Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Cell Physiol ; 236(12): 8197-8207, 2021 12.
Article in English | MEDLINE | ID: mdl-34224586

ABSTRACT

Age-related diseases such as cardiovascular diseases portend disability, increase health expenditures, and cause late-life mortality. Synthetic agonists of growth hormone-releasing hormone (GHRH) exhibit several favorable effects on heart function and remodeling. Here we assessed whether GHRH agonist MR409 can modulate heart function and systemic parameters in old mice. Starting at the age of 15 months, mice were injected subcutaneously with MR409 (10 µg/day, n = 8) or vehicle (n = 7) daily for 6 months. Mice treated with MR409 showed improvements in exercise activity, cardiac function, survival rate, immune function, and hair growth in comparison with the controls. More stem cell colonies were grown out of the bone marrow recovered from the MR409-treated mice. Mitochondrial functions of cardiomyocytes (CMs) from the MR409-treated mice were also significantly improved with more mitochondrial fusion. Fewer ß-gal positive cells were observed in endothelial cells after 10 passages with MR409. In Doxorubicin-treated H9C2 cardiomyocytes, cell senescence marker p21 and reactive oxygen species were significantly reduced after cultured with MR409. MR409 also improved cellular ATP production and oxygen consumption rate in Doxorubicin-treated H9C2 cells. Mitochondrial protein OPA1 long isoform was significantly increased after treatment with MR409. The effects of MR409 were mediated by GHRH receptor and protein kinase A (PKA). In short, GHRH agonist MR409 reversed the aging-associated changes with respect of heart function, mobility, hair growth, cellular energy production, and senescence biomarkers. The improvement of heart function may be related to a better mitochondrial functions through GHRH receptor/cAMP/PKA/OPA1 signaling pathway and relieved cardiac inflammation.


Subject(s)
Endothelial Cells/drug effects , Myocytes, Cardiac/drug effects , Peptide Fragments/pharmacology , Receptors, Neuropeptide/agonists , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/metabolism , Growth Hormone-Releasing Hormone/metabolism , Mice , Myocytes, Cardiac/metabolism , Signal Transduction/drug effects
2.
Endocrinology ; 160(7): 1600-1612, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31070727

ABSTRACT

In this article, we briefly review the identification of GHRH, provide an abridged overview of GHRH antagonists, and focus on studies with GHRH agonists. Potent GHRH agonists of JI and MR class were synthesized and evaluated biologically. Besides the induction of the release of pituitary GH, GHRH analogs promote cell proliferation and exert stimulatory effects on various tissues, which express GHRH receptors (GHRH-Rs). A large body of work shows that GHRH agonists, such as MR-409, improve pancreatic ß-cell proliferation and metabolic functions and facilitate engraftment of islets after transplantation in rodents. Accordingly, GHRH agonists offer a new therapeutic approach to treating diabetes. Various studies demonstrate that GHRH agonists promote repair of cardiac tissue, producing improvement of ejection fraction and reduction of infarct size in rats, reduction of infarct scar in swine, and attenuation of cardiac hypertrophy in mice, suggesting clinical applications. The presence of GHRH-Rs in ocular tissues and neuroprotective effects of GHRH analogs in experimental diabetic retinopathy indicates their possible therapeutic applications for eye diseases. Other effects of GHRH agonists, include acceleration of wound healing, activation of immune cells, and action on the central nervous system. As GHRH might function as a growth factor, we examined effects of GHRH agonists on tumors. In vitro, GHRH agonists stimulate growth of human cancer cells and upregulate GHRH-Rs. However, in vivo, GHRH agonists inhibit growth of human cancers xenografted into nude mice and downregulate pituitary and tumoral GHRH-Rs. Therapeutic applications of GHRH analogs are discussed. The development of GHRH analogs should lead to their clinical use.


Subject(s)
Diabetes Mellitus/drug therapy , Growth Hormone-Releasing Hormone/metabolism , Neoplasms/drug therapy , Receptors, Neuropeptide/agonists , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Animals , Cell Line, Tumor , Humans , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism
3.
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
4.
Eur J Pharm Sci ; 117: 341-350, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29526766

ABSTRACT

Extra-hypothalamic growth hormone-releasing hormone (GHRH) plays an important role in infertility. The female infertility models were formed by intraperitoneally injecting cyclophosphamide in 5-week-old Chinese hamster once in a week for 5 weeks. All the models mated with healthy male hamster in the ratio of 1:1 in the experimental 6-8th week and the couples were separated to breed in the 9-10th week. 20 mg/kg of cyclophosphamide induced temporary interference of reproduction and did not cause significant difference in the weight of body, bilateral ovaries, or liver. By intramuscularly injecting twice in a week during the experimental 4-10th week, 2, 4, 8 mg/kg of Grin induced 30, 42.9, 60% of total pregnancy rates in a dose-dependent manner whereas 200 U/kg of hMG induced 50% of total pregnancy rates. The single cyclophosphamide dose caused strongly eosinophilic ovarian cells, scattered early follicles, many atretic follicles, and no corpora luteum was observed. The hMG group individually presents many follicles at all levels, especially secondary ones in the ovarian cortex and medulla. Much of loose connective tissue, vacuoles, and sparse interstitial cells distribute in the medulla. Grin induced many follicles at all dose levels and corpora lutea in the cortex, and the compactly aligned interstitial cells occurred in the whole ovarian tissue. The less TUNEL staining and higher expression of ki67 showed the proliferation and protection effect of Grin on ovarian cells. Grin obviously promotes fertility by up-regulating ovarian GHRH receptor and strengthening the development and maturation of follicles without triggering central and ovarian GH secretion.


Subject(s)
Fertility Agents, Female/administration & dosage , Fertility/drug effects , Growth Hormone-Releasing Hormone/administration & dosage , Infertility, Female/drug therapy , Ovarian Follicle/drug effects , Ovary/drug effects , Receptors, Neuropeptide/agonists , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Animals , Cricetulus , Cyclophosphamide , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Growth Hormone/metabolism , Growth Hormone-Releasing Hormone/analogs & derivatives , Infertility, Female/chemically induced , Infertility, Female/metabolism , Infertility, Female/physiopathology , Injections, Intramuscular , Male , Ovarian Follicle/metabolism , Ovarian Follicle/physiopathology , Ovary/metabolism , Ovary/physiopathology , Pregnancy , Pregnancy Rate , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Signal Transduction/drug effects , Time Factors , Up-Regulation
5.
Proc Natl Acad Sci U S A ; 113(50): 14396-14401, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911838

ABSTRACT

Retinoblastoma (RB) is the most common intraocular cancer in children worldwide. Current treatments mainly involve combinations of chemotherapies, cryotherapies, and laser-based therapies. Severe or late-stage disease may require enucleation or lead to fatality. Recently, RB has been shown to arise from cone precursor cells, which have high MDM2 levels to suppress p53-mediated apoptosis. This finding leads to the hypothesis that restoring apoptosis mechanisms in RBs could specifically kill the cancer cells without affecting other retinal cells. We have previously reported involvement of an extrapituitary signaling pathway of the growth hormone-releasing hormone (GHRH) in the retina. Here we show that the GHRH receptor (GHRH-R) is highly expressed in RB cells but not in other retinal cells. We induced specific apoptosis with two different GHRH-R antagonists, MIA-602 and MIA-690. Importantly, these GHRH-R antagonists do not trigger apoptosis in other retinal cells such as retinal pigmented epithelial cells. We delineated the gene expression profiles regulated by GHRH-R antagonists and found that cell proliferation genes and apoptotic genes are down- and up-regulated, respectively. Our results reveal the involvement of GHRH-R in survival and proliferation of RB and demonstrate that GHRH-R antagonists can specifically kill the RB cells.


Subject(s)
Apoptosis/drug effects , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Pituitary Hormone-Regulating Hormone/antagonists & inhibitors , Retinal Neoplasms/drug therapy , Retinoblastoma/drug therapy , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Humans , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Retinoblastoma/metabolism , Retinoblastoma/pathology , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Signal Transduction/drug effects , Transcriptome
6.
Pharmacol Res ; 111: 859-866, 2016 09.
Article in English | MEDLINE | ID: mdl-27480202

ABSTRACT

Despite the great clinical significance of radiation-induced cardiac damage, experimental investigation of its mechanisms is an unmet need in medicine. Beneficial effects of growth hormone-releasing hormone (GHRH) agonists in regeneration of the heart have been demonstrated. The aim of this study was the evaluation of the potential of modern GHRH agonistic analogs in prevention of radiation damage in an in vitro cardiac myocyte-based model. Cultures of cardiac myocytes isolated from newborn rats (NRVM) were exposed to a radiation dose of 10Gy. The effects of the agonistic analogs, JI-34 and MR-356, of human GHRH on cell viability, proliferation, their mechanism of action and the protein expression of the GHRH/SV1 receptors were studied. JI-34 and MR-356, had no effect on cell viability or proliferation in unirradiated cultures. However, in irradiated cells JI-34 showed protective effects on cell viability at concentrations of 10 and 100nM, and MR-356 at 500nM; but no such protective effect was detected on cell proliferation. Both agonistic analogs decreased radiation-induced ROS level and JI-34 interfered with the activation of SAFE/RISK pathways. Using Western blot analysis, a 52kDa protein isoform of GHRHR was detected in the samples in both irradiated and unirradiated cells. Since GHRH agonistic analogs, JI-34 and MR-356 alleviated radiation-induced damage of cardiac myocytes, they should be tested in vivo as potential protective agents against radiogenic heart damage.


Subject(s)
Alprostadil/analogs & derivatives , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/agonists , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/radiation effects , Peptide Fragments/pharmacology , Radiation-Protective Agents/pharmacology , Alprostadil/pharmacology , Animals , Animals, Newborn , Cardiotoxicity , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Cytoprotection , Dose-Response Relationship, Drug , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects
7.
Arterioscler Thromb Vasc Biol ; 36(4): 663-672, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26868211

ABSTRACT

OBJECTIVE: The efficiency of cell therapy is limited by poor cell survival and engraftment. Here, we studied the effect of the growth hormone-releasing hormone agonist, JI-34, on mesenchymal stem cell (MSC) survival and angiogenic therapy in a mouse model of critical limb ischemia. APPROACH AND RESULTS: Mouse bone marrow-derived MSCs were incubated with or without 10(-8) mol/L JI-34 for 24 hours. MSCs were then exposed to hypoxia and serum deprivation to detect the effect of preconditioning on cell apoptosis, migration, and tube formation. For in vivo tests, critical limb ischemia was induced by femoral artery ligation. After surgery, mice received 50 µL phosphate-buffered saline or with 1×10(6) MSCs or with 1×10(6) JI-34-reconditioned MSCs. Treatment of MSCs with JI-34 improved MSC viability and mobility and markedly enhanced their capability to promote endothelial tube formation in vitro. These effects were paralleled by an increased phosphorylation and nuclear translocation of signal transducer and activator of transcription 3. In vivo, JI-34 pretreatment enhanced the engraftment of MSCs into ischemic hindlimb muscles and augmented reperfusion and limb salvage compared with untreated MSCs. Significantly more vasculature and proliferating CD31(+) and CD34(+) cells were detected in ischemic muscles that received MSCs treated with JI-34. CONCLUSIONS: Our studies demonstrate a novel role for JI-34 to markedly improve therapeutic angiogenesis in hindlimb ischemia by increasing the viability and mobility of MSCs. These findings support additional studies to explore the full potential of growth hormone-releasing hormone agonists to augment cell therapy in the management of ischemia.


Subject(s)
Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/agonists , Ischemia/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Muscle, Skeletal/blood supply , Peptide Fragments/pharmacology , Active Transport, Cell Nucleus , Animals , Antigens, CD34/metabolism , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Hindlimb , Ischemia/metabolism , Ischemia/physiopathology , Male , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Neovascularization, Physiologic , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , STAT3 Transcription Factor/metabolism , Time Factors
8.
Oncotarget ; 6(12): 9728-39, 2015.
Article in English | MEDLINE | ID: mdl-25797248

ABSTRACT

BACKGROUND: We previously showed that growth hormone-releasing hormone (GHRH) agonists are cardioprotective following myocardial infarction (MI). Here, our aim was to evaluate the in vitro and in vivo activities of highly potent new GHRH agonists, and elucidate their mechanisms of action in promoting cardiac repair. METHODS AND RESULTS: H9c2 cells were cultured in serum-free medium, mimicking nutritional deprivation. GHRH agonists decreased calcium influx and significantly improved cell survival. Rats with cardiac infarction were treated with GHRH agonists or placebo for four weeks. MI size was reduced by selected GHRH agonists (JI-38, MR-356, MR-409); this accompanied an increased number of cardiac c-kit+ cells, cellular mitotic divisions, and vascular density. One week post-MI, MR-409 significantly reduced plasma levels of IL-2, IL-6, IL-10 and TNF-α compared to placebo. Gene expression studies revealed favorable outcomes of MR-409 treatment partially result from inhibitory activity on pro-apoptotic molecules and pro-fibrotic systems, and by elevation of bone morphogenetic proteins. CONCLUSIONS: Treatment with GHRH agonists appears to reduce the inflammatory responses post-MI and may consequently improve mechanisms of healing and cardiac remodeling by regulating pathways involved in fibrosis, apoptosis and cardiac repair. Patients with cardiac dysfunction could benefit from treatment with novel GHRH agonists.


Subject(s)
Heart Failure/drug therapy , Myocardial Infarction/drug therapy , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/chemistry , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Receptors, Pituitary Hormone-Regulating Hormone/chemistry , Alprostadil/analogs & derivatives , Alprostadil/chemistry , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Gene Expression Regulation , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/chemistry , Humans , Inflammation , Interleukin-10/blood , Interleukin-2/blood , Interleukin-6/blood , Microscopy, Fluorescence , Mitosis , Rats , Sermorelin/analogs & derivatives , Sermorelin/chemistry , Tumor Necrosis Factor-alpha/blood
9.
Proc Natl Acad Sci U S A ; 111(48): 17260-5, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25404316

ABSTRACT

The beneficial effects of agonists of growth hormone-releasing hormone receptor (GHRH-R) in heart failure models are associated with an increase in the number of ckit(+) cardiac stem cells (CSCs). The goal of the present study was to determine the presence of GHRH-R in CSCs, the effect of GHRH-R agonists on their proliferation and survival, and the mechanisms involved. We investigated the expression of GHRH-R in CSCs of different species and the effect of GHRH-R agonists on their cell proliferation and survival. GHRH-R is expressed in ckit(+) CSCs isolated from mouse, rat, and pig. Treatment of porcine CSCs with the GHRH-R agonist JI-38 significantly increased the rate of cell division. Similar results were observed with other GHRH-R agonists, MR-356 and MR-409. JI-38 exerted a protective effect on survival of porcine CSCs under conditions of oxidative stress induced by exposure to hydrogen peroxide. Treatment with JI-38 before exposure to peroxide significantly reduced cell death. A similar effect was observed with MR-356. Addition of GHRH-R agonists to porcine CSCs induced activation of ERK and AKT pathways as determined by increased expression of phospho-ERK and phospho-AKT. Inhibitors of ERK and AKT pathways completely reversed the effect of GHRH-R agonists on CSC proliferation. Our findings extend the observations of the expression of GHRH-R by CSCs and demonstrate that GHRH-R agonists have a direct effect on proliferation and survival of CSCs. These results support the therapeutic use of GHRH-R agonists for stimulating endogenous mechanisms for myocardial repair or for preconditioning of stem cells before transplantation.


Subject(s)
Cell Proliferation/drug effects , Growth Hormone-Releasing Hormone/analogs & derivatives , Myocardium/cytology , Receptors, Neuropeptide/agonists , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Stem Cells/drug effects , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Growth Hormone-Releasing Hormone/pharmacology , HeLa Cells , Humans , MCF-7 Cells , Mice , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Signal Transduction/drug effects , Stem Cells/metabolism , Swine
10.
Endocrinology ; 154(9): 3305-18, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23825127

ABSTRACT

Botulinum neurotoxin is a potent inhibitor of acetylcholine secretion and acts by cleaving members of the soluble N-ethylmaleimide-sensitive factor-attachment protein receptor family, which are critical to exocytotic vesicular secretion. However, the potential of botulinum neurotoxin for treating secretory disease is limited both by its neural selectivity and the necessity for direct injection into the relevant target tissue. To circumvent these limitations, a technology platform called targeted secretion inhibitors (TSIs) is being developed. TSIs are derived from botulinum neurotoxin but are retargeted to specific cell types to inhibit aberrant secretion. A TSI called qGHRH-LHN/D, with a GHRH receptor targeting domain and designed to specifically inhibit pituitary somatotroph GH release through cleavage of the N-ethylmaleimide-sensitive factor-attachment protein receptor protein, vesicle-associated membrane protein (VAMP), has recently been described. Here we show this TSI activates GHRH receptors in primary cultured rat pituicytes is internalized into these cells, depletes VAMP-3, and inhibits phorbol-12-myristate-13-acetate-induced GH secretion. In vivo studies show that this TSI, but not one with an inactive catalytic unit, produces a dose-dependent inhibition of pulsatile GH secretion, thus confirming its mechanism of action through VAMP cleavage. Selectivity of action has been shown by the lack of effect of this TSI in vivo on secretion from thyrotrophs, corticotrophs, and gonadotrophs. In the absence of suitable in vivo models, these data provide proof of concept for the use of somatotroph-targeted TSIs in the treatment of acromegaly and moreover raise the potential that TSIs could be used to target other diseases characterized by hypersecretion.


Subject(s)
Botulinum Toxins/pharmacology , Down-Regulation/drug effects , Growth Hormone-Releasing Hormone/pharmacology , Growth Hormone/metabolism , Pituitary Gland, Anterior/drug effects , Receptors, Neuropeptide/agonists , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Recombinant Fusion Proteins/pharmacology , Animals , Biological Transport/drug effects , Botulinum Toxins/chemistry , Botulinum Toxins/genetics , Botulinum Toxins/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Growth Hormone/blood , Growth Hormone-Releasing Hormone/chemistry , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Male , Organ Specificity , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Periodicity , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Protein Interaction Domains and Motifs , Proteolysis/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/chemistry , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Somatotrophs/cytology , Somatotrophs/drug effects , Somatotrophs/metabolism , Vesicle-Associated Membrane Protein 3/antagonists & inhibitors , Vesicle-Associated Membrane Protein 3/metabolism
11.
J Endocrinol ; 195(3): 525-36, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18000314

ABSTRACT

In this study, two novel GHRHR receptor splice variants, named chicken GHRHR-v1 (cGHRHR-v1) and cGHRHR-v2 respectively, were identified from chicken pituitary using RT-PCR assay. cGHRHR-v1 is characterized by an N-terminal deletion of 36 amino acid residues, including an aspartate at position 56 (Asp(56)) conserved in G protein-coupled receptor B-I subfamily. cGHRHR-v2 is a carboxyl-terminal truncated receptor variant with four putative transmembrane domains, which arose from alternative use of a splice acceptor site on intron 8. Using the pGL3-CRE-luciferase reporter system, the functionality of the two variants was examined in Chinese hamster ovary cells. cGHRHR-v1 was shown to be capable of transmitting signal upon agonist stimulation, but cGHRHR-v2 could not. Both GHRH and pituitary adenylate cyclase-activating peptide (PACAP) could activate cGHRHR-v1 at high dosages (GHRH >/=10(-8) M; PACAP >/=10(-6) M) and GHRH was much more potent than PACAP, suggesting that cGHRHR-v1 is a functional membrane-spanning receptor with an impairment in high-affinity ligand binding, rather than in receptor activation and ligand-binding specificity. This finding also points out the possibility that Asp(56) is not a critical determinant for receptor activation and direct ligand-receptor interaction. To substantiate this hypothesis, using site-directed mutagenesis, two receptor mutants with replacement of Asp(56) by Ala or Gly were generated. Expectedly, chicken or human GHRH could still activate both receptor mutants with reduced potencies (about 2- to 14-fold less potent). Taken together, our findings not only suggest that cGHRHR variants may play a role in controlling normal pituitary functions, but also support that Asp(56) is nonessential for receptor activation and direct ligand-receptor interaction.


Subject(s)
Chickens/genetics , Chickens/metabolism , Protein Isoforms , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Amino Acid Sequence , Animals , Aspartic Acid , Base Sequence , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Humans , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pituitary Gland/metabolism , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/drug effects , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Receptors, Pituitary Hormone-Regulating Hormone/drug effects , Sheep , Signal Transduction
12.
Endocrine ; 14(1): 95-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11322508

ABSTRACT

One of the most active topics in the growth hormone-IGF-1 field is that of the so-called growth hormone secretagogues (GHS). At a time when the isolation of GHRH had not occurred, the GHS were developed as artificial tools to release GH. The interest in these groups of compounds was rekindled when it was realized that they were not surrogates of GHRH nor were they acting through the modulation of the release of either GHRH or somatostatin. With the subsequent cloning of the specific receptor of GHS, and today of the natural ligand for that receptor, named ghrelin, it soon become clear that GHS and the GHS-receptor were part of a new physiological system involved in GH regulation. The dual control of GH secretion became a trinity. GHS releases GH when administered by any route--oral, iv, sc, and even transdermally-with a surprising potency and reproductivity. In addition, GHS when administered together with GHRH exert a synergistic action on GH secretion and that combined administration is the most potent GH releaser to date. Clinical studies have demonstrated that the GHS-GHRH administration may be considered the new "gold standard" test of GH reserve in humans, as the GH secretion so elicited is not altered by gender, adiposity, or age. The combined administration of GHRH plus GHS is able to discriminate between healthy subjects and patients with adult GH deficiency, suggesting a considerable utility in the clinical setting.


Subject(s)
Growth Hormone-Releasing Hormone , Growth Hormone/deficiency , Growth Hormone/metabolism , Hormones , Oligopeptides , Peptide Hormones , Receptors, Neuropeptide/agonists , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Adult , Child , Clinical Trials as Topic , Drug Synergism , Female , Ghrelin , Humans , Male , Peptides/genetics , Peptides/physiology , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/physiology , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/physiology
13.
Pharmacol Toxicol ; 88(2): 81-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11169166

ABSTRACT

We have investigated the effect of short-term preexposure of growth hormone-releasing hormone (GHRH) on the subsequent response to GHRH in a baby hamster kidney (BHK) cell line expressing the human GHRH receptor and in primary rat pituitary cells. In the BHK cells the receptor was rapidly desensitised in a homologous fashion. Preexposure with agents directly stimulating the cAMP pathway like forskolin and db-cAMP had no effect. In rat pituitary cells we also observed a rapid desensitisation of the GHRH response in an apparently homologous fashion. In both systems the desensitisation was dose-dependent with no change in the potency of the hormone in a subsequent stimulation, only the efficacy was decreased. In the rat pituitary cell, the response measured as growth hormone release was more sensitive to the agonist-induced desensitisation than the cAMP response. No indication of depletion of growth hormone (GH) stores was seen. In rat pituitary cells, contrary to observations in BHK cells, preexposure with both forskolin and db-cAMP desensitised a subsequent growth hormone-releasing hormone stimulation, indicating a heterologous desensitisation. Phorbol-12-myristate 13-acetate (PMA), on the other hand, had no effect. In the baby hamster kidney cells it was demonstrated that the GHRH receptor surface expression decreased following preexposure with GHRH. This phenomenon was observed only in whole cells suggesting a rapid internalisation process. Together, these data indicate that after short-term GHRH preexposure, both in a human and rat system, the following GHRH response is desensitised. In BHK cells this desensitisation is strictly homologous. In rat pituitary cells, on the other hand, the desensitisation is a mixed homologous/ heterologous type.


Subject(s)
Down-Regulation , Kidney/drug effects , Pituitary Gland/drug effects , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Sermorelin/pharmacology , Adenylyl Cyclases/metabolism , Animals , Animals, Newborn , Bucladesine/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Colforsin/pharmacology , Cricetinae , Dose-Response Relationship, Drug , Humans , Kidney/cytology , Kidney/enzymology , Male , Pituitary Gland/cytology , Pituitary Gland/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide/agonists , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Tetradecanoylphorbol Acetate/pharmacology , Transfection
14.
Endocrine ; 9(1): 71-7, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9798733

ABSTRACT

Effect of growth hormone-releasing peptide-2 (GHRP-2) on ovine somatotrophs is abolished by a growth hormone-releasing factor (GRF) receptor antagonist, which raises the possibility that GHRP-2 may act on GRF receptors. In the present study, we used rat pituitary GC cells with or without stable transfection of cDNA coding for the human GRF receptor (GC/R+ or GC/R-) to determine whether or not GHRP-2 acts via the GRF receptor. Northern blot analysis indicated that GRF receptor mRNA was undetectable in GC/R-cells, whereas a high level of expression occurred in GC/R+ cells that were transfected by GRF receptor cDNA. In GC/R- cells, incubation with up to 10(-7)M of either hGRF or GHRP-2 did not alter the intracellular cAMP, [Ca2+]i, or GH secretion. In GC/R+ cells, hGRF (10(-11)-10(-7)M) increased cAMP levels in a concentration-dependent manner up to 20-fold. This increase in cAMP levels was blocked by a GRF receptor antagonist, [Ac-Tyr1, D-Arg2]-GRF 1-29, but not by a Ca2+ channel blocker, NiCl2 (0.5 mM). GH secretion and [Ca2+]i were, however, not increased by hGRF. Incubation of the transfected cells with 10(-1)-10(-8)MGH RP-2 did not modify intracellular cAMP levels. This result suggests that GHRP-2 does not act through the GRF receptor.


Subject(s)
Growth Hormone-Releasing Hormone/physiology , Oligopeptides/pharmacology , Pituitary Gland/physiology , Receptors, Neuropeptide/physiology , Receptors, Pituitary Hormone-Regulating Hormone/physiology , Animals , Base Sequence , Cell Line , Cells, Cultured , Humans , Molecular Sequence Data , Rats , Receptors, Neuropeptide/agonists , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Signal Transduction/drug effects , Transfection
15.
Neuroendocrinology ; 64(3): 169-76, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8875434

ABSTRACT

To assess the efficacy of a potent agonist analog of GH-releasing hormone (GH-RH), [Dat1,Gln8,Orn12,21,Abu15,Nle27,Asp28,A gm29]hGH-RH(1-29) (JI-38), we investigated the effects of its chronic administration on growth responses in monosodium glutamate (MSG)-lesioned and normal young rats. Body weight (BW), body length (BL), tibia length (TIL), and tail length (TAL) were monitored. Basal serum GH concentrations, GH responses to bolus injections of GH-RH, pituitary GH and serum IGF-I concentrations were measured by RIA. Pituitary GH-RH receptor concentration and binding affinity was also evaluated after the treatment. Neonatal treatment with MSG resulted, as expected, in blunted growth and a decrease in serum and pituitary GH concentration and serum IGF-I levels. A reduction in GH-RH receptor concentration, associated with increased binding affinity of the GH-RH receptor was also found. Chronic administration of GH-RH agonist JI-38 in doses of 2 micrograms at 12-hour intervals for 2 weeks markedly increased the GH responsiveness to GH-RH and stimulated growth, with MSG-treated animals achieving the growth rate of normal controls. Acceleration of growth was associated with stimulated GH synthesis and IGF-I secretion, although basal serum GH levels did not change. Pituitary GH-RH receptor concentration and binding affinity were not significantly modified by the treatment. Treatment of normal young growing rats with agonist JI-38 did not further increase the normal growth acceleration in these rats, but stimulated the GH synthesis and augmented the GH secretory responsiveness. The treatment of MSG-lesioned rats with GH-RH agonist was generally more effective in female than in male animals, and in some cases masked the sex differences in growth rate. Our findings provide the first evidence that the blunted growth rate of the MSG-lesioned rats is associated with a decreased pituitary GH-RH receptor concentration. Our work demonstrates that administration of GH-RH agonist JI-38 is able to restore the normal growth rate of the GH-deficient rats by stimulating GH synthesis and IGF-I secretion.


Subject(s)
Growth Disorders/drug therapy , Growth Hormone-Releasing Hormone/pharmacology , Growth Hormone/deficiency , Receptors, Neuropeptide/agonists , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Animals , Body Height/drug effects , Body Weight/drug effects , Drug Administration Schedule , Drug Evaluation, Preclinical , Female , Growth Hormone/metabolism , Growth Hormone-Releasing Hormone/analogs & derivatives , Insulin-Like Growth Factor I/metabolism , Male , Pituitary Gland/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Reference Values , Secretory Rate/drug effects , Sodium Glutamate , Tibia/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...