Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Biotech Histochem ; 93(6): 463-470, 2018.
Article in English | MEDLINE | ID: mdl-29693452

ABSTRACT

Many types of aflatoxin cause problems for both public and animal health. Aflatoxin B1 (AFB1) is the most toxic and commonly encountered fungal toxin that appears in poultry feed and in feeds stored under unsuitable conditions. AFB1 decreases feed quality, egg production and fertility of hatching eggs. Also, AFB1 alters the development of embryos by infecting eggs. We investigated using sequence analysis the changes caused by different concentrations of AFB1 on the promoter sequences of the growth hormone regulated gene-1 (GHRG-1) in chick embryo at 13, 17, 19 and 21 days incubation. DNA isolated from the liver of chick embryos treated with different concentrations of AFB1 was separated using agarose gel electrophoresis to detect apoptosis, and DNA interaction with AFB1 was investigated using plasmids to detect changes in electrophoretic mobility and their effects on DNA. Base changes of the promoter sequences of GHRG-1 in 5 ng/egg, 15 ng/egg and 40 ng/egg doses of AFB1 were increased on day 19 compared to base changes of the same AFB1 doses on day 13. We also found that AFB at different concentrations changed the mobility of DNA by binding to it, and that high doses of AFB1 destroyed DNA. The DNA interaction study using plasmid demonstrated that AFB1 at high doses was bound to plasmid DNA, slowed its mobility and inhibited restriction cuts.


Subject(s)
Aflatoxin B1/pharmacology , DNA/drug effects , Growth Hormone/drug effects , Liver/drug effects , Animal Feed , Animals , Chick Embryo , Chickens , DNA/metabolism , Growth Hormone-Releasing Hormone/drug effects , Growth Hormone-Releasing Hormone/metabolism , Liver/embryology
2.
Med Sport Sci ; 59: 36-46, 2012.
Article in English | MEDLINE | ID: mdl-23075553

ABSTRACT

The secretion of growth hormone (GH) is regulated through a complex neuroendocrine control system, especially by the functional interplay of two hypothalamic hormones, GH-releasing hormone and somatostatin. These hormones are subject to modulation by a host of neurotransmitters and are the final mediators of endocrine and neural influences for GH secretion. Interest in the possible role of γ-aminobutyric acid (GABA) in the control of GH secretion began decades ago. However, interest in its role as an ergogenic aid is only recent. It is well accepted that GABAergic neurons are found in the hypothalamus and recent evidence suggests its secretion within the pituitary itself. Inhibition of GABA degradation and blockade of GABA transmission as well as administration of GABA and GABA mimetic drugs have all been shown to affect GH secretion. However, there are many controversial findings. The effects may depend on the site of action within the hypothalamic-pituitary unit and the hormonal milieu. Experimental and clinical evidence support the presence of a dual action of GABA - one mediated centrally, the other exerted directly at the pituitary level. The two sites of action may be responsible for excitatory and inhibitory effects of GABA on GH secretion. This chapter will outline the anatomical basis for possible influences of GABA on GH secretion and present evidence for a role of GABA in the control of GH release by actions at either hypothalamic or pituitary sites. The potential ergogenic benefits of oral GABA supplementation will also be discussed.


Subject(s)
Dietary Supplements , GABA Agents/therapeutic use , Human Growth Hormone/drug effects , Pituitary Gland, Anterior/drug effects , gamma-Aminobutyric Acid/therapeutic use , Growth Hormone-Releasing Hormone/drug effects , Growth Hormone-Releasing Hormone/metabolism , Human Growth Hormone/metabolism , Humans , Hypothalamo-Hypophyseal System , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pituitary Gland, Anterior/metabolism , Receptors, GABA/drug effects , Receptors, GABA/metabolism
3.
Fundam Clin Pharmacol ; 23(3): 339-44, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19527301

ABSTRACT

To investigate the effect of chronic oral arginine aspartate on the growth hormone (GH), GH-releasing hormone (GHRH), insulin-like growth factor-1 (IGF-1) and IGF-binding protein-3 (IGFBP-3) secretions in healthy volunteers. Twenty-three healthy non-athlete volunteer males were administered arginine aspartate (30 g) orally once daily at 21:00 h for 21 consecutive days. Subjects were hospitalized on days 0, 1, 3, 5, 7, 14 and 21 of treatment. At each hospitalization, concentrations of GHRH, GH, IGF-1 and IGFBP-3 were measured over 4 h after arginine aspartate intake. GH, IGF-1 and IGFBP-3 concentrations were also determined over 12 h at days 0, 1 and 21. Compared with day 1, 4 h GH levels dropped at day 5 and subsequently rose to levels not significantly different from initial ones. The latter was substantiated by 12 h GH levels that did not significantly change from days 1 to 21. GHRH levels were not statistically different, although there was a trend in median values that seemed to inversely mirror those of GH. This dynamic over the course of the study for GH and GHRH was accompanied by a general decrease in IGF-1 and IGFBP-3. In healthy volunteers, a chronic oral treatment with 30 g/day arginine aspartate is followed by a decrease in IGF-1 and IGFBP-3 secretions.


Subject(s)
Arginine/pharmacology , Aspartic Acid/pharmacology , Insulin-Like Growth Factor Binding Protein 3/drug effects , Insulin-Like Growth Factor I/drug effects , Administration, Oral , Adult , Arginine/administration & dosage , Aspartic Acid/administration & dosage , Drug Administration Schedule , Growth Hormone/drug effects , Growth Hormone/metabolism , Growth Hormone-Releasing Hormone/drug effects , Growth Hormone-Releasing Hormone/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Time Factors , Young Adult
4.
Oncol Rep ; 20(5): 1289-94, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18949435

ABSTRACT

GHRH antagonists have been shown to inhibit growth of various human cancer cell lines xenografted into nude mice including estrogen receptor negative human breast cancers. Previous observations also suggest that GHRH locally produced in diverse neoplasms including breast cancer might directly affect proliferation of tumor cells. In the present study we demonstrate that a novel highly potent GHRH antagonist JMR-132 strongly inhibits the proliferation of both estrogen receptor negative SKBR 3 and estrogen receptor positive ZR 75 human breast cancer cell lines in vitro. The proliferation in vitro of ZR 75 and SKBR 3 was increased after direct stimulation with GHRH(1-29)NH2. The GHRH antagonist JMR-132 had a significant antiproliferative activity in the absence of GHRH and nullified the proliferative effect of GHRH in these cell lines. SKBR 3 and ZR 75 expressed the GHRH ligand as well as the pituitary type of GHRH-receptor, which likely appears to mediate the antiproliferative mechanisms in these cell lines. These in vitro results suggest that JMR-132 is a potent inhibitor of breast cancer growth, independent of the estrogen receptor status. Further investigations on the combination treatment with endocrine agents affecting the estrogen pathway and GRHR antagonists are needed in order to improve the treatment of breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Receptors, Estrogen/metabolism , Sermorelin/analogs & derivatives , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Growth Hormone-Releasing Hormone/biosynthesis , Growth Hormone-Releasing Hormone/drug effects , Humans , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sermorelin/pharmacology
5.
J Psychiatr Res ; 42(12): 991-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18226817

ABSTRACT

Acute administration of cortisol increases non-rapid-eye movement (non-REM) sleep, suppresses rapid-eye movement (REM) sleep and stimulates growth hormone (GH) release in healthy subjects. This study investigates whether cortisol has similar endocrine and electrophysiological effects in patients with depression who typically show a pathological overactivity of the hypothalamus-pituitary-adrenal (HPA) system. Fifteen depressed inpatients underwent the combined dexamethasone/corticotropin-releasing hormone test followed by three consecutive sleep EEG recordings in which the patients received placebo (saline) and hourly injections of cortisol (1mg/KG BW). Cortisol increased duration and intensity of non-REM sleep in particular in male patients and stimulated GH release. The activity of the HPA axis appeared to influence the cortisol-induced effects on non-REM sleep and GH levels. Stimulation of delta sleep was less pronounced in patients with dexamethasone nonsuppression. In contrast, REM sleep parameters were not affected by the treatment. These data demonstrate that the non-REM sleep-promoting effects of acute cortisol injections observed in healthy controls could be replicated in patients with depression. Our results suggest that non-REM and REM sleep abnormalities during the acute state of the disease are differentially linked to the activity of the HPA axis.


Subject(s)
Depressive Disorder, Major/physiopathology , Human Growth Hormone/blood , Hydrocortisone/pharmacology , Sleep/drug effects , Adult , Aged , Delta Rhythm/drug effects , Delta Rhythm/statistics & numerical data , Depressive Disorder, Major/blood , Dexamethasone/pharmacology , Electroencephalography/drug effects , Female , Growth Hormone-Releasing Hormone/blood , Growth Hormone-Releasing Hormone/drug effects , Growth Hormone-Releasing Hormone/physiology , Human Growth Hormone/physiology , Humans , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiopathology , Male , Middle Aged , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiopathology , Polysomnography/drug effects , Polysomnography/statistics & numerical data , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/physiology , Sleep/physiology , Sleep Stages/drug effects , Sleep Stages/physiology , Sleep, REM/drug effects , Sleep, REM/physiology
6.
J Pharmacol Sci ; 104(4): 319-28, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17666866

ABSTRACT

N(omega)-Nitro-L-arginine methyl ester (L-NAME) induces a pre-eclampsia-like syndrome in pregnant rats. We have previously reported the anti-hypertensive effects of several Japanese traditional (Kampo) medicines in this model, and one of these, Tokishakuyakusan (TS), also improved intrauterine growth retardation (IUGR). In the present study, we characterized the effect of TS on IUGR. TS administration reversed the decrease in fetal body weight and fetal blood glucose concentration induced by the infusion of L-NAME. Growth hormone (GH) levels in the fetal blood, which were decreased by L-NAME infusion, were also significantly elevated by TS; however, levels of GH releasing hormone (GHRH) and insulin-like growth factor I (IGF-I) were unchanged and only slightly changed, respectively. Treatment with L-NAME with or without TS had no apparent effect on GH, GHRH, and IGF-I levels of dams. In an immunocytochemical study, the number of GH-positive cells in the fetal pituitary gland was significantly increased in TS-treated rats. These data suggest that enhanced proliferation of somatotrope cells of the pituitary gland and the resultant increase in GH secretion in the fetus may be involved in the improvement of IUGR by TS.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Fetal Growth Retardation/drug therapy , Pre-Eclampsia/drug therapy , Animals , Blood Glucose/drug effects , Disease Models, Animal , Female , Fetal Weight/drug effects , Growth Hormone/drug effects , Growth Hormone-Releasing Hormone/drug effects , Insulin-Like Growth Factor I/drug effects , Male , NG-Nitroarginine Methyl Ester , Pituitary Gland/cytology , Pituitary Gland/drug effects , Placenta/drug effects , Placentation , Plants, Medicinal/chemistry , Pre-Eclampsia/chemically induced , Pregnancy , Rats , Rats, Sprague-Dawley
7.
Exp Biol Med (Maywood) ; 231(8): 1373-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16946406

ABSTRACT

The enzyme dipeptidyl peptidase-IV (DPP-IV) inactivates a variety of bioactive peptides, including glucagon-like peptide-1 (GLP-1) and growth hormone releasing hormone (GHRH). Inhibiting DPP-IV in order to increase circulating GLP-1 is of interest as a treatment for Type II diabetes. Inactivation of DPP-IV may also increase circulating GHRH, potentially enhancing growth in domestic animals. To test the hypothesis that inhibition of DPP-IV activity will influence the growth hormone/ IGF-1 axis, growing pigs (Sus scrofa domesticus, 78 kg) were treated with a DPP-IV inhibitor (Compound 1, the 2,5-difluor-ophenyl analog of the triazolopiperazine MK0431, sitagliptin), and plasma concentrations of IGF-1 were monitored. Pigs were administered either sterile saline (0.11 ml/kg followed by a continuous infusion at 2 ml/hr for 72 hrs, controls, n = 2), Compound 1 (2.78 mg/kg followed by a continuous infusion at 0.327 mg/kg x hr for 72 hrs, n = 4) or GHRH (0.11 ml/kg sterile saline, followed by a continuous infusion of GHRH at 2.5 microg/ kg x hr for 48 hrs, n = 4). Plasma concentrations of Compound 1 were maintained at 1 microM, which resulted in a 90% inhibition of circulating DPP-IV activity. Relative to the predose 24-hr period, area under the IGF-1 concentration curve (AUC) tended to be lower (P = 0.062) with Compound 1 (.79 +/- 130 ng/ml x hr) than controls (543 +/- 330 ng/ml x hr). GHRH treatment increased the IGF-1 AUC (1210 +/- 160 ng/ml x hr, P = 0.049 vs. controls and P = 0.001 vs. Compound 1). We conclude that inhibition of DPP-IV does not alter the circulating levels of IGF-1 in the growing pig.


Subject(s)
Cathepsin C/antagonists & inhibitors , Growth Hormone-Releasing Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Animals , Area Under Curve , Cathepsin C/blood , Cathepsin C/drug effects , Enzyme Inhibitors/pharmacology , Growth Hormone-Releasing Hormone/drug effects , Insulin-Like Growth Factor I/drug effects , Male , Pyrazines/pharmacology , Sitagliptin Phosphate , Swine , Triazoles/pharmacology
8.
J Mol Neurosci ; 22(3): 179-88, 2004.
Article in English | MEDLINE | ID: mdl-14997011

ABSTRACT

The effect of exogenous growth hormone (GH) and insulin-like growth factor I (IGF-I) on brain IGF-I binding sites (IGF-IR), and on the levels of growth hormone-releasing hormone (GHRH) and somatostatin was studied in hypophysectomized and intact juvenile male rats. Animals were injected subcutaneously twice daily (n = 5 each) with recombinant GH (rGH) (2.5 U/kg per day) or rIGF-I (500 microg/kg per day). In the hypophysectomized rats, serum GH and IGF-I levels were markedly suppressed and IGF-I levels were partially restored by GH treatment. There was a significant increase in IGF-IR binding capacity in the IGF-I-treated hypophysectomized rats compared to the saline-treated hypophysectomized animals (150.61 +/- 45.66 vs 41.32 +/- 12.42 fmol/mg, p < 0.05) but no significant difference in IGF-IR mRNA levels. GHRH levels in the saline-treated hypophysectomized group were significantly lower than in the saline-treated intact rats (31.2 +/- 11.2 vs 140.6 +/- 48.1 pg/mg tissue, respectively, p < 0.01); no effect was induced by GH or IGF-I (37.5 +/- 26.8 and 53.8 +/- 22.5 pg/mg tissue, respectively). However, in the intact rats, GH and IGF-I injection led to a decrease in GHRH content, which was significant in the GH-treated compared to the saline-treated animals (33.1 +/- 16.2 vs 140.6 +/- 48.1 pg/mg tissue, p < 0.01). No difference was found in somatostatin levels between intact and hypophysectomized rats (631.2 +/- 81.2 and 625.0 +/- 62.5 pg/mg tissue, respectively). However, in the hypophysectomized animals, GH and IGF-I treatment induced a significant increase in somatostatin levels (1300 +/- 193.7 pg/mg tissue, p < 0.01, and 912.5 +/- 81.2 pg/mg tissue, p < 0.05, respectively). Our findings suggest that the bioavailability of exogenous IGF-I is greater than that of GH-stimulated endogenous IGF-I. Because IGF-I is a potent neurotrophic agent, this effect may have important implications for states of neurodegenerative diseases.


Subject(s)
Growth Hormone-Releasing Hormone/metabolism , Growth Hormone/pharmacology , Hypothalamus/drug effects , Insulin-Like Growth Factor I/pharmacology , Somatostatin/metabolism , Age Factors , Animals , Binding Sites/drug effects , Binding Sites/physiology , Body Weight/drug effects , Body Weight/genetics , Growth Hormone/blood , Growth Hormone-Releasing Hormone/drug effects , Hypophysectomy , Hypothalamus/growth & development , Hypothalamus/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Organ Size/drug effects , Organ Size/genetics , Rats , Rats, Wistar , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 1/genetics , Recombinant Fusion Proteins/pharmacology , Somatostatin/drug effects
9.
Neuroendocrinology ; 77(6): 380-7, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12845224

ABSTRACT

Testosterone (T) is known to affect the growth hormone (GH) axis. However, the mechanisms underlying the activation of GH secretion by T still remain to be clarified. Available data in animals and humans have shown that withdrawal of somatostatin (SRIH) infusion induces a GH-releasing hormone (GHRH)-mediated rebound release of GH, and there is accumulating evidence that SRIH infusion withdrawal may be a useful test to probe the GHRH function in vivo. With the aim of investigating whether the stimulatory effect of androgens on GH release in man could be accounted for by activation of the hypothalamic GHRH tone, we evaluated the plasma GH response to SRIH withdrawal in 10 patients aged 29.6 +/- 2.4 years (mean +/- SEM), diagnosed with hypergonadotropic hypogonadism, before and after a 6-month replacement therapy with T enanthate (250 mg every 3 weeks, i.m.), and in 10 healthy men, aged 26.7 +/- 2.8 years. To verify whether the modulation of GH secretion by T could also be mediated through changes in SRIH tone and/or pituitary releasable pool, we examined GH secretory responses to combined GHRH and L-arginine (ARG) in the same individuals. Basal plasma concentrations of GH (0.48 +/- 0.11 microg/l) and IGF-I (23.79 +/- 1.83 nmol/l) were significantly lower in untreated hypogonadal patients than in healthy men, and significantly increased after T replacement therapy (GH 1.13 +/- 0.28 microg/l; IGF-I 28.71 +/- 1.46 nmol/l). The mean Delta GH peak after SRIH withdrawal recorded in untreated hypogonadal men (2.65 +/- 0.86 microg/l) was significantly (p < 0.05) lower than that observed in healthy men (6.53 +/- 1.33 microg/l) and significantly increased after T replacement therapy (5.52 +/- 1.25 microg/l). The GH responses to GHRH combined with ARG (a functional SRIH antagonist) were not significantly different between healthy men and untreated hypogonadal patients, and were not significantly affected by T treatment. Plasma T and estradiol (E(2)) levels significantly correlated with Delta GH peak after SRIH withdrawal in healthy men and in T-treated hypogonadal patients, whereas in untreated patients they did not. No significant correlation was found between GH areas under the curve after GHRH + ARG test and T and E(2) plasma levels in either healthy men or in hypogonadal patients (both before and after T replacement). These findings are consistent with the view that in humans the stimulatory action of T on the GH axis appears to be mediated at the hypothalamic level primarily by promoting GHRH function.


Subject(s)
Growth Hormone-Releasing Hormone/metabolism , Growth Hormone/metabolism , Hypogonadism/metabolism , Hypothalamus/metabolism , Pituitary Gland/metabolism , Testosterone/administration & dosage , Testosterone/metabolism , Adult , Area Under Curve , Arginine/administration & dosage , Arginine/metabolism , Case-Control Studies , Estradiol/blood , Growth Hormone/blood , Growth Hormone/drug effects , Growth Hormone-Releasing Hormone/administration & dosage , Growth Hormone-Releasing Hormone/drug effects , Heptanoates/administration & dosage , Heptanoates/metabolism , Humans , Hypogonadism/drug therapy , Hypothalamus/drug effects , Immunoradiometric Assay , Insulin-Like Growth Factor I/metabolism , Male , Pituitary Gland/drug effects , Radioimmunoassay , Somatostatin/metabolism
10.
Gerontology ; 49(3): 191-5, 2003.
Article in English | MEDLINE | ID: mdl-12679611

ABSTRACT

BACKGROUND: The growth hormone (GH) secretion declines by 14% with each decade of adult life. Several attempts have been made to reverse the manifestations of the senile GH deficiency, termed somatopause, but GH substitution treatment in old age has not yet developed an established regimen. Cholinesterase inhibitors like pyridostigmine are able to elicit GH secretion when administered alone and to enhance the GH response to growth hormone releasing hormone (GHRH), but its clinical use is limited due to the strong peripheral cholinergic side effects. OBJECTIVE: The aims of our experiments were to find out whether the GH response to GHRH can be augmented by rivastigmine, a new orally applicable and well-tolerated selective inhibitor of cerebral acetylchoinesterase. METHODS: Eight healthy volunteers (age range 65-69 years) were studied. After an overnight fast, GHRH tests were done: 1 microg/kg GHRH was injected as an intravenous bolus. Blood samples for an immunoradiometric GH assay were taken at the time of GHRH injection (time 0) and after 15, 30, 45, 60, and 120 min. First, the baseline experiment was done: it consisted of two subsequent GHRH tests which were carried out within an interval of 120 min. Four weeks later the rivastigmine experiment was done identically, but 60 min before performing the second GHRH test, rivastigmine (4.5 mg) was administered orally. The GH secretory responses were expressed as areas under the curve (AUC; median, interquartile range), Wilcoxon's signed-rank test was used for statistical comparisons. RESULTS: Baseline experiment: The GH AUC of the first GHRH test was 1040 (range 420-1250) ng/ml/h. The repeated GHRH stimulation after 120 min (second GHRH test) showed a 13-fold decrease to 80 (range 60-130) ng/ml/h. Rivastigmine experiment: The GH AUC of the first GHRH test was 950 (range 540-1430) ng/ml/h and, therefore, similar to that of the baseline experiment. 60 min after ingestion of the single oral dose of rivastigmine (4.5 mg), the following GHRH stimulation (second GHRH test) nearly doubled the GH AUC to 1580 (range 860-3330) ng/ml/h. Comparing the DeltaGH AUC values (DeltaGH AUC = GH AUC of the first GHRH test minus GH AUC of the second GHRH test), baseline experiment versus rivastigmine experiment, there was a 20-fold (p = 0.018) increase in GH AUC after rivastigmine pretreatment. CONCLUSIONS: Rivastigmine is a powerful drug to enhance GH release to repeated GHRH stimulation in healthy elderly human subjects. Future investigations are necessary to find out whether rivastigmine can restore the senile decline of the circadian GH secretion in the long term and, therefore, has new implications for patient treatment.


Subject(s)
Carbamates/pharmacology , Cholinesterase Inhibitors/pharmacology , Growth Hormone-Releasing Hormone/drug effects , Growth Hormone/drug effects , Phenylcarbamates , Aged , Alzheimer Disease/drug therapy , Female , Growth Hormone/biosynthesis , Growth Hormone-Releasing Hormone/physiology , Humans , Male , Rivastigmine
11.
Ginecol Obstet Mex ; 69: 379-85, 2001 Oct.
Article in Spanish | MEDLINE | ID: mdl-11816525

ABSTRACT

BACKGROUND: Great interest has sparked recently the role that plays the changes that the growth hormone undergoes in the menopausal woman, specially its involvement in the central nervous, cardiovascular, genitourinary, digestive and osteomuscular systems. OBJECTIVE: To evaluate the influence of transdermal administration of 17-beta estradiol on growth hormone secretion in menopausal women before and after treatment under the stimulus of growth-hormonereleasing hormone (GH-RH). MATERIAL AND METHODS: We studied 5 patients with a mean age of 51 +/- 4.1 yr. with clinical and biochemical evidence of menopause. Evolution time 5.4 +/- 4.61 (range: 1-13 yr.). We monitored the pulsatility of GH during the first 120 minutes and 3 hours after the administration of the GHRH-1-29-NH2, i.v. bolus (50 micrograms). There were obtained every 15 minutes for the determination of GH levels before and after the stimulus. Immediately thereafter hormone replacement therapy was initiated with transdermal beta-estradiol with 50 micrograms patches twice a week. Clinical evaluations and hormone dynamics with OHRH-1-29 were performed at baseline and at 1,3 and 6 months from the start of therapy as described previously. RESULTS: GH pulsatility before estrogen replacement therapy (ERT) in these 5 patients was: X: 0.48 +/- 0.22, 0.38 +/- 0.17, 0.45 +/- 0.25 and 0.29 (at baseline, 1, 3 and 6 months respectively) and 2.74 +/- o 1.21; 3.48 +/- 1.32 (p > 0.05) 4.91 +/- 1.57 (p < 0.05) and 6.04 +/- 1.69 (p < 0.05) (p in relation to baseline) post stimulus with GH-RH-1-29 at baseline 1, 3 and 6 months respectively after transdermal estrogen therapy. Gonadotrophins basal serum levels fall from X: 54.68 +/- 27 to 33.20 +/- 11.23 and 40.48 +/- 12 to 28.30 +/- 6.70 (FSH and LH respectively). Estradiol serum level were from 1.82 +/- 4.06 to 25.95 +/- 5.96 before and after treatment, respectively. COMMENTS AND CONCLUSIONS: These results demonstrate that transdermal estrogen therapy does not modify the pulsatility of growth hormone but it does increase the magnitude of response to the stimulus with GH-RH-1-29 proportional to the time of treatment. We consider that this tendency to increase the production of growth hormone could be explained by an endogenous deficit of growth hormone releasing hormone due to a number of factors including the lack of adequate estrogen serum levels in menopausal women. More investigations will be needed to support this hypothesis and to bring forth a new understanding of menopause and its treatment.


Subject(s)
Climacteric , Estradiol/administration & dosage , Growth Hormone-Releasing Hormone/drug effects , Growth Hormone-Releasing Hormone/metabolism , Administration, Cutaneous , Adult , Female , Humans , Middle Aged
12.
Alcohol ; 18(2-3): 109-22, 1999.
Article in English | MEDLINE | ID: mdl-10456561

ABSTRACT

When alcohol is a large proportion of daily nutrient energy, the network of signals for energy homeostasis appears to adapt with abnormal patterns of sleep and growth hormone (GH) release along with gradual acquisition of an addictive physical dependency on alcohol. Early relapse during treatment of alcoholism is associated with a lower GH response to challenge, perhaps reflecting an altered balance of somatostatin (SS) to somatropin releasing hormone (GHRH) that also affects slow wave sleep (SWS) in dependent patients. Normal patterns of sleep have progressively shorter SWS episodes and longer rapid eye movement (REM) episodes during the overall sleep period, but the early sleep cycles of alcoholics have truncated or non-existent SWS episodes, and the longer REM episodes occur in early cycles. During SWS delta wave activity, the hypothalamus releases GHRH, which causes the pituitary to release GH. Alcohol-dependent patients have lower levels of SWS power and GH release than normal subjects, and efforts to understand the molecular basis for this maladaptation and its relation to continued alcohol dependence merit encouragement. More needs to be learned about the possibility of decreasing alcohol dependency by increasing SWS or enhancing GHRH action.


Subject(s)
Alcohol Drinking/adverse effects , Alcoholism/physiopathology , Growth Hormone-Releasing Hormone/drug effects , Human Growth Hormone/drug effects , Sleep/drug effects , Adult , Aged , Female , Humans , Male , Middle Aged
13.
Biol Psychiatry ; 45(3): 334-9, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-10023511

ABSTRACT

BACKGROUND: According to the existence in anorexia nervosa (AN) of peripheral growth hormone (GH) resistance, low circulating insulinlike growth factor I (IGF-I) levels may be coupled with GH hypersecretion; however, there is also evidence for alterations in the neural control of GH secretion. In fact, reportedly GH secretion is partially refractory to the inhibitory effect of muscarinic cholinergic antagonists as well as to the stimulatory effect of muscarinic cholinergic agonists, which act via opposite modulation of hypothalamic somatostatin (SS) release. Thus, somatostatinergic activity could be impaired in AN. This could be due to an impaired hypothalamic SS release or, alternatively, an altered somatotroph sensitivity to SS. METHODS: We studied in 10 women with AN in acute phase (AN, age, mean +/- SEM: 18.7 +/- 0.8 years) the effect of exogenous SS1-14 (25 and 75 micrograms/hour i.v., infused from +15 to +75 min), at doses that had previously been shown capable of increasing circulating SS levels within the physiological range, on the GH response to GH-releasing hormone (GHRH) (1 microgram/kg i.v. at 0 min). The same study protocol was performed in 8 normal age-matched women (NW, 22.9 +/- 1.0 years). RESULTS: In AN patients, IGF-I levels were lower (p < .01) than those in NW, while basal GH levels were similar in both groups. The GHRH-induced GH rise in AN was higher (p < .01) than that in NW. In AN, the exaggerated GH response to GHRH was inhibited to the same extent by both SS doses (p < .05) and became similar to that after GHRH alone in NW. In NW both 25 and 75 micrograms/hour SS decreased the GHRH-induced GH response; however, the inhibitory effect of the lower dose did not attain statistical significance, whereas the higher dose did (p < .02). During SS infusion, the GHRH-induced GH response in NW was persistently lower (p < .02) than that in AN. The percent inhibitory effect of SS on the somatotroph responsiveness to GHRH was similar in both groups at each dose. CONCLUSIONS: Our present findings demonstrate that the sensitivity of somatotroph cells to exogenous SS given at physiological doses is preserved in patients with AN. It is noteworthy that, during the infusion of physiological SS doses, the GH response to GHRH in AN overlaps on that to GHRH alone under physiological conditions. Thus, in AN, the sensitivity of somatotroph cells to SS apparently being preserved, an impairment of somatostatinergic neurons cannot be ruled out.


Subject(s)
Anorexia Nervosa/physiopathology , Growth Hormone-Releasing Hormone , Hormone Antagonists/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Insulin-Like Growth Factor I/analysis , Somatostatin/pharmacology , Adolescent , Adult , Analysis of Variance , Anorexia Nervosa/blood , Area Under Curve , Case-Control Studies , Dose-Response Relationship, Drug , Drug Interactions , Feedback , Female , Growth Hormone-Releasing Hormone/drug effects , Humans , Hypothalamus/drug effects , Somatostatin/metabolism , Statistics, Nonparametric , Time Factors
14.
Neurosci Lett ; 276(2): 87-90, 1999 Dec 03.
Article in English | MEDLINE | ID: mdl-10624798

ABSTRACT

We examined the effect of insulin-like growth factor-I (IGF-I) on GH-releasing factor (GRF) receptor expression in the primary rat anterior pituitary cell culture. The levels of GRF receptor mRNA were dose-dependently reduced by IGF-I treatment for 24 h. To clarify whether altered levels of GRF receptor mRNA contribute to GRF receptor concentrations, we examined the GH response to GRF in vitro. There was no difference in basal GH secretion between control and IGF-I pretreated cells, while GRF-stimulated GH secretion in cells pretreated with IGF-I for 24 h was significantly lower than that in control cells. Moreover, specific [125I] Tyr10-human GRF binding to pituitary cells was reduced significantly by IGF-I treatment. These results suggest that IGF-I acts directly on the pituitary and participates in the regulation of GRF receptor expression.


Subject(s)
Growth Hormone-Releasing Hormone/drug effects , Insulin-Like Growth Factor I/pharmacology , Pituitary Gland, Anterior/drug effects , RNA, Messenger/drug effects , Receptors, Neuropeptide/drug effects , Receptors, Pituitary Hormone-Regulating Hormone/drug effects , Animals , Cells, Cultured , Growth Hormone-Releasing Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Pituitary Gland, Anterior/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism
15.
Physiol Behav ; 64(1): 7-13, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9661976

ABSTRACT

Five thousand and eighteen quadruplet daily measurements of lower-leg length of 62 female and 81 male rats, were performed in order to characterize short-term growth. Within a short time, growth proceeds irregularly and consists of multiple incremental bursts (mini growth spurts) with no evidence for strict periodic behavior. Mini growth spurts are S-shaped incremental patterns that can be characterized by double-exponential functions (Gompertz's functions). Gompertz's functions are S-shaped, and can be defined by three parameters that identify amplitude, inflection point (age at peak growth velocity), and slope. The latter not only refers to the rapidity of each incremental burst, but also alludes to the duration that one incremental burst needs for completion. In regard to these characteristics, mini growth spurts differ significantly between the sexes in rats. Mean amplitude of mini growth spurts was 2153 microm (SD 1034 microm) in female rats and 2958 microm (SD 1614 microm) in male rats. Peak growth velocity of mini growth spurts appeared lower in male rats than in female rats. Female rats showed mean gamma of -1.23 (SD 0.72), whereas male rats showed mean y of -0.96 (SD 0.72). Partial growth hormone deficiency led to a modification in rats that was reversed when exogenous growth hormone was administered. Mean intervals between subsequent mini growth spurts ranged between 4.2 and 4.6 days, but the large variation of these intervals (SD between 1.6 and 2.3 days) and the fact that neither spurt-spurt interval nor spurt amplitude appeared predictable, strongly suggest chaotic behavior of mini growth spurts.


Subject(s)
Hindlimb/growth & development , Nonlinear Dynamics , Algorithms , Animals , Computer Simulation , Female , Growth/drug effects , Growth Hormone/pharmacology , Growth Hormone/physiology , Growth Hormone-Releasing Hormone/biosynthesis , Growth Hormone-Releasing Hormone/drug effects , Male , Periodicity , Rats , Sex Distribution , Sodium Glutamate/pharmacology
16.
Acta Paediatr Suppl ; 423: 92-6, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9401552

ABSTRACT

Growth hormone-releasing peptides (GHRPs) are synthetic molecules with strong, dose-related and reproducible growth hormone (GH)-releasing activity in humans. GHRPs act at both the pituitary and the hypothalamic level, where specific receptors have been located. In adults, GHRPs release more GH than does GH-releasing hormone (GHRP), whilst their co-administration has a synergistic effect, indicating that they have, at least partially, different mechanisms of action. However, normal activity of GHRH-secreting neurones is needed to achieve the full GH-releasing effect of GHRPs. In contrast to GHRH, the GH-releasing activity of GHRPs is not further increased by substances acting via inhibition of hypothalamic somatostatin, and is only blunted by substances that stimulate hypothalamic somatostatin release. Even free fatty acids and exogenous somatostatin, which act directly on somatotrophs, do no more than blunt the effect of GHRPs. Thus, the GH-releasing activity of GHRPs is partially refractory to inhibitory influences, GHRPs act, at least in part, by antagonism of somatostatin activity, both at the pituitary and the hypothalamic level. The GH-releasing effect of GHRPs is not dependent on gender, but undergoes age-related variations. Gonadal steroids seem to influence the activity of GHRPs only in childhood. The reduced GH response to GHRPs in the elderly is probably due mainly to concomitant GHRH hypoactivity and somatostatinergic hyperactivity. A preserved GH-releasing effect of GHRPs has been reported in acromegaly, anorexia nervosa, hyperthyroidism and in critically ill patients. GHRPs have also been found to increase GH release in children with idiopathic short stature, in GH deficiency and in obese patients, in whom there is a well-known reduction of somatotroph function. The GH response to GHRPs is markedly reduced in hypothyroidism and Cushing's syndrome.


Subject(s)
Aging/metabolism , Growth Hormone-Releasing Hormone/physiology , Growth Substances/pharmacology , Human Growth Hormone/metabolism , Oligopeptides/pharmacology , Drug Synergism , Growth Hormone-Releasing Hormone/drug effects , Human Growth Hormone/physiology , Humans , Hypothalamus/drug effects , Pituitary Gland/drug effects
17.
Acta Paediatr Suppl ; 423: 102-6, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9401554

ABSTRACT

A diagnostic test was devised to evaluate pituitary growth hormone (GH) secretory potential. GH secretory dynamics were assessed in children with and without GH deficiency. The GH response was measured to GH-releasing hormone (GHRH) and the GH-releasing peptide GHRP-2, administered sequentially. The mean (+/- SEM) peak GH response to GHRP-2 was 20.1 +/- 5.5, 63.6 +/- 24.9 and 42.2 +/- 4.3 micrograms/l for GH-deficient, slowly growing non-GH-deficient and control children, respectively (p < 0.02 and p < 0.05 for GH-deficient vs controls and slowly growing children, respectively). Corresponding values for area under the curve (AUC) were 995 +/- 371. 2460 +/- 953 and 1598 +/- 274 micrograms/l x minute. Peak GH (and AUC) responses to GHRH were 19.6 +/- 5.1 micrograms/l (924 +/- 232 micrograms/l x minute), 31.4 +/- 8.4 micrograms/l (1544 +/- 449 micrograms/l x minute) and 39.8 +/- 7.8 micrograms/l (2201 +/- 437 micrograms/l x minute) for the same three groups, respectively (p < 0.05 for peak GH in GH-deficient patients vs controls, and p < 0.02 and p < 0.01 for AUC in GH-deficient vs slowly growing children and controls, respectively). The ratio of the peak GH response to GHRP-2 and GHRH was similar in all three groups. As these secretagogues stimulate different aspects of hypothalamic function (i.e., they are functional complements), robust GH secretion in response to GHRH or GHRP could suggest adequate endogenous GHRP or GHRH, respectively. A poor response to either GH secretagogue administered individually could represent inadequacy of its endogenous complement. The integrity of functional pituitary elements could be differentiated from inadequate complements by administering both GH secretagogues simultaneously. Application of these principles should allow a better definition of the underlying disorder and provide the basis for therapeutic strategies for those patients with abnormal GH production and/or secretion.


Subject(s)
Growth Disorders/metabolism , Human Growth Hormone/deficiency , Human Growth Hormone/metabolism , Oligopeptides/pharmacology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Area Under Curve , Case-Control Studies , Child, Preschool , Female , Growth Disorders/diagnosis , Growth Hormone-Releasing Hormone/drug effects , Humans , Male , Oligopeptides/pharmacokinetics , Reference Values
18.
Recenti Prog Med ; 88(6): 264-8, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9233053

ABSTRACT

Sumatriptan (SU), a specific 5-HT1D receptor agonist, was recently shown to be able to increase growth hormone (GH) levels in normals, but not in acromegalics, while no effect was seen on prolactine (PRL). SU is also able to produce an increase in GH response to growth hormone releasing hormone (GHRH) in prepubertal children. In this study we investigated whether SU administration influences GHRH-induced GH secretion in 8 acromegalics, and 6 age-matched normal volunteers, as a control group. We evaluated the effects of SU (6 mg s.c.) or placebo (PL) administration on GHRH (1 microgram/kg bw i.v.)-induced GH and PRL secretion. After SU priming the GH response to GHRH did not changed in acromegalics, but significantly decreased in controls, in comparison with that observed after PL plus GHRH. In acromegalics, no difference in GH peak was seen after SU plus GHRH and PL plus GHRH, nor was any difference seen in AUC between tests. In controls, no difference was seen in GH peaks, while SU priming significantly (P < 0.03) decreased the AUC 90-120 min of GH after GHRH administration. In acromegalics, SU did not change the slight GHRH-induced increase in PRL levels. Our study documents that 5-HT1 D receptors do not interfere with GHRH-stimulated GH secretion in acromegalic subjects. In normals, SU is able to decrease GH response to GHRH, thus confirming that 5-HT1D receptors are able to modulate GH secretion in normals.


Subject(s)
Acromegaly/blood , Growth Hormone-Releasing Hormone/drug effects , Growth Hormone/metabolism , Prolactin/metabolism , Serotonin Receptor Agonists/pharmacology , Sumatriptan/pharmacology , Adult , Aged , Case-Control Studies , Female , Growth Hormone/drug effects , Humans , Male , Middle Aged , Prolactin/drug effects
19.
J Endocrinol Invest ; 20(3): 144-50, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9186821

ABSTRACT

The individual role played by GH and IGF-I in the regulation of hypothalamic GHRH and SRIF gene expression is still object of debate. We have investigated the effect of exogenously administered recombinant hGH (rhGH) and recombinant hIGF-I (rhIGF-I) in ad libitum fed control and starved rats, the latter an animal model which is characterized by low circulating levels of endogenous GH and IGF-I. Adult male rats were fed ad libitum (C) or food-deprived (S) for 72 hours; rats in either C or S groups were treated with systemic administration of rhGH and rhIGF-I for 3 days. GHRH, SRIF and GH mRNA levels were evaluated by Northern and slot blot hybridization. Administration of rhGH (250 micrograms/kg/twice daily, sc) induced a significant inhibition of GHRH and a significant stimulation of SRIF mRNA levels in C rats; GH treatment was, however, ineffective on both neuropeptide mRNA levels in the S group. Continuous infusion of rhIGF-I (300 micrograms/kg/day, sc) induced a significant increase of SRIF levels in both C and S rats but did not modify GHRH mRNA levels in either group. In the pituitary, GH mRNA levels followed a pattern very similar to that of GHRH. These results provide evidence for a direct role of GH in the inhibition of GHRH mRNA levels; IGF-I appears more involved in the direct stimulation of SRIF mRNA levels.


Subject(s)
Food Deprivation/physiology , Growth Hormone-Releasing Hormone/drug effects , Human Growth Hormone/pharmacology , Insulin-Like Growth Factor I/pharmacology , RNA, Messenger/drug effects , Somatostatin/drug effects , Animals , Eating , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Human Growth Hormone/administration & dosage , Human Growth Hormone/genetics , Humans , Hypothalamus/chemistry , Hypothalamus/metabolism , Injections, Intraventricular , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/genetics , Male , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Somatostatin/genetics , Somatostatin/metabolism
20.
J Endocrinol Invest ; 20(3): 151-4, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9186822

ABSTRACT

It is generally accepted that growth hormone influences its own secretion by modulating the activity of GHRH and SRIF neurons. To investigate if GH feedback mechanisms are already operating in the early postnatal life of the rat, we have studied in 10-day-old pups the effects of rhGH and rhIGF-I administration on GHRH and somatostatin mRNA levels. The same experiment was also performed in pups passively immunized with an anti-GHRH antiserum from the day of birth. The latter animal model had been previously characterized for presenting reduced levels of circulating GH and IGF-I. In control pups, neither rhGH (250 micrograms/kg, b.i.d., sc) nor rhIGF-I (150 micrograms/kg, b.i.d., sc) administration induced significant changes of GHRH and SRIF gene expression. The passive immunization against GHRH induced per se a trend toward an increase and a reduction of GHRH and SRIF mRNA levels, respectively. Also in these rats the treatment for 3 days with rhGH and rhIGF-I did not further modify the GHRH and SRIF mRNA levels. Based on these results, we conclude that in the 10-day-old rat GH feedback mechanisms are poorly operative, though a direct ultra-short loop mechanism involving the GHRH and SRIF systems seems already operating.


Subject(s)
Animals, Newborn/physiology , Growth Hormone-Releasing Hormone/drug effects , Human Growth Hormone/pharmacology , Insulin-Like Growth Factor I/pharmacology , RNA, Messenger/drug effects , Somatostatin/drug effects , Animals , Animals, Newborn/growth & development , Female , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/immunology , Human Growth Hormone/administration & dosage , Injections, Subcutaneous , Insulin-Like Growth Factor I/administration & dosage , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Somatostatin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...