroblewska E,
    7. Kagnoff MF
    : A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bacterial invasion. J Clin Invest 95(1): 55-65, 1995.
    OpenUrlCrossRefPubMed
  36. ↵
    1. Serhan CN,
    2. Savill J
    : Resolution of inflammation: the beginning programs the end. Nat Immunol 6(12): 1191-1197, 2005.
    OpenUrlCrossRefPubMed
  37. ↵
    1. Gewirtz AT,
    2. McCormick B,
    3. Neish AS,
    4. Petasis NA,
    5. Gronert K,
    6. Serhan CN,
    7. Madara JL
    : Pathogen-induced chemokine secretion from model intestinal epithelium is inhibited by lipoxin A4 analogs. J Clin Invest 101(9): 1860-1869, 1998.
    OpenUrlCrossRefPubMed
  38. ↵
    1. An G,
    2. Wei B,
    3. Xia B,
    4. McDaniel JM,
    5. Ju T,
    6. Cummings RD,
    7. Braun J,
    8. Xia L
    : Increased susceptibility to colitis and colorectal tumors in mice lacking core 3-derived O-glycans. J Exp Med 204(6): 1417-1429, 2007.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    1. Byrd JC,
    2. Bresalier RS
    : Mucins and mucin binding proteins in colorectal cancer. Cancer Metastasis Rev 23(1-2): 77-99, 2004.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Jass JR,
    2. Walsh MD
    : Altered mucin expression in the gastrointestinal tract: a review. J Cell Mol Med 5(3): 327-351, 2001.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Van der Sluis M,
    2. De Koning BA,
    3. De Bruijn AC,
    4. Velcich A,
    5. Meijerink JP,
    6. Van Goudoever JB,
    7. Buller HA,
    8. Dekker J,
    9. Van Seuningen I,
    10. Renes IB,
    11. Einerhand AW
    : Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology 131(1): 117-129, 2006.
    OpenUrlCrossRefPubMed
    1. Velcich A,
    2. Yang W,
    3. Heyer J,
    4. Fragale A,
    5. Nicholas C,
    6. Viani S,
    7. Kucherlapati R,
    8. Lipkin M,
    9. Yang K,
    10. Augenlicht L
    : Colorectal cancer in mice genetically deficient in the mucin Muc2. Science 295(5560): 1726-1729, 2002.
    OpenUrlAbstract/FREE Full Text
    1. Hermiston ML,
    2. Gordon JI
    : Inflammatory bowel disease and adenomas in mice expressing a dominant negative N-cadherin. Science 270(5239): 1203-1207, 1995.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    1. Baribault H,
    2. Penner J,
    3. Iozzo RV,
    4. Wilson-Heiner M
    : Colorectal hyperplasia and inflammation in keratin 8-deficient FVB/N mice. Genes Dev 8(24): 2964-2973, 1994.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    1. Okayasu I,
    2. Hatakeyama S,
    3. Yamada M,
    4. Ohkusa T,
    5. Inagaki Y,
    6. Nakaya R
    : A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology 98(3): 694-702, 1990.
    OpenUrlPubMed
  44. ↵
    1. Tanaka T,
    2. Kohno H,
    3. Suzuki R,
    4. Yamada Y,
    5. Sugie S,
    6. Mori H
    : A novel inflammation-related mouse colon carcinogenesis model induced by azoxymethane and dextran sodium sulfate. Cancer Sci 94(11): 965-973, 2003.
    OpenUrlCrossRefPubMed
  45. ↵
    1. Izcue A,
    2. Powrie F
    : Special regulatory T-cell review: Regulatory T-cells and the intestinal tract–patrolling the frontier. Immunology 123(1): 6-10, 2008.
    OpenUrlCrossRefPubMed
    1. Powrie F,
    2. Leach MW,
    3. Mauze S,
    4. Caddle LB,
    5. Coffman RL
    : Phenotypically distinct subsets of CD4+ T-cells induce or protect from chronic intestinal inflammation in C. B-17 scid mice. Int Immunol 5(11): 1461-1471, 1993.
    OpenUrlAbstract/FREE Full Text
    1. Uhlig HH,
    2. Coombes J,
    3. Mottet C,
    4. Izcue A,
    5. Thompson C,
    6. Fanger A,
    7. Tannapfel A,
    8. Fontenot JD,
    9. Ramsdell F,
    10. Powrie F
    : Characterization of Foxp3+CD4+CD25+ and IL-10-secreting CD4+CD25+ T cells during cure of colitis. J Immunol 177(9): 5852-5860, 2006.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Powrie F,
    2. Read S,
    3. Mottet C,
    4. Uhlig H,
    5. Maloy K
    : Control of immune pathology by regulatory T cells. Novartis Found Symp 252: 92-98; discussion 98-105, 106-114, 2003.
    OpenUrlCrossRefPubMed
  47. ↵
    1. Murai M,
    2. Turovskaya O,
    3. Kim G,
    4. Madan R,
    5. Karp CL,
    6. Cheroutre H,
    7. Kronenberg M
    : Interleukin 10 acts on regulatory T-cells to maintain expression of the transcription factor Foxp3 and suppressive function in mice with colitis. Nat Immunol 10(11): 1178-1184, 2009.
    OpenUrlCrossRefPubMed
  48. ↵
    1. Glocker EO,
    2. Kotlarz D,
    3. Boztug K,
    4. Gertz EM,
    5. Schaffer AA,
    6. Noyan F,
    7. Perro M,
    8. Diestelhorst J,
    9. Allroth A,
    10. Murugan D,
    11. Hatscher N,
    12. Pfeifer D,
    13. Sykora KW,
    14. Sauer M,
    15. Kreipe H,
    16. Lacher M,
    17. Nustede R,
    18. Woellner C,
    19. Baumann U,
    20. Salzer U,
    21. Koletzko S,
    22. Shah N,
    23. Segal AW,
    24. Sauerbrey A,
    25. Buderus S,
    26. Snapper SB,
    27. Grimbacher B,
    28. Klein C
    : Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med 361(21): 2033-2045, 2009.
    OpenUrlCrossRefPubMed
  49. ↵
    1. Kuhn R,
    2. Lohler J,
    3. Rennick D,
    4. Rajewsky K,
    5. Muller W
    : Interleukin-10-deficient mice develop chronic enterocolitis. Cell 75(2): 263-274, 1993.
    OpenUrlCrossRefPubMed
  50. ↵
    1. Berg DJ,
    2. Davidson N,
    3. Kuhn R,
    4. Muller W,
    5. Menon S,
    6. Holland G,
    7. Thompson-Snipes L,
    8. Leach MW,
    9. Rennick D
    : Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4(+) TH1-like responses. J Clin Invest 98(4): 1010-1020, 1996.
    OpenUrlCrossRefPubMed
  51. ↵
    1. Davidson NJ,
    2. Leach MW,
    3. Fort MM,
    4. Thompson-Snipes L,
    5. Kuhn R,
    6. Muller W,
    7. Berg DJ,
    8. Rennick DM
    : T-Helper cell 1-type CD4+ T-cells, but not B-cells, mediate colitis in interleukin 10-deficient mice. J Exp Med 184(1): 241-251, 1996.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    1. Kullberg MC,
    2. Rothfuchs AG,
    3. Jankovic D,
    4. Caspar P,
    5. Wynn TA,
    6. Gorelick PL,
    7. Cheever AW,
    8. Sher A
    : Helicobacter hepaticus-induced colitis in interleukin-10-deficient mice: cytokine requirements for the induction and maintenance of intestinal inflammation. Infect Immun 69(7): 4232-4241, 2001.
    OpenUrlAbstract/FREE Full Text
  53. ↵
    1. Watanabe N,
    2. Ikuta K,
    3. Okazaki K,
    4. Nakase H,
    5. Tabata Y,
    6. Matsuura M,
    7. Tamaki H,
    8. Kawanami C,
    9. Honjo T,
    10. Chiba T
    : Elimination of local macrophages in intestine prevents chronic colitis in interleukin-10-deficient mice. Dig Dis Sci 48(2): 408-414, 2003.
    OpenUrlCrossRefPubMed
  54. ↵
    1. Xu W,
    2. Roos A,
    3. Schlagwein N,
    4. Woltman AM,
    5. Daha MR,
    6. van Kooten C
    : IL-10-producing macrophages preferentially clear early apoptotic cells. Blood 107(12): 4930-4937, 2006.
    OpenUrlAbstract/FREE Full Text
  55. ↵
    1. Glauben R,
    2. Batra A,
    3. Stroh T,
    4. Erben U,
    5. Fedke I,
    6. Lehr HA,
    7. Leoni F,
    8. Mascagni P,
    9. Dinarello CA,
    10. Zeitz M,
    11. Siegmund B
    : Histone deacetylases: novel targets for prevention of colitis-associated cancer in mice. Gut 57(5): 613-622, 2008.
    OpenUrlAbstract/FREE Full Text
    1. O'Mahony L,
    2. Feeney M,
    3. O'Halloran S,
    4. Murphy L,
    5. Kiely B,
    6. Fitzgibbon J,
    7. Lee G,
    8. O'Sullivan G,
    9. Shanahan F,
    10. Collins JK
    : Probiotic impact on microbial flora, inflammation and tumour development in IL-10 knockout mice. Aliment Pharmacol Ther 15(8): 1219-1225, 2001.
    OpenUrlCrossRefPubMed
  56. ↵
    1. Chichlowski M,
    2. Sharp JM,
    3. Vanderford DA,
    4. Myles MH,
    5. Hale LP
    : Helicobacter typhlonius and Helicobacter rodentium differentially affect the severity of colon inflammation and inflammation-associated neoplasia in Il10-deficient mice. Comp Med 58(6): 534-541, 2008.
    OpenUrlPubMed
  57. ↵
    1. Hegazi RA,
    2. Mady HH,
    3. Melhem MF,
    4. Sepulveda AR,
    5. Mohi M,
    6. Kandil HM
    : Celecoxib and rofecoxib potentiate chronic colitis and premalignant changes in interleukin 10 knockout mice. Inflamm Bowel Dis 9(4): 230-236, 2003.
    OpenUrlCrossRefPubMed
  58. ↵
    1. Li A,
    2. Goto M,
    3. Horinouchi M,
    4. Tanaka S,
    5. Imai K,
    6. Kim YS,
    7. Sato E,
    8. Yonezawa S
    : Expression of MUC1 and MUC2 mucins and relationship with cell proliferative activity in human colorectal neoplasia. Pathol Int 51(11): 853-860, 2001.
    OpenUrlCrossRefPubMed
  59. ↵
    1. Beatty PL,
    2. Plevy SE,
    3. Sepulveda AR,
    4. Finn OJ
    : Cutting edge: transgenic expression of human MUC1 in Il-10−/− mice accelerates inflammatory bowel disease and progression to colon cancer. J Immunol 179(2): 735-739, 2007.
    OpenUrlAbstract/FREE Full Text
  60. ↵
    1. Sturlan S,
    2. Oberhuber G,
    3. Beinhauer BG,
    4. Tichy B,
    5. Kappel S,
    6. Wang J,
    7. Rogy MA
    : Interleukin-10-deficient mice and inflammatory bowel disease associated cancer development. Carcinogenesis 22(4): 665-671, 2001.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    1. Li MO,
    2. Flavell RA
    : Contextual regulation of inflammation: a duet by transforming growth factor-beta and interleukin-10. Immunity 28(4): 468-476, 2008.
    OpenUrlCrossRefPubMed
  62. ↵
    1. Fantini MC,
    2. Rizzo A,
    3. Fina D,
    4. Caruso R,
    5. Sarra M,
    6. Stolfi C,
    7. Becker C,
    8. Macdonald TT,
    9. Pallone F,
    10. Neurath MF,
    11. Monteleone G
    : Smad7 controls resistance of colitogenic T-cells to regulatory T-cell-mediated suppression. Gastroenterology 136(4): 1308-1316, e1301-1303, 2009.
    OpenUrlCrossRefPubMed
  63. ↵
    1. Souza RF,
    2. Lei J,
    3. Yin J,
    4. Appel R,
    5. Zou TT,
    6. Zhou X,
    7. Wang S,
    8. Rhyu MG,
    9. Cymes K,
    10. Chan O,
    11. Park WS,
    12. Krasna MJ,
    13. Greenwald BD,
    14. Cottrell J,
    15. Abraham JM,
    16. Simms L,
    17. Leggett B,
    18. Young J,
    19. Harpaz N,
    20. Meltzer SJ
    : A transforming growth factor beta 1 receptor type II mutation in ulcerative colitis-associated neoplasms. Gastroenterology 112(1): 40-45, 1997.
    OpenUrlPubMed
  64. ↵
    1. Shull MM,
    2. Ormsby I,
    3. Kier AB,
    4. Pawlowski S,
    5. Diebold RJ,
    6. Yin M,
    7. Allen R,
    8. Sidman C,
    9. Proetzel G,
    10. Calvin D,
    11. et al
    .: Targeted disruption of the mouse transforming growth factor-beta 1 gene results in multifocal inflammatory disease. Nature 359(6397): 693-699, 1992.
    OpenUrlCrossRefPubMed
  65. ↵
    1. Engle SJ,
    2. Hoying JB,
    3. Boivin GP,
    4. Ormsby I,
    5. Gartside PS,
    6. Doetschman T
    : Transforming growth factor beta1 suppresses nonmetastatic colon cancer at an early stage of tumorigenesis. Cancer Res 59(14): 3379-3386, 1999.
    OpenUrlAbstract/FREE Full Text
  66. ↵
    1. Engle SJ,
    2. Ormsby I,
    3. Pawlowski S,
    4. Boivin GP,
    5. Croft J,
    6. Balish E,
    7. Doetschman T
    : Elimination of colon cancer in germ-free transforming growth factor beta 1-deficient mice. Cancer Res 62(22): 6362-6366, 2002.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    1. Graff JM,
    2. Bansal A,
    3. Melton DA
    : Xenopus Mad proteins transduce distinct subsets of signals for the TGF beta superfamily. Cell 85(4): 479-487, 1996.
    OpenUrlCrossRefPubMed
  68. ↵
    1. Zhu Y,
    2. Richardson JA,
    3. Parada LF,
    4. Graff JM
    : Smad3-mutant mice develop metastatic colorectal cancer. Cell 94(6): 703-714, 1998.
    OpenUrlCrossRefPubMed
  69. ↵
    1. Yang X,
    2. Letterio JJ,
    3. Lechleider RJ,
    4. Chen L,
    5. Hayman R,
    6. Gu H,
    7. Roberts AB,
    8. Deng C
    : Targeted disruption of SMAD3 results in impaired mucosal immunity and diminished T-cell responsiveness to TGF-beta. EMBO J 18(5): 1280-1291, 1999.
    OpenUrlAbstract
  70. ↵
    1. Maggio-Price L,
    2. Treuting P,
    3. Zeng W,
    4. Tsang M,
    5. Bielefeldt-Ohmann H,
    6. Iritani BM
    : Helicobacter infection is required for inflammation and colon cancer in SMAD3-deficient mice. Cancer Res 66(2): 828-838, 2006.
    OpenUrlAbstract/FREE Full Text
  71. ↵
    1. Erdman SE,
    2. Poutahidis T,
    3. Tomczak M,
    4. Rogers AB,
    5. Cormier K,
    6. Plank B,
    7. Horwitz BH,
    8. Fox JG
    : CD4+ CD25+ regulatory T-lymphocytes inhibit microbially induced colon cancer in Rag2-deficient mice. Am J Pathol 162(2): 691-702, 2003.
    OpenUrlCrossRefPubMed
    1. Erdman SE,
    2. Rao VP,
    3. Poutahidis T,
    4. Ihrig MM,
    5. Ge Z,
    6. Feng Y,
    7. Tomczak M,
    8. Rogers AB,
    9. Horwitz BH,
    10. Fox JG
    : CD4(+) CD25(+) regulatory lymphocytes require interleukin 10 to interrupt colon carcinogenesis in mice. Cancer Res 63(18): 6042-6050, 2003.
    OpenUrlAbstract/FREE Full Text
  72. ↵
    1. Poutahidis T,
    2. Haigis KM,
    3. Rao VP,
    4. Nambiar PR,
    5. Taylor CL,
    6. Ge Z,
    7. Watanabe K,
    8. Davidson A,
    9. Horwitz BH,
    10. Fox JG,
    11. Erdman SE
    : Rapid reversal of interleukin-6-dependent epithelial invasion in a mouse model of microbially induced colon carcinoma. Carcinogenesis 28(12): 2614-2623, 2007.
    OpenUrlAbstract/FREE Full Text
  73. ↵
    1. Kado S,
    2. Uchida K,
    3. Funabashi H,
    4. Iwata S,
    5. Nagata Y,
    6. Ando M,
    7. Onoue M,
    8. Matsuoka Y,
    9. Ohwaki M,
    10. Morotomi M
    : Intestinal microflora are necessary for development of spontaneous adenocarcinoma of the large intestine in T-cell receptor beta chain and p53 double-knockout mice. Cancer Res 61(6): 2395-2398, 2001.
    OpenUrlAbstract/FREE Full Text
  74. ↵
    1. Cote-Sierra J,
    2. Foucras G,
    3. Guo L,
    4. Chiodetti L,
    5. Young HA,
    6. Hu-Li J,
    7. Zhu J,
    8. Paul WE
    : Interleukin 2 plays a central role in Th2 differentiation. Proc Natl Acad Sci USA 101(11): 3880-3885, 2004.
    OpenUrlAbstract/FREE Full Text
  75. ↵
    1. Fontenot JD,
    2. Rasmussen JP,
    3. Gavin MA,
    4. Rudensky AY
    : A function for interleukin 2 in Foxp3-expressing regulatory T-cells. Nat Immunol 6(11): 1142-1151, 2005.
    OpenUrlCrossRefPubMed
  76. ↵
    1. Kundig TM,
    2. Schorle H,
    3. Bachmann MF,
    4. Hengartner H,
    5. Zinkernagel RM,
    6. Horak I
    : Immune responses in interleukin-2-deficient mice. Science 262(5136): 1059-1061, 1993.
    OpenUrlAbstract/FREE Full Text
    1. Schorle H,
    2. Holtschke T,
    3. Hunig T,
    4. Schimpl A,
    5. Horak I
    : Development and function of T-cells in mice rendered interleukin-2 deficient by gene targeting. Nature 352(6336): 621-624, 1991.
    OpenUrlCrossRefPubMed
  77. ↵
    1. Sadlack B,
    2. Merz H,
    3. Schorle H,
    4. Schimpl A,
    5. Feller AC,
    6. Horak I
    : Ulcerative colitis-like disease in mice with a disrupted interleukin-2 gene. Cell 75(2): 253-261, 1993.
    OpenUrlCrossRefPubMed
  78. ↵
    1. Kramer S,
    2. Schimpl A,
    3. Hunig T
    : Immunopathology of interleukin (IL) 2-deficient mice: thymus dependence and suppression by thymus-dependent cells with an intact IL-2 gene. J Exp Med 182(6): 1769-1776, 1995.
    OpenUrlAbstract/FREE Full Text
  79. ↵
    1. Contractor NV,
    2. Bassiri H,
    3. Reya T,
    4. Park AY,
    5. Baumgart DC,
    6. Wasik MA,
    7. Emerson SG,
    8. Carding SR
    : Lymphoid hyperplasia, autoimmunity, and compromised intestinal intraepithelial lymphocyte development in colitis-free gnotobiotic IL-2-deficient mice. J Immunol 160(1): 385-394, 1998.
    OpenUrlAbstract/FREE Full Text
  80. ↵
    1. McVay LD,
    2. Li B,
    3. Biancaniello R,
    4. Creighton MA,
    5. Bachwich D,
    6. Lichtenstein G,
    7. Rombeau JL,
    8. Carding SR
    : Changes in human mucosal gamma delta T-cell repertoire and function associated with the disease process in inflammatory bowel disease. Mol Med 3(3): 183-203, 1997.
    OpenUrlPubMed
  81. ↵
    1. Simpson SJ,
    2. Mizoguchi E,
    3. Allen D,
    4. Bhan AK,
    5. Terhorst C
    : Evidence that CD4+, but not CD8+ T-cells are responsible for murine interleukin-2-deficient colitis. Eur J Immunol 25(9): 2618-2625, 1995.
    OpenUrlCrossRefPubMed
  82. ↵
    1. Shah SA,
    2. Simpson SJ,
    3. Brown LF,
    4. Comiskey M,
    5. de Jong YP,
    6. Allen D,
    7. Terhorst C
    : Development of colonic adenocarcinomas in a mouse model of ulcerative colitis. Inflamm Bowel Dis 4(3): 196-202, 1998.
    OpenUrlPubMed
  83. ↵
    1. Sohn KJ,
    2. Shah SA,
    3. Reid S,
    4. Choi M,
    5. Carrier J,
    6. Comiskey M,
    7. Terhorst C,
    8. Kim YI
    : Molecular genetics of ulcerative colitis-associated colon cancer in the interleukin 2- and beta(2)-microglobulin-deficient mouse. Cancer Res 61(18): 6912-6917, 2001.
    OpenUrlAbstract/FREE Full Text
  84. ↵
    1. Bicknell DC,
    2. Rowan A,
    3. Bodmer WF
    : Beta 2-microglobulin gene mutations: a study of established colorectal cell lines and fresh tumors. Proc Natl Acad Sci USA 91(11): 4751-4755, 1994.
    OpenUrlAbstract/FREE Full Text
  85. ↵
    1. Fridman WH,
    2. Pages F,
    3. Sautes-Fridman C,
    4. Galon J
    : The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 12(4): 298-306, 2012.
    OpenUrlCrossRefPubMed
  86. ↵
    1. Neves SR,
    2. Ram PT,
    3. Iyengar R
    : G protein pathways. Science 296(5573): 1636-1639, 2002.
    OpenUrlAbstract/FREE Full Text
  87. ↵
    1. Rudolph U,
    2. Finegold MJ,
    3. Rich SS,
    4. Harriman GR,
    5. Srinivasan Y,
    6. Brabet P,
    7. Boulay G,
    8. Bradley A,
    9. Birnbaumer L
    : Ulcerative colitis and adenocarcinoma of the colon in G alpha i2-deficient mice. Nat Genet 10(2): 143-150, 1995.
    OpenUrlCrossRefPubMed
  88. ↵
    1. Edwards RA,
    2. Witherspoon M,
    3. Wang K,
    4. Afrasiabi K,
    5. Pham T,
    6. Birnbaumer L,
    7. Lipkin SM
    : Epigenetic repression of DNA mismatch repair by inflammation and hypoxia in inflammatory bowel disease-associated colorectal cancer. Cancer Res 69(16): 6423-6429, 2009.
    OpenUrlAbstract/FREE Full Text
  89. ↵
    1. Hanada T,
    2. Kobayashi T,
    3. Chinen T,
    4. Saeki K,
    5. Takaki H,
    6. Koga K,
    7. Minoda Y,
    8. Sanada T,
    9. Yoshioka T,
    10. Mimata H,
    11. Kato S,
    12. Yoshimura A
    : IFNgamma-dependent, spontaneous development of colorectal carcinomas in SOCS1-deficient mice. J Exp Med 203(6): 1391-1397, 2006.
    OpenUrlAbstract/FREE Full Text
  90. ↵
    1. Deng L,
    2. Zhou JF,
    3. Sellers RS,
    4. Li JF,
    5. Nguyen AV,
    6. Wang Y,
    7. Orlofsky A,
    8. Liu Q,
    9. Hume DA,
    10. Pollard JW,
    11. Augenlicht L,
    12. Lin EY
    : A novel mouse model of inflammatory bowel disease links mammalian target of rapamycin-dependent hyperproliferation of colonic epithelium to inflammation-associated tumorigenesis. Am J Pathol 176(2): 952-967, 2010.
    OpenUrlCrossRefPubMed
  91. ↵
    1. Coombes JL,
    2. Powrie F
    : Dendritic cells in intestinal immune regulation. Nat Rev Immunol 8(6): 435-446, 2008.
    OpenUrlCrossRefPubMed
  92. ↵
    1. Niess JH,
    2. Brand S,
    3. Gu X,
    4. Landsman L,
    5. Jung S,
    6. McCormick BA,
    7. Vyas JM,
    8. Boes M,
    9. Ploegh HL,
    10. Fox JG,
    11. Littman DR,
    12. Reinecker HC
    : CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 307(5707): 254-258, 2005.
    OpenUrlAbstract/FREE Full Text
    1. Rescigno M,
    2. Rotta G,
    3. Valzasina B,
    4. Ricciardi-Castagnoli P
    : Dendritic cells shuttle microbes across gut epithelial monolayers. Immunobiology 204(5): 572-581, 2001.
    OpenUrlCrossRefPubMed
  93. ↵
    1. Rescigno M,
    2. Urbano M,
    3. Valzasina B,
    4. Francolini M,
    5. Rotta G,
    6. Bonasio R,
    7. Granucci F,
    8. Kraehenbuhl JP,
    9. Ricciardi-Castagnoli P
    : Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol 2(4): 361-367, 2001.
    OpenUrlCrossRefPubMed
  94. ↵
    1. Saenz SA,
    2. Taylor BC,
    3. Artis D
    : Welcome to the neighborhood: epithelial cell-derived cytokines license innate and adaptive immune responses at mucosal sites. Immunol Rev 226: 172-190, 2008.
    OpenUrlCrossRefPubMed
  95. ↵
    1. Taylor BC,
    2. Zaph C,
    3. Troy AE,
    4. Du Y,
    5. Guild KJ,
    6. Comeau MR,
    7. Artis D
    : TSLP regulates intestinal immunity and inflammation in mouse models of helminth infection and colitis. J Exp Med 206(3): 655-667, 2009.
    OpenUrlAbstract/FREE Full Text
  96. ↵
    1. Glimcher LH
    : Trawling for treasure: tales of T-bet. Nat Immunol 8(5): 448-450, 2007.
    OpenUrlCrossRefPubMed
  97. ↵
    1. Lugo-Villarino G,
    2. Ito S,
    3. Klinman DM,
    4. Glimcher LH
    : The adjuvant activity of CpG DNA requires T-bet expression in dendritic cells. Proc Natl Acad Sci USA 102(37): 13248-13253, 2005.
    OpenUrlAbstract/FREE Full Text
  98. ↵
    1. Garrett WS,
    2. Punit S,
    3. Gallini CA,
    4. Michaud M,
    5. Zhang D,
    6. Sigrist KS,
    7. Lord GM,
    8. Glickman JN,
    9. Glimcher LH
    : Colitis-associated colorectal cancer driven by T-bet deficiency in dendritic cells. Cancer Cell 16(3): 208-219, 2009.
    OpenUrlCrossRefPubMed
  99. ↵
    1. Garrett WS,
    2. Glimcher LH
    : T-bet−/− RAG2−/− ulcerative colitis: the role of T-bet as a peacekeeper of host-commensal relationships. Cytokine 48(1-2): 144-147, 2009.
    OpenUrlCrossRefPubMed
  100. ↵
    1. Ermann J,
    2. Garrett WS,
    3. Kuchroo J,
    4. Rourida K,
    5. Glickman JN,
    6. Bleich A,
    7. Glimcher LH
    : Severity of innate immune-mediated colitis is controlled by the cytokine deficiency-induced colitis susceptibility-1 (Cdcs1) locus. Proc Natl Acad Sci USA 108(17): 7137-7141, 2011.
    OpenUrlAbstract/FREE Full Text
  101. ↵
    1. Okayasu I,
    2. Ohkusa T,
    3. Kajiura K,
    4. Kanno J,
    5. Sakamoto S
    : Promotion of colorectal neoplasia in experimental murine ulcerative colitis. Gut 39(1): 87-92, 1996.
    OpenUrlAbstract/FREE Full Text
  102. ↵
    1. Blackburn AC,
    2. Doe WF,
    3. Buffinton GD
    : Salicylate hydroxylation as an indicator of hydroxyl radical generation in dextran sulfate-induced colitis. Free Radic Biol Med 25(3): 305-313, 1998.
    OpenUrlCrossRefPubMed
  103. ↵
    1. Blackburn AC,
    2. Doe WF,
    3. Buffinton GD
    : Protein carbonyl formation on mucosal proteins in vitro and in dextran sulfate-induced colitis. Free Radic Biol Med 27(3-4): 262-270, 1999.
    OpenUrlCrossRefPubMed
  104. ↵
    1. Buffinton GD,
    2. Doe WF
    : Depleted mucosal antioxidant defences in inflammatory bowel disease. Free Radic Biol Med 19(6): 911-918, 1995.
    OpenUrlCrossRefPubMed
  105. ↵
    1. Buffinton GD,
    2. Doe WF
    : Altered ascorbic acid status in the mucosa from inflammatory bowel disease patients. Free Radic Res 22(2): 131-143, 1995.
    OpenUrlCrossRefPubMed
  106. ↵
    1. Kohonen-Corish MR,
    2. Daniel JJ,
    3. te Riele H,
    4. Buffinton GD,
    5. Dahlstrom JE
    : Susceptibility of Msh2-deficient mice to inflammation-associated colorectal tumors. Cancer Res 62(7): 2092-2097, 2002.
    OpenUrlAbstract/FREE Full Text
  107. ↵
    1. Cooper HS,
    2. Murthy S,
    3. Kido K,
    4. Yoshitake H,
    5. Flanigan A
    : Dysplasia and cancer in the dextran sulfate sodium mouse colitis model. Relevance to colitis-associated neoplasia in the human: a study of histopathology, β-catenin and p53 expression and the role of inflammation. Carcinogenesis 21(4): 757-768, 2000.
    OpenUrlAbstract/FREE Full Text
  108. ↵
    1. Chang WC,
    2. Coudry RA,
    3. Clapper ML,
    4. Zhang X,
    5. Williams KL,
    6. Spittle CS,
    7. Li T,
    8. Cooper HS
    : Loss of p53 enhances the induction of colitis-associated neoplasia by dextran sulfate sodium. Carcinogenesis 28(11): 2375-2381, 2007.
    OpenUrlAbstract/FREE Full Text
  109. ↵
    1. Tanaka T
    : Development of an inflammation-associated colorectal cancer model and its application for research on carcinogenesis and chemoprevention. Int J Inflam 2012: 658786, 2012.
    OpenUrlPubMed
  110. ↵
    1. Hardwick JC,
    2. van den Brink GR,
    3. Offerhaus GJ,
    4. van Deventer SJ,
    5. Peppelenbosch MP
    : NF-kappaB, p38 MAPK and JNK are highly expressed and active in the stroma of human colonic adenomatous polyps. Oncogene 20(7): 819-827, 2001.
    OpenUrlCrossRefPubMed
  111. ↵
    1. Lind DS,
    2. Hochwald SN,
    3. Malaty J,
    4. Rekkas S,
    5. Hebig P,
    6. Mishra G,
    7. Moldawer LL,
    8. Copeland EM 3rd.,
    9. Mackay S
    : Nuclear factor-kappa B is up-regulated in colorectal cancer. Surgery 130(2): 363-369, 2001.
    OpenUrlCrossRefPubMed
  112. ↵
    1. Rogler G,
    2. Brand K,
    3. Vogl D,
    4. Page S,
    5. Hofmeister R,
    6. Andus T,
    7. Knuechel R,
    8. Baeuerle PA,
    9. Scholmerich J,
    10. Gross V
    : Nuclear factor kappaB is activated in macrophages and epithelial cells of inflamed intestinal mucosa. Gastroenterology 115(2): 357-369, 1998.
    OpenUrlCrossRefPubMed
  113. ↵
    1. Neurath MF,
    2. Pettersson S,
    3. Meyer zum Buschenfelde KH,
    4. Strober W
    : Local administration of antisense phosphorothioate oligonucleotides to the p65 subunit of NF-kappa B abrogates established experimental colitis in mice. Nat Med 2(9): 998-1004, 1996.
    OpenUrlCrossRefPubMed
  114. ↵
    1. Karin M,
    2. Greten FR
    : NF-kappaB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol 5(10): 749-759, 2005.
    OpenUrlCrossRefPubMed
  115. ↵
    1. Greten FR,
    2. Eckmann L,
    3. Greten TF,
    4. Park JM,
    5. Li ZW,
    6. Egan LJ,
    7. Kagnoff MF,
    8. Karin M
    : IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 118(3): 285-296, 2004.
    OpenUrlCrossRefPubMed
  116. ↵
    1. Eckmann L,
    2. Nebelsiek T,
    3. Fingerle AA,
    4. Dann SM,
    5. Mages J,
    6. Lang R,
    7. Robine S,
    8. Kagnoff MF,
    9. Schmid RM,
    10. Karin M,
    11. Arkan MC,
    12. Greten FR
    : Opposing functions of IKKbeta during acute and chronic intestinal inflammation. Proc Natl Acad Sci USA 105(39): 15058-15063, 2008.
    OpenUrlAbstract/FREE Full Text
  117. ↵
    1. Grivennikov S,
    2. Karin E,
    3. Terzic J,
    4. Mucida D,
    5. Yu GY,
    6. Vallabhapurapu S,
    7. Scheller J,
    8. Rose-John S,
    9. Cheroutre H,
    10. Eckmann L,
    11. Karin M
    : IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell 15(2): 103-113, 2009.
    OpenUrlCrossRefPubMed
  118. ↵
    1. Li Y,
    2. de Haar C,
    3. Chen M,
    4. Deuring J,
    5. Gerrits MM,
    6. Smits R,
    7. Xia B,
    8. Kuipers EJ,
    9. van der Woude CJ
    : Disease-related expression of the IL6/STAT3/SOCS3 signalling pathway in ulcerative colitis and ulcerative colitis-related carcinogenesis. Gut 59(2): 227-235, 2010.
    OpenUrlAbstract/FREE Full Text
  119. ↵
    1. Rigby RJ,
    2. Simmons JG,
    3. Greenhalgh CJ,
    4. Alexander WS,
    5. Lund PK
    : Suppressor of cytokine signaling 3 (SOCS3) limits damage-induced crypt hyper-proliferation and inflammation-associated tumorigenesis in the colon. Oncogene 26(33): 4833-4841, 2007.
    OpenUrlCrossRefPubMed
  120. ↵
    1. Tebbutt NC,
    2. Giraud AS,
    3. Inglese M,
    4. Jenkins B,
    5. Waring P,
    6. Clay FJ,
    7. Malki S,
    8. Alderman BM,
    9. Grail D,
    10. Hollande F,
    11. Heath JK,
    12. Ernst M
    : Reciprocal regulation of gastrointestinal homeostasis by SHP2 and STAT-mediated trefoil gene activation in gp130 mutant mice. Nat Med 8(10): 1089-1097, 2002.
    OpenUrlCrossRefPubMed
    1. Dann SM,
    2. Spehlmann ME,
    3. Hammond DC,
    4. Iimura M,
    5. Hase K,
    6. Choi LJ,
    7. Hanson E,
    8. Eckmann L
    : IL-6-dependent mucosal protection prevents establishment of a microbial niche for attaching/effacing lesion-forming enteric bacterial pathogens. J Immunol 180(10): 6816-6826, 2008.
    OpenUrlAbstract/FREE Full Text
  121. ↵
    1. Kishimoto T
    : Interleukin-6: from basic science to medicine-40 years in immunology. Annu Rev Immunol 23(1-21, 2005.
    OpenUrlCrossRefPubMed
  122. ↵
    1. Bettelli E,
    2. Carrier Y,
    3. Gao W,
    4. Korn T,
    5. Strom TB,
    6. Oukka M,
    7. Weiner HL,
    8. Kuchroo VK
    : Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T-cells. Nature 441(7090): 235-238, 2006.
    OpenUrlCrossRefPubMed
  123. ↵
    1. Waldner MJ,
    2. Wirtz S,
    3. Jefremow A,
    4. Warntjen M,
    5. Neufert C,
    6. Atreya R,
    7. Becker C,
    8. Weigmann B,
    9. Vieth M,
    10. Rose-John S,
    11. Neurath MF
    : VEGF receptor signaling links inflammation and tumorigenesis in colitis-associated cancer. J Exp Med 207(13): 2855-2868, 2010.
    OpenUrlAbstract/FREE Full Text
  124. ↵
    1. Fina D,
    2. Sarra M,
    3. Fantini MC,
    4. Rizzo A,
    5. Caruso R,
    6. Caprioli F,
    7. Stolfi C,
    8. Cardolini I,
    9. Dottori M,
    10. Boirivant M,
    11. Pallone F,
    12. Macdonald TT,
    13. Monteleone G
    : Regulation of gut inflammation and th17 cell response by interleukin-21. Gastroenterology 134(4): 1038-1048, 2008.
    OpenUrlCrossRefPubMed
  125. ↵
    1. Stolfi C,
    2. Rizzo A,
    3. Franze E,
    4. Rotondi A,
    5. Fantini MC,
    6. Sarra M,
    7. Caruso R,
    8. Monteleone I,
    9. Sileri P,
    10. Franceschilli L,
    11. Caprioli F,
    12. Ferrero S,
    13. MacDonald TT,
    14. Pallone F,
    15. Monteleone G
    : Involvement of interleukin-21 in the regulation of colitis-associated colon cancer. J Exp Med 208(11): 2279-2290, 2011.
    OpenUrlAbstract/FREE Full Text
  126. ↵
    1. Balkwill F
    : Tumor necrosis factor or tumor promoting factor? Cytokine Growth Factor Rev 13(2): 135-141, 2002.
    OpenUrlCrossRefPubMed
  127. ↵
    1. Jaiswal M,
    2. LaRusso NF,
    3. Burgart LJ,
    4. Gores GJ
    : Inflammatory cytokines induce DNA damage and inhibit DNA repair in cholangiocarcinoma cells by a nitric oxide-dependent mechanism. Cancer Res 60(1): 184-190, 2000.
    OpenUrlAbstract/FREE Full Text
    1. Leber TM,
    2. Balkwill FR
    : Regulation of monocyte MMP-9 production by TNF-alpha and a tumour-derived soluble factor (MMPSF). Br J Cancer 78(6): 724-732, 1998.
    OpenUrlCrossRefPubMed
    1. Gordon HM,
    2. Kucera G,
    3. Salvo R,
    4. Boss JM
    : Tumor necrosis factor induces genes involved in inflammation, cellular and tissue repair, and metabolism in murine fibroblasts. J Immunol 148(12): 4021-4027, 1992.
    OpenUrlAbstract
    1. Yoshida S,
    2. Ono M,
    3. Shono T,
    4. Izumi H,
    5. Ishibashi T,
    6. Suzuki H,
    7. Kuwano M
    : Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor alpha-dependent angiogenesis. Mol Cell Biol 17(7): 4015-4023, 1997.
    OpenUrlAbstract/FREE Full Text
  128. ↵
    1. Grivennikov SI,
    2. Kuprash DV,
    3. Liu ZG,
    4. Nedospasov SA
    : Intracellular signals and events activated by cytokines of the tumor necrosis factor superfamily: From simple paradigms to complex mechanisms. Int Rev Cytol 252: 129-161, 2006.
    OpenUrlCrossRefPubMed
  129. ↵
    1. Popivanova BK,
    2. Kitamura K,
    3. Wu Y,
    4. Kondo T,
    5. Kagaya T,
    6. Kaneko S,
    7. Oshima M,
    8. Fujii C,
    9. Mukaida N
    : Blocking TNF-alpha in mice reduces colorectal carcinogenesis associated with chronic colitis. J Clin Invest 118(2): 560-570, 2008.
    OpenUrlPubMed
  130. ↵
    1. Medzhitov R
    : Toll-like receptors and innate immunity. Nat Rev Immunol 1(2): 135-145, 2001.
    OpenUrlCrossRefPubMed
  131. ↵
    1. Karin M,
    2. Lawrence T,
    3. Nizet V
    : Innate immunity gone awry: linking microbial infections to chronic inflammation and cancer. Cell 124(4): 823-835, 2006.
    OpenUrlCrossRefPubMed
  132. ↵
    1. Salcedo R,
    2. Worschech A,
    3. Cardone M,
    4. Jones Y,
    5. Gyulai Z,
    6. Dai RM,
    7. Wang E,
    8. Ma W,
    9. Haines D,
    10. O'HUigin C,
    11. Marincola FM,
    12. Trinchieri G
    : MyD88-mediated signaling prevents development of adenocarcinomas of the colon: role of interleukin 18. J Exp Med 207(8): 1625-1636, 2010.
    OpenUrlAbstract/FREE Full Text
  133. ↵
    1. Xiao H,
    2. Gulen MF,
    3. Qin J,
    4. Yao J,
    5. Bulek K,
    6. Kish D,
    7. Altuntas CZ,
    8. Wald D,
    9. Ma C,
    10. Zhou H,
    11. Tuohy VK,
    12. Fairchild RL,
    13. de la Motte C,
    14. Cua D,
    15. Vallance BA,
    16. Li X
    : The Toll-interleukin-1 receptor member SIGIRR regulates colonic epithelial homeostasis, inflammation, and tumorigenesis. Immunity 26(4): 461-475, 2007.
    OpenUrlCrossRefPubMed
  134. ↵
    1. Fukata M,
    2. Chen A,
    3. Vamadevan AS,
    4. Cohen J,
    5. Breglio K,
    6. Krishnareddy S,
    7. Hsu D,
    8. Xu R,
    9. Harpaz N,
    10. Dannenberg AJ,
    11. Subbaramaiah K,
    12. Cooper HS,
    13. Itzkowitz SH,
    14. Abreu MT
    : Toll-like receptor-4 promotes the development of colitis-associated colorectal tumors. Gastroenterology 133(6): 1869-1881, 2007.
    OpenUrlCrossRefPubMed
  135. ↵
    1. Nakanishi M,
    2. Menoret A,
    3. Tanaka T,
    4. Miyamoto S,
    5. Montrose DC,
    6. Vella AT,
    7. Rosenberg DW
    : Selective PGE(2) suppression inhibits colon carcinogenesis and modifies local mucosal immunity. Cancer Prev Res 4(8): 1198-1208, 2011.
    OpenUrlAbstract/FREE Full Text
  136. ↵
    1. Kanneganti TD,
    2. Lamkanfi M,
    3. Nunez G
    : Intracellular NOD-like receptors in host defense and disease. Immunity 27(4): 549-559, 2007.
    OpenUrlCrossRefPubMed
  137. ↵
    1. Ghayur T,
    2. Banerjee S,
    3. Hugunin M,
    4. Butler D,
    5. Herzog L,
    6. Carter A,
    7. Quintal L,
    8. Sekut L,
    9. Talanian R,
    10. Paskind M,
    11. Wong W,
    12. Kamen R,
    13. Tracey D,
    14. Allen H
    : Caspase-1 processes IFN-gamma-inducing factor and regulates LPS-induced IFN-gamma production. Nature 386(6625): 619-623, 1997.
    OpenUrlCrossRefPubMed
  138. ↵
    1. Kuida K,
    2. Lippke JA,
    3. Ku G,
    4. Harding MW,
    5. Livingston DJ,
    6. Su MS,
    7. Flavell RA
    : Altered cytokine export and apoptosis in mice deficient in interleukin-1 beta converting enzyme. Science 267(5206): 2000-2003, 1995.
    OpenUrlAbstract/FREE Full Text
  139. ↵
    1. Zitvogel L,
    2. Kepp O,
    3. Galluzzi L,
    4. Kroemer G
    : Inflammasomes in carcinogenesis and anticancer immune responses. Nat Immunol 13(4): 343-351, 2012.
    OpenUrlCrossRefPubMed
  140. ↵
    1. Villani AC,
    2. Lemire M,
    3. Fortin G,
    4. Louis E,
    5. Silverberg MS,
    6. Collette C,
    7. Baba N,
    8. Libioulle C,
    9. Belaiche J,
    10. Bitton A,
    11. Gaudet D,
    12. Cohen A,
    13. Langelier D,
    14. Fortin PR,
    15. Wither JE,
    16. Sarfati M,
    17. Rutgeerts P,
    18. Rioux JD,
    19. Vermeire S,
    20. Hudson TJ,
    21. Franchimont D
    : Common variants in the NLRP3 region contribute to Crohn's disease susceptibility. Nat Genet 41(1): 71-76, 2009.
    OpenUrlCrossRefPubMed
  141. ↵
    1. Zhernakova A,
    2. Festen EM,
    3. Franke L,
    4. Trynka G,
    5. van Diemen CC,
    6. Monsuur AJ,
    7. Bevova M,
    8. Nijmeijer RM,
    9. van ‘t Slot R,
    10. Heijmans R,
    11. Boezen HM,
    12. van Heel DA,
    13. van Bodegraven AA,
    14. Stokkers PC,
    15. Wijmenga C,
    16. Crusius JB,
    17. Weersma RK
    : Genetic analysis of innate immunity in Crohn's disease and ulcerative colitis identifies two susceptibility loci harboring CARD9 and IL18RAP. Am J Hum Genet 82(5): 1202-1210, 2008.
    OpenUrlCrossRefPubMed
  142. ↵
    1. Siegmund B
    : Interleukin-18 in intestinal inflammation: friend and foe? Immunity 32(3): 300-302, 2010.
    OpenUrlCrossRefPubMed
  143. ↵
    1. Hu B,
    2. Elinav E,
    3. Huber S,
    4. Booth CJ,
    5. Strowig T,
    6. Jin C,
    7. Eisenbarth SC,
    8. Flavell RA
    : Inflammation-induced tumorigenesis in the colon is regulated by caspase-1 and NLRC4. Proc Natl Acad Sci USA 107(50): 21635-21640, 2010.
    OpenUrlAbstract/FREE Full Text
  144. ↵
    1. Elinav E,
    2. Strowig T,
    3. Kau AL,
    4. Henao-Mejia J,
    5. Thaiss CA,
    6. Booth CJ,
    7. Peaper DR,
    8. Bertin J,
    9. Eisenbarth SC,
    10. Gordon JI,
    11. Flavell RA
    : NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 145(5): 745-757, 2011.
    OpenUrlCrossRefPubMed
  145. ↵
    1. Zaki MH,
    2. Vogel P,
    3. Body-Malapel M,
    4. Lamkanfi M,
    5. Kanneganti TD
    : IL-18 production downstream of the Nlrp3 inflammasome confers protection against colorectal tumor formation. J Immunol 185(8): 4912-4920, 2010.
    OpenUrlAbstract/FREE Full Text
  146. ↵
    1. Dunn GP,
    2. Koebel CM,
    3. Schreiber RD
    : Interferons, immunity and cancer immunoediting. Nat Rev Immunol 6(11): 836-848, 2006.
    OpenUrlCrossRefPubMed
  147. ↵
    1. Okamura H,
    2. Tsutsi H,
    3. Komatsu T,
    4. Yutsudo M,
    5. Hakura A,
    6. Tanimoto T,
    7. Torigoe K,
    8. Okura T,
    9. Nukada Y,
    10. Hattori K,
    11. et al
    : Cloning of a new cytokine that induces IFN-gamma production by T-cells. Nature 378(6552): 88-91, 1995.
    OpenUrlCrossRefPubMed
  148. ↵
    1. Takeda K,
    2. Tsutsui H,
    3. Yoshimoto T,
    4. Adachi O,
    5. Yoshida N,
    6. Kishimoto T,
    7. Okamura H,
    8. Nakanishi K,
    9. Akira S
    : Defective NK cell activity and Th1 response in Il-18-deficient mice. Immunity 8(3): 383-390, 1998.
    OpenUrlCrossRefPubMed
  149. ↵
    1. Allen IC,
    2. TeKippe EM,
    3. Woodford RM,
    4. Uronis JM,
    5. Holl EK,
    6. Rogers AB,
    7. Herfarth HH,
    8. Jobin C,
    9. Ting JP
    : The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med 207(5): 1045-1056, 2010.
    OpenUrlAbstract/FREE Full Text
  150. ↵
    1. Chen GY,
    2. Shaw MH,
    3. Redondo G,
    4. Nunez G
    : The innate immune receptor Nod1 protects the intestine from inflammation-induced tumorigenesis. Cancer Res 68(24): 10060-10067, 2008.
    OpenUrlAbstract/FREE Full Text
  151. ↵
    1. Colgan SP,
    2. Taylor CT
    : Hypoxia: an alarm signal during intestinal inflammation. Nat Rev Gastroenterol Hepatol 7(5): 281-287, 2010.
    OpenUrlCrossRefPubMed
  152. ↵
    1. Semenza GL
    : Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3(10): 721-732, 2003.
    OpenUrlCrossRefPubMed
  153. ↵
    1. Wang T,
    2. Marquardt C,
    3. Foker J
    : Aerobic glycolysis during lymphocyte proliferation. Nature 261(5562): 702-705, 1976.
    OpenUrlCrossRefPubMed
    1. Borregaard N,
    2. Herlin T
    : Energy metabolism of human neutrophils during phagocytosis. J Clin Invest 70(3): 550-557, 1982.
    OpenUrlCrossRefPubMed
    1. MacDonald HR,
    2. Koch CJ
    : Energy metabolism and T-cell-mediated cytolysis. I. Synergism between inhibitors of respiration and glycolysis. J Exp Med 146(3): 698-709, 1977.
    OpenUrlAbstract/FREE Full Text
  154. ↵
    1. Cramer T,
    2. Yamanishi Y,
    3. Clausen BE,
    4. Forster I,
    5. Pawlinski R,
    6. Mackman N,
    7. Haase VH,
    8. Jaenisch R,
    9. Corr M,
    10. Nizet V,
    11. Firestein GS,
    12. Gerber HP,
    13. Ferrara N,
    14. Johnson RS
    : HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell 112(5): 645-657, 2003.
    OpenUrlCrossRefPubMed
  155. ↵
    1. Makino Y,
    2. Nakamura H,
    3. Ikeda E,
    4. Ohnuma K,
    5. Yamauchi K,
    6. Yabe Y,
    7. Poellinger L,
    8. Okada Y,
    9. Morimoto C,
    10. Tanaka H
    : Hypoxia-inducible factor regulates survival of antigen receptor-driven T-cells. J Immunol 171(12): 6534-6540, 2003.
    OpenUrlAbstract/FREE Full Text
  156. ↵
    1. Biju MP,
    2. Neumann AK,
    3. Bensinger SJ,
    4. Johnson RS,
    5. Turka LA,
    6. Haase VH
    : Vhlh gene deletion induces Hif-1-mediated cell death in thymocytes. Mol Cell Biol 24(20): 9038-9047, 2004.
    OpenUrlAbstract/FREE Full Text
  157. ↵
    1. Kojima H,
    2. Gu H,
    3. Nomura S,
    4. Caldwell CC,
    5. Kobata T,
    6. Carmeliet P,
    7. Semenza GL,
    8. Sitkovsky MV
    : Abnormal B lymphocyte development and autoimmunity in hypoxia-inducible factor 1alpha -deficient chimeric mice. Proc Natl Acad Sci USA 99(4): 2170-2174, 2002.
    OpenUrlAbstract/FREE Full Text
  158. ↵
    1. Hellwig-Burgel T,
    2. Stiehl DP,
    3. Wagner AE,
    4. Metzen E,
    5. Jelkmann W
    : Review: hypoxia-inducible factor-1 (HIF-1): a novel transcription factor in immune reactions. J Interferon Cytokine Res 25(6): 297-310, 2005.
    OpenUrlCrossRefPubMed
  159. ↵
    1. Bracken CP,
    2. Whitelaw ML,
    3. Peet DJ
    : The hypoxia-inducible factors: key transcriptional regulators of hypoxic responses. Cell Mol Life Sci 60(7): 1376-1393, 2003.
    OpenUrlCrossRefPubMed
  160. ↵
    1. Peyssonnaux C,
    2. Cejudo-Martin P,
    3. Doedens A,
    4. Zinkernagel AS,
    5. Johnson RS,
    6. Nizet V
    : Cutting edge: Essential role of hypoxia inducible factor-1alpha in development of lipopolysaccharide-induced sepsis. J Immunol 178(12): 7516-7519, 2007.
    OpenUrlAbstract/FREE Full Text
  161. ↵
    1. Rius J,
    2. Guma M,
    3. Schachtrup C,
    4. Akassoglou K,
    5. Zinkernagel AS,
    6. Nizet V,
    7. Johnson RS,
    8. Haddad GG,
    9. Karin M
    : NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature 453(7196): 807-811, 2008.
    OpenUrlCrossRefPubMed
  162. ↵
    1. Taylor CT
    : Interdependent roles for hypoxia inducible factor and nuclear factor-kappaB in hypoxic inflammation. J Physiol 586(Pt 17): 4055-4059, 2008.
    OpenUrlCrossRefPubMed
  163. ↵
    1. Shah YM,
    2. Ito S,
    3. Morimura K,
    4. Chen C,
    5. Yim SH,
    6. Haase VH,
    7. Gonzalez FJ
    : Hypoxia-inducible factor augments experimental colitis through an MIF-dependent inflammatory signaling cascade. Gastroenterology 134(7): 2036-2048, 2048 e2031-2033, 2008.
    OpenUrlCrossRefPubMed
  164. ↵
    1. Hirota SA,
    2. Beck PL,
    3. MacDonald JA
    : Targeting hypoxia-inducible factor-1 (HIF-1) signaling in therapeutics: implications for the treatment of inflammatory bowel disease. Recent Pat Inflamm Allergy Drug Discov 3(1): 1-16, 2009.
    OpenUrlCrossRefPubMed
  165. ↵
    1. Cummins EP,
    2. Seeballuck F,
    3. Keely SJ,
    4. Mangan NE,
    5. Callanan JJ,
    6. Fallon PG,
    7. Taylor CT
    : The hydroxylase inhibitor dimethyloxalylglycine is protective in a murine model of colitis. Gastroenterology 134(1): 156-165, 2008.
    OpenUrlCrossRefPubMed
    1. Tambuwala MM,
    2. Cummins EP,
    3. Lenihan CR,
    4. Kiss J,
    5. Stauch M,
    6. Scholz CC,
    7. Fraisl P,
    8. Lasitschka F,
    9. Mollenhauer M,
    10. Saunders SP,
    11. Maxwell PH,
    12. Carmeliet P,
    13. Fallon PG,
    14. Schneider M,
    15. Taylor CT
    : Loss of Prolyl Hydroxylase-1 Protects Against Colitis Through Reduced Epithelial Cell Apoptosis and Increased Barrier Function. Gastroenterology 139(6): 2093-2101, 2010.
    OpenUrlCrossRefPubMed
  166. ↵
    1. Robinson A,
    2. Keely S,
    3. Karhausen J,
    4. Gerich ME,
    5. Furuta GT,
    6. Colgan SP
    : Mucosal protection by hypoxia-inducible factor prolyl hydroxylase inhibition. Gastroenterology 134(1): 145-155, 2008.
    OpenUrlCrossRefPubMed
  167. ↵
    1. Eltzschig HK,
    2. Thompson LF,
    3. Karhausen J,
    4. Cotta RJ,
    5. Ibla JC,
    6. Robson SC,
    7. Colgan SP
    : Endogenous adenosine produced during hypoxia attenuates neutrophil accumulation: coordination by extracellular nucleotide metabolism. Blood 104(13): 3986-3992, 2004.
    OpenUrlAbstract/FREE Full Text
    1. Furuta GT,
    2. Turner JR,
    3. Taylor CT,
    4. Hershberg RM,
    5. Comerford K,
    6. Narravula S,
    7. Podolsky DK,
    8. Colgan SP
    : Hypoxia-inducible factor 1-dependent induction of intestinal trefoil factor protects barrier function during hypoxia. J Exp Med 193(9): 1027-1034, 2001.
    OpenUrlAbstract/FREE Full Text
  168. ↵
    1. Karhausen J,
    2. Furuta GT,
    3. Tomaszewski JE,
    4. Johnson RS,
    5. Colgan SP,
    6. Haase VH
    : Epithelial hypoxia-inducible factor-1 is protective in murine experimental colitis. J Clin Invest 114(8): 1098-1106, 2004.
    OpenUrlCrossRefPubMed
  169. ↵
    1. Cummins EP,
    2. Berra E,
    3. Comerford KM,
    4. Ginouves A,
    5. Fitzgerald KT,
    6. Seeballuck F,
    7. Godson C,
    8. Nielsen JE,
    9. Moynagh P,
    10. Pouyssegur J,
    11. Taylor CT
    : Prolyl hydroxylase-1 negatively regulates IkappaB kinase-beta, giving insight into hypoxia-induced NFkappaB activity. Proc Natl Acad Sci USA 103(48): 18154-18159, 2006.
    OpenUrlAbstract/FREE Full Text
  170. ↵
    1. Zhong H,
    2. De Marzo AM,
    3. Laughner E,
    4. Lim M,
    5. Hilton DA,
    6. Zagzag D,
    7. Buechler P,
    8. Isaacs WB,
    9. Semenza GL,
    10. Simons JW
    : Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res 59(22): 5830-5835, 1999.
    OpenUrlAbstract/FREE Full Text
  171. ↵
    1. Olaru AV,
    2. Selaru FM,
    3. Mori Y,
    4. Vazquez C,
    5. David S,
    6. Paun B,
    7. Cheng Y,
    8. Jin Z,
    9. Yang J,
    10. Agarwal R,
    11. Abraham JM,
    12. Dassopoulos T,
    13. Harris M,
    14. Bayless TM,
    15. Kwon J,
    16. Harpaz N,
    17. Livak F,
    18. Meltzer SJ
    : Dynamic changes in the expression of MicroRNA-31 during inflammatory bowel disease-associated neoplastic transformation. Inflamm Bowel Dis 17(1): 221-231, 2011.
    OpenUrlCrossRefPubMed
  172. ↵
    1. Abraham C,
    2. Cho J
    : Interleukin-23/Th17 pathways and inflammatory bowel disease. Inflamm Bowel Dis 15(7): 1090-1100, 2009.
    OpenUrlCrossRefPubMed
  173. ↵
    1. Uhlig HH,
    2. Powrie F
    : Mouse models of intestinal inflammation as tools to understand the pathogenesis of inflammatory bowel disease. Eur J Immunol 39(8): 2021-2026, 2009.
    OpenUrlCrossRefPubMed
  174. ↵
    1. Wu S,
    2. Rhee KJ,
    3. Albesiano E,
    4. Rabizadeh S,
    5. Wu X,
    6. Yen HR,
    7. Huso DL,
    8. Brancati FL,
    9. Wick E,
    10. McAllister F,
    11. Housseau F,
    12. Pardoll DM,
    13. Sears CL
    : A human colonic commensal promotes colon tumorigenesis via activation of T-helper type 17 T-cell responses. Nat Med 15(9): 1016-1022, 2009.
    OpenUrlCrossRefPubMed
  175. ↵
    1. Dang EV,
    2. Barbi J,
    3. Yang HY,
    4. Jinasena D,
    5. Yu H,
    6. Zheng Y,
    7. Bordman Z,
    8. Fu J,
    9. Kim Y,
    10. Yen HR,
    11. Luo W,
    12. Zeller K,
    13. Shimoda L,
    14. Topalian SL,
    15. Semenza GL,
    16. Dang CV,
    17. Pardoll DM,
    18. Pan F
    : Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1. Cell 146(5): 772-784, 2011.
    OpenUrlCrossRefPubMed
  176. ↵
    1. Mladenova D,
    2. Daniel JJ,
    3. Dahlstrom JE,
    4. Bean E,
    5. Gupta R,
    6. Pickford R,
    7. Currey N,
    8. Musgrove EA,
    9. Kohonen-Corish MR
    : The NSAID sulindac is chemopreventive in the mouse distal colon but carcinogenic in the proximal colon. Gut 60(3): 350-360, 2011.
    OpenUrlAbstract/FREE Full Text
  177. ↵
    1. Sparmann A,
    2. Bar-Sagi D
    : Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell 6(5): 447-458, 2004.
    OpenUrlCrossRefPubMed
  178. ↵
    1. Waugh DJ,
    2. Wilson C
    : The interleukin-8 pathway in cancer. Clin Cancer Res 14(21): 6735-6741, 2008.
    OpenUrlAbstract/FREE Full Text
  179. ↵
    1. Laiho P,
    2. Kokko A,
    3. Vanharanta S,
    4. Salovaara R,
    5. Sammalkorpi H,
    6. Jarvinen H,
    7. Mecklin JP,
    8. Karttunen TJ,
    9. Tuppurainen K,
    10. Davalos V,
    11. Schwartz S Jr..,
    12. Arango D,
    13. Makinen MJ,
    14. Aaltonen LA
    : Serrated carcinomas form a subclass of colorectal cancer with distinct molecular basis. Oncogene 26(2): 312-320, 2007.
    OpenUrlCrossRefPubMed
  180. ↵
    1. Sekirov I,
    2. Russell SL,
    3. Antunes LC,
    4. Finlay BB
    : Gut microbiota in health and disease. Physiol Rev 90(3): 859-904, 2010.
    OpenUrlAbstract/FREE Full Text
    1. Wang Z,
    2. Klipfell E,
    3. Bennett BJ,
    4. Koeth R,
    5. Levison BS,
    6. Dugar B,
    7. Feldstein AE,
    8. Britt EB,
    9. Fu X,
    10. Chung YM,
    11. Wu Y,
    12. Schauer P,
    13. Smith JD,
    14. Allayee H,
    15. Tang WH,
    16. DiDonato JA,
    17. Lusis AJ,
    18. Hazen SL
    : Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 472(7341): 57-63, 2011.
    OpenUrlCrossRefPubMed
  181. ↵
    1. Sudo N,
    2. Chida Y,
    3. Aiba Y,
    4. Sonoda J,
    5. Oyama N,
    6. Yu XN,
    7. Kubo C,
    8. Koga Y
    : Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. J Physiol 558(Pt 1): 263-275, 2004.
    OpenUrlCrossRefPubMed
  182. ↵
    1. Turnbaugh PJ,
    2. Ridaura VK,
    3. Faith JJ,
    4. Rey FE,
    5. Knight R,
    6. Gordon JI
    : The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med 1(6): 6ra14, 2009.
    OpenUrlAbstract/FREE Full Text
  183. ↵
    1. Vijay-Kumar M,
    2. Aitken JD,
    3. Carvalho FA,
    4. Cullender TC,
    5. Mwangi S,
    6. Srinivasan S,
    7. Sitaraman SV,
    8. Knight R,
    9. Ley RE,
    10. Gewirtz AT
    : Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science 328(5975): 228-231, 2010.
    OpenUrlAbstract/FREE Full Text
  184. ↵
    1. Garrett WS,
    2. Lord GM,
    3. Punit S,
    4. Lugo-Villarino G,
    5. Mazmanian SK,
    6. Ito S,
    7. Glickman JN,
    8. Glimcher LH
    : Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell 131(1): 33-45, 2007.
    OpenUrlCrossRefPubMed
  185. ↵
    1. Lepage P,
    2. Hasler R,
    3. Spehlmann ME,
    4. Rehman A,
    5. Zvirbliene A,
    6. Begun A,
    7. Ott S,
    8. Kupcinskas L,
    9. Dore J,
    10. Raedler A,
    11. Schreiber S
    : Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology 141(1): 227-236, 2011.
    OpenUrlCrossRefPubMed
  186. ↵
    1. Uronis JM,
    2. Muhlbauer M,
    3. Herfarth HH,
    4. Rubinas TC,
    5. Jones GS,
    6. Jobin C
    : Modulation of the intestinal microbiota alters colitis-associated colorectal cancer susceptibility. PLoS One 4(6): e6026, 2009.
    OpenUrlCrossRefPubMed
  187. ↵
    1. Chu FF,
    2. Esworthy RS,
    3. Chu PG,
    4. Longmate JA,
    5. Huycke MM,
    6. Wilczynski S,
    7. Doroshow JH
    : Bacteria-induced intestinal cancer in mice with disrupted Gpx1 and Gpx2 genes. Cancer Res 64(3): 962-968, 2004.
    OpenUrlAbstract/FREE Full Text
  188. ↵
    1. Roth KA,
    2. Kapadia SB,
    3. Martin SM,
    4. Lorenz RG
    : Cellular immune responses are essential for the development of Helicobacter felis-associated gastric pathology. J Immunol 163(3): 1490-1497, 1999.
    OpenUrlAbstract/FREE Full Text
    1. Mannick EE,
    2. Bravo LE,
    3. Zarama G,
    4. Realpe JL,
    5. Zhang XJ,
    6. Ruiz B,
    7. Fontham ET,
    8. Mera R,
    9. Miller MJ,
    10. Correa P
    : Inducible nitric oxide synthase, nitrotyrosine, and apoptosis in Helicobacter pylori gastritis: effect of antibiotics and antioxidants. Cancer Res 56(14): 3238-3243, 1996.
    OpenUrlAbstract/FREE Full Text
  189. ↵
    1. Correa P,
    2. Houghton J
    : Carcinogenesis of Helicobacter pylori. Gastroenterology 133(2): 659-672, 2007.
    OpenUrlCrossRefPubMed
  190. ↵
    1. Swidsinski A,
    2. Loening-Baucke V,
    3. Lochs H,
    4. Hale LP
    : Spatial organization of bacterial flora in normal and inflamed intestine: a fluorescence in situ hybridization study in mice. World J Gastroenterol 11(8): 1131-1140, 2005.
    OpenUrlCrossRefPubMed
  191. ↵
    1. Maeda H,
    2. Akaike T
    : Nitric oxide and oxygen radicals in infection, inflammation, and cancer. Biochemistry (Mosc) 63(7): 854-865, 1998.
    OpenUrlCrossRefPubMed
  192. ↵
    1. Fulton AM,
    2. Loveless SE,
    3. Heppner GH
    : Mutagenic activity of tumor-associated macrophages in Salmonella typhimurium strains TA98 and TA 100. Cancer Res 44(10): 4308-4311, 1984.
    OpenUrlAbstract/FREE Full Text
  193. ↵
    1. Maeda S,
    2. Kamata H,
    3. Luo JL,
    4. Leffert H,
    5. Karin M
    : IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 121(7): 977-990, 2005.
    OpenUrlCrossRefPubMed
  194. ↵
    1. Fausto N
    : Liver regeneration. J Hepatol 32(1 Suppl): 19-31, 2000.
    OpenUrlPubMed
    1. Kanneganti M,
    2. Mino-Kenudson M,
    3. Mizoguchi E
    : Animal models of colitis-associated carcinogenesis. J Biomed Biotechnol 2011: 342637, 2011.
    OpenUrlPubMed
    1. Fujii S,
    2. Fujimori T,
    3. Kawamata H,
    4. Takeda J,
    5. Kitajima K,
    6. Omotehara F,
    7. Kaihara T,
    8. Kusaka T,
    9. Ichikawa K,
    10. Ohkura Y,
    11. Ono Y,
    12. Imura J,
    13. Yamaoka S,
    14. Sakamoto C,
    15. Ueda Y,
    16. Chiba T
    : Development of colonic neoplasia in p53-deficient mice with experimental colitis induced by dextran sulphate sodium. Gut 53(5): 710-716, 2004.
    OpenUrlAbstract/FREE Full Text
    1. Tanaka T,
    2. Kohno H,
    3. Suzuki R,
    4. Hata K,
    5. Sugie S,
    6. Niho N,
    7. Sakano K,
    8. Takahashi M,
    9. Wakabayashi K
    : Dextran sodium sulfate strongly promotes colorectal carcinogenesis in ApcMin/+ mice: inflammatory stimuli by dextran sodium sulfate results in development of multiple colonic neoplasms. Int J Cancer 118(1): 25-34, 2006.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

In Vivo: 26 (4)
In Vivo
Vol. 26, Issue 4
July-August 2012
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on In Vivo.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Mouse Models of Inflammatory Bowel Disease - Insights into the Mechanisms of Inflammation-associated Colorectal Cancer
(Your Name) has sent you a message from In Vivo
(Your Name) thought you would like to see the In Vivo web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
1 + 0 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Mouse Models of Inflammatory Bowel Disease - Insights into the Mechanisms of Inflammation-associated Colorectal Cancer
DESSISLAVA MLADENOVA, MAIJA R. J. KOHONEN-CORISH
In Vivo Jul 2012, 26 (4) 627-646;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Mouse Models of Inflammatory Bowel Disease - Insights into the Mechanisms of Inflammation-associated Colorectal Cancer
DESSISLAVA MLADENOVA, MAIJA R. J. KOHONEN-CORISH
In Vivo Jul 2012, 26 (4) 627-646;
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Inflammation-associated Colorectal Cancer
    • Molecular Alterations in Inflammation-associated Colorectal Cancer
    • The Role of the Colonic Epithelial Barrier in Maintaining Gut Health
    • Mice with Dysregulation of the Immunosuppressive IL-10 or Transforming Growth Factor beta (TGF-β) Pathways
    • Mice with Defects in the Adaptive Immunity
    • Mice with Defects in Signal Transduction and Transcription Factors
    • The Role of DCs in Inflammation and Inflammation-associated Colorectal Carcinogenesis
    • Mouse Models with Chemically-induced Colitis and Cancer
    • Insights into the Mechanism of Carcinogenesis from the CAC Model
    • Is HIF an Unappreciated Target of Inflammation-associated CRC?
    • The Role of the Microbiota in Inflammation-associated CRC
    • Perspective and Conclusion
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Rectus Sheath Block (RSB) Analgesia Could Enhance Significantly the Patient Satisfaction Following Midline Laparotomy in Benign Disease and in Cancer: A Prospective Study With Special Reference to Nitrosative Stress Marker Nitrotyrosine (NT) Plasma Concentrations
  • Impact of red cell distribution width on future risk of cancer and all-cause mortality among cancer patients - the Tromso Study
  • HIF1{alpha} deficiency reduces inflammation in a mouse model of proximal colon cancer
  • Google Scholar

Similar Articles

In Vivo

© 2023 In Vivo

Powered by HighWire