Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 300(2): G327-33, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21109593

ABSTRACT

The colonic mucus layer serves as an important barrier and prevents colonic bacteria from invading the mucosa and cause inflammation. The regulation of colonic mucus secretion is poorly understood. The aim of this study was to investigate the role of the mucus barrier in induction of colitis. Furthermore, regulation of mucus secretion by luminal bacterial products was studied. The colon of anesthetized Muc2(-/-), Muc1(-/-), wild-type (wt), and germ-free mice was exteriorized, the mucosal surface was visualized, and mucus thickness was measured with micropipettes. Colitis was induced by DSS (dextran sodium sulfate, 3%, in drinking water), and disease activity index (DAI) was assessed daily. The colonic mucosa of germ-free and conventionally housed mice was exposed to the bacterial products LPS (lipopolysaccharide) and PGN (peptidoglycan). After DSS induction of colitis, the thickness of the firmly adherent mucus layer was significantly thinner after 5 days and onward, which paralleled the increment of DAI. Muc2(-/-) mice, which lacked firmly adherent mucus, were predisposed to colitis, whereas Muc1(-/-) mice were protected with significantly lower DAI by DSS compared with wt mice. The mucus barrier increased in Muc1(-/-) mice in response to DSS, whereas significantly fewer T cells were recruited to the inflamed colon. Mice housed under germ-free conditions had an extremely thin adherent colonic mucus layer, but when exposed to bacterial products (PGN or LPS) the thickness of the adherent mucus layer was quickly restored to levels observed in conventionally housed mice. This study demonstrates a correlation between decreasing mucus barrier and increasing clinical symptoms during onset of colitis. Mice lacking colonic mucus (Muc2(-/-)) were hypersensitive to DSS-induced colitis, whereas Muc1(-/-) were protected, probably through the ability to increase the mucus barrier but also by decreased T cell recruitment to the afflicted site. Furthermore, the ability of bacteria to regulate the thickness of the colonic mucus was demonstrated.


Subject(s)
Colitis/physiopathology , Colon/metabolism , Intestinal Mucosa/metabolism , Mucin-1/metabolism , Mucin-2/metabolism , Animals , Bacterial Translocation , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Colon/drug effects , Dextran Sulfate , Disease Susceptibility , Down-Regulation , Germ-Free Life , Intestinal Mucosa/drug effects , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-1/genetics , Mucin-2/genetics , Peptidoglycan/pharmacology , Severity of Illness Index , T-Lymphocytes/pathology
2.
Cancer Sci ; 99(11): 2113-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19037975

ABSTRACT

Goblet cell depletion and down-regulation of MUC2 expression are observed in a significant percentage of human non-mucinous colorectal adenocarcinomas. Direct evidence for the role of MUC2 in gastrointestinal tumor formation was demonstrated by a knockout of Muc2 in mice that resulted in the development of adenocarcinomas in the small and large intestine. The secretory phospholipase Pla2g2a is a protein that confers resistance to Apc(Min/+)-induced intestinal tumorigenesis. Like Muc2, in the large intestine Pla2g2a is exclusively expressed by the goblet cells and Pla2g2a's tumor resistance is also strongest in the large intestine. Possible genetic interactions between Muc2 and Pla2g2a were examined by creating C57BL/6-Muc2(-/-)Pla2g2a transgenic mice. Expression of a Pla2g2a transgene reduced tumorigenesis in the large intestine by 90% in male Muc2(-/-) mice and by nearly 100% in female Muc2(-/-) mice. Expression of Pla2g2a also inhibited tumor progression. Microarray gene expression studies revealed Pla2g2a target genes that modulate intestinal energy metabolism, differentiation, inflammation, immune responses and proliferation. Overall, results of the present study demonstrate an Apc-independent role for Pla2g2a in tumor resistance and indicate that Pla2g2a plays an important role, along with Muc2, in protection of the intestinal mucosa.


Subject(s)
Cell Transformation, Neoplastic/genetics , Group II Phospholipases A2/genetics , Mucin-2/genetics , Animals , Female , Gene Expression , Group II Phospholipases A2/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucin-2/metabolism , Oligonucleotide Array Sequence Analysis
3.
Cancer Res ; 61(16): 6297-302, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507085

ABSTRACT

Sulindac, a nonsteroidal anti-inflammatory drug, inhibits intestinal tumorigenesis in humans and rodents. Sulindac induced complex alterations in gene expression, but only 0.1% of 8063 sequences assayed were altered similarly by the drug in rectal biopsies of patients treated for 1 month and during response of colonic cells in culture. Among these changes was induction of the cyclin-dependent kinase inhibitor, p21(WAF1/cip1). In Apc1638(+/-) mice, targeted inactivation of p21 increased intestinal tumor formation in a gene-dose-dependent manner, but inactivation of p21 completely eliminated the ability of sulindac to both inhibit mitotic activity in the duodenal mucosa and to inhibit Apc-initiated tumor formation. Thus, p21 is essential for tumor inhibition by this drug. The array data can be accessed on the Internet at http://sequence.aecom.yu.edu/genome/.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclins/physiology , Intestinal Mucosa/drug effects , Sulindac/pharmacology , Animals , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Duodenum/cytology , Duodenum/drug effects , Duodenum/physiology , Female , Gene Dosage , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Gene Silencing/physiology , Genes, APC , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Male , Mice , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/drug therapy , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Rectum/cytology , Rectum/drug effects , Rectum/physiology
4.
Cancer Res ; 61(8): 3465-71, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309309

ABSTRACT

The beta-catenin TCF pathway is implicated in the regulation of colonic epithelial cell proliferation, but its role in the regulation of cell differentiation is unknown. The colon carcinoma cell line, Caco-2, spontaneously undergoes G(0)/G(1) cell cycle arrest and differentiates along the absorptive cell lineage over 21 days in culture. In parallel, we show that beta-catenin-TCF activity and complex formation are significantly down-regulated. The down-regulation of beta-catenin-TCF signaling was independent of APC, which we characterized as having a nonsense mutation in codon 1367 in Caco-2 cells, but was associated with a decrease in TCF-4 protein levels. Total beta-catenin levels increased during Caco-2 cell differentiation, although this was attributable to an increase in the membrane, E-cadherin-associated, fraction of beta-catenin. Importantly, down-regulation of beta-catenin-TCF signaling in undifferentiated Caco-2 cells by three different mechanisms, ectopic expression of E-cadherin, wild-type APC, or dominant negative TCF-4, resulted in an increase in the promoter activities of two genes that are well-established markers of cell differentiation, alkaline phosphatase and intestinal fatty acid binding protein. These studies demonstrate, therefore, that in addition to its established role in the regulation of cell proliferation, down-regulation of the beta-catenin-TCF pathway is associated with the promotion of a more-differentiated phenotype in colonic epithelial cells.


Subject(s)
Colon/cytology , Cytoskeletal Proteins/physiology , Signal Transduction/physiology , Trans-Activators , Transcription Factors/physiology , Adenomatous Polyposis Coli Protein , Caco-2 Cells , Cadherins/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage , Colon/metabolism , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Intestinal Absorption/physiology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Promoter Regions, Genetic , TCF Transcription Factors , Transcription Factor 7-Like 2 Protein , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection , beta Catenin
5.
Gastroenterology ; 120(4): 889-99, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231943

ABSTRACT

BACKGROUND & AIMS: The short-chain fatty acid butyrate induces cell cycle arrest, differentiation, and apoptosis in colon cancer cells, but often induces opposite effects in normal colonic epithelial cells. We determined whether response to butyrate is dependent on the basal differentiation status of colonic epithelial cells. METHODS: Caco-2 cells at progressive stages of differentiation were treated with butyrate, and endpoints were measured. RESULTS: Response of Caco-2 cells to butyrate was dependent on their differentiation status. Butyrate maximally stimulated cell cycle arrest, apoptosis, alkaline phosphatase activity, transepithelial resistance, cell migration, urokinase receptor expression, and interleukin 8 secretion in undifferentiated Caco-2 cells, whereas differentiated Caco-2 cells were essentially resistant to these effects. Consistently, butyrate selectively induced histone hyperacetylation in undifferentiated Caco-2 cells. This resistance was also observed during HT29cl.19A cell differentiation, but not in the nondifferentiating SW620 cell line. Finally, the rate of butyrate use significantly increased as Caco-2 cells underwent spontaneous differentiation. CONCLUSIONS: Colonic epithelial cells become progressively more refractory to the effects of butyrate during absorptive cell differentiation. We postulate that this resistance is caused by the rapid use of butyrate by differentiated Caco-2 cells, which likely results in low intracellular concentrations and subsequently in its inability to inhibit histone deacetylase.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Colon/cytology , Intestinal Mucosa/cytology , Acetylation , Alkaline Phosphatase/metabolism , Caco-2 Cells , Cell Differentiation/drug effects , Cell Line , Cell Movement/physiology , Colon/metabolism , Colon/physiology , Drug Resistance , Electric Impedance , Histones/metabolism , Humans , Interleukin-8/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiology , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator
6.
Cancer Res ; 61(2): 565-9, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11212250

ABSTRACT

Elimination of both alleles of the gene that encodes the cyclin kinase inhibitor p21(WAF1/cip1) increases the frequency and size of intestinal tumors in Apc1638+/- mice that inherit a mutant allele of the Apc gene, and intermediate effects are seen if a single p21 allele is inactivated. The increased tumor formation is associated with altered cell maturation in the intestinal mucosa of the p21-deficient mice--increased cell proliferation, and decreased apoptosis, and goblet cell differentiation--that is also a function of p21 gene dosage. Moreover, a Western-style diet that mimics principal risk factors for colon cancer (high fat and phosphate, low calcium and vitamin D) accelerates tumor formation in Apc1638+/- mice, and the loss of a single or both p21 alleles is additive with the tumor-promoting effects of this diet, resulting in more and larger tumors, and a highly significant decrease in survival time. Thus, p21 normally suppresses Apc-initiated tumor formation and is haplo-insufficient in this regard. This is consistent with recent reports that Apc initiates tumor formation by up-regulating c-myc expression through altered beta-catenin-Tcf signaling and that c-myc then up-regulates cdk4, whose activity is inhibited by p21. Decreased expression of p21 is also a marker of poor prognosis in patients, and the data presented suggest that dietary alterations in patients undergoing treatment for colon cancer might be highly effective in improving outcome.


Subject(s)
Cyclins/genetics , Cytoskeletal Proteins/genetics , Diet/adverse effects , Gastrointestinal Neoplasms/genetics , Intestinal Mucosa/pathology , Adenomatous Polyposis Coli Protein , Animals , Apoptosis , Cell Differentiation/genetics , Cell Division/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Female , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/pathology , Gene Silencing , Genotype , Immunohistochemistry , Intestinal Mucosa/chemistry , Intestinal Mucosa/metabolism , Male , Mice , Mice, Mutant Strains , Mucins/analysis , Mutation , Proliferating Cell Nuclear Antigen/analysis , Survival Analysis
7.
Cancer Res ; 61(2): 570-6, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11212251

ABSTRACT

Modulation of mucin gene expression is an important component both in the early steps of colon cancer development and in later tumor progression. Previous work from our laboratory and others has suggested that the Sp family of transcription factors may play an important role in the regulation of the human MUC2 gene. To determine whether this was an essential element, we extended our work to the cloning and analysis of 3.5 kb of the 5'-flanking region of the mouse Muc2 (mMuc2) gene. Comparative analysis between the mouse and human MUC2 promoter regions has identified a strong sequence homology between the mouse and human genes, including the presence of GC-rich boxes, the location and composition of which are maintained in the mouse and human genes. We show that these GC boxes are binding sites for Sp-family transcription factors and are functionally important since mithramycin, an inhibitor of Sp1/Sp3 binding, blocks MUC2 gene expression in HT29 cells. Furthermore, by a combination of gel shift analysis and site-directed mutagenesis, we have identified the relative contribution of individual GC boxes, and of the factors they bind, to the regulation of the mouse Muc2 promoter, which appears to be different in the mouse and human genes. Finally, we demonstrate by overexpressing Sp1 and Sp3 that the functional difference between the proximal promoter region of the MUC2 gene in the two species is not attributable to differential ability of this region to bind members of the Sp family of transcription factors, but rather to the different anatomy of the individual GC boxes in the mouse and human proximal promoters.


Subject(s)
DNA-Binding Proteins/metabolism , Mucins/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites/genetics , Binding, Competitive , Cloning, Molecular , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred Strains , Molecular Sequence Data , Mucin-2 , Mucins/metabolism , Mutation , Oligonucleotides/metabolism , Plicamycin/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/physiology , Protein Synthesis Inhibitors/pharmacology , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Nucleic Acid , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor , Transcription Factors/genetics , Transcription, Genetic , Tumor Cells, Cultured
8.
Oncogene ; 18(16): 2599-606, 1999 Apr 22.
Article in English | MEDLINE | ID: mdl-10353603

ABSTRACT

On 18q, frequently deleted in late stage colorectal cancers, a gene, Deleted in Colon Cancer (DCC), has been identified and postulated to play a role as a tumor suppressor gene. DCC is retained in the majority of mucinous tumors, which produce high levels of mucins, and seems to be preferentially expressed in intestinal goblet cells. To investigate whether DCC is related to mucin expression and can modulate the transformed phenotype, we introduced a full-length DCC cDNA into HT29 cells, which can be induced in vitro to express MUC2, the gene that encodes the major colonic mucin. Expression of DCC did not modulate constitutive or induced expression of MUC2, nor did DCC induce a mature goblet cell phenotype. However, HT29 clones expressing high and low levels of DCC protein showed a significant decrease in cell proliferation and tumorigenicity. Furthermore, increased shedding and an elevated rate of spontaneous apoptosis were associated with higher levels of expression of DCC. In summary, while restoration of DCC expression in a human colon carcinoma cell line did not influence expression of differentiation markers, DCC expression did affect the growth and tumorigenic properties of the cells suggesting that DCC can modulate the malignant phenotype of colon cancer.


Subject(s)
Adenocarcinoma/genetics , Cell Adhesion Molecules/biosynthesis , Cell Transformation, Neoplastic , Colonic Neoplasms/genetics , Genes, DCC , Tumor Suppressor Proteins , Cell Adhesion Molecules/genetics , Cell Differentiation , DCC Receptor , Goblet Cells/cytology , HT29 Cells , Humans , Loss of Heterozygosity , Mucin-2 , Mucins/biosynthesis , Mucins/genetics , Phenotype , Receptors, Cell Surface , Recombinant Proteins/biosynthesis
10.
Ann N Y Acad Sci ; 889: 20-31, 1999.
Article in English | MEDLINE | ID: mdl-10668479

ABSTRACT

Our early work using the first array and imaging methods for the quantitative analysis of the expression of 4000 cDNA sequences suggested that modulation of mitochondrial gene expression was a factor in determining whether colonic epithelial cells displayed a differentiated or transformed phenotype. We have since dissected a pathway in which mitochondrial function is a key element in determining the probability of cells undergoing cell-cycle arrest, lineage-specific differentiation, and cell death. Moreover, this pathway is linked to signaling through beta-catenin-Tcf, but in a manner that is independent of effects of the APC gene on beta-catenin-Tcf activity. Utilization of unique mouse genetic models of intestinal tumorigenesis has confirmed that mitochondrial function is an important element in generation of apoptotic cells in the colon in vivo and has demonstrated that modulation of cell death may be involved in intestinal tumor progression rather than initiation. Normal spatial and temporal patterns of cell proliferation, differentiation, and apoptosis in the colonic mucosa are determined by developmentally programmed genetic signals and external signals generated by homo- and heterotypic cell interactions, humoral agents, and lumenal contents. Mitochondrial function may play a pivotal role in integrating these signals and in determining probability of cells entering different maturation pathways. How this is accomplished is under investigation using high-density cDNA microarrays.


Subject(s)
Cell Transformation, Neoplastic , Neoplasms, Experimental/pathology , Neoplasms/pathology , Animals , Apoptosis , Cell Death , Cell Division , Humans , Mice
11.
J Biol Chem ; 272(12): 7968-76, 1997 Mar 21.
Article in English | MEDLINE | ID: mdl-9065467

ABSTRACT

The human MUC2 gene maps to chromosome 11p15, where three additional mucin genes have been located, and encodes the most abundant gastrointestinal mucin normally expressed in the intestinal goblet cell lineage. However, in pathological conditions, including colorectal cancer, MUC2 can be abnormally expressed. Therefore, it is of considerable interest to understand the regulation of the MUC2 gene and how the mechanism is altered in colon cancer. Toward this goal, we have isolated a group of overlapping clones (contig) spanning 85 kilobases harboring the entire MUC2 locus, including sequences located upstream of the gene. Detection of two DNase I-hypersensitive sites in the 5' region of the MUC2 gene suggests the presence of DNA regulatory elements. To better characterize this region, we have sequenced 12 kilobases of the upstream region and analyzed it for functional activity by cloning portions of it into a luciferase reporter vector and assaying for promoter/enhancer activity using a transient transfection assay. A fragment from the AUG translational initiation codon +1 to -848 confers maximal transcriptional activity in several intestinal cell lines. Elements located further upstream exert a negative effect on the expression of the reporter gene when tested in conjunction with homologous or heterologous promoters. The same pattern of expression is observed when the MUC2/luciferase constructs are transfected into HeLa cells, which do not express the endogenous MUC2 gene. However, the level of activity in HeLa cells is at least an order of magnitude higher, suggesting that additional sequences singularly or in combination are responsible for the tissue- and cell lineage-specific expression of MUC2. Finally, we have identified an additional mucin-like gene (MUCX), located upstream of MUC2. We show that this MUCX gene, that is transcribed in opposite orientation to that of MUC2, is expressed with a pattern distinct from that of MUC2, yet similar to that of MUC5B and MUC6, two additional mucin genes located at chromosome 11p15. Recent information on the order of the mucin genes at chromosome 11p15 suggests that MUCX may be MUC6, one of the already identified mucin genes, or a novel one, yet to be fully characterized.


Subject(s)
Mucins/genetics , Amino Acid Sequence , Base Sequence , Chromosomes, Human, Pair 11 , DNA, Recombinant , HeLa Cells , Humans , Molecular Sequence Data , Mucin-2 , Promoter Regions, Genetic , Sequence Homology, Amino Acid , Transfection
12.
Cell Growth Differ ; 6(6): 749-57, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7669730

ABSTRACT

The four different cell types present in the mature colon, absorptive enterocytes, mucus-secreting goblet cells, Paneth cells, and enteroendocrine cells, are believed to derive from a common precursor, the stem cell, anchored near the base of the crypt. Stem cell descendants undergo several rounds of cell division, creating a pool of transit cells that are committed to differentiation. The mechanisms by which committed cells are allocated to the different cell lineages of the intestine are poorly understood. We have used the colon carcinoma cell line HT29 and Cl.16E cells, a clonal derivative of HT29 cells, to investigate the regulation and pattern of expression of several markers (MUC2, MUC3, carcinoembryonic antigen, and alkaline phosphatase) that are associated with a more differentiated phenotype and that, in the mature cells, are lineage restricted. HT29 cells can express, upon exposure to the appropriate inducers, distinct intestinal specific markers; they are, therefore, considered multipotent, similar to the stem cells of the crypt. Conversely, Cl.16E cells are lineage restricted and respond to cell contact inhibition by expressing a fully differentiated goblet cell phenotype. We show that, in HT29 cells, different inducers (12-O-tetradecanoylphorbol-13-acetate, forskolin, and sodium butyrate) modulate specific sets of markers. Forskolin induces the expression of both MUC2 and MUC3, whereas 12-O-tetradecanoylphorbol-13-acetate is capable of inducing only MUC2, and sodium butyrate, only MUC3 gene expression. Carcinoembryonic antigen, a marker common to enterocytes and goblet cells; can be induced by all the agents, whereas the alkaline phosphatase gene, the expression of which is characteristic of enterocytes, is responsive solely to sodium butyrate treatment.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antigens, Differentiation/biosynthesis , Antigens, Neoplasm/biosynthesis , Carcinoma/pathology , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Alkaline Phosphatase/biosynthesis , Alkaline Phosphatase/genetics , Antigens, Differentiation/genetics , Antigens, Neoplasm/genetics , Biomarkers , Butyrates/pharmacology , Butyric Acid , Carcinoembryonic Antigen/biosynthesis , Carcinoembryonic Antigen/genetics , Cell Differentiation/drug effects , Colforsin/pharmacology , Humans , Intestinal Mucosa/pathology , Mucin-2 , Mucin-3 , Mucins/biosynthesis , Mucins/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured/drug effects
14.
Eur J Pharmacol ; 267(1): 95-103, 1994 Mar 15.
Article in English | MEDLINE | ID: mdl-7515824

ABSTRACT

The regulation of intestinal mucin secretion by cytokines, soluble factors released by mucosal activated immune cells, is so far unknown. The aim of the present study was (1) to investigate the regulatory effects of interferon-gamma on baseline and stimulated mucin secretion elicited by an increase in intracellular cAMP, either a short-term increase (induced by vasoactive intestinal peptide or by forskolin) or a long-term increase (cholera toxin-induced), and (2) to attempt to delineate the site of action of interferon-gamma. The in vitro model used was the human colonic goblet cell line Cl.16E, which has already been shown to respond to physiological secretagogues in terms of mucin secretion. We examined the effects of interferon-gamma 1) on mucin exocytosis, measured as release of [3H]glucosamine-labeled macromolecules trapped at the stacking/running gel interface of polyacrylamide gels, and 2) on mucin biosynthesis, examined at the RNA level using a cDNA probe directed to the MUC2 mucin gene. We demonstrated that, while interferon-gamma did not alter baseline Cl.16E mucin secretion and MUC2 gene expression, it strongly inhibited the protein kinase A-dependent secretory response to VIP, forskolin, or cholera toxin. However, interferon-gamma had no effect on the protein kinase A-dependent MUC2 over-expression induced by cholera toxin. We thus concluded that the target for interferon-gamma inhibition of cAMP-stimulated Cl.16E mucin secretion is distal to protein kinase A and might be a component of the exocytotic machinery. Together, our results establish interferon-gamma as a pharmacologically powerful tool to specifically inhibit stimulated secretory processes without affecting baseline secretion.


Subject(s)
Colon/drug effects , Cyclic AMP/pharmacology , Exocytosis/drug effects , Interferon-gamma/pharmacology , Mucins/drug effects , Blotting, Northern , Cell Survival , Cells, Cultured , Cholera Toxin/pharmacology , Colforsin/pharmacology , Colon/cytology , Electrophysiology , Gene Expression/drug effects , Humans , Mucins/genetics , Mucins/metabolism , RNA/analysis , Thymidine/metabolism , Vasoactive Intestinal Peptide/pharmacology
15.
J Biol Chem ; 268(19): 13956-61, 1993 Jul 05.
Article in English | MEDLINE | ID: mdl-7686147

ABSTRACT

We have investigated the regulation of the intestinal mucin gene MUC2 in HT29 cells. Surprisingly, sodium butyrate, an effective inducer of aspects of colonic cell differentiation in HT29 cells, fails to induce MUC2 during short-term exposure, despite the fact that it has been used to select stably differentiated clones of HT29 that resemble goblet cells and produce mucin. However, 12-O-tetradecanoylphorbol-13-acetate and forskolin, which trigger the protein kinase C- and A-dependent signal transduction pathways, respectively, are potent inducers of MUC2 gene expression. 12-O-Tetradecanoylphorbol-13-acetate and forskolin operate through distinct mechanisms, with the former requiring de novo protein synthesis and the latter not. Experiments using specific protein kinase inhibitors suggest that both inducers operate by triggering their respective signal transduction pathways. Nuclear runoff analyses suggest that post-transcriptional (rather than transcriptional) mechanisms are important in the accumulation of MUC2 mRNA. Finally, we show that in several cell lines from human mucinous tumors, characterized by elevated levels of mucin production, MUC2 expression is very high and constitutive compared to forskolin-treated HT29 cells. Thus, the different regulation of MUC2 in HT29 cells and in mucinous tumor cell lines may reflect molecular pathways that characterize colon carcinomas of different histology and pathology.


Subject(s)
Gene Expression Regulation, Neoplastic , Intestinal Mucosa/metabolism , Mucins/genetics , RNA, Messenger/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Adenocarcinoma , Bucladesine/pharmacology , Calcimycin/pharmacology , Colforsin/pharmacology , Colonic Neoplasms , DNA Probes , Ethers, Cyclic/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ionomycin/pharmacology , Ionophores/pharmacology , Kinetics , Okadaic Acid , RNA, Messenger/biosynthesis , RNA, Neoplasm/isolation & purification , RNA, Neoplasm/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects , Tumor Cells, Cultured
16.
Mol Cell Biol ; 10(12): 6273-82, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2147223

ABSTRACT

A DNA element located at positions -295 to -289 of the c-fos promoter (FAP site) is highly homologous to a consensus 12-O-tetradecanoyl phorbol-13-acetate-responsive element (TRE) and to a cyclic AMP (cAMP)-responsive element (CRE). We found that an oligonucleotide containing the FAP element was a transcription regulator which was distinct from both the TRE and CRE. When cloned in multiple copies in front of a reporter gene in HeLa cells, the FAP oligonucleotide was a powerful constitutive activator sequence. Conversely, in the same cells, reporter plasmids containing multiple copies of the TRE of the human metallothionein gene required phorbol esters for their induction. In PC12 cells, the FAP oligonucleotide was cAMP responsive. Its activity was mediated through a cAMP-dependent protein kinase II and did not rely on ongoing protein synthesis for activation. Adenovirus E1a proteins activated viral promoters through ATF (activation transcription factor) consensus binding sequences identical to the CRE. However, E1a repressed the FAP oligonucleotide-associated transcriptional activity in HeLa cells. In PC12 cells, E1a neither transactivated nor transrepressed the basal and cAMP-stimulated FAP activity. In contrast, the CRE of the human c-fos promoter located at -60 was weakly induced by cAMP and E1a in both HeLa and PC12 cells. We suggest that the FAP oligonucleotide acts through a factor(s) distinct from those employed by the TRE and CRE and that the FAP-associated protein factor(s) may differ in HeLa and PC12 cells in expression or posttranslational regulation.


Subject(s)
Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Transcription, Genetic , Base Sequence , Cyclic AMP Response Element-Binding Protein , DNA-Binding Proteins/genetics , Globins/genetics , HeLa Cells/drug effects , HeLa Cells/metabolism , Humans , Molecular Sequence Data , Oligonucleotide Probes , Plasmids , Proto-Oncogene Proteins c-fos , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Sequence Homology, Nucleic Acid , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/genetics , Transfection
17.
Oncogene ; 4(6): 707-13, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2543941

ABSTRACT

F9 embryonal carcinoma (EC) cells serve as a model system for early mammalian development. We have investigated the effect of the E1a gene products on the F9 cell differentiation program by stably introducing into these cells plasmids which express wild type or mutant forms of E1a. We have found that expression of the 12S E1a mRNA product results in the expression of the endoA gene, a marker specific for the differentiated phenotype of F9 cells, as well as an altered cell morphology associated with the differentiated state. These alterations are not observed in F9 cells which express the 13S E1a product. Other markers specific for the differentiated state are regulated normally in the E1a transformants following exposure to retinoic acid (R.A.) and dibutyryl cyclic AMP (dbcAMP). We discuss these results in the context of the well established transcriptional activities of the E1a proteins.


Subject(s)
Gene Expression Regulation , Oncogene Proteins, Viral/physiology , Adenovirus Early Proteins , Bucladesine/pharmacology , Cell Differentiation , Genetic Markers , Immunoblotting , Molecular Weight , Oncogene Proteins, Viral/genetics , Plasmids , Transfection , Tretinoin/pharmacology , Tumor Cells, Cultured
18.
Oncogene Res ; 5(1): 31-7, 1989.
Article in English | MEDLINE | ID: mdl-2780052

ABSTRACT

Utilizing F9 embryonal carcinoma cells as a model system for early mammalian development, we have studied the pattern of expression of the endogenous murine homolog of the human K-fgf/hst oncogene, which encodes a new member of the fibroblast growth factors (FGFs) family. The K-fgf mRNA is expressed in undifferentiated F9 cells and its level becomes undetectable upon the induction of differentiation. Furthermore, a growth-promoting activity with properties identical to those of K-FGF is present in the conditioned medium of F9 cells, but absent in that of differentiated cells. Shut-off of K-fgf expression is mediated at the transcriptional level. The acidic FGF gene is also expressed in undifferentiated F9 cells and down modulated once differentiation is induced. In contrast, int-2, another member of the FGF gene family, is transcriptionally induced in differentiated F9 cells. Our data suggest that single members of the FGF gene family may perform distinct functions in vivo, and that the physiological role of K-FGF may be related to early development.


Subject(s)
Fibroblast Growth Factors/genetics , Gene Expression Regulation , Neoplastic Stem Cells/metabolism , Proto-Oncogenes , Cell Differentiation , Embryonal Carcinoma Stem Cells , Fibroblast Growth Factors/analysis , Neoplastic Stem Cells/pathology , RNA, Messenger/analysis
20.
Mol Cell Biol ; 8(5): 2177-83, 1988 May.
Article in English | MEDLINE | ID: mdl-2968500

ABSTRACT

The E1a gene of adenovirus encodes two proteins, 289 and 243 amino acids long, which have positive (transactivator) and negative (enhancer repressor) RNA polymerase II transcriptional regulatory properties and cell transformation activities including cooperation with an activated ras gene. The E1a transforming functions more closely correlate with the repressor property than with transactivation in that both E1a proteins express the repressor and transformation functions while only the 289-amino-acid protein is an efficient transactivator. To understand whether the transcriptional regulatory activities of E1a are related to its ras cooperation activity, we generated a series of mutant E1a expression vectors by linker insertion mutagenesis of the 289-amino-acid protein. Here we describe a new class of mutants which although defective for enhancer repression still can cooperate with the ras oncogene in cell transformation. The mutants are also defective in transcription transactivation. Our data suggest that enhancer repression and transformation via ras cooperation are separate E1a functions and that cooperation with ras does not rely on either of the RNA polymerase II transcription regulatory functions of E1a. We also show that mutations which inactivate enhancer repression are not confirmed to a single critical domain necessary for repression. We therefore propose that the integrity of the overall configuration of the E1a proteins is important for the repression activity.


Subject(s)
Adenoviridae/physiology , Oncogene Proteins, Viral/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Transcription, Genetic , Adenoviridae/genetics , Adenovirus Early Proteins , Cell Transformation, Neoplastic/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , Oncogene Proteins, Viral/genetics , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins p21(ras) , RNA Polymerase II/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...