Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Cell Death Dis ; 7: e2124, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26938296

ABSTRACT

Increasing studies suggest that ceramides differing in acyl chain length and/or degree of unsaturation have distinct roles in mediating biological responses. However, still much remains unclear about regulation and role of distinct ceramide species in the immune response. Here, we demonstrate that alkaline ceramidase 3 (Acer3) mediates the immune response by regulating the levels of C18:1-ceramide in cells of the innate immune system and that Acer3 deficiency aggravates colitis in a murine model by augmenting the expression of pro-inflammatory cytokines in myeloid and colonic epithelial cells (CECs). According to the NCBI Gene Expression Omnibus (GEO) database, ACER3 is downregulated in immune cells in response to lipopolysaccharides (LPS), a potent inducer of the innate immune response. Consistent with these data, we demonstrated that LPS downregulated both Acer3 mRNA levels and its enzymatic activity while elevating C(18:1)-ceramide, a substrate of Acer3, in murine immune cells or CECs. Knocking out Acer3 enhanced the elevation of C(18:1)-ceramide and the expression of pro-inflammatory cytokines in immune cells and CECs in response to LPS challenge. Similar to Acer3 knockout, treatment with C(18:1)-ceramide, but not C18:0-ceramide, potentiated LPS-induced expression of pro-inflammatory cytokines in immune cells. In the mouse model of dextran sulfate sodium-induced colitis, Acer3 deficiency augmented colitis-associated elevation of colonic C(18:1)-ceramide and pro-inflammatory cytokines. Acer3 deficiency aggravated diarrhea, rectal bleeding, weight loss and mortality. Pathological analyses revealed that Acer3 deficiency augmented colonic shortening, immune cell infiltration, colonic epithelial damage and systemic inflammation. Acer3 deficiency also aggravated colonic dysplasia in a mouse model of colitis-associated colorectal cancer. Taken together, these results suggest that Acer3 has an important anti-inflammatory role by suppressing cellular or tissue C(18:1)-ceramide, a potent pro-inflammatory bioactive lipid and that dysregulation of ACER3 and C(18:1)-ceramide may contribute to the pathogenesis of inflammatory diseases including cancer.


Subject(s)
Alkaline Ceramidase/genetics , Colitis/etiology , Immunity, Innate/drug effects , Lipopolysaccharides/toxicity , Alkaline Ceramidase/deficiency , Animals , Cell Transformation, Neoplastic , Ceramides/analysis , Ceramides/metabolism , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Cytokines/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Substrate Specificity , Up-Regulation/drug effects
2.
Cell Death Differ ; 18(4): 632-44, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21052096

ABSTRACT

We have previously shown that a non-toxic noscapinoid, EM011 binds tubulin without altering its monomer/polymer ratio. EM011 is more active than the parent molecule, noscapine, in inducing G2/M arrest, inhibiting cellular proliferation and tumor growth in various human xenograft models. However, the mechanisms of mitotic-block and subsequent cell death have remained elusive. Here, we show that EM011-induced attenuation of microtubule dynamics was associated with impaired association of microtubule plus-end tracking proteins, such as EB1 and CLIP-170. EM011 treatment then led to the formation of multipolar spindles containing 'real' centrioles indicating drug-induced centrosome amplification and persistent centrosome declustering. Centrosome amplification was accompanied by an upregulation of Aurora A and Plk4 protein levels, as well as a surge in the kinase activity of Aurora A, suggesting a deregulation of the centrosome duplication cycle. Cell-cycle phase-specific experiments showed that the 'cytotoxicity-window' of the drug encompasses the late S-G2 period. Drug-treatment, excluding S-phase, not only resulted in lower sub-G1 population but also attenuated centrosome amplification and spindle multipolarity, suggesting that drug-induced centrosome amplification is essential for maximal cell death. Subsequent to a robust mitotic arrest, EM011-treated cells displayed diverse cellular fates suggesting a high degree of intraline variation. Some 'apoptosis-evasive' cells underwent aberrant cytokinesis to generate rampant aneuploidy that perhaps contributed to drug-induced cell death. These data indicate that spindle multipolarity induction by means of centrosome amplification has an exciting chemotherapeutic potential that merits further investigation.


Subject(s)
Apoptosis , Centrosome/physiology , Dioxoles/pharmacology , Isoquinolines/pharmacology , Microtubules/metabolism , Spindle Apparatus/physiology , Tubulin Modulators/pharmacology , Aurora Kinases , Cell Line, Tumor , G1 Phase , G2 Phase , Humans , Microtubule-Associated Proteins/metabolism , Microtubules/ultrastructure , Mitosis , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , S Phase , Up-Regulation
3.
Oncogene ; 29(13): 1929-40, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20062083

ABSTRACT

The p53 tumor suppressor inhibits the proliferation of cells that undergo prolonged activation of the mitotic checkpoint. However, the function of this antiproliferative response is not well defined. Here, we report that p53 suppresses structural chromosome instability after mitotic arrest in human cells. In both HCT116 colon cancer cells and normal human fibroblasts, DNA breaks occurred during mitotic arrest in a p53-independent manner, but p53 was required to suppress the proliferation and structural chromosome instability of the resulting polyploid cells. In contrast, cells made polyploid without mitotic arrest exhibited neither significant structural chromosome instability nor p53-dependent cell cycle arrest. We also observed that p53 suppressed both the frequency and structural chromosome instability of spontaneous polyploids in HCT116 cells. Furthermore, time-lapse videomicroscopy revealed that polyploidization of p53(-/-) HCT116 cells is frequently accompanied by mitotic arrest. These data suggest that a function of the p53-dependent postmitotic response is the prevention of structural chromosome instability after prolonged activation of the mitotic checkpoint. Accordingly, our study suggests a novel mechanism of tumor suppression for p53, as well as a potential function for p53 in the outcome of antimitotic chemotherapy.


Subject(s)
Cell Cycle/physiology , Chromosomal Instability/genetics , Colonic Neoplasms/pathology , Mitosis , Polyploidy , Tumor Suppressor Protein p53/physiology , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Survival , Colonic Neoplasms/genetics , HCT116 Cells/pathology , Humans , Mitosis/physiology , Neoplasm Proteins/metabolism , Neoplasm Proteins/pharmacology , Tumor Suppressor Protein p53/genetics
4.
Oncogene ; 28(9): 1197-205, 2009 Mar 05.
Article in English | MEDLINE | ID: mdl-19137014

ABSTRACT

Krüppel-like factor 4 (KLF4) is a zinc-finger transcription factor with tumor suppressive activity in colorectal cancer. Here, we investigated whether KLF4 is involved in maintaining genetic stability in mouse embryonic fibroblasts (MEFs) isolated from mice wild type (+/+), heterozygous (+/-), or homozygous (-/-) for the Klf4 alleles. Compared to Klf4(+/+) and Klf4(+/-) MEFs, Klf4(-/-) MEFs had both a higher level of apoptosis and rate of proliferation. Quantification of chromosome numbers showed that Klf4(-/-) MEFs were aneuploid. A higher number of Klf4(-/-) MEFs exhibited gamma-H2AX foci and had higher amounts of gamma-H2AX compared to controls. Cytogenetic analysis demonstrated the presence of numerous chromosome aberrations including dicentric chromosomes, chromatid breaks, and double minute chromosomes in Klf4(-/-) cells but in few, if any, Klf4(+/+) or Klf4(+/-) MEFs. Approximately 25% of Klf4(-/-) MEFs exhibited centrosome amplification in contrast to the less than 5% of Klf4(+/+) or Klf4(+/-) MEFs. Finally, only Klf4(-/-) MEFs were capable of anchorage-independent growth. Taken together, these findings demonstrate that MEFs null for the Klf4 alleles are genetically unstable, as evidenced by the presence of aneuploidy, chromosome aberration and centrosome amplification. The results support a crucial role for KLF4 in maintaining genetic stability and as a tumor suppressor.


Subject(s)
Kruppel-Like Transcription Factors/genetics , Aneuploidy , Animals , Cell Division , Centrosome , Chromosome Aberrations , DNA Damage , Heterozygote , Homozygote , Kruppel-Like Factor 4 , Mice , Mice, Knockout
5.
Oncogene ; 26(16): 2365-73, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17016435

ABSTRACT

In response to gamma-radiation-induced DNA damage, organisms either activate cell cycle checkpoint and repair machinery or undergo apoptosis to eliminate damaged cells. Although previous studies indicated that the tumor suppressor p53 is critically involved in mediating both responses, how a cell decides which pathway to take is not well established. The zinc-finger-containing transcription factor, Krüppel-like factor 4 (KLF4), is a crucial mediator for the checkpoint functions of p53 after gamma-irradiation and does so by inhibiting the transition from the G(1) to S and G(2) to M phases of the cell cycle. Here, we determined the role of KLF4 in modulating the apoptotic response following gamma-irradiation. In three independent cell systems including colorectal cancer cells and mouse embryo fibroblasts in which expression of KLF4 could be manipulated, we observed that gamma-irradiated cells underwent apoptosis if KLF4 was absent. In the presence of KLF4, the degree of apoptosis was significantly reduced and cells resorted to checkpoint arrest. The mechanism by which KLF4 accomplished this antiapoptotic effect is by activating expression of the cell cycle arrest gene, p21(WAF1/CIP1), and by inhibiting the ability of p53 to transactivate expression of the proapoptotic gene, BAX. Results of our study illustrate an unexpected antiapoptotic function of KLF4, heretofore considered a tumor suppressor in colorectal cancer, and suggest that KLF4 may be an important determinant of cell fate following gamma-radiation-induced DNA damage.


Subject(s)
Apoptosis/radiation effects , DNA, Neoplasm/radiation effects , Kruppel-Like Transcription Factors/physiology , Animals , COS Cells , Cell Cycle/radiation effects , Cell Line, Tumor , Chlorocebus aethiops , DNA Damage , DNA Primers , DNA, Neoplasm/genetics , Flow Cytometry , Gamma Rays , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/radiation effects , Promoter Regions, Genetic , Transfection , Tumor Suppressor Protein p53/radiation effects , bcl-2-Associated X Protein/genetics
6.
Oncogene ; 20(35): 4884-90, 2001 Aug 09.
Article in English | MEDLINE | ID: mdl-11521200

ABSTRACT

Gut-enriched Krüppel-like factor (GKLF or KLF4) is a zinc finger-containing, epithelial-specific transcription factor, that functions as a suppressor of cell proliferation. We previously showed that GKLF expression is decreased in intestinal and colonic adenomas, respectively, from multiple intestinal neoplasia (Min) mice and familial adenomatous polyposis (FAP) patients. This study shows that GKLF is induced upon activation of the adenomatous polyposis coli (APC) gene. However, among several human colon cancer cell lines surveyed, expression of GKLF is lowest in RKO, a line with wild-type APC and beta-catenin. RKO contains a mutated allele that encodes the putative tumor suppressor homeodomain protein, CDX2. We show that wild-type CDX2 activates the GKLF promoter and that the mutated CDX2 has a dominant negative effect on wild-type function. Our results may help explain the exceedingly low levels of GKLF expression detected in this cell line, which may in turn contribute to the tumor phenotype.


Subject(s)
Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , HMGB Proteins , Homeodomain Proteins/physiology , Trans-Activators , Transcription Factors/genetics , CDX2 Transcription Factor , Cytoskeletal Proteins/physiology , Genes, APC , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Promoter Regions, Genetic , RNA, Messenger/analysis , TCF Transcription Factors , Transcription Factor 7-Like 1 Protein , Tumor Cells, Cultured , beta Catenin
7.
J Biol Chem ; 276(32): 30423-8, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11390382

ABSTRACT

Krüppel-like factor 4 (KLF4) is an epithelial cell-enriched, zinc finger-containing transcription factor, the expression of which is associated with growth arrest. Previous studies show that constitutive expression of KLF4 inhibits DNA synthesis but the manner by which KLF4 exerts this effect is unclear. In the present study, we developed a system in which expression of KLF4 is controlled by a promoter that is induced upon treatment of cells containing the receptors for the insect hormone, ecdysone, with ponasterone A, an ecdysone analogue. The rate of proliferation of a stably transfected colon cancer cell line, RKO, was significantly decreased following addition of ponasterone A when compared with untreated cells. Flow cytometric analyses indicated that the inducible expression of KLF4 caused a block in the G(1)/S phase of the cell cycle. A similar block was observed when ecdysone receptor-containing RKO cells were infected with a replication-defective recombinant adenovirus containing an inducible KLF4 and treated with ponasterone A. Results of these studies provide evidence that the inhibitory effect of KLF4 on cell proliferation is mainly exerted at the G(1)/S boundary of the cell cycle.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/pharmacology , Ecdysterone/analogs & derivatives , G1 Phase , S Phase , Transcription Factors/chemistry , Transcription Factors/pharmacology , Adenoviridae/genetics , Animals , Blotting, Northern , Blotting, Western , CHO Cells , Cell Cycle , Cell Division , Cell Line , Cell Separation , Cricetinae , DNA/metabolism , DNA-Binding Proteins/genetics , Drosophila , Ecdysone/pharmacology , Ecdysterone/pharmacology , Fibroblasts/metabolism , Flow Cytometry , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Plasmids/metabolism , Promoter Regions, Genetic , Time Factors , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured
8.
J Biol Chem ; 276(10): 6897-900, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11152667

ABSTRACT

Intestinal-enriched Krüppel-like factor (IKLF or KLF5) belongs to the family of mammalian Krüppel-like transcription factors. Previous studies indicate that expression of IKLF is enriched in the proliferating crypt epithelial cells of the intestinal tract. However, the biological function of IKLF is unknown. In the current study, we have shown that the level of IKLF mRNA was nearly undetectable in serum-deprived NIH3T3 fibroblasts but became acutely and significantly increased upon the addition of fetal bovine serum or the phorbol ester, PMA. This induction required protein synthesis because it was prevented by cycloheximide. Transfection of IKLF into NIH3T3 cells resulted in the formation of foci in a manner similar to that caused by the activated Ha-ras oncogene. Constitutive expression of IKLF in transfected NIH3T3 cells significantly increased the rate of proliferation when compared with cells transfected with an empty vector. The growth of IKLF-transfected cells was no longer inhibited by cell-cell contact or by low serum content. Moreover, these cells proliferated in an anchorage-independent fashion. We conclude that IKLF encodes a delayed early response gene product that positively regulates cellular proliferation and may give rise to a transformed phenotype when overexpressed.


Subject(s)
Trans-Activators/physiology , 3T3 Cells , Animals , Blotting, Northern , Blotting, Western , Cattle , Cell Communication , Cell Differentiation , Cell Division , Culture Media, Serum-Free/pharmacology , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Genetic Vectors , Intestinal Mucosa/metabolism , Kruppel-Like Transcription Factors , Mice , Phenotype , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Messenger/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transfection
9.
FEBS Lett ; 476(3): 203-7, 2000 Jul 07.
Article in English | MEDLINE | ID: mdl-10913614

ABSTRACT

Gut-enriched Krüppel-like factor (GKLF) is a zinc finger-containing transcription factor, the expression of which is associated with growth arrest. We compared Gklf expression in intestinal and colonic adenomas to normal mucosa in multiple intestinal neoplasia (Min) mice and familial adenomatous polyposis (FAP) patients, respectively, using semi-quantitative RT-PCR. In Min mice, the level of Gklf transcript is highest in normal-appearing intestinal tissues and decreases as the size of the adenoma increases. In FAP patients, the level of GKLF transcript is lower in adenomas compared to paired normal-appearing mucosa from the same patient or normal colonic mucosa from control individuals without FAP. The possibility of DNA methylation as a cause for the decreased expression of Gklf in adenomas of Min mice was investigated by methylation-specific PCR. Results indicate that the Gklf gene is not methylated in either normal or tumorous tissues. The findings of our study are therefore consistent with the potential role of GKLF as a negative growth regulator of gut epithelial cells.


Subject(s)
Adenoma/genetics , Adenomatous Polyposis Coli/genetics , DNA-Binding Proteins , Growth Inhibitors/genetics , Intestinal Neoplasms/genetics , Transcription Factors/genetics , Animals , Base Sequence , Case-Control Studies , DNA Methylation , DNA Primers/genetics , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Down-Regulation , Female , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Polymerase Chain Reaction , Zinc Fingers/genetics
10.
J Biol Chem ; 275(24): 18391-8, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10749849

ABSTRACT

An important mechanism by which the tumor suppressor p53 maintains genomic stability is to induce cell cycle arrest through activation of the cyclin-dependent kinase inhibitor p21(WAF1/Cip1) gene. We show that the gene encoding the gut-enriched Krüppel-like factor (GKLF, KLF4) is concurrently induced with p21(WAF1/Cip1) during serum deprivation and DNA damage elicited by methyl methanesulfonate. The increases in expression of both Gklf and p21(WAF1/Cip1) due to DNA damage are dependent on p53. Moreover, during the first 30 min of methyl methanesulfonate treatment, the rise in Gklf mRNA level precedes that in p21(WAF1/Cip1), suggesting that GKLF may be involved in the induction of p21(WAF1/Cip1). Indeed, GKLF activates p21(WAF1/Cip1) through a specific Sp1-like cis-element in the p21(WAF1/Cip1) proximal promoter. The same element is also required by p53 to activate the p21(WAF1/Cip1) promoter, although p53 does not bind to it. Potential mechanisms by which p53 activates the p21(WAF1/Cip1) promoter include a physical interaction between p53 and GKLF and the transcriptional induction of Gklf by p53. Consequently, the two transactivators cause a synergistic induction of the p21(WAF1/Cip1) promoter activity. The physiological relevance of GKLF in mediating p53-dependent induction of p21(WAF1/Cip1) is demonstrated by the ability of antisense Gklf oligonucleotides to block the production of p21(WAF1/Cip1) in response to p53 activation. These findings suggest that GKLF is an essential mediator of p53 in the transcriptional induction of p21(WAF1/Cip1) and may be part of a novel pathway by which cellular responses to stress are modulated.


Subject(s)
Cyclins/genetics , DNA-Binding Proteins , Growth Inhibitors/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Zinc Fingers , 3T3 Cells , Animals , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Methyl Methanesulfonate/pharmacology , Mice , Polymerase Chain Reaction , Rabbits , Sp1 Transcription Factor/metabolism
11.
Nucleic Acids Res ; 28(5): 1106-13, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10666450

ABSTRACT

Gut-enriched Krüppel-like factor (GKLF or KLF4) is a pleiotropic (activating and repressive) transcription factor. This study characterizes the mechanisms of transactivation by GKLF. Using a GAL4 fusion assay, the activating domain of murine GKLF was localized to the 109 amino acid residues in the N-terminus. Site-directed mutagenesis showed that two adjacent clusters of acidic residues within this region are responsible for the activating effect. Transactivation by GKLF involves intermolecular interactions as demonstrated by the ability of wild-type, but not mutated, GKLF to compete with the N-terminal activation domain. In addition, wild-type adenovirus E1A, but not a mutated E1A that failed to bind p300/CBP, inhibited transactivation by the N-terminal 109 amino acids of GKLF, suggesting that p300/CBP are GKLF's interacting partners. A physical interaction between GKLF and CBP was demonstrated by glutathione- S -transferase pull-down and by in vivo co-immuno-precipitation experiments. We also showed that the two acidic amino acid clusters are essential for this interaction, since GKLF with mutations in these residues failed to co-immunoprecipitate with CBP. Importantly, the same mutations abrogated the ability of GKLF to suppress cell growth as determined by a colony suppression assay. These studies therefore provide plausible evidence for a structural and functional correlation between the transactivating and growth-suppressing effects of GKLF.


Subject(s)
DNA-Binding Proteins , Transcription Factors/genetics , Transcriptional Activation , Amino Acids , Animals , COS Cells , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Mutation , Protein Binding , Protein Conformation , Transcription Factors/metabolism , Transfection , Zinc Fingers
12.
Prostaglandins Other Lipid Mediat ; 60(1-3): 83-96, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10680778

ABSTRACT

Recent studies indicate that sulindac, a nonsteroidal anti-inflammatory drug (NSAID), lowers mucosal prostanoid levels and regresses colorectal adenomas in patients with familial adenomatous polyposis (FAP). To determine whether they are biomarkers for sulindac-mediated chemoprevention of colorectal adenomas, levels of 5 prostanoids [prostaglandin (PG) D2, PGE2, PGF2alpha, thromboxane B2, and 6-keto-PGF1alpha] in the normal-appearing rectal mucosa from 7 FAP patients with a history of subtotal colectomy and ileorectal anastomosis and 4 FAP patients without surgery, were measured in the absence or presence of exogenously added arachidonic acid before the initiation and at the end of 3 months of sulindac treatment. The addition of arachidonic acid resulted in a uniform increase in the levels of all 5 prostanoids although this increase was selectively attenuated in patients with ileorectal anastomosis who took sulindac. In the latter patients, arachidonic acid also augmented the inhibition of prostanoid synthesis by sulindac. In contrast, sulindac failed to attenuate the increase in prostanoid levels resulting from arachidonic acid in patients without previous surgery. Importantly, when measured in the presence of arachidonic acid, the reduction in the levels of all 5 prostanoids due to sulindac was statistically correlated with a reduction in the size and number of adenomas in the two groups of patients combined. These results suggest that tissue prostanoids measured in the presence of arachidonic acid may serve as sensitive and reliable biomarkers in monitoring the clinical responsiveness of FAP patients undergoing chemoprevention for colorectal neoplasia with NSAIDs.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/prevention & control , Prostaglandins/metabolism , Sulindac/therapeutic use , Adenomatous Polyposis Coli/pathology , Adolescent , Adult , Colorectal Neoplasms/metabolism , Female , Humans , Male , Middle Aged , Prostaglandins/biosynthesis
13.
Int J Biochem Cell Biol ; 32(11-12): 1103-21, 2000.
Article in English | MEDLINE | ID: mdl-11137451

ABSTRACT

Recent advances in molecular cloning have led to the identification of a large number of mammalian zinc finger-containing transcription factors that exhibit homology to the Drosophila melanogaster protein, Krüppel. Although the amino acid sequences in the zinc finger domains of these Krüppel-like factors (KLFs) are closely related to one another, the regions outside the zinc fingers of the proteins are usually unique. KLFs display seemingly different and broad biological properties with each functioning as an activator of transcription, a repressor or both. This review article provides a current phylogenetic classification of the identified KLFs to date. More importantly, the currently known biological activities of the KLFs in regulating transcription, cell proliferation, differentiation and development are summarized and compared. Further characterization of this interesting protein family should provide additional insights into the their respective regulatory role in various important biological processes.


Subject(s)
DNA-Binding Proteins , Repressor Proteins , Transcription Factors , Transcription, Genetic , Animals , Humans , Kruppel-Like Transcription Factors , Mammals , Phylogeny , Transcription Factors/chemistry , Transcription Factors/classification , Transcription Factors/genetics , Transcription Factors/physiology , Zinc Fingers
14.
Nucleic Acids Res ; 27(23): 4562-9, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10556311

ABSTRACT

Gut-enriched Krüppel-like factor (GKLF, KLF4) is an epithelial-specific transcription factor whose expression is associated with growth arrest. In order to understand the mechanisms regulating expression of the gene encoding GKLF, we isolated a genomic clone containing murine GKLF. The gene spans 5.3 kb and contains four exons. A major start site of transcription was mapped to an adenine residue 601 nt 5' of the translation initiation codon. An additional 1 kb of the 5'-flanking region was sequenced and found to contain multiple cis -elements homologous to the binding sites of several established transcription factors including Sp1, AP-1, Cdx, GATA, and USF. In particular, three closely spaced GC-boxes 5' of the TATA box resemble the established binding site for GKLF. DNase I protection and electrophoretic mobility shift assays verified that recombinant GKLF bound to each of the three GC-boxes. In co-transfection experiments, GKLF transactivated a reporter gene linked to the GKLF 1 kb 5'-flanking region, as did Sp1, Sp3 and Cdx-2. Mutations of one or both of the first and second GC-boxes in the promoter resulted in diminished transactivation by GKLF. These results demonstrate that the 5'-flanking sequence of the mouse GKLF gene functions as a promoter and is subject to autoregulation by its own gene product.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation , Transcription Factors/chemistry , Transcription Factors/genetics , Animals , Base Sequence , CHO Cells , COS Cells , Cricetinae , DNA Footprinting , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Transcriptional Activation , Zinc Fingers
15.
J Biol Chem ; 274(35): 24906-13, 1999 Aug 27.
Article in English | MEDLINE | ID: mdl-10455165

ABSTRACT

Most mammalian cells and some pathogenic bacteria are capable of adhering to collagenous substrates in processes mediated by specific cell surface adherence molecules. Crystal structures of collagen-binding regions of the human integrin alpha(2)beta(1) and a Staphylococcus aureus adhesin reveal a "trench" on the surface of both of these proteins. This trench can accommodate a collagen triple-helical structure and presumably represents the ligand-binding site (Emsley, J., King, S. L., Bergelson, J. M., and Liddington, R. C. (1997) J. Biol. Chem. 272, 28512-28517; Symersky, J., Patti, J. M., Carson, M., House-Pompeo, K., Teale, M., Moore, D., Jin, L., Schneider, A., DeLucas, L. J., Höök, M., and Narayana, S. V. L. (1997) Nat. Struct. Biol. 4, 833-838). We report here the crystal structure of the alpha subunit I domain from the alpha(1)beta(1) integrin. This collagen-binding protein also contains a trench on one face in which the collagen triple helix may be docked. Furthermore, we compare the collagen-binding mechanisms of the human alpha(1) integrin I domain and the A domain from the S. aureus collagen adhesin, Cna. Although the S. aureus and human proteins have unrelated amino acid sequences, secondary structure composition, and cation requirements for effective ligand binding, both proteins bind at multiple sites within one collagen molecule, with the sites in collagen varying in their affinity for the adherence molecule. We propose that (i) these evolutionarily dissimilar adherence proteins recognize collagen via similar mechanisms, (ii) the multisite, multiclass protein/ligand interactions observed in these two systems result from a binding-site trench, and (iii) this unusual binding mechanism may be thematic for proteins binding extended, rigid ligands that contain repeating structural motifs.


Subject(s)
Bacterial Proteins/chemistry , Collagen/chemistry , Integrins/chemistry , Staphylococcus aureus/metabolism , Adhesins, Bacterial/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Integrin alpha1beta1 , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Staphylococcus aureus/pathogenicity , Surface Plasmon Resonance
16.
J Biol Chem ; 274(18): 12454-60, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10212220

ABSTRACT

Decorin is ubiquitously distributed in the extracellular matrix of mammals and a member of the proteoglycan family characterized by a core protein dominated by leucine-rich repeat motifs. We show here that decorin extracted from bovine tissues under denaturing conditions or produced in recombinant "native" form by cultured mammalian cells has a high affinity for Zn2+ as demonstrated by equilibrium dialyses. The Zn2+-binding sites are localized to the N-terminal domain of the core protein that contains 4 Cys residues in a spacing reminiscent of a zinc finger. A recombinant 41-amino acid long peptide representing the N-terminal domain of decorin has full Zn2+ binding activity and binds two Zn2+ ions with an average KD of 3 x 10(-7) M. Binding of Zn2+ to this peptide results in a change in secondary structure as shown by circular dichroism spectroscopy. Biglycan, a proteoglycan that is structurally closely related to decorin contains a similar high affinity Zn2+-binding segment, whereas the structurally more distantly related proteoglycans, epiphycan and osteoglycin, do not bind Zn2+ with high affinity.


Subject(s)
Metalloproteins/chemistry , Proteoglycans/chemistry , Zinc/chemistry , Amino Acid Sequence , Animals , Base Sequence , Biglycan , Cattle , Decorin , Extracellular Matrix Proteins , Protein Binding , Protein Conformation , Proteoglycans/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Zinc/metabolism
17.
Curr Gastroenterol Rep ; 1(5): 449-54, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10980985

ABSTRACT

Colorectal cancer is a major cause of morbidity and mortality among types of cancer in the United States. Significant progress has been made in understanding the molecular mechanisms that lead to it. Much knowledge was obtained through study of genetic changes that occur in individuals with a familial predisposition to colorectal cancer, including familial adenomatous polyposis (FAP) and hereditary nonpolyposis colorectal cancer (HNPCC) syndromes. The gene with mutations that result in FAP has been identified as adenomatous polyposis coli (APC). Similarly, mutations in several genes that normally function in DNA mismatch repair result in HNPCC. Colorectal cancer is the result of accumulated mutations in several additional oncogenes or tumor suppressor genes, and this information leads to the formulation of a genetic model for the disease. Recent studies have also identified a relatively prevalent polymorphism in the APC gene in Ashkenazi Jews that is associated with an increased risk for colorectal cancer. These studies present a paradigm based on the APC mutation (APC I1307K) for the screening of cancer susceptibility genes in the population at large. Currently available techniques for genetic testing of colorectal cancer are also discussed in this review, along with their ethical implications.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Colorectal Neoplasms/epidemiology , Female , Genetic Predisposition to Disease/epidemiology , Genetic Testing , Humans , Incidence , Male , Molecular Biology , Risk Assessment
19.
J Biol Chem ; 273(28): 17917-25, 1998 Jul 10.
Article in English | MEDLINE | ID: mdl-9651398

ABSTRACT

The gut-enriched Krüppel-like factor (GKLF) is a newly identified zinc finger-containing transcription factor. Recent studies indicate that GKLF binds to a core DNA sequence of 5'-(G/A)(G/A)GG(C/T)G(C/T)-3', which is found in an endogenous cis element, the basic transcription element (BTE) of the cytochrome P-450IA1 (CYP1A1) promoter. The present study characterizes the ability of GKLF to regulate CYP1A1 expression. By electrophoretic mobility gel shift assay (EMSA) and methylation interference assay, GKLF was found to bind BTE in a manner similar to several other transcription factors known to interact with BTE including Sp1 and BTEB. Cotransfection studies in Chinese hamster ovary cells showed that GKLF inhibited the CYP1A1 promoter in a dose- and BTE-dependent manner. The same experiments also revealed that BTE was responsible for a significant portion of the CYP1A1 promoter activity. EMSA of nuclear extracts from Chinese hamster ovary cells showed that Sp1 and Sp3 were two major proteins that interacted with BTE. Additional cotransfection studies showed that GKLF inhibited Sp1-mediated activation of the CYP1A1 promoter. In contrast, GKLF enhanced Sp3-dependent suppression of the same promoter. Moreover, the ability of GKLF to inhibit Sp1-dependent transactivation was in part due to physical interaction of the two proteins. These findings indicate that GKLF is a negative regulator of the CYP1A1 promoter in a BTE-dependent fashion and that this inhibitory effect is in part mediated by physical interaction with Sp1.


Subject(s)
Cytochrome P-450 CYP1A1/genetics , Gene Expression Regulation, Enzymologic/physiology , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Transcription Factors/physiology , Zinc Fingers , Animals , Base Sequence , Binding, Competitive , CHO Cells , Cricetinae , DNA Primers , DNA-Binding Proteins/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Protein Binding , Rats , Recombinant Proteins/metabolism , Sp3 Transcription Factor , Transcription Factors/metabolism , Transcription, Genetic
20.
Cancer Res ; 58(8): 1750-3, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9563494

ABSTRACT

Recent studies indicate that nonsteroidal anti-inflammatory drugs have a chemopreventive effect against colorectal neoplasia. Nonsteroidal anti-inflammatory drugs inhibit cyclooxygenases, principal enzymes that mediate the formation of prostanoids. To determine whether prostanoids are involved in the pathogenesis of colorectal adenomas, we compared the levels of five major stable metabolic products of the cyclooxygenase pathway in the normal-appearing mucosa and in adenomas of patients with familial adenomatosis polyposis. Of 12 patients tested, 6 had elevated levels of at least one prostanoid in the adenomas. More importantly, the relative levels of three prostanoids [prostaglandin (PG)D2, PGE2, and 6-keto-PGF1alpha] were elevated in adenomas compared to normal-appearing mucosa from the same patients, and the resulting ratios were correlated with the size of the adenoma. These results suggest a role for prostanoids in progression of colorectal polyposis in familial adenomatosis polyposis patients.


Subject(s)
Adenoma/metabolism , Adenomatous Polyposis Coli/metabolism , Prostaglandins/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Adenoma/genetics , Adenomatous Polyposis Coli/genetics , Adult , Dinoprost/metabolism , Dinoprostone/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Male , Oxytocics/metabolism , Prostaglandin D2/metabolism , Thromboxane B2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...