Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
J Mol Recognit ; 24(3): 429-35, 2011.
Article in English | MEDLINE | ID: mdl-21504020

ABSTRACT

In this paper we describe a new surface plasmon resonance (SPR) biosensor dedicated to potential estrogenic compounds prescreening, by developing an estrogen receptor (ER) specific DNA chip. Through the covalent binding of a DNA strain wearing the estrogen response element (ERE) to an activated 6-mercapto-1-hexadecanoic acid and 11-mercapto-1-undecanol self-assembled monolayer on gold surface, the SPR biosensor allows to detect specifically, quickly, and without any labeling the binding of ER in the presence of estrogen. In parallel, we investigated the ER interaction with itself, in order to study the formation of ER dimer apparently needed to activate the gene expression through ERE interaction. For that, we engaged force spectroscopy experiments that allowed us to prove that ER needs estrogen for its dimerization. Moreover, these ER/ER intermolecular measurements enabled to propose an innovative screening tool for anti-estrogenic compounds, molecules of interest for hormono-dependent cancer therapy.


Subject(s)
DNA/metabolism , Microscopy, Atomic Force/methods , Proteins/metabolism , Receptors, Estrogen/metabolism , Biosensing Techniques , Protein Binding , Surface Plasmon Resonance
2.
Br J Cancer ; 102(6): 1024-31, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20197771

ABSTRACT

BACKGROUND: This study evaluates the relation of the early oestrogen-regulated gene gabarapl1 to cellular growth and its prognostic significance in breast adenocarcinoma. METHODS: First, the relation between GABARAPL1 expression and MCF-7 growth rate was analysed. Thereafter, by performing macroarray and reverse transcriptase quantitative-polymerase chain reaction (RT-qPCR) experiments, gabarapl1 expression was quantified in several histological breast tumour types and in a retrospective cohort of 265 breast cancers. RESULTS: GABARAPL1 overexpression inhibited MCF-7 growth rate and gabarapl1 expression was downregulated in breast tumours. Gabarapl1 mRNA levels were found to be significantly lower in tumours presenting a high histological grade, with a lymph node-positive (pN+) and oestrogen and/or progesterone receptor-negative status. In univariate analysis, high gabarapl1 levels were associated with a lower risk of metastasis in all patients (hazard ratio (HR) 4.96), as well as in pN+ patients (HR 14.96). In multivariate analysis, gabarapl1 expression remained significant in all patients (HR 3.63), as well as in pN+ patients (HR 5.65). In univariate or multivariate analysis, gabarapl1 expression did not disclose any difference in metastasis risk in lymph node-negative patients. CONCLUSIONS: Our data show for the first time that the level of gabarapl1 mRNA expression in breast tumours is a good indicator of the risk of recurrence, specifically in pN+ patients.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Ductal, Breast/pathology , Microtubule-Associated Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Microtubule-Associated Proteins/metabolism , Middle Aged , Outcome Assessment, Health Care , Prognosis , Recurrence , Retrospective Studies , Tumor Cells, Cultured , Up-Regulation
3.
Biosens Bioelectron ; 21(9): 1830-7, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16406567

ABSTRACT

Many biotechnology applications use proteins immobilized on surface. For biosensor, the sensing layer is a key component interfacing the transducer and the sample. Strategies employed to activate the bidimensional surface act directly on the performance of the biosensor. In this paper we propose a novel strategy for engineered proteins self-assembly. Our original supramolecular structure allows a direct and fast covalent attachment of proteins onto bare gold substrate through a homobifunctional cross-linker, 1,4-di-([2'-pyridyldithio]propionamido)butane (DPDPB). In this work, engineered proteins and linker-protein complexes were synthesized and characterized by gel electrophoresis, chromatography and spectroscopy experiments. Macromolecular construction "DPDPB-GST tag-GEC1 protein" was conceived in order to guarantee a 2D architecture enhancing the capabilities of the target (tubulin) to recognize its partner (GEC1). Surface plasmon resonance measurements clearly showed potential of this particular self-assembled protein layer compared to a commercial immunosensor interface. At the concentrations tested, the recognition process occurs between tubulin and the immobilized GEC1; moreover enhanced binding was obtained with the home-made 2D sensing layer more than with 3D carboxymethyl dextran matrix.


Subject(s)
Biosensing Techniques/methods , Coated Materials, Biocompatible/chemistry , Crystallization/methods , Gold/chemistry , Immunoassay/methods , Protein Interaction Mapping/methods , Surface Plasmon Resonance/methods , Biosensing Techniques/instrumentation , Feasibility Studies , Macromolecular Substances/chemistry
4.
Clin Biochem ; 36(8): 621-8, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14636877

ABSTRACT

OBJECTIVES: To develop a sensitive telomeric repeat amplification protocol (TRAP)-silver staining assay for telomerase activity quantification. DESIGN AND METHODS: TRAP assays were performed by using a TRAPeze telomerase kit with or without [alpha-32P]-dCTP. Amplification products were electrophoresed in polyacrylamide gels and detected by autoradiography or a modified silver staining protocol. Telomerase activity was quantified from radioactive counts or optical density of telomerase products from test extracts and controls. RESULTS: TRAP-silver staining assay was at least as sensitive as radioactive TRAP assay and quantified telomerase activity within linearity from 10 to 3,000 cell equivalents. Both methods quantified a weak telomerase activity in normal endometrial glandular epithelial cells (GEC) and a strong increase in immortalized GEC. In human pathologic endometria (n=24), telomerase activity was correlated with lesion seriousness and distinguished simple hyperplasias from nonhyperplasic or cancerous lesions. CONCLUSIONS: TRAP-silver staining assay is suitable for cell and tissue telomerase activity routine quantification.


Subject(s)
Endometrium/chemistry , Endometrium/enzymology , Silver Staining/methods , Telomerase/analysis , Telomerase/metabolism , Adult , Aged , Aged, 80 and over , Animals , Endometrium/pathology , Female , Guinea Pigs , HeLa Cells , Humans , Middle Aged
5.
Recent Prog Horm Res ; 55: 163-93; discussion 194-5, 2000.
Article in English | MEDLINE | ID: mdl-11036937

ABSTRACT

The action of nuclear hormone receptors is tripartite, involving the receptor, its ligands, and its co-regulator proteins. The estrogen receptor (ER), a member of this superfamily, is a hormone-regulated transcription factor that mediates the effects of estrogens and anti-estrogens (e.g., tamoxifen) in breast cancer and other estrogen target cells. This chapter presents our recent work on several aspects of estrogen action and the function of the ER: 1) elucidation of ER structure-function relationships and development of ligands that are selective for one of the two ER subtypes, ERalpha or ERbeta; 2) identification of ER-selective co-regulators that potentiate the inhibitory effectiveness of anti-estrogens and dominant-negative ERs and modulate the activity of estrogens; 3) characterization of genes that are regulated by the anti-estrogen-ER versus the estrogen-ER complex; and 4) elucidation of the intriguing pharmacology of these ER complexes at different gene regulatory sites. These findings indicate that different residues of the ER hormone-binding domain are involved in the recognition of structurally distinct estrogens and anti-estrogens and highlight the exquisite precision of the regulation of ER activities by ligands, with small changes in ligand structure resulting in major changes in receptor character. Studies also explore the biology and distinct pharmacology mediated by ERalpha and ERbeta complexed with different ligands through different target genes. The upregulation of the anti-oxidant detoxifying phase II enzyme, quinone reductase, by the anti-estrogen-occupied ER, mediated via the electrophile response element in the QR gene, may contribute to the beneficial antioxidant effects of anti-estrogens in breast cancer and illustrates the activation of some genes by ER via non-estrogen response element sequences. The intriguing biology of estrogen in its diverse target cells is thus determined by the structure of the ligand, the ER subtype involved, the nature of the hormone-responsive gene promoter, and the character and balance of co-activators and co-repressors that modulate the cellular response to the ER-ligand complex. The continuing development of novel ligands and the study of how they function as selective agonists or antagonists through ERalpha or ERbeta should allow optimized tissue selectivity of these agents for hormone replacement therapy and treatment and prevention of breast cancer.


Subject(s)
Receptors, Estrogen/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Estrogens/chemistry , Estrogens/metabolism , Estrogens/pharmacology , Female , Humans , Ligands , Models, Biological , Models, Molecular , Molecular Sequence Data , Prohibitins , Receptors, Estrogen/chemistry , Receptors, Estrogen/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
6.
J Biol Chem ; 275(46): 35848-56, 2000 Nov 17.
Article in English | MEDLINE | ID: mdl-10960470

ABSTRACT

The transcriptional activity of nuclear hormone receptors is known to be modulated by coregulator proteins. We found that the repressor of estrogen receptor activity (REA), a protein recruited to the hormone-occupied estrogen receptor (ER), decreased the transcriptional activity of ER, both when ER was acting directly through DNA response elements as well as when it was tethered to other transcription factors. Administration of antisense REA resulted in a 2-4-fold increase in ER transactivation, implying that endogenous REA normally dampens the stimulatory response to estradiol. To define the interaction regions between ER and REA, we used glutathione S-transferase pull-down assays. We found that REA bound to the ligand-binding domain (E domain) of ER, but not to other regions of ER, and that REA interaction with ER involved a region in the C-terminal half of REA. REA and the coactivator SRC-1 were involved in a functional competition for regulation of ER transcriptional activity, which we show results from competition between these two coregulators for interaction with ER. REA contains an LXXLL motif near its N terminus, but this motif was not involved in its binding to ER. Rather, this sequence was required for the competitive binding of REA and SRC-1 to ER and thus for optimal repression of ER activity. Our findings show that the regions of REA required for its interaction with ER and for its repression of ER activity are different.


Subject(s)
Gene Expression Regulation , Receptors, Estrogen/metabolism , Repressor Proteins/metabolism , Amino Acid Motifs , Animals , Binding, Competitive , Consensus Sequence/genetics , Estradiol/metabolism , Genes, Reporter , Histone Acetyltransferases , Humans , Mutation , Nuclear Receptor Coactivator 1 , Oligonucleotides, Antisense/genetics , Prohibitins , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, Estrogen/genetics , Recombinant Fusion Proteins , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/chemistry , Repressor Proteins/genetics , Response Elements/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Tumor Cells, Cultured
7.
Mol Cell Biol ; 20(17): 6224-32, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10938099

ABSTRACT

We find that prothymosin alpha (PTalpha) selectively enhances transcriptional activation by the estrogen receptor (ER) but not transcriptional activity of other nuclear hormone receptors. This selectivity for ER is explained by PTalpha interaction not with ER, but with a 37-kDa protein denoted REA, for repressor of estrogen receptor activity, a protein that we have previously shown binds to ER, blocking coactivator binding to ER. We isolated PTalpha, known to be a chromatin-remodeling protein associated with cell proliferation, using REA as bait in a yeast two-hybrid screen with a cDNA library from MCF-7 human breast cancer cells. PTalpha increases the magnitude of ERalpha transcriptional activity three- to fourfold. It shows lesser enhancement of ERbeta transcriptional activity and has no influence on the transcriptional activity of other nuclear hormone receptors (progesterone receptor, glucocorticoid receptor, thyroid hormone receptor, or retinoic acid receptor) or on the basal activity of ERs. In contrast, the steroid receptor coactivator SRC-1 increases transcriptional activity of all of these receptors. Cotransfection of PTalpha or SRC-1 with increasing amounts of REA, as well as competitive glutathione S-transferase pulldown and mammalian two-hybrid studies, show that REA competes with PTalpha (or SRC-1) for regulation of ER transcriptional activity and suppresses the ER stimulation by PTalpha or SRC-1, indicating that REA can function as an anticoactivator in cells. Our data support a model in which PTalpha, which does not interact with ER, selectively enhances the transcriptional activity of the ER but not that of other nuclear receptors by recruiting the repressive REA protein away from ER, thereby allowing effective coactivation of ER with SRC-1 or other coregulators. The ability of PTalpha to directly interact in vitro and in vivo with REA, a selective coregulator of the ER, thereby enabling the interaction of ER with coactivators, appears to explain its ability to selectively enhance ER transcriptional activity. These findings highlight a new role for PTalpha as a coregulator activity-modulating protein that confers receptor specificity. Proteins such as PTalpha represent an additional regulatory component that defines a novel paradigm enabling receptor-selective enhancement of transcriptional activity by coactivators.


Subject(s)
Protein Precursors/metabolism , Protein Precursors/physiology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Thymosin/analogs & derivatives , Transcription, Genetic , Animals , Blotting, Northern , CHO Cells , Cricetinae , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Gene Library , Glutathione Transferase/metabolism , Histone Acetyltransferases , Humans , Intracellular Signaling Peptides and Proteins , Models, Biological , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivators , Plasmids , Precipitin Tests , Prohibitins , Protein Binding , Protein Biosynthesis , Protein Precursors/pharmacology , Proteins/metabolism , Receptors, Estrogen/genetics , Thymosin/metabolism , Thymosin/pharmacology , Thymosin/physiology , Transcription Factors/metabolism , Transcriptional Activation/drug effects , Transfection , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Two-Hybrid System Techniques
8.
J Steroid Biochem Mol Biol ; 74(5): 279-85, 2000 Nov 30.
Article in English | MEDLINE | ID: mdl-11162936

ABSTRACT

Estrogens exert profound effects on the physiology of diverse target cells and these effects appear to be mediated by two estrogen receptor (ER) subtypes, ERalpha and ERbeta. We have investigated how ER ligands, ranging from pure agonists to antagonists, interact with ERalpha and ERbeta, and regulate their transcriptional activity on different genes. Mutational mapping-structure activity studies indicate that different residues of the ER ligand binding domain are involved in the recognition of structurally distinct estrogens and antiestrogens. We have identified from ligands of diverse structure, several particularly interesting ones that are high potency selective agonists via ERalpha and others that are full agonists through ERalpha while being full antagonists through ERbeta. Antiestrogens such as hydroxytamoxifen, which are mixed agonist/antagonists through ERalpha, are pure antagonists through ERbeta at estrogen response element-containing gene sites. Studies with ERalpha/beta chimeric proteins reveal that tamoxifen agonism requires the activation function 1 region of ERalpha. Through two-hybrid assays, we have isolated an ER-specific coregulator that potentiates antiestrogen antagonist effectiveness and represses ER transcriptional activity. We have also focused on understanding the distinct pharmacologies of antiestrogen- and estrogen-regulated genes. Although antiestrogens are thought to largely act by antagonizing the actions of estrogens, we have found among several new ER-regulated genes, quinone reductase (QR), a detoxifying phase II antioxidant enzyme, that has its activity up-regulated by antiestrogens in an ER-dependent manner in breast cancer cells. This response is antagonized by estrogens, thus showing 'reversed pharmacology'. Increased QR activity by antiestrogens requires a functional ER (ERalpha or ERbeta) and is, interestingly, mediated via the electrophile response element in the QR gene 5' regulatory region. The up-regulation of QR may contribute to the beneficial effects of tamoxifen, raloxifene, and other antiestrogens in breast cancer prevention and treatment. Estrogens rapidly up-regulate expression of several genes associated with cell cytoarchitectural changes including NHE-RF, the sodium hydrogen exchanger regulatory factor, also known as EBP50. NHE-RF/EBP50 is enriched in microvilli, and may serve as a scaffold adaptor protein in regulating early changes in cell architecture and signal transduction events induced by estrogen. Analyses of the regulatory regions of these primary response genes, and the antioxidant and other signaling pathways involved, are providing considerable insight into the mechanisms by which ligands, that function as selective estrogen receptor modulators or SERMs, exert their marked effects on the activities and properties of target cells. The intriguing biology of estrogens in its diverse target cells is thus determined by the structure of the ligand, the ER subtype involved, the nature of the hormone-responsive gene promoter, and the character and balance of coactivators and corepressors that modulate the cellular response to the ER-ligand complex. The continuing development of ligands that function as selective estrogens or antiestrogens for ERalpha or ERbeta should allow optimized tissue selectivity of these agents for menopausal hormone replacement therapy and the treatment and prevention of breast cancer.


Subject(s)
Estrogens/pharmacology , Receptors, Estrogen/metabolism , Estrogen Antagonists/pharmacology , Estrogen Receptor Modulators/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Ligands , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/chemistry , Selective Estrogen Receptor Modulators/pharmacology , Structure-Activity Relationship , Substrate Specificity
9.
Proc Natl Acad Sci U S A ; 96(12): 6947-52, 1999 Jun 08.
Article in English | MEDLINE | ID: mdl-10359819

ABSTRACT

The action of nuclear hormone receptors is tripartite, involving the receptor, its ligands, and its coregulator proteins. The estrogen receptor (ER), a member of this superfamily, is a hormone-activated transcription factor that mediates the stimulatory effects of estrogens and the inhibitory effects of antiestrogens such as tamoxifen in breast cancer and other estrogen target cells. To understand how antiestrogens and dominant negative ERs suppress ER activity, we used a dominant negative ER as bait in two-hybrid screening assays from which we isolated a clone from breast cancer cells that potentiates the inhibitory activities of dominant negative ERs and antiestrogen-liganded ER. At higher concentrations, it also represses the transcriptional activity of the estradiol-liganded ER, while having no effect on other nuclear hormone receptors. This clone, denoted REA for "repressor of estrogen receptor activity," encodes a 37-kDa protein that is an ER-selective coregulator. Its competitive reversal of steroid receptor coactivator 1 enhancement of ER activity and its direct interaction with liganded ER suggest that it may play an important role in determining the sensitivity of estrogen target cells, including breast cancer cells, to antiestrogens and estrogens.


Subject(s)
Estrogens/metabolism , Hormone Antagonists/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Amino Acid Sequence , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CHO Cells , Cricetinae , Estrogens/genetics , Female , Humans , Molecular Sequence Data , Prohibitins , Receptors, Estrogen/agonists , Receptors, Estrogen/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
10.
Mol Cell Endocrinol ; 139(1-2): 37-43, 1998 Apr 30.
Article in English | MEDLINE | ID: mdl-9705072

ABSTRACT

Among the four isoforms of the calcitonin receptor (CTR) described in humans, two differ by the presence of h-CTR1 or absence of h-CTR2 of 16 amino acids in the first intracellular loop. Both receptors are biologically active. The TT cell line derived from a human medullary carcinoma of the thyroid is characterized by the secretion of large amounts of calcitonin. We have recently shown that this cell line expresses h-CTR2. In the present work we have studied the expression of CTR during TT cell proliferation and used dexamethasone to modify calcitonin expression in order to establish if an autocrine regulation involving calcitonin and its receptor was functional in the TT cells. The expression of this receptor and of calcitonin during TT cell proliferation was studied by reverse transcriptase-polymerase chain reaction (RT-PCR). Dexamethasone, a potent inhibitor of TT cell proliferation, levels (day 6 of culture) specifically increased receptor levels from day 8 onwards. CT peptide and CT mRNA levels decreased or were similar during experimental time. CTR regulation by glucocorticoids is suggested in TT cells. Autocrine regulation of CTR is also suggested by relation between CT mRNA levels and CTR mRNA.


Subject(s)
Autocrine Communication/genetics , Dexamethasone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glucocorticoids/pharmacology , Receptors, Calcitonin/genetics , Calcitonin/biosynthesis , Calcitonin/genetics , Carcinoma, Medullary/genetics , Carcinoma, Medullary/pathology , Cell Division , Humans , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Tumor Cells, Cultured
11.
Thyroid ; 8(2): 141-7, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9510122

ABSTRACT

We recently reported the presence of a truncated form (h-CTR2) of the human calcitonin receptor (CTR) in TT cells, a cell line derived from medullary thyroid carcinoma (MTC). This form (h-CTR2), characterized by the absence of 16 amino acids in the first intracellular domain, was also detected in two cases of MTC. In the present study we determined the expression of CTR mRNA in a larger sample, representative of the different clinical forms of MTC, and in normal thyroid. h-CTR2 was expressed in all MTC specimens (both sporadic and familial) and in the normal thyroid samples. The expression of the receptor mRNA was higher in MTC compared with normal thyroid. Moreover, CT and CTR mRNA levels were modified significantly during proliferation. This result suggests that CT may be involved in proliferation of MTC via autocrine/paracrine regulation. Calcitonin secretion by MTC may play a role in the development and spread of these tumors.


Subject(s)
Carcinoma, Medullary/metabolism , RNA, Messenger/metabolism , Receptors, Calcitonin/genetics , Thyroid Neoplasms/metabolism , Carcinoma, Medullary/pathology , Cell Division/physiology , Extracellular Space/metabolism , Female , Humans , Male , Receptors, Calcitonin/metabolism , Reference Values , Thyroid Gland/metabolism , Thyroid Neoplasms/pathology , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
12.
Blood ; 89(6): 2105-12, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9058733

ABSTRACT

1,25-Dihydroxyvitamin D3 [1,25-(OH)2D3] induces the differentiation of monocytes into macrophage-like cells in vitro. To identify the genes expressed during this process, we performed differential display polymerase chain reaction on RNA extracted from cord blood monocytes (CBMs) treated with 1,25-(OH)2D3. Treated CBMs expressed type-I 15-hydroxyprostaglandin dehydrogenase (type-I 15-PGDH), the key enzyme of prostaglandin E2 (PGE2) catabolism and a 15-PGDH-related mRNA (15-PGDHr). This newly described 15-PGDH-related mRNA was constitutively expressed in adult monocytes. 15-PGDH gene(s) transcription was accompanied by the appearance of the 15-PGDH activity in treated CBMs. In addition, the cyclooxygenase 2 mRNA level was decreased and PGE2 levels in the culture mediums were lowered (50%). Our results stress that 1,25-(OH)2D3, at least in neonatal monocytes, can exert, directly or indirectly, a dual control on key enzymes of PGE2 metabolism. In conclusion, we suggest that modifications in prostaglandin metabolism, induced by the expression of type-I 15-PGDH and the downregulation of cyclooxygenase 2, could be involved in monocytic differentiation.


Subject(s)
Calcitriol/pharmacology , Fetal Blood/enzymology , Hydroxyprostaglandin Dehydrogenases/biosynthesis , Monocytes/enzymology , Adult , Culture Media , Cyclooxygenase 2 , Dinoprostone/analysis , Enzyme Activation , Enzyme Induction , Fetal Blood/chemistry , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/isolation & purification , Isoenzymes/genetics , Membrane Proteins , Monocytes/chemistry , NAD/physiology , Polymerase Chain Reaction , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/analysis
13.
Gene ; 188(1): 143-8, 1997 Mar 25.
Article in English | MEDLINE | ID: mdl-9099873

ABSTRACT

NAD+-dependent 15-hydroxyprostaglandin dehydrogenase (type-I 15-PGDH) inactivates prostaglandins. We recently reported an mRNA sequence coding for a predicted isomer (PGDH(rI)) of this enzyme. The TT cell line, derived from medullary thyroid carcinoma (MTC), expresses mRNAs for both isomers. We report here the expression by TT cells and MTC of a third 15-PGDH related mRNA (PGDH(rII)), 241 nt shorter than type-I 15-PGDH. RNase protection assays confirmed that TT cells expressed this mRNA (PGDH(rII)). Thus different splicing patterns could be involved in the post-transcriptional regulation of type-I 15-PGDH gene in MTC.


Subject(s)
Hydroxyprostaglandin Dehydrogenases/genetics , RNA, Messenger/genetics , Amino Acid Sequence , Base Sequence , Blotting, Western , Cloning, Molecular , DNA, Complementary , Humans , Hydroxyprostaglandin Dehydrogenases/metabolism , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tissue Distribution , Tumor Cells, Cultured
14.
Hum Genet ; 99(2): 279-81, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9048936

ABSTRACT

The gene encoding the human NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase, designated type-I 15-PGDH, was mapped to chromosome 4 by analyzing its segregation in a panel of human-hamster somatic cell hybrids. This gene was further localized to bands 4q34-q35 by in situ hybridization on human chromosomes.


Subject(s)
Chromosomes, Human, Pair 4 , Hydroxyprostaglandin Dehydrogenases/genetics , Chromosome Banding , HL-60 Cells , Humans , Hybrid Cells , In Situ Hybridization
17.
Gene ; 162(2): 319-22, 1995 Sep 11.
Article in English | MEDLINE | ID: mdl-7557451

ABSTRACT

We amplified, using the polymerase chain reaction (PCR) and NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase (type-I 15-PGDH)-specific primers, RNA extracted from the HL-60 cell line. Two bands, differing in size by approx. 160 bp, were detected with ethidium bromide staining after electrophoresis of amplification products and hybridization with a 15-PGDH-specific probe. Sequencing these DNA bands revealed that the largest corresponded to the 15-PGDH cloned from human placenta [Ensor et al., J. Biol. Chem. 265 (1990) 14888-14891]. The smaller sequence coded for a predicted C-terminal-truncated form of 15-PGDH. This subtype of the type-I 15-PGDH mRNA was also found using RT-PCR in human liver, placenta and a cell line derived from a human medullary thyroid carcinoma (TT cells). Hybridization studies using specific probes indicated that this new mRNA form probably corresponded to the 3.4-kb mRNA, one of the two 15-PGDH mRNAs previously detected in Northern blot analysis.


Subject(s)
Hydroxyprostaglandin Dehydrogenases/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Gene Expression , HL-60 Cells , Humans , Isoenzymes/genetics , Molecular Sequence Data , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL