Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 380
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 114(6): 1359-1364, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28123062

ABSTRACT

The etiology of benign prostatic hyperplasia (BPH) is multifactorial, and chronic inflammation plays a pivotal role in its pathogenesis. Growth hormone-releasing hormone (GHRH) is a hypothalamic neuropeptide that has been shown to act as paracrine/autocrine factor in various malignancies including prostate cancer. GHRH and its receptors are expressed in experimental models of BPH, in which antagonists of GHRH suppressed the levels of proinflammatory cytokines and altered the expression of genes related to epithelial-to-mesenchymal transition (EMT). We investigated the effects of GHRH antagonist on prostatic enlargement induced by inflammation. Autoimmune prostatitis in Balb/C mice was induced by a homogenate of reproductive tissues of male rats. During the 8-wk induction of chronic prostatitis, we detected a progressive increase in prostatic volume reaching 92% at week 8 compared with control (P < 0.001). Daily treatment for 1 mo with GHRH antagonist MIA-690 caused a 30% reduction in prostate volume (P < 0.05). Conditioned medium derived from macrophages increased the average volume of spheres by 82.7% (P < 0.001) and elevated the expression of mRNA for N-cadherin, Snail, and GHRH GHRH antagonist reduced the average volume of spheres stimulated by inflammation by 75.5% (P < 0.05), and TGF-ß2 by 91.8% (P < 0.01). The proliferation of primary epithelial cells stimulated by IL-17A or TGF-ß2 was also inhibited by 124.1% and 69.9%, respectively. GHRH stimulated the growth of BPH-1 and primary prostate spheres. This study provides evidence that GHRH plays important roles in prostatic inflammation and EMT and suggests the merit of further investigation to elucidate the effects of GHRH antagonists in prostatitis and BPH.


Subject(s)
Cell Proliferation/drug effects , Epithelial Cells/drug effects , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Cell Proliferation/genetics , Cells, Cultured , Epithelial Cells/metabolism , Gene Expression/drug effects , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Humans , Male , Mice, Inbred BALB C , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatitis/genetics , Prostatitis/metabolism , Prostatitis/pathology , Rats , Transforming Growth Factor beta2/pharmacology
2.
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
3.
BMC Infect Dis ; 17(1): 551, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28793863

ABSTRACT

BACKGROUND: Aim of this review is to focus the attention on people living with HIV infection at risk of developing a cardiovascular event. What is or what would be the most suitable antiretroviral therapy? Which statin or fibrate to reduce the risk? How to influence behavior and lifestyles? DISCUSSION: Prevention of cardiovascular disease (CVD) risk remains the first and essential step in a medical intervention on these patients. The lifestyle modification, including smoking cessation, increased physical activity, weight reduction, and the education on healthy dietary practices are the main instruments. Statins are the cornerstone for the treatment of hypercholesterolemia. They have been shown to slow the progression or promote regression of coronary plaque, and could also exert an anti-inflammatory and immunomodulatory effect. However the current guidelines for the use of these drugs in general population are dissimilar, with important differences between American and European ones. The debate between American and European guidelines is still open and, also considering the independent risk factor represented by HIV, specific guidelines are warranted. Ezetimibe reduces the intestinal absorption of cholesterol. It is effective alone or in combination with rosuvastatin. It does not modify plasmatic concentrations of antiretrovirals. A number of experimental new classes of drugs for the treatment of hypercholesterolemia are being studied. Fibrates represent the first choice for treatment of hypertriglyceridemia, however, the renal toxicity of fibrates and statins should be considered. Omega 3 fatty acids have a good safety profile, but their efficacy is limited. Another concern is the high dose needed. Other drugs are acipimox and tesamorelin. Current antiretroviral therapies are less toxic and more effective than regimens used in the early years. Lipodistrophy and dyslipidemia are the main causes of long-term toxicities. Not all antiretrovirals have similar toxicities. Protease Inhibitors may cause dyslipidemia and lipodystrophy, while integrase inhibitors have a minimal impact on lipids profile, and no evidence of lipodystrophy. There is still much to be written with the introduction of new drugs in clinical practice. CONCLUSIONS: Cardiovascular risk among HIV infected patients, interventions on behavior and lifestyles, use of drugs to reduce the risk, and switch in antiretroviral therapy, remain nowadays major issues in the management of HIV-infected patients.


Subject(s)
Anti-HIV Agents/adverse effects , Cardiovascular Diseases/prevention & control , Dyslipidemias/chemically induced , HIV Infections/complications , Lipodystrophy/chemically induced , Anti-HIV Agents/therapeutic use , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/etiology , Cholesterol/therapeutic use , Dyslipidemias/complications , Dyslipidemias/prevention & control , Growth Hormone-Releasing Hormone/adverse effects , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/therapeutic use , HIV Infections/drug therapy , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipids , Lipodystrophy/prevention & control , Pyrazines/adverse effects , Pyrazines/therapeutic use , Risk Factors
4.
Proc Natl Acad Sci U S A ; 111(2): 781-6, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24379381

ABSTRACT

The dismal prognosis of malignant brain tumors drives the development of new treatment modalities. In view of the multiple activities of growth hormone-releasing hormone (GHRH), we hypothesized that pretreatment with a GHRH agonist, JI-34, might increase the susceptibility of U-87 MG glioblastoma multiforme (GBM) cells to subsequent treatment with the cytotoxic drug, doxorubicin (DOX). This concept was corroborated by our findings, in vivo, showing that the combination of the GHRH agonist, JI-34, and DOX inhibited the growth of GBM tumors, transplanted into nude mice, more than DOX alone. In vitro, the pretreatment of GBM cells with JI-34 potentiated inhibitory effects of DOX on cell proliferation, diminished cell size and viability, and promoted apoptotic processes, as shown by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide proliferation assay, ApoLive-Glo multiplex assay, and cell volumetric assay. Proteomic studies further revealed that the pretreatment with GHRH agonist evoked differentiation decreasing the expression of the neuroectodermal stem cell antigen, nestin, and up-regulating the glial maturation marker, GFAP. The GHRH agonist also reduced the release of humoral regulators of glial growth, such as FGF basic and TGFß. Proteomic and gene-expression (RT-PCR) studies confirmed the strong proapoptotic activity (increase in p53, decrease in v-myc and Bcl-2) and anti-invasive potential (decrease in integrin α3) of the combination of GHRH agonist and DOX. These findings indicate that the GHRH agonists can potentiate the anticancer activity of the traditional chemotherapeutic drug, DOX, by multiple mechanisms including the induction of differentiation of cancer cells.


Subject(s)
Drug Therapy/methods , Glioblastoma/drug therapy , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/agonists , Peptide Fragments/pharmacology , Animals , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Glial Fibrillary Acidic Protein , Growth Hormone-Releasing Hormone/pharmacology , Immunohistochemistry , Mice , Mice, Nude , Nerve Tissue Proteins/metabolism , Nestin/metabolism , Real-Time Polymerase Chain Reaction
5.
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
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.
Subst Use Misuse ; 51(1): 73-84, 2016 Jan 02.
Article in English | MEDLINE | ID: mdl-26771670

ABSTRACT

BACKGROUND: Communal online folk pharmacology fuels the drive for short cuts in attaining muscle enhancement, fat loss, and youthful skin. OBJECTIVES: The study used "netnography" to explore female use of CJC-1295, a synthetic growth hormone analogue from the perspectives contained in Internet forum activity. METHODS: A systematic Internet search was conducted using variation of the term "CJC-1295"; and combined with "forum." Ninety-six hits related to bodybuilding websites where CJC-1295 was mentioned. Following application of exclusion criteria to confine to female use and evidence of forum activity, 9 sites remained. These were searched internally for reference to CJC-1295. Twenty-three discussion threads relating to female use of CJC-1295 formed the end data set, and analyzed using the Empirical Phenomenological Psychological method. RESULTS: Forum users appeared well versed and experienced in the poly use of performance and image drug supplementation. Choice to use CJC-1295 centered on weight loss, muscle enhancement, youthful skin, improved sleep, and injury healing. Concerns were described relating to female consequences of use given gender variations in growth hormone pulses affecting estimation of dosage, cycling, and long-term consequences. CONCLUSIONS: Public health interventions should consider female self-medicating use of synthetic growth hormone within a repertoire of product supplementation, and related adverse health consequences.


Subject(s)
Growth Hormone-Releasing Hormone/analogs & derivatives , Human Growth Hormone/administration & dosage , Peptide Fragments/administration & dosage , Body Image/psychology , Decision Making , Female , Growth Hormone-Releasing Hormone/administration & dosage , Humans , Self Care
8.
Growth Factors ; 33(2): 160-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25798996

ABSTRACT

Activity and half-life play key roles in the application of GHRH analogues. The GHRH monomers produced in a solid synthesizer were incubated, respectively, in NH4OH solution and lyophilized to obtain their dimers. The activities, specificities, and receptor affinities of the GHRH dimers were evaluated in rGH release/inhibition, rACTH/LH/PRL release, pituitary homogenate binding, and fluorescent staining. Compared to hGHRH(1-44)NH2 (S), PP-hGHRH(1-44)-GGC-CGG-hGHRH(44-1)-PP (2D), P-hGHRH(1-44)-GGC-CGG-hGHRH(44-1)-P (2E), (1)P-hGHRH(2-44)-GGC-CGG-hGHRH(44-2)-(1)P (2F), or hGHRH(1-44)-GGC-CGG-hGHRH(44-1) (2Y) had potency of 104 ± 16.7%, 94 ± 32.6%, 114 ± 16.6%, or 122 ± 14.5% and similar specificities. The inhibition effect of GHIH on rGH stimulated by GHRH dimer was in dose-/time-dependent manner. The staining of FITC-labeled dimer showed cytomembrane distribution and the binding ranking was 2F>2D>2Y>2E>S. 2F presents the strongest activity and the highest affinity to pituitary cells. The dimer with (1)Pro-GHRH stimulates stronger rGH release than that with (1)Tyr-GHRH and the N-terminal single cyclic amino acid is required for the stimulation.


Subject(s)
Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/chemistry , Animals , Cell Membrane/metabolism , Female , Fluorescent Dyes/chemistry , Growth Hormone/metabolism , Growth Hormone-Releasing Hormone/chemical synthesis , Hormones/metabolism , Humans , Ligands , Peptide Fragments/chemistry , Pituitary Gland/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry
9.
Curr Atheroscler Rep ; 17(4): 493, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25702057
10.
J Pharmacokinet Pharmacodyn ; 42(3): 287-99, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25895899

ABSTRACT

The objective of this analysis was to characterize the time course of selected pharmacodynamic (PD) markers of tesamorelin: growth hormone (GH) and insulin-like growth factor (IGF-1) concentrations in HIV-infected patients and healthy volunteers. A total of 41 subjects in Phase I trials receiving subcutaneous daily doses of 1 or 2 mg of tesamorelin during 14 consecutive days were included in this analysis. A previous pharmacokinetic (PK) model of tesamorelin was used as the input function for the PD model of GH. Tesamorelin was hypothesized to stimulate the secretion of GH in an "episodic" manner, i.e., for a finite duration of time. The resulting PK/PD model of GH was used to describe the time course of IGF-1. The effect of age, body weight, body mass index, sex, race, and health status on the model parameters was evaluated. The model was qualified using predictive checks and non-parametric bootstrap. Within the range of the values evaluated no covariates were significantly associated with GH or IGF-1 model parameters. Model evaluation procedures indicated accurate prediction of the selected pharmacodynamic markers. The time course of GH and IGF-1 concentrations following multiple doses of tesamorelin were well predicted by the sequential PK/PD model developed using Phase I data.


Subject(s)
Growth Hormone-Releasing Hormone/analogs & derivatives , HIV Infections/metabolism , Models, Biological , Adult , Dose-Response Relationship, Drug , Female , Growth Hormone-Releasing Hormone/pharmacokinetics , Growth Hormone-Releasing Hormone/therapeutic use , HIV Infections/drug therapy , Healthy Volunteers , Human Growth Hormone/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Male , Middle Aged
11.
Proc Natl Acad Sci U S A ; 109(2): 559-63, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22203988

ABSTRACT

Both cardiac myocytes and cardiac stem cells (CSCs) express the receptor of growth hormone releasing hormone (GHRH), activation of which improves injury responses after myocardial infarction (MI). Here we show that a GHRH-agonist (GHRH-A; JI-38) reverses ventricular remodeling and enhances functional recovery in the setting of chronic MI. This response is mediated entirely by activation of GHRH receptor (GHRHR), as demonstrated by the use of a highly selective GHRH antagonist (MIA-602). One month after MI, animals were randomly assigned to receive: placebo, GHRH-A (JI-38), rat recombinant GH, MIA-602, or a combination of GHRH-A and MIA-602, for a 4-wk period. We assessed cardiac performance and hemodynamics by using echocardiography and micromanometry derived pressure-volume loops. Morphometric measurements were carried out to determine MI size and capillary density, and the expression of GHRHR was assessed by immunofluorescence and quantitative RT-PCR. GHRH-A markedly improved cardiac function as shown by echocardiographic and hemodynamic parameters. MI size was substantially reduced, whereas myocyte and nonmyocyte mitosis was markedly increased by GHRH-A. These effects occurred without increases in circulating levels of growth hormone and insulin-like growth factor I and were, at least partially, nullified by GHRH antagonism, confirming a receptor-mediated mechanism. GHRH-A stimulated CSCs proliferation ex vivo, in a manner offset by MIA-602. Collectively, our findings reveal the importance of the GHRH signaling pathway within the heart. Therapy with GHRH-A although initiated 1 mo after MI substantially improved cardiac performance and reduced infarct size, suggesting a regenerative process. Therefore, activation of GHRHR provides a unique therapeutic approach to reverse remodeling after MI.


Subject(s)
Growth Hormone-Releasing Hormone/analogs & derivatives , Myocardial Infarction/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Sermorelin/analogs & derivatives , Signal Transduction/physiology , Ventricular Remodeling/drug effects , Analysis of Variance , Animals , Cell Proliferation/drug effects , Echocardiography , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Growth Hormone/administration & dosage , Growth Hormone-Releasing Hormone/administration & dosage , Growth Hormone-Releasing Hormone/agonists , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Growth Hormone-Releasing Hormone/pharmacology , Hemodynamics/drug effects , Histological Techniques , Immunohistochemistry , In Situ Nick-End Labeling , Manometry , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Rats , Real-Time Polymerase Chain Reaction , Sermorelin/administration & dosage , Sermorelin/pharmacology
12.
Invest New Drugs ; 32(5): 871-82, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25000999

ABSTRACT

Growth hormone-releasing hormone (GHRH) and its receptors have been implicated in a variety of cellular phenotypes related with tumorigenesis process. Human epidermal growth factor receptor family members (HER) such as EGFR and HER2 are involved in mitogenic signaling pathways implicated in the progression of prostate cancer. We analyzed the cross-talk between GHRH and EGF receptors in prostate cancer. The effects of GHRH in HER signaling were evaluated on human androgen-independent PC3 prostate cancer cells in vitro and GHRH antagonist in vitro and in nude mice xenografts of PC3 prostate cancer. Time-course studies indicated that GHRH had a stimulatory activity on both the expression of EGFR and HER2. GHRH analogues, JMR-132 and JV-1-38, endowed with antagonistic activity for GHRH receptors, abrogated the response to GHRH in PC3 cells. GHRH stimulated a rapid ligand-independent activation of EGFR and HER2 involving at least cAMP/PKA and Src family signaling pathways. GHRH also stimulated a slow ligand-dependent activation of EGFR and HER2 involving an extracellular pathway with an important role for ADAM. Preliminary results also revealed an increase of mRNA for GHRH and GHRH receptor induced by EGF. The inhibition of tumor growth, in vivo, was associated with a substantial reduction in the expression of mRNA and protein levels of EGFR and HER2 in the tumors. GHRH antagonist JV-1-38, significantly decreased the phosphorylated Src levels. The cross-talk between HER and GHRH-R may be impeded by combining drugs acting upon GHRH receptors and HER family members in human advanced prostate cancer.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Sermorelin/analogs & derivatives , Animals , Cell Line, Tumor , Epidermal Growth Factor/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Humans , Male , Mice, Nude , RNA, Messenger/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Sermorelin/pharmacology , src-Family Kinases/metabolism
13.
JAMA ; 312(4): 380-9, 2014.
Article in English | MEDLINE | ID: mdl-25038357

ABSTRACT

IMPORTANCE: Among patients infected with human immunodeficiency virus (HIV), visceral adiposity is associated with metabolic dysregulation and ectopic fat accumulation. Tesamorelin, a growth hormone-releasing hormone analog, specifically targets visceral fat reduction but its effects on liver fat are unknown. OBJECTIVE: To investigate the effect of tesamorelin on visceral and liver fat. DESIGN, SETTING, AND PATIENTS: Double-blind, randomized, placebo-controlled trial conducted among 50 antiretroviral-treated HIV-infected men and women with abdominal fat accumulation at Massachusetts General Hospital in Boston. The first patient was enrolled on January 10, 2011; for the final patient, the 6-month study visit was completed on September 6, 2013. INTERVENTIONS: Participants were randomized to receive tesamorelin, 2 mg (n=28), or placebo (n=22), subcutaneously daily for 6 months. MAIN OUTCOMES AND MEASURES: Primary end points were changes in visceral adipose tissue and liver fat. Secondary end points included glucose levels and other metabolic end points. RESULTS: Forty-eight patients received treatment with study drug. Tesamorelin significantly reduced visceral adipose tissue (mean change, -34 cm2 [95% CI, -53 to -15 cm2] with tesamorelin vs 8 cm2 [95% CI, -14 to 30 cm2] with placebo; treatment effect, -42 cm2 [95% CI, -71 to -14 cm2]; P = .005) and liver fat (median change in lipid to water percentage, -2.0% [interquartile range {IQR}, -6.4% to 0.1%] with tesamorelin vs 0.9% [IQR, -0.6% to 3.7%] with placebo; P = .003) over 6 months, for a net treatment effect of -2.9% in lipid to water percentage. Fasting glucose increased in the tesamorelin group at 2 weeks (mean change, 9 mg/dL [95% CI, 5-13 mg/dL] vs 2 mg/dL [95% CI, -3 to 8 mg/dL] in the placebo group; treatment effect, 7 mg/dL [95% CI, 1-14 mg/dL]; P = .03), but changes at 6 months in fasting glucose (mean change, 4 mg/dL [95% CI, -2 to 10 mg/dL] with tesamorelin vs 2 mg/dL [95% CI, -4 to 7 mg/dL] with placebo; treatment effect, 2 mg/dL [95% CI, -6 to 10 mg/dL]; P = .72 overall across time points) and 2-hour glucose (mean change, -1 mg/dL [95% CI, -18 to 15 mg/dL] vs -8 mg/dL [95% CI, -24 to 8 mg/dL], respectively; treatment effect, 7 mg/dL [95% CI, -16 to 29 mg/dL]; P = .53 overall across time points) were not significant. CONCLUSIONS AND RELEVANCE: In this preliminary study of HIV-infected patients with abdominal fat accumulation, tesamorelin administered for 6 months was associated with reductions in visceral fat and additionally with modest reductions in liver fat. Further studies are needed to determine the clinical importance and long-term consequences of these findings. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT01263717.


Subject(s)
Adiposity/drug effects , Fatty Liver/drug therapy , Growth Hormone-Releasing Hormone/analogs & derivatives , HIV Infections/complications , Intra-Abdominal Fat/drug effects , Abdominal Fat/drug effects , Adult , Double-Blind Method , Fatty Liver/etiology , Female , Growth Hormone-Releasing Hormone/therapeutic use , Humans , Liver/drug effects , Liver/metabolism , Male , Middle Aged
14.
Int J Cancer ; 132(4): 755-65, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-22777643

ABSTRACT

New approaches are needed to the therapy of advanced prostate cancer. This study determined the effect of growth hormone-releasing hormone (GHRH) antagonists, JMR-132 and JV-1-38 on growth of PC3 tumors as well as on angiogenesis and metastasis through the evaluation of various factors that contribute largely to the progression of prostate cancer. Human PC3 androgen-independent prostate cancer cells were injected subcutaneously into nude mice. The treatment with JMR-132 (10 µg/day) or JV-1-38 (20 µg/day) lasted 41 days. We also evaluated the effects of JMR-132 and JV-1-38 on proliferation, cell adhesion and migration in PC-3 cells in vitro. Several techniques (Western blot, reverse transcription polymerase chain reaction, immunohistochemistry, ELISA and zymography) were used to evaluate the expression levels of GHRH receptors and its splice variants, GHRH, vascular endothelial growth factor (VEGF), hypoxia inducible factor (HIF)-1α, metalloproteinases (MMPs) -2 and -9, ß-catenin and E-cadherin. GHRH antagonists suppressed the proliferation of PC-3 cells in vitro and significantly inhibited growth of PC3 tumors. After treatment with these analogues, we found an increase in expression of GHRH receptor accompanied by a decrease of GHRH levels, a reduction in both VEGF and HIF-1α expression and in active forms of MMP-2 and MMP-9, a significant increase in levels of membrane-associated ß-catenin and a significant decline in E-cadherin. These results support that the blockade of GHRH receptors can modulate elements involved in angiogenesis and metastasis. Consequently, GHRH antagonists could be considered as suitable candidates for therapeutic trials in the management of androgen-independent prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Neoplasm Metastasis/drug therapy , Neovascularization, Pathologic/drug therapy , Prostatic Neoplasms/drug therapy , Sermorelin/analogs & derivatives , Animals , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Growth Hormone-Releasing Hormone/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Prostatic Neoplasms/pathology , Random Allocation , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Sermorelin/pharmacology , Vascular Endothelial Growth Factors/metabolism , Xenograft Model Antitumor Assays , beta Catenin/metabolism
15.
Methods ; 56(2): 230-5, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21871962

ABSTRACT

For most peptide hormones prohibited in elite sports the concentrations in plasma or urine are very low (pg/mL). Accordingly, hyphenated purification and enrichment steps prior to mass spectrometric detection are required to obtain sufficient doping control assays. Immunoaffinity purification in combination with nano-scale liquid chromatography coupled to high resolution/high accuracy mass spectrometry was found to have the potential of providing the necessary sensitivity and unambiguous specificity to produce reliable results. With the presented methodology 12 prohibited peptides (porcine insulin, Novolog, Apidra, Lantus DesB30-32 metabolite, Humalog and human insulin, Synacthen (synthetic ACTH analogue), luteinizing hormone-releasing hormone (LH-RH), growth hormone releasing hormone (GH-RH(1-29)) and CJC-1295 (GH-RH analogue), LongR(3)-IGF-1 and IFG-1) were simultaneously purified from plasma/serum or urine. With limits of detection for each target compound ranging in the low pg/mL level (urine), the method enables the determination of urinary peptides at physiologically relevant concentrations. For each class of peptides an appropriate antibody and a respective internal standard was implemented ensuring robust analysis conditions. Due to the fast and simple sample preparation procedure (∼25 samples per day) and the fact that all materials are commercial available, the implementation of the methodology to laboratories from other analytical fields (forensics, pharmacokinetic sciences, etc.) is enabled.


Subject(s)
Chromatography, Affinity/methods , Immunoassay/methods , Peptide Hormones/isolation & purification , Substance Abuse Detection/methods , Amino Acid Sequence , Chromatography, Affinity/standards , Doping in Sports , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/blood , Growth Hormone-Releasing Hormone/isolation & purification , Growth Hormone-Releasing Hormone/urine , Humans , Immunoassay/standards , Insulin Aspart/blood , Insulin Aspart/urine , Mass Spectrometry/methods , Mass Spectrometry/standards , Molecular Sequence Data , Peptide Fragments/blood , Peptide Fragments/isolation & purification , Peptide Fragments/urine , Peptide Hormones/blood , Peptide Hormones/urine , Performance-Enhancing Substances/blood , Performance-Enhancing Substances/isolation & purification , Performance-Enhancing Substances/urine , Reference Values , Reproducibility of Results , Sensitivity and Specificity , Time Factors
16.
Proc Natl Acad Sci U S A ; 107(43): 18611-5, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20937882

ABSTRACT

Despite the well-documented action of growth hormone-releasing hormone (GHRH) on the stimulation of production and release of growth hormone (GH), the effects of GHRH in peripheral tissues are incompletely explored. In this study, we show that GHRH plays a role in wound healing and tissue repair by acting primarily on wound-associated fibroblasts. Mouse embryonic fibroblasts (MEFs) in culture and wound-associated fibroblasts in mice expressed a splice variant of the receptors for GHRH (SV1). Exposure of MEFs to 100 nM and 500 nM GHRH or the GHRH agonist JI-38 stimulated the expression of α-smooth muscle actin (αSMA) based on immunoblot analyses as well as the expression of an αSMA-ß-galactosidase reporter transgene in primary cultures of fibroblasts isolated from transgenic mice. Consistent with this induction of αSMA expression, results of transwell-based migration assays and in vitro wound healing (scratch) assays showed that both GHRH and GHRH agonist JI-38 stimulated the migration of MEFs in vitro. In vivo, local application of GHRH or JI-38 accelerated healing in skin wounds of mice. Histological evaluation of skin biopsies showed that wounds treated with GHRH and JI-38 were both characterized by increased abundance of fibroblasts during the early stages of wound healing and accelerated reformation of the covering epithelium at later stages. These results identify another function of GHRH in promoting skin tissue wound healing and repair. Our findings suggest that GHRH may have clinical utility for augmenting healing of skin wounds resulting from trauma, surgery, or disease.


Subject(s)
Growth Hormone-Releasing Hormone/agonists , Growth Hormone-Releasing Hormone/pharmacology , Wound Healing/drug effects , Actins/genetics , Animals , Cell Movement/drug effects , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/physiology , Gene Expression/drug effects , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Wound Healing/genetics , Wound Healing/physiology
17.
Proc Natl Acad Sci U S A ; 107(6): 2604-9, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20133784

ABSTRACT

Whether the growth hormone (GH)/insulin-like growth factor 1(IGF-1) axis exerts cardioprotective effects remains controversial; and the underlying mechanism(s) for such actions are unclear. Here we tested the hypothesis that growth hormone-releasing hormone (GHRH) directly activates cellular reparative mechanisms within the injured heart, in a GH/IGF-1 independent fashion. After experimental myocardial infarction (MI), rats were randomly assigned to receive, during a 4-week period, either placebo (n = 14), rat recombinant GH (n = 8) or JI-38 (n = 8; 50 microg/kg per day), a potent GHRH agonist. JI-38 did not elevate serum levels of GH or IGF-1, but it markedly attenuated the degree of cardiac functional decline and remodeling after injury. In contrast, GH administration markedly elevated body weight, heart weight, and circulating GH and IGF-1, but it did not offset the decline in cardiac structure and function. Whereas both JI-38 and GH augmented levels of cardiac precursor cell proliferation, only JI-38 increased antiapoptotic gene expression. The receptor for GHRH was detectable on myocytes, supporting direct activation of cardiac signal transduction. Collectively, these findings demonstrate that within the heart, GHRH agonists can activate cardiac repair after MI, suggesting the existence of a potential signaling pathway based on GHRH in the heart. The phenotypic profile of the response to a potent GHRH agonist has therapeutic implications.


Subject(s)
Cardiotonic Agents/pharmacology , Growth Hormone-Releasing Hormone/agonists , Growth Hormone/pharmacology , Myocardial Infarction/prevention & control , Animals , Blotting, Western , Body Weight/drug effects , Echocardiography , Female , Growth Hormone/blood , Growth Hormone/genetics , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Heart/drug effects , Heart/physiopathology , Hemodynamics/drug effects , Immunohistochemistry , Insulin-Like Growth Factor I/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Organ Size/drug effects , Random Allocation , Rats , Rats, Inbred F344 , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Recombinant Proteins/pharmacology
18.
Clin Infect Dis ; 54(11): 1642-51, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22495074

ABSTRACT

BACKGROUND: Tesamorelin, a growth hormone-releasing hormone analogue, decreases visceral adipose tissue (VAT) by 15%-20% over 6-12 months in individuals with human immunodeficiency virus (HIV)-associated abdominal adiposity, but it is unknown whether VAT reduction is directly associated with endocrine and metabolic changes. METHODS: In 2 phase III, randomized, double-blind studies, men and women with HIV-associated abdominal fat accumulation were randomly assigned (ratio, 2:1) to receive tesamorelin or placebo for 26 weeks. At week 26, patients initially receiving tesamorelin were randomly assigned to continue receiving tesamorelin or to receive placebo for an additional 26 weeks. In per-protocol analysis of 402 subjects initially randomly assigned to receive tesamorelin, those with ≥8% reduction in VAT were defined a priori as responders per the statistical analysis plan. Post hoc analyses were performed to assess differences between responders and nonresponders. RESULTS: Compared with tesamorelin nonresponders, responders experienced greater mean (±SD) reduction in triglyceride levels (26 weeks: -0.6 ± 1.7 mmol/L vs -0.1 ± 1.2 mmol/L [P = .005]; 52 weeks: -0.8 ± 1.8 mmol/L vs 0.0 ± 1.1 mmol/L [P = .003]) and attenuated changes in fasting glucose levels (26 weeks: 1 ± 16 mg/dL vs 5 ± 14 mg/dL [P = .01]; 52 weeks: -1 ± 14 mg/dL vs 8 ± 17 mg/dL [P < .001]), hemoglobin A1c levels (26 weeks: 0.1 ± 0.3% vs 0.3 ± 0.4% [P < .001]; 52 weeks: 0.0 ± 0.3% vs 0.2 ± 0.5% [P = .003]), and other parameters of glucose homeostasis. Similar patterns were seen for adiponectin levels, with significant improvement in responders vs nonresponders. Changes in lipid levels and glucose homeostasis were significantly associated with percentage change in VAT. CONCLUSIONS: In contrast to nonresponders, HIV-infected patients receiving tesamorelin with ≥8% reduction in VAT have significantly improved triglyceride levels, adiponectin levels, and preservation of glucose homeostasis over 52 weeks of treatment. CLINICALTRIALS.GOV REGISTRATION: NCT00123253, NCT00435136, NCT00608023.


Subject(s)
Adiposity/drug effects , Anti-HIV Agents/adverse effects , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/administration & dosage , HIV Infections/drug therapy , HIV-Associated Lipodystrophy Syndrome/chemically induced , HIV-Associated Lipodystrophy Syndrome/prevention & control , Adolescent , Adult , Aged , Anti-HIV Agents/administration & dosage , Double-Blind Method , Female , HIV-Associated Lipodystrophy Syndrome/diagnosis , Humans , Male , Metabolome , Middle Aged , Placebos/administration & dosage , Treatment Outcome , Young Adult
19.
Neuroendocrinology ; 96(1): 81-8, 2012.
Article in English | MEDLINE | ID: mdl-22377963

ABSTRACT

Experimental data indicate that antagonists of growth hormone-releasing hormone (GHRH) could be used clinically in disorders characterized by excessive GHRH/growth hormone (GH) secretion, but direct evidence for the effectiveness of GHRH antagonists on human pituitary tissue is still lacking. In this study, we investigated the inhibitory effect of our GHRH antagonists MZ-4-71 and JV-1-36 and the somatostatin (SST) analog RC-160 on superfused pituitary cells obtained from a human GH-secreting adenoma. Using Western blot analysis and immunohistochemistry, we demonstrated profuse expression of the GHRH receptor and its major splice variant SV1 and an increase in the expression of Gsa protein in the adenoma tissue. Exposure of the tumor cells to exogenous pulses of GHRH induced definite GH responses, causing a 3- to 5-fold elevation of the basal GH level. The antagonists MZ-4-71 and JV-1-36 did not alter basal GH secretion, indicating that the adenoma cells did not secrete GHRH in an autocrine manner. However, both antagonists prevented the stimulatory effect of exogenous GHRH. Similarly to the GHRH antagonists, neither SST-14 nor the SST analog RC-160 had an effect on the basal GH secretion of the tumor cells, but both peptides inhibited the stimulatory effect of exogenous GHRH, with RC-160 being more potent than SST. Our study provides direct evidence for the effectiveness of potent GHRH antagonists such as MZ-4-71 and JV-1-36 on human pituitary GH-secreting adenoma tissue and strongly suggests that these drugs could be used for therapy of GHRH-associated forms of acromegaly, particularly for those patients in whom surgery fails or is not an option.


Subject(s)
Growth Hormone-Releasing Hormone/antagonists & inhibitors , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Hormone Antagonists/pharmacology , Pituitary Neoplasms/metabolism , Acromegaly/metabolism , Adult , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/pharmacology , Humans , Male , Sermorelin/analogs & derivatives , Sermorelin/pharmacology
20.
Ann Pharmacother ; 46(2): 240-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22298602

ABSTRACT

OBJECTIVE: To evaluate the efficacy and safety of tesamorelin, a growth hormone releasing factor analogue approved by the Food and Drug Administration in November 2010 for the treatment of lipodystrophy associated with HIV infection. DATA SOURCES: Literature was obtained through MEDLINE (1948-November 2011) and International Pharmaceutical Abstracts (1970-October 2011) using the search terms tesamorelin, TH9507, growth hormone releasing factor, and HIV-associated lipodystrophy syndrome. Additional publications were obtained through review of references within primary literature publications as well as pertinent Web sites. STUDY SELECTION AND DATA EXTRACTION: All articles published in English identified from the data sources were evaluated and all pertinent information was included. All studies relevant to the evaluation of efficacy and safety of tesamorelin in the management of HIV-associated lipodystrophy were included, with a focus on trials completed in humans. DATA SYNTHESIS: In 2 Phase 3 clinical trials and their pooled analyses, tesamorelin was proven to significantly decrease waist circumference and visceral adipose tissue (VAT) following 26 weeks of treatment. Both trials also demonstrated significant improvements in some subjective body image parameters. Both studies had 26-week extension phases that confirmed maintenance of VAT improvements on treatment without adverse impact on blood glucose and lipid parameters. Limited data support off-label uses of tesamorelin at this time. CONCLUSIONS: Tesamorelin is effective in improving visceral adiposity and body image in patients with HIV-associated lipodystrophy over 26-52 weeks of treatment. Potential limitations for its use include high cost and lack of long-term safety and adherence data. Tesamorelin provides a useful treatment option for management of patients with significant lipodystrophy related to HIV infection.


Subject(s)
Growth Hormone-Releasing Hormone/analogs & derivatives , HIV-Associated Lipodystrophy Syndrome/drug therapy , Animals , Drug Costs , Growth Hormone-Releasing Hormone/economics , Growth Hormone-Releasing Hormone/pharmacokinetics , Growth Hormone-Releasing Hormone/therapeutic use , HIV-Associated Lipodystrophy Syndrome/economics , Humans , Off-Label Use
SELECTION OF CITATIONS
SEARCH DETAIL