Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Protein Sci ; 32(9): e4727, 2023 09.
Article in English | MEDLINE | ID: mdl-37428391

ABSTRACT

Recombinant human growth hormone (rhGH) and GH receptor antagonists (GHAs) are used clinically to treat a range of disorders associated with GH deficiency or hypersecretion, respectively. However, these biotherapeutics can be difficult and expensive to manufacture with multiple challenges from recombinant protein generation through to the development of long-acting formulations required to improve the circulating half-life of the drug. In this review, we summarize methodologies and approaches used for making and purifying recombinant GH and GHA proteins, and strategies to improve pharmacokinetic and pharmacodynamic properties, including PEGylation and fusion proteins. Therapeutics that are in clinical use or are currently under development are also discussed.


Subject(s)
Human Growth Hormone , Receptors, Somatotropin , Humans , Human Growth Hormone/genetics , Human Growth Hormone/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Receptors, Somatotropin/agonists , Receptors, Somatotropin/antagonists & inhibitors
2.
Eur J Pharmacol ; 891: 173753, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33245901

ABSTRACT

The intracellular signaling pathway of growth hormone (GH)-stimulated DNA synthesis and proliferation was investigated in primary cultures of adult rat hepatocytes. DNA synthesis and cell proliferation were detected in hepatocyte parenchymal cells grown in serum-free, defined medium containing GH (100 ng/ml). GH-stimulated hepatocyte DNA synthesis and proliferation were almost completely blocked by TG101209 (10-6 M), a selective Janus kinase (JAK)2 inhibitor, U-73122 (10-6 M), a selective phospholipase C (PLC) inhibitor, and a monoclonal antibody to insulin-like growth factor-I (IGF-I) receptor (100 ng/ml) or anti-secretion agents such as somatostatin (10-6 M) and BAPTA/AM (10-7 M). In addition, blocking monoclonal antibodies to IGF-I, but not transforming growth factor-α, completely inhibited GH-induced hepatocyte DNA synthesis and proliferation. IGF-I levels in the culture medium increased rapidly versus baseline levels within 5 min in response to GH (100 ng/ml), and the maximum IGF-I level (100 pg/ml) was reached 20 min after GH stimulation. Autocrine secretion of IGF-I into the culture medium was inhibited by a growth-inhibitory dose of TG101209, U-73122, somatostatin, or BAPTA/AM. These data indicate that the proliferative mechanism of action of GH is mediated mainly through a GH receptor/JAK2/PLC-stimulated increase in the autocrine secretion of IGF-I by primary cultured hepatocytes, followed by stimulation of the 95 kDa IGF-I receptor tyrosine kinase signaling pathway.


Subject(s)
Autocrine Communication , Cell Proliferation/drug effects , DNA Replication/drug effects , Hepatocytes/drug effects , Human Growth Hormone/pharmacology , Insulin-Like Growth Factor I/metabolism , Animals , Cells, Cultured , Hepatocytes/metabolism , Janus Kinase 2/metabolism , Male , Phosphorylation , Primary Cell Culture , Rats, Wistar , Receptor, IGF Type 1/metabolism , Receptors, Somatotropin/agonists , Receptors, Somatotropin/metabolism , Secretory Pathway , Signal Transduction , Type C Phospholipases/metabolism
3.
J Cell Physiol ; 234(12): 23388-23397, 2019 12.
Article in English | MEDLINE | ID: mdl-31173363

ABSTRACT

It is well known that zinc ion (Zn2+ ) can regulate the biological activity of growth hormone (GH). However, until now, the mechanism by which Zn2+ regulates GH biological activity remains unclear. In the current study, we first performed molecular docking between Zn2+ and porcine GH (pGH) using computational biology. We then explored the effect of Zn2+ on the GH signaling ability in the cell model expressing porcine growth hormone receptor (GHR). It was found that the phosphorylation levels of Janus kinase 2, signal transducers and activators of transcription 5/3/1, and GHR increased significantly under Zn2+ treatment, indicating that Zn2+ can enhance the signaling ability of GH/GHR. On this basis, we further explored how Zn2+ regulates the biological activity of GH/GHR. The results showed that downregulation and turnover of GHR changed under Zn2+ /pGH treatment. Zn2+ enhanced the membrane residence time of pGH/GHR and delayed GHR downregulation. Further investigation showed that the internalization dynamic of pGH/GHR was changed by Zn2+ , which prolonged the residence time of pGH/GHR in the cell membrane. These factors acted together to upregulate the signaling of GH/GHR. This study lays a foundation for further exploration of the biological effects of Zn2+ on GH.


Subject(s)
Cell Membrane/drug effects , Chlorides/pharmacology , Hepatocytes/drug effects , Human Growth Hormone/pharmacology , Receptors, Somatotropin/agonists , Zinc Compounds/pharmacology , Animals , Binding Sites , CHO Cells , Cell Membrane/metabolism , Chlorides/metabolism , Cricetulus , Endocytosis , Hepatocytes/metabolism , Human Growth Hormone/metabolism , Janus Kinase 2/metabolism , Molecular Docking Simulation , Phosphorylation , Protein Binding , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Sus scrofa , Zinc Compounds/metabolism
4.
Article in English | MEDLINE | ID: mdl-30114526

ABSTRACT

Phoenixin (Pnx) is an endogenous peptide known to be involved in reproduction and food intake in rats, with two active isoforms, phoenixin-14 (Pnx-14) and phoenixin-20 (Pnx-20). However, little is known about the functions of Pnx in teleost. Here, pnx was cloned and was detected in all tissues of both male and female in spotted scat (Scatophagus argus), including growth axis, hypothalamus, pituitary, and liver. Real-time PCR analysis showed that pnx in the hypothalamus increased significantly after 2 d and 7 d fasting, while reduced significantly after re-feeding (P < 0.05). When pituitary and liver fragments were cultured in vitro with Pnx-14 and Pnx-20 (10 nM and 100 nM) for 6 h, the expression of ghrhr (growth hormone-releasing hormone receptor) and gh (growth hormone) in the pituitary, and ghr1 (growth hormone receptor 1) in the liver increased significantly, except ghr2 (growth hormone receptor 2) incubated with 10 nM and 100 nM Pnx-20 and ghr1 incubated with 10 nM Pnx-20. Similarly, the expression of ghrhr and gh in the pituitary, as well as ghr1 and ghr2 in the liver, increased significantly after injecting S. argus with Pnx-14 and Pnx-20 (10 ng/g and 100 ng/g body weight). These results indicate that Pnx is likely to be involved in the regulation of food intake, and also regulates the growth of S. argus by increasing ghrhr and gh expression in the pituitary, ghr1 and ghr2 in the liver, and ghr1 directly in the liver.


Subject(s)
Energy Intake , Fish Proteins/metabolism , Gene Expression Regulation, Developmental , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Peptide Hormones/metabolism , Perciformes/physiology , Animals , Aquaculture , China , Energy Intake/drug effects , Female , Fish Proteins/administration & dosage , Fish Proteins/genetics , Fish Proteins/pharmacology , Gene Expression Regulation, Developmental/drug effects , Growth Hormone/agonists , Growth Hormone/genetics , Growth Hormone/metabolism , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/genetics , Hypothalamic Hormones/pharmacology , Hypothalamus/drug effects , Injections, Intraperitoneal , Liver/drug effects , Liver/metabolism , Male , Organ Specificity , Peptide Hormones/administration & dosage , Peptide Hormones/genetics , Peptide Hormones/pharmacology , Perciformes/growth & development , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Protein Isoforms/administration & dosage , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , Random Allocation , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Receptors, Somatotropin/agonists , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Tissue Culture Techniques/veterinary , Weight Gain
5.
Eur J Endocrinol ; 178(5): 481-489, 2018 May.
Article in English | MEDLINE | ID: mdl-29500309

ABSTRACT

BACKGROUND: Patients with homozygous intronic pseudoexon GH receptor (GHR) mutations (6Ψ) have growth hormone insensitivity (GHI) (growth failure, IGF1 deficiency and normal/elevated serum GH). We report 9 patients in addition to previously described 11 GHR 6Ψ patients and their responses to rhIGF1 therapy. METHODS: 20 patients (12 males, 11 families, mean age 4.0 ± 2.2 years) were diagnosed genetically in our centre. Phenotypic data and responses to rhIGF1 treatment were provided by referring clinicians. Continuous parametric variables were compared using Student t-test or ANOVA. RESULTS: 10/20 (50%) had typical facial features of GHI, 19/20 (95%) from consanguineous families and 18/20 (90%) of Pakistani origin. At diagnosis, mean height SDS: -4.1 ± 0.95, IGF1 SDS: -2.8 ± 1.4; IGFBP3 SDS: -3.0 ± 2.1 and mean basal and peak GH levels: 11.9 µg/L and 32.9 µg/L, respectively. 1/12 who had IGF1 generation test, responded (IGF1: 132-255 ng/mL). 15/20 (75%; 11M) received rhIGF1 (mean dose: 114 µg/kg twice daily, mean duration: 5.3 ± 2.5 years). Mean baseline height velocity of 4.7 ± 1.1 cm/year increased to 7.4 ± 1.8 cm/year (P = 0.001) during year 1 of therapy. Year 3 mean height SDS (-3.2 ± 1.0) was higher than pre-treatment height SDS (-4.3 ± 0.8) (P = 0.03). Mean cumulative increase in height SDS after year 5 was 1.4 ± 0.9. Difference between target height (TH) SDS and adult or latest height SDS was less than that of TH SDS and pre-treatment height SDS (2.1 ± 1.2 vs 3.0 ± 0.8; P = 0.02). CONCLUSION: In addition to phenotypic heterogeneity in the cohort, there was mismatch between clinical and biochemical features in individual patients with 6Ψ GHR mutations. rhIGF1 treatment improved height outcomes.


Subject(s)
Growth Disorders/prevention & control , Insulin-Like Growth Factor I/therapeutic use , Laron Syndrome/drug therapy , Point Mutation , Receptors, Somatotropin/agonists , Receptors, Somatotropin/genetics , Body Height/drug effects , Child , Child, Preschool , Consanguinity , Drug Resistance , England , Family Health , Female , Growth Disorders/etiology , Homozygote , Humans , Insulin-Like Growth Factor I/genetics , Introns , Laron Syndrome/genetics , Laron Syndrome/metabolism , Laron Syndrome/physiopathology , Male , Pakistan/ethnology , Receptors, Somatotropin/metabolism , Recombinant Proteins/therapeutic use
6.
Eur J Endocrinol ; 177(6): R309-R321, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28904008

ABSTRACT

Pharmacogenetics aims to maximize the beneficial effects of a medical therapy by identifying genetic finger prints from responders and non-responders and, thereby improving safety and efficacy profile of the drug. Most subjects who are deficient in growth hormone (GHD) are candidates for recombinant human GH (rhGH) therapy. To date, it is well established that even after adjustments for several clinical variables, such as age, gender, body composition and the age at onset of the GHD, response to rhGH treatment is highly variable among individuals, part of which is believed to be due to genetic factors within the GH system. As the first genetic variant to potentially influence the individual response to rhGH therapy in children with growth disorders, polymorphism in the GH receptor (GHR) has attracted a great interest as a target for pharmacogenetics. Studies have been conducted to compare the functional and molecular effects of the full-length GHR (fl-GHR) isoform with the exon 3 deleted (d3-GHR) isoform in children and adults treated with rhGH therapy. Additionally, the impact of the GHR polymorphism has been investigated in relation to the clinical status and response to medical treatment in acromegaly, especially to the GHR antagonist drug pegvisomant. We have performed a narrative review of the studies performed to date on the association of GHR polymorphism with rhGH response in children and adults, and its potential influence in the medical management of acromegaly. In addition, data from studies on the general population and in other chronic diseases examining a role of this genetic variant in the regulation of growth and metabolism are summarized.


Subject(s)
Growth Disorders/drug therapy , Hormone Replacement Therapy/adverse effects , Human Growth Hormone/deficiency , Human Growth Hormone/therapeutic use , Pharmacogenomic Variants , Polymorphism, Genetic , Receptors, Somatotropin/genetics , Acromegaly/chemically induced , Acromegaly/genetics , Acromegaly/metabolism , Acromegaly/therapy , Adult , Child , Drug Resistance , Exons , Gene Deletion , Growth Disorders/etiology , Growth Disorders/genetics , Growth Disorders/metabolism , Human Growth Hormone/adverse effects , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/genetics , Humans , Peptide Fragments/adverse effects , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Protein Isoforms/adverse effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/therapeutic use , Receptors, Somatotropin/agonists , Receptors, Somatotropin/antagonists & inhibitors , Receptors, Somatotropin/metabolism , Recombinant Proteins/adverse effects , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use
7.
J Biol Chem ; 291(7): 3346-58, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26703468

ABSTRACT

After significant injury, the liver must maintain homeostasis during the regenerative process. We hypothesized the existence of mechanisms to limit hepatocyte proliferation after injury to maintain metabolic and synthetic function. A screen for candidates revealed suppressor of cytokine signaling 2 (SOCS2), an inhibitor of growth hormone (GH) signaling, was strongly induced after partial hepatectomy. Using genetic deletion and administration of various factors we investigated the role of SOCS2 during liver regeneration. SOCS2 preserves liver function by restraining the first round of hepatocyte proliferation after partial hepatectomy by preventing increases in growth hormone receptor (GHR) via ubiquitination, suppressing GH pathway activity. At later times, SOCS2 enhances hepatocyte proliferation by modulating a decrease in serum insulin-like growth factor 1 (IGF-1) that allows GH release from the pituitary. SOCS2, therefore, plays a dual role in modulating the rate of hepatocyte proliferation. In particular, this is the first demonstration of an endogenous mechanism to limit hepatocyte proliferation after injury.


Subject(s)
Insulin-Like Growth Factor I/antagonists & inhibitors , Liver Regeneration , Liver/physiology , Receptors, Somatotropin/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/metabolism , Ubiquitination , Animals , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Growth Hormone/antagonists & inhibitors , Growth Hormone/metabolism , Hepatectomy/adverse effects , Immunohistochemistry , Insulin-Like Growth Factor I/analysis , Liver/cytology , Liver/surgery , Male , Mice, Inbred C57BL , Mice, Knockout , Pituitary Gland/cytology , Pituitary Gland/metabolism , Protein Transport , Proteolysis , Receptors, Somatotropin/agonists , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Suppressor of Cytokine Signaling Proteins/genetics
8.
Mol Cell Endocrinol ; 390(1-2): 54-64, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24755421

ABSTRACT

A series of studies have reported that anti-GHR antibody can function as a GHR agonist and may serve as an attractive tool for studying the mechanisms of GHR activation. However, to date, there is relatively little information about intracellular signalling triggered by anti-GHR antibody. Therefore, in this work, we have developed a panel of monoclonal antibodies to GHBP, among which one Mab, termed CG-172, was selected for further characterisation because of its signalling properties. The results from FACS assays, receptor binding and immunoprecipitation assays and western blotting demonstrated that CG-172 specifically binds to GHR expressed on target cells. Subsequently, epitope mapping studies that used receptor binding analysis showed that CG-172 specifically binds subdomain 1 of GHR ECD. We next examined the resulting signal transduction pathways triggered by this antibody in CHO-GHR638 cells and rat hepatocytes. We found that CG-172 can activate JAK2, AKT, ERK1/2 and STAT1/3 but not STAT5. The phosphorylation kinetics of STAT1/3, AKT and ERK1/2 induced by either GH or CG-172 were analysed in dose-response and time course experiments. Our observations demonstrated that an anti-GHR monoclonal antibody (CG-172) can serve as an attractive tool to study the mechanism(s) of GHR-mediated intracellular signalling pathways and may lead to the production of signal-specific molecules that are capable of inducing different biochemical responses.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , MAP Kinase Signaling System , Receptors, Somatotropin/agonists , Animals , CHO Cells , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Epitope Mapping , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Human Growth Hormone/pharmacology , Humans , Mice, Inbred BALB C , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptors, Somatotropin/immunology , Receptors, Somatotropin/metabolism , STAT Transcription Factors/metabolism
9.
J Pediatr Endocrinol Metab ; 26(9-10): 833-40, 2013.
Article in English | MEDLINE | ID: mdl-23729600

ABSTRACT

AIM: Some cases of idiopathic short stature (ISS) may be caused by defects in the modulation of the negative feedback regulation of the growth hormone receptor (GHR)/ Janus kinase (JAK)2/signal transducers and activators of transcription (STAT)5 signaling pathway. The cytosolic tyrosine phosphatases, protein tyrosine phosphatase 1B (PTP1B) and Src homology 2 (SH2) domain-containing protein-tyrosine phosphatase-1 (SHP-1), the later which translocates to the nucleus after activation, interact with JAK2 in a GH-dependent manner. The possible contribution of PTP1B and SHP-1 to GH signaling in fibroblasts from ISS patients has not been studied. METHODS: We determined the basal protein content of PTP1B and SHP-1 in the presence of recombinant human GH (rhGH) for 24 h in skin fibroblast cultures, obtained from patients with ISS, and were compared with a normal height control children group. JAK2 activation was determined in both groups. RESULTS: JAK2 activation was delayed in fibroblasts from ISS patients compared to controls. Under basal conditions, the protein content of SHP-1 was lower in ISS, and after incubation with rhGH, it decreased in the non-nuclear and nuclear fraction of controls, but not in ISS patients. The protein content of PTP1B, however, increased in a similar fashion in fibroblasts from both ISS and control children. CONCLUSION: The delayed activation of JAK2 and the lack of response of SHP-1 after incubation with GH in fibroblasts from ISS patients, suggests that the growth retardation observed in some of these children may be mediated in part by this phosphotyrosine phosphatase.


Subject(s)
Growth Disorders/enzymology , Human Growth Hormone/metabolism , Janus Kinase 2/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Signal Transduction , Skin/enzymology , Body Height , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Cells, Cultured , Child , Child Development , Enzyme Activation , Growth Disorders/metabolism , Growth Disorders/pathology , Human Growth Hormone/genetics , Humans , Janus Kinase 2/chemistry , Kinetics , Male , Phosphorylation , Protein Processing, Post-Translational , Receptors, Somatotropin/agonists , Receptors, Somatotropin/metabolism , Recombinant Proteins/metabolism , Skin/metabolism , Skin/pathology
10.
Cell Signal ; 23(2): 417-24, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20946955

ABSTRACT

Epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) signal through EGF and PDGF receptors, which are important receptor tyrosine kinases (RTKs). Growth hormone (GH) and prolactin (PRL) are four helical bundle peptide hormones that signal via GHR and PRLR, members of the cytokine receptor superfamily. In this study, we examine crosstalk between signaling pathways emanating from these disparate receptor groups (RTKs and cytokine receptors). We find that GH and EGF specifically synergize for activation of ERK in murine preadipocytes. The locus of this synergy resides at the level of MEK activation, but not above this level (i.e., not at the level of EGFR, SHC, or Raf activation). Furthermore, dephosphorylation of the scaffold protein, KSR, at a critical serine residue is also synergistically promoted by GH and EGF, suggesting that GH sensitizes these cells to EGF-induced ERK activation by augmenting the actions of KSR in facilitating MEK-ERK activation. Similarly specific synergy in ERK activation is also detected in human T47D breast cancer cells by cotreatment with PRL and PDGF. This synergy also resides at the level of MEK activation. Consistent with this synergy, PRL and PDGF also synergized for c-fos-dependent transactivation of a luciferase reporter gene in T47D cells, indicating that events downstream of ERK activation reflect this signaling synergy. Important conceptual and physiological implications of these findings are discussed.


Subject(s)
ErbB Receptors/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, Platelet-Derived Growth Factor/physiology , Receptors, Prolactin/physiology , Receptors, Somatotropin/physiology , 3T3 Cells , Animals , Cells, Cultured , Enzyme Activation , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/physiology , ErbB Receptors/agonists , Genes, Reporter , Human Growth Hormone/pharmacology , Human Growth Hormone/physiology , Humans , Mice , Phospholipase C gamma/metabolism , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Platelet-Derived Growth Factor/physiology , Prolactin/pharmacology , Prolactin/physiology , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor Cross-Talk , Receptors, Platelet-Derived Growth Factor/agonists , Receptors, Prolactin/agonists , Receptors, Somatotropin/agonists , Signal Transduction
11.
Curr Diabetes Rev ; 7(1): 50-5, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21067510

ABSTRACT

Involvement of the growth hormone (GH) / insulin-like growth factor 1 (IGF-I) axis in the pathogenesis of diabetic nephropathy (DN) is strongly suggested by studies investigating the impact of GH excess and deficiency on renal structure and function. GH excess in both the human (acromegaly) and in transgenic animal models is characterized by significant structural and functional changes in the kidney. In the human a direct relationship has been noted between the activity of the GH/IGF-1 axis and renal hypertrophy, microalbuminuria, and glomerulosclerosis. Conversely, states of GH deficiency or deficiency or inhibition of GH receptor (GHR) activity confer a protective effect against DN. The glomerular podocyte plays a central and critical role in the structural and functional integrity of the glomerular filtration barrier and maintenance of normal renal function. Recent studies have revealed that the glomerular podocyte is a target of GH action and that GH's actions on the podocyte could be detrimental to the structure and function of the podocyte. These results provide a novel mechanism for GH's role in the pathogenesis of DN and offer the possibility of targeting the GH/IGF-1 axis for the prevention and treatment of DN.


Subject(s)
Diabetic Nephropathies/etiology , Human Growth Hormone/metabolism , Human Growth Hormone/physiology , Kidney Glomerulus/metabolism , Podocytes/metabolism , Animals , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Disease Progression , Humans , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/physiology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Models, Biological , Podocytes/pathology , Receptors, Somatotropin/agonists , Receptors, Somatotropin/metabolism
12.
Orv Hetil ; 151(34): 1384-93, 2010 Aug 22.
Article in Hungarian | MEDLINE | ID: mdl-20705553

ABSTRACT

Growth hormone (GH) hypersecretion leads to acromegaly which is associated with several co-morbidities and increased mortality. Despite of the typical clinical features and modern diagnostic tools, it often takes years from the onset of the disease until the diagnosis. The aims of the treatment are to reduce or control the tumor growth, inhibit the GH hypersecretion, normalize the insulin-like growth factor-I (IGF-I) levels, treat the co-morbidities and therefore reduce mortality. There are three approaches for therapy: surgery, medical management (dopamine agonist, somatostatin analogues and GH receptor antagonist), and radiotherapy. Efficient therapy of the disease is based on the appropriate multidisciplinary team management.


Subject(s)
Acromegaly/etiology , Pituitary Neoplasms/diagnosis , Pituitary Neoplasms/therapy , Acromegaly/diagnosis , Acromegaly/epidemiology , Acromegaly/metabolism , Acromegaly/therapy , Human Growth Hormone/metabolism , Humans , Interdisciplinary Communication , Patient Care Team , Pituitary Neoplasms/complications , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/radiotherapy , Pituitary Neoplasms/surgery , Receptors, Dopamine/drug effects , Receptors, Somatotropin/agonists , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use
13.
Nat Rev Endocrinol ; 6(9): 515-25, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20664532

ABSTRACT

Growth hormone is widely used clinically to promote growth and anabolism and for other purposes. Its actions are mediated via the growth hormone receptor, both directly by tyrosine kinase activation and indirectly by induction of insulin-like growth factor 1 (IGF-1). Insensitivity to growth hormone (Laron syndrome) can result from mutations in the growth hormone receptor and can be treated with IGF-1. This treatment is, however, not fully effective owing to the loss of the direct actions of growth hormone and altered availability of exogenous IGF-1. Excessive activation of the growth hormone receptor by circulating growth hormone results in gigantism and acromegaly, whereas cell transformation and cancer can occur in response to autocrine activation of the receptor. Advances in understanding the mechanism of receptor activation have led to a model in which the growth hormone receptor exists as a constitutive dimer. Binding of the hormone realigns the subunits by rotation and closer apposition, resulting in juxtaposition of the catalytic domains of the associated tyrosine-protein kinase JAK2 below the cell membrane. This change results in activation of JAK2 by transphosphorylation, then phosphorylation of receptor tyrosines in the cytoplasmic domain, which enables binding of adaptor proteins, as well as direct phosphorylation of target proteins. This model is discussed in the light of salient information from closely related class 1 cytokine receptors, such as the erythropoietin, prolactin and thrombopoietin receptors.


Subject(s)
Receptors, Somatotropin/agonists , Receptors, Somatotropin/physiology , Animals , Hormone Antagonists/pharmacology , Hormone Antagonists/therapeutic use , Human Growth Hormone/antagonists & inhibitors , Human Growth Hormone/metabolism , Human Growth Hormone/physiology , Human Growth Hormone/therapeutic use , Humans , Janus Kinase 2/metabolism , Models, Biological , Models, Molecular , Mutation/physiology , Phosphorylation , Receptors, Somatotropin/chemistry , Receptors, Somatotropin/genetics
14.
Growth Horm IGF Res ; 20(2): 118-26, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20022531

ABSTRACT

CONTEXT: Growth hormone (GH) is an anabolic hormone that regulates growth and metabolism. Ames dwarf mice are natural mutants for Prop1, with impaired development of anterior pituitary and undetectable levels of circulating GH, prolactin and TSH. They constitute an endocrine model of life-long GH-deficiency. The main signaling cascades activated by GH binding to its receptor are the JAK2/STATs, PI-3K/Akt and the MAPK Erk1/2 pathways. OBJECTIVES: We have previously reported that GH-induced STAT5 activation was higher in Ames dwarf mice liver compared to non-dwarf controls. The aim of this study was to evaluate the principal components of the main GH-signaling pathways under GH-deficiency in liver and skeletal muscle, another GH-target tissue. METHODS: Ames dwarf mice and their non-dwarf siblings were assessed. Animals were injected i.p. with GH or saline 15min before tissue removal. Protein content and phosphorylation of signaling mediators were determined by immunoblotting of tissue solubilizates. RESULTS: GH was able to induce STAT5 and STAT3 tyrosine phosphorylation in both liver and muscle, but the response was higher for Ames dwarf mice than for non-dwarf controls. When Erk1/2 activation was assessed in liver, only dwarf mice showed GH-induced phosphorylation, while in muscle no response to the hormone was found in either genotype. GH-induced Akt phosphorylation at Ser473 in liver was only detected in dwarf mice. In skeletal muscle, both normal and dwarf mice responded to a GH stimulus, although dwarf mice presented higher GH activation levels. The phosphorylation of GSK-3, a substrate of Akt, increased upon hormone stimulation only in dwarf mice in both tissues. In contrast, no differences in the phosphorylation of mTOR, another substrate of Akt, were observed after GH stimulus, either in normal or dwarf mice in liver, while we were unable to determine mTOR in muscle. Protein content of GH-receptor and of the signaling mediators studied did not vary between normal and dwarf animals in the assessed tissues. CONCLUSION: These results show that several components of the main GH-signaling pathways exhibit enhanced sensitivity to the hormone in liver and muscle of Ames dwarf mice.


Subject(s)
Dwarfism, Pituitary/metabolism , Growth Hormone/pharmacology , Homeodomain Proteins/genetics , Liver/drug effects , Muscle, Skeletal/drug effects , Signal Transduction/drug effects , Animals , Drug Resistance/drug effects , Drug Resistance/genetics , Dwarfism, Pituitary/genetics , Dwarfism, Pituitary/pathology , Glycogen Synthase Kinase 3/metabolism , Growth Hormone/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Somatotropin/agonists , Receptors, Somatotropin/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases
15.
J Endocrinol ; 202(3): 463-71, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19525340

ABSTRACT

Previously, we isolated and characterized two distinct GH receptor (GHR)-encoding mRNAs, ghr1 and ghr2, from rainbow trout. In this study, Chinese hamster ovary-K1 cells were individually transfected with plasmids that contained cDNAs encoding rainbow trout ghr1 or ghr2. High affinity binding of (125)I-salmonid GH (sGH) by the expressed receptors was saturable, displaceable, and ligand selective. Whole-cell binding analysis revealed a single class of binding site; for Ghr1 K(d)=8 nM, for Ghr2 K(d)=17 nM. While salmonid prolactin (sPrl) displaced (125)I-sGH from both Ghr1 and Ghr2, the affinity of either receptor subtype for sPrl was substantially less than for sGH; salmonid somatolactin, another member of the GH-PRL family, did not displace labeled sGH except at pharmacological concentrations. (125)I-sGH was internalized by Ghr1- and Ghr2-expressing cells in a time-dependent manner; the maximum internalization reached was 71% for Ghr1 and 55% for Ghr2. Long-term exposure (24 h) of transfected cells to sGH up-regulated surface expression of both Ghr1 and Ghr2; however, sGH induced surface expression of Ghr1 to a greater extent than that of Ghr2. These results indicate that rainbow trout ghrs display both overlapping and distinct characteristics that may be important for ligand selection and differential action in target organs.


Subject(s)
Oncorhynchus mykiss/physiology , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Animals , Binding, Competitive/physiology , CHO Cells , Cricetinae , Cricetulus , DNA, Complementary , Endocytosis/physiology , Gene Expression/physiology , Growth Hormone/metabolism , Iodine Radioisotopes , Ligands , Microsomes/physiology , Plasmids , Prolactin/metabolism , Receptors, Somatotropin/agonists , Species Specificity , Transfection
16.
Rev Endocr Metab Disord ; 10(2): 145-56, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18622706

ABSTRACT

Growth hormone (GH) regulates somatic growth, substrate metabolism and body composition. Its actions are elaborated through the GH receptor (GHR). GHR signalling involves the role of at least three major pathways, STATs, MAPK, and PI3-kinase/Akt. GH receptor function can be modulated by changes to the ligand, to the receptor or by factors regulating signal transduction. Insights on the physico-chemical basis of the binding of GH to its receptor and the stoichiometry required for activation of the GH receptor-dimer has led to the development of novel GH agonists and antagonists. Owing to the fact that GH has short half-life, several approaches have been taken to create long-acting GHR agonists. This includes the pegylation, sustained release formulations, and ligand-receptor fusion proteins. Pegylation of a GH analogue (pegvisomant) which binds but not activate signal transduction forms the basis of a new successful approach to the treatment of acromegaly. GH receptors can be regulated at a number of levels, by modifying receptor expression, surface availability and signalling. Insulin, thyroid hormones and sex hormones are among hormones that modulate GHR through some of these mechanisms. Estrogens inhibit GH signalling by stimulating the expression of SOCS proteins which are negative regulators of cytokine receptor signalling. This review of GHR modulators will cover the effects of ligand modification, and of factors regulating receptor expression and signalling.


Subject(s)
Receptors, Somatotropin/physiology , Animals , Estrogens/pharmacology , Gene Expression Regulation/drug effects , Growth Hormone/metabolism , Humans , Insulin/pharmacology , Receptors, Somatotropin/agonists , Receptors, Somatotropin/antagonists & inhibitors , Receptors, Somatotropin/metabolism , Thyroid Hormones/pharmacology
17.
Nat Cell Biol ; 10(6): 740-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18488018

ABSTRACT

The growth and metabolic actions of growth hormone (GH) are believed to be mediated through the GH receptor (GHR) by JAK2 activation. The GHR exists as a constitutive homodimer, with signal transduction by ligand-induced realignment of receptor subunits. Based on the crystal structures, we identify a conformational change in the F'G' loop of the lower cytokine module, which results from binding of hGH but not G120R hGH antagonist. Mutations disabling this conformational change cause impairment of ERK but not JAK2 and STAT5 activation by the GHR in FDC-P1 cells. This results from the use of two associated tyrosine kinases by the GHR, with JAK2 activating STAT5, and Lyn activating ERK1/2. We provide evidence that Lyn signals through phospholipase C gamma, leading to activation of Ras. Accordingly, mice with mutations in the JAK2 association motif respond to GH with activation of hepatic Src and ERK1/2, but not JAK2/STAT5. We suggest that F'G' loop movement alters the signalling choice between JAK2 and a Src family kinase by regulating TMD realignment. Our findings could explain debilitated ERK but not STAT5 signalling in some GH-resistant dwarfs and suggest pathway-specific cytokine agonists.


Subject(s)
Receptors, Somatotropin/agonists , Receptors, Somatotropin/chemistry , Animals , Dimerization , Humans , Janus Kinase 2/metabolism , Mice , Models, Biological , Molecular Conformation , Phospholipase C gamma/metabolism , Protein Conformation , Protein Structure, Tertiary , Rabbits , STAT5 Transcription Factor/metabolism , Signal Transduction , ras Proteins/metabolism
18.
Nat Med ; 13(9): 1108-13, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17721547

ABSTRACT

Cytokine hormones have a short plasma half-life and require frequent administration. For example, growth hormone replacement involves daily injections. In common with other cytokines, the extracellular domain of the growth hormone receptor circulates as a binding protein, which naturally prolongs the biological half-life of growth hormone. Here we have studied the biological actions of a ligand-receptor fusion of growth hormone and the extracellular domain of its receptor. The genetically engineered ligand-receptor fusion protein was purified from mammalian cell culture. In rats, the ligand-receptor fusion had a 300-times reduced clearance as compared to native growth hormone, and a single injection promoted growth for 10 d, far exceeding the growth seen after administration of native growth hormone. The ligand-receptor fusion forms a reciprocal, head-to-tail dimer that provides a reservoir of inactive hormone similar to the natural reservoir of growth hormone and its binding protein. In conclusion, a ligand-receptor fusion of cytokine to its extracellular receptor generates a potent, long-acting agonist with exceptionally slow absorption and elimination. This approach could be easily applied to other cytokines.


Subject(s)
Human Growth Hormone/chemistry , Receptors, Somatotropin/chemistry , Animals , Dimerization , Growth Hormone/chemistry , Growth Hormone/physiology , Humans , Hypophysectomy , Ligands , Models, Molecular , Mutant Proteins/chemistry , Protein Conformation , Rats , Receptors, Somatotropin/agonists , Receptors, Somatotropin/physiology
19.
Mol Endocrinol ; 17(11): 2228-39, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12907755

ABSTRACT

Protein tyrosine phosphatases (PTPs) play key roles in switching off tyrosine phosphorylation cascades, such as initiated by cytokine receptors. We have used substrate-trapping mutants of a large set of PTPs to identify members of the PTP family that have substrate specificity for the phosphorylated human GH receptor (GHR) intracellular domain. Among 31 PTPs tested, T cell (TC)-PTP, PTP-beta, PTP1B, stomach cancer-associated PTP 1 (SAP-1), Pyst-2, Meg-2, and PTP-H1 showed specificity for phosphorylated GHR that had been produced by coexpression with a kinase in bacteria. We then used GH-induced, phosphorylated GH receptor, purified from overexpressing mammalian cells, in a Far Western-based approach to test whether these seven PTPs were also capable of recognizing ligand-induced, physiologically phosphorylated GHR. In this assay, only TC-PTP, PTP1B, PTP-H1, and SAP-1 interacted with the mature form of the phosphorylated GHR. In parallel, we show that these PTPs recognize very different subsets of the seven GHR tyrosines that are potentially phosphorylated. Finally, mRNA tissue distribution of these PTPs by RT-PCR analysis and coexpression of the wild-type PTPs to test their ability to dephosphorylate ligand-activated GHR suggest PTP-H1 and PTP1B as potential candidates involved in GHR signaling.


Subject(s)
Protein Tyrosine Phosphatases/metabolism , Receptors, Somatotropin/agonists , Receptors, Somatotropin/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cell Line , Cricetinae , Growth Hormone/metabolism , Humans , Ligands , Molecular Sequence Data , Organ Specificity , Phosphotyrosine/metabolism , Receptors, Somatotropin/chemistry , Receptors, Somatotropin/genetics , Signal Transduction , Substrate Specificity , Tyrosine/metabolism
20.
Mol Endocrinol ; 17(11): 2240-50, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12907759

ABSTRACT

Monoclonal antibody (MAb) 263 is a widely used monoclonal antibody that recognizes the extracellular domain (ECD) of the GH receptor. It has been shown to act as a GH agonist both in vitro and in vivo, and we report here that it must be divalent to exert its effect on the full-length receptor. To understand the mechanism of its agonist action, we have determined the precise epitope for this antibody using a novel random PCR mutagenesis approach together with expression screening in yeast. A library of 5200 clones of rabbit GH receptor ECD mutants were screened both with MAb 263 and with an anticarboxy-tag antibody to verify complete ECD expression. Sequencing for clones that expressed complete ECD but were not MAb 263 positive identified 20 epitope residues distributed in a discontinuous manner throughout the ECD. The major part of the epitope, as revealed after mapping onto the crystal structure model of the ECD molecule, was located on the side and upper portion of domain 1, particularly within the D-E strand disulfide loop 79-96. Molecular dynamics docking of an antibody of the same isotype as MAb 263 was used to dock the bivalent antibody to the 1528-A2 epitope and to visualize the likely consequences of MAb binding. The minimized model enables the antibody to grasp two receptors in a pincer-like movement from opposite sides, facilitating alignment of the receptor dimerization domains in a manner similar to, but not identical with, GH.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Epitope Mapping , Receptors, Somatotropin/agonists , Receptors, Somatotropin/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibody Specificity , Crystallography, X-Ray , Disulfides/metabolism , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Mice , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Protein Conformation , Rabbits , Receptors, Somatotropin/chemistry , Receptors, Somatotropin/genetics , Sequence Alignment , Tryptophan/genetics
SELECTION OF CITATIONS
SEARCH DETAIL