Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Dig Dis Sci ; 54(11): 2488-96, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19757048

ABSTRACT

BACKGROUND: A number of agents, including aspirin, nonsteroidal antiinflammatory drugs, cyclooxygenase-2 inhibitors, folic acid, calcium, and vitamins, have been evaluated for their potential in chemoprevention of sporadic colorectal adenomas or cancer. Preclinical data suggest that 5-aminosalicylates also may have a chemopreventive effect. AIM: To investigate chemoprevention of colonic polyps with balsalazide, a 5-aminosalicylate prodrug. METHODS: In this randomized, double-blind, placebo-controlled study, adults diagnosed with small polyps in the rectosigmoid colon were treated with either balsalazide 3 g/d or placebo for 6 months. Follow-up lower endoscopy was performed, and all polyps were measured and analyzed histologically. The primary endpoint was reduction in mean size of the largest polyp per subject. RESULTS: Among 241 participants screened, 86 were randomized to treatment, with 75 subjects evaluable. Balsalazide 3 g/d (n = 38) did not significantly reduce the mean size of the largest colonic polyp or the number of polyps compared with placebo (n = 37). Although not significant, post-hoc analysis revealed that total adenoma burden per subject, calculated as the sum of the volumes of all adenomas in mm3, increased by 55% in the balsalazide group compared with 95% in the placebo group. CONCLUSIONS: Although balsalazide did not have significant chemopreventive effects on established colonic polyps, these results can aid in designing future prospective studies.


Subject(s)
Colonic Polyps/prevention & control , Gastrointestinal Agents/therapeutic use , Mesalamine/therapeutic use , Phenylhydrazines/therapeutic use , Adenoma/pathology , Adenoma/prevention & control , Aged , Apoptosis/drug effects , Colon, Sigmoid/pathology , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Colonic Polyps/pathology , Double-Blind Method , Gastrointestinal Agents/pharmacology , Humans , Male , Mesalamine/pharmacology , Middle Aged , Phenylhydrazines/pharmacology , Prospective Studies
2.
Int J Cancer ; 122(6): 1253-60, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18027866

ABSTRACT

Mucinous colorectal cancers are characterized by abundant production of intestinal goblet cell mucin, MUC2 and frequent ectopic expression of gastric foveolar mucin, MUC5AC. SOX2, an HMG-box transcription factor expressed in gastric mucosa but not in intestine is thought to play an important role in regulating transcription and expression of gastric differentiation related genes. Herein, we investigated the possible role of SOX2 in MUC5AC transcription and in the development of mucinous cancers. We observed good correlation between SOX2 and MUC5AC message levels in most colon cancer cell lines. SOX2 expression significantly transactivated MUC5AC promoter/reporter constructs in 3 of 5 colon cancer cell lines. We also examined SOX2 expression in normal stomach and colon, nonmucinous and mucinous colorectal cancers, serrated polyps and conventional adenomas using immunohistochemistry and in situ hybridization. SOX2 was expressed in the nuclei of both gastric foveolar cells and fundic glands by immunohistochemistry and in the cytoplasm by in situ hybridization. SOX2 was not expressed in normal colon but was strongly expressed in serrated polyps, mucinous and signet ring cell carcinomas, but rarely in nonmucinous carcinomas and tubular adenomas. Concordant expression of SOX2 with MUC5AC was observed in these lesions. Our results suggest that SOX2 is important in the upregulation of gastric foveolar mucin, MUC5AC in colorectal mucinous and signet ring cell carcinomas. In addition, the expression of both SOX2 and MUC5AC in serrated polyps supports the hypothesis that these polyps may be predominant precursors of mucinous and signet ring cell carcinomas of the colorectum.


Subject(s)
Colorectal Neoplasms/genetics , HMGB Proteins/physiology , Mucins/physiology , Transcription Factors/physiology , Up-Regulation/physiology , Base Sequence , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Primers , Gastric Mucosa/metabolism , Genes, Reporter , HMGB Proteins/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Mucin 5AC , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors , Transcription Factors/genetics
3.
Int J Oncol ; 31(4): 693-703, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17786299

ABSTRACT

Connective tissue growth factor (CTGF/CCN2) is thought to play a role in normal wound repair and bone remodeling, but also promotes fibrosis in several disease processes including diabetic nephropathy, sclerodoma and pancreatitis. A contribution to desmoplasia associated with pancreatic cancer progression has also been proposed. CTGF is induced by TGFbeta in diverse cell types, but TGFbeta receptor mediated signaling is impaired in pancreatic cancers and cell lines, usually due to DPC4/Smad4 mutations which arise during the later stages of intraepithelial neoplastic progression. Therefore, in order to define signaling pathways that mediate basal and TGFbeta-induced CTGF expression in normal and transformed cells, we compared CTGF gene regulation in pancreatic cancer cells and fibroblasts by measuring the effects of small molecule inhibitors and dominant negative mutants of signaling proteins on CTGF promoter reporter activity, message, and protein expression. We determined that the previously identified TEF-1 cis element is essential for CTGF promoter reporter activity in pancreatic cancer cell lines. Whereas p38 mediated CTGF induction by TGFbeta in fibroblasts, MEK/ERK signaling mediated TGFbeta-induced CTGF expression in pancreatic cancer cells and was also responsible for basal CTGF expression in pancreatic cancer cell lines with defective Smad signaling. Since activating Ras mutations occur in the earliest stages of pancreatic cancer, CTGF may be induced independent of Smad4 in pancreatic cancer cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Pancreatic Neoplasms/metabolism , Signal Transduction , 3T3 Cells , Animals , Blotting, Western , Cells, Cultured , Connective Tissue Growth Factor , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/physiology , Mice , Nuclear Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements , Smad Proteins/metabolism , Sp1 Transcription Factor/metabolism , TEA Domain Transcription Factors , Transcription Factors/metabolism , Transcription, Genetic , Transforming Growth Factor beta/metabolism
4.
Clin Cancer Res ; 12(18): 5403-10, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-17000673

ABSTRACT

PURPOSE: Mucinous cancers and signet ring carcinomas are distinct classes of colon cancers characterized by their production of copious quantities of intestinal goblet cell mucin, MUC2. Deletion of transcription factor HATH1 ablates the biogenesis of goblet cells in developing mouse intestine, and forced expression of HATH1 results in elevated expression of MUC2 in colon cancer cells. The aim of this study was to assess the possible role of HATH1 in the development of mucinous cancers and signet ring carcinomas. EXPERIMENTAL DESIGN: Immunohistochemistry and confocal microscopy was used to examine HATH1 expression and subcellular distribution in normal colon and small intestine, mucinous cancers, signet ring carcinomas, and nonmucinous cancers and in precursor lesions, including hyperplastic polyps, serrated adenomas, tubular adenomas, and villous adenomas. We also analyzed the transactivation of MUC2 promoter/reporter constructs by a HATH1 expression vector. RESULTS: HATH1 expression transactivated MUC2 promoter/reporter constructs, an activity that was significantly inhibited by mutation of putative HATH1-binding sites. HATH1 was expressed in the nuclei of goblet cells and in the cytoplasm and nuclei of enteroendocrine cells of the colon. In the small intestine, only cytoplasmic expression of HATH1 in enteroendocrine cells was detected. HATH1 was found to be strongly expressed in the nuclei of hyperplastic polyps, serrated adenomas, villous adenomas, mucinous cancers, and signet ring carcinomas but repressed in nonmucinous cancers and tubular adenomas. CONCLUSIONS: This study confirms the importance of HATH1 for the development of intestinal secretory cells. The results further suggest that HATH1 is an important factor in the up-regulation of MUC2 expression that occurs in mucinous cancers and signet ring carcinomas. In addition, the expression of HATH1 in hyperplastic polyps, serrated adenomas, and villous adenomas lends support to the hypothesis that these neoplasms are frequent precursors in mucinous cancer and signet ring carcinoma development.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Colorectal Neoplasms/metabolism , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma, Mucinous/ultrastructure , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , Colorectal Neoplasms/ultrastructure , Disease Progression , Female , Humans , Intestinal Polyps/pathology , Intestines/cytology , Intestines/ultrastructure , Male , Microscopy, Fluorescence , Middle Aged , Mucin-2 , Mucins/metabolism , Promoter Regions, Genetic , Tissue Distribution , Transcriptional Activation
5.
Int J Oncol ; 29(1): 33-40, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16773182

ABSTRACT

MUC5AC is a secretory mucin normally expressed by the surface mucous cells of the human stomach and in the bronchial tract. It is absent from normal pancreas, but de novo expression of this mucin occurs in early-stage pancreatic intraepithelial neoplasias and in the invasive ductal adenocarcinoma of the pancreas, prompting this study of MUC5AC gene regulation in pancreatic cancer cells. Promoter deletion constructs and EMSA studies revealed that transcription factors Sp1 and AP-1 are both involved in basal transcription of the MUC5AC gene. Phorbol 12-myrisate 13-acetate (PMA) increased MUC5AC mRNA expression and transcriptional activities of MUC5AC promoter-reporter deletion constructs containing AP-1 consensus sites. EMSA studies showed that Fos/Jun binding to putative AP-1 sites is increased by PMA treatment. Western blot analysis showed that ERK, JNK and p38 are all activated by PMA treatment in SW1990 cells. Inhibitors of mitogen-activated protein/extracellular signal regulated kinase (MEK), such as ERK inhibitor PD98059 and JNK inhibitors dicumarol and SP60015, but not p38 inhibitor SB203580, inhibited PMA-induced MUC5AC reporter activity. Our studies indicate that Sp1 is involved in basal MUC5AC promoter activity while AP-1 is involved in basal and PMA-induced MUC5AC promoter activation in pancreatic cancer cells. Furthermore, PMA-induced MUC5AC gene transcription appears to be mediated by activating Sp1, PKC/ERK/AP-1 and PKC/JNK/AP-1 pathways.


Subject(s)
Cell Line, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Mucins/metabolism , Carcinoma, Pancreatic Ductal , Electrophoretic Mobility Shift Assay , Genes, Reporter , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Luciferases/genetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mucin 5AC , Mucins/biosynthesis , Mucins/genetics , Pancreatic Neoplasms , Promoter Regions, Genetic , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/analysis , Sp1 Transcription Factor/metabolism , Tetradecanoylphorbol Acetate , Transcription Factor AP-1/metabolism , Transcription, Genetic , Transfection
6.
Int J Cancer ; 118(12): 2999-3005, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16425274

ABSTRACT

Regional DNA hypermethylation and global DNA hypomethylation are 2 epigenetic alterations associated with colorectal cancers. However, their correlation with microsatellite instability (MSI) and chromosomal instability (CIN) in colorectal cancer, and their relationship with chromatin conformation and histone modification are not clear. In this study, we analyzed regional and global methylation in 16 cell lines and 64 primary colorectal cancers. We found that MSI and CIN are 2 alternative events in most cell lines and tumors. Furthermore, regional hypermethylation and global hypomethylation are also alternative events in most cases. We also observed a strong correlation between MSI and regional hypermethylation and between CIN and global hypomethylation. We further analyzed chromatin conformation and histone acetylation in cell lines with CIN or MSI. CIN cancers had open chromatin conformation and enriched histone acetylation in repetitive as well as in gene-specific regions. MSI cancers, on the other hand, had closed chromatin conformation and low levels of histone acetylation. After a MSI cell line was treated with 5-aza-2'-deoxycytidine or trichostatin A, the closed chromatin conformation became open, and histone acetylation was enriched. These observations support our hypothesis that in colorectal cancer, regional hypermethylation and global hypomethylation are associated with altered chromatin conformation and histone acetylation, which might have a causal correlation with MSI and CIN, respectively.


Subject(s)
Chromatin/metabolism , Chromosomal Instability , Colorectal Neoplasms/metabolism , DNA Methylation , Histones/metabolism , Microsatellite Repeats , Acetylation , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/genetics , DNA, Neoplasm/metabolism , Epigenesis, Genetic , Humans , Hydroxamic Acids/pharmacology , Immunoprecipitation , Loss of Heterozygosity , Protein Conformation
7.
Am J Physiol Gastrointest Liver Physiol ; 289(5): G949-59, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16227528

ABSTRACT

VIP exerts a spectrum of effects as a potent anti-inflammatory factor. In addition, VIP increases expression of MUC2, a major intestinal secretory mucin. We therefore investigated the effects of VIP on the promoter activity of the 5'-flanking region of the MUC2 gene. VIP activated MUC2 transcription in human colonic epithelial cells via cAMP signaling to ERK and p38. cAMP/Epac/Rap1/B-Raf signaling was not involved in MUC2 reporter activation. Furthermore, activation of MUC2 transcription was independent of many of the reported downstream effectors of G protein-coupled receptors, such as PKC, Ras, Raf, Src, calcium, and phosphoinositide 3-kinase. VIP induced cAMP response element-binding protein (CREB)/ATF1 phosphorylation, and this was prevented by treatment with inhibitors of either MEK or p38 and by PKA and MSK1 inhibitor H89. CREB/ATF1 and c-Jun were shown to bind to an oligonucleotide encompassing a distal, conserved CREB/AP1 site in the 5'-flanking region of the MUC2 gene, and this cis element was shown to mediate promoter reporter activation by VIP. This study has identified a new, functional cis element within the MUC2 promoter and also a new pathway regulating MUC2 expression, thus providing further insight into the molecular mechanism of VIP action in the colon. These findings are relevant to the normal biology of the colonic mucosa as well as to the development of VIP as a therapeutic agent for treatment of inflammatory bowel disease.


Subject(s)
Activating Transcription Factor 1/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Mucins/biosynthesis , Mucins/genetics , Up-Regulation/drug effects , Vasoactive Intestinal Peptide/pharmacology , Cell Line , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mucin-2 , Phosphorylation , Promoter Regions, Genetic , Signal Transduction , Transcription, Genetic , Vasoactive Intestinal Peptide/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Am J Gastroenterol ; 100(5): 1143-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15842591

ABSTRACT

BACKGROUND AND AIM: Screening adenomas for microsatellite instability (MSI) in patients younger than 40 yr of age has been recommended by the Bethesda Guidelines as a means of identifying patients at risk for hereditary nonpolyposis colorectal cancer (HNPCC). We sought to determine the rate of MSI in adenomas removed from individuals under 40 yr of age over a 5-yr period in a university general gastroenterology practice. METHODS: We identified patients between 18 and 39 yr of age with endoscopically removed adenomatous colorectal polyps. Patients with polyposis syndromes, inflammatory bowel disease, or colorectal carcinoma were excluded. A three-generation family history was obtained via telephone interview. Endoscopic and histology reports were reviewed. Adenomas were tested for MSI using the BAT26 and BAT40 microsatellite markers, and expression of the MSH2 and MLH1 proteins was assessed by immunostaining. RESULTS: A total of 34 patients had 46 adenomas removed endoscopically. Out of 34 patients, 14 (41%) had a family history of colorectal cancer and 3 were from Amsterdam criteria positive families. A total of 28 of 46 adenomas (61%) were distal to the splenic flexure. Polyps ranged in size from 2 to 20 mm and averaged 6.6 mm. Five polyps (11%) were tubulovillous adenomas, and the remainder were tubular adenomas. None of the polyps were serrated adenomas and none demonstrated high-grade dysplasia. Among the 40 adenomas available for testing, none demonstrated MSI using either BAT26 (0/40) or BAT40 (0/21), nor did any of the polyps tested demonstrate loss of either MSH2 or MLH1 expression (0/16). CONCLUSION: Screening adenomas from patients younger than 40 yr of age for MSI was ineffective in identifying potentially new cases of HNPCC. New strategies that improve on the current clinical and molecular screening methods should be developed so that at-risk individuals can be identified and referred for germline testing before developing their first cancer.


Subject(s)
Adenoma/genetics , Chromosomal Instability/genetics , Colonic Neoplasms/genetics , Microsatellite Repeats/genetics , Rectal Neoplasms/genetics , Adaptor Proteins, Signal Transducing , Adenoma/pathology , Adenoma, Villous/genetics , Adenoma, Villous/pathology , Adolescent , Adult , Base Pair Mismatch/genetics , Carrier Proteins , Colonic Neoplasms/pathology , Colonic Polyps/genetics , Colonic Polyps/pathology , Colonoscopy , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA-Binding Proteins/genetics , Female , Genetic Predisposition to Disease/genetics , Genetic Testing , Humans , Intestinal Polyps/genetics , Intestinal Polyps/pathology , Male , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/genetics , Nuclear Proteins , Proctoscopy , Proto-Oncogene Proteins/genetics , Rectal Neoplasms/pathology , Risk Factors
9.
Mol Cancer Res ; 2(9): 504-13, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15383629

ABSTRACT

The colonic epithelium contains three major types of mature cells, namely, absorptive, goblet, and enteroendocrine cells. These cells are maintained by a complex process of cell renewal involving progenitor and stem cells, and colon cancers develop when this process goes awry. Much is known about the genetic and epigenetic changes that occur in cancer; however, little is known as to the specific cell types involved in carcinogenesis. In this study, we expressed the SV40 Tag oncogene in the intestinal epithelium under the control of an intestinal trefoil factor (ITF) promoter. This caused tumor formation in the proximal colon with remarkable efficiency. ITFTag tumors were rapidly growing, multifocal, and invasive. ITFTag tumor cells express synaptophysin and contain dense core secretory granules, markers of neuroendocrine differentiation. The cell type involved in the early steps of ITFTag tumorigenesis was studied by examining partially transformed crypts that contained populations of both normal and dysplastic cells. The dysplastic cell population always expressed both Tag and synaptophysin. Cells expressing Tag alone were never observed; however, normal enteroendocrine cells expressing synaptophysin but not Tag were readily visualized. This suggests that ITFTag tumor cells originate from the enteroendocrine cell lineage following a transforming event that results in Tag expression. ITFTag tumors closely resemble human small cell carcinomas of the colon, suggesting the possibility that these tumors might be derived from the enteroendocrine cell lineage as well.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Carcinoma, Small Cell/pathology , Colonic Neoplasms/pathology , Mucins/genetics , Muscle Proteins/genetics , Peptides/genetics , Promoter Regions, Genetic/genetics , Animals , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/ultrastructure , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/ultrastructure , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Mice , Mice, Transgenic , Oncogenes/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trefoil Factor-2
10.
Genomics ; 83(5): 936-9, 2004 May.
Article in English | MEDLINE | ID: mdl-15081123

ABSTRACT

The complete genomic organization of the two mucin genes MUC2 and MUC6 was obtained by comparison of new and published mRNA sequences with newly available human genomic sequence. The two genes are located 38.5 kb apart in a head-to-head orientation within a gene complex on chromosome 11p15.5. The N-terminal organization of MUC6 is highly similar to that of MUC2, containing the D1, D2, D', and D3 Von Willebrand factor domains followed by the large tandem repeat domains located in exons 31 and 30, respectively. MUC6 has a much smaller C-terminal domain (101 amino acids) encoded by 2 exons containing only the CK domain, compared with MUC2, which has a C-terminal domain of 859 amino acids containing the D4, C, D, and CK domains, encoded by 19 exons. The gene structures agreed partially but not completely with predictions from gene prediction programs.


Subject(s)
Exons/genetics , Introns/genetics , Mucins/genetics , Chromosomes, Human, Pair 11/genetics , Computational Biology , Genomics , Humans , Molecular Sequence Data , Mucin-2 , Mucin-6 , Multigene Family/genetics , RNA, Messenger/genetics
11.
Clin Cancer Res ; 10(1 Pt 1): 191-5, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14734469

ABSTRACT

PURPOSE: The BRAF gene encodes a serine/threonine kinase and plays an important role in the mitogen-activated protein kinase signaling pathway. BRAF mutations in sporadic colorectal cancer with microsatellite instability (MSI) are more frequently detected than those in microsatellite stable cancer. In this study, we sought to compare the frequencies of BRAF mutations in sporadic colorectal cancer with MSI with those in hereditary nonpolyposis colorectal cancer (HNPCC). EXPERIMENTAL DESIGN: We analyzed BRAF mutations in 26 colorectal cancer cell lines, 80 sporadic colorectal cancers, and 20 tumors from HNPCC patients by DNA sequencing and sequence-specific PCR. The methylation status of the hMLH1 gene was measured by either sequencing or restriction enzyme digestion after NaHSO(3) treatment. RESULTS: We observed a strong correlation of BRAF mutation with hMLH1 promoter methylation. BRAF mutations were present in 13 of 15 (87%) of the colorectal cell lines and cancers with methylated hMLH1, whereas only 4 of 91 (4%) of the cell lines and cancers with unmethylated hMLH1 carried the mutations (P < 0.00001). Sixteen of 17 mutations were at residue 599 (V599E). A BRAF mutation was also identified at residue 463 (G463V) in one cell line. In addition, BRAF mutations were not found in any cancers or cell lines with K-ras mutations. In 20 MSI+ cancers from HNPCC patients, however, BRAF mutations were not detectable, including a subset of 9 tumors with negative hMLH1 immunostaining and methylated hMLH1. CONCLUSIONS: BRAF mutations are frequently present in sporadic colorectal cancer with methylated hMLH1, but not in HNPCC-related cancers. This discrepancy of BRAF mutations between sporadic MSI+ cancer and HNPCC might be used in a strategy for the detection of HNPCC families.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms/genetics , DNA Methylation , Mutation/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-raf/genetics , Adaptor Proteins, Signal Transducing , Carrier Proteins , DNA Repair , DNA, Neoplasm/genetics , Genes, ras/genetics , Genomic Instability , Humans , Immunoenzyme Techniques , Microsatellite Repeats/genetics , MutL Protein Homolog 1 , Nuclear Proteins , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins B-raf , Sensitivity and Specificity , Tumor Cells, Cultured
12.
J Biol Chem ; 278(49): 49600-9, 2003 Dec 05.
Article in English | MEDLINE | ID: mdl-12958310

ABSTRACT

Human intestinal mucin genes MUC3A and MUC3B are members of a membrane mucin gene family residing at chromosome 7q22. In this paper, we utilized genomic and cDNA cloning to elucidate the sequence of the 5'-region of the MUC3A gene including the gene promoter and the amino terminus coding sequence. Following its 21-residue signal peptide, the amino terminus of the mucin consists of a 233-residue Thr-, Ser-, and Pro-rich nonrepetitive sequence that is contiguous with its hypervariable domain of 375-residue repeats. RNase protection analysis and 5'-GeneRacer PCR indicated that MUC3A gene transcripts initiate from multiple start sites along a region spanning approximately 180 bases. The 5'-flanking region of the gene had promoter activity when fused to a luciferase reporter gene in all of the tested cell lines. This region contained binding sites for several transcription factors, including those implicated in the regulation of intestinal genes, but lacked a cognate TATA box. These features of the gene promoter may enable the gene to be expressed at variable levels in several cell types with different repertoires of transcription factors. We also utilized 5'-GeneRacer PCR to determine the sequence of the 5'-terminus of the MUC3B message. The amino termini of the MUC3A and MUC3B mucins are 91% conserved at the amino acid level. Thus, MUC3A and MUC3B have highly conserved amino and carboxyl termini, suggesting a recent duplication of the entire ancestral gene. It remains to be determined whether other members of the 7q22 membrane mucin gene family have amino-terminal domains similar to MUC3A and MUC3B.


Subject(s)
Intestinal Mucosa/metabolism , Mucins/genetics , Promoter Regions, Genetic , TATA Box , Transcription, Genetic , Amino Acid Sequence , Base Sequence , DNA, Complementary , Humans , Molecular Sequence Data , Mucin-3 , Polymerase Chain Reaction , Sequence Homology, Amino Acid
13.
Int J Oncol ; 23(3): 585-92, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12888891

ABSTRACT

The nucleic acid sequence of the human gene, MUC17, indicates that this mucin contains an SEA domain, a transmembrane domain, and putative N-glycosylation sites in the carboxyl terminus. Mucins that possess an SEA domain are usually proteolytically cleaved within that domain to yield two subunits, the smaller of which is associated with the surface membrane. Homogenates of ASPC-1 pancreatic cancer cells showed three main bands of immunoreactivity with alpha-SEA (a polyclonal antibody directed against a site downstream of the postulated cleavage site) after SDS-PAGE and Western blotting (38, 45, and 49 kDa). Experiments utilizing N-glycan specific hydrolases showed that the 38 kDa band contained high mannose glycans whereas the 45 and 49 kDa bands contained complex-type glycans. Only two smaller alpha-SEA reactive bands (30 and 32 kDa) were present after cells had been treated with the N-glycosylation inhibitor tunicamycin. Surface biotinylation studies showed that only the forms possessing complex-type N-glycans were localized to the cell surface. Both tunicamycin and brefeldin A, an inhibitor of protein transport, reduced surface localization. In summary, our results indicate that the surface localization of the smaller subunit of MUC17 is dependent on its N-glycosylation status.


Subject(s)
Glycosylation , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/chemistry , Mucins/biosynthesis , Mucins/chemistry , Amino Acid Sequence , Animals , Anti-Bacterial Agents/pharmacology , Antibodies/chemistry , Biotinylation , Blotting, Western , Brefeldin A/pharmacology , Cell Line, Tumor , Cell Membrane/metabolism , Detergents/pharmacology , Electrophoresis, Polyacrylamide Gel , Glycoside Hydrolases/metabolism , Humans , Hydrolases/chemistry , Immunohistochemistry , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Tertiary , Protein Synthesis Inhibitors/pharmacology , RNA/chemistry , Rabbits , Tunicamycin/pharmacology
14.
Pancreas ; 26(3): e48-54, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12657964

ABSTRACT

INTRODUCTION: Ductal adenocarcinoma of the pancreas has recently been suggested to arise from histologically identifiable ductal lesions known as pancreatic intraepithelial neoplasia (PanINs). Altered levels and patterns of mucin gene expression have been reported to occur in epithelial cancers. AIM: To examine the pattern of expression of membrane-associated mucins, MUC3 and MUC4, and a mucin-associated carbohydrate tumor antigen, sialyl Le(x), in these precursor lesions and ductal adenocarcinoma of the pancreas. METHODOLOGY: A total of 144 PanIN lesions and 85 cases of ductal adenocarcinoma of the pancreas were examined by using immunohistochemistry and in situ hybridization methods. RESULTS: MUC3 showed a progressive increase in expression in PanINs of increasing dysplasia and was also highly expressed in ductal adenocarcinoma. In contrast, neoexpression of MUC4 and sialyl Le(x) antigen was observed, mainly in PanIN-3 and ductal adenocarcinoma. In addition, a decrease in the expression of MUC3 and MUC4 was correlated with the degree of de-differentiation of the tumor. CONCLUSION: Aberrant expression of membrane mucins MUC3 and MUC4 and of a mucin-associated carbohydrate tumor antigen Sialyl Le(x) in PanINs and adenocarcinoma further supports the progression model for pancreatic adenocarcinoma.


Subject(s)
Carcinoma in Situ/metabolism , Mucins/metabolism , Oligosaccharides/metabolism , Pancreatic Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation , Disease Progression , Gene Expression , Humans , Immunohistochemistry , In Situ Hybridization , Mucin-3 , Mucin-4 , Mucins/genetics , Mucins/immunology , Oligosaccharides/genetics , Oligosaccharides/immunology , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA, Messenger/metabolism , Sialyl Lewis X Antigen
15.
Biochem J ; 372(Pt 2): 335-45, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12597771

ABSTRACT

The entire cDNA corresponding to the C-terminal cysteine-rich domain of the human MUC2 apomucin, after the serine- and threonine-rich tandem repeat, was expressed in Chinese-hamster ovary-K1 cells and in the human colon carcinoma cell line, LS 174T. The C-terminus was expressed as a fusion protein with the green fluorescent protein and mycTag sequences and the murine immunoglobulin kappa-chain signal sequence to direct the protein to the secretory pathway. Pulse-chase studies showed a rapid conversion of the C-terminal monomer into a dimer in both Chinese-hamster ovary-K1 and LS 174T cells. Disulphide-bond-stabilized dimers secreted into the media of both cell lines had a higher apparent molecular mass compared with the intracellular forms. The MUC2 C-terminus was purified from the spent culture medium and visualized by molecular electron microscopy. The dimer nature of the molecule was visible clearly and revealed that each monomer was attached to the other by a large globular domain. Gold-labelled antibodies against the mycTag or green fluorescent protein revealed that these were localized to the ends opposite to the parts responsible for the dimerization. The C-terminus expressed in LS 174T cells formed heterodimers with the full-length wild-type MUC2, but not with the MUC5AC mucin, normally expressed in LS 174T cells. The homodimers of the MUC2 C-termini were secreted continuously from the LS 174T cells, but no wild-type MUC2 secretion has been observed from these cells. This suggests that the information for sorting the MUC2 mucin into the regulated secretory pathway in cells having this ability is present in parts other than the C-terminus of MUC2.


Subject(s)
Mucins/metabolism , Animals , Blotting, Western , CHO Cells , Colonic Neoplasms/chemistry , Colonic Neoplasms/metabolism , Cricetinae , DNA Primers/chemistry , Dimerization , Disulfides/metabolism , Electrophoresis, Polyacrylamide Gel , Gastric Mucins/chemistry , Gastric Mucins/metabolism , Glycoside Hydrolases/metabolism , Glycosylation , Green Fluorescent Proteins , Humans , Hydrofluoric Acid/pharmacology , Immunoglobulin Isotypes/metabolism , Luminescent Proteins/metabolism , Microscopy, Electron , Mucin 5AC , Mucin-2 , Mucins/genetics , Mucins/immunology , Mutagenesis, Site-Directed , Plasmids , Polymerase Chain Reaction , Precipitin Tests , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Fusion Proteins , Transfection
16.
J Biol Chem ; 277(49): 47248-56, 2002 Dec 06.
Article in English | MEDLINE | ID: mdl-12374796

ABSTRACT

The N terminus of the human MUC2 mucin (amino acids 1-1397) has been expressed as a recombinant tagged protein in Chinese hamster ovary cells. The intracellular form was found to be an endoglycosidase H-sensitive monomer, whereas the secreted form was an oligomer that gave monomers upon disulfide bond reduction. The secreted MUC2 N terminus contained a trypsin-resistant core fragment. Edman sequencing and mass spectrometry of the peptides obtained localized this core fragment to the C-terminal end of the recombinant protein. This core retained its oligomeric nature with an apparent mass of approximately 240 kDa. Upon reduction, peptides of approximately 85 kDa were found, suggesting that the N terminus forms trimers. This interpretation was also supported by gel electrophoresis and gel filtration of the intact MUC2 N terminus. Electron microscopy revealed three globular domains each linked via an extended and flexible region to a central part in a trefoil-like manner. Immunostaining with gold-labeled antibodies localized the N-terminal end to the three globular structures, and the antibodies directed against the Myc and green fluorescent protein tags attached at the C terminus localized these to the stalk side of the central trefoil. The N terminus of the MUC2 mucin is thus assembled into trimers that contain proteolytically stable parts, suggesting that MUC2 can only be partly degraded by intestinal proteases and thus is able to maintain a mucin network protecting the intestine.


Subject(s)
Mucins/chemistry , Amino Acid Sequence , Animals , CHO Cells , Chromatography, Gel , Cricetinae , Dimerization , Disulfides , Electrophoresis, Polyacrylamide Gel , Genetic Vectors , Glycosylation , Hydrofluoric Acid/pharmacology , Immunoblotting , Microscopy, Electron , Molecular Sequence Data , Mucin-2 , Mucins/metabolism , Mutagenesis, Site-Directed , Peptides/chemistry , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypsin/pharmacology
17.
J Biol Chem ; 277(47): 45547-57, 2002 Nov 22.
Article in English | MEDLINE | ID: mdl-12237307

ABSTRACT

Transforming growth factor-beta (TGF-beta) and related factors are multifunctional cytokines that regulate diverse cellular processes, including proliferation, differentiation, apoptosis, and immune response. The involvement of TGF-beta receptor-mediated signaling in bacteria-induced up-regulation of mucin, a primary innate defensive response for mammalian airways, however, still remains unknown. Here, we report that the bacterium nontypeable Haemophilus influenzae (NTHi), an important human respiratory pathogen, utilizes the TGF-beta-Smad signaling pathway together with the TLR2-MyD88-TAK1-NIK-IKKbeta/gamma-IkappaBalpha pathway to mediate NF-kappaB-dependent MUC2 mucin transcription. The NTHi-induced TGF-beta receptor Type II phosphorylation occurred at as early as 5 min. Pretreatment of NTHi with TGF-beta neutralization antibody reduced up-regulation of MUC2 transcription. Moreover, functional cooperation of NF-kappaB p65/p50 with Smad3/4 appears to positively mediate NF-kappaB-dependent MUC2 transcription. These data are the first to demonstrate the involvement of TGF-beta receptor-mediated signaling in bacteria-induced up-regulation of mucin transcription, bring insights into the novel role of TGF-beta signaling in bacterial pathogenesis, and may lead to new therapeutic intervention of NTHi infections.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins , Gene Expression Regulation , Haemophilus influenzae/metabolism , Mucins/genetics , NF-kappa B/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Adaptor Proteins, Signal Transducing , Antigens, Differentiation/metabolism , Autocrine Communication , Cell Line , Genes, Bacterial , Genes, Reporter , Haemophilus influenzae/genetics , Humans , I-kappa B Kinase , I-kappa B Proteins/metabolism , Membrane Glycoproteins/metabolism , Models, Biological , Mucin-2 , Mucins/metabolism , Myeloid Differentiation Factor 88 , NF-KappaB Inhibitor alpha , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Serotyping , Smad Proteins , Toll-Like Receptor 2 , Toll-Like Receptors , Transcription, Genetic , NF-kappaB-Inducing Kinase
18.
J Biol Chem ; 277(36): 32624-31, 2002 Sep 06.
Article in English | MEDLINE | ID: mdl-12077118

ABSTRACT

MUC2 is a secretory mucin normally expressed by goblet cells of the intestinal epithelium. It is overexpressed in mucinous type colorectal cancers but down-regulated in colorectal adenocarcinoma. Phorbol 12-myristate 13-acetate (PMA) treatment of colon cancer cell lines increases MUC2 expression, so we have undertaken a detailed analysis of the effects of PMA on the promoter activity of the 5'-flanking region of the MUC2 gene using stably and transiently transfected promoter reporter vectors. Protein kinase C inhibitors (bisindolylmaleimide, calphostin C) and inhibitors of mitogen-activated protein/extracellular signal regulated kinase kinase (MEK) (PD98059 and U0126) suppressed up-regulation of MUC2. Src tyrosine kinase inhibitor PP2, a protein kinase A inhibitor (KT5720), and a p38 inhibitor (SB 203580) did not affect transcription. Western blotting and reverse transcription-PCR analysis confirmed these results. In addition, co-transfections with mutants of Ras, Raf, and MEK showed that the induction of MUC2 promoter activity by PMA required these three signaling proteins. Our results demonstrate that PMA activates protein kinase C, stimulating MAP kinase through a Ras- and Raf-dependent mechanism. An important role for nuclear factor kappaB (NF-kappaB) was also demonstrated using the inhibitor caffeic acid phenethyl ester and electrophoretic mobility shift assays. Such identification of pathways involved in MUC2 up-regulation by PMA in the HM3 colon cancer cell line may serve as a model for the effects of cytokines and growth factors, which regulate MUC2 expression during the progression of colorectal cancer.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Mucins/biosynthesis , NF-kappa B/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation , ras Proteins/metabolism , Binding Sites , Cell Line , Cell Nucleus/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Genes, Dominant , Humans , Luciferases/metabolism , Models, Biological , Mucin-2 , Mucins/genetics , Mutation , Promoter Regions, Genetic , Protein Kinase C/metabolism , RNA/metabolism , Signal Transduction , Time Factors , Transcription, Genetic , Transcriptional Activation , Tumor Cells, Cultured
19.
Gastroenterology ; 122(4): 940-7, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11910346

ABSTRACT

BACKGROUND & AIMS: Early onset colorectal cancer (CRC) is an important feature of hereditary nonpolyposis colorectal cancer (HNPCC). We sought to compare rates of genetically defined HNPCC among individuals with early onset CRC drawn from a high-risk clinic and a population-based cancer registry. METHODS: Probands with CRC diagnosed before 36 years of age were enrolled from a high-risk CRC clinic at the University of California, San Francisco (UCSF), and a population-based Kaiser Permanente (KP) Health Plan cancer registry. Probands provided cancer family histories and tumors for microsatellite instability (MSI) testing and MSH2/MLH1 protein immunostaining. Germline MSH2 and MLH1 mutational analysis was performed. RESULTS: Forty-three probands were enrolled from UCSF and 23 from KP. The UCSF and KP probands had similar median age of onset of CRC (30 vs. 31 years) and the percentage with any personal or family history of another HNPCC-related cancer (70% vs. 74%). However, 28 of 40 (70%) of the UCSF tumors were MSI-H compared with 6 of 18 (33%) of KP tumors (P = 0.01), and 13 germline MSH2 or MLH1 mutations were found in the UCSF group compared with 0 in the KP group (P = 0.0001). In a multivariate analysis, institution (P = 0.002) and the total number of colorectal cancers in the family (P = 0.0001) were independent predictors of MSH2 or MLH1 mutation. CONCLUSIONS: Family history of cancer is an important feature of HNPCC, even among individuals with early onset CRC. Caution must be undertaken when extrapolating data regarding HNPCC from high-risk clinic populations to the general population.


Subject(s)
Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , DNA-Binding Proteins , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Age of Onset , Ambulatory Care Facilities , Carrier Proteins , Colorectal Neoplasms/chemistry , DNA Mutational Analysis , Germ-Line Mutation , Humans , Immunohistochemistry , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Nuclear Proteins , Predictive Value of Tests , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Registries , Risk Factors
20.
Biochem Biophys Res Commun ; 291(3): 466-75, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11855812

ABSTRACT

Membrane mucins have several functions in epithelial cells including cytoprotection, extravasation during metastases, maintenance of luminal structure, and signal transduction. In this paper we describe a large membrane mucin expressed in the normal intestine. This novel mucin, designated MUC17, contains an extended, repetitive extracellular glycosylation domain and a carboxyl terminus with two EGF-like domains, a SEA module domain, a transmembrane domain, and a cytoplasmic domain with potential serine and tyrosine phosphorylation sites. RNA blot analysis and in situ hybridization indicates that MUC17 is expressed in select pancreatic and colon cancer cell lines and in intestinal absorptive cells. Radiation hybrid mapping localized MUC17 to chromosome 7q22 where it resides in close proximity with three other membrane mucin genes, MUC3A, MUC3B, and MUC12. Thus, these membrane mucins reside together in a gene cluster, but are expressed in different tissues and are likely to have different functions as well.


Subject(s)
Mucins/biosynthesis , Mucins/genetics , Amino Acid Sequence , Animals , Chromosomes, Human, Pair 7 , Cloning, Molecular , Cricetinae , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/metabolism , Humans , In Situ Hybridization , Intestinal Mucosa/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , Mucins/chemistry , Phylogeny , Protein Structure, Tertiary , RNA, Messenger/biosynthesis , Radiation Hybrid Mapping , Sequence Alignment , Sequence Homology, Amino Acid , Tissue Distribution , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL