Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Diabetes Obes Metab ; 18(2): 142-51, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26462866

ABSTRACT

AIMS: To investigate the effects of specific retinoic acid receptor (RAR) agonists in diabetes and fatty liver disease. METHODS: Synthetic agonists for RARß2 were administered to wild-type (wt) mice in a model of high-fat-diet (HFD)-induced type 2 diabetes (T2D) and to ob/ob and db/db mice (genetic models of obesity-associated T2D). RESULTS: We show that administration of synthetic agonists for RARß2 to either wt mice in a model of HFD-induced T2D or to ob/ob and db/db mice reduces hyperglycaemia, peripheral insulin resistance and body weight. Furthermore, RARß2 agonists dramatically reduce steatosis, lipid peroxidation and oxidative stress in the liver, pancreas and kidneys of obese, diabetic mice. RARß2 agonists also lower levels of mRNAs involved in lipogenesis, such as sterol regulatory element-binding transcription factor 1 (SREBP1) and fatty acid synthase, and increase mRNAs that mediate mitochondrial fatty acid ß-oxidation, such as CPT1α, in these organs. RARß2 agonists lower triglyceride levels in these organs, and in muscle. CONCLUSIONS: Collectively, our data show that orally active, rapid-acting, high-affinity pharmacological agonists for RARß2 improve the diabetic phenotype while reducing lipid levels in key insulin target tissues. We suggest that RARß2 agonists should be useful drugs for T2D therapy and for treatment of hepatic steatosis.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hyperglycemia/prevention & control , Hypoglycemic Agents/therapeutic use , Liver/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Receptors, Retinoic Acid/agonists , Animals , Benzoates/therapeutic use , Biphenyl Compounds/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Drugs, Investigational/therapeutic use , Insulin Resistance , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Non-alcoholic Fatty Liver Disease/complications , Obesity/complications , Oxidative Stress/drug effects , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Receptors, Retinoic Acid/metabolism , Thiazoles/therapeutic use
2.
Curr Med Chem ; 18(29): 4440-53, 2011.
Article in English | MEDLINE | ID: mdl-21864277

ABSTRACT

Recurrent prostate cancer (PCa) remains a major clinical challenge. Invasive and metastatic PCa lesions often exhibit a partial and time-limited response to therapy before the cancer progresses and the patient succumbs to the disease. Despite recent advances in early diagnosis and treatment, approximately one-third of treated patients will relapse and become resistant to currently available treatments. In this review we evaluate current treatment practices and recent advances in therapy for localized prostate malignancy and advanced, metastatic prostate cancer. Some of the promising new drugs for PCa treatment include MDV3100, an androgen receptor (AR) antagonist that prevents androgens from binding to the AR and nuclear translocation and co-activator recruitment of the ligand-receptor complex; abiraterone, an orally administered drug that irreversibly inhibits a rate-limiting enzyme in androgen biosynthesis, CYP17; and several newer cytotoxic drugs (epothilones, satraplatin). Key new insights are that cancer stem cells play a role in PCa and that PCa cells are dependent on the AR for proliferation, even in the hormone refractory state of the disease. We also discuss potential molecular targets for new drug candidates for the treatment of metastatic PCa.


Subject(s)
Antineoplastic Agents/therapeutic use , Prostate/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/secondary , Androgen Receptor Antagonists/chemistry , Androgen Receptor Antagonists/pharmacology , Androgen Receptor Antagonists/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Signal Transduction/drug effects
3.
Neuroscience ; 126(4): 967-78, 2004.
Article in English | MEDLINE | ID: mdl-15207330

ABSTRACT

Laminins are the major glycoproteins present in basement membrane, a type of extracellular matrix. We showed that the LAMB1 gene, which encodes the laminin beta1 subunit, is transcriptionally activated by retinoic acid in embryonic stem cells. However, little information is available concerning LAMB1 developmental regulation and spatial expression in the adult mouse brain. In this study we used transgenic mice expressing different lengths of LAMB1 promoter driving beta-galactosidase to investigate developmental and adult transcriptional regulation in the regions of the brain in which the laminin beta1 protein is expressed. CNS expression was not observed in transgenic mice carrying a 1.4LAMB1betagal construct. Mice carrying a 2.5LAMB1betagal construct expressed the LAMB1 transgene, as assayed by X-gal staining, only in the molecular layer of the neonatal cerebellum. In contrast, a 3.9LAMB1betagal transgene showed broad regional expression in the adult mouse brain, including the hippocampus, entorhinal cortex, colliculi, striatum, and substantia nigra. Similar expression patterns were observed for the endogenous laminin beta1 protein and for the 3.9LAMB1betagal transgene, analyzed with an antibody against the beta-galactosidase protein. The 3.9LAMB1betagal transgene expression in the hippocampal tri-synaptic circuit suggests a role for the LAMB1 gene in learning and memory.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Laminin/metabolism , Promoter Regions, Genetic , Transgenes/physiology , Animals , Animals, Newborn , Brain/anatomy & histology , Brain/growth & development , Brain/metabolism , Female , Galactosides/metabolism , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry/methods , In Situ Hybridization/methods , Indoles/metabolism , Laminin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/genetics , Rats
4.
Mech Dev ; 103(1-2): 13-25, 2001 May.
Article in English | MEDLINE | ID: mdl-11335108

ABSTRACT

The LAMB1 gene encodes the laminin beta1 subunit of laminin, an extracellular matrix protein. Using several transgenic mouse lines containing various lengths of the LAMB1 promoter driving lacZ reporter gene expression, regions of LAMB1 promoter that contain cis-acting DNA regulatory element(s) have been identified. The 3.9LAMB1betagal transgene is expressed in various tissues during development. LAMB1 transgene expression is observed in a selective set of nephrons of the neonatal and adult kidneys. The cis-acting DNA regulatory elements responsible for LAMB1 transgene expression in ovaries and in juvenile kidneys are present between -'1.4 and -0.7 kb relative to the transcription start site, while those of adult kidneys are located between -2.5 and -1.4 kb. The LAMB1 transgene is also expressed in the epididymis of 1 week old transgenic mice. Mutation of the retinoic acid response element (RARE) in the context of the 3.9LAMB1betagal transgene results in loss of LAMB1 transgene expression in all tissues. Thus, sequences between -2.5 and -0.7 kb plus the RARE are required for appropriate expression of the LAMB1 transgene in mice.


Subject(s)
Gene Expression Regulation , Lac Operon , Laminin/biosynthesis , Laminin/chemistry , Promoter Regions, Genetic , Tretinoin/metabolism , Tretinoin/physiology , Animals , Animals, Newborn , Base Sequence , Embryo, Mammalian/metabolism , Enhancer Elements, Genetic , Epididymis/metabolism , Female , In Situ Hybridization , Kidney/embryology , Kidney/metabolism , Laminin/genetics , Male , Mice , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Ovary/embryology , Ovary/metabolism , Plasmids/metabolism , Testis/embryology , Testis/metabolism , Time Factors , Transcription, Genetic , Transfection , Transgenes
5.
Cancer Res ; 61(6): 2774-81, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11289161

ABSTRACT

Clinical and preclinical studies suggest that retinoids can inhibit the growth of a small percentage of human renal cancers (RCs), although the majority of RCs both in vitro and in vivo are retinoid resistant. Our recent studies indicate that the metabolism of retinol to retinyl esters is greatly reduced in human carcinoma cell lines of the oral cavity, skin, and breast as compared with their normal epithelial counterparts, suggesting that human carcinoma cells are retinoid deficient relative to normal epithelial cells. We considered whether retinoid resistance in RCs was related to an abnormality in retinoid metabolism. The metabolism of [3H]retinol and of [3H]retinoic acid (RA) was examined in RC cell lines and normal human kidney (NK) epithelial cells cultured in media, in RA, or in RA plus IFN-alpha. The expression of LRAT (lecithin:retinol acyltransferase) was assessed by Northern and Western analysis. Retinol and retinyl ester levels were determined in tissue samples of normal human kidney and renal cell carcinoma. NK cells esterified all of the 50 nM [3H]retinol in which they were cultured. In contrast, six of the seven RC cell lines metabolized only trace amounts of [3H]retinol to [3H]retinyl esters. Consistent with this relative lack of [3H]retinol esterification by the tumor cells, the tumor cells exhibited LRAT transcripts of aberrantly low sizes relative to those in normal epithelial cells. Moreover, the NK cells expressed abundant levels of LRAT protein by Western analysis, whereas the RC cells did not express LRAT protein. When samples of human kidney tumor tissue were compared with samples of normal kidney tissue from patients who had undergone surgery for primary RC, the normal kidney tissues contained much higher levels of retinol and retinyl esters (approximately 0.5-2 microg/gram wet weight) than the tumor tissues in all seven patients examined. Culture of the RC lines in IFN-alpha plus all-trans-RA, a combination therapy used clinically, resulted in higher intracellular levels of [3H]retinol and [3H]retinyl esters. The metabolism of [3H]RA was also examined in these RC lines versus NK cells. Although the NK epithelial cells metabolized [3H]RA, the majority of the RC lines metabolized [3H]RA at a much slower rate. Most of the RC lines metabolized only 10-30% of the 50 nM [3H]RA over 6 h of culture. These data indicate that RCs both in vitro and in vivo are retinol and retinyl ester deficient relative to the normal human kidney, and they suggest that the aberrant differentiation of the neoplastic renal cells results in part from a defect in retinoid metabolism.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Tretinoin/analogs & derivatives , Vitamin A/metabolism , Acyltransferases/biosynthesis , Acyltransferases/genetics , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Renal Cell/pathology , Cell Division/drug effects , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/genetics , Esters/metabolism , Female , Gene Expression , Humans , Interferon alpha-2 , Interferon-alpha/pharmacology , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/metabolism , Male , Middle Aged , Recombinant Proteins , Retinoic Acid 4-Hydroxylase , Tretinoin/metabolism , Tretinoin/pharmacology , Tumor Cells, Cultured
6.
Carcinogenesis ; 21(11): 1925-33, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11062150

ABSTRACT

When exogenous [(3)H]retinol (vitamin A) was added to culture medium, normal human epithelial cells from the oral cavity, skin, lung and breast took up and esterified essentially all of the [(3)H]retinol within a few hours. As shown by [(3)H]retinol pulse-chase experiments, normal epithelial cells then slowly hydrolyzed the [(3)H]retinyl esters to [(3)H]retinol, some of which was then oxidized to [(3)H]retinoic acid (RA) over a period of several days. In contrast, cultured normal human fibroblasts and human umbilical vein endothelial cells (HUVEC) did not esterify significant amounts of [(3)H]retinol; this lack of [(3)H]retinol esterification was correlated with a lack of expression of lecithin:retinol acyltransferase (LRAT) transcripts in normal fibroblast and HUVEC strains. These results indicate that normal, differentiated cell types differ in their ability to esterify retinol. Human carcinoma cells (neoplastically transformed epithelial cells) of the oral cavity, skin and breast did not esterify much [(3)H]retinol and showed greatly reduced LRAT expression. Transcripts of the neutral, bile salt-independent retinyl ester hydrolase and the bile salt-dependent retinyl ester hydrolase were undetectable in all of the normal cell types, including the epithelial cells. These experiments suggest that retinoid-deficiency in the tumor cells could develop because of the lack of retinyl esters, a storage form of retinol.


Subject(s)
Acyltransferases/biosynthesis , Carcinoma/metabolism , Epithelial Cells/metabolism , Vitamin A/metabolism , Acyltransferases/genetics , Breast/cytology , Breast/enzymology , Breast/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/enzymology , Carcinoma/genetics , Cell Line , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Epithelial Cells/enzymology , Esters/metabolism , Fibroblasts/enzymology , Fibroblasts/metabolism , Gene Expression , Humans , Kinetics , Mouth/cytology , Mouth/enzymology , Mouth/metabolism , Skin/cytology , Skin/enzymology , Skin/metabolism , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tritium , Tumor Cells, Cultured , Vitamin A/pharmacokinetics
7.
Mol Cell Endocrinol ; 164(1-2): 225-37, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11026574

ABSTRACT

Estrogens regulate the proliferation, cytoarchitectural, and invasive properties of estrogen receptor (ER)-containing breast cancer cells. To identify genes under direct regulation by estrogen in breast cancer cells, we have used representational difference analysis (RDA) of cDNAs. In this way, we have identified (cyto)keratin 19 (K19), a major component of cell intermediate filaments, as being under rapid and direct regulation by estrogen in MCF-7 cells. Stimulation by estradiol (E2) of K19 mRNA is rapid, with maximal increase at 3 h, and is not blocked by cycloheximide, suggesting that it is a primary response to the hormone. Increased accumulation of K19 protein is observable by 8 h after E2 and levels continue to increase at 24-48 h after E2 treatment. Suppression of E2-induced K19 gene expression by the antiestrogen ICI 182,780 suggests that ER mediates this regulation. Analysis of the human K19 chromosomal gene, by transient transfection assays employing reporter gene constructs with the 5' and 3' flanking regions and portions of the body of the K19 gene, has resulted in identification of a complex enhancer region in the first intron. This enhancer region consists of a near-consensus estrogen response element (K19 ERE, which differs by only 1 bp from the consensus ERE) and two ERE half sites, as well as two AP1-like sites. The results of transfections with either the K19 gene promoter or the heterologous thymidine kinase promoter and constructs containing mutated or deleted portions of the enhancer region show that the K19 ERE is responsible for the E2-dependent transactivation of the keratin 19 gene and for the synergism that is observed between E2 and TPA with both ER alpha and ER beta. These studies document ER regulation of the K19 gene, localize the estrogen responsive region, and suggest that up-regulation of keratin 19 gene expression by estrogen may contribute to the cytoskeletal and nuclear matrix reorganization, and increased metastatic potential of ER-containing breast cancer cells upon exposure to estrogens.


Subject(s)
Breast Neoplasms/genetics , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Keratins/genetics , Base Sequence , Enhancer Elements, Genetic , Female , Humans , Molecular Sequence Data , Tumor Cells, Cultured
8.
Exp Cell Res ; 259(1): 274-83, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10942599

ABSTRACT

The homeobox gene Hoxa-1 is transcriptionally regulated by retinoic acid (RA) and encodes a transcription factor which has been shown to play important roles in cell differentiation and embryogenesis. In order to clone and characterize target genes of Hoxa-1, we utilized differential hybridization screening and cDNA subtractive hybridization methods to identify genes which are differentially expressed in F9-10, a murine F9 teratocarcinoma stem cell line which expresses high levels of exogenous Hoxa-1, compared to F9 wild-type stem cells, which do not express endogenous Hoxa-1 mRNA in the absence of RA. Twenty-eight candidate genes were identified; these genes encode very diverse proteins, including signaling molecules such as BMP-4, the enzyme superoxide dismutase, the cell adhesion molecule cadherin-6, proteins involved in gene transcription such as HMG-1 and SAP18, homeodomain-containing proteins Gbx-2 and Evx-2, and cell cycle regulatory proteins such as the retinoblastoma binding protein-2. Clone 104 encodes a novel protein; the expression of the clone 104 mRNA is also regulated in a fashion very similar to that of the exogenous Hoxa-1 gene in another F9 cell line, called F9-tet-Hoxa1-8, in which the exogenous Hoxa-1 mRNA expression is tightly regulated by a Tet-off gene expression system. These data strongly suggest that clone 104 is a direct downstream target of the transcription factor Hoxa-1. The cDNA sequence of clone 104 is related to that of human ubiquitin carboxyl-terminal hydrolase T. Further characterization of these putative Hoxa-1 target genes will aid in delineating the functions of the Hoxa-1 protein in the differentiation processes which occur during embryogenesis.


Subject(s)
Cloning, Molecular/methods , Proteins , Trans-Activators/genetics , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Cadherins/genetics , Carrier Proteins/genetics , Cell Differentiation/genetics , Co-Repressor Proteins , Gene Expression/physiology , Genetic Testing/methods , HMGB1 Protein , High Mobility Group Proteins/genetics , Histone Deacetylases , Homeodomain Proteins , Humans , Mice , Phosphoproteins/genetics , RNA, Messenger/analysis , RNA-Binding Proteins , Receptors, Retinoic Acid/genetics , Retinoblastoma-Like Protein p130 , Stem Cells/cytology , Superoxide Dismutase/genetics , Teratocarcinoma , Transcription Factors/genetics , Transcription, Genetic/physiology , Tumor Cells, Cultured , Ubiquitins/genetics
9.
Cell Growth Differ ; 11(2): 71-82, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10714763

ABSTRACT

Retinoic acid (RA) induces the differentiation of many murine teratocarcinoma cell lines such as F9 and P19. In F9 cells, the level of the cellular retinoic acid binding protein I (CRABP I) mRNA is greatly reduced after exposure of the cultured cells to exogenous RA. In P19 cells, the level of CRABP I mRNA is greatly increased after RA exposure. We have identified a 176-bp region in the murine CRABP I promoter, between -2.9 and -2.7 kb 5' of the start site of transcription, which acts as an enhancer in undifferentiated F9 stem cells and through which RA effects inhibition of CRABP I transcription. Within this region are two footprinted sites at -2763 and -2834. This 176-bp regulatory region does not function to enhance CRABP I transcription in P19 stem cells. Several DNA sequences within these two footprinted regions bind proteins from F9 nuclear extracts but not from P19 nuclear extracts (e.g., FP1B, FP1A, and FP2B), as assessed by gel shift assays. This 176-bp CRABP I genomic region has not been sequenced previously and functionally analyzed in cultured cells because it was not present in the murine CRABP I clones used for the promoter analyses reported earlier by another laboratory. The function of this enhancer may be to reduce the expression of the CRABP I gene in specific embryonic cell types in order to regulate the amount of RA to which the cells are exposed.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Receptors, Retinoic Acid/genetics , Teratocarcinoma/genetics , Transcription, Genetic , Tretinoin/pharmacology , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Nucleus/chemistry , DNA Footprinting , Enhancer Elements, Genetic , Fibroblasts/cytology , Mice , Molecular Sequence Data , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptors, Retinoic Acid/biosynthesis , Teratocarcinoma/pathology , Transfection , Tumor Cells, Cultured
10.
Cell Growth Differ ; 11(1): 11-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10672899

ABSTRACT

Using a PCR-based cDNA subtractive hybridization method (L. Diatchenko et al., Proc. Natl. Acad. Sci. USA, 93: 6025-6030, 1996), we cloned a cDNA fragment of a novel gene that is highly expressed in F9-10; F9-10 is an F9 teratocarcinoma stem cell line that expresses high levels of exogenous Hoxa-1 mRNA and protein in comparison to F9 wild-type stem cells, which do not express endogenous Hoxa-1 mRNA in the absence of retinoic acid (RA). Rapid amplification of cDNA ends was used to clone the full-length cDNA of this gene, designated HA1R-62 (Hoxa1 regulated-62). We have shown that HA1R-62 is also a RA-responsive gene and that it is expressed (mRNA size, approximately 4.3 kb) in adult mouse thymus, lung, kidney, and ovary as well as in 12.5-day mouse embryos. DNA sequence analysis and in vitro translation experiments have shown that HA1R-62 encodes a protein with a molecular mass of approximately 26 kDa. Elucidation of the function of the HA1R-62 gene product will provide new insights into the functions of RA and homeobox genes.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/genetics , Proteins/genetics , Repressor Proteins , Tretinoin/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Genes, Homeobox/genetics , Mice , Molecular Sequence Data , Neoplasm Proteins/chemistry , Protein Biosynthesis , Proteins/chemistry , RNA, Messenger/metabolism , Reticulocytes/metabolism , Teratocarcinoma , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , Up-Regulation
11.
Proc Natl Acad Sci U S A ; 96(23): 13524-9, 1999 Nov 09.
Article in English | MEDLINE | ID: mdl-10557354

ABSTRACT

Retinoids, vitamin A (retinol) and its metabolic derivatives, are required for normal vertebrate development. In murine embryonic stem (ES) cells, which remain undifferentiated when cultured in the presence of LIF (leukemia inhibitory factor), little metabolism of exogenously added retinol takes place. After LIF removal, ES cells metabolize exogenously added retinol to 4-hydroxyretinol and 4-oxoretinol and concomitantly differentiate. The conversion of retinol to 4-oxoretinol is a high-capacity reaction because most of the exogenous retinol is metabolized rapidly, even when cells are exposed to physiological ( approximately 1 microM) concentrations of retinol in the medium. No retinoic acid or 4-oxoRA synthesis from retinol was detected in ES cells cultured with or without LIF. The cytochrome P450 enzyme CYP26 (retinoic acid hydroxylase) is responsible for the metabolism of retinol to 4-oxoretinol, and CYP26 mRNA is greatly induced (>15-fold) after LIF removal. Concomitant with the expression of CYP26, differentiating ES cells grown in the absence of LIF activate the expression of the differentiation marker gene FGF-5 whereas the expression of the stem cell marker gene FGF-4 decreases. The strong correlation between the production of polar metabolites of retinol and the differentiation of ES cells upon removal of LIF suggests that one important action of LIF in these cells is to prevent retinol metabolism to biologically active, polar metabolites such as 4-oxoretinol.


Subject(s)
Embryo, Mammalian/cytology , Embryo, Nonmammalian , Growth Inhibitors/metabolism , Interleukin-6 , Lymphokines/metabolism , Stem Cells/metabolism , Vitamin A/analogs & derivatives , Vitamin A/metabolism , Animals , COS Cells , Cell Differentiation , Cloning, Molecular , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Kinetics , Leukemia Inhibitory Factor , Retinoic Acid 4-Hydroxylase , Transfection , Vitamin A/biosynthesis
12.
J Biol Chem ; 274(38): 26783-8, 1999 Sep 17.
Article in English | MEDLINE | ID: mdl-10480883

ABSTRACT

F9 teratocarcinoma cell lines, carrying one or two disrupted alleles of the RARbeta(2) gene, were generated by homologous recombination to study the role of RARbeta(2) in mediating the effects of retinoids on cell growth and differentiation. Retinoic acid (RA) does not induce growth arrest of the RARbeta(2)-/- cells, whereas the F9 WT and RARbeta(2)+/- heterozygote lines undergo RA-induced growth arrest. The RARbeta(2)+/- lines also exhibit a faster cell cycle transit time in the absence of RA. The RARbeta(2)-/- stem cells exhibit an altered morphology when compared with the F9 WT parent line, and after RA treatment, the RARbeta(2)-/- cells do not exhibit a fully differentiated cell morphology. As compared with F9 WT cells, the RARbeta-/- cells exhibited a markedly lower induction of several early RA-responsive genes and no induction of laminin B1, a late response gene. The induction of RA metabolism in the F9 RARbeta(2)-/- cells following differentiation was not impaired. The research presented here, and prior research suggest that RARbeta is required for RA-induced growth arrest in a variety of cell types and that RARbeta also functions in mediating late responses to RA. These findings are significant in view of the reduced expression of RARbeta transcripts in a number of different types of human carcinomas.


Subject(s)
Alleles , Receptors, Retinoic Acid/genetics , Tretinoin/physiology , Cell Differentiation , Cell Division , Electrophoresis, Polyacrylamide Gel , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Tumor Cells, Cultured
13.
Nucleosides Nucleotides ; 18(3): 363-76, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10358941

ABSTRACT

A synthesis of the C-nucleoside, 2-amino-7-(2-deoxy-beta-D-erythro- pentofuranosyl)-3H,5H-pyrrolo[3,2-d]pyrimidin-4-one (9-deaza-2'-deoxyguanosine) was achieved starting from 2-amino-6-methyl-3H-pyrimidin-4-one (5) and methyl 2-deoxy-3,5-di-O-(p-nitrobenzoyl)-D-erythro-pento-furanoside (11). The anomeric configuration of the C-nucleoside was established by 1H NMR, NOEDS and ROESY. This C-nucleoside did not inhibit the growth of T-cell lymphoma cells.


Subject(s)
Antimetabolites, Antineoplastic/chemical synthesis , Deoxyguanosine/analogs & derivatives , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/toxicity , Cell Survival/drug effects , Deoxyguanosine/chemical synthesis , Deoxyguanosine/chemistry , Deoxyguanosine/toxicity , Drug Screening Assays, Antitumor , Indicators and Reagents , Leukemia L1210 , Lymphoma, T-Cell , Magnetic Resonance Spectroscopy , Mice , Molecular Conformation , Molecular Structure , Tumor Cells, Cultured
14.
Cell Growth Differ ; 9(12): 969-81, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9869297

ABSTRACT

Homeobox genes play key roles in specifying body part identity during vertebrate embryonic development. Retinoids are signaling molecules involved in the regulation of expression of homeobox genes. We have previously identified an retinoic acid (RA)-inducible enhancer (RAIDR5) located approximately 6.5 kb 3' of the coding region of the murine Hoxb1 gene. This 3' enhancer contains three sequences that are highly conserved in similar RA-inducible enhancers identified in the murine and human Hoxa1 genes and in the chicken Hoxb1 gene. One element, a DR5 RA response element, contributes to the RA inducibility of a Hoxb1 reporter gene construct in F9 cells. In this report, further analysis of the other two elements of the Hoxb1 3' enhancer is reported. The two other sequences, conserved element (CE) 1 and CE2, act as negative elements in cultured F9 cells; when either is mutated, an increase in the beta-galactosidase activity of a Hoxb1 reporter gene construct results. A single Hoxb1 CE2 DNA element:protein binding complex was detected in F9 stem cells, and experiments suggest that this is the same binding protein that recognizes the CE2 element of Hoxa1. In a variant F9 cell line in which both allelic copies of the RA receptor gamma (RARgamma) gene are disrupted, the CE2 binding complex is absent, and this absence correlates with the inability of the CE2 element to function as a repressor of Hoxb1 reporter gene expression in these cells. A single Hoxb1 CE1 binding complex is also detected by gel shift assays in nuclear extracts prepared from both stem and RA-treated F9 cells. This complex contains an Mr approximately 200,000 protein as shown by UV cross-linking. Although the sequences of the CE1 elements of Hoxb1 and Hoxa1 are highly conserved, they differ by two nucleotides. Gel shift analysis shows that either of these nucleotide changes prevents binding of F9 cell protein extracts. When gel shift assays were performed using nuclear extracts prepared from mouse embryos at a time when Hoxb1 mRNA is expressed, i.e., day 9.0, CE1 and CE2 binding complexes identical in mobility to those detected in F9 cells were observed. This suggests roles for both the CE1 and CE2 elements in regulating Hoxb1 gene expression during development.


Subject(s)
Enhancer Elements, Genetic/genetics , Homeodomain Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Base Sequence , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genes, Homeobox/genetics , Genes, Reporter/genetics , Mice , Molecular Sequence Data , Mutation/genetics , Nuclear Proteins/genetics , Oligodeoxyribonucleotides/genetics , RNA, Messenger/genetics , Receptors, Retinoic Acid/genetics , Sequence Alignment , Transfection/genetics , Tretinoin/pharmacology , Tumor Cells, Cultured , Ultraviolet Rays , Retinoic Acid Receptor gamma
15.
Mol Cell Endocrinol ; 143(1-2): 155-66, 1998 Aug 25.
Article in English | MEDLINE | ID: mdl-9806360

ABSTRACT

In the presence of retinoic acid (RA), F9 murine teratocarcinoma cells differentiate into cells resembling the extra-embryonic endoderm of the early mouse embryo. Using differential hybridization, we have cloned and characterized six cDNAs corresponding to mRNAs that exhibit reduced expression in F9 cells following RA treatment. Two of these cDNAs encode novel genes (REX-2 and REX-3). The other isolated cDNAs encode genes that have been previously described in other contexts: 1-4 (cyclin D3); 2-10 (pyruvate kinase); 2-12 (glutathione S-transferase); and 2-17 (GLUT 3). The mRNA levels of these genes are reduced by RA or RA plus theophylline and cAMP (RACT) only after 48 h of treatment, and continue to decrease at 96 h. The half-lives of these mRNAs are not changed by RA treatment, indicating that these mRNAs may be regulated through a transcriptional mechanism. In isoleucine-deprived cells, which are growth arrested but do not differentiate, the steady state mRNA levels of genes Rex 2, Rex 3, pyruvate kinase and GLUT 3 are not reduced, in contrast to cyclin D3 and glutathione S-transferase. The expression of the REX-2, REX-3, pyruvate kinase, glutathione S-transferase and GLUT 3 genes is reduced by RACT to the same extent in F9 RARgamma-/- and RARalpha-/- lines as in F9-Wt. In contrast, cyclin D3 exhibits lower mRNA expression in F9 RARgamma-/- and RARalpha-/- stem cells, and this mRNA is not decreased by RACT treatment. Overexpression of cyclin D3 blocks the RA-induced growth arrest of F9 cells, indicating that the downregulation of this gene following RA treatment may constitute a necessary step in the cascade of events leading to growth inhibition by RA.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclins/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Teratocarcinoma/genetics , Tretinoin/pharmacology , Amino Acid Sequence , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/drug effects , Cell Division/genetics , Cyclin D3 , DNA, Complementary/analysis , DNA, Complementary/genetics , Genes, Tumor Suppressor , Mice , Molecular Sequence Data , Sequence Alignment , Tumor Cells, Cultured
16.
Development ; 125(16): 3235-46, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9671595

ABSTRACT

The murine Hoxb-1 gene contains a homeobox sequence and is expressed in a spatiotemporal specific pattern in neuroectoderm, mesoderm and gut endoderm during development. We previously identified a conserved retinoic acid (RA)-inducible enhancer, named the RAIDR5, which contains a DR5 RARE; this RAIDR5 enhancer is located 3' of the Hoxb-1-coding region in both the mouse and chick. In the F9 murine teratocarcinoma cell line, this DR5 RARE is required for the RA response of the Hoxb-1 gene, suggesting a functional role of the DR5 RARE in Hoxb-1 gene expression during embryogenesis. From the analysis of Hoxb-1/lacZ reporter genes in transgenic mice, we have shown that a wild-type (WT) transgene with 15 kb of Hoxb-1 genomic DNA, including this Hoxb-1 3' RAIDR5, is expressed in the same tissues and at the same times as the endogenous Hoxb-1 gene. However, a transgene construct with point mutations in the DR5 RARE (DR5mu) was not expressed in the developing foregut, which gives rise to organs such as the esophagus, lung, stomach, liver and pancreas. Like the wild-type transgene, this DR5 RARE mutated transgene was expressed in rhombomere 4 in 9.5 day postcoitum (d.p.c.) embryos. Similarly, transgene staining in the foregut of animals carrying a deletion of the entire Hox-b1 RAIDR5 enhancer (3'-del) was greatly reduced relative to that seen with the WT transgene. We also demonstrated that expression of the WT transgene in the gut increases in response to exogenous RA, resulting in anterior expansion of the expression in the gut. These observations that the Hoxb-1 gene is expressed in the developing gut and that this expression is regulated through a DR5 RARE strongly suggest a role for Hoxb-1 in the anteroposterior axis patterning of the gut and a critical role for endogenous retinoids in early gut development.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/physiology , Intestines/growth & development , Tretinoin/physiology , Animals , Enhancer Elements, Genetic/genetics , Genes, Homeobox/genetics , Genes, Reporter/genetics , Histocytochemistry , Homeodomain Proteins/genetics , In Situ Hybridization , Lac Operon/genetics , Mice , Point Mutation/genetics , Sequence Deletion/genetics , Transfection/genetics , Tumor Cells, Cultured
17.
Cancer Res ; 58(9): 2007-13, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9581846

ABSTRACT

All-trans-retinoic acid (RA) is used as a differentiation therapy for acute promyelocytic leukemia. Patients can become resistant to RA, and this resistance is thought to be mediated in part by an increase in the rate of RA metabolism. We have characterized the metabolism of all-trans-retinol (ROL; vitamin A) in NB4 cells, which are human promyelocytic leukemia cells. NB4 cells metabolize ROL into a variety of compounds, including all-trans-4-hydroxyretinol, all-trans-4-oxoretinol (4-oxoROL), 14-hydroxy-4,14-retro-retinol, anhydroretinol, and several ROL esters. No metabolism of ROL to RA or to RA derivatives in NB4 cells was detected. The rate of ROL metabolism increased after cell differentiation; in a 24-h period, differentiated cells metabolized 2-fold more ROL than did undifferentiated cells. The major difference in the ROL metabolism pattern between undifferentiated and differentiated cells was an approximately 10-fold increase in the production of all-trans-4-hydroxyretinol and 4-oxoROL in differentiated cells. Furthermore, exogenously added 4-oxoROL was capable of eliciting NB4 cell differentiation, as measured by growth inhibition, nitroblue tetrazolium reduction, nuclear body relocalization of PML, and surface expression of CD11b. In addition, 4-oxoROL synergized with IFN-gamma in the promotion of NB4 cell growth arrest. Following treatment of NB4 cells with 4-oxoROL to induce differentiation, the production of 4-oxoROL from ROL was observed; this indicated that 4-oxoROL induces its own synthesis in NB4 cells. In addition, 48 h after the administration of 1 microM 4-oxoROL, NB4 cells maintained a high intracellular concentration (17 microM) of 4-oxoROL. These unique properties of 4-oxoROL may provide advantages over RA in the treatment of promyelocytic leukemia cells because it may be possible to maintain cytodifferentiating concentrations of 4-oxoROL in the cells for extended periods of time.


Subject(s)
Granulocytes/drug effects , Leukemia, Promyelocytic, Acute/pathology , Vitamin A/analogs & derivatives , Vitamin A/pharmacology , Cell Differentiation/drug effects , Cell Division/drug effects , Chromatography, High Pressure Liquid , Drug Synergism , Granulocytes/cytology , Humans , Interferon-gamma/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Vitamin A/metabolism
18.
Mol Cell Biol ; 18(4): 1866-78, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9528758

ABSTRACT

The Rex-1 (Zfp-42) gene, which encodes an acidic zinc finger protein, is expressed at high levels in embryonic stem (ES) and F9 teratocarcinoma cells. Prior analysis identified an octamer motif in the Rex-1 promoter which is required for promoter activity in undifferentiated F9 cells and is involved in retinoic acid (RA)-associated reduction in expression. We show here that the Oct-3/4 transcription factor, but not Oct-1, can either activate or repress the Rex-1 promoter, depending on the cellular environment. Rex-1 repression is enhanced by E1A. The protein domain required for Oct-3/4 activation was mapped to amino acids 1 to 35, whereas the domain required for Oct-3/4 repression was mapped to amino acids 61 to 126, suggesting that the molecular mechanisms underlying transcriptional activation and repression differ. Like Oct-3/4, Oct-6 can also lower the expression of the Rex-1 promoter via the octamer site, and the amino-terminal portion of Oct-6 mediates this repression. In addition to the octamer motif, a novel positive regulatory element, located immediately 5' of the octamer motif, was identified in the Rex-1 promoter. Mutations in this element greatly reduce Rex-1 promoter activity in F9 cells. High levels of a binding protein(s), designated Rox-1, recognize this novel DNA element in F9 cells, and this binding activity is reduced following RA treatment. Taken together, these results indicate that the Rex-1 promoter is regulated by specific octamer family members in early embryonic cells and that a novel element also contributes to Rex-1 expression.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , COS Cells , Octamer Transcription Factor-3 , Octamer Transcription Factor-6 , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins , Stem Cells/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Zinc Fingers
19.
Cancer Res ; 58(1): 166-76, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9426073

ABSTRACT

Retinoids, metabolites and synthetic derivatives of vitamin A (retinol), have been shown to inhibit carcinogenesis in various epithelial tissues in animal model systems and to have clinical efficacy as chemotherapeutic agents against certain types of cancer, including squamous cell carcinomas (SCCs). We examined the metabolism of [3H]retinol in normal human cell strains and SCC lines from the oral cavity and skin, and we report here that the cultured normal human epithelial cell strains esterified [3H]retinol to a much greater extent than the SCC lines. Furthermore, microsomal extracts of normal cell strains (e.g., OKF4) exhibited about 7-fold more palmityl-CoA-dependent, phenylmethylsulfonyl fluoride-resistant retinol esterification activity than extracts from SCC lines (e.g., SCC25). The fact that the esteriflcation of retinol was phenylmethylsulfonyl fluoride resistant suggests that the enzyme acyl-CoA:retinol acyltransferase is involved. Culture of both the normal and SCC lines in the presence of 1 microM all-trans-retinoic acid (RA) for 48 h enhanced the formation of [3H]retinyl esters from [3H]retinol. All of the cell lines examined can also metabolize [3H]retinol to [3H]RA, [3H]14-hydroxy-4,14-retroretinol, [3H]retinaldehyde, and [3H]3,4-didehydroretinol, but this metabolism occurs to varying extents in different cell lines. Culture of the cells in the presence of RA for 48 h did not affect the subsequent metabolism of [3H]retinol to [3H]RA and [3H]14-hydroxy-4,14-retroretinol, but it did reduce the metabolism of [3H]retinol to [3H]3,4-didehydroretinol. When cultured for 6-10 h in the presence of nanomolar concentrations of exogenous [3H]retinol, both the normal and SCC lines had much higher intracellular [3H]retinol concentrations, in the micromolar range. No correlation was seen between CRABP II or CRBP I mRNA levels and the levels of either intracellular [3H]retinol or [3H]retinol metabolism in these lines. The reduced ability to esterify retinol in these tumor cells may result in inappropriate cell growth and the loss of normal differentiation responses because of the lack of a sufficient amount of internal retinol stored as retinyl esters.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Mouth Neoplasms/metabolism , Skin Neoplasms/metabolism , Tretinoin/metabolism , Humans , Tumor Cells, Cultured
20.
Dev Dyn ; 211(1): 97-108, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9438427

ABSTRACT

The murine Hoxa1 gene is a member of the vertebrate Hox complex and plays a role in defining the body plan during development. At day 8.0-9.0 post coitus, Hoxa1 transcripts are detected extensively throughout the embryo in the neural tube, adjacent mesenchyme, paraxial mesoderm, somites and gut epithelium; expression extends from the most caudal region of the embryo to the rhombomere 3/4 border. This spatiotemporal expression of Hoxa1 mRNA is critical for normal embryonic development. We have previously identified a 10 bp element, called CE2, which is located approximately 3 kilobases 3' of the Hoxa1 coding region in the RAIDR5 enhancer, and which binds to an approximately 170 kd protein in retinoic acid treated P19 embryonal carcinoma cells. CE2 elements were also identified 3' of the murine Hoxb1 gene, the chicken Hoxb1 gene and the human Hoxa1 gene. To examine the role of this CE2 element in regulating Hoxa1 expression in vivo, transgenic mice were generated which express a Hoxa1 beta-galactosidase reporter gene that contains a mutation in the CE2 element. Relative to transgenic mice bearing a wild type CE2 element, the mutant CE2 construct recapitulated rhombomeric, neural, and gut epithelium expression but failed to show beta-galactosidase expression in somites and adjacent mesenchymal tissue. Gel shift analysis showed that binding activity similar to that detected in extracts prepared from retinoic acid treated P19 cells was present in nuclear extracts prepared from day 9.0 embryos. However, an additional binding complex not detected in P19 cells was also observed. These results indicate that in transgenic animals, the evolutionary conserved CE2 element is a somite and adjacent mesenchymal enhancer of Hoxa1 expression.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Mesoderm/chemistry , Somites/chemistry , Transcription Factors/genetics , Animals , Base Sequence , Cell Extracts , Cell Nucleus , Conserved Sequence/genetics , Embryonic and Fetal Development , Gene Expression Regulation, Developmental/drug effects , Genes, Homeobox/genetics , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Organ Specificity , RNA, Messenger/analysis , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...