Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Aug 22.
Published in final edited form as: Dig Dis Sci. 2011 Oct 8;56(12):3405–3420. doi: 10.1007/s10620-011-1885-6

Molecular Mechanism of Barrett’s Esophagus

Hao Chen 1, Yu Fang 1, Whitney Tevebaugh 1, Roy C Orlando 2, Nicholas J Shaheen 2, Xiaoxin Chen 1,2,3
PMCID: PMC3750118  NIHMSID: NIHMS496182  PMID: 21984436

Abstract

Barrett’s esophagus (BE) is defined as metaplastic conversion of esophageal squamous epithelium to intestinalized columnar epithelium. As a premalignant lesion of esophageal adenocarcinoma (EAC), it develops as a result of chronic gastroesophageal reflux disease (GERD). Many studies have been conducted to undertand the molecular mechanism of this disease. This review summarizes recent results of involving squamous transcription factors, intestinal transcription factors, signaling pathways, stromal factors, microRNAs, and other factors in the development of BE. A conceptual framework is proposed to guide future studies. We expect elucidation of the molecular mechanism of BE will help us develop proper management of GERD, BE, and EAC.

Keywords: Barrett’s esophagus, transcription factor, signaling pathway

Introduction

There are two major histological types of esophageal cancer, squamous cell carcinoma (ESCC) and adenocarcinoma (EAC). ESCC arises in squamous epithelial cells that line the esophagus, and it usually occurs in the upper and middle part of the esophagus. It is known that ESCC develops as a result of carcinogen exposure in a pathological sequence of hyperplasia, dysplasia and carcinoma. In contrast, EAC arises from metaplastic glandular tissue in the lower third of the esophagus. It is thought to develop as a result of long-term gastroesophageal reflux in a pathological sequence of reflux esophagitis (GERD), Barrett’s esophagus (BE), dysplasia and adenocarcinoma (1).

Baseline risk for EAC is quite low in the general population. It is 30–125 times higher in patients with BE (2, 3). Although there is still debate regarding whether BE is always needed for the development of EAC, recent studies have suggested that most, if not all, EAC develops from existing BE (4). On the other hand, BE is a common clinical identity, with as many as 3 million Americans harboring this lesion (5). The yearly risk for EAC in non-dysplastic BE is approximately 0.5% per person-year (6, 7), and the majority of subjects with this condition never progress to EAC. However, in the minority that do progress to cancer, EAC has a poor prognosis with a five-year survival rate of less than 15%, according to data from the NCI Surveillance Epidemiology and End Results. Our poor understanding of the pathogenesis of BE and EAC has limited our ability to stratify risk for BE among the enormous numbers of subjects with GERD, and treatments for the condition are limited. Therefore, it is very important to study the pathogenesis and molecular mechanism of BE in order to design evidence-based and scientifically sound interventions to prevent EAC.

Conceptual framework

In theory, BE, the metaplastic conversion of esophageal squamous epithelium to intestinalized columnar epithelium, may develop by two distinct mechanisms (8). One possibility is direct conversion of differentiated cells, a process called transdifferentiation. Alternatively, it may develop from altered differentiation of stem cells. In general, the stem cell theory is favored by most researchers, although there is no solid experimental evidence to exclude the possibility of transdifferentiation.

There are four potential cellular origins of BE in human esophagus, each supported by experimental data (9, 10). 1) Stem cells of squamous epithelium in the basal cell layer may undergo de novo metaplasia. The resulting metaplastic change produces stem cells for future BE. 2) Cells at the gastroesophageal junction or transitional zone may colonize the gastric cardia or distal esophagus in response to noxious luminal contents. 3) Stem cells in the neck of esophageal submucosal gland duct may colonize the esophagus when mucosal damage of the squamous epithelium takes place. 4) Bone marrow-derived stem cells may colonize the esophagus when there is inflammation or damage, and undergo metaplasia (1113).

At the cellular level, four major cell lineages are present in BE, columnar epithelial cells, Paneth cells, enteroendocrine cells, and goblet cells. Each individual cell lineage has a unique differentiation program as we have learned from intestinal development. These cells are expected to interact with each other and other cells in the stroma, such as inflammatory cells, fibroblasts.

At the molecular level, metaplasia is believed to be mediated by activation or inactivation of transcription factors (14). Therefore, transcription factors that regulate expression of target genes specific for certain cellular functions may be regarded as “drivers”. Genes involved in other functions such as celluar structures, metabolism and defense, are likely to be “passengers”. Through analysis of gene expression data some of these drivers and passengers can be fished out. The main challenge is to verify the true drivers. Because of the complexity and heterogeneity of the Barrett’s phenotype, we would expect: (1) multiple drivers may participate in the development of BE, yet some drivers may be essential and others dispensable; (2) drivers may regulate expression of themselves. To verify drivers, we would expect in vitro experiments help us distinguish drivers from passengers with gene expression, cellular morphology and functions as the end points. In vivo experiments are needed to verify true drivers with histopathology, gene expression and tissue functions as the end points. Besides the individual genes, several signaling pathways are known to be involved in BE. Linking signaling pathways and drivers into a molecular network is challenging, but making these connections will explain the molecular mechanism of BE.

With this conceptual framework in mind, we first analyzed microarray and SAGE datasets in the public databases with bioinformatics tools to identify potential drivers, passengers, and signaling pathways involved in the development of BE (15). Out of 14 published studies in the literature, three microarray datasets (two cDNA arrays and one oligo array) and one SAGE dataset met our criteria of data inclusion. SAM is used for identifying differentially expressed individual genes, SAGE (Poisson) for analysis of SAGE data, and GSEA for identifying an a priori defined set of genes differentially expressed in BE. These gene sets are either grouped according to a certain signaling pathway (GSEA curated), or the presence of a consensus binding sequence for a known transcripton factor in the promoter regions (GSEA motif). Fifty-five BE genes and thirteen NE (normal esophagus) genes were identified as differentially expressed genes (cutoff >4 fold) by both SAM and SAGE (Poisson). Using immunohistochemical staining, we further narrowed this list to four categories of genes: Category I contains 25 genes expressed in BE only; Category II contains 5 genes expressed in NE only; Category III contains 12 genes expressed more in BE than in NE; and Category IV contains 2 genes expressed more in NE than in BE. Furthermore, with immunohistochemical staining of human BE tissues, we confirmed loss or downregulation of transcription factors related to development of esophageal squamous epithelium (e.g., P63, Sox2, Pax9) and overexpression of transcription factors related to intestinal development (e.g., Cdx1, Cdx2, HNF1α, HNF3α, HNF3β, HNF3γ, HNF4α, GATA4, GATA6, Sox9, Math1). Besides individual genes, we also identified the TGFβ/BMP signaling pathway as an active pathway in BE.

Our data suggest that when esophageal epithelial stem cells are stimulated by gastroesophageal reflux, the squamous differentiation program may be inactivated through loss of expression of squamous transcription factors. Meanwhile, the columnar differentiation program may be activated through expression of intestinal transcription factors. These molecular events may lead to inactivation of squamous differentiation and activation of columnar differentiation, which produces four major cell lineages in BE: columnar epithelial cells, Paneth cells, enteroendocrine cells, and goblet cells (16). Since differentiation of each of these cell lineages and squamous epithelial cells may require different sets of drivers, the overall molecular mechanism is expected to be very complex, even before considering interactions among these cell lineages and the stroma.

1. Squamous transcription factors: P63, Sox2 and Pax9

P63 is a critical initiator of epithelial stratification and a key regulator of cell adhesion and survival of progenitor cells in squamous epithelium (1719). In mouse esophagus, when basal cells become differentiated, p63 is down-regulated and eventually turned off in terminally differentiated superficial cells (20, 21). In p63-deficient mice, embryonic esophageal epithelium appears columnar containing both ciliated and goblet-like cells (22). In human esophagus, p63 is expressed in the basal layer, suprabasal cell layer and submucosal glands of esophageal squamous epithelium (20, 23). When esophageal squamous epithelial cells are exposed to bile and acid, p63 becomes down-regulated (24), explaining why p63 is lost in BE (25).

Sox2 is a member of the Sry-like high mobility group domain protein family. It is expressed in the pharynx, esophagus, and stomach of chicken gut, but not in the lower gastrointestinal tract, where Cdx1 and Cdx2 are present (26, 27). Sox2 mutations are associated with esophageal atresia in anophthalmia-esophageal-genital syndrome (28). Sox2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinoma (29). Its down-regulation is associated with intestinal metaplasia in the stomach (30, 31). Mice with hypomorphic Sox2 developed mucus-producing cells in the esophagus, and had fewer p63-expressing cells. Interestingly, esophagus with a hypomorphic Sox2 expressed genes normally expressed in glandular stomach and intestine (TTF1, TFF1, TFF2, Agr2, etc.) (32). In fact, these genes which are negatively regulated by Sox2 were found to be overexpressed in human BE. These data suggest a potential role of loss of Sox2 in the development of BE. Indeed, Sox2 is not expressed in human BE or in a rat model (25). Although a repressive interaction between Cdx2 and Sox2 was found to occur at the prospective stomach-intestine border during development (33), Sox2 expression was upregulated in the intestinal metaplastic mucosa of Cdx2-transgenic mice (34). This study suggested that Cdx2 may mediate intestinal metaplasia by overriding the function of Sox2 without inactivating its expression.

Pax9 belongs to a group of nine transcription factors characterized by the presence of a DNA-binding “paired” domain. It is critical for development of mouse thymus, parathyroid, limbs, palate, teeth and vertebral column. In adult mice, its expression is confined to the tongue, esophagus, salivary glands and thymus (3537). Loss of Pax9 impairs terminal differentiation of squamous epithelial cells of mouse tongue (35). In humans, Pax9 is significantly reduced in dysplasia and squamous cell carcinoma, suggesting a role for Pax9 in the normal differentiation process of esophageal squamous epithelium (38). In human BE, Pax9 expression is significantly reduced (15), but it is unclear how down-regulation of Pax9 may contribute to BE.

2. Intestinal transcription factors

a. Caudal-related homeobox gene (Cdx1 and Cdx2)

As members of the Caudal-related homeobox gene family, Cdx1 and Cdx2 are critical for intestinal development. They have mostly overlapping, but also certain distinct, functions in intestinal development (39, 40). Relatively speaking, Cdx2 may play a dominant role in regulating intestinal cell differentiation (41). However, Cdx1 and Cdx2 may regulate expression of each other in a reciprocal manner (42).

Cdx2 plays an important regulatory role in the development of intestinal metaplasia in the foregut, and in colon cancer development (39, 43). Two independent studies have shown that stomach-specific Cdx2 transgenic overexpression induced intestinal metaplasia in the mouse stomach within weeks after birth (44, 45). Homozygous knockout of Cdx2 is embryonically lethal, and heterozygous knockout produces colonic harmatomas with squamous epithelium appearing in the colon (46). These studies suggest that Cdx2 might be a pivotal switch between intestinal columnar epithelium and squamous epithelium in the gastrointestinal tract.

Several lines of evidence have suggested an important role of Cdx2 in the development of human BE: 1) In normal intestinal epithelium, Cdx2 is expressed in most cell lineages with Paneth cells having a lower level of expression than other cells (39). Squamous epithelial cells of normal human esophagus do not express Cdx2, while submucosal glands weakly express Cdx2 in the cytoplasm. In human BE, Cdx2 is expressed in both goblet and non-goblet cells (47). Dysplasia and adenocarcinoma may have decreased levels of Cdx2, or even lose Cdx2 expression. In EAC, a high level of Cdx2 expression is usually associated with well or moderately differentiated tumors (4753).

2) A low level of Cdx2 mRNA was detectable by RT-PCR in biopsy samples of squamous epithelium of GERD patients, even before the appearance of Cdx2 protein, other marker genes, or histological metaplasia (48, 53).

3) Many “marker” genes of BE, such as villin, GCC, SI, are known to be transcriptionally regulated by Cdx2 (5458). The presence of goblet cells is diagnostic of human BE, and Cdx2 regulates expression of critical genes of goblet cell differentiation, such as Muc2 and TFF3 (59, 60). More importantly Cdx2 regulates expression of many transcription factors essential for intestinal development, such as Cdx1, Cdx2, Math1, Hox genes, KLF4, HNF1 β, HNF3β, HNF4α, GATA5, GATA6, and Hes1 (61). For example, Math1 (also called Atoh1 or Hath1) is a transcriptional target of Cdx2 (60), and plays a critical role in the development of goblet cells: Cdx2 transfection into IEC6 cells and human esophageal squamous epithelial cells upregulates Math1 (59, 62). Knockout of Hes1, a direct upstream negative regulator of Math1, leads to an increased number of goblet cells in mouse small intestine (63), whereas loss of Math1 completely prevents the development of goblet cells in mouse intestine. Development of Paneth cell and enteroendocrine cells is also suppressed (64).

4) Treatment of human and rodent esophageal squamous epithelial cells with either acid or bile acids, which mimics gastroesophageal reflux, induces expression of Cdx2 (59, 65, 66).

5) Transfection of Cdx2 into human esophageal squamous epithelial cells induces metaplastic changes in morphology and gene expression (59). HET1A cells with stable transfection of human Cdx2 form crypt-like structures in vitro. Microarray analysis and quantitative real-time PCR showed that stable transfection of Cdx2 up-regulated differentiation markers of intestinal columnar epithelial cells and goblet cells in HET1A cells. This may be partially due to modulation of Notch signaling pathway.

6) Transgenic overexpression of Cdx2 in mouse esophagus interferes with differentiation of normal esophageal squamous epithelial cells, and induces a potentially transitional cell type between squamous epithelial cells and columar epithelial cells (67).

Apart from Cdx2, Cdx1 is also involved in the development of human BE (68, 69), although Cdx2 expression precedes Cdx1 expression (70). Similar to Cdx2, transgenic overexpression of Cdx1 induces intestinal metaplasia in the stomach. However the Cdx1 phenotype is not exactly the same as Cdx2 phenotype (71). Cdx1 mRNA and protein are universally expressed in BE, but not in normal esophageal squamous epithelium. This tissue-specific expression is attributable to the methylation status of the Cdx1 promoter (68, 69).

Expression of Cdx1 and Cdx2 is regulated by promoter methylation and single nucleotide polymorphisms (SNPs):

  1. Promoter methylation is a well-known mechanism for gene silencing or activation. Embryonic mouse esophagus has the keratin 8 promoter methylated when columnar epithelium transdifferentiates into squamous epithelium during embryogeneis (72). Many genes on the TGFβ and WNT signaling pathways are frequently hyper- or hypomethylated in BE. Cdx1 and Cdx2, as well as GATA4, GATA6, Muc2, are all regulated by promoter methylation. When human esophageal epithelial cells are exposed to acid and/or bile treatment, activation of gene expression is commonly associated with reduced level of promoter methylation (59, 69). Even in normal esophagus, treatment with 5-aza-2′-deoxycytidine upregulates Cdx2 expression (67). Interestingly, loss of promoter methylation and activation of NFκB pathway synergize in inducing Cdx1 expression in human esophageal epithelial cells, when they are exposed to bile and proinflammatory cytokines (69).

  2. SNPs of Cdx1 and Cdx2 genes have not yet been well characterized. Two previous studies failed to associate several SNPs of Cdx2 with colon cancer (73, 74). These SNPs (IVS1+1020, IVS1+1476, IVS1−1575, IVS1−1042, IVS1−34, IVS1−33, IVS2−429, Exon3+877, and 3UTR+10) were mostly located in a short noncoding region. However, it is known that a roughly 9.5-kb 5′-flanking region from the human Cdx2 gene contained key cis elements for regulating transcription in colon cancer cells (75). Wong et al. sequenced a 400-bp region upstream of exon 1 and all three exons of Cdx1 in 37 colon cancer cell lines. Three SNPs were identified with one each one in the promoter, 5′UTR and coding region of exon 1. None of these were associated with Cdx1 expression in these cells (76). In fact, the major regulatory region of human Cdx1 gene is far upstream. Transgenic expression of the nucleotides −15,601 to +68 of the Cdx1 gene was restricted to the intestinal epithelium, which was identical to endogenous Cdx1 gene expression. DNase I hypersensitivity assays further narrowed two active chromatin regions at approximately −5.8 and −6.8 kb upstream of the Cdx1 gene, respectively (77, 78). Therefore, we believe that in order to find functionally important SNPs of Cdx1 and Cdx2 genes, larger areas in the regulatory regions need to be examined.

Two recent interesting observations deserve further studies. 1) Acid and bile treatment induced Cdx2 expression in esophageal squamous epithelial cells from patients with BE, but not from GERD patients without BE. This observation suggests that fine tuning of Cdx2 regulation in the esophagus might determine gene expression, and even susceptibility to BE (79). 2) miR-9 was found to regulate Cdx2 expression in gastric cancer cells (80), suggesting microRNAs may regulate, or be regulated by, Cdx2. Involvement of miRNAs addes a player of complexity to our understanding of the role of Cdx2 in BE.

b. Hepatocyte nuclear factors (HNFs)

HNFs are liver-enriched homeodomain-containing transcription factors that regulates many genes in the liver, pancreas and intestines (8183). HNF1α and 1β control terminal differentiation and cell fate commitment in the intestine (84). GATA4, HNF1α, and Cdx2 have been shown to play cooperative roles in regulating expression of marker genes in intestinal epithelium and human BE (55, 82, 85, 86). Exposure of esophageal epithelial cells to bile induced expression of Muc4 through HNF1α (87). HNF1α regulates expression of differentiation markers of columnar epithelial cells, such as FABP1 and lactase-phlorizine hydrolase (8891).

HNF3α is known to be amplified and overexpressed in human EAC (92). HNF3β plays a critical role in lung development (93). Knockout of HNF3β in the lung suppressed alveolarization and induced goblet cell hyperplasia (94). In the intestine, HNF3α and HNF3β regulate expression of Agr2 and Muc2, markers of goblet cells (9597).

HNF4α is essential for embryonic development as well as homeostasis and function of adult intestine (98). HNF4α knockout mice failed to form normal colonic crypts due to suppressed epithelial cell proliferation. Goblet cell maturation and expression of a set of genes were also perturbed (99, 100). HNF4α is regulated by HNF1α/β and GATA6 (101). In return, HNF1α expression is activated by HNF4α and HNF4β, which may mediate the stimulating effects of TGFβ pathway (102, 103).

c. GATA4 and GATA6

GATA4 and GATA6 belong to a subfamily of the GATA transcription factor family involved in differentiation of mesoderm and endoderm-derived tissues. They are expressed in the stomach and small intestine, but not in the esophagus (104, 105). GATA4 and GATA6 regulate expression of intestinal differential markers alone or in concert with other transcriptional factors such as HNFs, Cdx2, TTF1 (89, 106111). GATA4 regulates expression of HNF4 and is reciprocally regulated by TGFβ pathway (112, 113). GATA4 and the TGFβ signaling pathway cooperate in regulating intestinal epithelial gene expression (114), and GATA4/GATA6 activate BMP4 at the transcriptional level during early stages of embryogenesis (115). Normal esophagus does not express GATA4 and GATA6 (116). However, both are expressed in human BE and a rat model (15, 25). Aberrant promoter methylation and gene amplification of GATA4 and GATA6 have been reported in human EAC (117119).

3. Signaling pathways

a. TGFβ and BMP signaling pathway

An important recent finding is the increased expression of BMP4 and activation of its signaling pathway in BE and GERD. Treatment of primary squamous cell with BMP4 induced squamous dedifferentiation and columnar differentiation (120, 121). In the normal adult esophagus, BMP4 is not expressed. When stimulated by reflux, the stromal cells may produce inductive factors such as BMP4, which impact development and homeostasis of the overlying epithelium. This hypothesis is in agreement with the role of BMP signaling pathway in intestinal and esophageal development (122124).

Noggin, a BMP antagonist, is expressed in BE at a low level. It has been shown that 70% of homozygous noggin knockout mice developed esophageal atresia. Interestingly, reducing the gene dosage of BMP4 by 50% rescued the Noggin null phenotype (125). This study suggests that fine tuning of BMP4 expression has a critical role on esophageal morphogenesis. When BMP4 signaling is inhibited in the mouse intestine by transgenic overexpression of Noggin, villus morphogenesis is suppressed in the small intestine, and crypts become highly proliferative. Nevertheless, goblet cells, enteroendocrine cells and Paneth cells are normally differentiated (126).

Although EAC tends to have decreased levels of TGFβRII, Smad2 and Smad4 through promoter methylation (127, 128), it may not be the same at the stage of BE. Cdx2 was found to interact with SMAD3 independent of SMAD4, resulting stimulation of SMAD3 transcriptional activity. Cdx1 also interacts with SMAD3 by inhibiting the SMAD3/SMAD4-dependent transcription (129).

b. WNT signaling pathway

The WNT signaling pathway is well known for its critical role in gastrointestinal development and cancer development (130132). Many studies have clearly shown than the WNT signaling pathway and its key component β-catenin are involved in the development of EAC. Targeting this pathway may have therapeutic effects on EAC (133). Although mutations of APC and β-catenin are rare in EAC, promoter hypermethylation (APC, SFRP1), downregulation of E-cadherin, nuclear translocation of β-catenin and WNT2 overexpression, are commonly seen in EAC (134136).

Cdx1 and Cdx2 are downstream effectors of the activated WNT pathway that mediate intestinal metaplasia. Cdx1 is transcriptionally regulated by the WNT pathway (137, 138). In the stomach, CagA physically interacts with E-cadherin and impairs the complex formation between E-cadherin and β-catenin, causing cytoplasmic and nuclear accumulation of β-catenin. As a result, Cdx1 and a goblet cell marker (Muc2) are activated (139). In endometrial cancer cell lines, overexpression of an active form β-catenin resulted in a significant increase in endogenous Cdx2 expression, independent of TCF4. Cells overexpressing exogenous Cdx2 may inhibit β-catenin/TCF4-mediated transcriptional activation of target genes probably through indirect mechanisms (140). In the lung, transgenic overexpression of β-catenin disturbed normal development, and induced strong expression of Cdx1, Math1 and other genes characteristic of intestinal Paneth, goblet cells, and non-lung secretary cell types (141).

As a downstream target of WNT signaling pathway, Sox9 exerts negative feedback on the WNT pathway and Cdx2 (142144). Sox9 is known to be expressed in the intestinal epithelium, and specifically in stem cells and Paneth cells. Knockout of Sox9 affected differentiation throughout the intestinal epithelium with a disappearance of Paneth cells. Other cell lineages (goblet, enteroendocrine, columnar cells) were more or less affected (142, 145). Sox9 expression is also modulated by the TGFβ pathway (146), the NFκB pathway (147), and GATA4/FOG2 complex (148). A recent study has clearly shown a potential role of Sox9 in the development of BE (149).

SAM pointed domain-containing Ets transcription factor (Spdef) is a transcription factor responsive to Wnt signaling that has been found to regulate goblet cell differentiation in the airway (150, 151). A recent study on Spdef knockout mice demonstrated its involvement in maturation of goblet and Paneth cells in the intestine (152). In fact, Spdef was found to be overexpressed in BE as well (unpublished data). These data suggested that Wnt signaling plays a potentially critical role in the development of human BE through downstream effectors many of which are transcription factors.

c. NFκB signaling pathway

Corresponding to increased levels of IL8 and IL1β, there is a step-wise increase of NFκB activity in human GERD, BE, dysplasia and EAC (153). As a consequence, many downstream genes may be activated and contribute to BE (154, 155). Gastroesophageal reflux is known to activate NFκB pathway and induce proinflammatory cytokines in esophageal epithelial cells (156). Accumulating evidence has shown that NFkB, when activated, upregulates Cdx1 and Cdx2 expression. Two independent studies have shown that induction of Cdx2 expression in esophageal epithelial cells by bile was mediated by binding sites in the proximal promoter for NFκB (66, 79, 157). Not only Cdx2, but also Cdx1 is regulated in a similar way by NFκB pathway in the esophageal epithelial cells when stimulated by gastroesophageal reflux (69). Similar to regulation in the esophagus, bacterial components may upregulate Cdx2 and Muc2 expression in rat biliary epithelial cells through Toll-like receptors and the NFkB pathway. This mechanism may contribute to induction of intestinal metaplasia in the bile duct by bacterial infection (158).

d. Hedgehog signaling pathway

Hedgehog signaling pathway was initially found to play a critical role in foregut development (159, 160). It was then found to be involved in upper gastrointestinal cancer (161, 162). A recent study has shown that Hedgehog ligand expression can be induced by acid or bile exposure in BE epithelium. Transgenic expression of Sonic hedgehog in mouse esophageal epithelium induces expression of stromal Bmp4, epithelial Sox9, and columnar cytokeratins (149). An independent study has verified an association among Sonic hedgehog, BMP4, and Cdx2 in human BE tissues (163).

e. Notch signaling pathway

Notch signaling pathway is known to play a critical role in intestinal development. Genetic disruption of Notch signaling results in increased number of goblet cells in mouse small intestine (63, 164). Similary, blocking Notch signaling pathway with γ-secretase inhibitors enhances the development of goblet cells in the intestine (165, 166). This effect depends on the presence of Math1 (167). Ectopic Notch signaling in adult intestinal progenitor cells leads to bias against secretory fates (168).

In the esophagus, Notch1 and Notch3 coordinated esophageal squamous differentiation (169). Its interaction with p63 suggests that Notch signling pathway might promote squamous epithelial cell differentiation, while p63 maintains the progenitor cell population (170). In our previous study, a set of genes on the Notch signaling pathway were modulated by Cdx2 transfection into HET1A cells, with JAG1, Notch4, Notch3, PSEN2, MSI1, Math1, and DF up-regulated and Hes1 downregulated (59).Treatment with a bile acid was found to inhibit Notch signaling in human EAC cells (171). Immunohistochemistry confirmed the presence of an intact and activated Notch signaling pathway in metaplastic BE epithelium, but not in the normal human esophagus. Treatment of a rat model of reflux-induced BE with a γ-secretase inhibitor converted the proliferative BE epithelial cells into terminally differentiated goblet cells, whereas the squamous epithelium remained intact (172). These data suggest that inhibition of Notch signaling pathways favors goblet cell differentiation and potentially promote BE.

4. Stromal factors

Epithelial-mesenchymal interactions play a critical role in epithelial cell differentiation. Manipulation of stromal factors has been implicated in the development of BE (173, 174) and EAC (175). One of the most well-studied aspects is the association between inflammation and BE (176, 177). Pro-inflammatory cytokines are associated with the development of human BE (153, 178, 179). A recent study on the surgical rat model showed that inflammatory changes preceded histopathology of caustic damage in reflux esophagitis. Chemokines like IL8 and IL1β were secreted by esophageal squamous epithelial cells to mediate esophagitis (180). This observation is consistent with a recent study showing transgenic overexpression of IL1β in the squamous epithelium of mouse esophagus and forestomach causes spontaneous inflammation and intestinal metaplasia at the squamocolumnar junction in the stomach. Intestinal metaplasia was characterized by expression of TFF2 and Cdx2, and promoted by exposure to bile acid (181).

Extracellular matrix exerts a significant impact on differentiation of esophageal epithelial cells. Squamous esophageal epithelial cells cultured on connective tissue from the skin failed to show the expected pattern of differentiation (182). Furthermore, conversion of embryonic stem cells to columnar or squamous epithelia has been shown to depend on the components of the extracellular matrix (183). Cdx2 expression in colon cancer cells is adaptable and strongly dependent on the surrounding extracellular matrix (184, 185).

5. microRNAs

Several studies have been published to show changes in the microRNA profile of human BE and EAC (186194). Although the major goal of these studies was to identify biomarkers and mechanism of EAC, some data showed an involvement of microRNAs in the development of BE. Certain microRNAs (e.g., miR-203) are known to modulate expression of critical genes associated with BE (e.g., p63). On the other hand, transcription factors including p63 are known to modulate processing of microRNAs such as miR-21 (195, 196). It would be interesting to determine whether microRNAs are drivers or passengers in the development of BE.

6. Other factors

Retinoic acid is required for diverse developmental programs (197). Retinoic acid alone or in synergy with WNT signaling activates Cdx1 (198, 199), HNF1α and HNF4α (200), GATA4 (201), and Sox9 (202). Interestingly, the level of retinoic acid increased in BE tissue compared to normal esophagus, and then decreased in EAC (203, 204). Expression of retinoic acid receptors and retinoid X receptors was also altered in BE compared to normal esophagus (205, 206). More importantly, bile treatment increases retinoic acid receptor activity, which further induces columnar differentiation in esophageal epithelial cells (207, 208).

Runt-related transcriptional factor gene 3 (RUNX3) belongs to the runt domain family of transcriptional factors that plays an important role in celluar differentiation of the esophagus (209). Frequent silencing of RUNX3 by promoter methylation has been reported in both BE and EAC (210212). As in the esophagus, RUNX3 also plays an important role in the development of the stomach (213). Interestingly, RUNX3 attenuated Wnt signaling in the intestine and interacted with TGFβ signaling (214, 215). RUNX3 is frequently silenced in gastric cancer through promoter methylation or protein mislocalization (216, 217), and loss of RUNX3 allows gastric epithelial cells to differentiate into intestinal cell types (218, 219), suggesting that loss of RUNX3 may play a similar role in the development of BE.

Krüppel-like factor 4 and 5 (KLF4 and KLF5) are zinc-finger transcription factors that are expressed in the epithelial tissues. KLF4 is required for terminal differentiation of goblet cells in the colon (220). It is known to be regulated by transcription factors (TCF4 and Sox9) and the WNT signaling pathway in the intestine (221). The Notch signaling pathway negatively regulates KLF4 expression, and Notch inhibition increases KLF4 expression and goblet cell differentiation in the intestine (222, 223). KLF4 was recently found to be involved in TGFβ signaling as well (224). In the esophagus, KLF4 is essential for squamous epithelial differentiation and interacts with KLF5 to maintain normal epithelial homeostasis (225, 226). In a recent study, KLF4 was found to be expressed in Barrett’s epithelium, and its expression was shown to respond to bile acids through the NFκB signaling pathway. More interestingly, KLF4 interacted with Cdx2 through transcriptional regulation (227).

Summary

In summary, development of BE is complex at the molecular level. As we have discussed above, this process involves squamous transcription factors, intestinal transcription factors, signaling pathways, microRNAs, SNPs, epigenetics, and many other factors. It is challenging to distinguish drivers from passengers, and although a molecular network has started to emerge, we do not yet fully understand how gastroesophageal reflux may induce intestinal metaplasia in the esophagus.

In vivo animal models are essential for validation of drivers of BE. K14-Cdx2 transgenic mice failed to generate Barrett’s phenotype in mouse esophagus (67). Several reasons may explain this observation. Expression level of the transgene may not be optimal due to chromatin remodeling and promoter strength. Cdx2 overexpression alone may not be able to drive intestinal metaplasia. Cooperation with other factors (e.g., loss of p63) may be needed. K14 promoter is only active in squamous epithelial cells, and therefore may not be able to drive the whole process of intestinal metaplasia. Finally, rodent esophagus is covered by keratinized squamous epithelium, which is different from the non-keratinized squamous epithelium of human esophagus. It is unclear how such a difference between species may impact the value and use of animal models in BE.

Acknowledgments

Grant support: NIH U54 CA156735 and NCBC 2011-MRG-1101

Abbreviations

BE

Barrett’s esophagus

BMP

bone morphogenetic protein

EAC

esophageal adenocarcinoma

GERD

gastroesophageal reflux disease

HNF

hepatocyte nuclear factor

KLF

Krüppel-like factor

RUNX3

runt-related transcriptional factor 3

SNP

single nucleotide polymorphism

Spdef

SAM pointed domain-containing Ets transcription factor

TGF

transforming growth factor

Footnotes

Disclosure: The authors have no financial arrangements related to this manuscript to disclose.

References

  • 1.Siersema PD. Pathogenesis, diagnosis and therapeutic possibilities of esophageal cancer. Curr Opin Gastroenterol. 2007;23:456–61. doi: 10.1097/MOG.0b013e32816a91de. [DOI] [PubMed] [Google Scholar]
  • 2.O’Connor JB, Falk GW, Richter JE. The incidence of adenocarcinoma and dysplasia in Barrett’s esophagus: report on the Cleveland Clinic Barrett’s Esophagus Registry. Am J Gastroenterol. 1999;94:2037–42. doi: 10.1111/j.1572-0241.1999.01275.x. [DOI] [PubMed] [Google Scholar]
  • 3.Drewitz DJ, Sampliner RE, Garewal HS. The incidence of adenocarcinoma in Barrett’s esophagus: a prospective study of 170 patients followed 4. 8 years. Am J Gastroenterol. 1997;92:212–5. [PubMed] [Google Scholar]
  • 4.Chandrasoma P, Wickramasinghe K, Ma Y, DeMeester T. Is intestinal metaplasia a necessary precursor lesion for adenocarcinomas of the distal esophagus, gastroesophageal junction and gastric cardia? Dis Esophagus. 2007;20:36–41. doi: 10.1111/j.1442-2050.2007.00638.x. [DOI] [PubMed] [Google Scholar]
  • 5.Fennerty MB, Triadafilopoulos G. Barrett’s-related esophageal adenocarcinoma: is chemoprevention a potential option? Am J Gastroenterol. 2001;96:2302–5. doi: 10.1111/j.1572-0241.2001.04033.x. [DOI] [PubMed] [Google Scholar]
  • 6.Spechler SJ. Barrett’s esophagus and esophageal adenocarcinoma: pathogenesis, diagnosis, and therapy. Med Clin North Am. 2002;86:1423–45. vii. doi: 10.1016/s0025-7125(02)00082-2. [DOI] [PubMed] [Google Scholar]
  • 7.Shaheen NJ, Crosby MA, Bozymski EM, Sandler RS. Is there publication bias in the reporting of cancer risk in Barrett’s esophagus? Gastroenterology. 2000;119:333–8. doi: 10.1053/gast.2000.9302. [DOI] [PubMed] [Google Scholar]
  • 8.Fitzgerald RC. Molecular basis of Barrett’s oesophagus and oesophageal adenocarcinoma. Gut. 2006;55:1810–20. doi: 10.1136/gut.2005.089144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Shaheen NJ. Advances in Barrett’s esophagus and esophageal adenocarcinoma. Gastroenterology. 2005;128:1554–66. doi: 10.1053/j.gastro.2005.03.032. [DOI] [PubMed] [Google Scholar]
  • 10.Jankowski JA, Harrison RF, Perry I, Balkwill F, Tselepis C. Barrett’s metaplasia. Lancet. 2000;356:2079–85. doi: 10.1016/S0140-6736(00)03411-5. [DOI] [PubMed] [Google Scholar]
  • 11.Hutchinson L, Stenstrom B, Chen D, et al. Human Barrett’s adenocarcinoma of the esophagus, associated myofibroblasts, and endothelium can arise from bone marrow-derived cells after allogeneic stem cell transplant. Stem Cells Dev. 2010;20:11–7. doi: 10.1089/scd.2010.0139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sarosi G, Brown G, Jaiswal K, et al. Bone marrow progenitor cells contribute to esophageal regeneration and metaplasia in a rat model of Barrett’s esophagus. Dis Esophagus. 2008;21:43–50. doi: 10.1111/j.1442-2050.2007.00744.x. [DOI] [PubMed] [Google Scholar]
  • 13.Li Y, Wo JM, Ellis S, Ray MB, Jones W, Martin RC. Morphological transformation in esophageal submucosa by bone marrow cells: esophageal implantation under external esophageal perfusion. Stem Cells Dev. 2006;15:697–705. doi: 10.1089/scd.2006.15.697. [DOI] [PubMed] [Google Scholar]
  • 14.Slack JM, Tosh D. Transdifferentiation and metaplasia--switching cell types. Curr Opin Genet Dev. 2001;11:581–6. doi: 10.1016/s0959-437x(00)00236-7. [DOI] [PubMed] [Google Scholar]
  • 15.Wang J, Qin R, Ma Y, et al. Differential gene expression in normal esophagus and Barrett’s esophagus. J Gastroenterol. 2009 doi: 10.1007/s00535-009-0082-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chen X, Yang CS. Barrett’s esophagus: Preclinical Models for Investigation. In: Jobe BA, Thomas CR, Hunter JG, editors. Esophageal Cancer Principles and Practice. New York: Demos Medical; 2009. pp. 61–8. [Google Scholar]
  • 17.Koster MI, Kim S, Mills AA, DeMayo FJ, Roop DR. p63 is the molecular switch for initiation of an epithelial stratification program. Genes Dev. 2004;18:126–31. doi: 10.1101/gad.1165104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Carroll DK, Carroll JS, Leong CO, et al. p63 regulates an adhesion programme and cell survival in epithelial cells. Nat Cell Biol. 2006;8:551–61. doi: 10.1038/ncb1420. [DOI] [PubMed] [Google Scholar]
  • 19.Koster MI, Kim S, Roop DR. P63 deficiency: a failure of lineage commitment or stem cell maintenance? J Investig Dermatol Symp Proc. 2005;10:118–23. doi: 10.1111/j.1087-0024.2005.200416.x. [DOI] [PubMed] [Google Scholar]
  • 20.Glickman JN, Yang A, Shahsafaei A, McKeon F, Odze RD. Expression of p53-related protein p63 in the gastrointestinal tract and in esophageal metaplastic and neoplastic disorders. Hum Pathol. 2001;32:1157–65. doi: 10.1053/hupa.2001.28951. [DOI] [PubMed] [Google Scholar]
  • 21.Daniely Y, Liao G, Dixon D, et al. Critical role of p63 in the development of a normal esophageal and tracheobronchial epithelium. Am J Physiol Cell Physiol. 2004;287:C171–81. doi: 10.1152/ajpcell.00226.2003. [DOI] [PubMed] [Google Scholar]
  • 22.Yang A, Schweitzer R, Sun D, et al. p63 is essential for regenerative proliferation in limb, craniofacial and epithelial development. Nature. 1999;398:714–8. doi: 10.1038/19539. [DOI] [PubMed] [Google Scholar]
  • 23.Geddert H, Kiel S, Heep HJ, Gabbert HE, Sarbia M. The role of p63 and deltaNp63 (p40) protein expression and gene amplification in esophageal carcinogenesis. Hum Pathol. 2003;34:850–6. doi: 10.1016/s0046-8177(03)00342-3. [DOI] [PubMed] [Google Scholar]
  • 24.Roman S, Petre A, Thepot A, et al. Downregulation of p63 upon exposure to bile salts and acid in normal and cancer esophageal cells in culture. Am J Physiol Gastrointest Liver Physiol. 2007;293:G45–53. doi: 10.1152/ajpgi.00583.2006. [DOI] [PubMed] [Google Scholar]
  • 25.Chen X, Qin R, Liu B, et al. Multilayered epithelium in a rat model and human Barrett’s esophagus: Expression patterns of transcription factors and differentiation markers. BMC Gastroenterol. 2008;8:1. doi: 10.1186/1471-230X-8-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ishii Y, Rex M, Scotting PJ, Yasugi S. Region-specific expression of chicken Sox2 in the developing gut and lung epithelium: regulation by epithelial-mesenchymal interactions. Dev Dyn. 1998;213:464–75. doi: 10.1002/(SICI)1097-0177(199812)213:4<464::AID-AJA11>3.0.CO;2-Z. [DOI] [PubMed] [Google Scholar]
  • 27.Kamachi Y, Uchikawa M, Kondoh H. Pairing SOX off: with partners in the regulation of embryonic development. Trends Genet. 2000;16:182–7. doi: 10.1016/s0168-9525(99)01955-1. [DOI] [PubMed] [Google Scholar]
  • 28.Williamson KA, Hever AM, Rainger J, et al. Mutations in SOX2 cause anophthalmia-esophageal-genital (AEG) syndrome. Hum Mol Genet. 2006;15:1413–22. doi: 10.1093/hmg/ddl064. [DOI] [PubMed] [Google Scholar]
  • 29.Bass AJ, Watanabe H, Mermel CH, et al. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat Genet. 2009;41:1238–42. doi: 10.1038/ng.465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tsukamoto T, Inada K, Tanaka H, et al. Down-regulation of a gastric transcription factor, Sox2, and ectopic expression of intestinal homeobox genes, Cdx1 and Cdx2: inverse correlation during progression from gastric/intestinal-mixed to complete intestinal metaplasia. J Cancer Res Clin Oncol. 2004;130:135–45. doi: 10.1007/s00432-003-0519-6. [DOI] [PubMed] [Google Scholar]
  • 31.Tsukamoto T, Mizoshita T, Mihara M, et al. Sox2 expression in human stomach adenocarcinomas with gastric and gastric-and-intestinal-mixed phenotypes. Histopathology. 2005;46:649–58. doi: 10.1111/j.1365-2559.2005.02170.x. [DOI] [PubMed] [Google Scholar]
  • 32.Que J, Okubo T, Goldenring JR, et al. Multiple dose-dependent roles for Sox2 in the patterning and differentiation of anterior foregut endoderm. Development. 2007;134:2521–31. doi: 10.1242/dev.003855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sherwood RI, Chen TY, Melton DA. Transcriptional dynamics of endodermal organ formation. Dev Dyn. 2009;238:29–42. doi: 10.1002/dvdy.21810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Mutoh H, Sashikawa M, Sugano K. Sox2 expression is maintained while gastric phenotype is completely lost in Cdx2-induced intestinal metaplastic mucosa. Differentiation. 2010;81:92–8. doi: 10.1016/j.diff.2010.10.002. [DOI] [PubMed] [Google Scholar]
  • 35.Jonker L, Kist R, Aw A, Wappler I, Peters H. Pax9 is required for filiform papilla development and suppresses skin-specific differentiation of the mammalian tongue epithelium. Mech Dev. 2004;121:1313–22. doi: 10.1016/j.mod.2004.07.002. [DOI] [PubMed] [Google Scholar]
  • 36.Peters H, Neubuser A, Kratochwil K, Balling R. Pax9-deficient mice lack pharyngeal pouch derivatives and teeth and exhibit craniofacial and limb abnormalities. Genes Dev. 1998;12:2735–47. doi: 10.1101/gad.12.17.2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Peters H, Schuster G, Neubuser A, Richter T, Hofler H, Balling R. Isolation of the Pax9 cDNA from adult human esophagus. Mamm Genome. 1997;8:62–4. doi: 10.1007/s003359900351. [DOI] [PubMed] [Google Scholar]
  • 38.Gerber JK, Richter T, Kremmer E, et al. Progressive loss of PAX9 expression correlates with increasing malignancy of dysplastic and cancerous epithelium of the human oesophagus. J Pathol. 2002;197:293–7. doi: 10.1002/path.1115. [DOI] [PubMed] [Google Scholar]
  • 39.Silberg DG, Swain GP, Suh ER, Traber PG. Cdx1 and cdx2 expression during intestinal development. Gastroenterology. 2000;119:961–71. doi: 10.1053/gast.2000.18142. [DOI] [PubMed] [Google Scholar]
  • 40.Calon A, Gross I, Lhermitte B, et al. Different effects of the Cdx1 and Cdx2 homeobox genes in a murine model of intestinal inflammation. Gut. 2007 doi: 10.1136/gut.2007.125542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Verzi MP, Shin H, Ho LL, Liu XS, Shivdasani RA. Essential and redundant functions of Caudal family proteins in activating adult intestinal genes. Mol Cell Biol. 2011 doi: 10.1128/MCB.01250-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kazumori H, Ishihara S, Kinoshita Y. Roles of caudal-related homeobox gene Cdx1 in oesophageal epithelial cells in Barrett’s epithelium development. Gut. 2009;58:620–8. doi: 10.1136/gut.2008.152975. [DOI] [PubMed] [Google Scholar]
  • 43.Guo RJ, Suh ER, Lynch JP. The role of cdx proteins in intestinal development and cancer. Cancer Biol Ther. 2004;3:593–601. doi: 10.4161/cbt.3.7.913. [DOI] [PubMed] [Google Scholar]
  • 44.Mutoh H, Hakamata Y, Sato K, et al. Conversion of gastric mucosa to intestinal metaplasia in Cdx2-expressing transgenic mice. Biochem Biophys Res Commun. 2002;294:470–9. doi: 10.1016/S0006-291X(02)00480-1. [DOI] [PubMed] [Google Scholar]
  • 45.Silberg DG, Sullivan J, Kang E, et al. Cdx2 ectopic expression induces gastric intestinal metaplasia in transgenic mice. Gastroenterology. 2002;122:689–96. doi: 10.1053/gast.2002.31902. [DOI] [PubMed] [Google Scholar]
  • 46.Tamai Y, Nakajima R, Ishikawa T, Takaku K, Seldin MF, Taketo MM. Colonic hamartoma development by anomalous duplication in Cdx2 knockout mice. Cancer Res. 1999;59:2965–70. [PubMed] [Google Scholar]
  • 47.Groisman GM, Amar M, Meir A. Expression of the intestinal marker Cdx2 in the columnar-lined esophagus with and without intestinal (Barrett’s) metaplasia. Mod Pathol. 2004;17:1282–8. doi: 10.1038/modpathol.3800182. [DOI] [PubMed] [Google Scholar]
  • 48.Eda A, Osawa H, Satoh K, et al. Aberrant expression of CDX2 in Barrett’s epithelium and inflammatory esophageal mucosa. J Gastroenterol. 2003;38:14–22. doi: 10.1007/s005350300001. [DOI] [PubMed] [Google Scholar]
  • 49.Werling RW, Yaziji H, Bacchi CE, Gown AM. CDX2, a highly sensitive and specific marker of adenocarcinomas of intestinal origin: an immunohistochemical survey of 476 primary and metastatic carcinomas. Am J Surg Pathol. 2003;27:303–10. doi: 10.1097/00000478-200303000-00003. [DOI] [PubMed] [Google Scholar]
  • 50.Phillips RW, Frierson HF, Jr, Moskaluk CA. Cdx2 as a marker of epithelial intestinal differentiation in the esophagus. Am J Surg Pathol. 2003;27:1442–7. doi: 10.1097/00000478-200311000-00006. [DOI] [PubMed] [Google Scholar]
  • 51.Moskaluk CA, Zhang H, Powell SM, Cerilli LA, Hampton GM, Frierson HF., Jr Cdx2 protein expression in normal and malignant human tissues: an immunohistochemical survey using tissue microarrays. Mod Pathol. 2003;16:913–9. doi: 10.1097/01.MP.0000086073.92773.55. [DOI] [PubMed] [Google Scholar]
  • 52.Kaimaktchiev V, Terracciano L, Tornillo L, et al. The homeobox intestinal differentiation factor CDX2 is selectively expressed in gastrointestinal adenocarcinomas. Mod Pathol. 2004;17:1392–9. doi: 10.1038/modpathol.3800205. [DOI] [PubMed] [Google Scholar]
  • 53.Moons LM, Bax DA, Kuipers EJ, et al. The homeodomain protein CDX2 is an early marker of Barrett’s oesophagus. J Clin Pathol. 2004;57:1063–8. doi: 10.1136/jcp.2003.015727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Park J, Schulz S, Waldman SA. Intestine-specific activity of the human guanylyl cyclase C promoter is regulated by Cdx2. Gastroenterology. 2000;119:89–96. doi: 10.1053/gast.2000.8520. [DOI] [PubMed] [Google Scholar]
  • 55.Boudreau F, Rings EH, van Wering HM, et al. Hepatocyte nuclear factor-1 alpha, GATA-4, and caudal related homeodomain protein Cdx2 interact functionally to modulate intestinal gene transcription. Implication for the developmental regulation of the sucrase-isomaltase gene. J Biol Chem. 2002;277:31909–17. doi: 10.1074/jbc.M204622200. [DOI] [PubMed] [Google Scholar]
  • 56.Braunstein EM, Qiao XT, Madison B, Pinson K, Dunbar L, Gumucio DL. Villin: A marker for development of the epithelial pyloric border. Dev Dyn. 2002;224:90–102. doi: 10.1002/dvdy.10091. [DOI] [PubMed] [Google Scholar]
  • 57.Mesquita P, Jonckheere N, Almeida R, et al. Human MUC2 mucin gene is transcriptionally regulated by Cdx homeodomain proteins in gastrointestinal carcinoma cell lines. J Biol Chem. 2003;278:51549–56. doi: 10.1074/jbc.M309019200. [DOI] [PubMed] [Google Scholar]
  • 58.Shimada T, Koike T, Yamagata M, Yoneda M, Hiraishi H. Regulation of TFF3 expression by homeodomain protein CDX2. Regul Pept. 2007;140:81–7. doi: 10.1016/j.regpep.2006.11.014. [DOI] [PubMed] [Google Scholar]
  • 59.Liu T, Zhang X, So CK, et al. Regulation of Cdx2 expression by promoter methylation, and effects of Cdx2 transfection on morphology and gene expression of human esophageal epithelial cells. Carcinogenesis. 2007;28:488–96. doi: 10.1093/carcin/bgl176. [DOI] [PubMed] [Google Scholar]
  • 60.Mutoh H, Sakamoto H, Hayakawa H, et al. The intestine-specific homeobox gene Cdx2 induces expression of the basic helix-loop-helix transcription factor Math1. Differentiation. 2006;74:313–21. doi: 10.1111/j.1432-0436.2006.00074.x. [DOI] [PubMed] [Google Scholar]
  • 61.Boyd M, Hansen M, Jensen TG, et al. Genome-wide analysis of CDX2 binding in intestinal epithelial cells (Caco-2) J Biol Chem. 2010;285:25115–25. doi: 10.1074/jbc.M109.089516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Uesaka T, Kageyama N, Watanabe H. Identifying target genes regulated downstream of Cdx2 by microarray analysis. J Mol Biol. 2004;337:647–60. doi: 10.1016/j.jmb.2004.01.061. [DOI] [PubMed] [Google Scholar]
  • 63.Jensen J, Pedersen EE, Galante P, et al. Control of endodermal endocrine development by Hes-1. Nat Genet. 2000;24:36–44. doi: 10.1038/71657. [DOI] [PubMed] [Google Scholar]
  • 64.Yang Q, Bermingham NA, Finegold MJ, Zoghbi HY. Requirement of Math1 for secretory cell lineage commitment in the mouse intestine. Science. 2001;294:2155–8. doi: 10.1126/science.1065718. [DOI] [PubMed] [Google Scholar]
  • 65.Marchetti M, Caliot E, Pringault E. Chronic acid exposure leads to activation of the cdx2 intestinal homeobox gene in a long-term culture of mouse esophageal keratinocytes. J Cell Sci. 2003;116:1429–36. doi: 10.1242/jcs.00338. [DOI] [PubMed] [Google Scholar]
  • 66.Kazumori H, Ishihara S, Rumi MA, Kadowaki Y, Kinoshita Y. Bile acids directly augment caudal related homeobox gene Cdx2 expression in oesophageal keratinocytes in Barrett’s epithelium. Gut. 2006;55:16–25. doi: 10.1136/gut.2005.066209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kong J, Crissey MA, Funakoshi S, Kreindler JL, Lynch JP. Ectopic Cdx2 Expression in Murine Esophagus Models an Intermediate Stage in the Emergence of Barrett’s Esophagus. PLoS ONE. 2011;6:e18280. doi: 10.1371/journal.pone.0018280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Silberg DG, Furth EE, Taylor JK, Schuck T, Chiou T, Traber PG. CDX1 protein expression in normal, metaplastic, and neoplastic human alimentary tract epithelium. Gastroenterology. 1997;113:478–86. doi: 10.1053/gast.1997.v113.pm9247467. [DOI] [PubMed] [Google Scholar]
  • 69.Wong NA, Wilding J, Bartlett S, et al. CDX1 is an important molecular mediator of Barrett’s metaplasia. Proc Natl Acad Sci U S A. 2005;102:7565–70. doi: 10.1073/pnas.0502031102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Eda A, Osawa H, Yanaka I, et al. Expression of homeobox gene CDX2 precedes that of CDX1 during the progression of intestinal metaplasia. J Gastroenterol. 2002;37:94–100. doi: 10.1007/s005350200002. [DOI] [PubMed] [Google Scholar]
  • 71.Mutoh H, Sakurai S, Satoh K, et al. Cdx1 induced intestinal metaplasia in the transgenic mouse stomach: comparative study with Cdx2 transgenic mice. Gut. 2004;53:1416–23. doi: 10.1136/gut.2003.032482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yu WY, Slack JM, Tosh D. Conversion of columnar to stratified squamous epithelium in the developing mouse oesophagus. Dev Biol. 2005;284:157–70. doi: 10.1016/j.ydbio.2005.04.042. [DOI] [PubMed] [Google Scholar]
  • 73.Rozek LS, Lipkin SM, Fearon ER, et al. CDX2 polymorphisms, RNA expression, and risk of colorectal cancer. Cancer Res. 2005;65:5488–92. doi: 10.1158/0008-5472.CAN-04-3645. [DOI] [PubMed] [Google Scholar]
  • 74.Sivagnanasundaram S, Islam I, Talbot I, Drummond F, Walters JR, Edwards YH. The homeobox gene CDX2 in colorectal carcinoma: a genetic analysis. Br J Cancer. 2001;84:218–25. doi: 10.1054/bjoc.2000.1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hinoi T, Loda M, Fearon ER. Silencing of CDX2 expression in colon cancer via a dominant repression pathway. J Biol Chem. 2003;278:44608–16. doi: 10.1074/jbc.M307435200. [DOI] [PubMed] [Google Scholar]
  • 76.Wong NA, Britton MP, Choi GS, et al. Loss of CDX1 expression in colorectal carcinoma: promoter methylation, mutation, and loss of heterozygosity analyses of 37 cell lines. Proc Natl Acad Sci U S A. 2004;101:574–9. doi: 10.1073/pnas.0307190101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Lickert H, Kemler R. Functional analysis of cis-regulatory elements controlling initiation and maintenance of early Cdx1 gene expression in the mouse. Dev Dyn. 2002;225:216–20. doi: 10.1002/dvdy.10149. [DOI] [PubMed] [Google Scholar]
  • 78.Rankin EB, Xu W, Silberg DG, Suh E. Putative intestine-specific enhancers located in 5′ sequence of the CDX1 gene regulate CDX1 expression in the intestine. Am J Physiol Gastrointest Liver Physiol. 2004;286:G872–80. doi: 10.1152/ajpgi.00326.2003. [DOI] [PubMed] [Google Scholar]
  • 79.Huo X, Zhang HY, Zhang XI, et al. Acid and bile salt-induced CDX2 expression differs in esophageal squamous cells from patients with and without Barrett’s esophagus. Gastroenterology. 2010;139:194–203. e1. doi: 10.1053/j.gastro.2010.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Rotkrua P, Akiyama Y, Hashimoto Y, Otsubo T, Yuasa Y. MiR-9 down-regulates CDX2 expression in gastric cancer cells. Int J Cancer. 2011 doi: 10.1002/ijc.25923. [DOI] [PubMed] [Google Scholar]
  • 81.Gregory PA, Lewinsky RH, Gardner-Stephen DA, Mackenzie PI. Regulation of UDP glucuronosyltransferases in the gastrointestinal tract. Toxicol Appl Pharmacol. 2004;199:354–63. doi: 10.1016/j.taap.2004.01.008. [DOI] [PubMed] [Google Scholar]
  • 82.Odom DT, Zizlsperger N, Gordon DB, et al. Control of pancreas and liver gene expression by HNF transcription factors. Science. 2004;303:1378–81. doi: 10.1126/science.1089769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Watt AJ, Garrison WD, Duncan SA. HNF4: a central regulator of hepatocyte differentiation and function. Hepatology. 2003;37:1249–53. doi: 10.1053/jhep.2003.50273. [DOI] [PubMed] [Google Scholar]
  • 84.D’Angelo A, Bluteau O, Garcia-Gonzalez MA, et al. Hepatocyte nuclear factor 1alpha and beta control terminal differentiation and cell fate commitment in the gut epithelium. Development. 2010;137:1573–82. doi: 10.1242/dev.044420. [DOI] [PubMed] [Google Scholar]
  • 85.Gregory PA, Lewinsky RH, Gardner-Stephen DA, Mackenzie PI. Coordinate regulation of the human UDP-glucuronosyltransferase 1A8, 1A9, and 1A10 genes by hepatocyte nuclear factor 1alpha and the caudal-related homeodomain protein 2. Mol Pharmacol. 2004;65:953–63. doi: 10.1124/mol.65.4.953. [DOI] [PubMed] [Google Scholar]
  • 86.Benoit YD, Pare F, Francoeur C, et al. Cooperation between HNF-1alpha, Cdx2, and GATA-4 in initiating an enterocytic differentiation program in a normal human intestinal epithelial progenitor cell line. Am J Physiol Gastrointest Liver Physiol. 2010;298:G504–17. doi: 10.1152/ajpgi.00265.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Piessen G, Jonckheere N, Vincent A, et al. Regulation of the human mucin MUC4 by taurodeoxycholic and taurochenodeoxycholic bile acids in oesophageal cancer cells is mediated by hepatocyte nuclear factor 1alpha. Biochem J. 2007;402:81–91. doi: 10.1042/BJ20061461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Divine JK, McCaul SP, Simon TC. HNF-1alpha and endodermal transcription factors cooperatively activate Fabpl: MODY3 mutations abrogate cooperativity. Am J Physiol Gastrointest Liver Physiol. 2003;285:G62–72. doi: 10.1152/ajpgi.00074.2003. [DOI] [PubMed] [Google Scholar]
  • 89.Divine JK, Staloch LJ, Haveri H, et al. GATA-4, GATA-5, and GATA-6 activate the rat liver fatty acid binding protein gene in concert with HNF-1alpha. Am J Physiol Gastrointest Liver Physiol. 2004;287:G1086–99. doi: 10.1152/ajpgi.00421.2003. [DOI] [PubMed] [Google Scholar]
  • 90.Bosse T, Fialkovich JJ, Piaseckyj CM, et al. Gata4 and Hnf1alpha are partially required for the expression of specific intestinal genes during development. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1302–14. doi: 10.1152/ajpgi.00418.2006. [DOI] [PubMed] [Google Scholar]
  • 91.Bosse T, van Wering HM, Gielen M, et al. Hepatocyte nuclear factor-1alpha is required for expression but dispensable for histone acetylation of the lactase-phlorizin hydrolase gene in vivo. Am J Physiol Gastrointest Liver Physiol. 2006;290:G1016–24. doi: 10.1152/ajpgi.00359.2005. [DOI] [PubMed] [Google Scholar]
  • 92.Lin L, Miller CT, Contreras JI, et al. The hepatocyte nuclear factor 3 alpha gene, HNF3alpha (FOXA1), on chromosome band 14q13 is amplified and overexpressed in esophageal and lung adenocarcinomas. Cancer Res. 2002;62:5273–9. [PubMed] [Google Scholar]
  • 93.Crisera CA, Connelly PR, Marmureanu AR, et al. TTF-1 and HNF-3beta in the developing tracheoesophageal fistula: further evidence for the respiratory origin of the distal esophagus’. J Pediatr Surg. 1999;34:1322–6. doi: 10.1016/s0022-3468(99)90003-9. [DOI] [PubMed] [Google Scholar]
  • 94.Wan H, Kaestner KH, Ang SL, et al. Foxa2 regulates alveolarization and goblet cell hyperplasia. Development. 2004;131:953–64. doi: 10.1242/dev.00966. [DOI] [PubMed] [Google Scholar]
  • 95.Zheng W, Rosenstiel P, Huse K, et al. Evaluation of AGR2 and AGR3 as candidate genes for inflammatory bowel disease. Genes Immun. 2006;7:11–8. doi: 10.1038/sj.gene.6364263. [DOI] [PubMed] [Google Scholar]
  • 96.van der Sluis M, Vincent A, Bouma J, et al. Forkhead box transcription factors Foxa1 and Foxa2 are important regulators of Muc2 mucin expression in intestinal epithelial cells. Biochem Biophys Res Commun. 2008;369:1108–13. doi: 10.1016/j.bbrc.2008.02.158. [DOI] [PubMed] [Google Scholar]
  • 97.Ye DZ, Kaestner KH. Foxa1 and Foxa2 control the differentiation of goblet and enteroendocrine L- and D-cells in mice. Gastroenterology. 2009;137:2052–62. doi: 10.1053/j.gastro.2009.08.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Cattin AL, Le Beyec J, Barreau F, et al. Hepatocyte nuclear factor 4alpha, a key factor for homeostasis, cell architecture, and barrier function of the adult intestinal epithelium. Mol Cell Biol. 2009;29:6294–308. doi: 10.1128/MCB.00939-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Garrison WD, Battle MA, Yang C, Kaestner KH, Sladek FM, Duncan SA. Hepatocyte nuclear factor 4alpha is essential for embryonic development of the mouse colon. Gastroenterology. 2006;130:1207–20. doi: 10.1053/j.gastro.2006.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Babeu JP, Darsigny M, Lussier CR, Boudreau F. Hepatocyte nuclear factor 4alpha contributes to an intestinal epithelial phenotype in vitro and plays a partial role in mouse intestinal epithelium differentiation. Am J Physiol Gastrointest Liver Physiol. 2009;297:G124–34. doi: 10.1152/ajpgi.90690.2008. [DOI] [PubMed] [Google Scholar]
  • 101.Hatzis P, Talianidis I. Regulatory mechanisms controlling human hepatocyte nuclear factor 4alpha gene expression. Mol Cell Biol. 2001;21:7320–30. doi: 10.1128/MCB.21.21.7320-7330.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Nastos A, Pogge von Strandmann E, Weber H, Ryffel GU. The embryonic expression of the tissue-specific transcription factor HNF1alpha in Xenopus: rapid activation by HNF4 and delayed induction by mesoderm inducers. Nucleic Acids Res. 1998;26:5602–8. doi: 10.1093/nar/26.24.5602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Weber H, Holewa B, Jones EA, Ryffel GU. Mesoderm and endoderm differentiation in animal cap explants: identification of the HNF4-binding site as an activin A responsive element in the Xenopus HNF1alpha promoter. Development. 1996;122:1975–84. doi: 10.1242/dev.122.6.1975. [DOI] [PubMed] [Google Scholar]
  • 104.Molkentin JD. The zinc finger-containing transcription factors GATA-4, -5, and -6. Ubiquitously expressed regulators of tissue-specific gene expression. J Biol Chem. 2000;275:38949–52. doi: 10.1074/jbc.R000029200. [DOI] [PubMed] [Google Scholar]
  • 105.Jacobsen CM, Narita N, Bielinska M, Syder AJ, Gordon JI, Wilson DB. Genetic mosaic analysis reveals that GATA-4 is required for proper differentiation of mouse gastric epithelium. Dev Biol. 2002;241:34–46. doi: 10.1006/dbio.2001.0424. [DOI] [PubMed] [Google Scholar]
  • 106.Zhang Y, Rath N, Hannenhalli S, et al. GATA and Nkx factors synergistically regulate tissue-specific gene expression and development in vivo. Development. 2007;134:189–98. doi: 10.1242/dev.02720. [DOI] [PubMed] [Google Scholar]
  • 107.Al-azzeh ED, Fegert P, Blin N, Gott P. Transcription factor GATA-6 activates expression of gastroprotective trefoil genes TFF1 and TFF2. Biochim Biophys Acta. 2000;1490:324–32. doi: 10.1016/s0167-4781(00)00013-0. [DOI] [PubMed] [Google Scholar]
  • 108.van Wering HM, Bosse T, Musters A, et al. Complex regulation of the lactase-phlorizin hydrolase promoter by GATA-4. Am J Physiol Gastrointest Liver Physiol. 2004;287:G899–909. doi: 10.1152/ajpgi.00150.2004. [DOI] [PubMed] [Google Scholar]
  • 109.Bossard P, Zaret KS. GATA transcription factors as potentiators of gut endoderm differentiation. Development. 1998;125:4909–17. doi: 10.1242/dev.125.24.4909. [DOI] [PubMed] [Google Scholar]
  • 110.Gao X, Sedgwick T, Shi YB, Evans T. Distinct functions are implicated for the GATA-4, -5, and -6 transcription factors in the regulation of intestine epithelial cell differentiation. Mol Cell Biol. 1998;18:2901–11. doi: 10.1128/mcb.18.5.2901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Sumi K, Tanaka T, Uchida A, et al. Cooperative interaction between hepatocyte nuclear factor 4 alpha and GATA transcription factors regulates ATP-binding cassette sterol transporters ABCG5 and ABCG8. Mol Cell Biol. 2007;27:4248–60. doi: 10.1128/MCB.01894-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Afouda BA, Ciau-Uitz A, Patient R. GATA4, 5 and 6 mediate TGF{beta} maintenance of endodermal gene expression in Xenopus embryos. Development. 2005;132:763–74. doi: 10.1242/dev.01647. [DOI] [PubMed] [Google Scholar]
  • 113.Morrisey EE, Tang Z, Sigrist K, et al. GATA6 regulates HNF4 and is required for differentiation of visceral endoderm in the mouse embryo. Genes Dev. 1998;12:3579–90. doi: 10.1101/gad.12.22.3579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Belaguli NS, Zhang M, Rigi M, Aftab M, Berger DH. Cooperation between GATA4 and TGF-beta signaling regulates intestinal epithelial gene expression. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1520–33. doi: 10.1152/ajpgi.00236.2006. [DOI] [PubMed] [Google Scholar]
  • 115.Nemer G, Nemer M. Transcriptional activation of BMP-4 and regulation of mammalian organogenesis by GATA-4 and -6. Dev Biol. 2003;254:131–48. doi: 10.1016/s0012-1606(02)00026-x. [DOI] [PubMed] [Google Scholar]
  • 116.Haveri H, Westerholm-Ormio M, Lindfors K, et al. Transcription factors GATA-4 and GATA-6 in normal and neoplastic human gastrointestinal mucosa. BMC Gastroenterol. 2008;8:9. doi: 10.1186/1471-230X-8-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Miller CT, Moy JR, Lin L, et al. Gene amplification in esophageal adenocarcinomas and Barrett’s with high-grade dysplasia. Clin Cancer Res. 2003;9:4819–25. [PubMed] [Google Scholar]
  • 118.Guo M, House MG, Akiyama Y, et al. Hypermethylation of the GATA gene family in esophageal cancer. Int J Cancer. 2006;119:2078–83. doi: 10.1002/ijc.22092. [DOI] [PubMed] [Google Scholar]
  • 119.Alvarez H, Opalinska J, Zhou L, et al. Widespread Hypomethylation Occurs Early and Synergizes with Gene Amplification during Esophageal Carcinogenesis. PLoS Genet. 2011;7:e1001356. doi: 10.1371/journal.pgen.1001356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Krishnadath KK. Novel findings in the pathogenesis of esophageal columnar metaplasia or Barrett’s esophagus. Curr Opin Gastroenterol. 2007;23:440–5. doi: 10.1097/MOG.0b013e32814e6b4f. [DOI] [PubMed] [Google Scholar]
  • 121.Milano F, van Baal JW, Buttar NS, et al. Bone morphogenetic protein 4 expressed in esophagitis induces a columnar phenotype in esophageal squamous cells. Gastroenterology. 2007;132:2412–21. doi: 10.1053/j.gastro.2007.03.026. [DOI] [PubMed] [Google Scholar]
  • 122.Domyan ET, Ferretti E, Throckmorton K, Mishina Y, Nicolis SK, Sun X. Signaling through BMP receptors promotes respiratory identity in the foregut via repression of Sox2. Development. 2011;138:971–81. doi: 10.1242/dev.053694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Rodriguez P, Da Silva S, Oxburgh L, Wang F, Hogan BL, Que J. BMP signaling in the development of the mouse esophagus and forestomach. Development. 2010;137:4171–6. doi: 10.1242/dev.056077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Auclair BA, Benoit YD, Rivard N, Mishina Y, Perreault N. Bone morphogenetic protein signaling is essential for terminal differentiation of the intestinal secretory cell lineage. Gastroenterology. 2007;133:887–96. doi: 10.1053/j.gastro.2007.06.066. [DOI] [PubMed] [Google Scholar]
  • 125.Que J, Choi M, Ziel JW, Klingensmith J, Hogan BL. Morphogenesis of the trachea and esophagus: current players and new roles for noggin and Bmps. Differentiation. 2006;74:422–37. doi: 10.1111/j.1432-0436.2006.00096.x. [DOI] [PubMed] [Google Scholar]
  • 126.Batts LE, Polk DB, Dubois RN, Kulessa H. Bmp signaling is required for intestinal growth and morphogenesis. Dev Dyn. 2006;235:1563–70. doi: 10.1002/dvdy.20741. [DOI] [PubMed] [Google Scholar]
  • 127.Onwuegbusi BA, Aitchison A, Chin SF, et al. Impaired transforming growth factor beta signalling in Barrett’s carcinogenesis due to frequent SMAD4 inactivation. Gut. 2006;55:764–74. doi: 10.1136/gut.2005.076430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Onwuegbusi BA, Rees JR, Lao-Sirieix P, Fitzgerald RC. Selective loss of TGFbeta Smad-dependent signalling prevents cell cycle arrest and promotes invasion in oesophageal adenocarcinoma cell lines. PLoS ONE. 2007;2:e177. doi: 10.1371/journal.pone.0000177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Calon A, Gross I, Davidson I, et al. Functional interaction between the homeoprotein CDX1 and the transcriptional machinery containing the TATA-binding protein. Nucleic Acids Res. 2007;35:175–85. doi: 10.1093/nar/gkl1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Gregorieff A, Pinto D, Begthel H, Destree O, Kielman M, Clevers H. Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology. 2005;129:626–38. doi: 10.1016/j.gastro.2005.06.007. [DOI] [PubMed] [Google Scholar]
  • 131.van Es JH, Jay P, Gregorieff A, et al. Wnt signalling induces maturation of Paneth cells in intestinal crypts. Nat Cell Biol. 2005;7:381–6. doi: 10.1038/ncb1240. [DOI] [PubMed] [Google Scholar]
  • 132.Gregorieff A, Clevers H. Wnt signaling in the intestinal epithelium: from endoderm to cancer. Genes Dev. 2005;19:877–90. doi: 10.1101/gad.1295405. [DOI] [PubMed] [Google Scholar]
  • 133.Clement G, Jablons DM, Benhattar J. Targeting the Wnt signaling pathway to treat Barrett’s esophagus. Expert Opin Ther Targets. 2007;11:375–89. doi: 10.1517/14728222.11.3.375. [DOI] [PubMed] [Google Scholar]
  • 134.Clement G, Braunschweig R, Pasquier N, Bosman FT, Benhattar J. Alterations of the Wnt signaling pathway during the neoplastic progression of Barrett’s esophagus. Oncogene. 2006;25:3084–92. doi: 10.1038/sj.onc.1209338. [DOI] [PubMed] [Google Scholar]
  • 135.Eads CA, Lord RV, Kurumboor SK, et al. Fields of aberrant CpG island hypermethylation in Barrett’s esophagus and associated adenocarcinoma. Cancer Res. 2000;60:5021–6. [PubMed] [Google Scholar]
  • 136.Bailey T, Biddlestone L, Shepherd N, Barr H, Warner P, Jankowski J. Altered cadherin and catenin complexes in the Barrett’s esophagus-dysplasia-adenocarcinoma sequence: correlation with disease progression and dedifferentiation. Am J Pathol. 1998;152:135–44. [PMC free article] [PubMed] [Google Scholar]
  • 137.Lickert H, Domon C, Huls G, et al. Wnt/(beta)-catenin signaling regulates the expression of the homeobox gene Cdx1 in embryonic intestine. Development. 2000;127:3805–13. doi: 10.1242/dev.127.17.3805. [DOI] [PubMed] [Google Scholar]
  • 138.Pilon N, Oh K, Sylvestre JR, Savory JG, Lohnes D. Wnt signaling is a key mediator of Cdx1 expression in vivo. Development. 2007;134:2315–23. doi: 10.1242/dev.001206. [DOI] [PubMed] [Google Scholar]
  • 139.Murata-Kamiya N, Kurashima Y, Teishikata Y, et al. Helicobacter pylori CagA interacts with E-cadherin and deregulates the beta-catenin signal that promotes intestinal transdifferentiation in gastric epithelial cells. Oncogene. 2007;26:4617–26. doi: 10.1038/sj.onc.1210251. [DOI] [PubMed] [Google Scholar]
  • 140.Saegusa M, Hashimura M, Kuwata T, Hamano M, Wani Y, Okayasu I. A functional role of Cdx2 in {beta}-catenin signaling during transdifferentiation in endometrial carcinomas. Carcinogenesis. 2007;28:1885–92. doi: 10.1093/carcin/bgm105. [DOI] [PubMed] [Google Scholar]
  • 141.Okubo A, Miyoshi O, Baba K, et al. A novel GATA4 mutation completely segregated with atrial septal defect in a large Japanese family. J Med Genet. 2004;41:e97. doi: 10.1136/jmg.2004.018895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Bastide P, Darido C, Pannequin J, et al. Sox9 regulates cell proliferation and is required for Paneth cell differentiation in the intestinal epithelium. J Cell Biol. 2007;178:635–48. doi: 10.1083/jcb.200704152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Blache P, van de Wetering M, Duluc I, et al. SOX9 is an intestine crypt transcription factor, is regulated by the Wnt pathway, and represses the CDX2 and MUC2 genes. J Cell Biol. 2004;166:37–47. doi: 10.1083/jcb.200311021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Topol L, Chen W, Song H, Day TF, Yang Y. Sox9 inhibits Wnt signaling by promoting beta-catenin phosphorylation in the nucleus. J Biol Chem. 2009;284:3323–33. doi: 10.1074/jbc.M808048200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Mori-Akiyama Y, van den Born M, van Es JH, et al. SOX9 is required for the differentiation of paneth cells in the intestinal epithelium. Gastroenterology. 2007;133:539–46. doi: 10.1053/j.gastro.2007.05.020. [DOI] [PubMed] [Google Scholar]
  • 146.Sumi E, Iehara N, Akiyama H, et al. SRY-related HMG box 9 regulates the expression of Col4a2 through transactivating its enhancer element in mesangial cells. Am J Pathol. 2007;170:1854–64. doi: 10.2353/ajpath.2007.060899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Murakami S, Lefebvre V, de Crombrugghe B. Potent inhibition of the master chondrogenic factor Sox9 gene by interleukin-1 and tumor necrosis factor-alpha. J Biol Chem. 2000;275:3687–92. doi: 10.1074/jbc.275.5.3687. [DOI] [PubMed] [Google Scholar]
  • 148.Manuylov NL, Fujiwara Y, Adameyko, Poulat F, Tevosian SG. The regulation of Sox9 gene expression by the GATA4/FOG2 transcriptional complex in dominant XX sex reversal mouse models. Dev Biol. 2007;307:356–67. doi: 10.1016/j.ydbio.2007.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Wang DH, Clemons NJ, Miyashita T, et al. Aberrant epithelial-mesenchymal Hedgehog signaling characterizes Barrett’s metaplasia. Gastroenterology. 2010;138:1810–22. doi: 10.1053/j.gastro.2010.01.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Chen G, Korfhagen TR, Xu Y, et al. SPDEF is required for mouse pulmonary goblet cell differentiation and regulates a network of genes associated with mucus production. J Clin Invest. 2009;119:2914–24. doi: 10.1172/JCI39731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Park KS, Korfhagen TR, Bruno MD, et al. SPDEF regulates goblet cell hyperplasia in the airway epithelium. J Clin Invest. 2007;117:978–88. doi: 10.1172/JCI29176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Gregorieff A, Stange DE, Kujala P, et al. The ets-domain transcription factor Spdef promotes maturation of goblet and paneth cells in the intestinal epithelium. Gastroenterology. 2009;137:1333–45. e1–3. doi: 10.1053/j.gastro.2009.06.044. [DOI] [PubMed] [Google Scholar]
  • 153.O’Riordan JM, Abdel-latif MM, Ravi N, et al. Proinflammatory cytokine and nuclear factor kappa-B expression along the inflammation-metaplasia-dysplasia-adenocarcinoma sequence in the esophagus. Am J Gastroenterol. 2005;100:1257–64. doi: 10.1111/j.1572-0241.2005.41338.x. [DOI] [PubMed] [Google Scholar]
  • 154.Si J, Fu X, Behar J, et al. NADPH oxidase NOX5-S mediates acid-induced cyclooxygenase-2 expression via activation of NF-kappaB in Barrett’s esophageal adenocarcinoma cells. J Biol Chem. 2007;282:16244–55. doi: 10.1074/jbc.M700297200. [DOI] [PubMed] [Google Scholar]
  • 155.Konturek PC, Nikiforuk A, Kania J, Raithel M, Hahn EG, Muhldorfer S. Activation of NFkappaB represents the central event in the neoplastic progression associated with Barrett’s esophagus: a possible link to the inflammation and overexpression of COX-2, PPARgamma and growth factors. Dig Dis Sci. 2004;49:1075–83. doi: 10.1023/b:ddas.0000037790.11724.70. [DOI] [PubMed] [Google Scholar]
  • 156.Jenkins GJ, Harries K, Doak SH, et al. The bile acid deoxycholic acid (DCA) at neutral pH activates NF-kappaB and induces IL-8 expression in oesophageal cells in vitro. Carcinogenesis. 2004;25:317–23. doi: 10.1093/carcin/bgh032. [DOI] [PubMed] [Google Scholar]
  • 157.Debruyne PR, Witek M, Gong L, et al. Bile acids induce ectopic expression of intestinal guanylyl cyclase C Through nuclear factor-kappaB and Cdx2 in human esophageal cells. Gastroenterology. 2006;130:1191–206. doi: 10.1053/j.gastro.2005.12.032. [DOI] [PubMed] [Google Scholar]
  • 158.Ikeda H, Sasaki M, Ishikawa A, et al. Interaction of Toll-like receptors with bacterial components induces expression of CDX2 and MUC2 in rat biliary epithelium in vivo and in culture. Lab Invest. 2007;87:559–71. doi: 10.1038/labinvest.3700556. [DOI] [PubMed] [Google Scholar]
  • 159.Wallace KN, Pack M. Unique and conserved aspects of gut development in zebrafish. Dev Biol. 2003;255:12–29. doi: 10.1016/s0012-1606(02)00034-9. [DOI] [PubMed] [Google Scholar]
  • 160.Litingtung Y, Lei L, Westphal H, Chiang C. Sonic hedgehog is essential to foregut development. Nat Genet. 1998;20:58–61. doi: 10.1038/1717. [DOI] [PubMed] [Google Scholar]
  • 161.Xie K, Abbruzzese JL. Developmental biology informs cancer: the emerging role of the hedgehog signaling pathway in upper gastrointestinal cancers. Cancer Cell. 2003;4:245–7. doi: 10.1016/s1535-6108(03)00246-0. [DOI] [PubMed] [Google Scholar]
  • 162.Watkins DN, Peacock CD. Hedgehog signalling in foregut malignancy. Biochem Pharmacol. 2004;68:1055–60. doi: 10.1016/j.bcp.2004.04.025. [DOI] [PubMed] [Google Scholar]
  • 163.Yamanaka Y, Shiotani A, Fujimura Y, et al. Expression of Sonic hedgehog (SHH) and CDX2 in the columnar epithelium of the lower oesophagus. Dig Liver Dis. 2011;43:54–9. doi: 10.1016/j.dld.2010.04.014. [DOI] [PubMed] [Google Scholar]
  • 164.van Es JH, van Gijn ME, Riccio O, et al. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature. 2005;435:959–63. doi: 10.1038/nature03659. [DOI] [PubMed] [Google Scholar]
  • 165.Wong GT, Manfra D, Poulet FM, et al. Chronic treatment with the gamma-secretase inhibitor LY-411,575 inhibits beta-amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation. J Biol Chem. 2004;279:12876–82. doi: 10.1074/jbc.M311652200. [DOI] [PubMed] [Google Scholar]
  • 166.Milano J, McKay J, Dagenais C, et al. Modulation of notch processing by gamma-secretase inhibitors causes intestinal goblet cell metaplasia and induction of genes known to specify gut secretory lineage differentiation. Toxicol Sci. 2004;82:341–58. doi: 10.1093/toxsci/kfh254. [DOI] [PubMed] [Google Scholar]
  • 167.Kazanjian A, Noah T, Brown D, Burkart J, Shroyer NF. Atonal homolog 1 is required for growth and differentiation effects of notch/gamma-secretase inhibitors on normal and cancerous intestinal epithelial cells. Gastroenterology. 2010;139:918–28. 28, e1–6. doi: 10.1053/j.gastro.2010.05.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Stanger BZ, Datar R, Murtaugh LC, Melton DA. Direct regulation of intestinal fate by Notch. Proc Natl Acad Sci U S A. 2005;102:12443–8. doi: 10.1073/pnas.0505690102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Ohashi S, Natsuizaka M, Yashiro-Ohtani Y, et al. NOTCH1 and NOTCH3 coordinate esophageal squamous differentiation through a CSL-dependent transcriptional network. Gastroenterology. 2010;139:2113–23. doi: 10.1053/j.gastro.2010.08.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Nguyen BC, Lefort K, Mandinova A, et al. Cross-regulation between Notch and p63 in keratinocyte commitment to differentiation. Genes Dev. 2006;20:1028–42. doi: 10.1101/gad.1406006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Morrow DJ, Avissar NE, Toia L, et al. Pathogenesis of Barrett’s esophagus: bile acids inhibit the Notch signaling pathway with induction of CDX2 gene expression in human esophageal cells. Surgery. 2009;146:714–21. doi: 10.1016/j.surg.2009.06.050. discussion 21–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Menke V, van Es JH, de Lau W, et al. Conversion of metaplastic Barrett’s epithelium into post-mitotic goblet cells by gamma-secretase inhibition. Dis Model Mech. 2010;3:104–10. doi: 10.1242/dmm.003012. [DOI] [PubMed] [Google Scholar]
  • 173.Lao-Sirieix P, Fitzgerald RC. Role of the micro-environment in Barrett’s carcinogenesis. Biochem Soc Trans. 2010;38:327–30. doi: 10.1042/BST0380327. [DOI] [PubMed] [Google Scholar]
  • 174.Harrison RF, Perry I, Jankowski JA. Barrett’s mucosa: remodelling by the microenvironment. J Pathol. 2000;192:1–3. doi: 10.1002/1096-9896(200009)192:1<1::AID-PATH713>3.0.CO;2-O. [DOI] [PubMed] [Google Scholar]
  • 175.Saadi A, Shannon NB, Lao-Sirieix P, et al. Stromal genes discriminate preinvasive from invasive disease, predict outcome, and highlight inflammatory pathways in digestive cancers. Proc Natl Acad Sci U S A. 2010;107:2177–82. doi: 10.1073/pnas.0909797107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Colleypriest BJ, Ward SG, Tosh D. How does inflammation cause Barrett’s metaplasia? Curr Opin Pharmacol. 2009;9:721–6. doi: 10.1016/j.coph.2009.09.005. [DOI] [PubMed] [Google Scholar]
  • 177.Stairs DB, Kong J, Lynch JP. Cdx genes, inflammation, and the pathogenesis of intestinal metaplasia. Prog Mol Biol Transl Sci. 2010;96:231–70. doi: 10.1016/B978-0-12-381280-3.00010-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Moons LM, Kusters JG, van Delft JH, et al. A pro-inflammatory genotype predisposes to Barrett’s esophagus. Carcinogenesis. 2008;29:926–31. doi: 10.1093/carcin/bgm241. [DOI] [PubMed] [Google Scholar]
  • 179.Fitzgerald RC, Abdalla S, Onwuegbusi BA, et al. Inflammatory gradient in Barrett’s oesophagus: implications for disease complications. Gut. 2002;51:316–22. doi: 10.1136/gut.51.3.316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Souza RF, Huo X, Mittal V, et al. Gastroesophageal reflux might cause esophagitis through a cytokine-mediated mechanism rather than caustic acid injury. Gastroenterology. 2009;137:1776–84. doi: 10.1053/j.gastro.2009.07.055. [DOI] [PubMed] [Google Scholar]
  • 181.Quante M, Abrams JA, Marrache F, Wang TC. Barrett’s esophagus correlates with increased putative gastrointestinal stem cell markers DCLK1 and CCK2R in a IL1b mouse model and in humans. DDW. 2010 [Google Scholar]
  • 182.Seery JP, Watt FM. Asymmetric stem-cell divisions define the architecture of human oesophageal epithelium. Curr Biol. 2000;10:1447–50. doi: 10.1016/s0960-9822(00)00803-4. [DOI] [PubMed] [Google Scholar]
  • 183.Takito J, Al-Awqati Q. Conversion of ES cells to columnar epithelia by hensin and to squamous epithelia by laminin. J Cell Biol. 2004;166:1093–102. doi: 10.1083/jcb.200405159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Benahmed F, Gross I, Guenot D, et al. The microenvironment controls CDX2 homeobox gene expression in colorectal cancer cells. Am J Pathol. 2007;170:733–44. doi: 10.2353/ajpath.2007.060696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Lorentz O, Duluc I, Arcangelis AD, Simon-Assmann P, Kedinger M, Freund JN. Key role of the Cdx2 homeobox gene in extracellular matrix-mediated intestinal cell differentiation. J Cell Biol. 1997;139:1553–65. doi: 10.1083/jcb.139.6.1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Fassan M, Volinia S, Palatini J, et al. MicroRNA expression profiling in human Barrett’s carcinogenesis. Int J Cancer. 2010 doi: 10.1002/ijc.25823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Nguyen GH, Schetter AJ, Chou DB, et al. Inflammatory and microRNA gene expression as prognostic classifier of Barrett’s-associated esophageal adenocarcinoma. Clin Cancer Res. 2010;16:5824–34. doi: 10.1158/1078-0432.CCR-10-1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Wijnhoven BP, Hussey DJ, Watson DI, Tsykin A, Smith CM, Michael MZ. MicroRNA profiling of Barrett’s oesophagus and oesophageal adenocarcinoma. Br J Surg. 2010;97:853–61. doi: 10.1002/bjs.7000. [DOI] [PubMed] [Google Scholar]
  • 189.Yang H, Gu J, Wang KK, et al. MicroRNA expression signatures in Barrett’s esophagus and esophageal adenocarcinoma. Clin Cancer Res. 2009;15:5744–52. doi: 10.1158/1078-0432.CCR-09-0385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Maru DM, Singh RR, Hannah C, et al. MicroRNA-196a is a potential marker of progression during Barrett’s metaplasia-dysplasia-invasive adenocarcinoma sequence in esophagus. Am J Pathol. 2009;174:1940–8. doi: 10.2353/ajpath.2009.080718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Dijckmeester WA, Wijnhoven BP, Watson DI, et al. MicroRNA-143 and -205 expression in neosquamous esophageal epithelium following Argon plasma ablation of Barrett’s esophagus. J Gastrointest Surg. 2009;13:846–53. doi: 10.1007/s11605-009-0799-5. [DOI] [PubMed] [Google Scholar]
  • 192.Bansal A, Lee IH, Hong X, et al. Feasibility of MicroRNAs as Biomarkers for Barrett’s Esophagus Progression: A Pilot Cross-Sectional, Phase 2 Biomarker Study. Am J Gastroenterol. 2011 doi: 10.1038/ajg.2011.37. [DOI] [PubMed] [Google Scholar]
  • 193.Smith CM, Watson DI, Michael MZ, Hussey DJ. MicroRNAs, development of Barrett’s esophagus, and progression to esophageal adenocarcinoma. World J Gastroenterol. 2010;16:531–7. doi: 10.3748/wjg.v16.i5.531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Kan T, Meltzer SJ. MicroRNAs in Barrett’s esophagus and esophageal adenocarcinoma. Curr Opin Pharmacol. 2009;9:727–32. doi: 10.1016/j.coph.2009.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Boominathan L. The tumor suppressors p53, p63, and p73 are regulators of microRNA processing complex. PLoS ONE. 2010;5:e10615. doi: 10.1371/journal.pone.0010615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Blandino G, Moll UM. p63 regulation by microRNAs. Cell Cycle. 2009;8:1466–7. doi: 10.4161/cc.8.10.8766. [DOI] [PubMed] [Google Scholar]
  • 197.Gudas LJ, Wagner JA. Retinoids regulate stem cell differentiation. J Cell Physiol. 2011;226:322–30. doi: 10.1002/jcp.22417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Prinos P, Joseph S, Oh K, Meyer BI, Gruss P, Lohnes D. Multiple pathways governing Cdx1 expression during murine development. Dev Biol. 2001;239:257–69. doi: 10.1006/dbio.2001.0446. [DOI] [PubMed] [Google Scholar]
  • 199.Houle M, Sylvestre JR, Lohnes D. Retinoic acid regulates a subset of Cdx1 function in vivo. Development. 2003;130:6555–67. doi: 10.1242/dev.00889. [DOI] [PubMed] [Google Scholar]
  • 200.Qian A, Cai Y, Magee TR, Wan YJ. Identification of retinoic acid-responsive elements on the HNF1alpha and HNF4alpha genes. Biochem Biophys Res Commun. 2000;276:837–42. doi: 10.1006/bbrc.2000.3549. [DOI] [PubMed] [Google Scholar]
  • 201.Arceci RJ, King AA, Simon MC, Orkin SH, Wilson DB. Mouse GATA-4: a retinoic acid-inducible GATA-binding transcription factor expressed in endodermally derived tissues and heart. Mol Cell Biol. 1993;13:2235–46. doi: 10.1128/mcb.13.4.2235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Afonja O, Raaka BM, Huang A, et al. RAR agonists stimulate SOX9 gene expression in breast cancer cell lines: evidence for a role in retinoid-mediated growth inhibition. Oncogene. 2002;21:7850–60. doi: 10.1038/sj.onc.1205985. [DOI] [PubMed] [Google Scholar]
  • 203.Chang CL, Hong E, Lao-Sirieix P, Fitzgerald RC. A novel role for the retinoic acid-catabolizing enzyme CYP26A1 in Barrett’s associated adenocarcinoma. Oncogene. 2008;27:2951–60. doi: 10.1038/sj.onc.1210969. [DOI] [PubMed] [Google Scholar]
  • 204.Hormi-Carver K, Feagins LA, Spechler SJ, Souza RF. All trans-retinoic acid induces apoptosis via p38 and caspase pathways in metaplastic Barrett’s cells. Am J Physiol Gastrointest Liver Physiol. 2007;292:G18–27. doi: 10.1152/ajpgi.00237.2006. [DOI] [PubMed] [Google Scholar]
  • 205.Lord RV, Tsai PI, Danenberg KD, et al. Retinoic acid receptor-alpha messenger RNA expression is increased and retinoic acid receptor-gamma expression is decreased in Barrett’s intestinal metaplasia, dysplasia, adenocarcinoma sequence. Surgery. 2001;129:267–76. doi: 10.1067/msy.2001.110856. [DOI] [PubMed] [Google Scholar]
  • 206.Brabender J, Lord RV, Metzger R, et al. Role of retinoid X receptor mRNA expression in Barrett’s esophagus. J Gastrointest Surg. 2004;8:413–22. doi: 10.1016/j.gassur.2004.02.007. [DOI] [PubMed] [Google Scholar]
  • 207.Chang CL, Lao-Sirieix P, Save V, De La Cueva Mendez G, Laskey R, Fitzgerald RC. Retinoic acid-induced glandular differentiation of the oesophagus. Gut. 2007;56:906–17. doi: 10.1136/gut.2006.097915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Cooke G, Blanco-Fernandez A, Seery JP. The effect of retinoic acid and deoxycholic acid on the differentiation of primary human esophageal keratinocytes. Dig Dis Sci. 2008;53:2851–7. doi: 10.1007/s10620-008-0240-z. [DOI] [PubMed] [Google Scholar]
  • 209.Hiramatsu T, Osaki M, Ito Y, Tanji Y, Tokuyasu N, Ito H. Expression of RUNX3 protein in human esophageal mucosa and squamous cell carcinoma. Pathobiology. 2005;72:316–24. doi: 10.1159/000091329. [DOI] [PubMed] [Google Scholar]
  • 210.Schulmann K, Sterian A, Berki A, et al. Inactivation of p16, RUNX3, and HPP1 occurs early in Barrett’s-associated neoplastic progression and predicts progression risk. Oncogene. 2005;24:4138–48. doi: 10.1038/sj.onc.1208598. [DOI] [PubMed] [Google Scholar]
  • 211.Smith E, De Young NJ, Pavey SJ, et al. Similarity of aberrant DNA methylation in Barrett’s esophagus and esophageal adenocarcinoma. Mol Cancer. 2008;7:75. doi: 10.1186/1476-4598-7-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Jin Z, Cheng Y, Gu W, et al. A multicenter, double-blinded validation study of methylation biomarkers for progression prediction in Barrett’s esophagus. Cancer Res. 2009;69:4112–5. doi: 10.1158/0008-5472.CAN-09-0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Fukamachi H. Runx3 controls growth and differentiation of gastric epithelial cells in mammals. Dev Growth Differ. 2006;48:1–13. doi: 10.1111/j.1440-169X.2006.00832.x. [DOI] [PubMed] [Google Scholar]
  • 214.Torquati A, O’Rear L, Longobardi L, Spagnoli A, Richards WO, Daniel Beauchamp R. RUNX3 inhibits cell proliferation and induces apoptosis by reinstating transforming growth factor beta responsiveness in esophageal adenocarcinoma cells. Surgery. 2004;136:310–6. doi: 10.1016/j.surg.2004.05.005. [DOI] [PubMed] [Google Scholar]
  • 215.Ito K, Lim AC, Salto-Tellez M, et al. RUNX3 attenuates beta-catenin/T cell factors in intestinal tumorigenesis. Cancer Cell. 2008;14:226–37. doi: 10.1016/j.ccr.2008.08.004. [DOI] [PubMed] [Google Scholar]
  • 216.Ito K, Liu Q, Salto-Tellez M, et al. RUNX3, a novel tumor suppressor, is frequently inactivated in gastric cancer by protein mislocalization. Cancer Res. 2005;65:7743–50. doi: 10.1158/0008-5472.CAN-05-0743. [DOI] [PubMed] [Google Scholar]
  • 217.Guo WH, Weng LQ, Ito K, et al. Inhibition of growth of mouse gastric cancer cells by Runx3, a novel tumor suppressor. Oncogene. 2002;21:8351–5. doi: 10.1038/sj.onc.1206037. [DOI] [PubMed] [Google Scholar]
  • 218.Fukamachi H, Mimata A, Tanaka I, Ito K, Ito Y, Yuasa Y. In vitro differentiation of Runx3−/− p53−/− gastric epithelial cells into intestinal type cells. Cancer Sci. 2008;99:671–6. doi: 10.1111/j.1349-7006.2008.00732.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Fukamachi H, Ito K, Ito Y. Runx3−/− gastric epithelial cells differentiate into intestinal type cells. Biochem Biophys Res Commun. 2004;321:58–64. doi: 10.1016/j.bbrc.2004.06.099. [DOI] [PubMed] [Google Scholar]
  • 220.Katz JP, Perreault N, Goldstein BG, et al. The zinc-finger transcription factor Klf4 is required for terminal differentiation of goblet cells in the colon. Development. 2002;129:2619–28. doi: 10.1242/dev.129.11.2619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Flandez M, Guilmeau S, Blache P, Augenlicht LH. KLF4 regulation in intestinal epithelial cell maturation. Exp Cell Res. 2008;314:3712–23. doi: 10.1016/j.yexcr.2008.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Ghaleb AM, Aggarwal G, Bialkowska AB, Nandan MO, Yang VW. Notch inhibits expression of the Kruppel-like factor 4 tumor suppressor in the intestinal epithelium. Mol Cancer Res. 2008;6:1920–7. doi: 10.1158/1541-7786.MCR-08-0224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Zheng H, Pritchard DM, Yang X, et al. KLF4 gene expression is inhibited by the notch signaling pathway that controls goblet cell differentiation in mouse gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol. 2009;296:G490–8. doi: 10.1152/ajpgi.90393.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Hu D, Wan Y. Regulation of Kruppel-like factor 4 by the anaphase promoting complex pathway is involved in TGF-beta signaling. J Biol Chem. 2011;286:6890–901. doi: 10.1074/jbc.M110.179952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Tetreault MP, Yang Y, Travis J, et al. Esophageal squamous cell dysplasia and delayed differentiation with deletion of kruppel-like factor 4 in murine esophagus. Gastroenterology. 2010;139:171–81. e9. doi: 10.1053/j.gastro.2010.03.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Goldstein BG, Chao HH, Yang Y, Yermolina YA, Tobias JW, Katz JP. Overexpression of Kruppel-like factor 5 in esophageal epithelia in vivo leads to increased proliferation in basal but not suprabasal cells. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1784–92. doi: 10.1152/ajpgi.00541.2006. [DOI] [PubMed] [Google Scholar]
  • 227.Kazumori H, Ishihara S, Takahashi Y, Amano Y, Kinoshita Y. Roles of Kruppel-like factor 4 in oesophageal epithelial cells in Barrett’s epithelium development. Gut. 2011;60:608–17. doi: 10.1136/gut.2010.221648. [DOI] [PubMed] [Google Scholar]

RESOURCES