Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Mol Endocrinol ; 23(3): 307-13, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10601976

ABSTRACT

We describe the properties of three monoclonal antibodies (MAbs) to ovine GH, two of which have previously been shown to enhance, in vivo, the biological activity of bovine and ovine growth hormone. We have examined the effects of these MAbs on GH activity in two appropriate GH-responsive cell culture systems, investigating both acute signalling effects (Janus-activated kinase (Jak)-2 tyrosine phosphorylation -5 min) and longer-term (MTT-formazan production -24 h) effects of hormone-antibody complexes. In the 3T3-F442A pre-adipocyte cell line (which has been demonstrated to be GH responsive), we show that complexation of recombinant bovine (rb) GH with either of the two enhancing anti-ovine GH MAbs (OA11 and OA15) and the non-enhancing MAb, OA14, attenuates the ability of GH to stimulate tyrosine phosphorylation of Jak-2 at a 5-min time point. Using the mouse myeloid cell line, FDC-P1, stably transfected with the full-length ovine GH receptor (oGHR), we demonstrate that rbGH causes a dose-dependent increase in MTT-formazan production by these cells. Further, we demonstrate that OA11 and OA14, but not OA15, cause a decrease in this stimulatory activity of rbGH over a hormone concentration range of 5-50 ng/ml at both 24 and 48 h. We conclude that the different in vitro activities of the two in vivo enhancing MAbs are most probably related to the time-courses over which these two assays are performed, and also to the relative affinities between antibody, hormone and receptor. In addition, the in vitro inhibitory activity of the enhancing MAb OA11 in both short- and long-term bioassay lends further support to an exclusively in vivo model for MAb-mediated enhancement of GH action.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigen-Antibody Complex/pharmacology , Growth Hormone/agonists , Growth Hormone/pharmacology , Proto-Oncogene Proteins , Signal Transduction/drug effects , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/enzymology , Adipocytes/metabolism , Animals , Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/immunology , Binding Sites , Cattle , Cell Line , Formazans/metabolism , Growth Hormone/antagonists & inhibitors , Growth Hormone/immunology , Janus Kinase 2 , Mice , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Prolactin/pharmacology , Protein-Tyrosine Kinases/metabolism , Rats , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Sheep , Substrate Specificity , Tetrazolium Salts/metabolism , Time Factors , Transfection
2.
Mol Pharmacol ; 54(5): 864-73, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9804621

ABSTRACT

Somatostatin (SRIF) is the main inhibitory peptide regulating growth hormone (GH) secretion. It has been difficult to establish the role of endogenous SRIF release in the absence of pure SRIF antagonists. Although several SRIF antagonists have recently been described, none have been shown to possess in vivo activity in the absence of added SRIF. Here, an SRIF antagonist with no detectable agonist activity has been identified from a synthetic combinatorial hexapeptide library containing 6.4 x 10(7) unique peptides. Each peptide in the library is amino-terminally acetylated and carboxyl-terminally amidated and consists entirely of D-amino acids. A SRIF-responsive yeast growth assay was used as a primary screening tool, and cAMP accumulation, competitive binding, and microphysiometry also were used to confirm and further characterize SRIF antagonist activity. The hexapeptide library was screened in stepwise iterative fashion to identify AC-178,335, a pure SRIF antagonist of the sequence Ac-hfirwf-NH2. This D-hexapeptide bound SRIF receptor type 2 with an affinity constant (Ki) of 172 +/- 12 nM, blocked SRIF inhibition of adenylate cyclase in vitro (IC50 = 5.1 +/- 1.4 microM), and induced GH release when given alone (50 micrograms intravenously) to anesthetized rats with or without pretreatment with a long-acting SRIF agonist.


Subject(s)
Growth Hormone/metabolism , Oligopeptides/pharmacology , Somatostatin/antagonists & inhibitors , Somatostatin/physiology , Animals , Binding, Competitive , Cell Line , Cyclic AMP/metabolism , Male , Oligopeptides/metabolism , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/physiology
3.
Endocrinology ; 139(6): 2824-30, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9607790

ABSTRACT

Two isoforms of the GH receptor, the full-length receptor (GHRL) and a short isoform (GHRS) that lacks the transmembrane and intracellular domains of GHRL, have been analyzed in rat tissue extracts by Western blotting and immunoprecipitation. Although quantitative estimates of GHRS and GHRL based on coprecipitation of [125I]GH indicated similar amounts of both isoforms in tissue extracts, the 110 kDa band corresponding to GHRL was generally not detected on Western blots without enrichment by immunoprecipitation. Two bands with electrophoretic mobilities corresponding to 38 and 42 kDa were present in extracts prepared from liver, muscle, and adipocytes. Western blots of the GH binding protein in rat serum also revealed two bands, but these had electrophoretic mobilities corresponding to 44 and 52 kDa. After digestion by endoglycosidase F, a single band with an electrophoretic mobility corresponding to 31 kDa was detected in samples from adipocytes, liver or serum, indicating that GHRS retained in tissues is glycosylated less extensively than that in rat serum. Digestion with neuraminidase indicated that the smaller glycoproteins in tissue extracts lack sialic acid residues that are present in serum samples. Furthermore, endoglycosidase H degraded GHRS in liver extracts to a 31 kDa band but did not degrade serum samples, suggesting that tissues retain a high mannose form of GHRS. The abundance of GHRS or GHRL in tissues from male, virgin female, and pregnant rats was estimated from the amount of 125I-GH that was bound to each isoform after immunoprecipitation. Liver contained more than 10 times as much GHRS per gram of tissue as fat or muscle. In liver, muscle, and fat, the amount of GHRS exceeded that of GHRL, sometimes by as much as 6-fold. GHBP levels in serum of females exceeded those in males, and rose even higher in pregnant females. The abundance of GHRS in all tissue extracts paralleled serum levels. In muscle and fat, the levels of GHRL did not differ in male, female and pregnant rats, whereas in liver, the pattern was similar to the GHRS pattern. In all tissues, pools of GHRS exceeded those of GHRL by a factor that grew larger as tissue and serum levels increased. The half life of GHBP in serum was estimated to be 2.4 h in rats treated with cycloheximide, whereas that of GHRS was 20 min in liver and 8.5 h in fat. These results suggest that GHRS is synthesized in liver 8 times faster than it is released into serum, whereas synthesis in fat is less than 30% of the rate at which it is released into serum by all tissues. Therefore, liver appears to be the major source of GHBP in serum. Although secretion into the circulatory system accounts for little or perhaps none of its turnover in some tissues, GHRS pools in tissues do appear to be regulated, suggesting that GHRS may function primarily in the cells in which it is synthesized.


Subject(s)
Receptors, Somatotropin/metabolism , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Blotting, Western , Cycloheximide/pharmacology , Female , Glycosylation , Isomerism , Liver/metabolism , Male , Muscles/metabolism , Precipitin Tests , Pregnancy , Protein Synthesis Inhibitors/pharmacology , Rats , Receptors, Somatotropin/blood , Tissue Distribution
4.
J Biol Chem ; 273(9): 5307-14, 1998 Feb 27.
Article in English | MEDLINE | ID: mdl-9478989

ABSTRACT

Signal transduction by the growth hormone receptor (GHR) occurs through growth hormone (GH)-induced dimerization of two GHRs to form a trimeric complex. It is thought that dimerization alone is sufficient for signaling, since monoclonal antibodies (mAbs) against the extracellular domain of the GHR elicit proliferation of FDC-P1 cells transfected with a chimeric receptor comprising the extracellular domain of the GHR and the fibronectin and cytoplasmic domains of the murine granulocyte colony-stimulating factor receptor. We have screened 14 GHR mAbs for proliferative activity against characterized FDC-P1 and BaF-B03 cell lines stably expressing the full-length human, rabbit, or rat GHR, or the chimeric human GHR/granulocyte colony-stimulating factor receptor, and for transactivation of the c-fos promoter and STAT activation. With the chimeric receptor, eight mAbs were able to elicit proliferation, although there was no correlation between inhibition of hormone binding and agonist activity. In contrast, no mAbs were able to act as agonists with the full-length GHR FDC-P1 cell lines, although nine competed with GH for binding. A weak proliferative response was observed in the BaF-B03 cell lines with two of the mAbs (263 and 1C9), and the addition of anti-mouse F(ab)2 resulted in increased signaling in the hGHR BaF-B03 cell line to a plateau of 28 +/- 4% of the GH maximum for mAb 263. These data could indicate considerable stringency in the ability of mAbs to correctly dimerize the full-length GHR. However, the ability of mAb 263 to stimulate a mutant hGHR altered in the F'-G' loop of domain 2 was nearly abolished, concurrent with an increased affinity of this mAb for the receptor. Since the F'-G' loop undergoes a conformational change on GH binding and is necessary for full proliferative signaling, we propose that in addition to promoting receptor dimerization, mAb 263 may induce specific changes in receptor conformation similar to GH, which are required for the biological response.


Subject(s)
Receptors, Somatotropin/agonists , Receptors, Somatotropin/immunology , Animals , Antibodies, Monoclonal/pharmacology , Cell Division , Cell Line , Dose-Response Relationship, Drug , Humans , Peptide Fragments/agonists , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Proto-Oncogene Proteins c-fos/genetics , Rabbits , Rats , Receptors, Granulocyte Colony-Stimulating Factor/agonists , Receptors, Somatotropin/chemistry , Receptors, Somatotropin/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Signal Transduction , Transcriptional Activation
5.
Comb Chem High Throughput Screen ; 1(4): 171-83, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10499123

ABSTRACT

The development of high throughput, homogeneous pharmacologic and functional assays and their implementation in screening combinatorial libraries has increased the pace of stochastic drug discovery in recent years. New, noninvasive approaches involving radiometric proximity assays, an array of fluorescence-based technologies, and reporter gene constructs in mammalian and nonmammalian systems are providing more options for the selection of specific therapeutic targets. The increasing sophistication of homogeneous assay designs has also served as a springboard to better lead validation in drug discovery initiatives. This review examines these approaches in the context of new drug discovery strategies which combine a growing repertoire of high throughput screening techniques. The utility and importance of cell-based assays vis-a-vis pharmacologic (cell-free) assays is considered with specific reference given to yeast-based functional screens and homogeneous binding methodologies used to search for somatostatin antagonists and other potential growth hormone secretagogues. Also considered is the custom tailoring of specific chemical libraries which provide yet another level of target selectivity. The advantages and shortcomings of these various technologies are discussed in light of emerging trends toward higher throughput and nanoscale formats which are pushing the limits of measurable response at the cellular and molecular level.


Subject(s)
Drug Evaluation, Preclinical/methods , Somatostatin/antagonists & inhibitors , Animals , Cell-Free System , Cells, Cultured , Humans , Peptide Library , Spectrometry, Fluorescence
6.
Proc Natl Acad Sci U S A ; 94(21): 11381-6, 1997 Oct 14.
Article in English | MEDLINE | ID: mdl-9326618

ABSTRACT

We previously have demonstrated that insulin and insulin-like growth factor-I (IGF-I) down-regulate growth hormone (GH) binding in osteoblasts by reducing the number of surface GH receptors (GHRs). The present study was undertaken to investigate the mechanism of GHR down-regulation. Treatment with 5 nM insulin or IGF-I for 18 hr significantly decreased surface GH binding to 26.4 +/- 2.9% and 23.0 +/- 2.7% of control (mean +/- SE; P < 0.05), respectively. No corresponding reductions in the mRNA level and total cellular content of GHR were found, nor was the rate of receptor internalization affected. The effects on GHR translocation were assessed by measuring the reappearance of GH binding of whole cells after trypsinization to remove the surface receptors. GH binding of control cultures significantly increased (P < 0.05) over 2 hr after trypsinization, whereas no recovery of binding activity was detected in insulin and IGF-I-treated cultures, indicating that GHR translocation was impaired. Studies on the time course of GHR down-regulation revealed that surface GH binding was reduced significantly by 3-hr treatment (P

Subject(s)
Human Growth Hormone/metabolism , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Osteoblasts/metabolism , Receptors, Somatotropin/metabolism , Androstadienes/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Line , Cell Membrane/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Homeostasis , Humans , Kinetics , Phospholipases A/antagonists & inhibitors , RNA, Messenger/biosynthesis , Rats , Receptors, Somatotropin/biosynthesis , Recombinant Proteins/metabolism , Transcription, Genetic/drug effects , Trypsin , Wortmannin
7.
Mol Cell Endocrinol ; 135(1): 1-10, 1997 Nov 30.
Article in English | MEDLINE | ID: mdl-9453235

ABSTRACT

Growth hormone receptor (GH-R) gene expression was evaluated in avian growth-plates in situ and in cultured chondrocytes. In the epiphyseal growth-plate, chondrocytes at different stages of differentiation located at the proliferative and upper hypertrophic zones express the GH-R gene. In culture, addition of ascorbic acid facilitated chondrocyte differentiation as evaluated by decrease in collagen type II gene expression and increase in alkaline phosphatase activity and osteopontin gene expression. Both the ascorbic acid-treated and untreated chondrocytes expressed the gene coding for the chicken growth hormone receptor (cGH-R), but only the undifferentiated cells were capable of binding the hormone. This reduction in GH-binding resulted in alteration in GH-dependent regulation of the GH-R gene expression: only the undifferentiated chondrocytes responded to chicken GH (cGH) by down-regulation of the cGH-R gene expression. Chondrocyte differentiation induced by either ascorbic acid or retinoic acid was associated with the appearance of two growth hormone binding-proteins (GHBPs) in the culture medium with estimated MWs of 32 and 70 kDa, respectively. These GHBPs differ in their MW from the major GHBP found in chicken plasma. Chondrocyte GHBPs specifically bind [125I]cGH, which can be displaced by an excess of unlabeled cGH. The differentiation-dependent increase in the 70 kDa GHBP was observed also using specific chicken GHBP antiserum. Our data suggest that the reduction of the differentiated chondrocytes response to GH is due to differentiation-dependent loss of the extracellular domain of the GH-R, resulting in a lack of functional receptors on the cell surface and generation of GHBP.


Subject(s)
Cell Differentiation , Growth Plate/metabolism , Receptors, Somatotropin/biosynthesis , Animals , Ascorbic Acid/pharmacology , Binding, Competitive , Cells, Cultured , Chickens , Collagen/genetics , Culture Media, Conditioned , Gene Expression , Growth Hormone/metabolism , Molecular Weight , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism
8.
Growth Regul ; 6(3): 165-75, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8894650

ABSTRACT

The presence of growth hormone (GH) receptor (GHR) gene transcripts and GH-binding sites in guinea pig liver suggests normal expression and translation of a GHR gene in these animals. Guinea pigs are, however, resistant to GH action and appear to lack the circulating GH-binding proteins (GHBPs) that result from alternate splicing of the GHR message or from cleavage of the extracellular binding domain of membrane GHRs. The paradoxical absence of circulating GHBPs in guinea pigs was therefore examined. The presence of GHR/GHBP mRNA in guinea pig liver was confirmed by Northern blotting. In addition to a 4.4 kb transcript that probably encodes a full-length receptor, an additional 1.9 kb transcript was detected that may encode a binding protein, although this transcript is larger than rat GHBP mRNA. The possibility that these transcripts may be translated into GHBPs was assessed immunologically. A 46 kDa protein, identical in size to rat GHBP, was specifically detected in guinea pig liver by a monoclonal antibody (MAb 4.3) raised against the hydrophilic tail of rat GHBP. A single protein of approximately 48 kDa was also detected by MAb 4.3 in proteins precipitated from guinea pig serum by a polyclonal antibody raised against the rat GHBP. This protein was slightly larger than the two proteins (46 kDa and 40 kDa) in rat serum labelled by the same method. The presence of a putative GHBP in guinea pig serum was also supported by the cross-reactivity of guinea pig serum with a monoclonal antibody (MAb 263) raised against rat GHBP. The binding of radioiodinated hGHBP to this antibody was inhibited, in a dose-related way and parallel to that of the standard, by serial dilutions of guinea pig serum, indicating immunoreactive GHBP concentrations > 500 ng/ml. Immunoreactive GHBP concentrations in other mammalian serum (from rats, rabbits, pigs, cattle, horses, goats, dogs and humans) were, in contrast, < 30 ng/ml. Guinea pig sera similarly cross-reacted, but to a lesser degree, in other radioimmunoassays for rGHBP, in which p(Ab)1 or MAb 4.3 were used as the primary antibodies. Nevertheless, despite these immunological findings, hGH binding activity could not be detected in guinea pig serum using a number of different radioligand binding assays. These results suggest the novel presence of abundant, but possibly defective, GHBP-like proteins in guinea pig serum. The immunological detection of the hydrophilic sequence of rat GHBP in guinea pig hepatic and serum proteins also suggests that GHBPs in this species arise from the truncated GHR gene transcript identified in guinea pig liver.


Subject(s)
Carrier Proteins/biosynthesis , Growth Hormone/pharmacology , Liver/metabolism , Receptors, Somatotropin/biosynthesis , Animals , Blotting, Northern , Carrier Proteins/metabolism , Cattle , Cross Reactions , Dogs , Drug Resistance , Female , Goats , Growth Hormone/metabolism , Guinea Pigs , Horses , Humans , Male , Rabbits , Radioimmunoassay , Radioligand Assay , Rats , Receptors, Somatotropin/metabolism , Sheep , Species Specificity , Swine , Transcription, Genetic
9.
Biochem Biophys Res Commun ; 221(1): 133-9, 1996 Apr 05.
Article in English | MEDLINE | ID: mdl-8660323

ABSTRACT

The receptor for growth hormone-releasing hormone (GHRH) has been purified from bovine pituitary tissue and HEK293 cells transfected with human or porcine receptor using a retrievable biotinylated GHRH analog. Custom synthesized [His1, Nle27, Biotin-Lys41]-human GHRH-(1-41)-NH2 (GHRHb) bound to pituitary membranes with affinity comparable to human GHRH. GHRHb which has the biotinyl group on the C-terminus of the peptide allowed simultaneous binding to both the receptor and streptavidin agarose. This analog was used directly in the purification of the receptor from pituitary tissue or was modified by incorporation of the photoaffinity group ANBNOS (GHRHlambdab), radioiodinated and used to demonstrate purification of the GHRH receptor from transfected HEK293 cell membranes. Membranes were prepared and prebound with the respective ligand followed by CHAPS-solubilization and application of the solubilized complex to a streptavidin agarose column. Analysis of eluates from the pituitary tissue purification by silver stained SDS PAGE or of autoradiographs of gels from HEK293 eluates revealed specific bands of 52 and 55 kDa, respectively. The higher size of the latter band is expected for the ligand-crosslinked receptor. Both bands displayed similar mobility shifts of 10 kDa upon treatment with N-glycosidase, a method previously used to characterize this receptor. A 45 kDa band corresponding to the size of the Gs alpha subunit was also detected in eluates of the silver stained gels, suggesting that the GHRH receptor was retrieved as a heterotrimeric complex. Fold purification and yield for this procedure were estimated to be greater than 50,000 and 2.6-9%, respectively.


Subject(s)
Receptors, Neuropeptide/isolation & purification , Receptors, Pituitary Hormone-Regulating Hormone/isolation & purification , Animals , Autoradiography , Cattle , Cell Line , Chromatography, Liquid , Electrophoresis, Polyacrylamide Gel , Humans , Ligands , Pituitary Gland/metabolism , Protein Binding , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Swine
10.
Endocrinology ; 136(10): 4701-4, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7664692

ABSTRACT

A binding assay for growth hormone releasing factor (GRF) has been developed using scintillation proximity assay (SPA) technology. Binding conditions were validated by several criteria. Equilibrium binding was attained within three hours at 22 degrees C in crude membrane fractions of HEK293 (293-P2) and GH4C1 (GH4-P1) cells transfected with the porcine GRF receptor. Saturation binding isotherms produced a KD of 296 pM and a Bmax of 4.7 pmols/mg membrane protein in 293-P2 cells. Cells not expressing the GRF receptor displayed no specific binding for the ligand. Competition binding curves produced the following rank order of potency for tested peptides: GRF analogs D-Ala2 = D-Arg2 (IC50 approximately 1 nM) >> PACAP > secretin, VIP (EC50 > 100 nM). Somatostatin (SRIF) binding was also adapted to the SPA format in a GH4C1 cell line transfected with the SRIF receptor subtype 2 (SSTR2) and in HEK293 cells transfected with the SRIF receptor subtype 5 (SSTR5). This assay represents a major improvement for binding measurements of these and potentially many other ligands for G-protein linked receptors, requiring no separation of bound from free hormone, allowing detailed pharmacological evaluations and enabling measurement of equilibrium binding in real time. In the 96-well format, it is suitable for high throughput screening.


Subject(s)
Growth Hormone-Releasing Hormone/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Animals , Cell Line , Humans , Rats , Somatostatin/metabolism , Swine
11.
J Endocrinol ; 146(3): 449-58, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7595140

ABSTRACT

Thyroid hormones inhibit the synthesis and release of GH in avian species. This may represent a feedback mechanism, since GH enhances the peripheral production of tri-iodothyronine (T3). The possibility that GH may also have direct effects on thyroidal function was therefore investigated. The basal and thyrotrophin-induced release of thyroxine (T4) from incubated chicken thyroid glands was not enhanced, however, in the presence of chicken GH. Contrarily, GH impaired T4 release in a dose-related way. These actions were probably mediated by specific receptors, since binding sites for radiolabelled GH were demonstrated on the plasma membranes of chicken thyroid glands. Expression of the GH receptor gene in these tissues was also demonstrated using a cRNA probe for the rabbit liver GH receptor, which specifically hybridized with RNA moieties of 4.4 kb, 2.7 kb and 1.0 kb. Moreover, reverse transcription of thyroidal RNA and its amplification in the presence of 3'- and 5'-oligonucleotide primers coding for the extracellular or intracellular domains of the GH receptor generated electrophoretically separable fragments of 500 bp and 800 bp respectively, as would be expected from analysis of the hepatic GH receptor cDNA sequence. Digestion of the 500 bp fragment with NcoI or EcoRI also produced moieties of expected size (350 bp and 150 bp or 325 bp and 175 bp respectively), as did BamHI or HaeIII digestion of the 800 bp fragment (yielding fragments of 550 bp and 275 bp or 469 bp and 337 bp respectively). Translation of the GH receptor mRNA was also indicated by the immunocytochemical demonstration of GH receptors in thyroid follicular and parafollicular cells, using a specific polyclonal antibody raised against the chicken GH-binding protein. These results therefore provide evidence, for the first time, of GH receptor gene expression in thyroid tissue and the translation of functional GH receptors in thyroid glands. These results also demonstrate differential effects of GH on the extracellular concentrations of T3 and T4, which may permit subtle regulation within the somatotroph-thyroid axis.


Subject(s)
Growth Hormone/physiology , Thyroid Gland/physiology , Thyroxine/metabolism , Animals , Base Sequence , Blotting, Northern , Cell Membrane/metabolism , Chickens , Depression, Chemical , Dose-Response Relationship, Drug , Gene Expression , Molecular Sequence Data , Oligonucleotide Probes/genetics , Polymerase Chain Reaction , Radioimmunoassay , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyrotropin/pharmacology
12.
Mol Cell Endocrinol ; 113(1): 11-7, 1995 Aug 30.
Article in English | MEDLINE | ID: mdl-8674809

ABSTRACT

At least two classes of mRNA for the GH receptor (GHR) and GH binding protein (GH BP) with different 5' untranslated first exons exist in the rat. One such class, the GHR1 is predominantly expressed in the liver of female rats. The hepatic expression of the GHR1 mRNA in normal and hypophsectomized rats of both sexes was studied by employing an RNase protection/solution hybridization assay. Normal females expressed 10-fold more GHR1 mRNA than males, hypophysectomy of female rats decreased the GHR1 level to that observed in male rats. Continuous GH treatment of hypophysectomized male and female rats for 6 days increased the expression of GHR1 mRNA to levels found in normal females, whereas intermittent GH treatment without effect. Bovine GH(bGH) induced the GHR1 expression in a time- and dose-dependent manner in primary cultures of adult rat hepatocytes as determined by solution hybridization. Maximal induction was achieved after 72 h of treatment with 50 ng bGH/ml medium. Female enriched expression of receptor and binding protein mRNAs raises the possibility that they participate in determining the ability of the liver to respond differently to the male and female GH secretory patterns. Our in vitro model utilizing cultures of primary adult rat hepatocytes could be used to address this issue as well as explore a hormonal interplay in regulation of GHR1 expression.


Subject(s)
Carrier Proteins/genetics , Liver/metabolism , RNA, Messenger/metabolism , Receptors, Somatotropin/genetics , Sex Characteristics , Animals , Blotting, Northern , Cells, Cultured , Female , Gene Expression , Growth Hormone/pharmacology , Hypophysectomy , Kinetics , Male , Nucleic Acid Hybridization , Rats , Rats, Sprague-Dawley , Ribonucleases
13.
Endocrinology ; 136(2): 379-85, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7835268

ABSTRACT

We have developed a ligand immunofunctional assay (LIFA) for quantifying the circulating functional GH-binding protein (GHBP) in the rat. This two-site solid-phase assay uses a capture monoclonal antibody (4.3) specific to the hydrophilic C-terminal segment of rat GHBP (rGHBP), saturation of binding with human GH, and a detection system of rabbit antihuman GH polyclonal antibody and peroxidase-conjugated antirabbit immunoglobulin G antibody. Results were compared with Scatchard estimates derived by immuno-precipitation with monoclonal antibody 4.3. This assay was used to determine the GHBP levels in male and female rats and to investigate the diurnal properties and dynamics of GH and GHBP interaction in 15-min blood sampling over a 6-h period. The dynamic range of the rLIFA was 0.15-20.0 nM recombinant rGHBP, with intraassay and interassay coefficients of variation of 10.5% (n = 20) and 12.9% (n = 12), respectively. Serum GHBP levels determined by the rLIFA and those derived from Scatchard estimates were strongly correlated (n = 8; beta = 0.55; r2 = 0.89; P = 0.0005). Male rats had lower GHBP levels (6.5 +/- 0.7 nM; mean +/- SE; n = 14) than female rats (35.4 +/- 2.7 nM; n = 15; P = 0.0001). In the diurnal study, male rats had higher GH peaks (312.5 +/- 121.6 ng/ml; n = 7) than female rats (96.5 +/- 15.4 ng/ml; n = 9; P < 0.0001). In contrast to the pulsatile secretion of GH, GHBP levels in both sexes remained stable and showed no relationship to secretory pulses of GH. However, the GH bursts significantly altered the distribution of the GH-GHBP complex in male rats. By saturation and mass analysis, the greater GH pulsatile secretion in male rats resulted in occupancy of GHBP from less than 5% at nadir to about 80% at secretory peaks, in contrast to the less than 5-15% range of GHBP occupancy in female rats. In male rats, greater than 80% of GH at secretory peaks existed in the free form, whereas in female rats, 16-23% of GH existed in the free form during pulsatile secretion. In summary, the rLIFA shows good correlation to Scatchard analysis using an identical antibody. We conclude that this assay provides a rapid, sensitive, and accurate measurement of the circulating functional GHBP in the rat, and that it facilitates the study of GH and GHBP dynamics under a range of physiological conditions.


Subject(s)
Carrier Proteins/blood , Growth Hormone/blood , Immunoenzyme Techniques , Animals , Antibodies, Monoclonal , Carrier Proteins/immunology , Circadian Rhythm , Female , Male , Precipitin Tests , Rats , Rats, Sprague-Dawley , Rats, Wistar , Recombinant Proteins/immunology , Sex Factors
14.
Endocrinology ; 136(2): 749-60, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7835307

ABSTRACT

In the rat, alternatively spliced messenger RNA (mRNA) species encode GH receptor (GHR) and GH-binding protein (GHBP). Additionally, these mRNAs are alternatively spliced in the 5'-untranslated region, resulting in at least two classes of GHR and GHBP mRNA with distinct first exons and identical coding regions. These alternative first exons define two unique classes of GHR and GHBP mRNA (called GHR1 and GHR2). The GHR1 class of RNA is expressed only in the liver, is far more abundant in females than males, and is particularly abundant during pregnancy. GHR1 RNA is induced later in development than is GHR2. Additional classes of GHR and GHBP RNA may also exist. The genomic structure of the GHR1 first exon reveals a putative promotor region with no TATA box, CAAT box, or other sequence elements suggesting specific responses. An in vivo approach was used to investigate the regulation of GHR1 expression. In female rats, gonadectomy was found to reduce the percentage of steady state GHR1 RNA levels in the liver, whereas male castration resulted in an induction of GHR1 RNA. However, short-term treatment with estrogen or testosterone had little effect, suggesting that direct regulation of GHR1 expression may occur through effector(s) other than gonadal steroids. Hypophysectomy abolished GHR1 RNA in females. Treatment of hypophysectomized females and castrated males with GH by single injection did not significantly induce GHR1 RNA, but treatment by continuous infusion of GH did. Little change in non-GHR1 RNA levels was observed for each of these treatments. The results suggest that: 1) the sexual dimorphism observed in total GHR and GHBP RNA in rat liver is attributable to the sexually dimorphic expression of the GHR1 class of RNA; 2) the sexually dimorphic pattern of GH release in rats regulates the GHR1 class of RNA; 3) changes in GHR and GHBP expression observed on gonadectomy, hypophysectomy, GH treatment, and pregnancy are best attributed to GHR1 regulation; and 4) since GHR1 is liver specific, the observed increases in serum GHBP concentration in response to sex steroids, GH pattern, and pregnancy are likely to originate from the liver.


Subject(s)
Carrier Proteins/metabolism , Liver/metabolism , RNA, Messenger/analysis , Receptors, Somatotropin/metabolism , Alternative Splicing , Animals , Base Sequence , Carrier Proteins/genetics , DNA Probes , Exons , Female , Growth Hormone/metabolism , Growth Hormone/pharmacology , Lactation/metabolism , Male , Molecular Sequence Data , Organ Specificity , Pregnancy , RNA, Messenger/genetics , Rats , Receptors, Somatotropin/genetics , Sex Characteristics
15.
Growth Regul ; 4(4): 147-54, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7756972

ABSTRACT

A radioimmunoassay (RIA) for the rat growth hormone binding protein (GHBP) was developed using a synthetic peptide (corresponding to the hydrophilic carboxyl-terminal sequence of mouse GHBP) as standard and a monoclonal antibody (MAb 4.3) reactive with this peptide as the primary antibody. The values for GHBP concentration obtained for normal rats using this assay compare favourably with those obtained by gel filtration and ELISA methods. The concentration of GHBP in normal male rats at 11 weeks of age (680 +/- 30 ng/ml, SEM, n = 9) was significantly less than the concentration in normal females (943 +/- 47 ng/ml, SEM, n = 25). In 11-week-old dwarf male rats the concentration of GHBP was 423 +/- 35 ng/ml (n = 8); less than in dwarf females (542 +/- 32, P < 0.05, n = 9) and normal males (680 +/- 30, P < 0.001, n = 9). The GHBP concentration in dwarf rats was not age-dependent, whereas in normal females the concentration of GHBP increased with age. The availability of an RIA which is not susceptible to interference by endogenous GH, will facilitate further studies on hormonal and nutritional regulation of the rat GHBP. The assay was applied to studying the effects of IGF-I infusion (240 micrograms/day for 1 week) and GH injection (65 micrograms/100 g body weight, twice daily for 1 week and 4 weeks) on the serum concentration of GHBP in 11-week-old Lewis dwarf rats. Hepatic GH binding sites were also measured in desaturated membranes from the same animals.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Carrier Proteins/metabolism , Dwarfism/metabolism , Growth Hormone/pharmacology , Insulin-Like Growth Factor I/pharmacology , Liver/metabolism , Receptors, Somatotropin/metabolism , Aging/metabolism , Animals , Binding Sites , Chromatography, Gel , Dwarfism/blood , Eating/physiology , Female , Liver/drug effects , Male , Radioimmunoassay , Rats , Rats, Inbred Lew , Rats, Wistar , Receptors, Somatotropin/drug effects , Sex Characteristics , Weight Gain/physiology
16.
Proc Soc Exp Biol Med ; 206(3): 195-9, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8016153

ABSTRACT

Growth hormone receptor (GHR) expression has been analyzed at the RNA level. In the rat, relative expression of the RNA species encoding the GHR and the GH-binding protein (GHBP) appears to be sensitive to endocrine status. Full-length GHR cDNA clones from ovine, porcine, and chicken were used as probes to investigate the existence of unique RNAs for GHBPs in these species. In the sheep and pig, only a single, approximately 4.5-kb RNA is apparent. Although quite high levels of GH binding activity are found in pig serum, a variety of methods failed to isolate a separate GHBP message, suggesting that porcine GHBP is produced via a mechanism different from that which is known for rat. One class of chicken GHR cDNA, resulting from alternative use of a splice acceptor 17 bases upstream of the intron 6/Exon 7 junction, is also presented.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation , Receptors, Somatotropin/genetics , Animals , Base Sequence , Chickens , Cloning, Molecular , DNA Probes , DNA, Complementary/genetics , Liver/metabolism , Molecular Sequence Data , RNA, Messenger/metabolism , Rats , Sheep , Swine
17.
J Endocrinol ; 139(3): 495-501, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7510771

ABSTRACT

The properties of four independent lines of monoclonal antibodies (MAbs) specific to rat GH-binding protein (GHBP) were examined. Three MAbs, designated GHR-12, GHR-13 and GHR-16, were raised against the entire GHBP molecule. The fourth MAb, designated as GHBP4.3, was raised against the 17 amino acid residues at the C-terminal end of rat GHBP. The interaction of these antibodies with GHBP and their effect on GH binding to GHBP were analysed by conventional competition binding assays and surface plasmon resonance, i.e. with a Biospecific Interaction Analysis (BIAcore) instrument. The binding affinity of these MAbs to GHBP ranged from 29 nmol/l to 30.9 pmol/l. The pair-wise antibody binding to GHBP on BIAcore suggested that GHR-13 and GHR-16 recognized different antigenic determinants while part of the GHR-12 epitope might be shared with the other antibodies. The antibodies inhibited the interaction of GH with GHBP in the competition binding assay. However, in sequential binding on the BIAcore instrument, they were able to bind GHBP after its interaction with GH, indicating that the inhibition observed in the competition binding assay resulted from steric hindrance rather than direct interference with the GH-binding site of GHBP. The present findings, therefore, suggest that these antibodies are useful for investigating GHBP and its interaction with GH.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigen-Antibody Complex/metabolism , Carrier Proteins/metabolism , Growth Hormone/metabolism , Animals , Binding, Competitive , Carrier Proteins/immunology , Epitopes/metabolism , Rats
18.
Mol Endocrinol ; 7(11): 1379-90, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8114753

ABSTRACT

A study of chicken GH receptor (cGHR) expression has revealed that the two major liver and skeletal muscle transcripts of the cGHR are developmentally expressed. Expression of the larger (4.7 kilobases) transcript increases with age. The smaller transcript (0.7 kilobases) is a truncation product, resulting from alternative usage of a functional polyadenylation [poly(A)] signal embedded in the coding sequence. The extent to which alternative cleavage and polyadenylation occur displays some tissue and sex specificity. Cleavage and polyadenylation occur down-stream of the AATAAA portion of the poly(A) signal (cGHR positions 304-309) and up-stream of a GT-rich sequence. The truncated transcript appears to be translated, based on its association in vivo with polyribosomes, although the physiological role of the putative protein product of this truncated transcript is as yet unknown. Three other avian species (quail, turkey, and duck) also show a polyadenylated truncation of the GHR message due to a poly(A) signal at the same location in the coding sequence. In cell culture expression, mutation of AATAAA to AACAAG prevents production of the truncated transcript. In a chimeric construct, the signal and neighboring sequence from the cGHR are sufficient to confer cleavage and polyadenylation upon the rat GHR, a gene that otherwise lacks the internal poly(A) signal. Alternative polyadenylation within the coding region of a structural gene is discussed as a heretofore unknown means of post-transcriptional regulation of a gene product.


Subject(s)
Chickens/genetics , Genes , Poly A/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/genetics , Receptors, Somatotropin/genetics , Regulatory Sequences, Nucleic Acid , Age Factors , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Female , Gene Expression Regulation , Humans , Liver/metabolism , Male , Molecular Sequence Data , Muscle Proteins/genetics , Muscles/metabolism , Organ Specificity , Poultry/genetics , Rats/genetics , Recombinant Fusion Proteins/genetics , Sequence Alignment , Sequence Homology, Nucleic Acid , Species Specificity , Transcription, Genetic , Transfection
19.
Endocrinology ; 133(3): 1125-30, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8396011

ABSTRACT

GH receptor immunoreactivity is widely distributed within the rat pituitary gland, although apart from somatotrophs the cell types with GH receptor immunoreactivity have yet to be identified. It is also unknown whether this immunoreactivity reflects the presence of GH binding proteins (GHBPs) or authentic receptors. The possible colocalization of GHBPs and pituitary hormones in somatotrophs, lactotrophs, gonadotrophs, thyrotrophs, and corticotrophs was therefore examined using immunogold electron microscopy. Pituitary sections were indirectly immunostained with antibodies for GH, PRL, LH, FSH, TSH, or ACTH using gold-labeled immunoglobulin G. The same grids were also immunostained with a polyclonal antibody raised against rat GHBP using protein A conjugated to gold particles of a different size. Some sections were gold-labeled using a monoclonal antibody (MAb 4.3) raised against the unique hydrophobic tail of the GHBP, using gold-labeled immunoglobulin G. The cellular and ultrastructural distribution of immunoreactivity within the pituitary gland was similar after labeling with either GHBP antiserum. In all immunoreactive cells the labeling was most intense in secretory granules. Specific staining was not, however, demonstrated in the nucleus, in contrast with earlier findings using MAb 263. Moreover, while staining with MAb 263 appeared to be ubiquitous, some pituitary cells were not labeled by the polyclonal GHBP antiserum or by MAb 4.3. GHBP immunoreactivity was, however, colocalized with hormones in all pituitary cell-types, although in some cells GHBP immunoreactivity was not present in all secretory granules. These results clearly demonstrate the presence of GHBPs in adenohypophyseal cells, in which they appear to be stored or secreted with GH, PRL, LH, FSH, TSH, and ACTH. The function of GHBPs in the pituitary gland is, however, uncertain.


Subject(s)
Carrier Proteins/analysis , Pituitary Gland, Anterior/ultrastructure , Pituitary Hormones/analysis , Adrenocorticotropic Hormone/analysis , Animals , Antibodies, Monoclonal , Cytoplasmic Granules/chemistry , Follicle Stimulating Hormone/analysis , Growth Hormone/analysis , Immunohistochemistry , Luteinizing Hormone/analysis , Microscopy, Immunoelectron , Pituitary Gland, Anterior/chemistry , Prolactin/analysis , Rats , Thyrotropin/analysis
20.
Endocrinology ; 132(3): 1071-7, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8440172

ABSTRACT

While circulating GH concentrations are high in fetal life, skeletal growth is only slightly reduced by GH deficiency in utero. This has been explained by the relatively low binding of GH to fetal hepatic tissue, suggesting a lack of GH receptors (GHR). The GHR also recognizes ovine placental lactogen (oPL), which may have a specific role either as a fetal growth-promoting hormone or in regulating fetal metabolism. We investigated GHR expression and membrane binding of ovine (o) GH and oPL in various ovine fetal tissues and in maternal liver at different gestational stages. Singleton-bearing ewes at 51, 95, and 120 days gestation were killed. Liver, muscle, kidney, and brain samples were taken from the fetuses as well as placentas and livers from the ewes (n = 3/gestational age). GHR mRNA measured by Northern blot analysis was expressed at high levels in maternal liver at all gestational stages. A major band was observed at 4.4 kilobases (kb), and three minor bands were observed at 2.5, 1.7, and 8.1 kb. In fetal and placental tissue, only the 4.4-kb band was detected. This was present as early as day 51 of gestation in liver, kidney, lung, heart, and placenta and increased slightly with advancing gestation. On day 51, the expression of GHR mRNA in muscle was negligible, but by day 95, muscle expressed higher concentrations than fetal liver. Placental samples showed only a slight signal, with no change over the gestational range studied. In situ hybridization revealed the placental mRNA to be primarily associated with the decidua. Hepatic tissue showed specific binding to [125I]oGH and [125I]oPL from 51 days gestation. [125I]oPL showed a higher [51 days, 17.9 +/- 1.9% (mean +/- SEM); 95 days, 11.5 +/- 1.6%; 120 days, 16.3 +/- 0.9%] specific binding to the liver membranes than [125I]oGH (51 days, 2.1 +/- 0.7%; 95 days, 2.6 +/- 0.3%; 120 days, 3.5 +/- 0.4%). We conclude that oGHR are present as early as day 51 of gestation in various tissues, including liver. The message appears later in skeletal muscle than in liver. As the GH receptor binds oPL with higher potency than oGH, the parallel ontogenic changes in [125]oGH and [125]oPL binding in the liver do not support the presence of a PL receptor under independent developmental regulation.


Subject(s)
Liver/metabolism , RNA, Messenger/metabolism , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Animals , Blotting, Northern , Cell Membrane , Cloning, Molecular , Female , Fetus , Gene Library , Gestational Age , Growth Hormone/metabolism , In Situ Hybridization , Ligands , Liver/embryology , Muscles/embryology , Muscles/metabolism , Organ Specificity , Placenta/metabolism , Pregnancy , Prolactin/metabolism , Receptors, Prolactin/metabolism , Receptors, Somatotropin/biosynthesis , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...