Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Endocrine ; 84(3): 1116-1124, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703329

ABSTRACT

PURPOSE: The separation between the inside and outside through the skin was fundamental for the evolution of prevertebrates, which grow through extrapituitary circuits, to vertebrates, which grow through the somatotrophic axis, namely pituitary growth hormone (GH). and circulating IGF1.Individuals with untreated isolated growth hormone (GH) deficiency (IGHD) due to a mutation in the GH-releasing hormone receptor (GHRH) gene, residing in Itabaianinha, Brazil, are vulnerable to skin cancer and have reduced sweating. However other aspects of their skin physiology are still unknown. Our objectives were to evaluate the number of skin cancers, skin aging, and functional aspects of the skin in this IGHD cohort. METHODS: Twenty-six IGHD individuals and 26 controls matched by age, sex, ethnicity, and occupation were submitted to a biochemical, dermatological and a functional skin assessment by the Multi Probe Adapter Cutometer® MPA 580. RESULTS: There was no difference in the number of skin cancers and in the degrees of photodamage between the groups. The melanin content in the forearm was similar between the groups but was lower in the buttocks (p = 0.005), as well as skin resistance (p < 0.0001) and elasticity (p = 0.003), lower in the IGHD. There was no difference in hydration and sebum content between the two groups. CONCLUSION: IGHD is apparently associated with a neutral profile in terms of skin cancer and photodamage, with similar melanin on the forearm and lower buttocks, lower skin resistance and elasticity, with hydration and sebum similar to controls.


Subject(s)
Human Growth Hormone , Skin , Humans , Male , Female , Adult , Skin/metabolism , Human Growth Hormone/deficiency , Middle Aged , Skin Neoplasms , Skin Aging/physiology , Young Adult , Skin Physiological Phenomena , Dwarfism, Pituitary/epidemiology , Adolescent
2.
Clin Endocrinol (Oxf) ; 100(5): 441-446, 2024 May.
Article in English | MEDLINE | ID: mdl-38463009

ABSTRACT

OBJECTIVES: Various biases pertaining to stature account for a male sex predominance in growth hormone deficiency (GHD) cases diagnosed by endocrinology clinics. This manuscript will assess the sex distribution when biases are minimised. METHODS: Retrospective chart review was conducted on patients diagnosed with GHD between 3 and 16 years of age. The sex distribution of cases was ascertained according to: (1) peak GH (pGH) by groups; based on growth hormone provocative testing, (2) pituitary gland imaging results, and (3) isolated GHD (IGHD) versus multiple pituitary hormone deficiencies (MPHD). The relative frequency of each sex was compared according to these subgroups with significance evaluated at α = .05 level. RESULTS: Of the 5880 clinic referrals for short stature, there were 3709 boys (63%) and 2171 girls (37%). Of these, 20% of boys (n = 745) and 15.3% of girls (n = 332) underwent provocative testing for GHD. Of those tested, 39.2% of boys (n = 292) and 32.2% of girls (n = 107) were diagnosed with GHD, all p < .001. There was a male predominance in GHD cases based on pGH or GHD severity. Though not significant, girls were more likely than boys to have MPHD (p = .056), even across pGH groups (p = .06). Both boys and girls had a similar distribution of imaging abnormalities. CONCLUSION: Stratifying by sex, we found similar percentages of pituitary imaging abnormalities (including tumours) and the number of pituitary hormone deficiencies in boys and girls as the cause of GHD. For these classifications, we did not find the historically reported male sex predominance.


Subject(s)
Dwarfism, Pituitary , Human Growth Hormone , Hypopituitarism , Female , Humans , Child , Male , Retrospective Studies , Hypopituitarism/epidemiology , Dwarfism, Pituitary/epidemiology , Growth Hormone , Sex Distribution
3.
Endocrine ; 85(1): 91-98, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38498128

ABSTRACT

OBJECTIVE: Growth hormone deficiency (GHD) is the most common pituitary hormone deficiency and is one of the main causes of short stature in children and adolescents. The aim of this study is to evaluate the epidemiology of pediatric GHD worldwide, since no other systematic review has been published so far. METHODS: We searched PubMed, Embase, and Web of Science up to July 2023 to find epidemiological studies involving children with GHD. Two review authors independently screened articles, extracted data and performed the quality assessment. RESULTS: We selected 9 epidemiological studies published from 1974 to 2022. The range of prevalence was 1/1107-1/8,646. A study based on a registry of GH users in the Piedmont region (Italy) reported the highest mean prevalence. In the included studies, the mean incidence ranged from 1/28,800 to 1/46,700 cases per year. One study reported a 20-year cumulative incidence of 127/100,000 for boys and 93/100,000 for girls. Studies were heterogeneous in terms of population (age and GHD etiology) and diagnostic criteria. As for the methodological quality of included studies, all but one study satisfied the majority of the checklist items. CONCLUSIONS: The included studies are mostly European, so the provided estimates cannot be considered global. International multicentre studies are needed to compare epidemiological estimates of GHD among different ethnical groups. Considering the considerable cost of human recombinant GH, the only available therapy to treat GHD, understanding accurate epidemiological estimates of GHD in each country is fundamental for resource allocation.


Subject(s)
Human Growth Hormone , Humans , Adolescent , Child , Human Growth Hormone/deficiency , Human Growth Hormone/therapeutic use , Prevalence , Male , Female , Growth Disorders/epidemiology , Incidence , Dwarfism, Pituitary/epidemiology
4.
Endocrine ; 85(1): 347-355, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38536548

ABSTRACT

PURPOSE: Bone mineral density (BMD) impairment is one of the critical factors for long-term quality of life in adults growth hormone deficiency (AGHD). This study aims to investigate the annual changes in BMD in AGHD patients with different ages of onset and to identify predicting factors that influence BMD. METHODS: AGHD patients (n = 160) with available data for 4 years follow-up from a major tertiary medical center in China were retrospectively included (110 [68.8%] childhood-onset, 119 [74.4%] male). BMD of the axial bone (including total hip, neck of femur, and L1-4) derived from dual X-ray absorptiometry and final height were investigated at the first visit, 12 months, 24 months, 36 months, and 48 months thereafter. Low BMD was defined as Z-score ≤ -2. RESULTS: The prevalence of low BMD was 30.0% at baseline and 12.5% at 4 years of follow-up. The CO AGHD group presented a significantly lower BMD than the AO AGHD group at the baseline (P = 0.009). In contrast, the CO AGHD group had significantly greater median annual BMD change than the AO AGHD group (0.044 vs. -0.0003 g/cm2/year in L1-4, P < 0.001), indicating a significant difference in the overall BMD trend between CO and AO groups. Childhood-onset (odds ratio [OR] 0.326, P = 0.012), low serum testosterone (OR 0.847; P = 0.004) and FT4 (OR 0.595; P = 0.039) level were independent risk factors for BMD loss. CONCLUSION: The annual changes of BMD show a different pattern in AGHD patients with varying ages of onset. Patients with CO AGHD have a lower bone mass, and in general, appropriate replacement therapy is necessary for long-term bone health in AGHD patients.


Subject(s)
Age of Onset , Bone Density , Human Growth Hormone , Humans , Male , Bone Density/physiology , Retrospective Studies , Female , Adult , Human Growth Hormone/deficiency , Human Growth Hormone/blood , Absorptiometry, Photon , Young Adult , Middle Aged , China/epidemiology , Dwarfism, Pituitary/blood , Dwarfism, Pituitary/epidemiology , Follow-Up Studies , Adolescent
5.
J Endocrinol Invest ; 47(7): 1743-1750, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38198073

ABSTRACT

PURPOSE: Growth hormone deficiency (GHD) is a rare condition with a worldwide prevalence of 1 patient in 4000 to 10,000 live births, placing a significant economic burden on healthcare systems. The aim of this study is to generate evidence on the economic burden of children and adolescents with GHD treated with rhGH and their parents in Italy. METHODS: A cost of illness analysis, adopting the prevalence approach, has been developed, producing evidence on the total annual cost sustained by the Italian National Health System (NHS) and by the society. The study is based on original data collected from a survey conducted among Italian children and adolescents with GHD and their parents. RESULTS: 143 children/adolescents with GHD and their parents participated to the survey, conducted from May to October 2021. Patients had a mean age of 12.2 years (SD: 3.1) and were mostly males (68.5%). The average direct healthcare cost sustained by the NHS was € 8,497.2 per patient/year; adding the out-of-pocket expenses (co-payments and expenses for private healthcare service), the total expense was € 8,568.6. The indirect costs, assessed with the human capital approach, were € 847.9 per patient/year. The total of direct and indirect cost is € 9,345.1 from the NHS perspective, and € 9,416.5 from a social perspective. The total cost incurred by the Italian NHS for children with GHD (range: 5,708-8,354) was estimated in € 48.5-71.0 million, corresponding to 0.04-0.06% of the total Italian public health expense in the year 2020. CONCLUSIONS: The total annual cost for GHD children is close to € 10,000, and is mainly due to the cost of rhGH treatment. This cost is almost entirely sustained by the NHS, with negligible out-of-pocket expenses. The economic burden on the Italian NHS for the health care of established GHD children is fourfold higher than the prevalence of the disease in the overall Italian population.


Subject(s)
Cost of Illness , Health Care Costs , Human Growth Hormone , Humans , Male , Italy/epidemiology , Child , Female , Adolescent , Human Growth Hormone/deficiency , Human Growth Hormone/economics , Human Growth Hormone/therapeutic use , Health Care Costs/statistics & numerical data , Health Expenditures/statistics & numerical data , Dwarfism, Pituitary/economics , Dwarfism, Pituitary/epidemiology , Dwarfism, Pituitary/drug therapy , Growth Disorders/economics , Growth Disorders/epidemiology , Prevalence , Parents
6.
Horm Res Paediatr ; 97(2): 126-133, 2024.
Article in English | MEDLINE | ID: mdl-37315542

ABSTRACT

INTRODUCTION: Genetic forms of growth hormone deficiency (GHD) may occur as isolated GHD (IGHD) or as a component of multiple pituitary hormone deficiency (MPHD). This study aimed to present the clinical and molecular characteristics of patients with IGHD/MPHD due to the GH1 gene variants. METHODS: A gene panel accommodating 25 genes associated with MPHD and short stature was used to search for small sequence variants. Multiplex ligation-dependent probe amplification was performed in patients with normal panel results to investigate gross deletion/duplications. Segregation in the family was performed by Sanger sequencing. RESULTS: The GH1 gene variants were detected in 5 patients from four unrelated families. One patient had IGHD IA due to homozygous whole GH1 gene deletion and one had IGHD IB due to novel homozygous c.162C>G/p.(Tyr54*) variant. Two patients from a family had previously reported heterozygous c.291+1G>A/p.(?) variant in which clinical and genetic characteristics were compatible with IGHD II accompanying MPHD. One patient had clinical and laboratory characteristics of IGHD II with MPHD but the heterozygous c.468 C>T/p.(R160W) variant had conflicting results about the relationship with the phenotype. CONCLUSION: Expanding our knowledge of the spectrum of GH1 gene variants by apprehending clinical and molecular data of more cases, helps to identify the genotype-phenotype correlation of IGHD/MPHD and the GH1 gene variants. These patients must be regularly followed up for the occurrence of additional pituitary hormone deficiencies.


Subject(s)
Dwarfism, Pituitary , Human Growth Hormone , Hypopituitarism , Humans , Dwarfism, Pituitary/genetics , Dwarfism, Pituitary/epidemiology , Human Growth Hormone/genetics , Hypopituitarism/genetics , Homozygote , Phenotype , Genetic Association Studies , Growth Hormone/genetics
7.
J Endocrinol Invest ; 46(12): 2513-2523, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37209402

ABSTRACT

PURPOSE: The aim of this study was to produce evidence on quality of life (QoL) among Italian growth hormone deficiency (GHD) children and adolescents treated with growth hormone (GH) and their parents. METHODS: A survey was conducted among Italian children and adolescents aged 4-18 with a confirmed diagnosis of GHD and treated with GH therapy and their parents. The European Quality of Life 5 Dimensions 3 Level Version (EQ-5D-3L) and the Quality of Life in Short Stature Youth (QoLISSY) questionnaires were administered between May and October 2021 through the Computer-Assisted Personal Interview (CAPI) method. Results were compared with national and international reference values. RESULTS: The survey included 142 GHD children/adolescents and their parents. The mean EQ-5D-3L score was 0.95 [standard deviation (SD) 0.09], while the mean visual analogue scale (VAS) score was 86.2 (SD 14.2); the scores are similar to those of a reference Italian population aged 18-24 of healthy subjects. As for the QoLISSY child-version, compared to the international reference values for GHD/ idiopathic short stature (ISS) patients, we found a significantly higher score for the physical domain, and lower scores for coping and treatment; compared to the specific reference values for GHD patients, our mean scores were significantly lower for all domains except the physical one. As for the parents, we found a significantly higher score for the physical domain, and a lower score for treatment; compared to reference values GHD-specific, we found lower score in the social, emotional, treatment, parental effects, and total score domains. CONCLUSIONS: Our results suggest that the generic health-related quality of life (HRQoL) in treated GHD patients is high, comparable to that of healthy people. The QoL elicited by a disease specific questionnaire is also good, and comparable with that of international reference values of GHD/ISS patients.


Subject(s)
Dwarfism, Pituitary , Human Growth Hormone , Humans , Adolescent , Quality of Life/psychology , Caregivers , Dwarfism, Pituitary/drug therapy , Dwarfism, Pituitary/epidemiology , Dwarfism, Pituitary/psychology , Italy/epidemiology , Human Growth Hormone/therapeutic use , Growth Hormone , Surveys and Questionnaires
8.
Front Endocrinol (Lausanne) ; 14: 1066182, 2023.
Article in English | MEDLINE | ID: mdl-36960394

ABSTRACT

Background: Isolated growth hormone deficiency (IGHD) is caused by a severe shortage or absence of growth hormone (GH), which results in aberrant growth and development. Patients with IGHD type IV (IGHD4) have a short stature, reduced serum GH levels, and delayed bone age. Objectives: To identify the causative mutation of IGHD in a consanguineous family comprising four affected patients with IGHD4 (MIM#618157) and explore its functional impact in silico. Methods: Clinical and radiological studies were performed to determine the phenotypic spectrum and hormonal profile of the disease, while whole-exome sequencing (WES) and Sanger sequencing were performed to identify the disease-causing mutation. In-silico studies involved protein structural modeling and docking, and molecular dynamic simulation analyses using computational tools. Finally, data from the Qatar Genome Program (QGP) were screened for the presence of the founder variant in the Qatari population. Results: All affected individuals presented with a short stature without gross skeletal anomalies and significantly reduced serum GH levels. Genetic mapping revealed a homozygous nonsense mutation [NM_000823:c.G214T:p.(Glu72*)] in the third exon of the growth-hormone-releasing hormone receptor gene GHRHR (MIM#139191) that was segregated in all patients. The substituted amber codon (UAG) seems to truncate the protein by deleting the C-terminus GPCR domain, thus markedly disturbing the GHRHR receptor and its interaction with the growth hormone-releasing hormone. Conclusion: These data support that a p.Glu72* founder mutation in GHRHR perturbs growth hormone signaling and causes IGHD type IV. In-silico and biochemical analyses support the pathogenic effect of this nonsense mutation, while our comprehensive phenotype and hormonal profiling has established the genotype-phenotype correlation. Based on the current study, early detection of GHRHR may help in better therapeutic intervention.


Subject(s)
Dwarfism, Pituitary , Human Growth Hormone , Humans , Dwarfism, Pituitary/genetics , Dwarfism, Pituitary/epidemiology , Codon, Nonsense , Pakistan , Human Growth Hormone/genetics , Growth Hormone/genetics , Mutation
9.
Front Endocrinol (Lausanne) ; 13: 1014743, 2022.
Article in English | MEDLINE | ID: mdl-36407317

ABSTRACT

Background: Children with growth hormone deficiency (GHD) are treated with daily somatropin injections; however, poor treatment persistence and adherence have been recognized previously and have been shown to negatively impact growth outcomes. A recent real-world study of a US pediatric GHD population found that a substantial proportion of children discontinued somatropin therapy, but similar data for a real-world UK population are lacking. Objectives: To describe the discontinuation of, and persistence with, daily somatropin treatment among children with GHD in the UK. Methods: This was a retrospective cohort study of children (≥3 and <16 years old) with ≥1 medication prescription for daily injectable somatropin from 1 July 2000 to 31 December 2020 in the IQVIA Medical Research DATA (IMRD) database. Early persistence was defined as the proportion of children prescribed ≥1 somatropin refill (≥2 prescriptions). Discontinuation was defined as the first date at which a medication gap for somatropin (of >60 or >90 days between prescriptions) occurred. Kaplan-Meier methods were used to evaluate persistence (non-discontinuation) over time to assess time to first discontinuation event. Cox proportional hazards models were used to evaluate the relationship between patient characteristics and time to medication discontinuation. Results: Among the cohort identified in this study (n = 117), the majority (n = 84, 71.8%) had 48 months of available follow-up; 56.4% were boys and the mean (median) age was 8.6 (8.0) years. About 98% exhibited early persistence, but persistence over the follow-up period decreased with follow-up duration. Using the conservative 90-day gap definition of persistence, an estimated 72.4%, 52.8%, and 43.3% were persistent at 12, 36, and 48 months. Lower persistence rates were observed using the 60-day definition. No significant patient predictors of time to discontinuation were identified. Conclusions: Despite high early persistence with somatropin, a high percentage of children with GHD were increasingly non-persistent over time. More than 1 in 4 were non-persistent at 12 months and more than 1 in 2 were non-persistent at 48 months of follow-up. These results suggest that strategies to support improved medication-taking behavior among children with GHD in the UK are warranted.


Subject(s)
Dwarfism, Pituitary , Human Growth Hormone , Humans , Adolescent , Male , Child , Female , Human Growth Hormone/therapeutic use , Growth Hormone , Retrospective Studies , Dwarfism, Pituitary/drug therapy , Dwarfism, Pituitary/epidemiology , United Kingdom/epidemiology
10.
J Pediatr Endocrinol Metab ; 35(8): 1059-1068, 2022 Aug 26.
Article in English | MEDLINE | ID: mdl-35822711

ABSTRACT

OBJECTIVES: Growth hormone deficiency (GHD) in adults is associated with an increased risk of cardiovascular morbidity and mortality. Although children with GHD are also believed to have a similar cardiovascular disease (CVD) risk beginning at an early age, the available data in children is scarce. We aimed to determine the various CVD risk parameters in children with isolated GHD (IGHD). METHODS: A cross-sectional case-control study was conducted at a tertiary care centre in North India comparing various auxological, biochemical, and echocardiographic parameters between 20 IGHD children aged 5-15 years and their age and sex-matched healthy controls. RESULTS: The mean age of children with IGHD and controls was similar (10.5 ± 2.6 yr vs. 9.9 ± 2.7 yr, p=0.48). Children with IGHD had significantly higher waist-hip-ratio (p=0.01), total cholesterol (p=0.02), non-high-density lipoprotein-cholesterol (p=0.02), serum homocysteine (p<0.001), C-reactive protein (CRP) (p=0.01) and pro-brain natriuretic peptide (pro-BNP) (p=0.04) levels as compared to healthy controls. Left ventricular mass (LVM) and interventricular septal thickness were significantly lower (p=0.04; p=0.02) in IGHD children. Correlation analysis showed that pro-BNP and CRP levels had negative correlation (p<0.001, r=-0.70; and p=0.04, r=-0.44, respectively) and LVM had a positive correlation (p=0.02, r=0.53) with height SDS among IGHD children. CONCLUSIONS: Children with IGHD showed abnormalities in several biochemical and cardiac parameters that may be associated with an increased CVD risk in later life. More extensive studies, including younger children with IGHD, are needed to determine the lower ages at which the CVD risk is detectable.


Subject(s)
Cardiovascular Diseases , Dwarfism, Pituitary , Human Growth Hormone , Adult , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Case-Control Studies , Child , Cholesterol , Cross-Sectional Studies , Dwarfism, Pituitary/complications , Dwarfism, Pituitary/epidemiology , Humans
11.
J Endocrinol Invest ; 45(9): 1709-1717, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35567736

ABSTRACT

PURPOSE: We aimed to evaluate the near-final height (nFHt) in a large cohort of pediatricpatients with growth hormone deficiency (GHD) and to elaborate a new predictive method of nFHt. METHODS: We recruited GHD patients diagnosed between 1987 and 2014 and followed-up until nFHt. To predict the values of nFHt, each predictor was run in a univariable spline. RESULTS: We enrolled 1051 patients. Pre-treatment height was -2.43 SDS, lower than parental height (THt) (-1.09 SDS, p < 0.001). The dose of recombinant human GH (rhGH) was 0.21mg/kg/week at start of treatment. nFHt was -1.08 SDS (height gain 1.27 SDS), higher than pre-treatment height (p < 0.001) and comparable to THt. 1.6% of the patients were shorter than -2 SDS from THt. The rhGH dose at nFHt was 0.19 mg/kg/week, lower than at the start (p < 0.001). The polynomial regression showed that nFHt was affected by gender, THt, age at puberty, height at puberty, age at the end of treatment (F = 325.37, p < 0.0001, R2 87.2%). CONCLUSION: This large national study shows that GHD children can reach their THt. The rhGH/kg/day dose significantly decreased from the start to the end of the treatment. Our model suggests the importance of a timely diagnosis, possibly before puberty, the beneficial effect of long-term treatment with rhGH, and the key-role of THt. Our prediction model has a very acceptable error compared to the majority of other published studies.


Subject(s)
Dwarfism, Pituitary , Human Growth Hormone , Body Height , Child , Cohort Studies , Dwarfism, Pituitary/diagnosis , Dwarfism, Pituitary/drug therapy , Dwarfism, Pituitary/epidemiology , Growth Hormone/therapeutic use , Humans , Puberty
12.
J Clin Endocrinol Metab ; 107(7): 1906-1919, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35368070

ABSTRACT

CONTEXT: Data on long-term safety of growth hormone (GH) replacement in adults with GH deficiency (GHD) are needed. OBJECTIVE: We aimed to evaluate the safety of GH in the full KIMS (Pfizer International Metabolic Database) cohort. METHODS: The worldwide, observational KIMS study included adults and adolescents with confirmed GHD. Patients were treated with GH (Genotropin [somatropin]; Pfizer, NY) and followed through routine clinical practice. Adverse events (AEs) and clinical characteristics (eg, lipid profile, glucose) were collected. RESULTS: A cohort of 15 809 GH-treated patients were analyzed (mean follow-up of 5.3 years). AEs were reported in 51.2% of patients (treatment-related in 18.8%). Crude AE rate was higher in patients who were older, had GHD due to pituitary/hypothalamic tumors, or adult-onset GHD. AE rate analysis adjusted for age, gender, etiology, and follow-up time showed no correlation with GH dose. A total of 606 deaths (3.8%) were reported (146 by neoplasms, 71 by cardiac/vascular disorders, 48 by cerebrovascular disorders). Overall, de novo cancer incidence was comparable to that in the general population (standard incidence ratio 0.92; 95% CI, 0.83-1.01). De novo cancer risk was significantly lower in patients with idiopathic/congenital GHD (0.64; 0.43-0.91), but similar in those with pituitary/hypothalamic tumors or other etiologies versus the general population. Neither adult-onset nor childhood-onset GHD was associated with increased de novo cancer risks. Neutral effects were observed in lipids/fasting blood glucose levels. CONCLUSION: These final KIMS cohort data support the safety of long-term GH replacement in adults with GHD as prescribed in routine clinical practice.


Subject(s)
Dwarfism, Pituitary , Human Growth Hormone , Hypopituitarism , Pituitary Diseases , Pituitary Neoplasms , Adolescent , Adult , Child , Dwarfism, Pituitary/complications , Dwarfism, Pituitary/drug therapy , Dwarfism, Pituitary/epidemiology , Growth Hormone/therapeutic use , Hormone Replacement Therapy/adverse effects , Human Growth Hormone/adverse effects , Humans , Hypopituitarism/drug therapy , Hypopituitarism/epidemiology , Pituitary Diseases/etiology , Pituitary Neoplasms/drug therapy
13.
Front Endocrinol (Lausanne) ; 12: 721812, 2021.
Article in English | MEDLINE | ID: mdl-34603204

ABSTRACT

Objective: Idiopathic short stature (ISS), an endocrine-related disease, is difficult to diagnose. Previous studies have shown that many children with some inflammation-related diseases often have short stature, but whether inflammation is the underlying mechanism of ISS has not been studied. Here, we attempt to explore the role of inflammation in the occurrence and development of ISS and to demonstrate an available clinical diagnostic model of ISS. Methods: Frozen serum samples were collected from ISS patients (n = 4) and control individuals (n = 4). Isobaric tags for relative and absolute quantitation (iTRAQ) combined with LC-MS/MS analysis were applied to quantitative proteomics analysis. To assess clusters of potentially interacting proteins, functional enrichment (GO and KEGG) and protein-protein interaction network analyses were performed, and the crucial proteins were detected by Molecular Complex Detection (MCODE). Furthermore, serum levels of two selected proteins were measured by ELISA between ISS patients (n = 80) and controls (n = 80). In addition, experiments in vitro were used to further explore the effects of crucial proteins on endochondral ossification. Results: A total of 437 proteins were quantified, and 84 DEPs (60 upregulated and 24 downregulated) were identified between patients with ISS and controls. Functional enrichment analysis showed that the DEPs were primarily enriched in blood microparticle, acute inflammatory response, protein activation cascade, collagen-containing extracellular matrix, platelet degranulation, etc. According to the results of top 10 fold change DEPs and MCODE analysis, C1QA and C1QB were selected to further experiment. The expression levels of C1QA and C1QB were validated in serum samples. Based on the logistic regression analysis and ROC curve analysis, we constructed a novel diagnostic model by serum levels of C1QA and C1QB with a specificity of 91.2% and a sensitivity of 75% (AUC = 0.900, p <0.001). Finally, the western blotting analysis confirmed the expression levels of OCN, OPN, RUNX2, and Collagen X were downregulated in chondrocytes, and the outcome of Collagen II was upregulated. Conclusion: Our study is the first to demonstrate the significant role of inflammation in the development of ISS. In addition, we identify C1QA and C1QB as novel serum biomarkers for the diagnosis of ISS.


Subject(s)
Dwarfism, Pituitary/diagnosis , Models, Theoretical , Adolescent , Biomarkers/blood , Blood Chemical Analysis/methods , Blood Proteins/analysis , Blood Proteins/chemistry , Blood Proteins/metabolism , Case-Control Studies , Child , Child, Preschool , Chondrocytes/metabolism , Chromatography, Liquid/methods , Complement C1q/analysis , Complement C1q/metabolism , Decision Support Techniques , Dwarfism, Pituitary/blood , Dwarfism, Pituitary/epidemiology , Female , Humans , Male , Osteogenesis , Prognosis , Proteome/analysis , Proteome/metabolism , Proteomics/methods , Tandem Mass Spectrometry/methods
14.
Hormones (Athens) ; 20(4): 679-689, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34195937

ABSTRACT

Anxiety and depression are among the commonest emotional problems in children and young adolescents. They are encountered with even higher prevalence in children and adults with growth hormone deficiency (GHD). Alterations in the somatotropic axis, as observed in both GH/IGF1 deficiency and excess, can produce permanent changes in brain tissue structure. The growth hormone/insulin-like growth factor 1 (GH/IGF1) axis seems to exert a regulatory effect on brain function and neurogenesis, especially in the hippocampus, a brain region associated with mental and emotional disorders, such as depression and anxiety. There is evidence from animal models of the possible interrelationship of the endocrine system with the pathogenesis of emotional disorders. Moreover, clinical data support the association of GHD and mood disorders, which are often reversed by GH replacement therapy. However, the causal relationship and the mechanism underlying this association are to date obscure and remain to be clarified. The present review reports experimental data from animal models regarding the role of GH/IGF1 in emotional disorders and focuses on clinical data on the presence of these disorders in children with GHD and their response to GH therapy.


Subject(s)
Anxiety , Depression , Dwarfism, Pituitary , Human Growth Hormone , Adolescent , Animals , Anxiety/epidemiology , Child , Depression/epidemiology , Dwarfism, Pituitary/epidemiology , Human Growth Hormone/deficiency , Humans , Insulin-Like Growth Factor I
15.
J Clin Endocrinol Metab ; 106(6): 1728-1741, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33571362

ABSTRACT

CONTEXT: Growth hormone (GH) treatment has a generally good safety profile; however, concerns about increased mortality risk in adulthood have been raised. OBJECTIVE: This work aims to assess the long-term safety of GH treatment in clinical practice. METHODS: Data were collected from 676 clinics participating in 2 multicenter longitudinal observational studies: the NordiNet International Outcome Study (2006-2016, Europe) and ANSWER Program (2002-2016, USA). Pediatric patients treated with GH were classified into 3 risk groups based on diagnosis. Intervention consisted of daily GH treatment, and main outcome measures included incidence rates (events/1000 patient-years) of adverse drug reactions (ADRs), serious adverse events (SAEs), and serious ADRs, and their relationship to GH dose. RESULTS: The combined studies comprised 37 702 patients (68.4% in low-risk, 27.5% in intermediate-risk, and 4.1% in high-risk groups) and 130 476 patient-years of exposure. The low-risk group included children born small for gestational age (SGA; 20.7%) and non-SGA children (eg, with GH deficiency; 79.3%). Average GH dose up to the first adverse event (AE) decreased with increasing risk category. Patients without AEs received higher average GH doses than patients with more than one AE across all groups. A significant inverse relationship with GH dose was shown for ADR and SAE incidence rates in the low-risk group (P = .003 and P = .001, respectively) and the non-SGA subgroup (both P = .002), and for SAEs in the intermediate- and high-risk groups (P = .002 and P = .05, respectively). CONCLUSIONS: We observed no indication of increased mortality risk nor AE incidence related to GH dose in any risk group. A short visual summary of our work is available (1).


Subject(s)
Drug-Related Side Effects and Adverse Reactions/epidemiology , Growth Disorders/drug therapy , Human Growth Hormone/adverse effects , Adolescent , Child , Child, Preschool , Dwarfism, Pituitary/drug therapy , Dwarfism, Pituitary/epidemiology , Europe/epidemiology , Female , Growth Disorders/epidemiology , Human Growth Hormone/deficiency , Human Growth Hormone/therapeutic use , Humans , Incidence , Longitudinal Studies , Male , Mortality , Outcome Assessment, Health Care , Registries , United States/epidemiology
16.
Am J Med Genet A ; 182(9): 2077-2084, 2020 09.
Article in English | MEDLINE | ID: mdl-32656927

ABSTRACT

Potocki-Lupski Syndrome (PTLS, MIM 610883), or duplication of chromosome 17p11.2, is a clinically recognizable condition characterized by infantile hypotonia, failure to thrive, developmental delay, intellectual disability, and congenital anomalies. Short stature, classified as greater than two standard deviations below the mean, has not previously been considered a major feature of PTLS. Retrospective chart review on a cohort of 37 individuals with PTLS was performed to investigate the etiology of short stature. Relevant data included anthropometric measurements, insulin growth factor-1 (IGF-1), insulin-like growth factor binding protein 3 (IGFBP-3), growth hormone (GH) stimulation testing, blood glucose levels, brain MRI, and bone age. Approximately 25% (9/37) of individuals with PTLS had short stature. Growth hormone deficiency (GHD) was definitively identified in two individuals. These two PTLS patients with growth hormone deficiency, as well as three others with short stature and no documented GHD, received growth hormone and obtained improvement in linear growth. One individual was identified to have pituitary abnormalities on MRI and had complications of hypoglycemia due to unrecognized GHD. Individuals with PTLS can benefit from undergoing evaluation for GHD should they present with short stature or hypoglycemia. Early identification of GHD could facilitate potential therapeutic benefit for individuals with PTLS, including linear growth, musculoskeletal, and in cases of hypoglycemia, potentially cognitive development as well.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Disorders/genetics , Chromosome Duplication/genetics , Dwarfism, Pituitary/genetics , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/genetics , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/epidemiology , Abnormalities, Multiple/pathology , Adolescent , Adult , Blood Glucose/genetics , Brain/diagnostic imaging , Brain/pathology , Child , Child, Preschool , Chromosome Disorders/diagnostic imaging , Chromosome Disorders/epidemiology , Chromosome Disorders/pathology , Comparative Genomic Hybridization , Dwarfism, Pituitary/diagnostic imaging , Dwarfism, Pituitary/epidemiology , Dwarfism, Pituitary/pathology , Failure to Thrive/epidemiology , Failure to Thrive/genetics , Failure to Thrive/pathology , Female , Human Growth Hormone/deficiency , Human Growth Hormone/genetics , Humans , Infant , Magnetic Resonance Imaging , Male , Phenotype , Smith-Magenis Syndrome/diagnostic imaging , Smith-Magenis Syndrome/epidemiology , Smith-Magenis Syndrome/genetics , Smith-Magenis Syndrome/pathology , Young Adult
17.
J Endocrinol Invest ; 43(10): 1447-1452, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32239476

ABSTRACT

BACKGROUND: Assessing adherence to growth hormone (GH) is challenging. The Easypod™ connect device delivers pre-set doses of recombinant human GH (r-hGH) and stores a digital record of adherence that can be shared with healthcare provider. We assessed adherence to r-hGH delivered with Easypod™ according to the approved pediatric indications for r-hGH: growth hormone deficiency (GHD), born small for gestational age (SGA) who failed to show catch-up growth and Turner syndrome (TS). METHODS: ECOS (NCT01555528) was a multicenter (24 countries), 5-year, longitudinal, observational study, which aimed to evaluate country-specific adherence to r-hGH therapy prescribed via the Easypod™ electronic injection device. The primary endpoint was yearly adherence. Secondary endpoints were height velocity, height velocity standard deviation scores (SDS), height, height SDS and IGF-1 concentrations. Clinical and auxological data were obtained from medical records and adherence from Easypod™ logs. RESULTS: This study included 147 Easypod™-naïve Mexican children assessed during 3 years (mean age: 9.96 ± 3.41 years, 56.8% boys, mean height SDS at baseline: - 2.17 ± 0.97): 118 with GHD, 24 SGA and 5 with TS. A total of 105 (71.4%) patients were GH naïve. Overall median adherence was > 90% over the first year of treatment and > 80% at 3 years. Adherence was not different by r-hGH indication or between GH-naïve or experienced patients. At 1-year follow-up, mean change in height SDS was 0.57 ± 0.34, whereas mean height velocity SDS was 2.85 ± 2.51. In all, 84.7% patients had normal IGF-1 concentrations at 1-year follow-up. Adherence was associated with change in height SDS (r = 0.239, p = 0.005) and height velocity SDS (r = 0.194, p = 0.027). CONCLUSION: Adherence rates with the Easypod™ device are high and maintained over time in GHD, SGA and TS Easypod™-naïve Mexican patients. High adherence is associated with better outcomes. Easypod™ assists physicians in monitoring adherence to r-hGH.


Subject(s)
Growth Disorders/drug therapy , Human Growth Hormone/therapeutic use , Medication Adherence/statistics & numerical data , Adolescent , Child , Dwarfism, Pituitary/drug therapy , Dwarfism, Pituitary/epidemiology , Female , Growth Disorders/epidemiology , Hormone Replacement Therapy/methods , Hormone Replacement Therapy/statistics & numerical data , Humans , Infant, Newborn , Infant, Small for Gestational Age/growth & development , Longitudinal Studies , Male , Mexico/epidemiology , Recombinant Proteins/therapeutic use , Turner Syndrome/drug therapy , Turner Syndrome/epidemiology
18.
J Clin Res Pediatr Endocrinol ; 12(3): 295-302, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32157854

ABSTRACT

Objective: The aim was to determine the final adult height (FAH) achieved by recombinant human growth hormone (rhGH) treatment, the factors affecting FAH and the success of attaining the genetic potential. Methods: Data of 133 patients treated with rhGH therapy were reviewed retrospectively. Patients were grouped according to diagnosis, either isolated GH deficiency (IGHD) or multiple pituitary hormone deficiency (MPHD), and by sex, and pubertal status at the beginning of treatment. Results: The mean age of initiation of treatment was 12.3±2.18 years, and the mean duration of rhGH treatment was 3.65±1.5 years. The mean height standard deviation score (SDS) at diagnosis was -3.11±0.75 SD. All patients received a standardized GH dose of 0.033 mg/kg/day. Mean FAH-SDS was -1.8±0.77 and delta height-SDS (the change in height SDS between the beginning and end of treatment) was 1.28±0.94 SD. FAH-SDS was -1.79±0.86 SD in males; -1.82±0.64 in females (p=0.857); -1.94±0.71 at the beginning of treatment in pubertal patients and -1.68±0.81 in prepubertal patients (p=0.056); -1.84±0.89 in patients with IGHD and -0.47±0.2 in patients with MPHD (p˃0.05). In multiple regression analysis, First year delta height-SDS was the most predictive factor for both FAH-SDS and delta height-SDS. Conclusion: The majority of our patients achieved a final height compatible with their genetic potential as well as population standards when treated with rhGH even having started at a relatively late age. First year delta height-SDS was a predictive factor for FAH.


Subject(s)
Body Height , Growth Disorders/drug therapy , Growth Disorders/epidemiology , Growth Hormone/deficiency , Human Growth Hormone/therapeutic use , Adolescent , Adult , Body Height/drug effects , Child , Dwarfism, Pituitary/drug therapy , Dwarfism, Pituitary/epidemiology , Female , Growth Disorders/complications , Growth Hormone/therapeutic use , Hormone Replacement Therapy , Humans , Hypopituitarism/complications , Hypopituitarism/drug therapy , Hypopituitarism/epidemiology , Male , Puberty/drug effects , Puberty/physiology , Retrospective Studies , Turkey/epidemiology
20.
J Clin Res Pediatr Endocrinol ; 12(3): 261-268, 2020 09 02.
Article in English | MEDLINE | ID: mdl-31948187

ABSTRACT

Objective: Mutations of the genes encoding transcription factors which play important roles in pituitary morphogenesis, differentiation and maturation may lead to combined pituitary hormone deficiency (CPHD). PROP1 gene mutations are reported as the most frequent genetic aetiology of CHPD. The aim of this study was to describe the phenotypes of Turkish CPHD patients and define the frequency of PROP1 mutations. Methods: Fifty-seven CPHD patients from 50 families were screened for PROP1 mutations. The patients were affected by growth hormone (GH) and additional anterior pituitary hormone deficiencies. Results: All patients had GH deficiency. In addition, 98.2% had central hypothyroidism, 45.6% had hypogonadotropic hypogonadism, 43.8% had adrenocorticotropic hormone deficiency and 7.1% had prolactin deficiency. Parental consanguinity rate was 50.9% and 14 cases were familial. Mean height standard deviation score (SDS) and weight SDS were -3.8±1.4 and -3.1±2.0, respectively. Of 53 patients with available pituitary imaging, 32 (60.4%) showed abnormalities. None had extra-pituitary abnormalities. Eight index patients had PROP1 gene mutations. Five sporadic patients were homozygous for c.301_302delAG (p.Leu102CysfsTer8) mutation, two siblings had exon 2 deletion, two siblings had complete gene deletion and two siblings were homozygous for the novel c.353A>G (p.Q118R) mutation. The frequency of the PROP1 mutations was 16% in our cohort. Mutation rate was significantly higher in familial cases compared to sporadic cases (42.8% vs 11.6%; p<0.01). Conclusion: Phenotype of patients regarding hormonal deficiencies, pituitary morphology, presence of extra-pituitary findings, family history of CPHD and parental consanguinity are important for deciding which pituitary transcription factor deficiency should be investigated. PROP1 mutation frequencies vary in different populations and its prevalence is high in Turkish CPHD patients.


Subject(s)
Homeodomain Proteins/genetics , Hypopituitarism/genetics , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Consanguinity , DNA Mutational Analysis , Dwarfism, Pituitary/drug therapy , Dwarfism, Pituitary/epidemiology , Dwarfism, Pituitary/genetics , Family , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Hormone Replacement Therapy , Human Growth Hormone/therapeutic use , Humans , Hypopituitarism/drug therapy , Hypopituitarism/epidemiology , Infant , Male , Mutation , Retrospective Studies , Turkey/epidemiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL