Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Endocr Connect ; 9(7): 676-686, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32567553

ABSTRACT

PURPOSE: Augmented survival of childhood nephroblastoma and neuroblastoma has increased long-term side effects such as metabolic syndrome (MetS). Risk stratification is difficult after abdominal radiation because waist circumference underestimates adiposity. We aimed to develop a strategy for determining MetS in irradiated survivors using an integrated biomarker profile and vascular ultrasonography. METHODS: The NCEP-ATPIII MetS-components, 14 additional serum biomarkers and 9 vascular measurements were assessed in a single-centre cohort of childhood nephroblastoma (n = 67) and neuroblastoma (n = 36) survivors and controls (n = 61). Multivariable regression models were used to study treatment effects. Principal component analysis (PCA) was used to study all biomarkers in a combined analysis, to identify patterns and correlations. RESULTS: After 27.5 years of follow-up, MetS occurred more often in survivors (14%) than controls (3%). Abdominal radiotherapy and nephrectomy, to a lesser extent, were associated with MetS and separate components and with several biomarker abnormalities. PCA of biomarkers revealed a pattern on PC1 from favourable lipid markers (HDL-cholesterol, adiponectin) towards unfavourable markers (triglycerides, LDL-cholesterol, apoB, uric acid). Abdominal radiotherapy was associated with the unfavourable biomarker profile (ß = 1.45, P = 0.001). Vascular measurements were not of added diagnostic value. CONCLUSIONS: Long-term childhood nephro- and neuroblastoma survivors frequently develop MetS. Additional assessment of biomarkers identified in PCA - adiponectin, LDL, apoB, and uric acid - may be used especially in abdominally irradiated survivors, to classify MetS as alternative for waist circumference. Vascular ultrasonography was not of added value.

3.
Int J Obes (Lond) ; 41(2): 210-216, 2017 02.
Article in English | MEDLINE | ID: mdl-27795552

ABSTRACT

BACKGROUND: Hypothalamic obesity is a devastating consequence of craniopharyngioma. Bariatric surgery could be a promising therapeutic option. However, its efficacy and safety in patients with craniopharyngioma-related hypothalamic obesity remain largely unknown. OBJECTIVES: We investigated the efficacy of bariatric surgery for inducing weight loss in patients with craniopharyngioma-related hypothalamic obesity. In addition, we studied the safety of bariatric surgery regarding its effects on hormone replacement therapy for pituitary insufficiency. METHODS: In this retrospective matched case-control study, we compared weight loss after bariatric surgery (that is, Roux-en-Y gastric bypass and sleeve gastrectomy) between eight patients with craniopharyngioma-related hypothalamic obesity and 75 controls with 'common' obesity during 2 years of follow-up. We validated our results at 1 year of follow-up in a meta-analysis. In addition, we studied alterations in hormone replacement therapy after bariatric surgery in patients with craniopharyngioma. RESULTS: Mean weight loss after bariatric surgery was 19% vs 25% (difference -6%, 95% confidence of interval (CI) -14.1 to 4.6; P=0.091) at 2 years of follow-up in patients with craniopharyngioma-related hypothalamic obesity compared with control subjects with 'common' obesity. Mean weight loss was 25% vs 29% (difference -4%, 95% CI -11.6 to 8.1; P=0.419) after Roux-en-Y gastric bypass and 10% vs 20% (difference -10%, 95% CI -14.1 to -6.2; P=0.003) after sleeve gastrectomy at 2 years of follow-up in patients with craniopharyngioma-related hypothalamic obesity vs control subjects with 'common' obesity. Our meta-analysis demonstrated significant weight loss 1 year after Roux-en-Y gastric bypass, but not after sleeve gastrectomy. Seven patients with craniopharyngioma suffered from pituitary insufficiency; three of them required minor adjustments in hormone replacement therapy after bariatric surgery. CONCLUSIONS: Weight loss after Roux-en-Y gastric bypass, but not sleeve gastrectomy, was comparable between patients with craniopharyngioma-related hypothalamic obesity and control subjects with 'common' obesity at 2 years of follow-up. Bariatric surgery seems safe regarding its effects on hormone replacement therapy.


Subject(s)
Craniopharyngioma/complications , Gastrectomy , Gastric Bypass , Obesity/etiology , Pituitary Neoplasms/complications , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Craniopharyngioma/drug therapy , Craniopharyngioma/surgery , Female , Follow-Up Studies , Humans , Male , Netherlands/epidemiology , Obesity/surgery , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/surgery , Retrospective Studies , Sweden/epidemiology , Treatment Outcome , Weight Loss , Young Adult
5.
Clin Endocrinol (Oxf) ; 85(3): 488-94, 2016 09.
Article in English | MEDLINE | ID: mdl-26850227

ABSTRACT

BACKGROUND: Prader-Willi syndrome (PWS) is characterized by hyperphagia with impaired satiety. PWS patients have very high acylated ghrelin (AG) with normal unacylated ghrelin (UAG) levels, resulting in an elevated AG/UAG ratio, suggesting an intrinsic defect in the ghrelin regulation. Normally, food intake induces satiety and a drop in AG and UAG levels, but it is unknown if these levels also decline in PWS. OBJECTIVE: To evaluate whether the high AG levels in PWS decline in response to glucose intake during an oral glucose tolerance test (OGTT), and to investigate the effects of growth hormone (GH) treatment on this response. METHOD: Serum levels of AG, UAG and AG/UAG ratio during an OGTT were determined in 24 GH-treated patients with PWS (median age 19·0, range 14·2-25·9 years) and in 10 GH-stop patients (of whom five were in GH-treated group; 18·5, 14·5-20·3 years). RESULTS: In GH-treated and GH-stop young adults with PWS, there was a sharp decline of AG levels and a decrease of UAG levels in the first 30 min after the glucose load, which resulted in a lower AG/UAG ratio. GH-treated patients had significantly lower AG levels than GH-stop patients at baseline and during the OGTT. All UAG levels and AG/UAG ratios were lower in the GH-treated patients, although not significantly. CONCLUSIONS: In young adults with PWS, an oral glucose load significantly reduces AG and UAG levels, suggesting normal regulation of the ghrelin axis by food intake. GH treatment results in lower AG levels at baseline and during OGTT, suggesting a more favourable metabolic profile. Our findings might suggest that the impaired satiety is not the result of an abnormal response of the orexigenic ghrelin to food intake.


Subject(s)
Eating , Ghrelin/blood , Glucose Tolerance Test , Prader-Willi Syndrome/blood , Acylation , Adolescent , Adult , Blood Glucose , Ghrelin/metabolism , Human Growth Hormone/pharmacology , Human Growth Hormone/therapeutic use , Humans , Young Adult
6.
Eur J Endocrinol ; 174(5): 663-7, 2016 May.
Article in English | MEDLINE | ID: mdl-26903550

ABSTRACT

CONTEXT: Although combination therapy of acromegaly with long-acting somatostatin analogs (LA-SSAs) and pegvisomant (PEGV) normalizes insulin-like growth factor-1 (IGF1) levels in the majority of patients, it requires long-term adherence. Switching from combination therapy to monotherapy with weekly PEGV could improve patients' comfort, but the efficacy is unknown. OBJECTIVE: To assess the efficacy of switching to PEGV monotherapy in patients well controlled on combination therapy of LA-SSAs and PEGV. DESIGN: Single-center, open-label observational pilot study. LA-SSA therapy was discontinued at baseline and all patients were switched to PEGV monotherapy for 12 months. If IGF1 levels exceeded 1.0 times upper limit of normal (ULN), PEGV dose was increased by 20 mg weekly. SUBJECTS AND METHODS: The study included 15 subjects (eight males), with a median age of 58 years (range 35-80) on combination therapy of high-dose LA-SSAs and weekly PEGV for >6 months, and IGF1 levels within the normal range. Treatment efficacy was assessed by measuring serum IGF1 levels. RESULTS: After 12 months of weekly PEGV monotherapy, serum IGF1 levels of 73% of the subjects remained controlled. In one patient, LA-SSA had to be restarted due to recurrence of headache. IGF1 levels increased from a baseline level of 0.62 × ULN (range 0.30-0.84) to 0.83 × ULN (0.30-1.75) after 12 months, while the median weekly PEGV dose increased from 60 (30-80) mg to 80 (50-120) mg. CONCLUSION: Our results suggest that switching from combination therapy of LA-SSAs and PEGV to PEGV monotherapy can be a viable treatment option for acromegaly patients without compromising efficacy.


Subject(s)
Acromegaly/blood , Acromegaly/drug therapy , Human Growth Hormone/analogs & derivatives , Insulin-Like Growth Factor I/analysis , Outcome Assessment, Health Care , Somatostatin/pharmacology , Adult , Aged , Aged, 80 and over , Drug Therapy, Combination , Female , Human Growth Hormone/administration & dosage , Human Growth Hormone/pharmacology , Humans , Male , Middle Aged , Pilot Projects , Somatostatin/administration & dosage , Somatostatin/analogs & derivatives
7.
Eur J Endocrinol ; 173(5): 553-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26243033

ABSTRACT

BACKGROUND: Doses of the GH receptor (GHR) antagonist pegvisomant (PEGV) that normalize insulin-like growth factor 1 (IGF1) levels vary widely among acromegaly patients. Predictors for PEGV response are baseline IGF1 levels, sex, body weight and previous radiotherapy. A GHR polymorphism lacking exon 3 (d3-GHR) is frequent in the general population. The influence of d3-GHR on PEGV responsiveness in acromegaly is unclear. OBJECTIVE: To assess the influence of d3-GHR on IGF1 levels and PEGV responsiveness in acromegaly patients using combined PEGV and long-acting somatostatin receptor ligand (LA-SRIF) treatment. DESIGN: Data were collected at the Rotterdam Pituitary Centre between 2004 and 2013. Patients with elevated IGF1 levels (>1.2 upper limit of normal; n=112) and over 6 months of high-dose LA-SRIF treatment were co-treated with PEGV. GHR genotype was assessed using genomic DNA in 104 patients. RESULTS: D3-GHR was observed in 51 (49.0%) of the patients (7.7% homozygous, 41.3% heterozygous) and was in Hardy-Weinberg equilibrium (P=0.859). Baseline characteristics were similar in d3-GHR and full-length (fl)-GHR genotypes. During PEGV/LA-SRIF treatment IGF1 levels were not different between d3-carriers and non-carriers. Similarly, no difference in PEGV dose required to normalize IGF1 (P=0.337) or PEGV serum levels (P=0.433) was observed between the two groups. However, adenoma size decreased significantly (>20% of largest diameter) in 25.6% of the fl-GHR genotype but only in 7.5% of d3-carriers (P=0.034, OR: 4.6 (CI: 1.1-18.9)). CONCLUSIONS: GHR genotype does not predict the IGF1 normalizing dose of PEGV in acromegaly patients using combination PEGV/LA-SRIF treatment. However, fewer d3-carriers showed significant reductions in adenoma size.


Subject(s)
Acromegaly/drug therapy , Adenoma/drug therapy , Human Growth Hormone/analogs & derivatives , Insulin-Like Growth Factor I/metabolism , Membrane Proteins/genetics , Pituitary Neoplasms/drug therapy , Somatostatin/pharmacology , Adult , Delayed-Action Preparations , Drug Therapy, Combination , Exons , Female , Human Growth Hormone/pharmacology , Humans , Male , Membrane Proteins/antagonists & inhibitors , Middle Aged , Somatostatin/analogs & derivatives , Treatment Outcome
8.
Endocrine ; 50(3): 633-42, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25989955

ABSTRACT

Prader-Willi syndrome (PWS) is characterized by a switch from failure to thrive to excessive weight gain and hyperphagia in early childhood. Hyperghrelinemia may be involved in the underlying mechanisms of the switch. The purpose of this study is to evaluate acylated ghrelin (AG) and unacylated ghrelin (UAG) levels in PWS and investigate their associations with hyperphagia. This is a cross-sectional clinical study conducted in three PWS expert centers in the Netherlands and France. Levels of AG and UAG and the AG/UAG ratio were determined in 138 patients with PWS (0.2-29.4 years) and compared with 50 age-matched obese subjects (4.3-16.9 years) and 39 healthy controls (0.8-28.6 years). AEBSF was used to inhibit deacylation of AG. As a group, PWS patients had higher AG but similar UAG levels as healthy controls (AG 129.1 vs 82.4 pg/ml, p = 0.016; UAG 135.3 vs 157.3 pg/ml, resp.), resulting in a significantly higher AG/UAG ratio (1.00 vs 0.61, p = 0.001, resp.). Obese subjects had significantly lower AG and UAG levels than PWS and controls (40.3 and 35.3 pg/ml, resp.), but also a high AG/UAG ratio (1.16). The reason for the higher AG/UAG ratio in PWS and obese was, however, completely different, as PWS had a high AG and obese a very low UAG. PWS patients without weight gain or hyperphagia had a similar AG/UAG ratio as age-matched controls, in contrast to those with weight gain and/or hyperphagia who had an elevated AG/UAG ratio. The switch to excessive weight gain in PWS seems to coincide with an increase in the AG/UAG ratio, even prior to the start of hyperphagia.


Subject(s)
Ghrelin/blood , Prader-Willi Syndrome/blood , Acylation , Adolescent , Age Factors , Body Mass Index , Case-Control Studies , Child , Child, Preschool , Cross-Sectional Studies , Eating , Feeding Behavior , Female , Genotype , Humans , Hyperphagia/blood , Insulin-Like Growth Factor I/metabolism , Male , Obesity/blood
9.
Int J Obes (Lond) ; 38(2): 163-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23774329

ABSTRACT

Obesity is one of the greatest public health challenges of the 21st century. Obesity is currently responsible for ∼0.7-2.8% of a country's health costs worldwide. Treatment is often not effective because weight regulation is complex. Appetite and energy control are regulated in the brain. Melanocortin-4 receptor (MC4R) has a central role in this regulation. MC4R defects lead to a severe clinical phenotype with lack of satiety and early-onset severe obesity. Preclinical research has been carried out to understand the mechanism of MC4R regulation and possible effectors. The objective of this study is to systematically review the literature for emerging pharmacological obesity treatment options. A systematic literature search was performed in PubMed and Embase for articles published until June 2012. The search resulted in 664 papers matching the search terms, of which 15 papers remained after elimination, based on the specific inclusion and exclusion criteria. In these 15 papers, different MC4R agonists were studied in vivo in animal and human studies. Almost all studies are in the preclinical phase. There are currently no effective clinical treatments for MC4R-deficient obese patients, although MC4R agonists are being developed and are entering phase I and II trials.


Subject(s)
Anti-Obesity Agents/therapeutic use , Appetite Regulation/drug effects , Energy Intake/drug effects , Energy Metabolism/drug effects , Obesity/drug therapy , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/metabolism , Acetamides/therapeutic use , Animals , Appetite Regulation/genetics , Body Mass Index , Energy Intake/genetics , Energy Metabolism/genetics , Gene Frequency , Genotype , Humans , Obesity/genetics , Peptides, Cyclic/therapeutic use , Phenotype , Pyrrolidines/therapeutic use , Receptor, Melanocortin, Type 4/deficiency , Receptor, Melanocortin, Type 4/genetics , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/metabolism , Spiro Compounds/therapeutic use , alpha-MSH/analogs & derivatives , alpha-MSH/therapeutic use
10.
Growth Horm IGF Res ; 23(5): 193-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23850060

ABSTRACT

OBJECTIVE: Ghrelin, a gut-brain peptide, regulates energy homeostasis and glucose metabolism and is present in acylated and nonacylated form in the circulation. Although desacyl ghrelin (DAG), the predominant form of ghrelin, is associated with insulin sensitivity and improved metabolic state, not much is known about its direct regulation by insulin. We aimed to assess changes in DAG in response to the rapid increase in insulin concentration during an insulin tolerance test (ITT) in normal weight and obese subjects. DESIGN: We performed an observational single center study. An ITT was assessed in eight subjects (four males), median age of 29.9 years (range 19.6-42.0). DAG concentrations were measured at 20, 40, 60 and 90 min after insulin infusion. Homeostatic Model Assessment (HOMA) was calculated from fasting insulin and glucose. Body mass index (BMI) and waist circumference were assessed. RESULTS: Three subjects were obese (BMI ≥ 30 kg/m(2)), one subject was overweight (BMI = 25-30 kg/m(2)) and four subjects had normal weight (BMI = 18.5-25 kg/m(2)). Median DAG decreased after insulin infusion (90 pg/mL, p = 0.028), especially in normal weight subjects. Baseline DAG was lower in subjects with higher BMI (ρ = -0.76, p = 0.028) and higher fasting insulin (ρ = -0.76, p = 0.030). DAG changes correlated with fasting insulin levels (ρ = -0.85, p = 0.007), HOMA (ρ = -0.86, p = 0.007), BMI (ρ = -0.83, p = 0.010) and waist circumference (ρ = -0.93, p < 0.001). CONCLUSION: DAG levels rapidly decreased in response to insulin administration in normal subjects, but not in insulin-resistant obese who are in a state of relative DAG deficiency.


Subject(s)
Diagnostic Techniques, Endocrine , Ghrelin/blood , Insulin Resistance , Insulin/administration & dosage , Insulin/blood , Adult , Dose-Response Relationship, Drug , Fasting , Female , Glucose Clamp Technique , Humans , Ideal Body Weight/physiology , Male , Obesity/blood , Overweight/blood , Young Adult
11.
Peptides ; 32(11): 2309-18, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21396419

ABSTRACT

Ghrelin plays an important physiological role in modulating GH secretion, insulin secretion and glucose metabolism. Ghrelin has direct effects on pancreatic islet function. Also, ghrelin is part of a mechanism that integrates the physiological response to fasting. However, pharmacologic studies indicate the important obesogenic/diabetogenic properties of ghrelin. This is very likely of physiological relevance, deriving from a requirement to protect against seasonal periods of food scarcity by building energy reserves, predominantly in the form of fat. Available data indicate the potential of ghrelin blockade as a means to prevent its diabetogenic effects. Several studies indicate a negative correlation between ghrelin levels and the incidence of type 2 diabetes and insulin resistance. However, it is unclear if low ghrelin levels are a risk factor or a compensatory response. Direct antagonism of the receptor does not always have the desired effects, however, since it can cause increased body weight gain. Pharmacological suppression of the ghrelin/des-acyl ghrelin ratio by treatment with des-acyl ghrelin may also be a viable alternative approach which appears to improve insulin sensitivity. A promising recently developed approach appears to be through the blockade of GOAT activity, although the longer term effects of this treatment remain to be investigated.


Subject(s)
Acyltransferases/metabolism , Ghrelin , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/drug effects , Peptides/pharmacology , Receptors, Ghrelin/metabolism , Acylation , Acyltransferases/antagonists & inhibitors , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Expression , Ghrelin/antagonists & inhibitors , Ghrelin/blood , Ghrelin/genetics , Ghrelin/pharmacology , Humans , Insulin Resistance , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Islets of Langerhans/metabolism , Liver/metabolism , Mice , Obesity/drug therapy , Obesity/metabolism , Peptides/therapeutic use , Rats , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/genetics , Signal Transduction
12.
Am J Physiol Endocrinol Metab ; 293(1): E302-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17405826

ABSTRACT

Ghrelin is expressed in normal human adrenocortical cells and induces their proliferation through growth hormone secretagogue receptor 1a (GHS-R1a). Consequently, it was of interest to us to determine whether acylated ghrelin and its predominant serum isoform, unacylated ghrelin, also act as factors for adrenocortical carcinoma cell growth. To examine a potential ghrelin-regulated system in adrenocortical tumors, we measured proliferative effects of acylated and unacylated ghrelin in the adrenocortical carcinoma cell lines SW-13 and NCI-H295R. We also examined the expression of ghrelin, GHS-R1a, and corticotrophin-releasing factor receptor 2 (CRF-R2). Acylated and unacylated ghrelin in the nanomolar range dose-dependently induced adrenocortical cell growth up to 200% of untreated controls, as measured by thymidine uptake and WST1 assay. The proliferative effects of acylated and unacylated ghrelin in SW-13 cells was blocked by [D-Lys(3)]growth hormone-releasing peptide 6 (GHRP6), but a CRF-R2 antagonist had no effect on unacylated ghrelin growth stimulation. Cell cycle analysis suggests that acylated and unacylated ghrelin suppress the sub-G(0)/apoptotic fraction by up to 50%. Measurement of DNA fragmentation and caspase-3 and -7 activity in SW-13 cells confirmed that acylated and unacylated ghrelin suppress apoptotic rate. SW-13 cells express preproghrelin mRNA and secrete ghrelin, and [D-Lys(3)]GHRP6 suppresses their basal proliferation rate, strongly suggesting that ghrelin could act as an auto/paracrine growth factor. Acylated and unacylated ghrelin are potential auto/paracrine factors acting through an antiapoptotic pathway to stimulate adrenocortical tumor cell growth. Unacylated ghrelin-stimulated growth is suppressed by an antagonist of GHS-R1a, suggesting either that unacylated ghrelin is acylated before its action or that ghrelin, unacylated ghrelin, and [D-Lys(3)]GHRP-6 bind to a novel receptor in these cells.


Subject(s)
Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/pathology , Apoptosis/drug effects , Cell Proliferation/drug effects , Peptide Hormones/pharmacology , Acetylation , Cell Cycle/drug effects , Ghrelin , Humans , Peptide Hormones/metabolism , Protein Isoforms/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Ghrelin , Signal Transduction/drug effects , Tumor Cells, Cultured
13.
J Endocrinol ; 188(1): 37-47, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16394173

ABSTRACT

Recent studies demonstrate widespread expression of ghrelin among tissues and have uncovered its pleiotropic nature. We have examined gene expression of ghrelin and its two receptor splice variants, growth hormone secretagogue receptors (GHS-R) 1a and 1b, in human bone biopsies and in the human pre-osteoblastic SV-HFO cell line during differentiation. Additionally, we examined proliferative effects of ghrelin and unacylated ghrelin (UAG) in differentiating and non-differentiating cells. We detected GHS-R1b mRNA in human bone and osteoblasts but not ghrelin's cognate receptor GHS-R1a, using two different real-time PCR assays and both total RNA and mRNA. In osteoblasts GHS-R1b mRNA expression remained low during the first 14 days of culture, but increased 300% in differentiating cells by day 21. Both human bone biopsies and osteoblasts expressed ghrelin mRNA, and osteoblasts were found to secrete ghrelin. Overall, ghrelin gene expression was greater in differentiating than non-differentiating osteoblasts, but was not increased during culture in either group. Ghrelin and UAG induced thymidine uptake dose-dependently, peaking at 1 and 10 nM respectively, at day 6 of culture in both non-differentiating and differentiating osteoblasts. The proliferative response to ghrelin and UAG declined with culture time and state of differentiation. The proliferative effects of ghrelin and UAG were suppressed by inhibitors of extracellular-signal-regulated kinase (ERK) and phosphoinositide-3 kinase, and both peptides rapidly induced ERK phosphorylation. Overall, our data suggest new roles for ghrelin and UAG in modulating human osteoblast proliferation via a novel signal transduction pathway.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/cytology , Peptide Hormones/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Alkaline Phosphatase/metabolism , Analysis of Variance , Biomarkers/analysis , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , DNA/biosynthesis , Femur Head , Ghrelin , Humans , Osteoblasts/drug effects , Receptors, Ghrelin , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical , Thymidine/analysis , Thymidine/metabolism
14.
J Clin Endocrinol Metab ; 89(10): 5035-42, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15472202

ABSTRACT

We investigated the metabolic actions of ghrelin in humans by examining the effects of acute administration of acylated ghrelin, unacylated ghrelin, and the combination in eight adult-onset GH-deficient patients. We followed glucose, insulin, and free fatty acid concentrations before and after lunch and with or without the presence of GH in the circulation. We found that acylated ghrelin, which is rapidly cleared from the circulation, induced a rapid rise in glucose and insulin levels. Unacylated ghrelin, however, prevented the acylated ghrelin-induced rise in insulin and glucose when it was coadministered with acylated ghrelin. Surprisingly, the injection of acylated ghrelin induced an acute increase in unacylated ghrelin and therefore total ghrelin levels. Finally, acylated ghrelin decreased insulin sensitivity up to the end of a period of 6 h after administration. This decrease in insulin sensitivity was prevented by coinjection of unacylated ghrelin. This combined administration of acylated and unacylated ghrelin even significantly improved insulin sensitivity, compared with placebo, for at least 6 h, which warrants studies to investigate the long-term efficacy of this combination in the treatment of disorders with disturbed insulin sensitivity.


Subject(s)
Human Growth Hormone/deficiency , Hypopituitarism/drug therapy , Insulin Resistance , Peptide Hormones/administration & dosage , Acylation , Adult , Age of Onset , Aged , Blood Glucose/drug effects , Blood Glucose/metabolism , Eating , Fatty Acids, Nonesterified/metabolism , Ghrelin , Humans , Hypopituitarism/metabolism , Insulin/blood , Male , Middle Aged , Peptide Hormones/blood
15.
Am J Physiol Endocrinol Metab ; 280(2): E287-95, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158932

ABSTRACT

The initiation of liver regeneration is regulated by endogenously produced growth factors and cytokines and is accompanied by suppression of growth hormone (GH) binding to hepatocytes. We have demonstrated some of these factors, particularly GH, which modulate acid-labile subunit (ALS) expression in vitro. Consequently, we investigated ALS hepatic mRNA and serum levels in rats for 24 h after partial hepatectomy (PHx). There was a significant suppression of ALS gene expression (approximately 50%, P < 0.005) and serum levels (approximately 30%, P < 0.02) by 12 h in PHx rats relative to controls. Relative to intact animals, hepatic mRNA and serum levels of ALS were suppressed by approximately 60% at 24 h. Similarly, hepatic GH receptor mRNA levels were significantly reduced in PHx animals. Moreover, hepatocytes isolated from PHx animals were less responsive to GH than those from controls. Overall, our results demonstrate that suppression of ALS gene expression and serum levels during liver regeneration relates to lowered hepatic GH sensitivity. Suppressed circulating ALS may alter insulin-like growth factor bioavailability and constitute a mechanism to maintain relatively normal glucoregulation after loss of liver mass.


Subject(s)
Carrier Proteins/metabolism , Glucose/metabolism , Glycoproteins/metabolism , Liver Regeneration/physiology , Milk Proteins , Animals , Biomarkers , Blood Glucose/analysis , DNA-Binding Proteins/metabolism , Female , Gene Expression , Growth Hormone/physiology , Hepatectomy/methods , Hepatocytes/metabolism , Insulin/blood , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Rats , Rats, Wistar , Receptors, Somatotropin/genetics , STAT5 Transcription Factor , Trans-Activators/metabolism
16.
Gene ; 256(1-2): 283-91, 2000 Oct 03.
Article in English | MEDLINE | ID: mdl-11054558

ABSTRACT

The RET proto-oncogene plays an important role in the initiation and progression of tumors derived from the neural crest. The cis-regulatory elements responsible for RET basal promoter activity have not been identified. To characterize these elements, a RET promoter DNA fragment (-453 to +227bp) was fused to a luciferase reporter and introduced into TT, a neural crest-derived cell line. Sequential 5' deletions of the promoter revealed that optimal expression of the RET promoter in TT cells required only 70bp of sequence upstream of the transcription start site, and contains two Sp1 binding sites. DNase I footprinting, electrophoretic mobility shift analysis (EMSA), and supershift assays revealed that this region binds both Sp1 and its related protein, Sp3. Additionally, RET basal promoter activity was abrogated by removal of these Sp1/Sp3 binding sites. The proximal two GC boxes were sufficient to allow transactivation of the RET promoter in Drosophila SL2 cells. Sp3 expression in these cells caused an additional activation of the promoter. These results demonstrate that the transactivation of the RET promoter within a neural crest-derived cell line is dependent on Sp1 and Sp3.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Animals , Binding Sites/genetics , Cell Line , DNA/genetics , DNA/metabolism , DNA Footprinting , Deoxyribonucleases/metabolism , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation , Luciferases/genetics , Luciferases/metabolism , Molecular Sequence Data , Mutation , Protein Binding , Proto-Oncogene Proteins c-ret , RNA/genetics , RNA/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Ribonucleases/metabolism , Sequence Deletion , Sp3 Transcription Factor , Transcription, Genetic , Transcriptional Activation
17.
J Biol Chem ; 274(33): 23328-32, 1999 Aug 13.
Article in English | MEDLINE | ID: mdl-10438509

ABSTRACT

The acid-labile subunit (ALS) is a glycosylated 85-kDa member of the leucine-rich repeat (LRR) protein superfamily and circulates in ternary complexes with the insulin-like growth factors (IGFs) and their binding proteins (IGFBPs). These complexes are thought to regulate the serum IGFs by restricting IGF movement out of the circulation. However, little is known about how ALS binds to IGFBP-3 or -5, which link the IGFs to ALS. To investigate potential sites of interaction, the ALS structure has been modeled with the crystal structure of the LRR protein porcine ribonuclease inhibitor as a template. ALS is predicted to be a donut-shaped molecule with an internal diameter of 1.7 nm, an external diameter of 7.2 nm, and a thickness of 3.6 nm. These dimensions are supported by rotary shadowing electron microscopy of ALS. The internal face is lined with a substantial region of electronegative surface potential that could interact with the positively charged region on IGFBP-3 known to be involved in ALS binding. The model also predicts that three potential N-linked oligosaccharide sites within the LRR domain are clustered together, which may be important in light of recent studies showing ALS glycan involvement in complex formation with IGFBP-3.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/chemistry , Insulin-Like Growth Factor Binding Protein 5/chemistry , Acids , Amino Acid Sequence , Carbohydrates/chemistry , Humans , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Protein Conformation , Static Electricity
18.
Am J Physiol ; 277(2): E245-52, 1999 08.
Article in English | MEDLINE | ID: mdl-10444419

ABSTRACT

There is little information on free insulin-like growth factor I (IGF-I) and its regulatory proteins during fasting and refeeding. Therefore, we examined rats during fasting (0, 1, 2, and 3 days) and refeeding (3, 6, and 12 h and 1, 2, 3, and 7 days) (n = 6-9). Serum was analyzed for insulin, C-peptide, growth hormone (GH), free and total IGF-I, IGF-binding protein (IGFBP)-1 and -3, and the acid-labile subunit (ALS). Additionally, liver mRNA for IGF-I, IGFBP-1, and ALS was determined. Fasting reduced serum levels of GH, free and total IGF-I, IGFBP-3, and ALS, whereas IGFBP-1 was increased (P < 0.0001). Refeeding normalized IGFBP-1 at 3 h and GH at 12 h. Free IGF-I changed in parallel with total IGF-I, ALS, and IGFBP-3, being normalized at 48 h of refeeding. IGFBP-1 (peptide and mRNA) correlated inversely with insulin and C-peptide (P < 0.001). The correlation between peptide and mRNA was relatively strong for IGFBP-1 (r(2) = 0.36; P < 0.0001), moderate for IGF-I (r(2) = 0.18; P < 0.0005), and insignificant for ALS. In conclusion, insulin appears to regulate IGFBP-1 in fasted and refed rats. However, the normal inverse relationship between free IGF-I and IGFBP-1 was absent, and free IGF-I changed in parallel with total IGF-I and thus ALS and IGFBP-3. Finally, the regulation of the hepatic synthesis of IGF-I, IGFBP-1, and ALS seems to differ substantially.


Subject(s)
Animal Feed , Fasting/physiology , Somatomedins/metabolism , Animals , C-Peptide/blood , Carrier Proteins/blood , Carrier Proteins/genetics , Glycoproteins/blood , Glycoproteins/genetics , Human Growth Hormone/blood , Insulin/blood , Insulin-Like Growth Factor Binding Protein 1/blood , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/genetics , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors
19.
J Clin Endocrinol Metab ; 84(2): 553-60, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10022415

ABSTRACT

We investigated the acute (4-5 h) and short-term (5 days) effects of GH treatment on hepatic messenger RNA (mRNA) levels of the genes for the insulin-like growth factors (IGFs), insulin-like growth factor binding protein-1, -2, and -3 (IGFBPs), and the acid labile subunit (ALS), as well as serum levels of these proteins in humans. At the mRNA level, we observed an increase in IGF-1 transcription (+173%) following GH treatment in the acute group, which remained elevated in the short-term treatment group. IGFBP-2 mRNA decreased after short-term GH treatment, without changes in IGFBP-1 or -3 expression. The ALS transcript level increased after 5 days. In serum, we found increased levels of IGF-I and insulin, and decreased levels of IGF-II, in the short-term treatment group. IGFBP-1 decreased in both treatment groups, whereas IGFBP-2 was reduced after 5 days treatment. ALS increased in the short-term group. We observed increased IGFBP-3 serum levels after 5 days of GH treatment, likely due to increased formation of the ternary complex. Our results show that the metabolic effects by GH on the IGF axis are complex. In addition to a direct stimulation of IGF-I and ALS expression, GH inhibits IGFBP-1 serum levels and IGFBP-2 expression in an indirect manner, possibly facilitating enhanced IGF bioavailability to target tissues.


Subject(s)
Human Growth Hormone/pharmacology , Insulin-Like Growth Factor Binding Proteins/blood , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , RNA, Messenger/metabolism , Adult , Carrier Proteins/blood , Carrier Proteins/genetics , Female , Glycoproteins/blood , Glycoproteins/genetics , Human Growth Hormone/administration & dosage , Humans , Insulin-Like Growth Factor Binding Protein 1/blood , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 2/blood , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor II/genetics , Male , Middle Aged
20.
J Biol Chem ; 274(9): 5292-8, 1999 Feb 26.
Article in English | MEDLINE | ID: mdl-10026136

ABSTRACT

Over 75% of the circulating insulin-like growth factors (IGF-I and -II) are bound in 140-kDa ternary complexes with IGF-binding protein-3 (IGFBP-3) and the 84-86-kDa acid-labile subunit (ALS), a glycoprotein containing 20 kDa of carbohydrate. The ternary complexes regulate IGF availability to the tissues. Since interactions of glycoproteins can be influenced by their glycan moieties, this study aimed to determine the role of ALS glycosylation in ternary complex formation. Complete deglycosylation abolished the ability of ALS to associate with IGFBP-3. To examine this further, seven recombinant ALS mutants each lacking one of the seven glycan attachment sites were expressed in CHO cells. All the mutants bound IGFBP-3, demonstrating that this interaction is not dependent on any single glycan chain. Enzymatic desialylation of ALS caused a shift in isoelectric point from 4.5 toward 7, demonstrating a substantial contribution of anionic charge by sialic acid. Ionic interactions are known to be involved in the association between ALS and IGFBP-3. Desialylation reduced the affinity of ALS for IGFBP-3. IGF complexes by 50-80%. Since serum protein glycosylation is often modified in disease states, the dependence of IGF ternary complex formation on the glycosylation state of ALS suggests a novel mechanism for regulation of IGF bioavailability.


Subject(s)
Carrier Proteins/metabolism , Glycoproteins/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , N-Acetylneuraminic Acid/metabolism , Somatomedins/metabolism , Animals , Base Sequence , CHO Cells , Cricetinae , DNA Primers , Glycoside Hydrolases/metabolism , Glycosylation , Neuraminidase/metabolism , Protein Binding , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...