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










Publication year range
1.
J Steroid Biochem Mol Biol ; 142: 39-47, 2014 Jul.
Article in English | MEDLINE | ID: mdl-23954501

ABSTRACT

Progestogens are widely used in contraception and in hormone therapy. Biochemical and molecular biological evidence suggests that progestogens differ widely in their affinities and transcriptional effects via different steroid receptors, and hence cannot be considered as a single class of compounds. Consistent with these observations, recent clinical evidence suggests that, despite their similar progestogenic actions, these differences underlie different side-effect profiles for cardiovascular disease and susceptibility to infectious diseases. However, choice of progestogen for maximal benefit and minimal side-effects is hampered by insufficient comparative clinical and molecular studies to understand their relative mechanisms of action, as well as their relative potencies for different assays and clinical effects. This review evaluates the usage, meaning and significance of the terms affinity, potency and efficacy in different models systems, with a view to improved understanding of their physiological and pharmacological significance. This article is part of a Special Issue entitled 'Menopause'.


Subject(s)
Estrogen Replacement Therapy/methods , Progestins/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Humans , Progestins/administration & dosage , Progestins/metabolism , Receptors, Steroid/metabolism
2.
Cochrane Database Syst Rev ; (3): CD005214, 2006 Jul 19.
Article in English | MEDLINE | ID: mdl-16856087

ABSTRACT

BACKGROUND: There are two injectable progestogen-only contraceptives (IPCs) that have been available in many countries in the world since 1983. They are both still extensively used in many developing countries, forming a large proportion of the health system's expenditure on contraception. These are depot medroxyprogesterone acetate (DMPA) and norethisterone oenanthate (NET-EN). These are both highly effective contraceptives that receive wide acceptance amongst women in their fertile years. They differ in frequency of administration that has implications on patient uptake. They also differ in cost that may significantly affect budgeting in the health system. A systematic comparison will aid to ensure their rational use. OBJECTIVES: To determine if there are differences between depot medroxyprogesterone acetate given at a dose of 150 mg IM every 3 months and norethisterone oenanthate given at a dose of 200mg IM every 2 months, in terms of contraceptive effectiveness, reversibility and discontinuation patterns, minor effects and major effects. SEARCH STRATEGY: We searched the computerized databases MEDLINE using PubMed, Popline, Cochrane Controlled Trials Register, Biblioline, LILACS, EMBASE and PASCAL for randomised controlled trials of DMPA versus NET-EN for long-acting progestogenic contraception. Studies were included regardless of language, and all databases were reviewed from the time that injectable progestogens have been in use. SELECTION CRITERIA: All randomised controlled comparisons of DMPA acetate given at a dose of 150 mg IM every 3 months versus NET-EN given at a dose of 200mg IM every 2 months, used for contraception, were included. Trials had to report on contraceptive efficiency and return to fertility, discontinuation risks and reasons for discontinuation, and clinical effects, both menstrual and non-menstrual. DATA COLLECTION AND ANALYSIS: BD and CM evaluated the titles and abstracts obtained through applying the search strategy and applied the eligibility criteria. BD attempted to contact authors where clarification of the data was required, and contacted all main manufacturers of the contraceptives. After inclusion of the two studies, the data was abstracted and analysed with RevMan 4.2. MAIN RESULTS: Two trials were included in this review. There was no significant difference between the two treatment groups for the frequency of discontinuation for either contraceptive, although the women on NET-EN were 4% more likely to discontinue for personal reasons than those on DPMA. Discontinuation because of accidental pregnancy did not differ between the groups. Although the duration of bleeding and spotting events was the same in each group, women on DPMA were 21% more likely to develop amenorrhoea. Mean changes in body weight at 12 and 24 months, and in systolic and diastolic blood pressure at 12 months did not differ significantly between the studies. AUTHORS' CONCLUSIONS: While the choice between DPMA and NET-EN as injectable progestogen contraceptives may vary between both health providers and patients, data from randomized controlled trials indicate little difference between the effects of these methods, except that women on DMPA are more likely to develop amenorrhoea. There is inadequate data to detect differences in some non-menstrual major and minor clinical effects.


Subject(s)
Contraception/methods , Contraceptive Agents, Female/administration & dosage , Medroxyprogesterone Acetate/administration & dosage , Norethindrone/analogs & derivatives , Delayed-Action Preparations/administration & dosage , Drug Administration Schedule , Female , Humans , Norethindrone/administration & dosage , Randomized Controlled Trials as Topic
3.
J Neuroendocrinol ; 17(10): 619-38, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16159375

ABSTRACT

Gonadotrophin-releasing hormone (GnRH), acting via its cognate GnRH receptor (GnRHR), is the primary regulator of mammalian reproductive function, and hence GnRH analogues are extensively used in the treatment of hormone-dependent diseases, as well as for assisted reproductive techniques. In addition to its established endocrine role in gonadotrophin regulation in the pituitary, evidence is rapidly accumulating to support the expression and functional roles for two forms of GnRHR (GnRHR I and GnRHR II) in multiple and diverse extra-pituitary mammalian tissues and cells. These findings, together with findings indicating that mutations of the GnRHR are linked to the disease hypogonadotrophic hypogonadism and that GnRHRs play a direct role in neuronal migration and reproductive cancers, have presented new therapeutic targets and intensified research into the structure, function and mechanisms of regulation of expression of GnRHR genes. The present review focuses on the current knowledge on tissue-specific and hormonal regulation of transcription of mammalian GnRH receptor genes. Emerging insights, such as the discovery of diverse regulatory mechanisms in pituitary and extra-pituitary cell types, nonclassical mechanisms of steroid regulation, the use of composite elements for cell-specific expression, the increasing profile of hormones involved in regulation, the complexity of kinase pathways that target the GnRHR I gene, as well as species-differences, are highlighted. Although further research is necessary to understand the mechanisms of regulation of expression of GnRHR I and GnRHR II genes, the GnRHR is emerging as a potential target gene for facilitating cross-talk between neuroendocrine, immune and stress-response systems in multiple tissues via autocrine, paracrine and endocrine signalling.


Subject(s)
Gene Expression Regulation/physiology , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Reproduction/genetics , Signal Transduction/genetics , Animals , Gonadotropin-Releasing Hormone/metabolism , Humans , Mammals/genetics , Mammals/physiology , Mice , Pituitary Gland/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , Rats , Signal Transduction/physiology , Species Specificity , Tissue Distribution , Transcription, Genetic/physiology
4.
Mol Cell Endocrinol ; 201(1-2): 155-64, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12706303

ABSTRACT

We investigated the interactions between Compound A (CpdA), an analog of a hydroxyphenyl aziridine precursor found in an African shrub, and the androgen receptor (AR). CpdA represses androgen-induced activation of both specific and non-specific androgen DNA response elements. While a similar effect was obtained for the progesterone receptor (PR) via a non-specific hormone response element, CpdA had no effect on the actions of the glucocorticoid and mineralocorticoid receptors. CpdA represses the ligand-dependent interaction between the NH(2)- and COOH-terminal domains of the AR, similar to well-characterised anti-androgens. CpdA also interferes with the interaction of steroid receptor co-activator 1 (SRC1) with the activation domain AF2 but not with AF1. However, CpdA does not compete with androgen for binding to the AR. These results demonstrate that CpdA elicits anti-androgenic actions by a mechanism other than competitive binding for the AR.


Subject(s)
Acetates/pharmacology , Androgen Antagonists/pharmacology , Androgens/metabolism , Ethylamines/pharmacology , Receptors, Androgen/metabolism , Animals , Binding Sites , Cells, Cultured , DNA/metabolism , Gene Expression Regulation , Genes, Reporter , Haplorhini , Histone Acetyltransferases , Humans , Ligands , Male , Nuclear Receptor Coactivator 1 , Plants/chemistry , Promoter Regions, Genetic , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Structure, Tertiary , Receptors, Androgen/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Tyramine/analogs & derivatives
5.
Biol Reprod ; 67(6): 1741-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12444048

ABSTRACT

GnRH regulates reproduction via the well-characterized mammalian pituitary GnRH receptor (type I). In addition, two homologous genes for a second form of the GnRH receptor (type II) are present in the human genome, one on chromosome 14 and the second on chromosome 1. The chromosome 14 gene is ubiquitously transcribed at high levels in the antisense orientation but lacks exon 1, required to encode a full-length receptor. In comparison, the chromosome 1 gene contains all three exons. The issue of whether this gene is transcribed in any human tissue(s), and whether these transcripts encode a functional receptor protein, remains unresolved. We have directly addressed this by screening a panel of human RNAs by hybridization and RT-PCR. These analyses showed that, unlike the chromosome 14 gene, chromosome 1 gene expression is limited and of low abundance. Exon 1-containing transcripts were detected by in situ hybridization in mature sperm and in human postmeiotic testicular cells. Further sequence analysis revealed that although all the potential coding segments were present, the human transcripts, like the gene, contain a stop codon within the coding region and a frame-shift relative to other mammalian GnRH receptors. Although this suggests that the human gene may be a transcribed pseudogene, a functional type II GnRH receptor cDNA has recently been cloned from monkeys. Given the well-established role of GnRH in spermatogenesis and reported evidence of type II GnRH receptor immunoreactivity in human tissues, it is possible that the chromosome 1 gene is functional.


Subject(s)
RNA, Messenger/analysis , Receptors, LHRH/genetics , Spermatozoa/chemistry , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 14 , Cloning, Molecular , Exons , Humans , In Situ Hybridization , Male , Molecular Sequence Data , Pseudogenes , RNA Editing , Receptors, LHRH/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, DNA
6.
Cell Biol Int ; 25(1): 17-31, 2001.
Article in English | MEDLINE | ID: mdl-11237405

ABSTRACT

GC-rich DNA cis elements are important transcriptional regulatory elements present in the promoter, enhancer and locus control regions of many eukaryotic genes from several species. This review attempts to examine the structure, function and biological significance of GC-rich cis -regulatory elements and their cognate binding proteins, with a view to understanding their role in regulation of gene expression.


Subject(s)
Cytosine , Enhancer Elements, Genetic , Gene Expression Regulation , Guanine , Animals , Cell Cycle/genetics , Chromatin/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Humans , Promoter Regions, Genetic , Transcription, Genetic
7.
J Biol Chem ; 276(11): 7754-61, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11112780

ABSTRACT

Mammalian receptors for gonadotropin-releasing hormone (GnRH) have over 85% sequence homology and similar ligand selectivity. Biological studies indicated that the chicken GnRH receptor has a distinct pharmacology, and certain antagonists of mammalian GnRH receptors function as agonists. To explore the structural determinants of this, we have cloned a chicken pituitary GnRH receptor and demonstrated that it has marked differences in primary amino acid sequence (59% homology) and in its interactions with GnRH analogs. The chicken GnRH receptor had high affinity for mammalian GnRH (K(i) 4.1 +/- 1.2 nM), similar to the human receptor (K(i) 4.8 +/- 1.2 nM). But, in contrast to the human receptor, it also had high affinity for chicken GnRH ([Gln(8)]GnRH) and GnRH II ([His(5),Trp(7),Tyr(8)]GnRH) (K(i) 5.3 +/- 0.5 and 0.6 +/- 0.01 nM). Three mammalian receptor antagonists were also pure antagonists in the chicken GnRH receptor. Another three, characterized by D-Lys(6) or D-isopropyl-Lys(6) moieties, functioned as pure antagonists in the human receptor but were full or partial agonists in the chicken receptor. This suggests that the Lys side chain interacts with functional groups of the chicken GnRH receptor to stabilize it in the active conformation and that these groups are not available in the activated human GnRH receptor. Substitution of the human receptor extracellular loop two with the chicken extracellular loop two identified this domain as capable of conferring agonist activity to mammalian antagonists. Although functioning of antagonists as agonists has been shown to be species-dependent for several GPCRs, the dependence of this on an extracellular domain has not been described.


Subject(s)
Receptors, LHRH/chemistry , Amino Acid Sequence , Animals , Binding Sites , Chickens , Cloning, Molecular , Lysine , Molecular Sequence Data , Protein Conformation , Receptors, LHRH/agonists , Receptors, LHRH/antagonists & inhibitors , Species Specificity , Structure-Activity Relationship
8.
Endocrinology ; 141(5): 1764-71, 2000 May.
Article in English | MEDLINE | ID: mdl-10803587

ABSTRACT

We have cloned the full-length complementary DNA (cDNA) for a GnRH receptor from Xenopus laevis pituitary cDNA and determined its gene structure. The cDNA encodes a 368-amino acid protein that has a 46% amino acid identity to the human GnRH receptor. The X laevis GnRH receptor has all of the amino acids identified in the mammalian GnRH receptors as sites of interaction with the GnRH ligand. However, this receptor cDNA shares the same distinguishing structural features of the GnRH receptor that have been characterized from other nonmammalian vertebrates. These include the pair of aspartate residues in the transmembrane domains II and VII compared with the aspartate/asparagine arrangement in mammalian receptors, the amino acid PEY motif in extracellular loop III (SEP in mammals), and the presence of a carboxyl-terminal tail. Previous studies have reported that mammalian GnRH was equipotent to other naturally occurring GnRH subtypes in stimulating LH release from the amphibian pituitary. However, in this study we show that the X. laevis GnRH receptor has ligand selectivity for the naturally occurring GnRHs similar to other nonmammalian GnRH receptors. The order of potency of the GnRHs in stimulating inositol phosphate production in COS-1 cells transiently transfected with the X. laevis GnRH receptor cDNA was chicken GnRH II>salmon GnRH>mammalian GnRH. Transcripts of this GnRH receptor are expressed in the pituitary and midbrain of X. laevis.


Subject(s)
DNA, Complementary/genetics , Gene Expression Regulation , Mesencephalon/metabolism , Pituitary Gland/metabolism , Receptors, LHRH/genetics , Xenopus laevis/metabolism , Amino Acid Sequence , Animals , Goldfish , Gonadotropin-Releasing Hormone/metabolism , Humans , Ligands , Molecular Sequence Data , Receptors, LHRH/biosynthesis , Sequence Alignment , Xenopus laevis/genetics
9.
J Endocrinol ; 162(1): 117-26, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10396028

ABSTRACT

Gonadotropin releasing hormone (GnRH) regulates the reproductive system through a specific G-protein-coupled receptor (GPCR) in pituitary gonadotropes. The existence of two (or more) forms of GnRH in most vertebrates suggested the existence of GnRH receptor subtypes (I and II). Using sequence information for extracellular loop 3 of a putative Type II GnRH receptor from a reptile species, we have looked for a Type II GnRH receptor gene in the human genome EST (expressed sequence tag) database. A homolog was identified which has 45% and 41% amino acid identity with exons 2 and 3 of the known human GnRH pituitary receptor (designated Type I) and much lower homology with all other GPCRs. A total of 27 contiguous ESTs was found and comprised a continuous sequence of 1642 nucleotides. The EST sequences were confirmed in the cloned human gene and in PCR products of cDNA from several tissues. All EST transcripts detected were in the antisense orientation with respect to the novel GnRH receptor sequence and were highly expressed in a wide range of human brain and peripheral tissues. PCR of cDNA from a wide range of tissues revealed that intronic sequence equivalent to intron 2 of the Type I GnRH receptor was retained. The failure to splice out putative intron sequences in transcripts which spanned exon-intron boundaries is expected in antisense transcripts, as candidate donor and acceptor sites were only present in the gene when transcribed in the orientation encoding the GnRH receptor homolog. No transcripts extended 5' to the sequence corresponding to intron 2 of the Type I GnRH as the antisense transcripts terminated in poly A due to the presence of a polyadenylation signal sequence in the putative intron 2 when transcribed in the antisense orientation. These findings suggest that a Type II GnRH receptor gene has arisen during vertebrate evolution and is also present in the human. However, the receptor may have become vestigial in the human, possibly due to the abundant and universal tissue transcription of the opposite DNA strand to produce antisense RNA.


Subject(s)
Expressed Sequence Tags , Gonadotropin-Releasing Hormone/genetics , Receptors, LHRH/genetics , Base Sequence , Blotting, Northern , DNA, Antisense , Humans , Molecular Sequence Data , Polymerase Chain Reaction/methods
10.
Proc Natl Acad Sci U S A ; 96(5): 2526-31, 1999 Mar 02.
Article in English | MEDLINE | ID: mdl-10051676

ABSTRACT

In the goldfish (Carassius auratus) the two endogenous forms of gonadotropin-releasing hormone (GnRH), namely chicken GnRH II ([His5, Trp7,Tyr8]GnRH) and salmon GnRH ([Trp7,Leu8]GnRH), stimulate the release of both gonadotropins and growth hormone from the pituitary. This control is thought to occur by means of the stimulation of distinct GnRH receptors. These receptors can be distinguished on the basis of differential gonadotropin and growth hormone releasing activities of naturally occurring GnRHs and GnRHs with variant amino acids in position 8. We have cloned the cDNAs of two GnRH receptors, GfA and GfB, from goldfish brain and pituitary. Although the receptors share 71% identity, there are marked differences in their ligand selectivity. Both receptors are expressed in the pituitary but are differentially expressed in the brain, ovary, and liver. Thus we have found and cloned two full-length cDNAs that appear to correspond to different forms of GnRH receptor, with distinct pharmacological characteristics and tissue distribution, in a single species.


Subject(s)
Brain/physiology , Goldfish/physiology , Pituitary Gland/physiology , Receptors, LHRH/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , DNA Primers , Genetic Variation , Humans , In Situ Hybridization , Molecular Sequence Data , Organ Specificity , Polymerase Chain Reaction , Receptors, LHRH/chemistry , Receptors, LHRH/classification , Sequence Alignment , Sequence Homology, Amino Acid
11.
Gen Comp Endocrinol ; 112(3): 296-302, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9843635

ABSTRACT

The majority of vertebrates have two or three forms of gonadotropin-releasing hormone (GnRH), which appear to have arisen by successive gene duplication events. This suggests the possibility of concordant gene duplications of the GnRH receptor to produce two or more cognate receptors. Since the extracellular loop 3 (EC3) domain of mammalian GnRH receptors plays a role in distinguishing the different forms of GnRH, we have contemplated that the sequence of this domain will differ significantly in the putative cognate receptors. Degenerate oliognucleotides encoding the sequences of the transmembrane domains preceding and following EC3 were used for PCR amplification of genomic DNA from zebrafish (Brachydanio rerio), goldfish (Carassius auratus), African clawed frog (Xenopus laevis), chicken (Gallus domesticus), and lizard (Agama atra). Isolation and sequencing of specific clones revealed that they fell into three groups. Two of these were most similar to the mammalian pituitary GnRH receptor and were therefore designated Type IA and Type IB. The third form (designated Type II) was most different from the others and was identified in Xenopus, lizard, and human DNA. These findings support the concept of the existence of three distinct GnRH receptors, which have evolved in conjunction with three distinct GnRH ligand classes present in many vertebrates.


Subject(s)
Receptors, LHRH/metabolism , Vertebrates/metabolism , Amino Acid Sequence , Animals , Base Sequence , Chickens , Conserved Sequence , Goldfish , Humans , Lizards , Molecular Sequence Data , Polymerase Chain Reaction , Receptors, LHRH/genetics , Species Specificity , Xenopus laevis , Zebrafish
12.
Eur J Endocrinol ; 139(4): 438-47, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9820623

ABSTRACT

OBJECTIVE: Construction of constitutively active mutants of the GnRH receptor, a member of the G-protein coupled receptor superfamily, would facilitate investigation of the mechanism of receptor activation. DESIGN: Point mutations were introduced in the human GnRH receptor in positions corresponding to those which caused constitutive activity in other G-protein coupled receptors. The effects of these mutations on ligand binding, receptor intracellular signaling and receptor expression were determined. METHODS: Wild type and mutated receptor cDNAs were expressed in COS-1 cells. Basal and agonist-stimulated inositol phosphate production and ligand binding were determined. In addition, receptor mRNA levels, cell surface receptor stability and rate of internalization were measured. RESULTS AND CONCLUSIONS: Although none of the mutant receptors exhibited constitutive activity, mutation of Phe-2 72 in transmembrane helix VI to Leu increased cell surface receptor numbers, with unchanged affinities for radiolabeled agonist, superagonist and antagonist peptides compared with wild type receptor. The cell surface receptor stability and rate of internalization were similar for wild type and F272L GnRH receptors. Thus a single amino acid mutation in transmembrane helix VI causes an increase in cell surface receptor numbers, which appears to result from an increased rate of receptor protein translation, processing or insertion into membranes.


Subject(s)
Amino Acid Substitution , Gene Expression , Protein Structure, Secondary , Receptors, LHRH/chemistry , Receptors, LHRH/genetics , Animals , Blotting, Northern , COS Cells , Cell Membrane/chemistry , Humans , Mutagenesis, Site-Directed , Phenylalanine/genetics , Radioligand Assay , Receptors, LHRH/metabolism , Structure-Activity Relationship , Transfection
13.
Biochem J ; 331 ( Pt 3): 893-6, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9560319

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is a decapeptide that regulates reproductive function via binding to the GnRH receptor, which is a G-protein-coupled receptor (GPCR). For several members of this family, the C-terminal domain of intracellular loop III is important in ligand-mediated coupling to G-proteins; mutations in that region can lead to constitutive activity. A specific alanine residue is involved in certain GPCRs, the equivalent of which is Ala-261 in the GnRH receptor. Mutation of this residue to Leu, Ile, Lys, Glu or Phe in the human GnRH receptor did not result in constitutive activity and instead led to complete uncoupling of the receptor (failure to support GnRH-stimulated inositol phosphate production). When this residue was mutated to Gly, Pro, Ser or Val, inositol phosphate production was still supported. All the mutants retained the ability to bind ligand, and the affinity for ligand, where measured, was unchanged. These results show that Ala-261 cannot be involved in ligand binding but is critical for coupling of the receptor to its cognate G-protein. Coupling is also dependent on the size of the residue in position 261. When the amino acid side chain has a molecular mass of less than 40 Da efficient coupling is still possible, but when its molecular mass exceeds 50 Da the receptor is uncoupled. Internalization studies on the Ala261-->Lys mutant showed a marked decrease in receptor internalization compared with the wild type, indicating that coupling is necessary for effective receptor internalization in the GnRH receptor system. Activation of protein kinase C (with PMA), but not protein kinase A (with forskolin) markedly increased the internalization of the mutant receptor while having a small effect on the wild-type receptor.


Subject(s)
Alanine/chemistry , GTP-Binding Proteins/physiology , Receptors, LHRH/chemistry , Animals , Binding Sites/genetics , Binding, Competitive , COS Cells , Colforsin/pharmacology , Endocytosis/physiology , Gonadotropin-Releasing Hormone/analogs & derivatives , Humans , Inositol Phosphates/metabolism , Ligands , Mutagenesis, Site-Directed/genetics , Protein Binding/genetics , Receptors, LHRH/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transfection/genetics
14.
Biochemistry ; 36(42): 12881-9, 1997 Oct 21.
Article in English | MEDLINE | ID: mdl-9335546

ABSTRACT

Photoaffinity cross-linking with [azidobenzoyl-d-Lys6]GnRH leads to irreversible activation of the gonadotropin-releasing hormone (GnRH) receptor. In order to localize the cross-linking site, the disulfide bridge structure was initially probed by mutagenesis. A consistent pattern of changes in the ability of GnRH to stimulate signal transduction after Ser substitutions of extracellularly located Cys residues indicated that Cys14 in the N-terminal domain is connected to Cys200 in the second extracellular loop, while Cys196 in this loop is connected to the highly conserved Cys114 at the extracellular end of transmembrane helix 3. Protein chemical analysis of radioactive fragments of cross-linked GnRH receptor following deglycosylation and enzymatic digest with endoproteinase Glu-C and trypsin before and after introduction or elimination of potential protease cleavage sites indicated that 125I[azidobenzoyl-d-Lys6]GnRH cross-links to a segment comprising residues 12-18 of the N-terminal domain. The existence of the Cys114-Cys196 bridge was directly confirmed as a labeled fragment, including that Cys114 was resolvable only under reducing conditions. The observation that the cross-linked N-terminal enzymatic fragments had identical apparent size under non-reducing conditions shows that the cross-linking reaction disconnected the disulfide bridge between Cys14 and Cys200 and indicates that Cys14 is probably the residue involved in cross-linking of the ligand. It is concluded that covalent tethering of GnRH through a photoreactive side chain located at position 6 in the middle of the peptide leads to continued activation of the receptor presumably through covalent binding to Cys14 in the N-terminal domain of the receptor.


Subject(s)
Azides/metabolism , Cysteine , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/metabolism , Protein Conformation , Receptors, LHRH/chemistry , Receptors, LHRH/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Azides/pharmacology , Binding Sites , COS Cells , Cloning, Molecular , Cross-Linking Reagents , Gonadotropin-Releasing Hormone/pharmacology , Humans , Inositol Phosphates/metabolism , Kinetics , Mice , Models, Structural , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sheep , Transfection
15.
Clin Endocrinol (Oxf) ; 47(5): 549-54, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9425394

ABSTRACT

BACKGROUND AND OBJECTIVE: The mechanisms of tumourogenesis for the majority of pituitary tumours are unknown. Mutations of G-protein coupled receptors (GPCRs) have recently been described as important in diverse human diseases, including thyroid adenomas. To test this hypothesis in pituitary gonadotroph adenomas, we amplified and sequenced the GnRH receptor gene in 12 human tumours. We restricted our analysis to the third exon, since this represents the hotspot for activating mutations in other GPCRs. PATIENTS: Pituitary adenoma tissue was identified from patients who had tumours resected and where a diagnosis of gonadotroph adenoma had been made on the basis of immunohistochemical demonstration of LH and/or FSH. METHODS: Genomic DNA was extracted from paraffin-embedded tissue of 18 gonadotroph adenomas. The third exon was successfully amplified by PCR in 12 cases and directly sequenced. RESULTS: We found no missense point mutations or even silent polymorphisms in any tumour studied. CONCLUSION: We conclude that activating mutations of the GnRH receptor gene do not represent an important mechanism of pituitary gonadotroph tumourogenesis.


Subject(s)
Adenoma/genetics , Pituitary Neoplasms/genetics , Point Mutation , Receptors, LHRH/genetics , Adenoma/metabolism , Adolescent , Adult , Electrophoresis, Agar Gel , Exons , Female , Follicle Stimulating Hormone/metabolism , Humans , Luteinizing Hormone/metabolism , Male , Middle Aged , Pituitary Neoplasms/metabolism , Polymerase Chain Reaction
16.
Nucleic Acids Res ; 24(21): 4349-55, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8932393

ABSTRACT

The sea urchin G-string binding factor (suGF1) is one of several proteins that bind sequence-specifically to oligo(dGxdC) motifs, frequently present upstream of eukaryotic genes. In this study we investigate the interaction of suGF1, purified to near homogeneity, with its oligo(dGxdC) binding site in a reconstituted nucleosome core in vitro. We show that the in vitro reconstitution of a 214 bp fragment containing a suGF1 binding site results in the appearance of five distinct nucleosome core species. These species contain the histone octamer in an identical rotational setting but in different translational frames. The resulting different nucleosomal locations of the suGF1 binding site in the five core species are shown to modulate the ability of suGF1 to bind to nucleosomal DNA, even though the rotational setting of the DNA in the nucleosome cores maximally exposes the suGF1 binding site. We propose that a direct protein-protein steric clash between suGF1 and the histone octamer is the most likely determinant in modulating the binding of suGF1 to its nucleosomally wrapped binding site. This result suggests that in vivo suGF1, like TBP, NF1 and heat shock factor, may require a complementary nucleosome disrupting activity or that suGF1 binds to free nascent replicated DNA prior to nucleosome deposition.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Protein Biosynthesis , Trans-Activators/metabolism , Animals , Histones/genetics , Nucleic Acid Conformation , Oligodeoxyribonucleotides/metabolism , Protein Binding , Protein Conformation , Sea Urchins
17.
Endocrine ; 4(3): 207-12, 1996 Jun.
Article in English | MEDLINE | ID: mdl-21153276

ABSTRACT

Mutation ofN-glycosylation sites in the mouse gonadotropin-releasing hormone receptor was previously shown to impair its expression in COS-1 cells. We therefore investigated the effects of adding an extra glycosylation site to the human gonadotropin-releasing hormone receptor, as a means for increasing its expression. Covalent labeling of the mutant receptor expressed in COS-1 cells with a gonadotropin-releasing hormone (GnRH) photoreactive analog demonstrated a shift in apparent molecular weight, indicating that the new site was in fact glycosylated. The receptor with extra glycosylation site displayed normal binding affinities for agonists buserelin and [D: -Ala(6)-Pro(9)-NHEt]-GnRH, and the antagonist antide, and a slightly increased affinity for GnRH. Receptor number was increased by 1.7-fold in membrane preparations from cells expressing the mutant receptor, compared with wild-type. Photoaffinity labeling of cell-surface receptors in intact cells demonstrated a 1.8-fold increase in binding sites on the cell surface. The GnRH receptor (GnRHR) with extra glycosylation site conferred a markedly enhanced signaling response to agonist. Dose-response curves for GnRH-stimulated inositol phosphate production were left-shifted by an average of 4.4-fold, and maximal inositol phosphate responses were increased by 1.2 fold, in cells transfected with mutant compared with wild-type receptor, indicating that the increase in binding sites represented functional receptors. These results demonstrate that addition of an extra glycosylation site enhances expression of the human GnRHR, a strategy that may be applicable to other cell-surface receptors.

18.
Mol Cell Biol ; 14(2): 1402-9, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8289815

ABSTRACT

Contiguous deoxyguanosine residues (G strings) have been implicated in regulation of gene expression in several organisms via the binding of G-string factors. Regulation of expression of the chicken adult beta-globin gene may involve the interplay between binding of an erythrocyte-specific G-string factor, BGP1, and the stability of a positioned nucleosome (C. D. Lewis, S. P. Clark, G. Felsenfeld, and H. Gould, Genes Dev. 2:863-873, 1988). We have purified a 59.5-kDa nuclear protein (suGF1) from sea urchin embryos by DNA affinity chromatography. suGF1 has high binding affinity and specificity for oligo(dG).oligo(dC). The identity of the purified protein was confirmed by renaturation of sequence-specific DNA-binding activity from a sodium dodecyl sulfate-polyacrylamide gel slice and by Southwestern (DNA-protein) blotting. suGF1 binds in vitro to a G11 string present in the H1-H4 intergenic region of a sea urchin early histone gene battery. This suGF1 DNA recognition site occurs within a homopurine-homopyrimidine stretch previously shown to be incorporated into a positioned nucleosome core in vitro. DNase I footprinting shows that suGF1 protects the same base pairs on the promoter of the chicken beta A-globin gene as does BGP1. We show that a G-string cis-regulatory element of a sea urchin cell lineage-specific gene LpS1 (M. Xiang, S.-Y. Lu, M. Musso, G. Karsenty, and W. H. Klein, Development 113:1345-1355, 1991) also represents a high-affinity recognition site for suGF1. suGF1 may be a member of a family of G-string factors involved in the regulation of expression of unrelated genes during development of a number of different organisms.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides/metabolism , Sea Urchins/metabolism , Animals , Base Sequence , Cell Nucleus/metabolism , Chromatography, Affinity , Chromatography, Ion Exchange , DNA/isolation & purification , DNA/metabolism , DNA-Binding Proteins/isolation & purification , Deoxyguanosine , Electrophoresis, Polyacrylamide Gel , Embryo, Nonmammalian/metabolism , Kinetics , Molecular Sequence Data , Nuclear Proteins/isolation & purification , Sea Urchins/embryology
19.
Mol Cell Biol ; 14(2): 1410-8, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8289816

ABSTRACT

We have elsewhere reported the purification of a poly(dG).poly(dC)-binding nuclear protein (suGF1) from sea urchin embryos (J. Hapgood and D. Patterton, Mol. Cell. Biol. 14:this issue, 1994). We proposed that suGF1 may be a member of a family of G-string factors involved in developmental gene regulation, possibly via alterations in chromatin structure. In this article, we characterize the binding of purified suGF1 to 11 contiguous Gs in the H1-H4 intergenic region of a sea urchin early histone gene battery in vitro. It is shown that suGF1-DNA binding is dependent on ionic strength and requires divalent cations. Purified suGF1 forms discrete protein-DNA multimers, consistent with suGF1-suGF1 interactions. In a model for the suGF1-DNA complex derived from our footprinting and methylation interference data, suGF1 contacts the Gs in the major groove as well as one of the bordering phosphate backbones. The data are consistent with the direction of curvature of the DNA in the suGF1-DNA complex being the same as that preferred by the free DNA and exhibited by the DNA when bent around a positioned nucleosome core in vitro. However, on the basis of steric considerations, the binding of suGF1 and that of the histone octamer are predicted to be mutually exclusive. We show that suGF1 is indeed unable to bind to the G string when occupied by a histone octamer located in the major in vitro positioning frame in the H1-H4 intergenic region.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Polydeoxyribonucleotides/metabolism , Animals , Base Sequence , Binding Sites , Chromatin/metabolism , DNA/isolation & purification , DNA/metabolism , DNA-Binding Proteins/isolation & purification , Deoxyribonuclease I , Histones/biosynthesis , Histones/genetics , Methylation , Models, Structural , Molecular Sequence Data , Nuclear Proteins/isolation & purification , Nucleic Acid Conformation , Sea Urchins/metabolism
20.
Mol Cell Biol ; 11(9): 4314-23, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1652054

ABSTRACT

Dioxin stimulates transcription from the cytochrome P-450IA1 promoter by interaction with the intracellular dioxin receptor. Upon binding of ligand, the receptor is converted to a form which specifically interacts in vitro with two dioxin-responsive positive control elements located in close proximity to each other about 1 kb upstream of the rat cytochrome P-450IA1 gene transcription start point. In rat liver, the cytochrome P-450IA1 gene is marked at the chromatin level by two DNase I-hypersensitive sites that map to the location of the response elements and exist prior to induction of transcription by the dioxin receptor ligand beta-naphthoflavone. In addition, a DNase I-hypersensitive site is detected near the transcription initiation site and is altered in nuclease sensitivity by induction. The presence of the constitutive DNase I-hypersensitive sites at the dioxin response elements correlates with the presence of a constitutive, labile factor which specifically recognizes these elements in vitro. This factor appears to be distinct from the dioxin receptor, which is observed only in nuclear extract from treated cells. In conclusion, these data suggest that a certain protein-DNA architecture may be maintained at the response elements at different stages of gene expression.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , DNA/metabolism , Deoxyribonuclease I/metabolism , Liver/enzymology , Receptors, Drug/metabolism , Animals , Base Sequence , Benzoflavones/pharmacology , Chromatin/ultrastructure , Cytochrome P-450 Enzyme System/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Liver/cytology , Liver/metabolism , Male , Molecular Sequence Data , Rats , Rats, Inbred Strains , Receptors, Aryl Hydrocarbon , Receptors, Drug/genetics , Transcription, Genetic , Xenobiotics , beta-Naphthoflavone
SELECTION OF CITATIONS
SEARCH DETAIL
...