Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.440
Filter
1.
Front Endocrinol (Lausanne) ; 14: 1026187, 2023.
Article in English | MEDLINE | ID: mdl-36864831

ABSTRACT

Background: Gene expression (GE) data have shown promise as a novel tool to aid in the diagnosis of childhood growth hormone deficiency (GHD) when comparing GHD children to normal children. The aim of this study was to assess the utility of GE data in the diagnosis of GHD in childhood and adolescence using non-GHD short stature children as a control group. Methods: GE data was obtained from patients undergoing growth hormone stimulation testing. Data were taken for the 271 genes whose expression was utilized in our previous study. The synthetic minority oversampling technique was used to balance the dataset and a random forest algorithm applied to predict GHD status. Results: 24 patients were recruited to the study and eight subsequently diagnosed with GHD. There were no significant differences in gender, age, auxology (height SDS, weight SDS, BMI SDS) or biochemistry (IGF-I SDS, IGFBP-3 SDS) between the GHD and non-GHD subjects. A random forest algorithm gave an AUC of 0.97 (95% CI 0.93 - 1.0) for the diagnosis of GHD. Conclusion: This study demonstrates highly accurate diagnosis of childhood GHD using a combination of GE data and random forest analysis.


Subject(s)
Dwarfism , Growth Hormone , Transcriptome , Adolescent , Child , Humans , Control Groups , Gene Expression Profiling , Growth Hormone/deficiency
2.
Eur J Endocrinol ; 188(1)2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36651164

ABSTRACT

OBJECTIVES: Insulin resistance is associated with ectopic lipid deposition. Growth hormone (GH) status also modulates ectopic lipid accumulation, but how this associates with insulin resistance in patients with GH disorders is not well established. DESIGN AND METHODS: Twenty-one patients diagnosed with acromegaly and 12 patients with adult GH deficiency (GHD) were studied at diagnosis and after treatment. A reference group of 12 subjects was included. Each study day comprised assessment of body composition with dual-energy X-ray absorptiometry, ectopic lipid deposition in the liver by MR spectroscopy, and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR). RESULTS: Disease control of acromegaly decreased lean body mass (LBM) (P < .000) and increased the percentage of total body fat (TBF) (P < .000). GH replacement increased LBM in the GHD patients (P = .007) and decreased the percentage of TBF (P = .010). The intrahepatic lipid (IHL) content increased after disease control in acromegaly (P = .004), whereas IHL did not change significantly after GH replacement in GHD (P = .34). Insulin resistance (HOMA-IR) improved after disease control of acromegaly (P < .000) and remained unaltered after GH replacement in the GHD patients (P = .829). CONCLUSIONS: GH status is a significant modulator of body composition and insulin sensitivity.GH excess reduces total fat mass and intrahepatic lipid content together with induction of insulin resistance.The data support the notion that GH-induced insulin resistance is unassociated with hepatic lipid accumulation.


Subject(s)
Acromegaly , Growth Hormone , Human Growth Hormone , Insulin Resistance , Adult , Humans , Acromegaly/drug therapy , Acromegaly/complications , Body Composition , Growth Hormone/deficiency , Human Growth Hormone/therapeutic use , Insulin-Like Growth Factor I/metabolism , Lipids
4.
Horm Metab Res ; 54(12): 827-832, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36257327

ABSTRACT

Adult growth hormone deficiency (AGHD) is associated with increased cardiovascular risks. The primary endpoint of this retrospective cohort study was to compare metabolic profile and echocardiographic parameters in childhood-onset (CO) and adulthood-onset (AO) AGHD patients. 26 patients with CO AGHD (19 males, 26.8±8.5 years) and 41 patients with AO AGHD (23 males, 35.1±6.8 years) were included. The standard deviation score of insulin-like growth factor-1 (IGF-1 SDS), metabolic profile, liver sonography and echocardiographic parameters were compared. The IGF-1 SDS were significantly lower in CO AGHD patients (p<0.05). AO AGHD patients showed much profound glycolipid aberrations, elevated C-reactive protein levels (p=0.012), and proportionally higher prevalence of non-alcoholic fatty liver disease (95.2% vs.8.3%, p<0.001). In all subjects, IGF-1 SDS was negatively related to low-density lipoprotein cholesterol, and positively related to total cholesterol and lipoprotein a (Lpa). There were significant differences between the two group with regard to the correlations between IGF-1 SDS and high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, LPa, uric acid and creatinine (all p for interaction<0.05). CO AGHD patients exhibited smaller heart, but similar ventricular ejection fraction compared to AO AGHD patients. AGHD may be a group of heterogenous entity based on the onset ages of disease. AO patients had prominent metabolic disorders, while CO patients had smaller heart but similar cardiac performance. Whether growth hormone replacement therapy will equally benefit both CO and AO AGHD patients needs further investigations.


Subject(s)
Growth Hormone , Insulin-Like Growth Factor I , Metabolome , Adult , Humans , Cholesterol , Growth Hormone/deficiency , Lipoproteins, LDL , Retrospective Studies , Lipoprotein(a) , Male , Female , Adolescent , Young Adult
5.
Contrast Media Mol Imaging ; 2022: 3111585, 2022.
Article in English | MEDLINE | ID: mdl-36003997

ABSTRACT

This study aims to explore the magnetic resonance imaging (MRI) findings of the pituitary gland (PG) in children with growth hormone deficiency (GHD) and their correlation with the growth hormone (GH) peak during clinical GH stimulation tests. Sixty-one children with GHD diagnosed and treated between December 2018 and December 2021 were retrospectively analyzed in terms of clinical and pituitary morphological MRI data. MRI measurements of various diameters of the adenohypophysis (AH) were obtained to analyze the differences of the measured values in different genders and age groups, as well as their relationship with the GH peak in GH stimulation tests. Among the 61 children with GHD, the superior PG margin was protuberant in 2 cases, flat in 13 cases, and concave in 46 cases. The three age groups showed similar pituitary morphology and stalk (P > 0.05). On T1-weighted images, the proportion of isointensity was lower while the proportion of slightly-low signal intensity was higher in the anterior pituitary gland (APG) of children aged >10 compared with those aged 7-10. The comparison of AH linear parameters and GH peak values of male patients among different age groups showed that the anteroposterior (sagittal) diameter of AH and GH peak were the highest in the >10-year-old group and the lowest in the ≤6-year-old group, with those of the 7-10-year-old group in between (P < 0.05). In females, the anteroposterior (sagittal) diameter and GH peak were higher in the 7-10-year-old group and >10-year-old group compared with the ≤6-year-old group (P < 0.05). The MRI coronal and sagittal heights of PG in children with GHD were positively correlated with the GH peak value. In conclusion, in GHD patients, the coronal and sagittal heights as well as the coronal width of AH do not change with sex or age, but the coronal and sagittal heights of PG are positively correlated with the GH peak of GH stimulation tests, which has high application value in the diagnosis of children with GHD.


Subject(s)
Growth Hormone , Human Growth Hormone , Pituitary Gland , Child , Female , Growth Hormone/deficiency , Growth Hormone/pharmacology , Human Growth Hormone/deficiency , Human Growth Hormone/pharmacology , Humans , Magnetic Resonance Imaging , Male , Pituitary Gland/diagnostic imaging , Pituitary Gland/pathology , Retrospective Studies
6.
Geroscience ; 44(6): 2885-2893, 2022 12.
Article in English | MEDLINE | ID: mdl-35900661

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia, affecting approximately 6.5 million Americans age 65 or older. AD is characterized by increased cognitive impairment and treatment options available provide minimal disease attenuation. Additionally, diagnostic methods for AD are not conclusive with definitive diagnoses requiring postmortem brain evaluations. Therefore, miRNAs, a class of small, non-coding RNAs, have garnered attention for their ability to regulate a variety of mRNAs and their potential to serve as both therapeutic targets and biomarkers of AD. Several miRNAs have already been implicated with AD and have been found to directly target genes associated with AD pathology. The APP/PS1 mice is an AD model that expresses the human mutated form of the amyloid precursor protein (APP) and presenilin-1 (PS1) genes. In a previous study, it was identified that crossing long-living growth hormone (GH)-deficient Ames dwarf (df/df) mice with APP/PS1 mice provided protection from AD through a reduction in IGF-1, amyloid-ß (Aß) deposition, and gliosis. Hence, we hypothesized that changes in the expression of miRNAs associated with AD mediated such benefits. To test this hypothesis, we sequenced miRNAs in hippocampi of df/df, wild type (+ / +), df/ + /APP/PS1 (phenotypically normal APP/PS1), and df/df/APP/PS1 mice. Results of this study demonstrated significantly upregulated and downregulated miRNAs between df/df/APP/PS1 and df/ + /APP/PS1 mice that suggest the df/df mutation provides protection from AD progression. Additionally, changes in miRNA expression with age were identified in both df/df and wild-type mice as well as df/df/APP/PS1 and APP/PS1 mice, with predictive functional roles in the Pi3k-AKT/mTOR/FOXO pathways potentially contributing to disease pathogenesis.


Subject(s)
Alzheimer Disease , MicroRNAs , Aged , Animals , Humans , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases , Growth Hormone/deficiency
7.
Growth Horm IGF Res ; 65: 101469, 2022 08.
Article in English | MEDLINE | ID: mdl-35696752

ABSTRACT

OBJECTIVE: To understand the growth of teeth and mandibular and maxillary bones in subjects with isolated growth hormone deficiency (IGHD). MATERIAL AND METHODS: Mesiodistal tooth width of 28 maxillary and mandibular dental models of 14 adult IGHD subjects (9 men) were digitalized and compared to 40 models of 20 normal-statured controls (11 men). The mean SDS of the maxillary and mandibular teeth were compared with height, cephalic perimeter, total anterior facial height, total maxillary and mandibular length, and maxillary and mandibular arches. RESULTS: All average mesiodistal dimensions in absolute values of the 14 dental pairs were reduced in the IGHD group. Eight of 28 (28.6%) mesiodistal dimensions in IGHD subjects of both sexes had an average SDS below -2, thirteen of them (46.4%) had mean SDS between -1 and - 2, and seven of them (25.0%) had SDS above -1. The highest SDS values were the upper lateral incisor (-0.32 in women), and the upper canine (-0.91 in men). The lowest SDS values were the 2nd upper molar (-3.51 in men), and the 2nd upper premolar (-2.64 in women). The ascending order of the mean SDS was height, total maxillary length, total mandibular length, total anterior height of the face, cephalic perimeter, the maxillary arches width, the mesiodistal width of the mandibular teeth, the mesiodistal width of the maxillary teeth and the mandibular arches width. CONCLUSIONS: Reduction in mesiodistal width is present in untreated IGHD adults with magnitude of tooth size reduction being lower than height, cephalic perimeter, total anterior facial height, and most jaw measurements. IGHD abolishes the sexual dimorphism in mesiodistal dental measures.


Subject(s)
Bone Development , Dwarfism, Pituitary , Growth Hormone/deficiency , Tooth/growth & development , Female , Humans , Incisor , Male , Mandible , Maxilla
9.
Endocrinol Metab (Seoul) ; 37(2): 359-368, 2022 04.
Article in English | MEDLINE | ID: mdl-35504604

ABSTRACT

BACKGROUND: Discontinuing growth hormone (GH) treatment during the transition to adulthood has been associated with adverse health outcomes in patients with childhood-onset growth hormone deficiency (CO-GHD). This study investigated the metabolic changes associated with interrupting GH treatment in adolescents with CO-GHD during the transition period. METHODS: This study included 187 patients with CO-GHD who were confirmed to have adult GHD and were treated at six academic centers in Korea. Data on clinical parameters, including anthropometric measurements, metabolic profiles, and bone mineral density (BMD) at the end of childhood GH treatment, were collected at the time of re-evaluation for GHD and 1 year after treatment resumption. RESULTS: Most patients (n=182, 97.3%) had organic GHD. The median age at treatment discontinuation and re-evaluation was 15.6 and 18.7 years, respectively. The median duration of treatment interruption was 2.8 years. During treatment discontinuation, body mass index Z-scores and total cholesterol, low-density lipoprotein, and non-high-density lipoprotein (HDL) cholesterol levels increased, whereas fasting glucose levels decreased. One year after GH treatment resumption, fasting glucose levels, HDL cholesterol levels, and femoral neck BMD increased significantly. Longer GH interruption (>2 years, 60.4%) resulted in worse lipid profiles at re-evaluation. The duration of interruption was positively correlated with fasting glucose and non-HDL cholesterol levels after adjusting for covariates. CONCLUSION: GH treatment interruption during the transition period resulted in worse metabolic parameters, and a longer interruption period was correlated with poorer outcomes. GH treatment should be resumed early in patients with CO-GHD during the transition period.


Subject(s)
Growth Hormone/deficiency , Human Growth Hormone , Adolescent , Adult , Bone Density , Cholesterol , Glucose , Human Growth Hormone/administration & dosage , Human Growth Hormone/adverse effects , Humans , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects
10.
Physiol Res ; 70(Suppl 1): S61-S68, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34918530

ABSTRACT

There are only few studies concerning about long-term effect of growth hormone (GH) replacement therapy on bone mineral density and bone microstructure. To assess effect of GH replacement therapy on bone mineral density (BMD) and trabecular bone score (TBS) in adult GH deficient (AGHD) subjects over period of 10 years. From 2005 to 2018, a prospective study of AGHD patients was conducted in national referral center for treatment of GHD. All patients received subcutaneous recombinant human GH in an IGF 1-normalizing regimen once a day. Lumbar spine (L-spine) and total hip (TH) BMD using Hologic densitometers were measured at baseline and every two years during treatment with rhGH. TBS was derived from L1-L4 DXA using iNsight® software (Medimaps, France) at each time point. Periods of measurement were baseline, year 2; 4; 6; 8 and 10. In total, 63 patients (38 males, 25 females, mean age 25.1±16 years) were included in the study. After 10 years of GH treatment, IGF-1 significantly increased (~35 %), with greatest increase at year 2. During 10-year follow-up, L-spine BMD increased approximately of 7 % (NS). TH BMD increase of 11 % during follow-up (p=0.0003). The greatest increment of BMD was achieved at year 6 on both sites, L-spine (+6 %) and TH BMD (+13 %) (p<0.05). There was no significant change of TBS during whole follow-up. In this study, sustaining positive effect of GH replacement therapy on bone density in subjects with adult GH deficiency over 10 years of follow-up was observed. The study did not show effect on TBS, as indirect measure of trabecular bone microarchitecture.


Subject(s)
Bone Density/drug effects , Cancellous Bone/drug effects , Growth Hormone/deficiency , Human Growth Hormone/pharmacology , Adolescent , Adult , Child , Female , Follow-Up Studies , Hormone Replacement Therapy , Human Growth Hormone/therapeutic use , Humans , Male , Prospective Studies , Young Adult
11.
Mol Genet Genomic Med ; 9(11): e1821, 2021 11.
Article in English | MEDLINE | ID: mdl-34623774

ABSTRACT

BACKGROUND: Unbalanced translocations between the q arm of chromosomes 5 and 13 are exceedingly rare and there is only one reported case with distal trisomy 5q/monosomy 13q. In this report, we describe a second patient with a similar rearrangement arising from a paternal balanced translocation. METHODS: Karyotype analysis was performed on the proband and their parents. Microarray was also conducted on the proband. RESULTS: Our patient was found to have global developmental delay, distinct facial features, short stature, growth hormone deficiency, delayed puberty, and brain anomalies including a small pituitary. Karyotype and microarray analysis revealed a terminal duplication of chromosome regions 5q33.3 to 5qter and a terminal deletion of chromosome regions 13q34 to 13qter that resulted from a balanced translocation in her father. The endocrine abnormalities and neuroimaging findings have not been previously described in patients with either copy number change. CONCLUSIONS: This case helps expand on the phenotype of patients with distal trisomy 5q/monosomy 13q as well as possibly providing useful information on the more common individual copy number changes.


Subject(s)
Brain , Chromosome Disorders , Growth Hormone , Translocation, Genetic , Brain/diagnostic imaging , Brain/pathology , Chromosome Deletion , Chromosome Disorders/diagnosis , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 5/genetics , Female , Growth Hormone/deficiency , Humans , Trisomy
12.
Life Sci ; 285: 119970, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34562435

ABSTRACT

Growth hormone (GH) deficiency is a common cause of late sexual maturation and fertility issues. To determine whether GH-induced effects on reproduction are associated with alterations in hypothalamic kisspeptin system, we studied the male reproduction in two distinct GH deficiency mouse models. In the first model, mice present GH deficiency secondary to arcuate nucleus of the hypothalamus (ARH) lesions induced by posnatal monosodium glutamate (MSG) injections. MSG-induced ARH lesions led to significant reductions in hypothalamic Ghrh mRNA expression and consequently growth. Hypothalamic Kiss1 mRNA expression and Kiss1-expressing cells in the ARH were disrupted in the MSG-treated mice. In contrast, kisspeptin immunoreactivity remained preserved in the anteroventral periventricular and rostral periventricular nuclei (AVPV/PeN) of MSG-treated mice. Importantly, ARH lesions caused late sexual maturation and infertility in male mice. In our second mouse model, we studied animals profound GH deficiency due to a loss-of-function mutation in the Ghrhr gene (Ghrhrlit/lit mice). Interestingly, although Ghrhrlit/lit mice exhibited late puberty onset, hypothalamic Kiss1 mRNA expression and hypothalamic kisspeptin fiber density were normal in Ghrhrlit/lit mice. Despite presenting dwarfism, the majority of Ghrhrlit/lit male mice were fertile. These findings suggest that spontaneous GH deficiency during development does not compromise the kisspeptin system. Furthermore, ARH Kiss1-expressing neurons are required for fertility, while AVPV/PeN kisspeptin expression is sufficient to allow maturation of the hypothalamic-pituitary-gonadal axis in male mice.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Growth Hormone/deficiency , Hypothalamo-Hypophyseal System/metabolism , Kisspeptins/metabolism , Reproduction , Sexual Maturation , Animals , Dwarfism/genetics , Dwarfism/metabolism , Fertility , Kisspeptins/genetics , Male , Mice , Neurons/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism
13.
Int J Mol Sci ; 22(16)2021 Aug 23.
Article in English | MEDLINE | ID: mdl-34445772

ABSTRACT

In this review we described the interactions between ghrelin and the growth hormone (GH)-insulin-like growth factor 1 (IGF-1) axis in children and adults with growth hormone deficiency (GHD). A possible involvement of these interactions in the pathogenesis of unexplained cases of GHD was suggested. Current research provides more and more details to the knowledge on the circadian rhythm of ghrelin. We gathered reports on the decreasing effect of Helicobacter pylori-related chronic gastritis on the number of ghrelin immunopositive cells and the consequent decrease in ghrelin serum concentration. The gastrointestinal tract microflora modification of the ghrelin action, by the mechanism of molecular mimicry, was also stressed. Moreover, the mutual relationships between ghrelin and the TSH-FT4/FT3 axis in growth and metabolic processes are described. It is to be recalled that FT4 and FT3 exert a permissive impact on IGF-1 action and, in turn, GH, in reaction mediated by IGF-1, enhances the monodeiodination of FT4 to FT3. Finally, we discussed the latest attempts to use the GH secretagogue receptor (GHS-R) analogues for possible diagnostic and therapeutic purposes.


Subject(s)
Ghrelin/metabolism , Growth Hormone/deficiency , Growth Hormone/metabolism , Animals , Humans , Insulin-Like Growth Factor I/metabolism , Receptors, Ghrelin/metabolism
14.
Horm Res Paediatr ; 94(7-8): 239-250, 2021.
Article in English | MEDLINE | ID: mdl-34438400

ABSTRACT

BACKGROUND/AIMS: Diagnosis of growth hormone deficiency (GHD) in children requires the use of provocative growth hormone (GH) stimulation tests, which can have limited reliability and are potentially contraindicated in some patients. This is the first paediatric study to test the safety, tolerability, and pharmacokinetics (PK)/pharmacodynamics (PD) of macimorelin, an oral GH secretagogue, approved for diagnosis of adult GHD. METHODS: In this open-label, group comparison, single-dose escalation trial (EudraCT 2018-001988-23), sequential cohorts of patients (C1-C3) received ascending single doses of macimorelin: 0.25 (C1), 0.5 (C2), and 1.0 (C3) mg/kg. Primary endpoints were safety and tolerability, and secondary endpoints were PK/PD. RESULTS: Twenty-four patients aged between 2 and <18 with suspected GHD participated in the study. No macimorelin-related adverse events were reported, and macimorelin was well tolerated. Plasma macimorelin concentrations increased with dose: mean areas under the curve were 6.69 (C1), 18.02 (C2), and 30.92 (C3) h × ng/mL; mean maximum concentrations were 3.46 (C1), 8.13 (C2), and 12.87 (C3) ng/mL. GH concentration increased following macimorelin administration: mean times of maximum measured concentration were 52.5 (C1), 37.5 (C2), and 37.5 (C3) min. CONCLUSION: All 3 doses of macimorelin had excellent safety and tolerability with PK/PD profiles in expected ranges. These results support the use of 1.0 mg/mL macimorelin in a Phase 3 test validation trial in children.


Subject(s)
Dose-Response Relationship, Drug , Growth Hormone , Indoles/administration & dosage , Pediatrics , Tryptophan/analogs & derivatives , Child , Female , Ghrelin , Growth Hormone/deficiency , Growth Hormone/drug effects , Humans , Indoles/pharmacokinetics , Male , Reproducibility of Results , Surveys and Questionnaires , Tryptophan/administration & dosage , Tryptophan/pharmacokinetics
15.
J Clin Lipidol ; 15(5): 712-723, 2021.
Article in English | MEDLINE | ID: mdl-34462238

ABSTRACT

BACKGROUND: Elevated plasma concentrations of hepatic- and intestinally-derived triglyceride-rich lipoproteins (TRL) are implicated in the pathogenesis of atherosclerotic cardiovascular disease and all-cause mortality. Excess of TRL is the driving cause of atherogenic dyslipidemia commonly occurring in insulin-resistant individuals such as patients with obesity, type 2 diabetes and metabolic syndrome. Interestingly, growth hormone (GH)-deficient individuals display similar atherogenic dyslipidemia, suggesting an important role of GH and GH deficiency in the regulation of TRL metabolism. OBJECTIVE: We aimed to examine the direct and/or indirect role of GH on TRL metabolism. METHODS: We investigated the effect on fasting and postprandial hepatic-TRL and intestinal-TRL metabolism of short-term (one month) withdrawal of GH in 10 GH-deficient adults. RESULTS: After GH withdrawal, we found a reduction in fasting plasma TRL concentration (significant decrease in TRL-TG, TRL-cholesterol, TRL-apoB-100, TRL-apoC-III and TRL-apoC-II) but not in postprandial TRL response. This reduction was due to fewer fasting TRL particles without a change in TG per particle and was not accompanied by a change in postprandial TRL-apoB-48 response. Individual reductions in TRL correlated strongly with increases in insulin sensitivity and decreases in TRL-apoC-III. CONCLUSION: In this relatively short term 'loss of function' human experimental model, we have shown an unanticipated reduction of hepatic-TRL particles despite increase in total body fat mass and reduction in lean mass. These findings contrast with the atherogenic dyslipidemia previously described in chronic GH deficient states, providing a new perspective for the role of GH in lipoprotein metabolism.


Subject(s)
Coronary Artery Disease/etiology , Dyslipidemias/etiology , Growth Hormone/physiology , Intestines/metabolism , Lipoproteins/blood , Lipoproteins/metabolism , Liver/metabolism , Triglycerides/blood , Triglycerides/metabolism , Adult , Cause of Death , Coronary Artery Disease/mortality , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Female , Growth Hormone/deficiency , Humans , Insulin Resistance/physiology , Male , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Middle Aged , Obesity/complications , Obesity/metabolism
16.
J Korean Med Sci ; 36(18): e133, 2021 May 10.
Article in English | MEDLINE | ID: mdl-33975400

ABSTRACT

Weiss-Kruszka syndrome (WSKA), caused by heterozygous loss-of-function variants in ZNF462 gene, is a recently described and extremely rare genetic disorder. The main phenotypes include characteristic craniofacial features, ptosis, dysgenesis of the corpus callosum, and neurodevelopmental impairment. We report the first Korean boy with molecularly confirmed WSKA presenting with an atypical manifestation. A 16-year-old boy with a history of bilateral ptosis surgery presented with short stature (-3.49 standard deviation score) and delayed puberty. The patient showed characteristic craniofacial features including an inverted triangular-shaped head, exaggerated Cupid's bow, arched eyebrows, down-slanting palpebral fissures, and poorly expressive face. He had a mild degree of intellectual disability and mild hypotonia. Endocrine studies in the patient demonstrated complete growth hormone deficiency (GHD) associated with empty sella syndrome (ESS), based on a magnetic resonance imaging study for the brain that showed a flattened pituitary gland and cerebrospinal fluid space herniated into the sella turcica. To identify the genetic cause, we performed whole exome sequencing (WES). Through WES, a novel de novo heterozygous nonsense variant, c.4185del; p.(Met1396Ter) in ZNF462 was identified. This is the first case of WSKA accompanied by primary ESS associated with GHD. More clinical and functional studies are needed to elucidate this association.


Subject(s)
Empty Sella Syndrome/complications , Growth Hormone/deficiency , Intellectual Disability/genetics , Adolescent , DNA-Binding Proteins/genetics , Empty Sella Syndrome/diagnosis , Growth Hormone/genetics , Humans , Male , Nerve Tissue Proteins/genetics , Transcription Factors/genetics
17.
Am J Physiol Endocrinol Metab ; 320(6): E1158-E1172, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33938235

ABSTRACT

In the endocrine pancreas, growth hormone (GH) is known to promote pancreatic islet growth and insulin secretion. In this study, we show that GH receptor (GHR) loss in the germline and in adulthood impacts islet mass in general but more profoundly in male mice. GHR knockout (GHRKO) mice have enhanced insulin sensitivity and low circulating insulin. We show that the total cross-sectional area of isolated islets (estimated islet mass) was reduced by 72% in male but by only 29% in female GHRKO mice compared with wild-type controls. Also, islets from GHRKO mice secreted ∼50% less glucose-stimulated insulin compared with size-matched islets from wild-type mice. We next used mice with a floxed Ghr gene to knock down the GHR in adult mice at 6 mo of age (6mGHRKO) and examined the impact on glucose and islet metabolism. By 12 mo of age, female 6mGHRKO mice had increased body fat and reduced islet mass but had no change in glucose tolerance or insulin sensitivity. However, male 6mGHRKO mice had nearly twice as much body fat, substantially reduced islet mass, and enhanced insulin sensitivity, but no change in glucose tolerance. Despite large losses in islet mass, glucose-stimulated insulin secretion from isolated islets was not significantly different between male 6mGHRKO and controls, whereas isolated islets from female 6mGHRKO mice showed increased glucose-stimulated insulin release. Our findings demonstrate the importance of GH to islet mass throughout life and that unique sex-specific adaptations to the loss of GH signaling allow mice to maintain normal glucose metabolism.NEW & NOTEWORTHY Growth hormone (GH) is important for more than just growth. GH helps to maintain pancreatic islet mass and insulin secretion throughout life. Sex-specific adaptations to the loss of GH signaling allow mice to maintain normal glucose regulation despite losing islet mass.


Subject(s)
Germ Cells/metabolism , Growth Hormone/deficiency , Islets of Langerhans/growth & development , Islets of Langerhans/physiology , Receptors, Somatotropin/genetics , Age Factors , Animals , Cell Proliferation/genetics , Female , Germ Cells/physiology , Growth Hormone/genetics , Growth Hormone/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/genetics , Receptors, Somatotropin/deficiency , Receptors, Somatotropin/metabolism , Sex Characteristics , Signal Transduction/genetics
18.
J Endocrinol Invest ; 44(12): 2809-2817, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34003462

ABSTRACT

PURPOSE: Many patients who undergo bone marrow transplantation (BMT) in adulthood experience unexplained chronic fatigue which can have a major impact on their health-related quality of life (QoL). Pre-BMT treatment regimens increase the risk of developing acquired growth hormone deficiency (GHD), which results in a clinical syndrome with decreased energy and has additionally been linked to metabolic syndrome. METHODS: Using the gold-standard insulin hypoglycemic test (IHT), we evaluated the prevalence of GHD in 18 post-BMT adult patients with unexplained chronic fatigue, as well as the correlation between peak serum GH response and QoL scores, the metabolic syndrome, and insulin resistance. Peak serum GH cut-point less than 3.0 ug/L was used for the diagnosis of severe GHD. The Fatigue Severity Scale and Quality of Life in Adult GHD Assessment questionnaires were used to quantify fatigue symptoms. RESULTS: The prevalence of severe GHD within this sample of 18 patients was 50%. A trend between lower peak serum GH response and higher fatigue and QoL-AGHDA scores was observed. CONCLUSIONS: GHD may represent a remediable contributor to post-BMT chronic fatigue in adults, further studies are needed to evaluate the potential role of screening and GH replacement therapy in this vulnerable patient population. IMPLICATIONS FOR CANCER SURVIVORS: GHD may be a treatable explanation for disabling post-BMT fatigue pending results of intervention studies.


Subject(s)
Bone Marrow Transplantation/adverse effects , Fatigue Syndrome, Chronic , Growth Hormone , Metabolic Syndrome , Postoperative Complications , Quality of Life , Bone Marrow Transplantation/methods , Cancer Survivors , Cross-Sectional Studies , Fatigue Syndrome, Chronic/etiology , Fatigue Syndrome, Chronic/metabolism , Fatigue Syndrome, Chronic/psychology , Fatigue Syndrome, Chronic/therapy , Female , Growth Hormone/blood , Growth Hormone/deficiency , Hormone Replacement Therapy/methods , Humans , Insulin Resistance , Male , Metabolic Syndrome/diagnosis , Metabolic Syndrome/etiology , Middle Aged , Postoperative Complications/diagnosis , Postoperative Complications/metabolism , Postoperative Complications/psychology , Postoperative Complications/therapy , Prevalence , Surveys and Questionnaires , Symptom Assessment/methods
19.
J Manag Care Spec Pharm ; 27(8): 1118-1128, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33896224

ABSTRACT

BACKGROUND: Pediatric growth hormone deficiency (GHD) is a rare disorder of short stature that is currently treated with daily injections of somatropin. In addition to short stature, GHD is associated with other comorbidities such as impaired musculoskeletal development, cardiovascular disease, and decreased quality of life. OBJECTIVE: To analyze somatropin utilization, adherence, and health care costs among children with GHD who had either Medicaid or commercial health insurance. METHODS: Children (aged < 18 years) with a GHD diagnosis between January 1, 2008, and December 31, 2017, were identified in the IBM MarketScan Commercial and Medicaid databases. Patients with at least 12- and 6-month continuous enrollment pre- and postdiagnosis were eligible. Children with GHD were direct matched (1:3) to controls without GHD (or other short stature-related disorders) on age, gender, plan type, region, and race (Medicaid only). Index date was the date of the first GHD diagnosis during the selection window for GHD patients and using random assignment for controls. Patients were followed until the end of continuous database enrollment or December 31, 2018. Baseline comorbidities and medications were measured during the 12 months pre-index, whereas somatropin treatment patterns along with all-cause and GHD-related health care costs were measured during the variable follow-up period. Multivariable modeling was used to compare costs between GHD patients and controls and between somatropin-treated and -untreated GHD patients while adjusting for baseline characteristics. RESULTS: There were 6,820 Medicaid and 14,070 commercial patients with GHD who met the study inclusion criteria. Mean (SD) age at index was 9.5 (4.5) years for Medicaid patients and 11.1 (3.7) years for commercial patients. The majority of patients were male (> 65%), and mean follow-up time for all cases and controls was 3-4 years. Overall, 63.2% of Medicaid and 68.4% of commercial GHD patients were treated with somatropin at some point during follow-up. Among Medicaid GHD patients, the treatment rate was highest among White males and lowest among Black females. Adherence was low as the proportion of days covered was ≥ 80% for only 18.4% of Medicaid patients and 32.3% of commercial patients and 49.1% of treated Medicaid and 24.3% of treated commercial patients discontinued before turning age 13. After adjusting for baseline characteristics, all-cause non-somatropin annualized costs were 5.67 times higher (Δ$19,309) for Medicaid GHD patients and 5.46 times higher (Δ$12,305) for commercial GHD patients than matched non-GHD controls. Adjusted all-cause non-somatropin annualized costs were 0.59 times lower (Δ$14,416) for treated Medicaid patients and 0.69 times lower (Δ$7,650) for treated commercial patients than for untreated patients. CONCLUSIONS: Pediatric GHD presents a significant health care burden, and many patients remain untreated or undertreated. Untreated GHD was associated with higher non-somatropin health care costs than treated GHD. Strategies to optimize treatment and improve adherence may reduce the health care burden faced by these patients. DISCLOSURES: This study was funded by Ascendis Pharma, Inc. Smith and Pitukcheewanont are employed by Ascendis Pharma, Inc. Manjelievskaia, Lopez-Gonzalez, and Morrow are employed by IBM Watson Health, which received funding from Ascendis Pharma, Inc., to conduct this study. Kaplowitz is a paid consultant of Ascendis Pharma, Inc.


Subject(s)
Cost of Illness , Growth Hormone/deficiency , Health Care Costs , Human Growth Hormone/economics , Adolescent , Child , Child, Preschool , Databases, Factual , Female , Human Growth Hormone/therapeutic use , Humans , Insurance Coverage , Male , Medicaid , United States
20.
Eur J Endocrinol ; 185(1): 1-12, 2021 May 21.
Article in English | MEDLINE | ID: mdl-33886498

ABSTRACT

BACKGROUND: POU1F1 encodes both PIT-1α, which plays pivotal roles in pituitary development and GH, PRL and TSHB expression, and the alternatively spliced isoform PIT-1ß, which contains an insertion of 26-amino acids (ß-domain) in the transactivation domain of PIT-1α due to the use of an alternative splice acceptor at the end of the first intron. PIT-1ß is expressed at much lower levels than PIT-1α and represses endogenous PIT-1α transcriptional activity. Although POU1F1 mutations lead to combined pituitary hormone deficiency (CPHD), no patients with ß-domain mutations have been reported. RESULTS: Here, we report that a three-generation family exhibited different degrees of CPHD, including growth hormone deficiency with intrafamilial variability of prolactin/TSH insufficiency and unexpected prolactinoma occurrence. The CPHD was due to a novel POU1F1 heterozygous variant (c.143-69T>G) in intron 1 of PIT-1α (RefSeq number NM_000306) or as c.152T>G (p.Ile51Ser) in exon 2 of PIT-1ß (NM_001122757). Gene splicing experiments showed that this mutation yielded the PIT-1ß transcript without other transcripts. The lymphocyte PIT-1ß mRNA expression was significantly higher in the patients with the heterozygous mutation than a control. A luciferase reporter assay revealed that the PIT-1ß-Ile51Ser mutant repressed PIT-1α and abolished transactivation capacity for the rat prolactin promoter in GH3 pituitary cells. CONCLUSIONS: We describe, for the first time, that the PIT-1ß mutation can cause CPHD through a novel genetic mechanism, such as PIT-1ß overexpression, and that POU1F1 mutation might be associated with a prolactinoma. Analysis of new patients and long-term follow-up are needed to clarify the characteristics of PIT-1ß mutations.


Subject(s)
Hypopituitarism/genetics , Hypothyroidism/genetics , Transcription Factor Pit-1/genetics , Adolescent , Adult , Aged , Alternative Splicing , Animals , Cell Line, Tumor , Female , Growth Hormone/deficiency , HeLa Cells , Heterozygote , Humans , In Vitro Techniques , Lymphocytes/metabolism , Male , Middle Aged , Mutation , Pedigree , Pituitary Neoplasms/genetics , Prolactin/genetics , Prolactinoma/genetics , Promoter Regions, Genetic , Protein Isoforms , RNA, Messenger/metabolism , Rats , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...