101
|
Cell lineage dynamics in the process leading to intestinal metaplasia. J Gastroenterol 2011; 46:620-8. [PMID: 21384254 DOI: 10.1007/s00535-011-0391-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 01/30/2011] [Indexed: 02/04/2023]
Abstract
BACKGROUND Gene expression in the early stage of the transition to intestinal metaplasia in human gastric mucosa has not been determined. In this study, we investigated the temporal relationship between cell lineage changes and intestine-specific gene expression in the process leading to intestinal metaplasia, using Cdx2-transgenic mice. METHODS Cellular phenotypes were analyzed by immunohistochemistry and were compared with the gene expression profiles of cell lineage markers by real-time polymerase chain reaction. RESULTS Up to postnatal day (PD) 20, the gastric mucosae of Cdx2-transgenic mice were histologically similar to those of their normal littermates. However, at approximately PD 20, we observed the sporadic appearance of glands in which all the epithelial cells expressed Cdx2 (Cdx2-diffuse positive glands). In the Cdx2-diffuse positive glands, parietal cells had disappeared, the proliferating zone had moved from the isthmus to the base, and absorptive cells and goblet cells were recognized. In contrast, the surrounding mucosa retained the phenotype of the gastric gland in which only some of the epithelial cells expressed Cdx2. During PDs 30 and 40, the entire fundic mucosa changed to transdifferentiated mucosa that was a composite of intestinal metaplasia and spasmolytic polypeptide-expressing metaplasia. An increase in the expression of intestine-specific genes, with a reciprocal decrease in gastric-specific gene expression, began much earlier than the emergence of Cdx2-diffuse positive glands. CONCLUSIONS A dramatic increase in intestine-specific gene expression precedes the morphological appearance of intestinal metaplasia and spasmolytic polypeptide-expressing metaplasia.
Collapse
|
102
|
Greenspon J, Li R, Xiao L, Rao JN, Sun R, Strauch ED, Shea-Donohue T, Wang JY, Turner DJ. Sphingosine-1-phosphate regulates the expression of adherens junction protein E-cadherin and enhances intestinal epithelial cell barrier function. Dig Dis Sci 2011; 56:1342-53. [PMID: 20936358 PMCID: PMC4140085 DOI: 10.1007/s10620-010-1421-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Accepted: 09/02/2010] [Indexed: 12/27/2022]
Abstract
BACKGROUND The regulation of intestinal barrier permeability is important in the maintenance of normal intestinal physiology. Sphingosine-1-phosphate (S1P) has been shown to play a pivotal role in enhancing barrier function in several non-intestinal tissues. The current study determined whether S1P regulated function of the intestinal epithelial barrier by altering expression of E-cadherin, an important protein in adherens junctions. METHODS Studies were performed upon cultured differentiated IECs (IEC-Cdx2L1 line) using standard techniques. RESULTS S1P treatment significantly increased levels of E-cadherin protein and mRNA in intestinal epithelial cells (IECs) and also led to E-cadherin localizing strongly to the cell-cell border. S1P also improved the barrier function as indicated by a decrease in 14C-mannitol paracellular permeability and an increase in transepithelial electrical resistance (TEER) in vitro. CONCLUSIONS These results indicate that S1P increases levels of E-cadherin, both in cellular amounts and at the cell-cell junctions, and leads to improved barrier integrity in cultured intestinal epithelial cells.
Collapse
Affiliation(s)
- Jose Greenspon
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ruiyun Li
- Department of Surgery, Baltimore Veterans Affairs Medical Center, 10 N. Greene Street, Baltimore, MD 21201, USA. Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lan Xiao
- Department of Surgery, Baltimore Veterans Affairs Medical Center, 10 N. Greene Street, Baltimore, MD 21201, USA. Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jaladanki N. Rao
- Department of Surgery, Baltimore Veterans Affairs Medical Center, 10 N. Greene Street, Baltimore, MD 21201, USA. Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rex Sun
- Department of Gastroenterology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eric D. Strauch
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Terez Shea-Donohue
- Department of Gastroenterology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jian-Ying Wang
- Department of Surgery, Baltimore Veterans Affairs Medical Center, 10 N. Greene Street, Baltimore, MD 21201, USA. Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA. Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Douglas J. Turner
- Department of Surgery, Baltimore Veterans Affairs Medical Center, 10 N. Greene Street, Baltimore, MD 21201, USA. Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
103
|
Rotkrua P, Akiyama Y, Hashimoto Y, Otsubo T, Yuasa Y. MiR-9 downregulates CDX2 expression in gastric cancer cells. Int J Cancer 2011; 129:2611-20. [PMID: 21225631 DOI: 10.1002/ijc.25923] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 11/30/2010] [Accepted: 12/30/2010] [Indexed: 12/14/2022]
Abstract
Ectopic expression of CDX2, a caudal-related homeobox protein, is known to be associated with the development of intestinal metaplasia in the stomach and gastric carcinogenesis. Previously, we reported that DNA methylation was partly responsible for CDX2 silencing in gastric cancer (GC). However, the mechanism underlying the aberrant expression of CDX2 during malignant transformation remained unclear. MicroRNAs (miRNAs) are small non-coding RNAs that function as post-transcriptional regulators. To elucidate the role of miRNAs in CDX2 downregulation in GC cells, putative miRNAs, such as miR-9, were computationally predicted. After exogenous pre-miR-9 precursor transfection, the luciferase activity of a reporter vector containing a part of the 3'-UTR of CDX2 was downregulated in HEK-293T cells. The inverse correlation between the miR-9 and CDX2 protein levels was demonstrated in GC cell lines. By means of miR-9 overexpression and knockdown techniques, the expression levels of the CDX2 protein and downstream target genes (p21, MUC2 and TFF3) were responsively altered in MKN45 and NUGC-3 cells. Transfection of an anti-miR-9 molecule significantly inhibited cell growth by promoting G(1) cell cycle arrest in MKN45 cells similarly to the effect of CDX2 overexpression. Moreover, examination of the miR-9 levels in primary GC tissues revealed that the amounts of miR-9 in the CDX2-negative group were significantly higher than those in the CDX2-positive group (p = 0.004). Therefore, miR-9 might repress CDX2 expression via the binding site in the 3'-UTR, resulting in the promotion of cell proliferation in GCs.
Collapse
Affiliation(s)
- Pichayanoot Rotkrua
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | |
Collapse
|
104
|
Affiliation(s)
- Rao N. Jaladanki
- University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center
| | - Jian-Ying Wang
- University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center
| |
Collapse
|
105
|
Feltes BC, de Faria Poloni J, Bonatto D. The developmental aging and origins of health and disease hypotheses explained by different protein networks. Biogerontology 2011; 12:293-308. [DOI: 10.1007/s10522-011-9325-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 02/23/2011] [Indexed: 12/18/2022]
|
106
|
Barros R, da Costa LT, Pinto-de-Sousa J, Duluc I, Freund JN, David L, Almeida R. CDX2 autoregulation in human intestinal metaplasia of the stomach: impact on the stability of the phenotype. Gut 2011; 60:290-8. [PMID: 21148572 PMCID: PMC3034084 DOI: 10.1136/gut.2010.222323] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Intestinal metaplasia (IM) is a gastric preneoplastic lesion that appears following Helicobacter pylori infection and confers an increased risk for development of cancer. It is induced by gastric expression of the intestine-specific transcription factor CDX2. The regulatory mechanisms involved in triggering and maintaining gastric CDX2 expression have not been fully elucidated. The Cdx2(+/-) mouse develops intestinal polyps with gastric differentiation and total loss of Cdx2 expression in the absence of structural loss of the second allele, suggesting a regulatory defect. This putative haplo-insufficiency, together with the apparent stability of IM, led to the hypothesis that CDX2 regulates its own expression through an autoregulatory loop in both contexts. METHODS Gastrointestinal cell lines were co-transfected with wild-type or mutated Cdx2 promoter constructs and CDX2 expression vector for luciferase assays. Transfection experiments were also used to assess endogenous CDX2 autoregulation, evaluated by RT-PCR, qPCR and western blotting. Chromatin immunoprecipitation was performed in a cell line, mouse ileum and human IM. RESULTS CDX2 binds to and transactivates its own promoter and positively regulates its expression in gastrointestinal human carcinoma cell lines. Furthermore, CDX2 is bound to its promoter in the mouse ileum and in human gastric IM, providing a major contribution to understanding the relevance of this autoregulatory pathway in vivo. CONCLUSION The results of this study demonstrate another layer of complexity in CDX2 regulation by an effective autoregulatory loop which may have a major impact on the stability of human IM, possibly resulting in the inevitable progression of the gastric carcinogenesis pathway.
Collapse
Affiliation(s)
- Rita Barros
- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Luis Teixeira da Costa
- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,ICAAM, Universidade de Évora, Évora, Portugal
| | - João Pinto-de-Sousa
- Department of Surgery, Hospital S. João, Porto, Portugal,Faculty of Medicine of the University of Porto, Porto, Portugal
| | | | | | - Leonor David
- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Raquel Almeida
- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine of the University of Porto, Porto, Portugal
| |
Collapse
|
107
|
Zhou H. HIV protease inhibitors induce endoplasmic reticulum stress and disrupt barrier integrity in intestinal epithelial cells. Methods Enzymol 2011; 490:107-19. [PMID: 21266246 DOI: 10.1016/b978-0-12-385114-7.00006-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The integrity of the intestinal epithelial barrier plays a crucial role in maintaining symbiotic homeostasis between microbes in the gut lumen and eukaryotic cells. Disruption of intestinal epithelial barrier function occurs commonly under various pathological conditions, including trauma, inflammatory bowel disease, and drug-induced gastrointestinal toxicity, exhibiting increased intestinal epithelial paracellular permeability or "leakiness" of the intestinal mucosa. Endoplasmic reticulum (ER) stress has recently been linked to various pathological conditions, including intestinal inflammation. Our previous studies have shown that HIV protease inhibitors (PIs) induce ER stress and activate the unfolded protein response (UPR) in different types of cells, and HIV PI-induced UPR activation contributes to the disruption of barrier function in intestinal epithelial cells and the increase of intestinal permeability. This chapter will discuss the commonly used methods for analysis of ER stress activation and epithelial barrier function. Both in vitro cell culture models and in vivo animal models are useful tools to examine general drug-induced ER stress and intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Huiping Zhou
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
108
|
Kim JH, Eom DY, Kim CW, Choi NK, Kwak JH, Choi GM, Jang HJ, Han MS. Expression of E-cadherin, β-catenin, Cdx2 and MMP7 in pT2 and N1/N2 Gastric Cancer: Relationship with Tumor Recurrence within 2-Year Period. JOURNAL OF THE KOREAN SURGICAL SOCIETY 2011. [DOI: 10.4174/jkss.2011.80.1.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Ji Hoon Kim
- Department of Surgery, College of Medicine, University of Ulsan, Gangneung Asan Hospital, Gangneung, Korea
| | - Dae Yoon Eom
- Department of Pathology, College of Medicine, University of Ulsan, Gangneung Asan Hospital, Gangneung, Korea
| | - Chan Wook Kim
- Department of Surgery, College of Medicine, University of Ulsan, Gangneung Asan Hospital, Gangneung, Korea
| | - Nam Kyu Choi
- Department of Surgery, College of Medicine, University of Ulsan, Gangneung Asan Hospital, Gangneung, Korea
| | - Jin Ho Kwak
- Department of Surgery, College of Medicine, University of Ulsan, Gangneung Asan Hospital, Gangneung, Korea
| | - Gun Moo Choi
- Department of Surgery, College of Medicine, University of Ulsan, Gangneung Asan Hospital, Gangneung, Korea
| | - Hyuck Jae Jang
- Department of Surgery, College of Medicine, University of Ulsan, Gangneung Asan Hospital, Gangneung, Korea
| | - Myung Sik Han
- Department of Surgery, College of Medicine, University of Ulsan, Gangneung Asan Hospital, Gangneung, Korea
| |
Collapse
|
109
|
Laurent C, Svrcek M, Flejou JF, Chenard MP, Duclos B, Freund JN, Reimund JM. Immunohistochemical expression of CDX2, β-catenin, and TP53 in inflammatory bowel disease-associated colorectal cancer. Inflamm Bowel Dis 2011; 17:232-40. [PMID: 20815042 DOI: 10.1002/ibd.21451] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 07/07/2010] [Indexed: 12/23/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) exposes patients to an increased risk of colorectal cancer (i-CRC) and differences between i-CRC and sporadic colorectal cancer (s-CRC) pathogenesis were reported. In s-CRC, studies indicate abnormalities in the tumor-suppressor gene Cdx2. This study compared CDX2, β-catenin, and TP53 expression in i-CRC, s-CRC, noncancer IBD, and normal control colonic mucosa. METHODS Expression was investigated by immunohistochemistry in 10 normal, 20 s-CRC, 11 noncancer colonic IBD and 30 i-CRC samples, and in four samples of Crohn's disease (CD)-associated small bowel adenocarcinoma (i-SBA). RESULTS In normal and noncancer IBD samples, CDX2 was confined to the colonocytes nuclei. CDX2 expression was normal in 90% of i-CRC, regardless of tumor differentiation or inflammation intensity. By contrast, CDX2 expression was altered in 45% s-CRC, particularly at the front of invasion in undifferentiated tumors. β-Catenin was restricted to cell membrane in all controls, in 91% noncancer IBD, and in 84% i-CRC samples, whereas 35% s-CRC showed cytoplasmic redistribution and exclusive nuclear staining at the front of invasion. TP53 was strongly and homogeneously expressed in i-CRC nuclei compared to normal control or s-CRC, and increases with inflammation intensity. Nested or diffuse TP53 was found in 81.8% of noncancer IBD samples with a higher proportion of TP53-expressing cells in the most inflamed samples. CDX2, β-catenin, and TP53 expression in CD-associated SBA appears similar to that of i-CRC. Neither Cdx2 nor β-catenin alterations are prominent features of i-CRC. CONCLUSIONS In i-CRC and CD-associated SBA, carcinogenesis is associated early with p53 mutations and to inflammation intensity.
Collapse
Affiliation(s)
- Camille Laurent
- Département de Pathologie, Pôle de Biologie, Hôpitaux Universitaires de Strasbourg, Hôpital de Hautepierre, Strasbourg, France.
| | | | | | | | | | | | | |
Collapse
|
110
|
The role of CDX2 in intestinal homeostasis and inflammation. Biochim Biophys Acta Mol Basis Dis 2010; 1812:283-9. [PMID: 21126581 DOI: 10.1016/j.bbadis.2010.11.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 11/19/2010] [Accepted: 11/22/2010] [Indexed: 12/17/2022]
Abstract
Many transcription factors are known to control transcription at several promoters, while others are only active at a few places. However, due to their importance in controlling cellular functions, aberrant transcription factor function and inappropriate gene regulation have been shown to play a causal role in a large number of diseases and developmental disorders. Inflammatory bowel disease (IBD) is characterized by a chronically inflamed mucosa caused by dysregulation of the intestinal immune homeostasis. The aetiology of IBD is thought to be a combination of genetic and environmental factors, including luminal bacteria. The Caudal-related homeobox transcription factor 2 (CDX2) is critical in early intestinal differentiation and has been implicated as a master regulator of the intestinal homeostasis and permeability in adults. When expressed, CDX2 modulates a diverse set of processes including cell proliferation, differentiation, cell adhesion, migration, and tumorigenesis. In addition to these critical cellular processes, there is increasing evidence for linking CDX2 to intestinal inflammation. The aim of the present paper was to review the current knowledge of CDX2 in regulation of the intestinal homeostasis and further to reveal its potential role in inflammation.
Collapse
|
111
|
Shakya A, Kang J, Chumley J, Williams MA, Tantin D. Oct1 is a switchable, bipotential stabilizer of repressed and inducible transcriptional states. J Biol Chem 2010; 286:450-9. [PMID: 21051540 DOI: 10.1074/jbc.m110.174045] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Little is known regarding how the Oct1 transcription factor regulates target gene expression. Using murine fibroblasts and two target genes, Polr2a and Ahcy, we show that Oct1 recruits the Jmjd1a/KDM3A lysine demethylase to catalyze the removal of the inhibitory histone H3K9 dimethyl mark and block repression. Using purified murine T cells and the Il2 target locus, and a colon cancer cell line and the Cdx2 target locus, we show that Oct1 recruits the NuRD chromatin-remodeling complex to promote a repressed state, but in a regulated manner can switch to a different capacity and mediate Jmjd1a recruitment to block repression. These findings indicate that Oct1 maintains repression through a mechanism involving NuRD and maintains poised gene expression states through an antirepression mechanism involving Jmjd1a. We propose that, rather than acting as a primary trigger of gene activation or repression, Oct1 is a switchable stabilizer of repressed and inducible states.
Collapse
Affiliation(s)
- Arvind Shakya
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| | | | | | | | | |
Collapse
|
112
|
Sakamoto H, Mutoh H, Sugano K. Expression of Claudin-2 in intestinal metaplastic mucosa of Cdx2-transgenic mouse stomach. Scand J Gastroenterol 2010; 45:1273-80. [PMID: 20602571 DOI: 10.3109/00365521.2010.501522] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Cdx2 is expressed in human intestinal metaplastic mucosa and induces intestinal metaplastic mucosa in Cdx2-transgenic mouse stomach. Claudin-2 is a structural component of tight junctions in the intestine and Cdx2 activates the Claudin-2 promoter in the human intestinal epithelial cell line Caco-2. Our aim is to evaluate the expression of Claudin-2 in intestinal metaplastic mucosa of Cdx2-transgenic mouse stomach. MATERIAL AND METHODS The Claudin-2 expression in the normal gastric mucosa and normal intestinal mucosa of wild type mice and the intestinal metaplastic mucosa of Cdx2-transgenic mice was analyzed by immunohistochemistry, Western blotting and quantitative real-time PCR (qRT-PCR). RESULTS Claudin-2 was expressed in the base of the glands in intestine and intestinal metaplasia while it was not expressed in the body of stomach. Claudin-2 expression was found in the antrum of stomach, while it was weaker than that in the intestine and the intestinal metaplasia. Claudin-2 was also detected in intestinal metaplasia, colon and ileum by both Western blotting and qRT-PCR while it was not detected in gastric body. CONCLUSION These results suggest that Cdx2 plays an important role in the expression of Claudin-2 in vivo.
Collapse
Affiliation(s)
- Hirotsugu Sakamoto
- Department of Medicine, Division of Gastroenterology, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | | | | |
Collapse
|
113
|
Ruan W, Zhu S, Wang H, Xu F, Deng H, Ma Y, Lai M. IGFBP-rP1, a potential molecule associated with colon cancer differentiation. Mol Cancer 2010; 9:281. [PMID: 20977730 PMCID: PMC2987981 DOI: 10.1186/1476-4598-9-281] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 10/26/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In our previous studies, we have demonstrated that insulin-like growth factor binding protein-related protein1 (IGFBP-rP1) played its potential tumor suppressor role in colon cancer cells through apoptosis and senescence induction. In this study, we will further uncover the role of IGFBP-rP1 in colon cancer differentiation and a possible mechanism by revealing responsible genes. RESULTS In normal colon epithelium, immunohistochemistry staining detected a gradient IGFBP-rP1 expression along the axis of the crypt. IGFBP-rP1 strongly expressed in the differentiated cells at the surface of the colon epithelium, while weakly expressed at the crypt base. In colon cancer tissues, the expression of IGFBP-rP1 correlated positively with the differentiation status. IGFBP-rP1 strongly expressed in low grade colorectal carcinoma and weakly expressed in high grade colorectal carcinoma. In vitro, transfection of PcDNA3.1(IGFBP-rP1) into RKO, SW620 and CW2 cells induced a more pronounced anterior-posterior polarity morphology, accompanied by upregulation with alkaline phosphatase (AKP) activity. Upregulation of carcino-embryonic antigen (CEA) was also observed in SW620 and CW2 transfectants. The addition of IGFBP-rP1 protein into the medium could mimic most but not all effects of IGFBP-rP1 cDNA transfection. Seventy-eight reproducibly differentially expressed genes were detected in PcDNA3.1(IGFBP-rP1)-RKO transfectants, using Affymetrix 133 plus 2.0 expression chip platform. Directed Acyclic Graph (DAG) of the enriched GO categories demonstrated that differential expression of the enzyme regulator activity genes together with cytoskeleton and actin binding genes were significant. IGFBP-rP1 could upreguate Transgelin (TAGLN), downregulate SRY (sex determining region Y)-box 9(campomelic dysplasia, autosomal sex-reversal) (SOX9), insulin receptor substrate 1(IRS1), cyclin-dependent kinase inhibitor 2B (p15, inhibits CDK4) (CDKN2B), amphiregulin(schwannoma-derived growth factor) (AREG) and immediate early response 5-like(IER5L) in RKO, SW620 and CW2 colon cancer cells, verified by Real time Reverse Transcription Polymerase Chain Reaction (rtRT-PCR). During sodium butyrate-induced Caco2 cell differentiation, IGFBP-rP1 was upregulated and the expression showed significant correlation with the AKP activity. The downregulation of IRS1 and SOX9 were also induced by sodium butyrate. CONCLUSION IGFBP-rP1 was a potential key molecule associated with colon cancer differentiation. Downregulation of IRS1 and SOX9 may the possible key downstream genes involved in the process.
Collapse
Affiliation(s)
- Wenjing Ruan
- Department of Pathology, School of Medicine, Zhejiang University, 388 Yuhangtang Road, Hangzhou 310058, Zhejiang Province, China.
| | | | | | | | | | | | | |
Collapse
|
114
|
Weimann A, Rieger A, Zimmermann M, Gross M, Hoffmann P, Slevogt H, Morawietz L. Comparison of six immunohistochemical markers for the histologic diagnosis of neoplasia in Barrett's esophagus. Virchows Arch 2010; 457:537-45. [PMID: 20844891 DOI: 10.1007/s00428-010-0972-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 08/20/2010] [Accepted: 08/24/2010] [Indexed: 12/28/2022]
Abstract
In esophageal neoplasms, the histopathologic differentiation between Barrett's esophagus with or without intraepithelial neoplasia and adenocarcinoma is often challenging. Immunohistochemistry might help to differentiate between these lesions. The expression of CDX2, LI-cadherin, mucin 2 (MUC2), blood group 8 (BG8, Lewis(y)), claudin-2, and villin was investigated in normal gastroesophageal (n = 23) and in Barrett's (n = 17) mucosa, in low-grade (n = 12) and high-grade (n = 9) intraepithelial neoplasia (IEN) as well as in esophageal adenocarcinoma (n = 16), using immunohistochemistry. For CDX2 and LI-cadherin, the immunoreactivity score was highest in IEN while for MUC2, BG8, and villin, it dropped gradually from Barrett's via IEN to adenocarcinoma, and expression of Claudin-2 was only weak and focal in all lesions. The expression of MUC2 and LI-cadherin differed significantly between all examined lesions except between low-grade and high-grade IEN. MUC2 and LI-cadherin are useful immunohistochemical markers for the differentiation between normal glandular mucosa, Barrett's mucosa, IEN, and invasive carcinoma of the esophagus; however, none of the examined markers was helpful for the differentiation between low-grade and high-grade IEN.
Collapse
Affiliation(s)
- Andreas Weimann
- Institute for Laboratory Medicine and Pathobiochemistry, Charité-University Medical Center, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
115
|
Rao JN, Rathor N, Zou T, Liu L, Xiao L, Yu TX, Cui YH, Wang JY. STIM1 translocation to the plasma membrane enhances intestinal epithelial restitution by inducing TRPC1-mediated Ca2+ signaling after wounding. Am J Physiol Cell Physiol 2010; 299:C579-88. [PMID: 20631248 PMCID: PMC2944314 DOI: 10.1152/ajpcell.00066.2010] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 06/11/2010] [Indexed: 11/22/2022]
Abstract
Early epithelial restitution is an important repair modality in the gut mucosa and occurs as a consequence of epithelial cell migration. Canonical transient receptor potential-1 (TRPC1) functions as a store-operated Ca2+ channel (SOCs) in intestinal epithelial cells (IECs) and regulates intestinal restitution, but the exact upstream signals initiating TRPC1 activation after mucosal injury remain elusive. Stromal interaction molecule 1 (STIM1) is a single membrane-spanning protein and is recently identified as essential components of SOC activation. The current study was performed to determine whether STIM1 plays a role in the regulation of intestinal epithelial restitution by activating TRPC1 channels. STIM1 translocation to the plasma membrane increased after wounding, which was followed by an increase in IEC migration to reseal wounds. Increased STIM1 levels at the plasma membrane by overexpressing EF-hand mutant STIM1 enhanced Ca2+ influx through SOCs and stimulated IEC migration after wounding. STIM1 interacted with TRPC1 and formed STIM1/TRPC1 complex, whereas inactivation of STIM1 by STIM1 silencing decreased SOC-mediated Ca2+ influx and inhibited epithelial restitution. In cells overexpressing EF-hand mutant STIM1, TRPC1 silencing also decreased STIM1/TRPC1 complex, reduced SOC-mediated Ca2+ influx, and repressed cell migration after wounding. Our findings demonstrate that induced STIM1 translocation to the plasma membrane promotes IEC migration after wounding by enhancing TRPC1-mediated Ca2+ signaling and provide new insight into the mechanism of intestinal epithelial restitution.
Collapse
Affiliation(s)
- Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
116
|
Verzi MP, Hatzis P, Sulahian R, Philips J, Schuijers J, Shin H, Freed E, Lynch JP, Dang DT, Brown M, Clevers H, Liu XS, Shivdasani RA. TCF4 and CDX2, major transcription factors for intestinal function, converge on the same cis-regulatory regions. Proc Natl Acad Sci U S A 2010; 107:15157-62. [PMID: 20696899 PMCID: PMC2930576 DOI: 10.1073/pnas.1003822107] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Surprisingly few pathways signal between cells, raising questions about mechanisms for tissue-specific responses. In particular, Wnt ligands signal in many mammalian tissues, including the intestinal epithelium, where constitutive signaling causes cancer. Genome-wide analysis of DNA cis-regulatory regions bound by the intestine-restricted transcription factor CDX2 in colonic cells uncovered highly significant overrepresentation of sequences that bind TCF4, a transcriptional effector of intestinal Wnt signaling. Chromatin immunoprecipitation confirmed TCF4 occupancy at most such sites and co-occupancy of CDX2 and TCF4 across short distances. A region spanning the single nucleotide polymorphism rs6983267, which lies within a MYC enhancer and confers colorectal cancer risk in humans, represented one of many co-occupied sites. Co-occupancy correlated with intestine-specific gene expression and CDX2 loss reduced TCF4 binding. These results implicate CDX2 in directing TCF4 binding in intestinal cells. Co-occupancy of regulatory regions by signal-effector and tissue-restricted transcription factors may represent a general mechanism for ubiquitous signaling pathways to achieve tissue-specific outcomes.
Collapse
Affiliation(s)
- Michael P. Verzi
- Department of Medical Oncology and
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Pantelis Hatzis
- Netherlands Institute of Developmental Biology and Hubrecht Institute, 3508 AD, Utrecht, The Netherlands
| | - Rita Sulahian
- Department of Medical Oncology and
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | | | - Jurian Schuijers
- Netherlands Institute of Developmental Biology and Hubrecht Institute, 3508 AD, Utrecht, The Netherlands
| | - Hyunjin Shin
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02115
| | | | - John P. Lynch
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; and
| | - Duyen T. Dang
- Department of Medicine, University of Michigan School of Medicine, Ann Arbor, MI 48109
| | - Myles Brown
- Department of Medical Oncology and
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Hans Clevers
- Netherlands Institute of Developmental Biology and Hubrecht Institute, 3508 AD, Utrecht, The Netherlands
| | - X. Shirley Liu
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Ramesh A. Shivdasani
- Department of Medical Oncology and
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
117
|
Boyd M, Hansen M, Jensen TGK, Perearnau A, Olsen AK, Bram LL, Bak M, Tommerup N, Olsen J, Troelsen JT. Genome-wide analysis of CDX2 binding in intestinal epithelial cells (Caco-2). J Biol Chem 2010; 285:25115-25. [PMID: 20551321 DOI: 10.1074/jbc.m109.089516] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The CDX2 transcription factor is known to play a crucial role in inhibiting proliferation, promoting differentiation and the expression of intestinal specific genes in intestinal cells. The overall effect of CDX2 in intestinal cells has previously been investigated in conditional knock-out mice, revealing a critical role of CDX2 in the formation of the normal intestinal identity. The identification of direct targets of transcription factors is a key problem in the study of gene regulatory networks. The ChIP-seq technique combines chromatin immunoprecipitation (ChIP) with next generation sequencing resulting in a high throughput experimental method of identifying direct targets of specific transcription factors. The method was applied to CDX2, leading to the identification of the direct binding of CDX2 to several known and novel target genes in the intestinal cell. Examination of the transcript levels of selected genes verified the regulatory role of CDX2 binding. The results place CDX2 as a key node in a transcription factor network controlling the proliferation and differentiation of intestinal cells.
Collapse
Affiliation(s)
- Mette Boyd
- Department of Cellular and Molecular Medicine, Panum Institute, Building 6.4, University of Copenhagen, Blegdamsvej 3. 2200 Copenhagen N, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Abstract
The Cdx (Caudal-type homeobox) group of ParaHox genes (Cdx1, Cdx2 and Cdx4 in the mouse) perform multiple functions in mammalian development. Cdx1 is concerned with axial positional information, and its deletion appears to have no important effect other than a disturbance of axial patterning. In contrast, Cdx2 is required for trophoblast differentiation, axial patterning and extension, as well as for morphological specification (i.e. patterning) of gut endoderm. Cdx4-knockout animals do not present an abnormal phenotype, but, when combined with Cdx2 haploinsufficiency, present a dramatic picture involving abnormal cloacal specification. The latter is probably due in large part to defective paraxial mesodermal development in the caudal region, but may also involve defective endodermal growth. A significant degree of redundancy is apparent between the Cdx genes with respect to caudal extension and possibly also during gut development.
Collapse
|
119
|
Pathophysiology of intestinal metaplasia of the stomach: emphasis on CDX2 regulation. Biochem Soc Trans 2010; 38:358-63. [PMID: 20298183 DOI: 10.1042/bst0380358] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
IM (intestinal metaplasia) of the stomach is a pre-neoplastic lesion that usually follows Helicobacter pylori infection and that confers increased risk for gastric cancer development. After setting the role played by CDX2 (Caudal-type homeobox 2) in the establishment of gastric IM, it became of foremost importance to unravel the regulatory mechanisms behind its de novo expression in the stomach. In the present paper, we review the basic pathology of gastric IM as well as the current knowledge on molecular pathways involved in CDX2 regulation in the gastric context.
Collapse
|
120
|
Weimann A, Zimmermann M, Gross M, Slevogt H, Rieger A, Morawietz L. CDX2 and LI-cadherin expression in esophageal mucosa: use of both markers can facilitate the histologic diagnosis of Barrett's esophagus and carcinoma. Int J Surg Pathol 2010; 18:330-7. [PMID: 20444732 DOI: 10.1177/1066896910364228] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Barrett's mucosa is a risk factor for esophageal adenocarcinoma and should be detected at an early stage. CDX2 and liver-intestine (LI)-cadherin are intestine-specific markers. Aberrant CDX2 expression has been demonstrated in Barrett's metaplasia, esophagitis, and intestinal metaplasia of the stomach. METHODS The relationship between CDX2 and LI-cadherin expression was investigated in normal gastroesophageal (n = 24) and in Barrett's (n = 20) mucosa, in low-grade (n = 15) and high-grade (n = 13) intraepithelial neoplasia (IEN) as well as in esophageal adenocarcinoma (n = 16), using immunohistochemistry. RESULTS Nuclear positivity for CDX2 coupled with membranous expression of LI-cadherin was observed in about 70% of the epithelial cells of Barrett's mucosa. The intensity of staining and the percentage of positive cells increased within the sequential steps of low-grade to high-grade IEN, whereas the normal cylindric epithelium lacked the expression of both. In adenocarcinoma, the expression of LI-cadherin and CDX2 was significantly weaker or absent. CONCLUSIONS CDX2 and LI-cadherin are sensitive markers of intestinal metaplasia with or without dysplasia in the upper gastrointestinal tract. Both can be helpful for the early histologic diagnosis of Barrett's esophagus and its subsequent lesions; however, they do not significantly discern between different grades of dysplasia.
Collapse
|
121
|
D'Angelo A, Bluteau O, Garcia-Gonzalez MA, Gresh L, Doyen A, Garbay S, Robine S, Pontoglio M. Hepatocyte nuclear factor 1alpha and beta control terminal differentiation and cell fate commitment in the gut epithelium. Development 2010; 137:1573-82. [PMID: 20388655 DOI: 10.1242/dev.044420] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The intestinal epithelium is a complex system characterized by massive and continuous cell renewal and differentiation. In this context, cell-type-specific transcription factors are thought to play a crucial role by modulating specific transcription networks and signalling pathways. Hnf1alpha and beta are closely related atypical homeoprotein transcription factors expressed in several epithelia, including the gut. With the use of a conditional inactivation system, we generated mice in which Hnf1b is specifically inactivated in the intestinal epithelium on a wild-type or Hnf1a(-/-) genetic background. Whereas the inactivation of Hnf1a or Hnf1b alone did not lead to any major intestinal dysfunction, the concomitant inactivation of both genes resulted in a lethal phenotype. Double-mutant animals had defective differentiation and cell fate commitment. The expression levels of markers of all the differentiated cell types, both enterocytes and secretory cells, were affected. In addition, the number of goblet cells was increased, whereas mature Paneth cells were missing. At the molecular level, we show that Hnf1alpha and beta act upstream of the Notch pathway controlling directly the expression of two crucial components: Jag1 and Atoh1. We demonstrate that the double-mutant mice present with a defect in intestinal water absorption and that Hnf1alpha and beta directly control the expression of Slc26a3, a gene whose mutations are associated with chloride diarrhoea in human patients. Our study identifies new direct target genes of the Hnf1 transcription factors and shows that they play crucial roles in both defining cell fate and controlling terminal functions in the gut epithelium.
Collapse
Affiliation(s)
- Anna D'Angelo
- Expression Génique, Développement et Maladies Equipe 26/ INSERM U567/ CNRS UMR 8104 / Université Paris-Descartes Institut Cochin Dpt. Génétique et Développement, 75014 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
122
|
Sipos F, Muzes G, Valcz G, Galamb O, Tóth K, Leiszter K, Krenács T, Tulassay Z, Molnár B. Regeneration associated growth factor receptor and epithelial marker expression in lymphoid aggregates of ulcerative colitis. Scand J Gastroenterol 2010; 45:440-448. [PMID: 20132083 DOI: 10.3109/00365521003624144] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Mesenchymal-epithelial transition may have crucial role in mucosal regeneration, hence we assayed epithelial growth factor receptor (EGFR), insulin-like growth factor receptor-1 (IGF1R), hepatocyte-derived growth factor receptor (HGFR), CDX2 and cytokeratin (CK) expression in lymphoid aggregates (LA) of ulcerative colitis (UC). MATERIAL AND METHODS Tissue microarrays (TMAs) made of biopsy samples from 20 mildly, 20 moderately and 20 severely active UC, 12 non-specific colitis (NSC) and 20 healthy colon were prepared, and immunolabelled with anti-EGFR, -IGF1R, -HGFR, -CDX2, -CK antibodies. After virtual microscopic evaluation, one-way ANOVA and correlation analysis were performed. For validation, TaqMan real-time RT-PCR was performed by using RNA from laser microdissected LA from 10 healthy colon and 10 endoscopically active UC biopsies. RESULTS The number of LA was in tight positive correlation with the severity of inflammation (r=0.9). The number of EGFR/HGFR positive subepithelial cells was found to be significantly elevated in severe (21.6+/-2.1%/21.3+/-1.9%), moderate (14.3+/-1.7%/14.6+/-1.6%) and mild (7.2+/-1.6%/7.4+/-1.3%) inflammation compared to healthy colon mucosa (2.6+/-1.4%/2.4+/-1.03%) (p < 0.005). Some alterations were found between UC and NSC samples regarding EGFR and HGFR expression. IGF1R immunoreactive cells were only found in a trace number in all cases. Increasing trend of CDX2 and CK positive subepithelial cells was found in active UC, but it was not in significant correlation with the severity of inflammation. CONCLUSION EGFR and HGFR positive subepithelial cells in LA may be involved in the induction of the regenerative mucosal processes. The presence of CDX2/CK positive subepithelial cells suggests that mesenchymal-to-epithelial transition may be located to lymphoid aggregates.
Collapse
Affiliation(s)
- Ferenc Sipos
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary.
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Abstract
Metaplasia (or transdifferentiation) is defined as the transformation of one tissue type to another. Clues to the molecular mechanisms that control the development of metaplasia are implied from knowledge of the transcription factors that specify tissue identity during normal embryonic development. Barrett's metaplasia describes the development of a columnar/intestinal phenotype in the squamous oesophageal epithelium and is the major risk factor for oesophageal adenocarcinoma. This particular type of cancer has a rapidly rising incidence and a dismal prognosis. The homoeotic transcription factor Cdx2 (Caudal-type homeobox 2) has been implicated as a master switch gene for intestine and therefore for Barrett's metaplasia. Normally, Cdx2 expression is restricted to the epithelium of the small and large intestine. Loss of Cdx2 function, or conditional deletion in the intestine, results in replacement of intestinal cells with a stratified squamous phenotype. In addition, Cdx2 is sufficient to provoke intestinal metaplasia in the stomach. In the present paper, we review the evidence for the role of Cdx2 in the development of Barrett's metaplasia.
Collapse
Affiliation(s)
- Benjamin J Colleypriest
- Centre for Regenerative Medicine, Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| | | | | | | |
Collapse
|
124
|
Benoit YD, Paré F, Francoeur C, Jean D, Tremblay E, Boudreau F, Escaffit F, Beaulieu JF. 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-G517. [PMID: 20133952 PMCID: PMC2907224 DOI: 10.1152/ajpgi.00265.2009] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 01/29/2010] [Indexed: 01/31/2023]
Abstract
In the intestinal epithelium, the Cdx, GATA, and HNF transcription factor families are responsible for the expression of differentiation markers such as sucrase-isomaltase. Although previous studies have shown that Cdx2 can induce differentiation in rat intestinal IEC-6 cells, no data are available concerning the direct implication of transcription factors on differentiation in human normal intestinal epithelial cell types. We investigated the role of Cdx2, GATA-4, and HNF-1alpha using the undifferentiated human intestinal epithelial crypt cell line HIEC. These transcription factors were tested on proliferation and expression of polarization and differentiation markers. Ectopic expression of Cdx2 or HNF-1alpha, alone or in combination, altered cell proliferation abilities through the regulation of cyclin D1 and p27 expression. HNF-1alpha and GATA-4 together induced morphological modifications of the cells toward polarization, resulting in the appearance of functional features such as microvilli. HNF-1alpha was also sufficient to induce the expression of cadherins and dipeptidylpeptidase, whereas in combination with Cdx2 it allowed the expression of the late differentiation marker sucrase-isomaltase. Large-scale analysis of gene expression confirmed the cooperative effect of these factors. Finally, although DcamKL1 and Musashi-1 expression were downregulated in differentiated HIEC, other intestinal stem cell markers, such as Bmi1, were unaffected. These observations show that, in cooperation with Cdx2, HNF-1alpha acts as a key factor on human intestinal cells to trigger the onset of their functional differentiation program whereas GATA-4 appears to promote morphological changes.
Collapse
Affiliation(s)
- Yannick D Benoit
- CIHR Team on Digestive Epithelium, Département d'anatomie et de biologie cellulaire, Faculté de médecine et des sciences de santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
125
|
Abstract
Barrett's oesophagus is the replacement of normal squamous oesophageal epithelium with an intestinalized columnar epithelium. Although some insight has been gained as to what Barrett's oesophagus is, how this columnar epithelium emerges from within a stratified squamous epithelium remains an unanswered question. We have sought to determine whether oesophageal keratinocytes can be trans-differentiated into Barrett's oesophagus cells. Using an Affymetrix microarray, we found unexpectedly that gene-expression patterns in the Barrett's oesophagus were only slightly more similar to the normal small intestine than they were to the normal oesophagus. Thus gene-expression patterns suggest significant molecular similarities remain between Barrett's oesophagus cells and normal squamous oesophageal epithelium, despite their histological resemblance with intestine. We next determined whether directed expression of intestine-specific transcription factors could induce intestinalization of keratinocytes. Retroviral-mediated Cdx2 (Caudal-type homeobox 2) expression in immortalized human oesophageal keratinocytes engineered with human telomerase reverse transcriptase (EPC2-hTERT cells) could be established transiently, but not maintained, and was associated with a reduction in cell proliferation. Co-expression of cyclin D1 rescued proliferation in the Cdx2-expressing cells, but co-expression of dominant-negative p53 did not. Cdx2 expression in the EPC2-hTERT.D1 cells did not induce intestinalization. However, when combined with treatments that induce chromatin remodelling, there was a significant induction of Barrett's oesophagus-associated genes. Studies are ongoing to determine whether other intestinal transcription factors, either alone or in combination, can provoke greater intestinalization of oesophageal keratinocytes. We conclude that, on the basis of gene-expression patterns, Barrett's oesophagus epithelial cells may represent an intermediate between oesophageal keratinocytes and intestinal epithelial cells. Moreover, our findings suggest that it may be possible to induce Barrett's oesophagus epithelial cells from oesophageal keratinocytes by altering the expression of certain critical genes.
Collapse
|
126
|
Peters JH, Avisar N. The molecular pathogenesis of Barrett's esophagus: common signaling pathways in embryogenesis metaplasia and neoplasia. J Gastrointest Surg 2010; 14 Suppl 1:S81-7. [PMID: 19760304 DOI: 10.1007/s11605-009-1011-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Accepted: 08/25/2009] [Indexed: 01/31/2023]
Abstract
Although Barrett's esophagus has been recognized for over 50 years, the cellular and molecular mechanisms leading to the replacement of squamous esophageal epithelium with a columnar type are largely unknown. Barrett's is known to be an acquired process secondary to chronic gastroesophageal reflux disease and occurs in the presence of severe disruption of the gastroesophageal barrier and reflux of a mixture of gastric and duodenal content. Current hypothesis suggest that epithelial change occurs due to stimulation of esophageal stem cells present in the basal layers of the epithelium or submucosal glands, toward a columnar epithelial differentiation pathway. The transcription factor CDX2 seems to play a key role in promoting the cellular biology necessary for columnar differentiation, and can be induced by bile salt and acid stimulation. Several cellular signaling pathways responsible for modulation of intestinal differentiation have also been identified and include WNT, Notch, BMP, Sonic HH and TGFB. These also have been shown to respond to stimulation by bile acids, acid or both and may influence CDX2 expression. Their relative activity within the stem cell population is almost certainly responsible for the development of the esophageal columnar epithelial phenotype we know as Barrett's esophagus.
Collapse
Affiliation(s)
- Jeffrey H Peters
- Department of Surgery, University of Rochester, Rochester, NY 14642, USA.
| | | |
Collapse
|
127
|
Zheng L, Tong Q, Weng M, He J, Lv Q, Wu Z, Du Z, Mei H, Hou X. Expression of resistin-like molecule beta in Barrett's esophagus: a novel biomarker for metaplastic epithelium. Dig Dis Sci 2010; 55:32-39. [PMID: 19225886 DOI: 10.1007/s10620-009-0719-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Accepted: 01/12/2009] [Indexed: 12/15/2022]
Abstract
The formation of goblet cells characterizes the intestinal metaplasia of Barrett's esophagus (BE). Hematoxylin-eosin (HE) staining may fail to show intestinal metaplasia in BE, and PAS-Alcian Blue may present difficulties of interpretation due to its more heterogeneous staining. Recent evidence indicates that expression of resistin-like molecule beta (RELMbeta), a goblet cell-specific protein, is uniquely restricted to intestinal epithelium. However, it still remains largely unknown whether RELMbeta can be served as a biomarker for metaplastic epithelium of BE. In this study, 104 biopsy specimens of the distal esophagus from 88 suspected BE patients were collected, including 56 suspected intestinal metaplasia, 26 gastric type mucosa, and 22 squamous epithelium. We evaluated the RELMbeta expression in these biopsy specimens, and compared with those of CDX-2 immunostaining and PAS-Alcian Blue staining (pH 2.5). Of the suspected intestinal metaplasia specimens, 46 presented intestinal-type goblet cells and were immunostaining positive for RELMbeta and CDX-2, the remaining ten possessed only goblet cell mimickers and were not reactive with RELMbeta and CDX-2. Of the gastric-type mucosa specimens, none reacted with either RELMbeta or CDX-2. Moreover, the squamous epithelium was not reactive with RELMbeta and CDX-2. Acid mucin was present in goblet cells in all cases of BE and columnar cells in ten gastric specimens. In addition, the reactivity of RELMbeta was enhanced in six BE specimens with dysplasia. These results provide evidence that RELMbeta protein may be a novel biomarker to distinguish the intestinal-type goblet cells and goblet cell mimickers, and useful in the correct diagnosis of BE.
Collapse
Affiliation(s)
- Liduan Zheng
- Department of Pathology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Stairs DB, Kong J, Lynch JP. Cdx genes, inflammation, and the pathogenesis of intestinal metaplasia. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 96:231-70. [PMID: 21075347 PMCID: PMC6005371 DOI: 10.1016/b978-0-12-381280-3.00010-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intestinal metaplasia (IM) is a biologically interesting and clinically relevant condition in which one differentiated type of epithelium is replaced by another that is morphologically similar to normal intestinal epithelium. Two classic examples of this are gastric IM and Barrett's esophagus (BE). In both, a chronic inflammatory microenvironment, provoked either by Helicobacter pylori infection of the stomach or acid and bile reflux into the esophagus, precedes the metaplasia. The Caudal-related homeodomain transcription factors Cdx1 and Cdx2 are critical regulators of the normal intestinal epithelial cell phenotype. Ectopic expression of Cdx1 and Cdx2 occurs in both gastric IM as well as in BE. This expression precedes the onset of the metaplasia and implies a causal role for these factors in this process. We review the observations regarding the role of chronic inflammation and the Cdx transcription factors in the pathogenesis of gastric IM and BE.
Collapse
Affiliation(s)
- Douglas B Stairs
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
129
|
WU XUDONG, SUN LIXIN, ZHA WEIBIN, STUDER ELAINE, GURLEY EMILY, CHEN LI, WANG XUAN, HYLEMON PHILLIPB, PANDAK WILLIAMM, SANYAL ARUNJ, ZHANG LUYONG, WANG GUANGJI, CHEN JIE, WANG JIAN, ZHOU HUIPING. HIV protease inhibitors induce endoplasmic reticulum stress and disrupt barrier integrity in intestinal epithelial cells. Gastroenterology 2010; 138:197-209. [PMID: 19732776 PMCID: PMC4644065 DOI: 10.1053/j.gastro.2009.08.054] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 07/28/2009] [Accepted: 08/20/2009] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Human immunodeficiency virus (HIV) protease inhibitor (PI)-induced adverse effects have become a serious clinical problem. In addition to their metabolic and cardiovascular complications, these drugs also frequently cause severe gastrointestinal disorders, including mucosal erosions, epithelial barrier dysfunction, and diarrhea. However, the exact mechanisms underlying gastrointestinal adverse effects of HIV PIs remain unknown. This study investigated whether HIV PIs disrupt intestinal epithelial barrier integrity by activating endoplasmic reticulum (ER) stress. METHODS The most commonly used HIV PIs (lopinavir, ritonavir, and amprenavir) were used; their effects on ER stress activation and epithelial paracellular permeability were examined in vitro as well as in vivo using wild-type and CHOP(-)/(-) mice. RESULTS Treatment with lopinavir and ritonavir, but not amprenavir, induced ER stress, as indicated by a decrease in secreted alkaline phosphatase activities and an increase in the unfolded protein response. This activated ER stress partially impaired the epithelial barrier integrity by promoting intestinal epithelial cell apoptosis. CHOP silencing by specific small hairpin RNA prevented lopinavir- and ritonavir-induced barrier dysfunction in cultured intestinal epithelial cells, whereas CHOP(-)/(-) mice exhibited decreased mucosal injury after exposure to lopinavir and ritonavir. CONCLUSIONS HIV PIs induce ER stress and activate the unfolded protein response in intestinal epithelial cells, thus resulting in disruption of the epithelial barrier integrity.
Collapse
Affiliation(s)
- XUDONG WU
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - LIXIN SUN
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - WEIBIN ZHA
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - ELAINE STUDER
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - EMILY GURLEY
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - LI CHEN
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - XUAN WANG
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia
| | - PHILLIP B. HYLEMON
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia,Department of Internal Medicine/Gastroenterology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia
| | - WILLIAM M. PANDAK
- Department of Internal Medicine/Gastroenterology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia
| | - ARUN J. SANYAL
- Department of Internal Medicine/Gastroenterology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia
| | - LUYONG ZHANG
- China Pharmaceutical University, Nanjing, Jiangsu, China
| | - GUANGJI WANG
- China Pharmaceutical University, Nanjing, Jiangsu, China
| | - JIE CHEN
- Department of Surgery, University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - JIAN–YING WANG
- Department of Surgery, University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - HUIPING ZHOU
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia,Department of Internal Medicine/Gastroenterology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia,China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
130
|
Xu Y, Watanabe T, Tanigawa T, Machida H, Okazaki H, Yamagami H, Watanabe K, Tominaga K, Fujiwara Y, Oshitani N, Arakawa T. Bile acids induce cdx2 expression through the farnesoid x receptor in gastric epithelial cells. J Clin Biochem Nutr 2009; 46:81-6. [PMID: 20104269 PMCID: PMC2803137 DOI: 10.3164/jcbn.09-71] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 08/19/2009] [Indexed: 01/26/2023] Open
Abstract
Clinical and experimental studies showed that the reflux of bile into the stomach contributes to the induction of intestinal metaplasia of the stomach and gastric carcinogenesis. Caudal-type homeobox 2 (Cdx2) plays a key role in the exhibition of intestinal phenotypes by regulating the expression of intestine-specific genes such as goblet-specific gene mucin 2 (MUC2). We investigated the involvement of the farnesoid X receptor (FXR), a nuclear receptor for bile acids, in the chenodeoxycholic acid (CDCA)-induced expression of Cdx2 and MUC2 in normal rat gastric epithelial cells (RGM-1 cells). RGM-1 cells were treated with CDCA or GW4064, an FXR agonist, in the presence or absence of guggulsterone, an FXR antagonist. CDCA induced dose-dependent expression of Cdx2 and MUC2 at both the mRNA and protein levels. The maximum stimulation of Cdx2 and MUC2 mRNA induced by CDCA was observed at 3 h and by 6 h, respectively. GW4064 also induced expression of these molecules. The effects of CDCA and GW4064 on expression of Cdx2 and MUC2 were abolished by guggulsterone. These findings suggest that bile acids may induce gastric intestinal metaplasia and carcinogenesis through the FXR.
Collapse
Affiliation(s)
- Yingji Xu
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka 545-8585, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Zhang X, Zou T, Rao JN, Liu L, Xiao L, Wang PY, Cui YH, Gorospe M, Wang JY. Stabilization of XIAP mRNA through the RNA binding protein HuR regulated by cellular polyamines. Nucleic Acids Res 2009; 37:7623-37. [PMID: 19825980 PMCID: PMC2794158 DOI: 10.1093/nar/gkp755] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 08/26/2009] [Accepted: 08/27/2009] [Indexed: 12/23/2022] Open
Abstract
The X chromosome-linked inhibitor of apoptosis protein (XIAP) is the most potent intrinsic caspase inhibitor and plays an important role in the maintenance of intestinal epithelial integrity. The RNA binding protein, HuR, regulates the stability and translation of many target transcripts. Here, we report that HuR associated with both the 3'-untranslated region and coding sequence of the mRNA encoding XIAP, stabilized the XIAP transcript and elevated its expression in intestinal epithelial cells. Ectopic HuR overexpression or elevated cytoplasmic levels of endogenous HuR by decreasing cellular polyamines increased [HuR/XIAP mRNA] complexes, in turn promoting XIAP mRNA stability and increasing XIAP protein abundance. Conversely, HuR silencing in normal and polyamine-deficient cells rendered the XIAP mRNA unstable, thus reducing the steady state levels of XIAP. Inhibition of XIAP expression by XIAP silencing or by HuR silencing reversed the resistance of polyamine-deficient cells to apoptosis. Our findings demonstrate that HuR regulates XIAP expression by stabilizing its mRNA and implicates HuR-mediated XIAP in the control of intestinal epithelial apoptosis.
Collapse
Affiliation(s)
- Xian Zhang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore Veterans Affairs Medical Center, Department of Pathology, University of Maryland School of Medicine and Laboratory of Cellular and Molecular Biology, National Institute on Aging-IRP, NIH, Baltimore, MD 21224, USA
| | - Tongtong Zou
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore Veterans Affairs Medical Center, Department of Pathology, University of Maryland School of Medicine and Laboratory of Cellular and Molecular Biology, National Institute on Aging-IRP, NIH, Baltimore, MD 21224, USA
| | - Jaladanki N. Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore Veterans Affairs Medical Center, Department of Pathology, University of Maryland School of Medicine and Laboratory of Cellular and Molecular Biology, National Institute on Aging-IRP, NIH, Baltimore, MD 21224, USA
| | - Lan Liu
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore Veterans Affairs Medical Center, Department of Pathology, University of Maryland School of Medicine and Laboratory of Cellular and Molecular Biology, National Institute on Aging-IRP, NIH, Baltimore, MD 21224, USA
| | - Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore Veterans Affairs Medical Center, Department of Pathology, University of Maryland School of Medicine and Laboratory of Cellular and Molecular Biology, National Institute on Aging-IRP, NIH, Baltimore, MD 21224, USA
| | - Peng-Yuan Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore Veterans Affairs Medical Center, Department of Pathology, University of Maryland School of Medicine and Laboratory of Cellular and Molecular Biology, National Institute on Aging-IRP, NIH, Baltimore, MD 21224, USA
| | - Yu-Hong Cui
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore Veterans Affairs Medical Center, Department of Pathology, University of Maryland School of Medicine and Laboratory of Cellular and Molecular Biology, National Institute on Aging-IRP, NIH, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore Veterans Affairs Medical Center, Department of Pathology, University of Maryland School of Medicine and Laboratory of Cellular and Molecular Biology, National Institute on Aging-IRP, NIH, Baltimore, MD 21224, USA
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore Veterans Affairs Medical Center, Department of Pathology, University of Maryland School of Medicine and Laboratory of Cellular and Molecular Biology, National Institute on Aging-IRP, NIH, Baltimore, MD 21224, USA
| |
Collapse
|
132
|
Colonic carcinoma with a pancreatic acinar cell differentiation. A case report. Virchows Arch 2009; 455:527-31. [PMID: 19908063 DOI: 10.1007/s00428-009-0852-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2009] [Revised: 09/23/2009] [Accepted: 10/16/2009] [Indexed: 02/08/2023]
Abstract
A case of a colonic carcinoma showing a pancreatic acinar cell differentiation is described for the first time. A 65-year-old woman underwent surgical resection for an ulcerated protruding tumour of 4 x 2.5 cm in size on the anterior wall of the sigmoid colon. Histologically, tumour cells were organized in acinar structures resembling pancreatic acini and in solid nests and ribbons or diffusely infiltrated as poorly cohesive cells. Lymph nodes and femur metastases displayed the same histological features. The ultrastructural analysis of the primary tumour indicated the presence of zymogen-like granules in the cytoplasm of tumour cells. Immunohistochemically, both acinar and diffuse patterns of growth showed an intense staining for trypsin, chymotrypsin and BCL10 and a weaker immunoreactivity for lipase and carboxyl ester hydrolase. Most tumour cells were cytokeratin 20, CDX2 and p53 positive; whereas, mucin (MUC)2 immunoreactivity was observed only in the signet ring cells present in the diffuse pattern and chromogranin A in rare isolated tumour cells. No immunoreactivity was observed for cytokeratin 7, MUC1, MUC5AC, pancreatic amylase or PDX1. There was no evidence of a pancreatic acinar cell carcinoma or of heterotopic pancreatic tissue. A colonic origin ought to be suspected when a metastatic carcinoma of unknown primary shows an acinar differentiation.
Collapse
|
133
|
Fu Z, Kim J, Vidrich A, Sturgill TW, Cohn SM. Intestinal cell kinase, a MAP kinase-related kinase, regulates proliferation and G1 cell cycle progression of intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2009; 297:G632-40. [PMID: 19696144 PMCID: PMC2763805 DOI: 10.1152/ajpgi.00066.2009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal cell kinase (ICK), originally cloned from the intestine and expressed in the intestinal crypt epithelium, is a highly conserved serine/threonine protein kinase that is similar to mitogen-activated protein kinases (MAPKs) in the catalytic domain and requires dual phosphorylation within a MAPK-like TDY motif for full activation. Despite these similarities to MAPKs, the biological functions of ICK remain unknown. In this study, we report that suppression of ICK expression in cultured intestinal epithelial cells by short hairpin RNA (shRNA) interference significantly impaired cellular proliferation and induced features of gene expression characteristic of colonic or enterocytic differentiation. Downregulation of ICK altered expression of cell cycle regulators (cyclin D1, c-Myc, and p21(Cip1/WAF1)) of G(1)-S transition, consistent with the G(1) cell cycle delay induced by ICK shRNA. ICK deficiency also led to a significant decrease in the expression and/or activity of p70 ribosomal protein S6 kinase (S6K1) and eukaryotic initiation factor 4E (eIF4E), concomitant with reduced expression of their upstream regulators, the mammalian target of rapamycin (mTOR) and the regulatory associated protein of mTOR (Raptor). Furthermore, ICK interacts with the mTOR/Raptor complex in vivo and phosphorylates Raptor in vitro. These results suggest that disrupting ICK function may downregulate protein translation of specific downstream targets of eIF4E and S6K1 such as cyclin D1 and c-Myc through the mTOR/Raptor signaling pathway. Taken together, our findings demonstrate an important role for ICK in proliferation and differentiation of intestinal epithelial cells.
Collapse
Affiliation(s)
- Zheng Fu
- Digestive Health Center of Excellence, Univ. of Virginia Health System, PO Box 800708, Charlottesville, VA 22908, USA.
| | - Jungeun Kim
- 1Digestive Health Center of Excellence and Department of Medicine and
| | - Alda Vidrich
- 1Digestive Health Center of Excellence and Department of Medicine and
| | - Thomas W. Sturgill
- 2Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Steven M. Cohn
- 1Digestive Health Center of Excellence and Department of Medicine and
| |
Collapse
|
134
|
Koslowski M, Türeci O, Huber C, Sahin U. Selective activation of tumor growth-promoting Ca2+ channel MS4A12 in colon cancer by caudal type homeobox transcription factor CDX2. Mol Cancer 2009; 8:77. [PMID: 19781065 PMCID: PMC2759907 DOI: 10.1186/1476-4598-8-77] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Accepted: 09/25/2009] [Indexed: 12/27/2022] Open
Abstract
Colon cancer-associated MS4A12 is a novel colon-specific component of store-operated Ca2+ (SOC) entry sensitizing cells for epidermal growth factor (EGF)-mediated effects on proliferation and chemotaxis. In the present study, we investigated regulation of the MS4A12 promoter to understand the mechanisms responsible for strict transcriptional restriction of this gene to the colonic epithelial cell lineage. DNA-binding assays and luciferase reporter assays showed that MS4A12 promoter activity is governed by a single CDX homeobox transcription factor binding element. RNA interference (RNAi)-mediated silencing of intestine-specific transcription factors CDX1 and CDX2 and chromatin immunoprecipitation (ChIP) in LoVo and SW48 colon cancer cells revealed that MS4A12 transcript and protein expression is essentially dependent on the presence of endogenous CDX2. In summary, our findings provide a rationale for colon-specific expression of MS4A12. Moreover, this is the first report establishing CDX2 as transactivator of tumor growth-promoting gene expression in colon cancer, adding to untangle the complex and conflicting biological functions of CDX2 in colon cancer and supporting MS4A12 as important factor for normal colonic development as well as for the biology and treatment of colon cancer.
Collapse
Affiliation(s)
- Michael Koslowski
- Department of Internal Medicine III, Experimental and Translational Oncology, Johannes Gutenberg University, Obere Zahlbacherstr, 63, 55131 Mainz, Germany.
| | | | | | | |
Collapse
|
135
|
Boyd M, Bressendorff S, Møller J, Olsen J, Troelsen JT. Mapping of HNF4alpha target genes in intestinal epithelial cells. BMC Gastroenterol 2009; 9:68. [PMID: 19761587 PMCID: PMC2761415 DOI: 10.1186/1471-230x-9-68] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Accepted: 09/17/2009] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The role of HNF4alpha has been extensively studied in hepatocytes and pancreatic beta-cells, and HNF4alpha is also regarded as a key regulator of intestinal epithelial cell differentiation. The aim of the present work is to identify novel HNF4alpha target genes in the human intestinal epithelial cells in order to elucidate the role of HNF4alpha in the intestinal differentiation progress. METHODS We have performed a ChIP-chip analysis of the human intestinal cell line Caco-2 in order to make a genome-wide identification of HNF4alpha binding to promoter regions. The HNF4alpha ChIP-chip data was matched with gene expression and histone H3 acetylation status of the promoters in order to identify HNF4alpha binding to actively transcribed genes with an open chromatin structure. RESULTS 1,541 genes were identified as potential HNF4alpha targets, many of which have not previously been described as being regulated by HNF4alpha. The 1,541 genes contributed significantly to gene ontology (GO) pathways categorized by lipid and amino acid transport and metabolism. An analysis of the homeodomain transcription factor Cdx-2 (CDX2), the disaccharidase trehalase (TREH), and the tight junction protein cingulin (CGN) promoters verified that these genes are bound by HNF4alpha in Caco2 cells. For the Cdx-2 and trehalase promoters the HNF4alpha binding was verified in mouse small intestine epithelium. CONCLUSION The HNF4alpha regulation of the Cdx-2 promoter unravels a transcription factor network also including HNF1alpha, all of which are transcription factors involved in intestinal development and gene expression.
Collapse
Affiliation(s)
- Mette Boyd
- Department of Cellular and Molecular Medicine, Panum Institute, Building 6,4, University of Copenhagen, Blegdamsvej 3B 2200 Copenhagen N, Denmark.
| | | | | | | | | |
Collapse
|
136
|
Guo RJ, Funakoshi S, Lee HH, Kong J, Lynch JP. The intestine-specific transcription factor Cdx2 inhibits beta-catenin/TCF transcriptional activity by disrupting the beta-catenin-TCF protein complex. Carcinogenesis 2009; 31:159-66. [PMID: 19734199 DOI: 10.1093/carcin/bgp213] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cdx2 is an intestine-specific transcription factor known to regulate proliferation and differentiation. We have reported previously that Cdx2 limits the proliferation of human colon cancer cells by inhibiting the transcriptional activity of the beta-catenin-T-cell factor (TCF) bipartite complex. Herein we further elucidate this mechanism. Studies with a classic Cdx2 target gene and a canonical Wnt/beta-catenin/TCF reporter suggest that Cdx2 regulates these promoters by distinctly different processes. Specifically, inhibition of beta-catenin/TCF activity by Cdx2 does not require Cdx2 transcriptional activity. Instead, Cdx2 binds beta-catenin and disrupts its interaction with the DNA-binding TCF factors, thereby silencing beta-catenin/TCF target gene expression. Using Cdx2 mutants, we map the Cdx2 domains required for the inhibition of beta-catenin/TCF activity. We identify a subdomain in the N-terminus that is highly conserved and when mutated significantly reduces Cdx2 inhibition of beta-catenin/TCF transcriptional activity. Mutation of this subdomain also abrogates Cdx2's anti-proliferative effects in colon cancer cells. In summary, we conclude that Cdx2 binds beta-catenin and disrupts the beta-catenin-TCF complex. Considering the pivotal role of beta-catenin/TCF activity in driving proliferation of normal intestinal epithelial and colon cancer cells, our findings suggest a novel mechanism for Cdx2-mediated regulation of Wnt/beta-catenin signaling and cell proliferation.
Collapse
Affiliation(s)
- Rong-Jun Guo
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
137
|
Krueger F, Madeja Z, Hemberger M, McMahon M, Cook SJ, Gaunt SJ. Down-regulation of Cdx2 in colorectal carcinoma cells by the Raf-MEK-ERK 1/2 pathway. Cell Signal 2009; 21:1846-56. [PMID: 19686845 DOI: 10.1016/j.cellsig.2009.07.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 07/06/2009] [Accepted: 07/29/2009] [Indexed: 12/30/2022]
Abstract
Cdx2 is a homeodomain transcription factor that regulates normal intestinal cell differentiation. Cdx2 is frequently lost during progression of colorectal cancer (CRC) and is widely viewed as a colorectal tumour suppressor. A previous study suggested that activation of protein kinase C (PKC) may be responsible for Cdx2 down-regulation in CRC cells. Here we show that activation of PKC does indeed promote down-regulation of Cdx2 at both the mRNA and protein levels. However, PKC-dependent loss of Cdx2 is dependent upon activation of the Raf-MEK-ERK1/2 pathway. Indeed, specific activation of the ERK1/2 pathway using the conditional kinase DeltaRaf-1:ER is sufficient to inhibit Cdx2 transcription. The Raf-MEK-ERK1/2 pathway is hyper-activated in a large fraction of colorectal cancers due to mutations in K-Ras and we show that treatment of CRC cell lines with MEK inhibitors causes an increase in Cdx2 expression. Furthermore, activation of the ERK1/2 pathway promotes the phosphorylation and proteasome-dependent degradation of the Cdx2 protein. The inhibitory effect of ERK1/2 upon Cdx2 in CRC cells is in sharp contrast to its stimulatory effect upon Cdx2 expression in trophectoderm and trophoblast stem cells. These results provide important new insights into the regulation of the Cdx2 tumour suppressor by linking it to ERK1/2, a pathway which is frequently activated in CRC.
Collapse
Affiliation(s)
- Felix Krueger
- Laboratory of Developmental Genetics & Imprinting, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | | | | | | | | | | |
Collapse
|
138
|
Sun YG, Wang XW, Yang SM, Zhou G, Wang WQ, Wang HB, Wang RQ, Fang DC. Inhibition of nucleostemin upregulates CDX2 expression in HT29 cells in response to bile acid exposure: implications in the pathogenesis of Barrett's esophagus. J Gastrointest Surg 2009; 13:1430-9. [PMID: 19449081 DOI: 10.1007/s11605-009-0899-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Accepted: 04/15/2009] [Indexed: 01/31/2023]
Abstract
BACKGROUND Barrett's esophagus (BE), a squamous-to-columnar metaplasia, may originate from growth-promoting mutations in metaplastic stem cells. Nucleostemin is a protein highly expressed in undifferentiated embryonic stem cells. The objectives of this study were to explore the potential role of nucleostemin in the pathogenesis of BE METHODS: The expression profiles of 30,968 genes were compared between BE and normal esophageal tissues (n = 6 in each group) by using oligo microarray. Three siRNA plasmid expression vectors against nucleostemin, pRNAi-1, pRNAi-2, and pRNAi-3, were constructed and transfected into HT29 cells. In addition, HT29 cells were exposed to 100-1,000 microM chenodeoxycholic acid (CDC), a bile acid, for 2, 12, and 24 h, and then messenger RNA and protein expressions of nucleostemin and CDX2 were determined by reverse-transcriptase polymerase chain reaction and Western blotting. RESULTS Four hundred and twenty-six differentially expressed genes were detected in BE; 142 were upregulated and 284 downregulated. Nucleostemin was downregulated while CDX2 was upregulated. In vitro, all the recombinant plasmids inhibited the nucleostemin expression in transfected HT29 cells, with pRNAi-1 being the most effective. CDX2 expression was significantly increased in pRNAi-1-transfected HT29 cells, compared with that in the empty plasmid (pRNAT-U6.1/Neo) transfected or untransfected HT29 cells. In addition, CDX2 expression was increased whereas nucleostemin expression was decreased in a dose- and time-dependent manner in HT29 cells treated with CDC. CONCLUSION These findings suggest that the inhibition of nucleostemin expression in "esophageal stem cells" in response to bile acid exposure may be involved in the pathogenesis of BE through upregulating CDX2 expression.
Collapse
Affiliation(s)
- Yong-Gang Sun
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | | | | | | | | | | | | | | |
Collapse
|
139
|
Chen YH, Lu YF, Ko TY, Tsai MY, Lin CY, Lin CC, Hwang SPL. Zebrafish cdx1b regulates differentiation of various intestinal cell lineages. Dev Dyn 2009; 238:1021-32. [PMID: 19253392 DOI: 10.1002/dvdy.21908] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Both antisense morpholino oligonucleotide (MO)-mediated knockdown and overexpression experiments were performed to analyze zebrafish cdx1b's function in intestinal cell differentiation. Substantial reductions in goblet cell numbers were detected in intestines of 102- and 120-hours post-fertilization (hpf) cdx1b MO-injected embryos (morphants) compared to cdx1b-4-base mismatched (4mm)-MO-injected and wild type embryos. A significant decrease in enteroendocrine cell numbers was also observed in intestines of 96-hpf cdx1b morphants. Furthermore, ectopic cdx1b expression caused notable increases in respective cell numbers of enteroendocrine and goblet cells in intestines of 96- and 98-hpf injected embryos. Decreased PepT1 expression was detected in enterocytes of intestines in cdx1b morphants from 80 to 102 hr of development. In addition, increased cell proliferation was detected in intestines of cdx1b morphants. Overall, our results suggest that zebrafish cdx1b plays important roles in regulating intestinal cell proliferation and the differentiation of various intestinal cell lineages.
Collapse
Affiliation(s)
- Yi-Hua Chen
- Institute of Cellular and Organismic Biology (formerly Institute of Zoology), Academia Sinica, Nankang, Taipei, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
140
|
Doyon G, St-Jean S, Darsigny M, Asselin C, Boudreau F. Nuclear receptor co-repressor is required to maintain proliferation of normal intestinal epithelial cells in culture and down-modulates the expression of pigment epithelium-derived factor. J Biol Chem 2009; 284:25220-9. [PMID: 19608741 DOI: 10.1074/jbc.m109.022632] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Stem cells of the gut epithelium constantly produce precursors that progressively undergo a succession of molecular changes resulting in growth arrest and commitment to a specific differentiation program. Few transcriptional repressors have been identified that maintain the normal intestinal epithelial cell (IEC) proliferation state. Herein, we show that the nuclear receptor co-repressor (NCoR1) is differentially expressed during the proliferation-to-differentiation IEC transition. Silencing of NCoR1 expression in proliferating cells of crypt origin resulted in a rapid growth arrest without associated cell death. A genechip profiling analysis identified several candidate genes to be up-regulated in NCoR1-deficient IEC. Pigment epithelium-derived factor (PEDF, also known as serpinf1), a suspected tumor suppressor gene that plays a key role in the inhibition of epithelial tissue growth, was significantly up-regulated in these cells. Chromatin immunoprecipitation experiments showed that the PEDF gene promoter was occupied by NCoR1 in proliferating epithelial cells. Multiple retinoid X receptor (RXR) heterodimers interacting sites of the PEDF promoter were confirmed to interact with RXR and retinoid acid receptor (RAR). Cotransfection assays showed that RXR and RAR were able to transactivate the PEDF promoter and that NCoR1 was repressing this effect. Finally, forced expression of PEDF in IEC resulted in a slower rate of proliferation. These observations suggest that NCoR1 expression is required to maintain IEC in a proliferative state and identify PEDF as a novel transcriptional target for NCoR1 repressive action.
Collapse
Affiliation(s)
- Geneviève Doyon
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | | | | | | | | |
Collapse
|
141
|
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. [PMID: 19389805 DOI: 10.1152/ajpgi.90690.2008] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatocyte nuclear factor 4alpha (HNF4alpha) is a regulator of hepatocyte and pancreatic transcription. Hnf4alpha deletion in the mouse is embryonically lethal with severe defects in visceral endoderm formation. It has been concluded in the past that the role of Hnf4alpha in the developing colon was much less important than in the liver. However, the precise role of Hnf4alpha in the homeostasis of the small intestinal epithelium remains unclear. Our aim was to evaluate the potential of Hnf4alpha to support an intestinal epithelial phenotype. First, Hnf4alpha potential to dictate this phenotype was assessed in nonintestinal cell lines in vitro. Forced expression of Hnf4alpha in fibroblasts showed an induction of features normally restricted to epithelial cells. Combinatory expression of Hnf4alpha with specific transcriptional regulators of the intestine resulted in the induction of intestinal epithelial genes in this context. Second, the importance of Hnf4alpha in maintaining the homeostasis of the intestinal epithelium was investigated in mice. Mice conditionally deficient for intestinal Hnf4alpha developed normally throughout adulthood with an epithelium displaying normal morphological and functional structures with minor alterations. Subtle but statistical differences were observed at the proliferation and the cytodifferentiation levels. Hnf4alpha mutant mice displayed an increase in the number of goblet and enteroendocrine cells compared with controls. Given the fundamental role of this transcription factor in other tissues, these findings dispute the crucial role for this regulator in the maintenance of intestinal epithelial cell function at a period of time that follows cytodifferentiation but may suggest a functional role in instructing cells to become specific to the intestinal epithelium.
Collapse
Affiliation(s)
- Jean-Philippe Babeu
- Département d'Anatomie et de Biologie Cellulaire, Université de Sherbrooke,Canadian Institutes of Health Research Team on Digestive Epithelium, Sherbrooke, QC J1H 5N4, Canada
| | | | | | | |
Collapse
|
142
|
Abstract
One of the main engines that drives cellular transformation is the loss of proper control of the mammalian cell cycle. The cyclin-dependent kinase inhibitor p21 (also known as p21WAF1/Cip1) promotes cell cycle arrest in response to many stimuli. It is well positioned to function as both a sensor and an effector of multiple anti-proliferative signals. This Review focuses on recent advances in our understanding of the regulation of p21 and its biological functions with emphasis on its p53-independent tumour suppressor activities and paradoxical tumour-promoting activities, and their implications in cancer.
Collapse
Affiliation(s)
- Tarek Abbas
- Department of Biochemistry and Molecular Genetics, University of Virginia, School of Medicine, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, USA. e-mails: ;
| | - Anindya Dutta
- Department of Biochemistry and Molecular Genetics, University of Virginia, School of Medicine, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, USA. e-mails: ;
| |
Collapse
|
143
|
Herfs M, Hubert P, Delvenne P. Epithelial metaplasia: adult stem cell reprogramming and (pre)neoplastic transformation mediated by inflammation? Trends Mol Med 2009; 15:245-53. [PMID: 19457719 DOI: 10.1016/j.molmed.2009.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 04/03/2009] [Accepted: 04/03/2009] [Indexed: 12/17/2022]
Abstract
Throughout adult life, new developmental commitment of adult stem cells causes metaplastic conversions to occur frequently in some organs. These reversible epithelial replacements are almost always observed in association with chronic inflammation and persistent irritation. Although metaplasia is not synonymous with dysplasia, clinical surveillance has demonstrated that these adaptive processes have an increased susceptibility to evolve into cancer. We propose that cytokines and other soluble factors released by both epithelial and inflammatory cells might alter the transcription-factor expression profile of stem cells and lead to the development of metaplasia. Furthermore, inflammatory mediators might also promote the malignant transformation of epithelial metaplasia by inducing genetic and epigenetic changes and by preventing the immune system from mounting an efficient anti-tumour immune response. A better understanding of the molecular mechanisms leading to metaplasia might help in the design of new therapies for neoplastic and degenerative diseases.
Collapse
Affiliation(s)
- Michael Herfs
- Laboratory of Experimental Pathology, GIGA-Cancer (Centre for Experimental Cancer Research), University of Liege, Liege, Belgium
| | | | | |
Collapse
|
144
|
Primary ovarian carcinoid tumors may express CDX-2: a potential pitfall in distinction from metastatic intestinal carcinoid tumors involving the ovary. Int J Gynecol Pathol 2009; 28:41-8. [PMID: 19047909 DOI: 10.1097/pgp.0b013e31817a8f51] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Carcinoid tumors of the ovary are rare neoplasms that may be primary or metastatic. Clinicopathologic features such as unilaterality and early stage favor a primary ovarian neoplasm but in the absence of other teratomatous elements it may be difficult or impossible to determine whether an ovarian carcinoid is primary or metastatic. CDX-2 is a marker of intestinal differentiation that has been proposed as a marker of midgut origin for metastatic carcinoids. Its expression has not been tested in ovarian carcinoids. Additional markers of potential help in defining the origin of a carcinoid include cytokeratin (CK) 20, CK7, and thyroid transcription factor (TTF-1), none of which have been studied in ovarian carcinoids. We evaluated the diagnostic utility of CDX-2, CK20, CK7, and TTF-1 as well as conventional clinicopathologic features in determining the site of origin in 26 ovarian carcinoids (16 primary and 10 metastatic from midgut). Non-neoplastic premenopausal ovaries (n=10) served as controls. All primary ovarian carcinoids were unilateral whereas only 3/10 metastatic carcinoids were unilateral. Multinodular growth occurred in 6/10 metastatic carcinoids but not in any primary carcinoid. The average size of primary ovarian carcinoids was 3.4 cm (range: 0.2-13.5 cm) versus 10.2 cm for metastatic carcinoids (range: 4-32 cm). Of the primary ovarian carcinoids, 12/16 were 3 cm or smaller whereas all metastatic carcinoids were 4 cm or larger. Teratomatous elements were present in association with 10/16 primary ovarian carcinoids, whereas none were present in any metastatic carcinoid. The primary ovarian carcinoid types were insular (n=6), trabecular (n=3), strumal (n=6, of which 5 were trabecular pattern and 1 was insular pattern) or mucinous (n=1). CDX-2 was not expressed in any cells in normal ovaries. Among primary ovarian neoplasms, there was diffuse nuclear CDX-2 expression in 4/6 insular, 0/3 trabecular, 1/6 strumal (1/1 insular pattern and 0/5 trabecular pattern strumal carcinoids), and 1/1 mucinous carcinoids. All metastatic carcinoids, except for two of mucinous type, were insular. CDX-2 was diffusely and strongly expressed in all 8 metastatic insular carcinoids and in both metastatic mucinous carcinoids. None of the metastases was trabecular in type but 12 primary hindgut or foregut trabecular carcinoids were evaluated and all were negative for CDX-2. None of the ovarian carcinoids expressed TTF-1, CK7, or CK20, except for the primary and metastatic mucinous carcinoids, all of which were CK20-positive. These results demonstrate that CDX-2 cannot be used to determine if a carcinoid is primary in the ovary or metastatic from the intestine as insular and mucinous types of either origin express this marker. Trabecular carcinoids of either origin lack CDX-2 expression. CK20, CK7, or TTF-1 do not have diagnostic utility in this context. Conventional clinicopathologic features (unilaterality, lack of multinodular growth, early stage, presence of teratomatous elements, and size 3 cm or smaller) are the most helpful findings in suggesting a primary origin for an ovarian carcinoid tumor.
Collapse
|
145
|
Greenspon J, Li R, Xiao L, Rao JN, Marasa BS, Strauch ED, Wang JY, Turner DJ. Sphingosine-1-phosphate protects intestinal epithelial cells from apoptosis through the Akt signaling pathway. Dig Dis Sci 2009; 54:499-510. [PMID: 18654850 PMCID: PMC2696985 DOI: 10.1007/s10620-008-0393-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Accepted: 06/17/2008] [Indexed: 12/09/2022]
Abstract
OBJECTIVE The regulation of apoptosis of intestinal mucosal cells is important in maintenance of normal intestinal physiology. SUMMARY Sphingosine-1-phosphate (S1P) has been shown to play a critical role in cellular protection to otherwise lethal stimuli in several nonintestinal tissues. METHODS The current study determines whether S1P protected normal intestinal epithelial cells (IECs) from apoptosis and whether Akt activation was the central pathway for this effect. RESULTS S1P demonstrated significantly reduced levels of apoptosis induced by tumor necrosis factor-alpha (TNF-alpha)/cycloheximide (CHX). S1P induced increased levels of phosphorylated Akt and increased Akt activity, but did not affect total amounts of Akt. This activation of Akt was associated with decreased levels of both caspase-3 protein levels and of caspase-3 activity. Inactivation of Akt by treatment with the PI3K chemical inhibitor LY294002 or by overexpression of the dominant negative mutant of Akt (DNMAkt) prevented the protective effect of S1P on apoptosis. Additionally, silencing of the S1P-1 receptor by specific siRNA demonstrated a lesser decrease in apoptosis to S1P exposure. CONCLUSION These results indicate that S1P protects intestinal epithelial cells from apoptosis via an Akt-dependent pathway.
Collapse
Affiliation(s)
- Jose Greenspon
- Department of Surgery, Cell Biology Group, Baltimore Veterans Affairs Medical Center, University of Maryland School of Medicine, 10 N. Greene Street, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Thoene S, Rawat VPS, Heilmeier B, Hoster E, Metzeler KH, Herold T, Hiddemann W, Gökbuget N, Hoelzer D, Bohlander SK, Feuring-Buske M, Buske C. The homeobox gene CDX2 is aberrantly expressed and associated with an inferior prognosis in patients with acute lymphoblastic leukemia. Leukemia 2009; 23:649-55. [DOI: 10.1038/leu.2008.355] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
147
|
Homeobox gene CDX2 inhibits human pancreatic cancer cell proliferation by down-regulating cyclin D1 transcriptional activity. Pancreas 2009; 38:49-57. [PMID: 19106744 DOI: 10.1097/mpa.0b013e31817fa2ba] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Homeobox gene caudal related homeobox gene 2 (CDX2) is an intestine-specific tumor suppressor gene. This study is intended to investigate the effect of CDX2 expression on cell proliferation and cyclin D1 expression in pancreatic cancer cells. METHODS Four pancreatic ductal adenocarcinoma cell lines (PancQGO-1, BxPC-3, MIAPaCa-2, CFPAC-1), 1 islet carcinoma cell line (QGP-1), and 1 adenosquamous carcinoma cell line (KP-3) were analyzed for CDX1 and CDX2 expression using real-time reverse transcription-polymerase chain reaction and Western blot analysis. Proliferation of pancreatic cancer cells was analyzed using WST-1 assay after CDX2 transfection. Luciferase assay was performed to examine the effects of CDX2 on cyclin D1 transcriptional activity. RESULTS CDX2 was expressed at a significantly higher level in QGP-1 cells than in KP-3 cells. Moreover, CDX2 was expressed at a middle level in 4 pancreatic ductal adenocarcinoma cells. Cell proliferation and cyclin D1 mRNA level were inhibited significantly after CDX2 transfection in pancreatic cancer cells. Furthermore, CDX2 inhibited exogenous nuclear factor kappaB-p65-induced luciferase gene expression in a dose-dependent manner. In addition, CDX2 inhibited pGL2HIVD1kappaB2-luciferase activity. CONCLUSIONS CDX2 might play a role in inhibiting cell proliferation and repressing cyclin D1 transcriptional activity through the proximal nuclear factor kappaB binding site in pancreatic cancer cells.
Collapse
|
148
|
Kong J, Nakagawa H, Isariyawongse BK, Funakoshi S, Silberg DG, Rustgi AK, Lynch JP. Induction of intestinalization in human esophageal keratinocytes is a multistep process. Carcinogenesis 2009; 30:122-30. [PMID: 18845559 PMCID: PMC2722140 DOI: 10.1093/carcin/bgn227] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 08/28/2008] [Accepted: 09/24/2008] [Indexed: 12/22/2022] Open
Abstract
Barrett's esophagus (BE) is the replacement of normal squamous esophageal mucosa with an intestinalized columnar epithelium. The molecular mechanisms underlying its development are not understood. Cdx2 is an intestine-specific transcription factor that is ectopically expressed in BE, but its role in this process is unclear. Herein, we describe a novel cell culture model for BE. Retroviral-mediated Cdx2 expression in immortalized human esophageal keratinocytes [EPC-human telomerase reverse transcriptase (hTERT)] could transiently be established but not maintained and was associated with a reduction in cell proliferation. Coexpression of cyclin D1, but not a dominant-negative p53, rescued proliferation in the Cdx2-expressing cells. Cdx2 expression in the EPC-hTERT.D1 cells decreased cell proliferation but did not induce intestinalization. We investigated for other treatments to enhance intestinalization and found that acidic culture conditions uniformly killed EPC-hTERT.D1.Cdx2 cells. However, treatment with 5-aza-2-deoxycytidine (5-AzaC) to demethylate epigenetically silenced genes did appear to be tolerated. Multiple Cdx2 target genes, markers of intestinal differentiation and markers of BE, were induced by this 5-AzaC treatment. More interestingly, the expression level of several of these genes was enhanced only in the EPC-hTERT.D1-Cdx2 cells treated with 5-AzaC. Two of these, SLC26a3/DRA (downregulated in adenoma) and Na+/H+ exchanger 2 (NHE2), were not previously known to be elevated in BE; however, we confirmed their elevation in BE tissue samples. 5-AzaC treatment also induced cell senescence, even at low doses. We conclude that ectopic proliferation signals, alterations in epigenetic gene regulation and the inhibition of tumor suppressor mechanisms are required for Cdx2-mediated intestinalization of human esophageal keratinocytes in BE.
Collapse
Affiliation(s)
- Jianping Kong
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hiroshi Nakagawa
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brandon K. Isariyawongse
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shinsuke Funakoshi
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Debra G. Silberg
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA 19104, USA
- AstraZeneca LP, Wilmington, DE 19850-5437, USA
| | - Anil K. Rustgi
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John P. Lynch
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
149
|
Immunoprofile of Adenocarcinomas of the Endometrium, Endocervix, and Ovary With Mucinous Differentiation. Appl Immunohistochem Mol Morphol 2009; 17:8-11. [DOI: 10.1097/pai.0b013e318174f012] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
150
|
Funakoshi S, Ezaki T, Kong J, Guo RJ, Lynch JP. Repression of the desmocollin 2 gene expression in human colon cancer cells is relieved by the homeodomain transcription factors Cdx1 and Cdx2. Mol Cancer Res 2008; 6:1478-90. [PMID: 18819935 DOI: 10.1158/1541-7786.mcr-07-2161] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Desmosomes are intracellular junctions that provide strong cell-cell adhesion in epithelia and cardiac muscle. Their disruption causes several human diseases and contributes to the epithelial-to-mesenchymal transition observed in cancer. Desmocollin 2 (DSC2) is a cadherin superfamily member and a critical component of desmosomes found in intestinal epithelium. However, the mechanism regulating DSC2 gene expression in intestinal cells is not known. Cdx1 and Cdx2 are homeodomain transcription factors that regulate intestine-specific gene expression. Cdx expression in the past has been associated with the induction of desmosomes. We now show that the DSC2 gene is a transcriptional target for Cdx1 and Cdx2. Colon cancer cell lines retaining Cdx2 expression typically express DSC2. Restoration of Cdx expression in Colo 205 cells induced DSC2 mRNA and protein and the formation of desmosomes. The 5'-flanking region of the DSC2 promoter contains two consensus Cdx-binding sites. Electrophoretic mobility shift assays show that Cdx1 and Cdx2 bind these sites in vitro, and chromatin immunoprecipitation confirmed Cdx2 binding in vivo. DSC2 promoter truncations established that these regions are Cdx responsive. The truncations also identify a region of the promoter in which potent transcriptional repressors act. This repressor activity is relieved by Cdx binding. We conclude that the homeodomain transcription factors Cdx1 and Cdx2 regulate DSC2 gene expression in intestinal epithelia by reversing the actions of a transcriptional repressor. The regulation of desmosomal junctions by Cdx contributes to normal intestinal epithelial columnar morphology and likely antagonizes the epithelial-to-mesenchymal transition necessary for the metastasis of colon cancer cells in humans.
Collapse
Affiliation(s)
- Shinsuke Funakoshi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|