Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 188
Filter
Add more filters

Publication year range
1.
Reproduction ; 149(2): 147-54, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25376626

ABSTRACT

Declining female fecundity at later age and the increasing tendency for women to delay childbirth have lead to a drastic rise in the number of women seeking assisted reproductive technology. Many women fail to respond adequately to standard ovarian stimulation regimens, raising a significant therapeutic challenge. Recently, we have demonstrated that the administration of GH, as an adjunct to ovarian stimulation, has improved the clinical outcomes by enhancing the oocyte quality. However, the mechanism(s) by which GH facilitated this improvement is yet to be understood. This study aimed to determine these potential mechanism(s) through the use of immunofluorescent localisation of GH receptors (GHRs) on the human oocyte and unbiased computer-based quantification to assess and compare oocyte quality between women of varying ages, with or without GH treatment. This study demonstrates for the first time, the presence of GHRs on the human oocyte. The oocytes retrieved from older women showed significant decrease in the expression of GHRs and amount of functional mitochondria when compared with those from younger patients. More interestingly, when older patients were treated with GH, a significant increase in functional mitochondria was observed in their oocytes. We conclude that GH exerts a direct mode of action, enabling the improvement of oocyte quality observed in our previous study, via the upregulation of its own receptors and enhancement of mitochondrial activity. This result, together with recent observations, provides scientific evidence in support of the use of GH supplementation for the clinical management of poor ovarian response.


Subject(s)
Human Growth Hormone/administration & dosage , Ovulation Induction/methods , Adult , Female , Fluorescent Antibody Technique , Humans , Middle Aged , Mitochondria/ultrastructure , Oocytes/chemistry , Oocytes/physiology , Oocytes/ultrastructure , Pregnancy , Receptors, Somatotropin/analysis , Sperm Injections, Intracytoplasmic/methods
2.
BMC Vet Res ; 9: 247, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24314007

ABSTRACT

BACKGROUND: Biomarkers are useful tools in research and clinical practice where they are often used to detect and monitor differences in the physiological state of an animal. The proteins IGF-1, IGFBP-3, GHR, CRP, SAA, Hp, IFN-α, IFN-γ, TNF-α, IL-1ß, IL-6, IL-10, and IL-18 have been proposed as potential biomarkers for monitoring growth in livestock. The objective of this study was to determine whether hepatic gene expression of these proposed biomarkers is associated with growth performance in nursery pigs. Herd information and growth parameters were collected for 168 piglets from 8 commercial farms in southern Ontario. From these pigs, a subset of liver tissue samples (n = 74) was used for gene expression analysis of the proposed biomarkers. Multivariable linear regression methods were used to determine whether genetic expression of the proposed biomarkers was associated with growth performance in the nursery. RESULTS: Modelling the herd information and individual piglet traits in relation to growth performance revealed that the weight at weaning and the age at weaning are significantly associated with nursery performance. Average daily gain (ADG) was significantly associated with hepatic IGFBP-3 and GHR expression in the liver (P < 0.05), and tended to be associated with hepatic IGF-1 expression (P = 0.071). Similarly, 9-week body weight was significantly associated with hepatic expression of IGFBP-3 and GHR expression (P < 0.05), and tended to be associated with hepatic expression of IGF-1 (P = 0.055). CONCLUSION: The age and weight at which pigs are weaned is an important determinant for nursery performance. Hepatic gene expression of IGF-1, IGFBP-3, and GHR can be useful biomarkers for monitoring growth performance in nursery pigs.


Subject(s)
Biomarkers/analysis , Swine/growth & development , Age Factors , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Female , Gene Expression Profiling , Insulin-Like Growth Factor Binding Protein 3/analysis , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/physiology , Liver/chemistry , Male , Real-Time Polymerase Chain Reaction/veterinary , Receptors, Somatotropin/analysis , Receptors, Somatotropin/physiology , Swine/genetics
3.
J Dairy Sci ; 95(7): 3711-21, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22720928

ABSTRACT

The objective of this study was to characterize the circulating concentrations of insulin-like growth factor-I (IGF-I) and the hepatic expression of key genes regulating the somatotropic axis in cows divergent in genetic merit for fertility traits but with similar genetic merit for milk production traits. A total of 11 cows with good genetic merit for fertility (Fert+) and 12 cows with poor genetic merit for fertility (Fert-) underwent liver biopsy by percutaneous punch technique on d 20 (±6.7 d) prepartum and on d 2 (±1.5 d), d 58 (±3.7 d), d 145 (±13 d), and d 245 (±17.1 d) postpartum. Total RNA was isolated and the mRNA expression of growth hormone receptor (GHR 1A and GHRtot), IGF-I, janus tyrosine kinase 2 (JAK2), signal transducer and activator of transcription 5B (STAT5B), suppressor of cytokine signaling 3 (SOCS-3), acid-labile subunit (ALS), and IGF-binding proteins (IGFBP1 to IGFBP6) were measured by real-time quantitative PCR. During lactation, the circulating concentrations of IGF-I were 34% greater in Fert+ cows. The Fert+ cows had increased mean expression of IGF-I mRNA during the study; however, the difference in IGF-I mRNA abundance between Fert+ and Fert- cows was most pronounced at d 145 and 245. The expression of IGFBP3 and ALS transcript was similar in Fert+ and Fert- cows for the duration of the study. The Fert- cows, however, had greater expression of IGFBP2, IGFBP4, IGFBP5, and IGFBP6. Genotype had no effect on mRNA abundance of GHR 1A, STAT5B, JAK2, or SOCS-3. Genetic merit for fertility traits affects hepatic expression of key genes of the somatotropic axis regulating the synthesis, bioavailability, and stability of circulating IGF-I.


Subject(s)
Cattle/genetics , Fertility/genetics , Lactation/genetics , Liver/metabolism , Pregnancy, Animal/genetics , Quantitative Trait, Heritable , Animals , Carrier Proteins/analysis , Carrier Proteins/genetics , Cattle/physiology , Female , Fertility/physiology , Genes/genetics , Genes/physiology , Glycoproteins/analysis , Glycoproteins/genetics , Insulin-Like Growth Factor Binding Proteins/analysis , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor I/analysis , Janus Kinase 2/analysis , Janus Kinase 2/genetics , Lactation/physiology , Liver/chemistry , Pregnancy , Pregnancy, Animal/physiology , Receptors, Somatotropin/analysis , Receptors, Somatotropin/genetics , STAT5 Transcription Factor/analysis , STAT5 Transcription Factor/genetics , Suppressor of Cytokine Signaling Proteins/analysis , Suppressor of Cytokine Signaling Proteins/genetics
4.
Gen Comp Endocrinol ; 172(3): 344-57, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21458459

ABSTRACT

Endocrine actions of growth hormone (GH) have been implicated during the development of adult testicular function in several mammalian species, and recently intracrine, autocrine, and paracrine effects have been proposed for locally expressed GH. Previous reports have shown the distribution of GH mRNA and the molecular heterogeneity of GH protein in both adult chicken testes and vas deferens. This study provides evidence of the presence and distribution of GH and its receptor (GHR) during all stages of spermatogenesis in adult chicken testes. This hormone and its receptor are not restricted to the cytoplasm; they are also found in the nuclei of spermatogonia, spermatocytes, and spermatids. The pattern of GH isoforms was characterized in the different, isolated germ cell subpopulations, and the major molecular variant in all subpopulations was 17 kDa GH, as reported in other chicken extra-pituitary tissues. Another molecular variant, the 29 kDa moiety, was found mainly in the enriched spermatocyte population, suggesting that it acts at specific developmental stages. The co-localization of GH with the proliferative cell nuclear antigen PCNA (a DNA replication marker present in spermatogonial cells) was demonstrated by immunohistochemistry. These results show for the first time that GH and GHR are present in the nuclei of adult chicken germinal cells, and suggest that GH could participate in proliferation and differentiation during the complex process of spermatogenesis.


Subject(s)
Chickens/metabolism , Growth Hormone/metabolism , Testis/metabolism , Animals , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Growth Hormone/analysis , Growth Hormone/genetics , Immunohistochemistry , Male , Proliferating Cell Nuclear Antigen/analysis , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/metabolism , Receptors, Somatotropin/analysis , Receptors, Somatotropin/metabolism , Sexual Maturation , Spermatogenesis , Testis/cytology , Testis/ultrastructure
5.
J Comp Neurol ; 529(6): 1228-1239, 2021 04 15.
Article in English | MEDLINE | ID: mdl-32844436

ABSTRACT

Multiple neuroendocrine, autonomic and behavioral responses are regulated by the paraventricular nucleus of the hypothalamus (PVH). Previous studies have shown that PVH neurons express the growth hormone (GH) receptor (GHR), although the role of GH signaling on PVH neurons is still unknown. Given the great heterogeneity of cell types located in the PVH, we performed a detailed analysis of the neurochemical identity of GH-responsive cells to understand the possible physiological importance of GH action on PVH neurons. GH-responsive cells were detected via the phosphorylated form of the signal transducer and activator of transcription-5 (pSTAT5) in adult male mice that received an intraperitoneal GH injection. Approximately 51% of GH-responsive cells in the PVH co-localized with the vesicular glutamate transporter 2. Rare co-localizations between pSTAT5 and vesicular GABA transporter or vasopressin were observed, whereas approximately 20% and 38% of oxytocin and tyrosine hydroxylase (TH) cells, respectively, were responsive to GH in the PVH. Approximately 55%, 35% and 63% of somatostatin, thyrotropin-releasing hormone (TRH) and corticotropin-releasing hormone (CRH) neurons expressed GH-induced pSTAT5, respectively. Additionally, 8%, 49% and 75% of neuroendocrine TH, TRH and CRH neurons, and 67%, 32% and 74% of nonneuroendocrine TH, TRH and CRH neurons were responsive to GH in the PVH of Fluoro-Gold-injected mice. Our findings suggest that GH action on PVH neurons is involved in the regulation of the thyroid, somatotropic and adrenal endocrine axes, possibly influencing homeostatic and stress responses.


Subject(s)
Growth Hormone/metabolism , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/metabolism , Phenotype , Receptors, Somatotropin/metabolism , Animals , Growth Hormone/analysis , Male , Mice , Mice, Inbred C57BL , Paraventricular Hypothalamic Nucleus/cytology , Receptors, Somatotropin/analysis
6.
Brain Res ; 1751: 147189, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33152340

ABSTRACT

A growth hormone (GH) injection is able to induce the phosphorylated form of the signal transducer and activator of transcription 5 (pSTAT5) in a large number of cells throughout the mouse brain. The present study had the objective to map the distribution of GH-responsive cells in the brain of rats that received an intracerebroventricular injection of GH and compare it to the pattern found in mice. We observed that rats and mice exhibited a similar distribution of GH-induced pSTAT5 in the majority of areas of the telencephalon, hypothalamus and brainstem. However, rats exhibited a higher density of GH-responsive cells than mice in the horizontal limb of the diagonal band of Broca (HDB), supraoptic and suprachiasmatic nuclei, whereas mice displayed more GH-responsive cells than rats in the hippocampus, lateral hypothalamic area and dorsal motor nucleus of the vagus (DMX). Since both HDB and DMX contain acetylcholine-producing neurons, pSTAT5 was co-localized with choline acetyltransferase in GH-injected animals. We found that 50.0 ± 4.5% of cholinergic neurons in the rat HDB coexpressed GH-induced pSTAT5, whereas very few co-localizations were observed in the mouse HDB. In contrast, rats displayed fewer cholinergic neurons responsive to GH in the DMX at the level of the area postrema. In summary, pSTAT5 can be used as a marker of GH-responsive cells in the rat brain. Although rats and mice exhibit a relatively similar distribution of GH-responsive neurons, some species-specific differences exist, as exemplified for the responsiveness to GH in distinct populations of cholinergic neurons.


Subject(s)
Brain Mapping/methods , Receptors, Somatotropin/analysis , STAT5 Transcription Factor/analysis , Acetylcholine , Animals , Brain/metabolism , Brain Stem/metabolism , Choline O-Acetyltransferase/metabolism , Cholinergic Neurons/metabolism , Growth Hormone/metabolism , Growth Hormone/pharmacology , Hippocampus/metabolism , Hypothalamus/metabolism , Infusions, Intraventricular , Male , Medulla Oblongata/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Rats , Rats, Long-Evans , Receptors, Somatotropin/metabolism , STAT5 Transcription Factor/metabolism
7.
Tumori ; 96(2): 282-8, 2010.
Article in English | MEDLINE | ID: mdl-20572587

ABSTRACT

AIMS AND BACKGROUND: Recombinant human growth hormone (rhGH) is increasingly used in the clinic because it promotes the synthesis of proteins. However, rhGH is able to increase malignant transformation and tumor recurrence. The aim of this study was to investigate the effects of rhGH on hepatocellular carcinoma (HCC) cells with positive and negative growth hormone receptors (GHR) in order to guide its clinical application. METHODS AND STUDY DESIGN: Cells of the human HCC cell lines Bel-7402 (GHR+) and SMMC-7721 (GHR-) as well as human umbilical vein endothelial cell line ECV304 cells in the exponential growth phase were harvested and divided into experimental and control groups. After the human HCC cells were cultured alone or co-cultured with ECV304 cells under the different treatments, cell cycle phase, proliferation index, and expression levels of vascular endothelial growth factor (VEGF) mRNA and proteins were determined. RESULTS: In the Bel-7402 GHR+ cells treated with rhGH, both the percentage of cell in G2-M phase and the proliferation index were higher than those of controls (P < 0.05); this was not the case in the SMMC-7721 GHR- cells treated with rhGH (P > 0.05). Although there was no difference in the cell doubling times between ECV304 cells co-incubated with Bel-7721 GHR-cells treated with rhGH and without rhGH, the doubling times of ECV304 cells co-incubated with Bel-7402 GHR+ cells, when treated with rhGH, were significantly shortened compared to those of controls (P < 0.05). The cell doubling times of ECV304 cells co-incubated with Bel-7721 GHR- or Bel-7402 GHR+ cells which were treated with bevacizumab were longer than those of controls and of cells with rhGH (P < 0.05). The VEGF mRNA and protein expression levels were higher in Bel-7402 GHR+ cells treated with different doses of rhGH than controls (P < 0.05 or P < 0.01); however, there was no statistically significant difference in the expression levels of VEGF mRNA and proteins between SMMC-7721 GHR- cells treated with rhGH and controls. CONCLUSIONS: rhGH can induce VEGF secretion and stimulate proliferation of Bel-7402 GHR+ cells in vitro, but has little effect on the proliferation of SMMC-7721 GHR-cells, suggesting that rhGH may be applied safely to treatment for the catabolic state in patients with GHR-negative HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Human Growth Hormone/pharmacology , Liver Neoplasms/pathology , Receptors, Somatotropin/analysis , Carcinoma, Hepatocellular/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Liver Neoplasms/chemistry , RNA, Messenger/analysis , Vascular Endothelial Growth Factor A/genetics
8.
J Clin Endocrinol Metab ; 93(4): 1379-85, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18182451

ABSTRACT

CONTEXT: Endothelial dysfunction is common in patients with GH deficiency who are at increased risk for premature cardiovascular death. GH regulates vascular tone and reactivity in humans. OBJECTIVE: Our objective was to explore the mechanisms underlying the GH's acute vascular effects. DESIGN AND STUDY SETTING: There were 10 healthy, lean and young, volunteers studied after an overnight fast. GH was infused systemically for 6 h at 0.06 microg/kg.min. Biopsy of the vastus lateralis muscle was done in seven subjects before and after GH infusion. Human aortic endothelial cells (HAECs) were incubated with GH in vitro. RESULTS: GH infusion increased plasma GH to 32.9 +/- 1.5 ng/ml and forearm blood flow by 66% (P < 0.001). GH infusion did not significantly change plasma IGF-I concentrations, muscle IGF-I mRNA expression, and muscle Akt phosphorylation, suggesting a lack of IGF-I action in muscle. Because it was reported that GH exerts an acute vascular effect via a nitric oxide (NO)-dependent mechanism, we performed additional in vitro experiments using HAECs. HAECs express abundant GH receptors. Incubating HAECs with GH at 30 ng/ml for 3 or 6 h did not alter endothelial NO synthase (eNOS) protein content but time dependently increased the phosphorylation and activity of eNOS, thus demonstrating a direct effect of GH on endothelial cells. CONCLUSIONS: GH exerts an acute vascular effect independent of both systemic and local IGF-I production, and this effect is likely via direct action on GH receptors and eNOS in the vascular endothelium.


Subject(s)
Blood Vessels/drug effects , Human Growth Hormone/pharmacology , Insulin-Like Growth Factor I/physiology , Adult , Blood Glucose/analysis , Cells, Cultured , Endothelial Cells/drug effects , Female , Forearm/blood supply , Humans , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/genetics , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Somatotropin/analysis , Regional Blood Flow/drug effects
9.
Endocr Rev ; 23(5): 623-46, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12372843

ABSTRACT

An understanding of the events that occur during GH receptor (GHR) signaling has facilitated the development of a GHR antagonist (pegvisomant) for use in humans. This molecule has been designed to compete with native GH for the GHR and to prevent its proper or functional dimerization-a process that is critical for GH signal transduction and IGF-I synthesis and secretion. Clinical trials in patients with acromegaly show GHR blockade to be an exciting new mode of therapy for this condition, and pegvisomant may have a therapeutic role in diseases, such as diabetes and malignancy, in which abnormalities of the GH/IGF-I axis have been observed. This review charts the discovery and development of GHR antagonists and details the experience gained in patients with acromegaly.


Subject(s)
Acromegaly/drug therapy , Human Growth Hormone/analogs & derivatives , Receptors, Somatotropin/antagonists & inhibitors , Animals , Clinical Trials as Topic , Drug Design , Human Growth Hormone/chemistry , Human Growth Hormone/genetics , Human Growth Hormone/physiology , Human Growth Hormone/therapeutic use , Humans , Models, Molecular , Molecular Structure , Receptors, Somatotropin/analysis , Receptors, Somatotropin/genetics , Receptors, Somatotropin/physiology , Structure-Activity Relationship
10.
Endocr Rev ; 12(3): 235-51, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1935820

ABSTRACT

PRL and GH are hormones with a wide spectrum of actions. Specific receptors are widely distributed in a number of classical target organs, but other tissues that are not known targets also contain measurable binding sites or receptor mRNA. The most likely explanation is that PRL and GH cause effects that have not yet been characterized in certain tissues. Cloning of the cDNAs encoding PRL and GH receptors has led to the discovery that the receptors, like the hormones themselves, form a gene family. Multiple receptor forms have been identified, including a short form, which for PRL is a membrane-bound receptor or for GH is a soluble BP, and a long form, which for both PRL and GH is a membrane-bound receptor. PRL and GH receptors, and the mRNAs encoding them, can be up- and down-regulated. GH induces an up-regulation of both GH and PRL receptors, whereas PRL stimulates an increase of only its own receptor. High concentrations of either hormone induce a homologous down-regulation of receptor expression. An assay has been developed to measure the functional activity of different forms of PRL receptor by cotransfecting a milk protein fusion gene specific to PRL coupled to a reporter-gene along with the cDNA of the PRL receptor. Although the short form represents the major form present in rat mammary gland, only the long form of receptor is able to stimulate milk protein gene transcription. For GH, increased expression of the receptor in some target cells is accompanied by a modest enhancement of the response to GH. No single second messenger mediating the action of either PRL or GH has been identified. Several potential components of the signal transduction pathways have been identified, but as yet none has clearly been shown to be able to mimic the effect of PRL or GH. Because of the wide range of biological actions associated with PRL, and the existence of various forms of PRL receptors, it is doubtful that one unifying mechanism of action will be found for this hormone. No human or animal model of a genetic defect of the PRL receptor has thus far been published. Mutations in the GH receptor gene have been demonstrated in Laron type dwarfism. Different exon deletions or point or nonsense mutations resulting in modifications in the extracellular, GH binding region of the GH receptor have been reported.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Receptors, Prolactin , Receptors, Somatotropin , Animals , Gene Expression , Growth Hormone/pharmacology , Growth Hormone/physiology , Humans , Prolactin/pharmacology , Prolactin/physiology , Receptors, Prolactin/analysis , Receptors, Prolactin/genetics , Receptors, Prolactin/physiology , Receptors, Somatotropin/analysis , Receptors, Somatotropin/genetics , Receptors, Somatotropin/physiology , Tissue Distribution
11.
J Dairy Sci ; 91(5): 1802-13, 2008 May.
Article in English | MEDLINE | ID: mdl-18420611

ABSTRACT

The growth hormone/insulin-like growth factor (IGF) system plays a critical endocrine role controlling nutrient metabolism in dairy cattle. In liver, growth hormone receptor (GHR) and IGF-1 are dynamically regulated by lactation and energy balance. Less is known about the regulation of GHR, IGF-1, and IGF-binding protein mRNA in reproductive tissues (uterus, ovarian follicle, and corpus luteum). The objective was to determine expression patterns for GHR, IGF-1, and IGF-binding protein (IGFBP)-2 mRNA in the liver, uterus, dominant follicle, and corpus luteum in Holstein cows (n = 21) sampled at 3 times during early lactation. The first postpartum ovulation was induced with an injection of GnRH within 15 d of calving. Nine days after ovulation [23 +/- 1 d postpartum; 20 d in milk (DIM)], the liver, uterus, dominant follicle, and corpus luteum were biopsied. Prostaglandin F(2alpha) and GnRH were injected 7 and 9 d after each biopsy to synchronize the second (41 +/- 1 d postpartum; 40 DIM) and third (60 +/- 1 d postpartum; 60 DIM) tissue collections. Total RNA was isolated and used for mRNA analysis by real-time quantitative reverse transcription PCR. Liver had more GHR, IGF-1, and IGFBP-2 mRNA than the reproductive tissues that were tested. Gene expression for GHR, IGF-1, and IGFPB-2 within tissues did not change across the sampling interval (20 to 60 DIM). The only detected change in gene expression across days was for cyclophilin in uterus (increased after 20 DIM). Parity had an effect on gene expression for GHR in corpus luteum. Neither level of milk production nor body condition score affected the amount of GHR, IGF-1, or IGFBP-2 mRNA in the respective tissues. The repeatability of gene expression within a tissue was 0.25 to 0.5 for most genes. In most instances, expression of a single gene within a tissue was correlated with other genes in the same tissue but was not correlated with the same gene in a different tissue. We did not find evidence for major changes in gene expression within reproductive tissues in postpartum cows. Differences between cows (independent of their BCS and milk production) accounted for a major portion of the variation that we observed.


Subject(s)
Cattle/metabolism , Genitalia, Female/metabolism , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor I/genetics , Postpartum Period/metabolism , Receptors, Somatotropin/genetics , Animals , Body Composition , Body Weight , Corpus Luteum/anatomy & histology , Corpus Luteum/chemistry , Cyclophilins/analysis , Female , Gene Expression , Genitalia, Female/chemistry , Growth Hormone/blood , Insulin-Like Growth Factor Binding Protein 2/analysis , Insulin-Like Growth Factor I/analysis , Lactation/physiology , Ovarian Follicle/anatomy & histology , Ovarian Follicle/chemistry , RNA, Messenger/analysis , Receptors, Somatotropin/analysis , Reverse Transcriptase Polymerase Chain Reaction , Uterus/chemistry
12.
Clinics (Sao Paulo) ; 63(1): 39-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18297205

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the presence of growth hormone receptor in plexiform neurofibromas of neurofibromatosis type 1 patients. INTRODUCTION: The development of multiple neurofibromas is one of the major features of neurofibromatosis type 1. Since neurofibromas commonly grow during periods of hormonal change, especially during puberty and pregnancy, it has been suggested that hormones may influence neurofibromatosis type 1 neurofibromas. A recent study showed that the majority of localized neurofibromas from neurofibromatosis type 1 patients have growth hormone receptor. METHODS: Growth hormone receptor expression was investigated in 5 plexiform neurofibromas using immunohistochemistry. RESULTS: Four of the 5 plexiform neurofibromas were immunopositive for growth hormone receptor. CONCLUSION: This study suggests that growth hormone may influence the development of plexiform neurofibromas in patients with neurofibromatosis type 1.


Subject(s)
Biomarkers, Tumor/analysis , Neurofibroma, Plexiform/chemistry , Neurofibromatosis 1/metabolism , Receptors, Somatotropin/analysis , Adolescent , Adult , Child , Female , Humans , Immunohistochemistry , Neurofibroma, Plexiform/etiology , Neurofibromatosis 1/complications
13.
Growth Horm IGF Res ; 17(2): 104-12, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17321774

ABSTRACT

Growth hormone (GH) binding to GH receptor (GHR) is the initial step that leads to the physiological functions of the hormone. Proteolytical cleavage of the GHR in humans and rabbits and alternative processing of the GHR transcript in rodents generates circulating growth hormone binding protein (GHBP). Moreover, other GHR truncated forms that result from alternative processing of the GHR mRNA transcript have been described. These GHR short forms are inserted in the plasma membrane but they are unable to transduce the signal. In rodents, membrane associated-GHBP (MA-GHBP), which accounts for a significant proportion of liver GH binding capacity, represents the main GHR short form found in membranes, and may therefore function as a negative form of the receptor. In the present study, GHR and MA-GHBP content in liver were analyzed using mutant and transgenic mice expressing different concentrations of growth hormone to evaluate the correlation between GH levels, body weight (BW), GHR and MA-GHBP expression. It was found that GH deficiency was associated with diminished BW, GHR and MA-GHBP expression, while increased GH concentration led to increased BW, GHR and MA-GHBP expression, but MA-GHBP upregulation was more pronounced than the observed increase in GHR expression. Since GHR and MA-GHBP both contribute to liver GH binding capacity, GH-induced enrichment of the dominant negative form would represent a compensatory mechanism triggered by high levels of the hormone. This attempt to attenuate the effects of supraphysiological concentrations of GH may be critical to reduce or prevent their plausible damaging effects on the organism.


Subject(s)
Carrier Proteins/metabolism , Growth Hormone/physiology , Liver/metabolism , Receptors, Somatotropin/metabolism , Animals , Carrier Proteins/analysis , Cattle , Cell Membrane/chemistry , Female , Growth Hormone/genetics , Liver/chemistry , Mice , Mice, Transgenic , Receptors, Somatotropin/analysis , Up-Regulation
14.
Growth Horm IGF Res ; 17(1): 47-53, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17161642

ABSTRACT

IGF-I is regarded as the most sensitive marker of growth hormone (GH) secretion in both GH deficient individuals and in individuals with excessive GH production. Studies on the effect of inhibitors of GH action in normal experimental animals are difficult to evaluate due to the complex relationship and feed back mechanisms of the GH/IGF-I system and the hypothalamo-pituitary axis. To circumvent the GH/IGF-I feedback mechanisms, we have used hypophysectomized (HX) rats treated with GH to assess the potential of a new low molecular weight compound, BVT-A ((N-[5-(aminosulfonyl)-2-methylphenyl]-5-bromo-2-furamide), to act as a GH receptor antagonist in vivo. GH treatment of HX rats induced serum IGF-I, body weight and hepatic mRNA levels of IGF-I, IGFBP-3, ALS and the IGF-I and GH receptors. Co-treatment with BVT-A suppressed all the GH-induced effects. We conclude that the GH substituted HX rat is a useful model for studies on GH receptor antagonists, and for the first time, a small molecule GH receptor antagonist with in vivo activity has been revealed. This opens up for development of new drugs for diseases in which lowering of GH receptor activity would be beneficial.


Subject(s)
Acromegaly/drug therapy , Receptors, Somatotropin/antagonists & inhibitors , Sulfonamides/administration & dosage , Acromegaly/pathology , Administration, Oral , Animals , Body Weight/drug effects , Drug Evaluation, Preclinical , Human Growth Hormone/administration & dosage , Hypophysectomy , Insulin-Like Growth Factor Binding Protein 3/analysis , Insulin-Like Growth Factor I/analysis , Liver/chemistry , Male , Molecular Weight , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/analysis , Receptors, Somatotropin/analysis
15.
J Endocrinol ; 189(1): 147-54, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16614389

ABSTRACT

Diabetic retinopathy and acromegaly are diseases associated with excess action of GH and its effector IGF-I, and there is a need for improved therapies. We have designed an optimised 2'-O-(2-methoxyethyl)-modified phosphorothioate oligodeoxynucleotide, ATL 227446, and demonstrated its ability to suppress GH receptor mRNA in vitro. Subcutaneous injections of ATL 227446 reduced GH receptor mRNA levels, GH binding activity and serum IGF-I levels in mice after seven days of dosing. The reduction in serum IGF-I could be sustained for over ten weeks of dosing at therapeutically relevant levels, during which there was also a significant decrease in body weight gain in antisense-treated mice relative to saline and mismatch control-treated mice. The findings indicate that administration of an antisense oligonucleotide to the GH receptor may be applicable to human diseases in which suppression of GH action provides therapeutic benefit.


Subject(s)
Insulin-Like Growth Factor I/analysis , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides/administration & dosage , Receptors, Somatotropin/analysis , Weight Gain/drug effects , Animals , Cells, Cultured , Gene Expression/genetics , Growth Hormone/metabolism , Injections, Subcutaneous , Insulin-Like Growth Factor I/antagonists & inhibitors , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/analysis , Receptors, Somatotropin/antagonists & inhibitors
16.
Obesity (Silver Spring) ; 24(5): 1090-1096, 2016 05.
Article in English | MEDLINE | ID: mdl-27015877

ABSTRACT

OBJECTIVE: Growth hormone (GH) administration reduces abdominal, but not lower body, fat mass. To gain insight into the underlying mechanisms, this study examined the expression of the GH receptor (GHR) and some of its targets in abdominal and gluteal adipose tissue. METHODS: GHR and GH targets in the lipolytic pathway were assessed (quantitative PCR/Western blotting) in adipose aspirates from premenopausal women [n = 15, age 26.9 ± 6.1 years, body mass index (BMI) 28.0 ± 6.8 kg/m(2) ] and men (n = 28, age 29.2 ± 7.0 years, BMI 26.9 ± 3.7 kg/m(2) ). RESULTS: GHR mRNA expression was lower in the gluteal depot when compared with the abdominal depot (P = 0.01). Abdominal GHR correlated negatively with age and BMI, whereas gluteal GHR was associated with lower waist-to-hip ratio (WHR), that is, pear shape. In both sites, GHR mRNA correlated strongly with genes important for the regulation of lipolysis: adipose tissue triglyceride lipase (ATGL), hormone-sensitive lipase, perilipin, and CIDEA (all P < 0.001), independently of BMI, WHR, age, and sex. GHR protein was lower in the gluteal fat when compared with the abdominal fat (P = 0.03) and correlated with ATGL protein in the gluteal depot (P < 0.001). CONCLUSIONS: GHR levels correlate with levels of lipases and lipid droplet-associated proteins crucial for lipolysis. Thus, higher GHR expression in the abdominal depot when compared with the gluteal depot may underlie the in vivo effect of GH to specifically reduce abdominal adipose tissue mass.


Subject(s)
Adipose Tissue/metabolism , Gene Expression , Receptors, Somatotropin/genetics , Subcutaneous Fat, Abdominal/metabolism , Adiposity , Adult , Aging , Body Mass Index , Buttocks , Carrier Proteins , Female , Human Growth Hormone/physiology , Humans , Lipase/analysis , Lipolysis/genetics , Male , Obesity/metabolism , Premenopause , RNA, Messenger/analysis , Receptors, Somatotropin/analysis , Waist-Hip Ratio
17.
Biochim Biophys Acta ; 1013(3): 223-30, 1989 Oct 09.
Article in English | MEDLINE | ID: mdl-2804083

ABSTRACT

Lactogenic and somatogenic receptors present in rat liver have been examined by cross-linking with a derivative of human somatotropin (AP-hGH1) followed by SDS-polyacrylamide gel electrophoresis. AP-hGH1, which has a content of 2.2 azidophenacyl groups per molecule, mainly linked to half Cys-182 and half Cys-189, exerted a specificity similar to that of the native hormone (hGH), with an ability of 46% with respect to hGH to compete with the radiolabelled hormone for the binding sites of microsomal preparations. Photolysis of the 125I-labelled derivative bound to the lactogenic receptors present in either microsomal membranes or Triton X-100 solubilized preparations gave rise to a 63 kDa species. In addition, 30% of the covalent complexes formed in microsomal membranes belonged to a species with a molecular mass of 70 kDa. Incubation of viable rat hepatocytes with the radiolabelled derivative at either 0 degrees C for 3 h or 15 degrees C for 1.5 h and subjection to irradiation, yielded covalent complexes of molecular masses estimated at 130, 73, 63, 45 and 35 kDa. Experiments performed in the presence of 1 mM NaCN, gave rise to the previous species in a similar yield as that obtained in the absence of cyanide. The 130 kDa complex is related to the somatogenic binding sites, since it was not visualized in the presence of unlabelled bovine somatotropin, while the 70-73, 63, 45 and 35 kDa bands disappeared when the incubations were performed in the presence of unlabelled ovine prolactin.


Subject(s)
Liver/analysis , Microsomes, Liver/analysis , Receptors, Prolactin/analysis , Receptors, Somatotropin/analysis , Animals , Binding, Competitive , Cross-Linking Reagents , Electrophoresis, Polyacrylamide Gel , Female , Growth Hormone/metabolism , Intracellular Membranes/analysis , Liver/ultrastructure , Microsomes, Liver/metabolism , Molecular Weight , Photolysis , Rats , Rats, Inbred Strains , Receptors, Prolactin/metabolism , Receptors, Somatotropin/metabolism , Sodium Cyanide/pharmacology , Solubility
18.
Endocrinology ; 146(5): 2434-44, 2005 May.
Article in English | MEDLINE | ID: mdl-15718272

ABSTRACT

A critical step in establishment of human pregnancy is the invasion of the uterus wall by the extravillous cytotrophoblast (EVCT), a process regulated by multiple autocrine and paracrine factors. Hormones belonging to the GH/prolactin family are expressed at the maternofetal interface. Because they are involved in cell motility in various models, we examined the possible regulatory role of human placental GH (hPGH) in EVCT invasiveness. By using an in vitro invasion model, we found that EVCT isolated from first-trimester chorionic villi and cultured on Matrigel secreted hPGH and expressed human GH receptor (hGHR). These data were confirmed by in situ immunohistochemistry. EVCT expressed the full-length and truncated forms of hGHR, and the Janus kinase-2/signal transducer and activator of transcription factor-5 signaling pathway was activated in EVCT by hPGH treatment. Strong hPGH and hGHR expression was observed when EVCT invaded Matrigel and moved through the pores of the filter on which they were cultured. hPGH stimulated EVCT invasiveness, and this effect was inhibited by a Janus kinase-2 inhibitor. Interestingly, hPGH was more efficient than pituitary GH in stimulating EVCT invasiveness. These results offer the first evidence for a placental role of hPGH and suggest an autocrine/paracrine role of hPGH in the regulation of trophoblast invasion.


Subject(s)
Growth Hormone/pharmacology , Placental Hormones/pharmacology , Trophoblasts/physiology , Cells, Cultured , Chorionic Villi , DNA-Binding Proteins/metabolism , Female , Gene Expression/physiology , Growth Hormone/genetics , Growth Hormone/metabolism , Humans , Immunohistochemistry , Janus Kinase 2 , Milk Proteins/metabolism , Models, Biological , Placenta/physiology , Placental Hormones/genetics , Placental Hormones/metabolism , Placental Lactogen/genetics , Pregnancy , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Somatotropin/analysis , Receptors, Somatotropin/genetics , Recombinant Fusion Proteins , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor , Trans-Activators/metabolism , Transfection
19.
J Clin Endocrinol Metab ; 90(5): 2493-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15713716

ABSTRACT

Human GH has two disulfide bridges linking Cys-53 to Cys-165 and Cys-182 to Cys-189. Although absence of the first disulfide bridge has been shown to affect the bioactivity of GH in transgenic mice, little is known of the importance of this bridge in mediating the GH/GH-receptor (GHR) interaction in humans. However, we have identified a missense mutation (G705C) in the GH1 gene of a Serbian patient. This mutation was found in the homozygous state and leads to the absence of the disulfide bridge Cys-53 to Cys-165. To study the impact of this mutation in vitro, GHR binding and Janus kinase (Jak)2/signal transducer and activator of transcription (Stat)5 activation experiments were performed, in which it was observed that at physiological concentrations (3-50 ng/ml) both GHR binding and Jak2/Stat5 signaling pathway activation were significantly reduced in the mutant GH-C53S, compared with wild-type (wt)-GH. Higher concentrations (400 ng/ml) were required for this mutant to elicit responses similar to wt-GH. These results demonstrate that the absence of the disulfide bridge Cys-53 to Cys-165 affects the binding affinity of GH for the GHR and subsequently the potency of GH to activate the Jak2/Stat5 signaling pathway. In conclusion, we have demonstrated that GH-C53S is a bioinactive GH at the physiological range and that the disulfide bridge Cys-53 to Cys-163 is required for mediating the biological effects of GH.


Subject(s)
Body Height , Growth Disorders/genetics , Human Growth Hormone/genetics , Mutation, Missense , Child , DNA-Binding Proteins/metabolism , Female , Humans , Janus Kinase 2 , Male , Milk Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Somatotropin/analysis , STAT5 Transcription Factor , Trans-Activators/metabolism
20.
Diabetes ; 41(5): 605-9, 1992 May.
Article in English | MEDLINE | ID: mdl-1568530

ABSTRACT

Poorly controlled insulin-dependent diabetes mellitus (IDDM) is associated with elevated basal plasma growth hormone (GH), disproportionally low insulin-like growth factor I (IGF-I) levels, and impaired somatic growth. These derangements in the GH-IGF axis imply a state of GH resistance. The mechanism of GH resistance is unknown; it may involve a defect at the level of the GH receptor, unresponsiveness due to a postreceptor defect in GH action, or both. To investigate a potential receptor involvement, we measured plasma high-affinity GH-binding protein (GHBP), which represents a truncated GH receptor and may reflect GH receptor levels in tissues, in patients with IDDM, patients with non-insulin-dependent diabetes (NIDDM), and nondiabetic control subjects. Patients with IDDM had significantly lower plasma GHBP levels than either patients with NIDDM or nondiabetic control subjects (mean value 18.2 vs. 24.6 and 23.8% GH bound/ml plasma, respectively, P less than 0.001). This difference persisted when only lean patients (less than 115% ideal body wt) were included in the analysis. Basal plasma GH levels were significantly elevated in IDDM compared with either patients with NIDDM or nondiabetic control subjects (mean 6.9 vs. 2.1 and 2.0 micrograms/L, respectively, P less than 0.001), whereas IFG-I levels were not significantly different in IDDM and NIDDM. No correlations were found between levels of GHBP and HbA1, duration of diabetes, or plasma GH. GHBP and IGF-I levels were significantly correlated in NIDDM but not in IDDM. We conclude that IDDM is associated with low GHBP levels and that GH resistance found in this disorder may be mediated, at least in part, by a decrease in GH receptor levels.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Carrier Proteins/blood , Diabetes Mellitus, Type 1/blood , Adolescent , Adult , Aged , Aged, 80 and over , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Female , Growth Hormone/blood , Humans , Insulin-Like Growth Factor I/analysis , Male , Middle Aged , Radioimmunoassay , Receptors, Somatotropin/analysis , Receptors, Somatotropin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL