Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters

Publication year range
1.
Pol J Pathol ; 69(1): 48-52, 2018.
Article in English | MEDLINE | ID: mdl-29895126

ABSTRACT

Encouraged by our previous finding of growth hormone-releasing hormone receptor (GHRH-R) expression in metaplastic and neoplastic apocrine breast epithelium, we examined a small series of skin adnexal tumours with various degrees of apocrine (oxyphilic) differentiation, as well as normal axillary and anogenital apocrine sweat glands, for the expression of GHRH-R. Sections of formalin-fixed paraffin-embedded tissue blocks were immunostained for gross cystic disease fluid protein-15 (GCDFP-15) and androgen receptor (AR), to prove apocrine differentiation and correlate it with areas of GHRH-R expression. All but one of 19 tumours with apocrine epithelium and all five benign apocrine glands stained with both anti-GHRH-R antibodies used, each labelling a different domain of the receptor. Non-apocrine areas of the tumours and four eccrine tumours without oxyphilic features did not stain, but most sebaceous glands and some eccrine glands were labelled. Our data suggest that anti-GHRH-R antibodies highlight apocrine differentiation at extramammary sites also. Although GHRH-R seems to have a sensitivity comparable to classic apocrine markers such as AR and GCDFP-15, it seems to be inferior in specificity. The GHRH-R labelling of apocrine glands and neoplastic epithelium might also interfere with the emerging anti-GHRH targeted treatment of some malignancies acting on these receptors.


Subject(s)
Apocrine Glands/chemistry , Biomarkers, Tumor/analysis , Epithelial Cells/chemistry , Neoplasms, Adnexal and Skin Appendage/chemistry , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Sweat Gland Neoplasms/chemistry , Apocrine Glands/pathology , Biopsy , Carrier Proteins/analysis , Diagnosis, Differential , Epithelial Cells/pathology , Glycoproteins/analysis , Humans , Immunohistochemistry , Membrane Transport Proteins , Neoplasms, Adnexal and Skin Appendage/pathology , Predictive Value of Tests , Receptors, Androgen/analysis , Sweat Gland Neoplasms/pathology
2.
Int J Gynecol Pathol ; 35(2): 142-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26535981

ABSTRACT

Malignant mixed Müllerian tumors (MMMTs) are aggressive malignant neoplasms with a high recurrence rate and poor prognosis. Despite advances in adjuvant therapies in recent years, the prognosis of these tumors has not improved. Growth hormone-releasing hormone (GHRH) is produced by a variety of malignant tumors and acts as a growth factor in an autocrine/paracrine manner. Its function requires the presence of its receptors to exert its effects on neoplastic cells. In this study, we evaluated the expression of GHRH receptors (GHRH-R) in a group of MMMTs. Thirty-one examples of MMMTs from endometrium, ovary, uterine tube, and pelvic peritoneum were retrieved from the files of Department of Pathology at the University of Miami, Jackson Memorial Hospital. Immunohistochemistry for GHRH-R was performed on paraffin sections and the staining results were evaluated separately in both epithelial and mesenchymal components of each tumor. The presence of pituitary type growth hormone-releasing hormone receptor mRNA and that of its biologically active splice variant were also evaluated by RT-PCR in 6 of the tumors. Positive immunohistochemical reaction for GHRH-R was detected in 30 tumors (96%). The epithelial and sarcomatous components were positive in 30 (96%), whereas one endometrial tumor was negative in both components. The mRNA for GHRH-R and its splice variant was found in all 6 tested tumors. This study shows that GHRH-R is expressed by the majority of MMMTs in both epithelial and mesenchymal components. This finding could potentially serve as a basis for therapeutic approaches using synthetic peptide antagonists of GHRH-R that have shown significant efficacy with minimal side effects in experimental models.


Subject(s)
Biomarkers, Tumor/analysis , Mixed Tumor, Mullerian/metabolism , Mixed Tumor, Mullerian/pathology , Receptors, Neuropeptide/biosynthesis , Receptors, Pituitary Hormone-Regulating Hormone/biosynthesis , Adult , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry , Middle Aged , Polymerase Chain Reaction , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis
3.
Int J Oncol ; 33(1): 137-43, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18575759

ABSTRACT

Recent evidence indicates that growth hormone-releasing hormone (GHRH) functions as a growth factor for gastrointestinal (GI) tumours. The tumourigenic effects of GHRH appear to be mediated by the splice variant 1 (SV-1) of GHRH receptor as well as the full length pituitary type receptor for GHRH (GHRH-R). We examined the protein and mRNA expression of GHRH-R and SV-1 in normal human tissues and tumours of the gastrointestinal (GI-) tract by immunohistochemical staining and reverse transcriptase (RT)-PCR. Squamous cells and squamous cell carcinoma of the oesophagus were negative for GHRH-R and SV-1, while Barrett's mucosa and adenocarcinomas of the oesophagus showed a strong expression of both receptors. The expression of GHRH-R was absent in normal colonic mucosa other than neuroendocrine cells (NE) and lining epithelium (LE) but strong in tubular adenomas of the colon, while the staining for SV-1 was absent in cells other than NE. However, the expression of both receptors was significantly increased in tubulovillous adenomas and colorectal cancers. No differences were seen in protein levels for both receptors between normal and neoplastic tissues of the stomach, pancreas and liver. Because of low mRNA levels for both receptors in all samples tested, only a qualitative assessment could be made. However, mRNA for GHRH-R and SV-1 showed a near-perfect correlation with the assessment of receptor proteins by immunostaining. Our study shows that in contrast to normal mucosa, transformed mucosa of the oesophagus and the colon expresses GHRH-R and SV-1. This aberrant expression of GHRH-R and SV-1 in oesophageal and colorectal malignancies may provide a molecular target for a therapeutic approach based on GHRH antagonists.


Subject(s)
Colon/chemistry , Colonic Neoplasms/chemistry , Esophageal Neoplasms/chemistry , Esophagus/chemistry , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Colonic Neoplasms/drug therapy , Cyclin D1/physiology , Esophageal Neoplasms/drug therapy , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Humans , Immunohistochemistry , Intestinal Mucosa/chemistry , RNA Splicing , RNA, Messenger/analysis , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/chemistry , Receptors, Pituitary Hormone-Regulating Hormone/genetics
4.
Hum Pathol ; 64: 164-170, 2017 06.
Article in English | MEDLINE | ID: mdl-28438614

ABSTRACT

Apocrine breast carcinomas were evaluated for the expression of components of the growth hormone-releasing hormone (GHRH) autocrine/paracrine pathway: GHRH and its receptors (GHRH-R), as mammary apocrine carcinomas and epithelium seemed to be uniformly positive for GHRH-R in a pilot study. The apocrine phenotype was determined on the basis of hematoxylin-eosin morphology and a congruent immunohistochemical profile (estrogen receptor negativity, androgen receptor and gross cystic disease fluid protein-15 positivity). Thirty-five formalin-fixed, paraffin-embedded apocrine breast cancers in tissue microarrays and 24 cases using whole-tissue sections were evaluated for GHRH-R and GHRH expression by immunohistochemistry using polyclonal antibodies raised against various domains of GHRH-R and one polyclonal antibody specific for GHRH. GHRH-R positivity was detected in the overwhelming majority (ranging from 90% to 100%) of apocrine breast carcinomas with all but one of the antibodies applied. The expression was usually diffuse with only isolated cases showing positivity in less than 50% of tumor cells. With the PA5-33583 antibody, GHRH-R positivity was seen only in 73% of the cases in at least 50% of the tumor cells. GHRH expression was also present in all but one case tested, with more than 50% of the cells expressing it in 30/34 cases. These results support a high rate of GHRH-R and GHRH expression in apocrine breast carcinomas. Whether these findings can be exploited for the targeted treatment of apocrine breast carcinomas with GHRH antagonists requires further study.


Subject(s)
Apocrine Glands/chemistry , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Carcinoma/chemistry , Growth Hormone-Releasing Hormone/analysis , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Apocrine Glands/pathology , Biopsy , Breast Neoplasms/pathology , Carcinoma/pathology , Cell Differentiation , Female , Humans , Immunohistochemistry , Retrospective Studies , Tissue Array Analysis , Up-Regulation
5.
J Endocrinol ; 190(3): 611-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17003262

ABSTRACT

Recently, the importance of the dopaminergic D2 receptor (D2R) subtype in normal body growth and neonatal GH secretion has been highlighted. Disruption of D2R alters the GHRH-GH-IGF-I axis and impairs body growth in adult male mice. The D2R knockout (KO) dwarf mouse has not been well characterized; we therefore sought to determine somatotrope function in the adult pituitary. Using immunohistochemistry and confocal microscopy, we found a significant decrease in the somatotrope population in pituitaries from KO mice (P=0.043), which was paralleled by a decreased GH output from pituitary cells cultured in vitro. In cells from adult mice the response amplitude to GHRH differed between genotypes (lower in KO), but this difference was less dramatic after taking into account the lower basal release and hormone content in the KO cells. Furthermore, there were no significant differences in cAMP generation in response to GHRH between genotypes. By Western blot, GHRH-receptor in pituitary membranes from KO mice was reduced to 46% of the level found in wildtype (WT) mice (P=0.016). Somatostatin induced a concentration-dependent decrease in GH and prolactin (PRL) secretion in both genotypes, and 1x10(-7) M ghrelin released GH in cells from both genotypes (P=0.017) in a proportionate manner to basal levels. These results suggest that KO somatotropes maintain a regulated secretory function. Finally, we tested the direct effect of dopamine on GH and PRL secretion in cells from both genotypes at 20 days and 6 months of life. As expected, we found that dopamine could reduce PRL levels at both ages in WT mice but not in KO mice, but there was no consistent effect of the neurotransmitter on GH release in either genotype at the ages studied. The present study demonstrates that in the adult male D2R KO mouse, there is a reduction in pituitary GH content and secretory activity. Our results point to an involvement of D2R signaling at the hypothalamic level as dopamine did not release GH acting at the pituitary level either in 1-month-old or adult mice. The similarity of the pituitary defect in the D2R KO mouse to that of GHRH-deficient models suggests a probable mechanism. A loss of dopamine signaling via hypothalamic D2Rs at a critical age causes the reduced release of GHRH from hypophyseotropic neurons leading to inadequate clonal expansion of the somatotrope population. Our data also reveal that somatotrope cell number is much more sensitive to changes in neonatal GHRH input than their capacity to develop properly regulated GH-secretory function.


Subject(s)
Dwarfism/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Growth Hormone/metabolism , Pituitary Gland/metabolism , Receptors, Dopamine D2/genetics , Somatostatin/pharmacology , Animals , Blotting, Western/methods , Cells, Cultured , Cyclic AMP/analysis , Cyclic AMP/biosynthesis , Dopamine/pharmacology , Dose-Response Relationship, Drug , Ghrelin , Immunohistochemistry/methods , Male , Mice , Mice, Knockout , Microscopy, Confocal , Peptide Hormones/pharmacology , Pituitary Gland/cytology , Pituitary Gland/drug effects , Prolactin/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Neuropeptide/analysis , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Receptors, Pituitary Hormone-Regulating Hormone/metabolism
6.
Mol Endocrinol ; 12(5): 750-65, 1998 May.
Article in English | MEDLINE | ID: mdl-9605937

ABSTRACT

The hypothalamic peptide GH-releasing hormone (GHRH) stimulates the release of GH from the pituitary through binding and activation of the GHRH receptor, which belongs to the family of G protein-coupled receptors. The objective of this study was to identify regions of the receptor critical for interaction with the ligand by expressing and analyzing truncated and chimeric epitope-tagged GHRH receptors. Two truncated receptors, GHRHdeltaN, in which part of the N-terminal domain between the putative signal sequence and the first transmembrane domain was deleted, and GHRHdeltaC, which was truncated downstream of the first intracellular loop, were generated. Both the receptors were deficient in ligand binding, indicating that neither the N-terminal extracellular domain (N terminus) nor the membrane-spanning domains with the associated extracellular loops (C terminus) are alone sufficient for interaction with GHRH. In subsequent studies, chimeric proteins between the receptors for GHRH and vasoactive intestinal peptide (VIP) or secretin were generated, using the predicted start of the first transmembrane domain as the junction for the exchange of the N terminus between receptors. The chimeras having the N terminus of the GHRH receptor and the C terminus of either the VIP or secretin receptor (GNVC and GNSC) did not bind GHRH or activate adenylate cyclase after GHRH treatment. The reciprocal chimeras having the N terminus of either the VIP or secretin receptors and the C terminus of the GHRH receptor (VNGC and SNGC) bound GHRH and stimulated cAMP accumulation after GHRH treatment. These results suggest that although the N-terminal extracellular domain is essential for ligand binding, the transmembrane domains and associated extracellular loop regions of the GHRH receptor provide critical information necessary for specific interaction with GHRH.


Subject(s)
Mutagenesis, Site-Directed , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Recombinant Fusion Proteins/metabolism , Antigens, Viral/genetics , Binding Sites/genetics , Epitopes/genetics , HeLa Cells , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Ligands , Models, Molecular , Protein Binding/genetics , Protein Structure, Tertiary , Receptors, G-Protein-Coupled , Receptors, Gastrointestinal Hormone/analysis , Receptors, Gastrointestinal Hormone/biosynthesis , Receptors, Gastrointestinal Hormone/genetics , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Receptors, Vasoactive Intestinal Peptide/analysis , Receptors, Vasoactive Intestinal Peptide/biosynthesis , Receptors, Vasoactive Intestinal Peptide/genetics , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/biosynthesis , Secretin/metabolism
7.
Endocrinology ; 140(6): 2836-42, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10342875

ABSTRACT

In aging, alterations of pituitary GH-releasing hormone (GHRH) receptor (GHRH-R)-binding sites have been proposed as one of the initiating factors contributing to the loss of somatotroph responsiveness to GHRH. Changes in the characteristics and/or concentration of the functional GHRH-R could take place in the course of aging and reduce the sensitivity of the somatotroph axis to GHRH. Because chronic exposure to GHRH has been proposed to resensitize aged somatotroph cells, better knowledge of its effects on the regulation of the somatotroph axis is required, particularly at the level of GHRH-R. Two- and 18-month-old male Sprague Dawley rats were treated for 14 days with a daily s.c. injection of 0.5 or 1.0 mg/kg BW human GHRH-(1-29)NH2 or saline. In 2-month-old rats, treatment with 0.5 mg/kg GHRH increased the number of high affinity pituitary GHRH-R-binding sites by 2-fold (P < 0.05) and hypothalamic somatostatin (SRIF) content by 45% (P < 0.05). It did not affect hypothalamic GHRH content, serum total insulin-like growth factor I (IGF-I), or body weight gain. Treatment with 1.0 mg/kg GHRH decreased the number of high affinity pituitary GHRH-R-binding sites by 2.4-fold compared with that in rats treated with 0.5 mg/kg BW (P < 0.05) and increased hypothalamic SRIF content by 45% (P < 0.05), but did not affect GHRH content. It also decreased circulating levels of IGF-I by 13% (P < 0.05) and slowed the growth rate by 17% (P < 0.05). In 18-month-old rats, treatment with 0.5 mg/kg GHRH for 14 days was not sufficient to rejuvenate pituitary GHRH binding parameters. However, treatment with 1.0 mg/kg GHRH restored the affinities of high and low affinity classes of GHRH-binding sites to values similar to those found in 2-month-old rats. Binding capacities of the high and low affinity classes of sites were increased by 1.8- and 3-fold, respectively, although significance was only reached for the low affinity site (P < 0.05). These changes were associated with a normalization of the level of 2.5-kb GHRH-R messenger RNA transcript, which was decreased by 31% in aging rats (P < 0.05), and by a trend for an increase in the 4-kb GHRH-R messenger RNA transcript, which was already increased by 49% in 18-month-old rats (P < 0.05). A normalization of serum IGF-I levels, which were decreased by 11% in 18-month-old control rats (P < 0.01), was also observed. No treatment effect was detected on body weight or hypothalamic SRIF and GHRH contents. We conclude that a 14-day administration of GHRH induces a differential GHRH-R-mediated regulation at the level of the pituitary and probably the hypothalamus as a function of age.


Subject(s)
Aging/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Pituitary Gland, Anterior/chemistry , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Animals , Body Weight , Insulin-Like Growth Factor I/analysis , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Somatostatin/analysis
8.
Endocrinology ; 140(11): 5066-74, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10537133

ABSTRACT

The little mouse is a dwarf strain characterized by low levels of GH, pituitary hypoplasia, and an unresponsiveness to treatment with exogenous GHRH. The defect has been mapped to a missense mutation in the GHRH receptor gene that abolishes the function of the receptor, but the mechanism of this inactivation is unknown. Receptor function might be affected at the level of protein expression, maturation and processing, localization to the cell surface, ligand binding, or signaling. In this study, Western blots, using antiserum raised against the GHRH receptor and immunoprecipitation analysis of epitope-tagged receptors, demonstrate that both wild-type and mutant receptor proteins are expressed at equivalent levels in transfected cells. Immunofluorescence analysis of intact and permeabilized cells expressing the epitope-tagged receptors suggests that wild-type and little mouse receptors are similarly localized to the cell surface. A species homologous binding assay was developed and used to show that 125I-mouse GHRH binds with high affinity to the wild-type mouse receptor but not to the little mutant receptor. Consistent with this, the mutant receptor fails to stimulate intracellular cAMP accumulation. Our results demonstrate that the little mutation does not dramatically affect the expression level, glycosylation, or cellular localization of the receptor protein but that it blocks specific GHRH binding, and therefore, signaling does not take place.


Subject(s)
Growth Hormone-Releasing Hormone/metabolism , Mutation, Missense , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Membrane/chemistry , Cell Membrane/metabolism , Dwarfism, Pituitary/genetics , Fluorescent Antibody Technique, Indirect , Glycosylation , Humans , Immunosorbent Techniques , Mice , Mice, Mutant Strains , Molecular Sequence Data , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Signal Transduction , Structure-Activity Relationship
9.
Endocrinology ; 136(9): 4044-8, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7649113

ABSTRACT

Transgenic mice expressing a tyrosine hydroxylase-human (h) GH fusion gene in the hypothalamus exhibit a dwarf phenotype. The GH feedback mechanism(s) underlying the growth retardation in these animals was investigated by assessing peptide and messenger RNA (mRNA) levels of the hormones of the hypothalamic-GH-IGF-I axis. Pituitary GH content, hypothalamic GH-releasing hormone (GHRH) and somatostatin (SRIH) content, and serum IGF-I levels were measured by RIA. mRNA levels of hypothalamic GHRH and SRIH and of pituitary GH and the GHRH receptor were measured by Northern blot hybridization. Transgenic mice of both sexes and their wild-type littermates were studied at 2-4 months of age. The pituitary GH content was markedly reduced by 85% in male and by 87% in female transgenic mice compared to that in wild-type controls (P < 0.01 for both). The pituitary GH mRNA content was also decreased by 73% (P = 0.002) in transgenic male mice. Circulating IGF-I levels were significantly reduced by 66% and 68% in male and female transgenic mice, respectively (P = 0.001). The hypothalamic GHRH content was significantly reduced by 19% and 33% (P < 0.05) in male and female transgenic mice, respectively. No significant difference was detected, however, in the hypothalamic SRIH content between wild-type and transgenic mice. Hypothalamic GHRH mRNA levels were significantly decreased by 35% (P = 0.002) in transgenic male mice compared to those in wild-type littermates. In contrast, SRIH mRNA was not significantly changed. An even greater reduction (61%; P = 0.003) was observed in pituitary GHRH receptor mRNA in transgenic mice. These data indicate that the GH deficiency and dwarf phenotype of the tyrosine hydroxylase-hGH transgenic mouse can be attributed primarily to impaired hypothalamic GHRH production. The mechanism of GH feedback inhibition appears to involve direct suppression of GHRH gene expression by locally produced hGH in the hypothalamus.


Subject(s)
Genes, Reporter/genetics , Growth Hormone/genetics , Growth Hormone/metabolism , Hypothalamus/chemistry , Tyrosine 3-Monooxygenase/genetics , Animals , Base Sequence , Blotting, Northern , Corticotropin-Releasing Hormone/analysis , Corticotropin-Releasing Hormone/genetics , DNA/analysis , DNA/chemistry , DNA/genetics , DNA Primers/analysis , DNA Primers/chemistry , DNA Primers/genetics , Feedback/physiology , Female , Growth Hormone/analysis , Hypothalamus/physiology , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/physiology , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Pituitary Gland/chemistry , Pituitary Gland/physiology , RNA, Messenger/analysis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Receptors, Neuropeptide/analysis , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Sex Characteristics , Somatostatin/analysis , Somatostatin/genetics , Tyrosine 3-Monooxygenase/analysis
10.
Endocrinology ; 136(9): 4147-50, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7649123

ABSTRACT

Growth hormone (GH)-releasing hormone (GRH) acts on specific receptors in the anterior pituitary to stimulate the synthesis and release of GH. Recent reports suggest that GRH is also synthesized in extrahypothalamic tissues. To evaluate the potential roles of extrahypothalamic GRH, we studied the gene expression of GRH and GRH receptors in various rat tissues by reverse transcribed (RT)-polymerase chain reaction (PCR). Total RNA was extracted from twenty-three rat organs and RT-PCR was performed with GRH and GRH receptor primers. Highly-sensitive RT-PCR-Southern blotting showed that GRH and GRH receptor mRNA coexist in the widespread tissues (14 of 25 tissues). GRH mRNA was relatively abundant in the cerebral cortex, brain stem, testis, and placenta, while GRH receptor mRNA was abundant in renal medulla and renal pelvis. Northern blot hybridization using poly A+ RNA indicated that the transcript of GRH receptor gene found in the renal medulla was similar to the longer transcript (about 4 Kb) of pituitary GRH receptor in the size. These results suggest that GRH plays a potential role not only in the neuroendocrine axis, but also in the autocrine and paracrine systems in extrahypothalamic tissues.


Subject(s)
Brain Chemistry , Growth Hormone-Releasing Hormone/genetics , Kidney/chemistry , Placenta/chemistry , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Testis/chemistry , Animals , Base Sequence , Blotting, Northern , Blotting, Southern , Brain/metabolism , Brain/ultrastructure , Brain Stem/chemistry , Brain Stem/metabolism , Brain Stem/ultrastructure , Cerebral Cortex/chemistry , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Female , Gene Expression , Growth Hormone-Releasing Hormone/analysis , Growth Hormone-Releasing Hormone/metabolism , Hypothalamus/chemistry , Hypothalamus/metabolism , Hypothalamus/ultrastructure , Kidney/metabolism , Kidney/ultrastructure , Male , Molecular Sequence Data , Placenta/metabolism , Placenta/ultrastructure , Polymerase Chain Reaction , Pregnancy , RNA, Messenger/analysis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, Neuropeptide/analysis , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Testis/metabolism , Testis/ultrastructure
11.
J Clin Endocrinol Metab ; 87(10): 4707-14, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12364462

ABSTRACT

Antagonists of GHRH inhibit the growth of various human tumors, including prostate cancer, but the tumoral receptors mediating the antiproliferative effect of GHRH antagonists have not been clearly identified. Recently, we demonstrated that human cancer cell lines express splice variants (SVs) of receptors for GHRH, of which SV1 exhibits the greatest similarity to the pituitary GHRH receptors. In this study we investigated the expression of GHRH and SVs of GHRH receptor and the binding characteristics of the GHRH receptor isoform in 20 surgical specimens of organ-confined and locally advanced human prostatic adenocarcinomas. The mRNA expression of GHRH and SVs of GHRH receptor was investigated by RT-PCR. The affinity and density of receptors for GHRH were determined by ligand competition assays based on binding of (125)I-labeled GHRH antagonist JV-1-42 to tumor membranes. Twelve of 20 tumors (60%) exhibited specific, high affinity binding for JV-1-42, with a mean dissociation constant (K(d)) of 0.81 nmol/liter and a mean maximal binding capacity of 185.2 fmol/mg membrane protein. The mRNA of SV1 was detected in 13 of 20 (65%) prostate cancer specimens and was consistent with the presence of GHRH binding. RT-PCR analyses also revealed the expression of mRNA for GHRH in 13 of 15 (86%) prostatic carcinoma specimens examined. The presence of GHRH and its tumoral receptor SVs in prostate cancers suggests the possible existence of an autocrine mitogenic loop. The antitumor effects of GHRH antagonists in prostate cancer could be exerted in part by interference with this local GHRH system.


Subject(s)
Adenocarcinoma/metabolism , Alternative Splicing , Gene Expression , Growth Hormone-Releasing Hormone/genetics , Prostatic Neoplasms/metabolism , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Aged , Aged, 80 and over , Binding, Competitive , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Humans , Iodine Radioisotopes , Male , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/analysis , Radioligand Assay , Receptors, Neuropeptide/analysis , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
J Clin Endocrinol Metab ; 89(9): 4545-52, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15356060

ABSTRACT

Pituitary adenomas are very common in humans. They are of monoclonal origin, very heterogeneous, and produce frequently paradoxical secretion. The normal anterior pituitary (AP) contains some unorthodox multifunctional cells able to store more than one AP hormone (polyhormonal) and/or to express multiple hypothalamic-releasing hormone receptors (multiresponsive). Multifunctional AP cells seem to be involved in plasticity processes such as transdifferentiation or paradoxical secretion. Here, we have characterized the single-cell phenotypes of 15 human pituitary tumors, including prolactinomas, nonfunctioning adenomas, and adenomas from multiple endocrine neoplasia type I (MEN-I) and pituitary Cushing's disease patients. Individual tumor cells were typed according to expression of AP hormones and hypothalamic-releasing hormone receptors by combination of calcium imaging and multiple sequential immunocytochemistry in the same cells. We found a large heterogeneity among the different tumors. In eight of the 15 tumors studied, more than 80% of the cells presented a multifunctional phenotype. This may explain the occurrence of paradoxical secretion. In addition, our results suggest that human pituitary adenomas might derive from multifunctional cells. This is consistent with the existence of a link between pituitary plasticity and tumorigenesis.


Subject(s)
Adenoma/pathology , Pituitary Hormones, Anterior/metabolism , Pituitary Neoplasms/pathology , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Adenoma/etiology , Adenoma/metabolism , Adult , Aged , Cell Line, Tumor , Cushing Syndrome/metabolism , Cushing Syndrome/pathology , Female , Humans , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/metabolism , Multiple Endocrine Neoplasia Type 1/pathology , Phenotype , Pituitary Neoplasms/etiology , Pituitary Neoplasms/metabolism , Prolactinoma/metabolism , Prolactinoma/pathology
13.
FEBS Lett ; 394(1): 1-4, 1996 Sep 23.
Article in English | MEDLINE | ID: mdl-8925914

ABSTRACT

A site-directed anti-peptide antibody (anti-hGHRHRc18) was generated against the cytoplasmic tail of human GHRH receptor. The dissociation constant (Kd) and the antibody binding site (AbT) of anti-hGHRHRc18 were 2.5 nmol/l and 0.54 nmol/l, respectively. In an immunoblotting experiment, affinity-purified anti-hGHRHRc18 specifically recognized a single 50-kDa protein in human pituitary. In a screening of the expression of GHRH receptor protein in extra-pituitary tissues, only human kidney showed a single 52-kDa protein. Our results suggest that the GHRH receptor protein exhibits tissue-specific molecular heterogeneity.


Subject(s)
Receptors, Neuropeptide/analysis , Receptors, Neuropeptide/chemistry , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Receptors, Pituitary Hormone-Regulating Hormone/chemistry , Amino Acid Sequence , Animals , Antibodies/immunology , Chromatography, Affinity , Female , Humans , Immunoblotting , Kidney/chemistry , Molecular Sequence Data , Pituitary Gland/chemistry , Protein Binding , Radioimmunoassay , Rats , Receptors, Neuropeptide/immunology , Receptors, Pituitary Hormone-Regulating Hormone/immunology
14.
J Endocrinol ; 180(3): 369-78, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15012591

ABSTRACT

Fasting results in a reciprocal shift in hypothalamic neuropeptide Y (NPY) and GH-releasing hormone (GHRH) expression in the adult male rat. It is hypothesized that the fasting-induced rise in NPY is responsible for the GHRH decline and subsequent attenuation of pulsatile GH release. Fasting also leads to a decrease in circulating IGF-I, attributed to both reduced GH release and peripheral GH resistance. Although pituitary GH output is suppressed in the fasted rat, we report herein that pituitary GHRH receptor (GHRH-R) and GH secretagogue receptor (GHS-R) mRNA levels are increased, while pituitary expression of the somatostatin receptor subtype 2 (sst2) and 5 (sst5) is decreased, as determined by real-time reverse transcription (RT)-PCR. A shift in the expression of pituitary receptor subtypes to favor GH synthesis and release may be due, at least in part, to a decline in GH/IGF-I negative feedback. In order to test this hypothesis, we compared hypothalamic and pituitary response to fasting (72 h) in normal male rats and rats with isolated GH deficiency (spontaneous dwarf rats (SDR)). Circulating GH levels were undetectable in SDR, and IGF-I levels were less than 10% of normal controls. Fasting stimulated NPY mRNA levels in SDR; however, the rise in NPY mRNA levels was not accompanied by a fall in GHRH mRNA, as observed in fasted normal rats. In fact, GHRH mRNA levels paradoxically rose in the fasted SDR to 135% of fed controls. At the pituitary level, fasting did not alter sst2 and sst5 mRNA levels in SDR but did stimulate the expression of GHRH-R and GHS-R to 165% and 149% of fed controls, respectively. These results demonstrate that the fasting-induced changes in pituitary expression of sst2 and sst5, but not GHRH-R and GHS-R, are GH/IGF-I dependent. In addition, these results argue against the theory that the negative association of NPY and GHRH expression observed following fasting represents a simple cause-and-effect relationship and suggest that GH, either directly or indirectly, mediates the effects of fasting on hypothalamic GHRH expression.


Subject(s)
Dwarfism/metabolism , Fasting , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone/deficiency , Neuropeptide Y/metabolism , Actins/analysis , Animals , Base Sequence , Corticosterone/blood , DNA Primers , Growth Hormone/genetics , Growth Hormone-Releasing Hormone/genetics , Insulin-Like Growth Factor I/analysis , Male , Molecular Sequence Data , Neuropeptide Y/genetics , RNA, Messenger/analysis , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/analysis , Receptors, Ghrelin , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Receptors, Somatostatin/analysis , Reverse Transcriptase Polymerase Chain Reaction , Somatostatin/analysis
15.
Mol Cell Endocrinol ; 150(1-2): 65-72, 1999 Apr 25.
Article in English | MEDLINE | ID: mdl-10411301

ABSTRACT

Growth hormone releasing hormone (GHRH) receptors are members of the G-protein receptor family that use cAMP as a second messenger. A human fetal kidney 293-derived cell line stably expressing the porcine GHRH receptor (pGHRHr/293 cells) and a cAMP-responsive reporter system were used to develop a bioassay for human GHRH. The reporter system (ph alpha180SEAP) was constructed by subcloning the tandem cAMP response elements from the human glycoprotein hormone alpha subunit gene promoter (h alpha180) upstream from the secreted alkaline phosphatase cDNA of reporter plasmid pSEAP-Basic. To generate a stable cell line expressing both the GHRH receptor and SEAP reporter system, a DNA fragment from pPUR that confers puromycin resistance was subcloned downstream from the reporter construct of ph alpha180SEAP. Tranfection of ph alpha180SEAPpur into pGHRHr/293 cells yielded pGHRHr/SEAP/293 cell lines that responded to recombinant GHRH with dose-dependent increases in SEAP activity. The GHRH receptor-SEAP reporter bioassay was compared to a conventional bioassay using cultured rat anterior pituitary cells. Synthetic and recombinant GHRH induced a 3.1-fold increase in growth hormone release by rat pituitary cells with ED50's of 3.6 and 2.2 x 10(-10) M, respectively. Recombinant GHRH was 1.7 +/- 0.7 times more potent than synthetic GHRH in the pituitary cell bioassay. In an analogous experiment, pGHRHr/SEAP/293 cells responded to synthetic and recombinant GHRH with a 9.1-fold increase in SEAP activity. The ED50's were 7.8 and 4.3 x 10(-11) M, respectively, with recombinant GHRH being 1.8 +/- 0.1 times more potent than the synthetic preparation. Thus, the GHRH receptor-SEAP reporter bioassay is a sensitive, accurate, precise and efficient method for measuring GHRH biological activity.


Subject(s)
Biological Assay , Growth Hormone-Releasing Hormone/analysis , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Animals , Cell Line , Cyclic AMP , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Humans , Rats , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
16.
J Gerontol A Biol Sci Med Sci ; 55(6): B274-9, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10843343

ABSTRACT

Aging impairs and dietary restriction may modulate pituitary response to growth hormone (GH)-releasing hormone (GHRH) and somatostatin (SRIH) for GH secretion. Using the semiquantitative reverse-transcription polymerase chain reaction method, we analyzed the mRNA levels of the GHRH receptor (grfr) and SRIH receptor subtype 2 (sstr2) and subtype 5 (sstr5) in anterior pituitaries of male rats fed ad libitum or 30% dietary restricted. Aging reduced the mRNA levels of these receptors in a slightly different manner. The levels of grfr progressively decreased between 6 and 24 months, whereas those of sstr2 and sstr5 declined after 16 months. Dietary restriction did not diminish the aging-dependent changes, although it slightly augmented the levels of grfr, but not sstr2 and sstr5. The present results suggest that the aging-dependent impairment in pituitary response for GH secretion could result mostly from a decline in grfr rather than relative increase of sstrs. Although DR could slightly enhance the pituitary sensitivity to GHRH, the antiaging action may be minor at the level of gene expression.


Subject(s)
Aging/physiology , Diet , Pituitary Gland/metabolism , RNA, Messenger/biosynthesis , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Somatostatin/genetics , Animals , Male , Pituitary Gland/chemistry , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Receptors, Somatostatin/analysis
17.
Peptides ; 19(1): 15-20, 1998.
Article in English | MEDLINE | ID: mdl-9437732

ABSTRACT

We cloned a fragment of the rat GH-releasing peptide (GHRP) receptor homologue and examined the tissue distribution of GHRP receptor mRNA in rats. Sequence analysis showed that the open reading frame is well conserved between rat and human with 96% identity in a 364-amino acid overlap. By reverse transcription-polymerase chain reaction we detected GHRP receptor mRNAs in the rat brain including the hypothalamus, anterior pituitary, and renal pelvis in twenty-eight tissues tested. Microdissection revealed that GHRP receptor mRNAs were localized predominantly in the arcuate nucleus and ventromedial hypothalamus.


Subject(s)
Brain Chemistry , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Conserved Sequence/genetics , Gene Expression/genetics , Humans , Hypothalamus/chemistry , Molecular Sequence Data , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution
18.
Domest Anim Endocrinol ; 14(4): 241-9, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9260062

ABSTRACT

The objective of this study was to evaluate developmental changes in somatotroph function and related gene expression in neonatal pigs. Male piglets were sacrificed at 1, 7, 14, 21, 28, 35, and 42 d of age (8/age group) for the collection of tissue and blood. Serum concentrations of GH were determined. Quantitations of mRNA were performed for pituitary Pit-1, GH, and GHRH receptor. Cultures of pituitary cells from each pig were stimulated with 0, 0.1, 1, or 10 nM GHRH; 2 mM 8-Br-cAMP; or 100 nM phorbol myristate acetate. Elevated serum concentrations of GH were observed at 1 d of age, followed by a pronounced decrease to basal levels thereafter (P < 0.0001). A mild transient increase in circulating GH occurred at Day 28. In vitro GH secretion was significantly stimulated by secretagogue treatments (P < 0.0001). Age-related declines in in vitro GH secretion were observed regardless of if the cells were stimulated by GHRH or by secretagogues that bypass the GHRH receptor (P < 0.001). Similarly, cellular GH content varied with age (P = 0.01). Levels of pituitary GH mRNA (P = 0.01) and GHRH receptor mRNA (P = 0.0002) decreased with age. The quantity of GHRH receptor mRNA was correlated with GH mRNA levels (r = 0.55, P = 0.02), serum GH concentrations (r = 0.55, P = 0.02), and in vitro GH secretion (r = 0.66, P = 0.001). Pituitary Pit-1 mRNA levels at 7 and 14 d of age were significantly elevated relative to all other sampling times (P = 0.0002). Levels of Pit-1 and GH mRNAs were significantly correlated (r = 0.64, P = 0.003). These results demonstrate a strong developmental regulation of somatotrophic function and related gene expression during the early neonatal period of the pig. Age-related decreases in secretory function may be mediated by concurrent mechanisms relating to the expression of the GHRH receptor and of GH.


Subject(s)
Animals, Newborn/physiology , Gene Expression Regulation, Developmental , Growth Hormone/metabolism , Membrane Proteins , Pituitary Gland, Anterior/physiology , Swine/physiology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Aging/metabolism , Animals , Animals, Newborn/metabolism , Base Sequence , Carrier Proteins/analysis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , DNA Primers/analysis , DNA Primers/chemistry , DNA Primers/genetics , DNA, Complementary/analysis , DNA, Complementary/chemistry , DNA, Complementary/genetics , Dose-Response Relationship, Drug , Growth Hormone/analysis , Growth Hormone/genetics , Growth Hormone-Releasing Hormone/pharmacology , Linear Models , Male , Phospholipid Transfer Proteins , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Polymerase Chain Reaction/methods , RNA, Messenger/analysis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Radioimmunoassay , Receptors, Neuropeptide/analysis , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Swine/metabolism , Tetradecanoylphorbol Acetate/pharmacology
19.
J Cancer Res Clin Oncol ; 138(3): 387-95, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22160161

ABSTRACT

PURPOSE: The response to neoadjuvant chemoradiotherapy (CRT) varies greatly in patients suffering from locally advanced rectal cancer. Our aim was to correlate the response to CRT with the pre-treatment expression of heat shock protein 90 (Hsp90), small heat shock protein 16.2 (sHsp 16.2), phospho-Akt (p-Akt), growth hormone-releasing hormone receptor (GHRH-R) and heme-binding protein 2 (SOUL) in order to try to identify one or more as a predictive marker. MATERIALS AND METHODS: Sixty-nine patients receiving combined CRT for locally advanced rectal cancer were examined retrospectively. Surgical resection was carried out 6-9 weeks following CRT. The histopathological response to neoadjuvant treatment was determined according to the modified Mandard score. Using immunohistochemistry, we investigated the relationship between the expression of the five cited proteins in the pre-operative samples as well as various clinical parameters and the histopathological regression of the tumors. RESULTS: Thirty-one patients (48%) were good responders, and 33 patients (52%) were found to respond poorly to neoadjuvant therapy. Among patients undergoing surgery 7 weeks following CRT, there were significantly more good responders than among patients who underwent surgery sooner (63% vs. 37%). High levels of expression of GHRH-R and Hsp90 were shown to be significantly correlated with minor or absent histological regression. CONCLUSIONS: GHRH-R and Hsp90 were found to be independent predictive factors of histopathological response to neoadjuvant RCT. Since GHRH-R antagonists and Hsp90 inhibitors are currently being tested as potential anticancer agents, our study implies the possible elaboration of an effective and individualized treatment of poor responders.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , HSP90 Heat-Shock Proteins/analysis , Neoadjuvant Therapy/methods , Receptors, Neuropeptide/analysis , Receptors, Pituitary Hormone-Regulating Hormone/analysis , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy , Adult , Aged , Chemoradiotherapy, Adjuvant , Female , Heat-Shock Proteins, Small/analysis , Heme-Binding Proteins , Hemeproteins/analysis , Humans , Male , Middle Aged , Predictive Value of Tests , Pregnancy Proteins/analysis , Proto-Oncogene Proteins c-akt/analysis , Rectal Neoplasms/chemistry , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL