Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 259
Filtrar
1.
Physiol Rep ; 12(17): e70027, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39227324

RESUMEN

The Snell dwarf mouse (Pit1dw/dw), an animal model of congenital combined pituitary hormone deficiency, displays skeletal muscle weakness. While enhanced responsivity to repeated exposures of muscle contractions have been documented for Snell dwarf mice, the response following single exposure to distinct contraction protocols remained uncharacterized. The purpose of this study was to investigate the muscle recovery of Snell dwarf and control littermate mice following a single exposure to two separate protocols-an intermittent slow velocity (30°/s) contraction protocol or a continuous rapid velocity (500°/s) contraction protocol. Following both protocols for control mice, torque values were 30% and 80% of pre-protocol values at 5 min and 3 days, respectively. At 10 days, performance returned to baseline for the 30°/s protocol and were depressed for the 500°/s protocol. For Snell dwarf mice following both protocols, torques were depressed to 5% of pre-protocol values at 5 min and returned to baseline by 3 days. Recovery following the 30°/s protocol for control mice and both protocols for Snell dwarf mice coincided with increased transcriptional output, upregulation of cytokine-mediated signaling genes, and a distribution shift to smaller muscle fibers with reduced area per nucleus. These features represent efficacious remodeling ubiquitous across distinct contraction paradigms in the context of the Pit1 mutation.


Asunto(s)
Contracción Muscular , Músculo Esquelético , Animales , Ratones , Músculo Esquelético/fisiología , Músculo Esquelético/metabolismo , Enanismo Hipofisario/genética , Enanismo Hipofisario/fisiopatología , Enanismo Hipofisario/metabolismo , Masculino , Femenino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
2.
Pediatr Res ; 96(1): 115-123, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38582946

RESUMEN

BACKGROUND: Growth hormone deficiency(GHD) and idiopathic short stature(ISS) are the primary causes of short stature in children. Animal experiments have revealed a link between growth hormone(GH), gut microbiota and metabolism, however, limited information is available from human trials. METHODS: Fecal samples collected from GHD (n = 36), ISS (n = 32) and healthy control (HC) children(n = 16) were subjected to microbiome (16 S rRNA gene sequencing) and metabolome (nuclear magnetic resonance,NMR) analyses. RESULTS: GHD, ISS and HC exhibit distinct differences in beta diversity of gut microbiota.In addition, short stature (GHD and ISS) exhibit higher relative abundance of Prevotellaceae_NK3B31_group at genus level compared to HC, whereas Rodentibacter, Rothia, and Pelomonas showed lower abundance. Additionally,Fusobacterium_mortiferum was identified as the characteristic species of GHD. Moreover, glucose metabolism, pyruvate metabolism and pyrimidine metabolism might play significant roles for distinguishing between GHD and normal GH groups (ISS and HC). Furthermore, a disease prediction model based on differential bacteria and metabolites between GHD and ISS exhibited high diagnostic value. CONCLUSION: These findings highlight the characteristics of different GH levels on the gut microbiota and metabolism in children, providing novel perspectives for early diagnosis and prognostic treatment of short stature with abnormal GH levels. IMPACT: The key message of our study is to identify human-relevant gut microbiota and host metabolic patterns that are interfered with growth hormone levels, and to develop biomarker models to identify short stature associated with growth hormone deficiency. We used idiopathic short stature as a control group for growth hormone deficiency, complementing the absence of height as a factor in the existing literature. Our study ultimately hopes to shed new light on the diagnosis and treatment of short stature children associated with growth hormone deficiency.


Asunto(s)
Heces , Microbioma Gastrointestinal , Trastornos del Crecimiento , Hormona de Crecimiento Humana , Humanos , Niño , Masculino , Femenino , Heces/microbiología , Hormona de Crecimiento Humana/metabolismo , Trastornos del Crecimiento/microbiología , Estudios de Casos y Controles , Metaboloma , Estatura , ARN Ribosómico 16S/genética , Enanismo Hipofisario/metabolismo , Enanismo Hipofisario/tratamiento farmacológico , Hormona del Crecimiento/metabolismo , Hormona del Crecimiento/sangre , Bacterias/metabolismo
3.
Nat Rev Endocrinol ; 18(9): 558-573, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35750929

RESUMEN

Since its discovery nearly a century ago, over 100,000 studies of growth hormone (GH) have investigated its structure, how it interacts with the GH receptor and its multiple actions. These include effects on growth, substrate metabolism, body composition, bone mineral density, the cardiovascular system and brain function, among many others. Recombinant human GH is approved for use to promote growth in children with GH deficiency (GHD), along with several additional clinical indications. Studies of humans and animals with altered levels of GH, from complete or partial GHD to GH excess, have revealed several covert or hidden actions of GH, such as effects on fibrosis, cardiovascular function and cancer. In this Review, we do not concentrate on the classic and controversial indications for GH therapy, nor do we cover all covert actions of GH. Instead, we stress the importance of the relationship between GH and fibrosis, and how fibrosis (or lack thereof) might be an emerging factor in both cardiovascular and cancer pathologies. We highlight clinical data from patients with acromegaly or GHD, alongside data from cellular and animal studies, to reveal novel phenotypes and molecular pathways responsible for these actions of GH in fibrosis, cardiovascular function and cancer.


Asunto(s)
Enfermedades Cardiovasculares , Fibrosis/metabolismo , Hormona de Crecimiento Humana/metabolismo , Neoplasias , Animales , Enfermedades Cardiovasculares/metabolismo , Niño , Enanismo Hipofisario/metabolismo , Hormona del Crecimiento , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Neoplasias/metabolismo
4.
Eur J Endocrinol ; 187(1): 27-38, 2022 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-35521713

RESUMEN

Objective: Growth hormone (GH) replacement therapy in patients with adult growth hormone deficiency (AGHD) is individually titrated due to variable dose-responses among patients. The aim of this study was to provide clinical guidance on dosing and titration of the novel long-acting GH derivative somapacitan based on analyses of somapacitan dose-insulin-like growth factor I (IGF-I) responses in AGHD patients. Design: Analyses of dosing information, 4364 somapacitan concentration samples and 4880 IGF-I samples from 330 AGHD patients treated with somapacitan in three phase 3 trials. Methods: Pharmacokinetic/pharmacodynamic modelling was used to evaluate starting dose groups by age and oral oestrogen therapy, characterise the dose-IGF-I response in the overall AGHD population and patient subgroups, predict the IGF-I response to dose changes and simulate missed dosing. Results: The analyses supported the clinical recommendations of higher starting doses for younger patients and women on oral oestrogen replacement therapy. For patients switching from daily GH treatment, the mean maintenance dose ratio between somapacitan (mg/week) and somatropin (mg/day) was predicted to be 8.2 (observed interquartile range of 6.7-9.1). Simulations of IGF-I SDS profiles confirmed the appropriate time for IGF-I sampling to be 3-4 days after somapacitan dosing and supported somapacitan administration with up to 3 days delay in case of missed dosing. Subgroup analyses characterised the dose-exposure-IGF-I response in patient subgroups and indicated that dose requirements are mainly influenced by sex and oral oestrogen treatment. Conclusions: This study extends the knowledge of the somapacitan dose-IGF-I response and provides information on clinical dosing of once-weekly somapacitan in patients with AGHD.


Asunto(s)
Enanismo Hipofisario , Hormona de Crecimiento Humana , Hormonas Adenohipofisarias , Adulto , Enanismo Hipofisario/tratamiento farmacológico , Enanismo Hipofisario/metabolismo , Estrógenos/uso terapéutico , Femenino , Histidina , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Manitol , Fenol
5.
J Vet Med Sci ; 84(5): 680-688, 2022 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-35387959

RESUMEN

Spontaneous dwarf rat (SDR) is a primary experimental animal model for the study of pituitary dwarfism with a point mutation in the Gh gene encoding growth hormone (GH). In previous studies, SDR has been reported to be associated with the GH deficiency as well as combined hormone deficiencies, the cause of which is unknown. In this study, we focused on the characteristics of pituitary stem/progenitor cell populations, which are a source of hormone-producing cells, in SDR. Immunofluorescence and quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) analyses confirmed the defects in GH-producing cells, the decreased number of prolactin- and thyroid-stimulating hormone-producing cells, and the increased number of adrenocorticotropic hormone- and luteinizing hormone-producing cells. Additionally, qRT-PCR analysis showed increased Prop1 (an embryonic stem/progenitor cell marker) expression and decreased S100b (a putative adult stem/progenitor cell marker) expression in SDRs. In the pituitary stem/progenitor cell niche, the marginal cell layer, the proportion of SOX2/PROP1-double positive cells was higher in adult SDRs than in adult Sprague Dawley (SD) rats but that of SOX2/S100ß-double positive cells was much lower. Furthermore, the number of SOX2/PROP1-double positive cells in SD rats significantly decreased with growth; however, the decrease was smaller in SDRs. In contrast, the number of SOX2/S100ß-double positive cells in SD rats significantly increased with growth; however, they were few in SDRs. Thus, S100ß-positive pituitary stem/progenitor cells failed to settle in pituitary dwarfism with the Gh gene mutation, leading to multiple hypopituitarism including GH deficiency.


Asunto(s)
Enanismo Hipofisario , Enfermedades de los Roedores , Animales , Enanismo Hipofisario/metabolismo , Enanismo Hipofisario/veterinaria , Hormona del Crecimiento/genética , Hormona del Crecimiento/metabolismo , Hipófisis/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre
6.
J Clin Endocrinol Metab ; 106(11): 3184-3195, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34272849

RESUMEN

CONTEXT: For children with growth hormone deficiency (GHD), treatment burden with daily somatropin injections [human growth hormone (hGH)] is high, which may lead to poor adherence and suboptimal overall treatment outcomes. Lonapegsomatropin (TransCon hGH) is an investigational long-acting, once-weekly prodrug for the treatment of GHD. OBJECTIVE: The objective of this study was to evaluate the efficacy and safety of once-weekly lonapegsomatropin vs daily somatropin. DESIGN: The heiGHt trial was a randomized, open-label, active-controlled, 52-week Phase 3 trial (NCT02781727). SETTING: This trial took place at 73 sites across 15 countries. PATIENTS: This trial enrolled and dosed 161 treatment-naïve, prepubertal patients with GHD. INTERVENTIONS: Patients were randomized 2:1 to receive lonapegsomatropin 0.24 mg hGH/kg/week or an equivalent weekly dose of somatropin delivered daily. MAIN OUTCOME MEASURE: The primary end point was annualized height velocity (AHV) at week 52. Secondary efficacy end points included change from baseline in height SD scores (SDS). RESULTS: Least squares (LS) mean (SE) AHV at 52 weeks was 11.2 (0.2) cm/year for lonapegsomatropin vs 10.3 (0.3) cm/year for daily somatropin (P = 0.009), with lonapegsomatropin demonstrating both noninferiority and superiority over daily somatropin. LS mean (SE) height SDS increased from baseline to week 52 by 1.10 (0.04) vs 0.96 (0.05) in the weekly lonapegsomatropin vs daily somatropin groups (P = 0.01). Bone age/chronological age ratio, adverse events, tolerability, and immunogenicity were similar between groups. CONCLUSIONS: The trial met its primary objective of noninferiority in AHV and further showed superiority of lonapegsomatropin compared to daily somatropin, with similar safety, in treatment-naïve children with GHD.


Asunto(s)
Enanismo Hipofisario/tratamiento farmacológico , Hormona de Crecimiento Humana/administración & dosificación , Hormona de Crecimiento Humana/deficiencia , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Niño , Enanismo Hipofisario/metabolismo , Enanismo Hipofisario/patología , Femenino , Estudios de Seguimiento , Terapia de Reemplazo de Hormonas , Hormona de Crecimiento Humana/química , Humanos , Masculino , Pronóstico
7.
Horm Res Paediatr ; 94(3-4): 81-104, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34091447

RESUMEN

The current differential diagnosis for a short child with low insulin-like growth factor I (IGF-I) and a normal growth hormone (GH) peak in a GH stimulation test (GHST), after exclusion of acquired causes, includes the following disorders: (1) a decreased spontaneous GH secretion in contrast to a normal stimulated GH peak ("GH neurosecretory dysfunction," GHND) and (2) genetic conditions with a normal GH sensitivity (e.g., pathogenic variants of GH1 or GHSR) and (3) GH insensitivity (GHI). We present a critical appraisal of the concept of GHND and the role of 12- or 24-h GH profiles in the selection of children for GH treatment. The mean 24-h GH concentration in healthy children overlaps with that in those with GH deficiency, indicating that the previously proposed cutoff limit (3.0-3.2 µg/L) is too high. The main advantage of performing a GH profile is that it prevents about 20% of false-positive test results of the GHST, while it also detects a low spontaneous GH secretion in children who would be considered GH sufficient based on a stimulation test. However, due to a considerable burden for patients and the health budget, GH profiles are only used in few centres. Regarding genetic causes, there is good evidence of the existence of Kowarski syndrome (due to GH1 variants) but less on the role of GHSR variants. Several genetic causes of (partial) GHI are known (GHR, STAT5B, STAT3, IGF1, IGFALS defects, and Noonan and 3M syndromes), some responding positively to GH therapy. In the final section, we speculate on hypothetical causes.


Asunto(s)
Enanismo Hipofisario , Enanismo , Hormona de Crecimiento Humana/metabolismo , Factor I del Crecimiento Similar a la Insulina/deficiencia , Hipotonía Muscular , Síndrome de Noonan , Columna Vertebral/anomalías , Niño , Preescolar , Diagnóstico Diferencial , Enanismo/diagnóstico , Enanismo/genética , Enanismo/metabolismo , Enanismo Hipofisario/diagnóstico , Enanismo Hipofisario/genética , Enanismo Hipofisario/metabolismo , Hormona de Crecimiento Humana/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Hipotonía Muscular/diagnóstico , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Síndrome de Noonan/diagnóstico , Síndrome de Noonan/genética , Síndrome de Noonan/metabolismo , Columna Vertebral/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-32621723

RESUMEN

Several different proteins regulate, directly or indirectly, the production of growth hormones from the pituitary gland, thereby complex genetics is involved. Defects in these genes are related to the deficiency of growth hormones solely, or deficiency of other hormones, secreted from the pituitary gland including growth hormones. These studies can aid clinicians to trace the pattern of the disease between the families, start early treatment and predict possible future consequences. This paper highlights some of the most common and novel genetic anomalies concerning growth hormones, which are responsible for various genetic defects in isolated growth and combined pituitary hormone deficiency disease.


Asunto(s)
Hormona de Crecimiento Humana/deficiencia , Hormona de Crecimiento Humana/genética , Pediatría/métodos , Enfermedades de la Hipófisis/genética , Enfermedades de la Hipófisis/metabolismo , Enanismo Hipofisario/diagnóstico , Enanismo Hipofisario/genética , Enanismo Hipofisario/metabolismo , Humanos , Hipopituitarismo/diagnóstico , Hipopituitarismo/genética , Hipopituitarismo/metabolismo , Enfermedades de la Hipófisis/diagnóstico
9.
Aging (Albany NY) ; 13(2): 1633-1648, 2020 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-33378746

RESUMEN

Histone modifications, specifically in the lysine residues of histone H3, have been implicated in lifespan regulation in several model organisms. Our previous studies showed that growth hormone (GH) treatment during early life can dramatically influence lifespan in long-lived Ames dwarf mice. However, the effects of this hormonal intervention on epigenetic modifications have never been examined. In this study, we sought to compare tissue-specific histone H3 lysine methylation and acetylation markers in Ames dwarf and wild type (WT) mice and to determine how these markers are affected by early-life GH intervention. Ames dwarf mice exhibited suppressed H3K4me in both hepatic and brain tissues, while showing elevated H3K27me in the brain. Early-life GH intervention significantly altered the histone H3 markers in those tissues. Furthermore, early GH intervention increased expression of histone H3 acetylation at multiple lysine residues in a tissue-specific manner. This included changes in H3K14ac and H3K18ac in the liver and brain, H3K18ac in visceral adipose tissue and H3K9ac, H3K14ac and H3K27ac in subcutaneous adipose tissue. This study serves as an initial, but important step in elucidating the epigenetic mechanisms by which hormonal signals during early life can influence aging and longevity in mammals.


Asunto(s)
Encéfalo/efectos de los fármacos , Enanismo Hipofisario/metabolismo , Epigénesis Genética/efectos de los fármacos , Hormona del Crecimiento/farmacología , Histonas/efectos de los fármacos , Hígado/efectos de los fármacos , Acetilación/efectos de los fármacos , Animales , Encéfalo/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasas/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Modelos Animales de Enfermedad , Enanismo Hipofisario/genética , Proteína Potenciadora del Homólogo Zeste 2 , Hormona del Crecimiento/deficiencia , Código de Histonas/efectos de los fármacos , Histonas/metabolismo , Proteínas de Homeodominio/genética , Terapia de Reemplazo de Hormonas , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/metabolismo , Hígado/metabolismo , Longevidad/genética , Metilación/efectos de los fármacos , Ratones , Grasa Subcutánea/efectos de los fármacos , Grasa Subcutánea/metabolismo , ADN Metiltransferasa 3B
10.
Growth Horm IGF Res ; 52: 101318, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32252003

RESUMEN

Both rhGH and rhIGF-I are signaling molecules with the capacity to restore the rate of growth in certain subsets of slowly growing children. In some instances, heights attained at or near the time of cessation of linear growth are indistinguishable from the height distribution of the community as a whole or from the height distribution expected based on the heights of biological parents. The GH: IGF-I signaling system is sequential, forming a continuous loop wherein GH will stimulate production of IGF-I and IGF-I will inhibit production of GH. This feature suggests that a deficiency of GH will be accompanied by a deficiency of IGF-I and that treatment of GH deficiency with rhGH will restore IGF-I and the subnormal growth of combined GH: IGF-I deficiency. Although logical, this proposition is not always true. rhGH and rhIGF-I are distinct polypeptides, with distinct cell surface receptors and distinct intracellular signaling pathways both capable of amplifying distinct, yet overlapping, patterns of gene replication, protein synthesis and metabolic activities. These features suggest that neither treatment with rhGH nor rhIGF-I alone will invariably recapitulate the combined activities of the GH: IGF-I system, At the present time, this proposition appears both logical and true. The possibility that combined rhGH and rhIGF-I treatment can accomplish that which neither monotherapy can has been examined in gene knock-out experiments in animals and direct comparisons of GH, IGF-I and combined GH: IGF- treatments in animals and in children with short stature, normal GH and low IGF-I (primary IGF-I deficiency). In these experimental models, the growth rates with combined rhGH and rhIGF-I treatment exceed those of either monotherapy. The extent to which this proposition can be generalized to various short stature populations remains to be determined.


Asunto(s)
Enanismo Hipofisario/tratamiento farmacológico , Hormona de Crecimiento Humana/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Animales , Niño , Quimioterapia Combinada , Enanismo Hipofisario/metabolismo , Humanos
11.
Artículo en Inglés | MEDLINE | ID: mdl-32328036

RESUMEN

Objective: To explore the most suitable calculation method for insulin dosage in an insulin tolerance test (ITT) and to evaluate the clinical application value of the optimization coefficient (γ). Methods: In this study, 140 adult patients with congenital growth hormone deficiency (GHD) or acquired hypopituitarism were randomized into the following two groups: the conventional group (n = 70) and the optimized group (n = 70). Oral glucose tolerance tests (OGTTs), insulin release tests (IRTs), and ITTs were conducted. For ITTs, insulin doses were the product of body weight (kg) and related coefficient (0.15 IU/kg for the control group and γ IU/kg for the optimized group, respectively). Notably, γ was defined as -0.034 + 0.000176 × AUCINS + 0.009846 × BMI, which was based on our previous study. Results: In the ITTs, the rate of achieving adequate hypoglycemia with a single insulin dose was significantly higher for the optimized group compared with the conventional group (92.9 vs. 60.0%, P < 0.001). The optimized group required higher initial doses of insulin (0.23 IU/kg). Meanwhile, the two groups did not differ significantly in their nadir blood glucose (1.9 vs. 1.9 mmol/L, P = 0.828). Conclusion: This study confirmed that the proposed optimized calculation method for insulin dosage in ITTs led to more efficient hypoglycemia achievement, without increasing the incidence of serious adverse events.


Asunto(s)
Glucemia/efectos de los fármacos , Técnicas de Diagnóstico Endocrino/normas , Cálculo de Dosificación de Drogas , Resistencia a la Insulina , Insulina/administración & dosificación , Adulto , Glucemia/metabolismo , Calibración , Enanismo Hipofisario/sangre , Enanismo Hipofisario/diagnóstico , Enanismo Hipofisario/metabolismo , Femenino , Prueba de Tolerancia a la Glucosa/métodos , Prueba de Tolerancia a la Glucosa/normas , Humanos , Hipoglucemia/sangre , Hipoglucemia/diagnóstico , Hipoglucemia/metabolismo , Hipopituitarismo/sangre , Hipopituitarismo/diagnóstico , Hipopituitarismo/metabolismo , Masculino , Adulto Joven
12.
Endocrinology ; 161(4)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32100023

RESUMEN

The gut microbiome has been implicated in host metabolism, endocrinology, and pathophysiology. Furthermore, several studies have shown that gut bacteria impact host growth, partially mediated through the growth hormone (GH)/insulin-like growth factor 1 (IGF-1) axis. Yet, no study to date has examined the specific role of GH on the gut microbiome. Our study thus characterized the adult gut microbial profile and intestinal phenotype in GH gene-disrupted (GH-/-) mice (a model of GH deficiency) and bovine GH transgenic (bGH) mice (a model of chronic, excess GH action) at 6 months of age. Both the GH-/- and bGH mice had altered microbial signatures, in opposing directions at the phylum and genus levels. For example, GH-/- mice had significantly reduced abundance in the Proteobacteria, Campylobacterota, and Actinobacteria phyla, whereas bGH mice exhibited a trending increase in those phyla compared with respective controls. Analysis of maturity of the microbial community demonstrated that lack of GH results in a significantly more immature microbiome while excess GH increases microbial maturity. Several common bacterial genera were shared, although in opposing directions, between the 2 mouse lines (e.g., decreased in GH-/- mice and increased in bGH mice), suggesting an association with GH. Similarly, metabolic pathways like acetate, butyrate, heme B, and folate biosynthesis were predicted to be impacted by GH. This study is the first to characterize the gut microbiome in mouse lines with altered GH action and indicates that GH may play a role in the growth of certain microbiota thus impacting microbial maturation and metabolic function.


Asunto(s)
Enanismo Hipofisario/microbiología , Microbioma Gastrointestinal/fisiología , Hormona del Crecimiento/metabolismo , Animales , Enanismo Hipofisario/genética , Enanismo Hipofisario/metabolismo , Hormona del Crecimiento/genética , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos
13.
J Clin Endocrinol Metab ; 105(4)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31917835

RESUMEN

CONTEXT: Daily growth hormone (GH) injections can be burdensome for patients and carers. Somapacitan is a long-acting, reversible albumin-binding GH derivative in development for once-weekly administration in patients with growth hormone deficiency (GHD). OBJECTIVE: The objective of this study is to evaluate the efficacy, safety, and tolerability of once-weekly somapacitan vs once-daily GH. DESIGN: REAL 3 is a multicenter, randomized, controlled, double-blind (somapacitan doses), phase 2 study with a 26-week main and 26-week extension phase (NCT02616562). SETTING: This study took place at 29 sites in 11 countries. PATIENTS: Fifty-nine GH treatment-naive prepubertal children with GHD were randomly assigned; 58 completed the trial. INTERVENTIONS: Interventions comprised 3 somapacitan doses (0.04 [n = 16], 0.08 [n = 15], or 0.16 mg/kg/wk [n = 14]) and daily GH (0.034 mg/kg/d [n = 14]), administered subcutaneously. MAIN OUTCOME MEASURES: The primary end point was height velocity (HV) at week 26. Secondary efficacy end points included HV SD score (SDS) and insulin-like growth factor-I (IGF-I) SDS. RESULTS: At week 26, mean (SD) annualized HV for the somapacitan groups was 8.0 (2.0), 10.9 (1.9), and 12.9 (3.5) cm/year, respectively, vs 11.4 (3.3) cm/year for daily GH; estimated treatment difference (somapacitan 0.16 mg/kg/week-daily GH): 1.7 [95% CI -0.2 to 3.6] cm/year. HV was sustained at week 52, and significantly greater with somapacitan 0.16 mg/kg/week vs daily GH. Mean (SD) change from baseline in HV SDS at week 52 was 4.72 (2.79), 6.14 (3.36), and 8.60 (3.15) for the somapacitan groups, respectively, vs 7.41 (4.08) for daily GH. Model-derived mean (SD) IGF-I SDS for the somapacitan groups was -1.62 (0.86), -1.09 (0.78), and 0.31 (1.06), respectively, vs -0.40 (1.50) observed for daily GH. Safety and tolerability were consistent with the profile of daily GH. CONCLUSIONS: In children with GHD, once-weekly somapacitan 0.16 mg/kg/week provided the closest efficacy match with similar safety and tolerability to daily GH after 26 and 52 weeks of treatment. A short visual summary of our work is available (1).


Asunto(s)
Biomarcadores/análisis , Enanismo Hipofisario/tratamiento farmacológico , Hormona de Crecimiento Humana/administración & dosificación , Niño , Preescolar , Método Doble Ciego , Esquema de Medicación , Enanismo Hipofisario/metabolismo , Enanismo Hipofisario/patología , Femenino , Estudios de Seguimiento , Hormona de Crecimiento Humana/clasificación , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/análisis , Factor I del Crecimiento Similar a la Insulina/análisis , Masculino , Pronóstico
15.
Growth Horm IGF Res ; 50: 23-26, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31835104

RESUMEN

OBJECTIVE: to describe the marked variability in clinical and biochemical patterns that are associated with a p.R209H GH1 missense variant in a large Argentinean pedigree, which makes the diagnosis of GHD elusive. DESIGN: We describe a non-consanguineous pedigree composed by several individuals with short stature, including 2 pediatric patients with typical diagnosis of isolated growth hormone deficiency (IGHD) and 4 other siblings with severe short stature, low serum IGF-1 and IGFBP-3, but normal stimulated GH levels, suggesting growth hormone insensitivity (GHI) in the latter group. RESULTS: Patients with classical IGHD phenotype carried a heterozygous variant in GH1: c.626G>A (p.R209H). Data from the extended pedigree suggested GH1 as the initial candidate gene, which showed the same pathogenic heterozygous GH1 variant in the four siblings with short stature and a biochemical pattern of GHI. CONCLUSIONS: We suggest considering GH1 sequencing in children with short stature associated to low IGF-1 and IGFBP-3 serum levels, even in the context of normal response to growth hormone provocative testing (GHPT).


Asunto(s)
Estatura , Enanismo Hipofisario/genética , Hormona de Crecimiento Humana/genética , Mutación Missense , Adolescente , Adulto , Argentina , Niño , Preescolar , Técnicas de Diagnóstico Endocrino , Enanismo Hipofisario/metabolismo , Enanismo Hipofisario/fisiopatología , Femenino , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/fisiopatología , Heterocigoto , Homocigoto , Hormona de Crecimiento Humana/metabolismo , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Persona de Mediana Edad , Linaje , Adulto Joven
16.
Horm Res Paediatr ; 92(2): 115-123, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31715605

RESUMEN

BACKGROUND: Floating-Harbor syndrome (FHS) is a rare condition characterized by dysmorphic facial features, short stature, and expressive language delay. OBJECTIVE: The aim of this study was to describe a cohort of patients with FHS and review the literature about the response to recombinant human growth hormone (rhGH) therapy. METHODS: Anthropometric and laboratory data from 7 patients with FHS were described. The molecular diagnosis was established by multigene analysis. Moreover, we reviewed the literature concerning patients with FHS treated with rhGH. RESULTS: All 7 patients were born small for gestational age. At first evaluation, 6 patients had a height standard deviation score (SDS) ≤-2 and 1 had short stature in relation to their target height. Bone age was usually delayed, which rapidly advanced during puberty. Nonspecific skeletal abnormalities were frequently noticed, and normal to elevated plasma IGF-I levels were observed in all except 1 patient with growth hormone deficiency. Information about 20 patients with FHS treated with rhGH was analyzed (4 from our cohort and 16 from the literature). The median height changes during the treatment period (approx. 2.9 years) were 1.1 SDS (range from -0.4 to 3.1). Nontreated patients had an adult height SDS of -4.1 ± 1.2 (n = 10) versus -2.6 ± 0.8 SDS (n = 7, p 0.012) for treated patients. CONCLUSION: We observed a laboratory profile compatible with IGF-1 insensitivity in some patients with FHS. Nevertheless, our study suggests that children with FHS may be considered as candidates for rhGH therapy. Further studies are necessary to establish the real benefit and safety of rhGH therapy in these patients.


Asunto(s)
Anomalías Múltiples , Desarrollo del Adolescente/efectos de los fármacos , Desarrollo Infantil/efectos de los fármacos , Anomalías Craneofaciales , Enanismo Hipofisario , Trastornos del Crecimiento , Defectos del Tabique Interventricular , Hormona de Crecimiento Humana/uso terapéutico , Pubertad/efectos de los fármacos , Anomalías Múltiples/tratamiento farmacológico , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Anomalías Múltiples/fisiopatología , Adolescente , Estatura/efectos de los fármacos , Niño , Preescolar , Anomalías Craneofaciales/tratamiento farmacológico , Anomalías Craneofaciales/metabolismo , Anomalías Craneofaciales/patología , Anomalías Craneofaciales/fisiopatología , Enanismo Hipofisario/tratamiento farmacológico , Enanismo Hipofisario/metabolismo , Enanismo Hipofisario/patología , Enanismo Hipofisario/fisiopatología , Femenino , Trastornos del Crecimiento/tratamiento farmacológico , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/patología , Trastornos del Crecimiento/fisiopatología , Defectos del Tabique Interventricular/tratamiento farmacológico , Defectos del Tabique Interventricular/metabolismo , Defectos del Tabique Interventricular/patología , Defectos del Tabique Interventricular/fisiopatología , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino
17.
Artículo en Inglés | MEDLINE | ID: mdl-31544067

RESUMEN

Isolated growth hormone (GH) deficiency (IGHD) affects approximately 1 in 4,000 to 1 in 10,000 individuals worldwide. We have previously described a large cohort of subjects with IGHD due to a homozygous mutation in the GH releasing hormone (GHRH) receptor gene. These subjects exhibit throughout the life very low levels of GH and its principal mediator, the Insulin Growth Factor-I (IGF-I). The facilitating role of IGF-I in the infection of mouse macrophages by different Leishmania strains is well-known. Nevertheless, the role of IGF-I in Leishmania infection of human macrophages has not been studied. This study aimed to evaluate the behavior of Leishmania infection in vitro in macrophages from untreated IGHD subjects. To this end, blood samples were collected from 14 IGHD individuals and 14 age and sex-matched healthy controls. Monocytes were isolated and derived into macrophages and infected with a strain of Leishmania amazonensis. In addition, IGF-I was added to culture medium to evaluate its effect on the infection. Cytokines were measured in the culture supernatants. We found that macrophages from IGHD subjects were less prone to Leishmania infection compared to GH sufficient controls. Both inflammatory and anti-inflammatory cytokines increase only in the supernatants of the control macrophages. Addition of IGF-I to the culture medium increased infection rates. In conclusion, we demonstrated that IGF-I is crucial for Leishmania infection of human macrophages.


Asunto(s)
Enanismo Hipofisario/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Leishmania mexicana/metabolismo , Leishmaniasis/inmunología , Macrófagos/metabolismo , Mutación , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Adulto , Animales , Citocinas/metabolismo , Femenino , Humanos , Leishmaniasis/microbiología , Masculino , Ratones , Persona de Mediana Edad , Fagocitosis , ARN Mensajero/metabolismo , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Adulto Joven
18.
J Mol Graph Model ; 92: 1-7, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31279174

RESUMEN

Isolated growth hormone deficiency (IGHD) is the most common pituitary hormone deficiency and can result from congenital or acquired causes. Among the known factors, genetic mutations in human growth hormone (hGH) remain the most frequent cause of IGHD, which influence the binding of hGH to its cognate receptor (hGHbp). Although previous studies have systematically investigated the residue importance at hGH-hGHbp complex interface, the molecular role of IGHD-associated residue mutations in the complex function still remains largely unexplored. Here, a total of 21 known hGH naturally-occurring missence mutations that have been clinically observed to be involved in IGHD disorder are collected and confirmed by original literature; they effects on the conformation, energetics and dynamics of hGH-hGHbp recognition and interaction are dissected at molecular level by using atomistic dynamics simulations, binding energy calculations and fluorescence spectroscopy assays. A systematic profile of hGH-hGHbp binding response to these clinical missence mutations is created, based on which it is revealed that (i) most mutations have appreciably unfavorable effect on the binding, which potentially destabilize the complex interaction, while only very few are predicted as moderate stabilizers for the complex system, and (ii) these disease-related mutations can locate either at complex interface or in hGH protein interior far away from the interface; both can influence the complex binding through either direct interaction or indirect allostericity. Two mutations, E100K (non-interface) and G146R (interface), are identified to address potent destabilization effect on hGH-hGHbp complex system; they can reduce the complex binding affinity by 8-fold (Kd changes from 0.76 to 5.9 nM) and 46-fold (Kd changes from 0.76 to 34.7 nM), respectively.


Asunto(s)
Enanismo Hipofisario/genética , Hormona de Crecimiento Humana/química , Hormona de Crecimiento Humana/genética , Modelos Moleculares , Mutación Missense , Receptores de Somatotropina/química , Secuencia de Aminoácidos , Sitios de Unión , Enanismo Hipofisario/metabolismo , Hormona de Crecimiento Humana/metabolismo , Humanos , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Receptores de Somatotropina/metabolismo , Relación Estructura-Actividad
19.
J Clin Endocrinol Metab ; 104(7): 2777-2784, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30860584

RESUMEN

CONTEXT: GH and IGF-1 are crucial for attainment of normal body size and regulation of food intake, nutrient storage, and insulin sensitivity. Enteroendocrine connections exist between the GH-IGF-1 axis and insulin, ghrelin, and glucagon-like peptide 1 (GLP-1). The status of these connections in GH deficiency (GHD) is unknown. OBJECTIVE: To study the enteroendocrine connections before and after a standard meal test in a homogeneous population of adults with congenital untreated isolated GHD (IGHD) due to a mutation in the GHRH receptor gene. DESIGN: In a cross-sectional study of 20 individuals with IGHD and 20 control subjects, we measured glucose, insulin, ghrelin, and GLP-1 before and 30, 60, 120, and 180 minutes after a standardized test meal. Homeostasis model assessment index of insulin resistance (HOMA-IR) and homeostasis model assessment (HOMA)-ß were calculated. Participants scored feelings of hunger, fullness, and prospective food consumption on a visual analog scale. MAIN OUTCOME MEASURES: Area under the curve (AUC) values of glucose, insulin, ghrelin, GLP-1, hunger, fullness, and prospective food consumption. RESULTS: Fasting HOMA-IR and HOMA-ß were lower in individuals with IGHD than in control subjects (P = 0.002 and P = 0.023, respectively). AUC was higher for hunger (P < 0.0001), glucose (P = 0.0157), ghrelin (P < 0.0001), and GLP-1 (P < 0.0001) and smaller for fullness (P < 0.0001) in individuals with IGHD compared with control subjects. There was no difference in AUC for prospective food consumption or insulin. CONCLUSIONS: Untreated IGHD is associated with increased GLP-1 secretion and reduced postprandial ghrelin and hunger attenuation in response to a mixed meal. These enteroendocrine connections can result in a favorable outcome in terms of environmental adaptation and guaranteeing appropriate food intake and can confer metabolic benefits.


Asunto(s)
Glucemia/metabolismo , Enanismo Hipofisario/metabolismo , Ghrelina/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Periodo Posprandial , Receptores LHRH/genética , Adulto , Área Bajo la Curva , Estudios de Casos y Controles , Enanismo Hipofisario/genética , Ingestión de Alimentos , Femenino , Humanos , Hambre , Masculino , Persona de Mediana Edad , Mutación , Respuesta de Saciedad
20.
Psychoneuroendocrinology ; 103: 67-75, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30658340

RESUMEN

PURPOSE: The aim of the present study was to investigate the differences in the topological organization of functional brain networks between children with growth hormone deficiency (GHD) and those with idiopathic short stature (ISS). METHODS: Thirty-one children with GHD and fifty-three children with ISS were recruited based on the results of GH stimulation tests. Resting-state fMRI data were acquired from all children. Whole brain functional connectivity density (FCD) analysis and subsequent seed-based functional connectivity analysis were used to explore the differences in functional brain networks between the children with ISS and GHD. Correlation analyses among the results of clinical laboratory examinations, neuropsychological scales and FCD values of different brain regions were applied. RESULTS: Compared with the ISS group, the GHD group exhibited significantly decreased FCDs in the left postcentral gyrus, right precentral gyrus and left cerebellar lobules 7b and 6. The subsequent functional connectivity analysis found decreased functional connectivity between lobules 7b and 6 of the left cerebellum as well as the left postcentral gyrus and right precentral gyrus in the GHD group compared to that in the ISS group. In addition, the FCD values of region 6 of the left cerebellum in the GHD group were negatively correlated with the scores on the Symptom Checklist-90 and Eysenck Personality Questionnaire. The FCD value of the left postcentral gyrus in children with ISS positively correlated with IGFBP-3 levels and was approximately correlated with IGF-1 levels. CONCLUSIONS: These findings highlight the impact of growth hormone deficiency on the brain network that mainly involves the somatosensory, somatic motor and cerebellum networks, which may contribute to the behavioural problems observed in these children.


Asunto(s)
Enanismo Hipofisario/metabolismo , Hormona de Crecimiento Humana/deficiencia , Encéfalo/metabolismo , Encéfalo/fisiología , Niño , Preescolar , Conectoma/métodos , Enanismo Hipofisario/fisiopatología , Femenino , Hormona del Crecimiento/deficiencia , Hormona del Crecimiento/metabolismo , Hormona de Crecimiento Humana/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Imagen por Resonancia Magnética/métodos , Masculino , Red Nerviosa/metabolismo , Red Nerviosa/fisiología , Descanso , Corteza Somatosensorial/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA