Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Mol Nutr Food Res ; 68(5): e2300270, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38389198

RESUMEN

SCOPE: The disturbance of the hypothalamic-pituitary-gonadal (HPG) axis, gut microbiota (GM) community, and short-chain fatty acids (SCFAs) is a triggering factor for pubertal onset. The study investigates the effects of the long-term intake of aspartame on puberty and GM in animals and humans. METHODS AND RESULTS: Aspartame-fed female offspring rats result in vaginal opening time prolongation, serum estrogen reduction, and serum luteinizing hormone elevation. , 60 mg kg-1 aspartame treatment decreases the mRNA levels of gonadotropin-releasing hormone (GnRH), Kiss1, and G protein-coupled receptor 54 (GPR54), increases the mRNA level of RFamide-related peptide-3 (RFRP-3), and decreases the expression of GnRH neurons in the hypothalamus. Significant differences in relative bacterial abundance at the genus levels and decreased fecal SCFA levels are noted by 60 mg kg-1 aspartame treatment. Among which, Escherichia-Shigella is negatively correlated with several SCFAs. In girls, high-dose aspartame consumption decreases the risk of precocious puberty. CONCLUSIONS: Aspartame reduces the chance of puberty occurring earlier than usual in female offspring and girls. Particularly, 60 mg kg-1 aspartame-fed female offspring delays pubertal onset through the dysregulation of HPG axis and GM composition by inhibiting the Kiss1/GPR54 system and inducing the RFRP-3. An acceptable dose of aspartame should be recommended during childhood.


Asunto(s)
Kisspeptinas , Pubertad Tardía , Humanos , Ratas , Femenino , Animales , Kisspeptinas/metabolismo , Kisspeptinas/farmacología , Aspartame/efectos adversos , Aspartame/metabolismo , Pubertad Tardía/metabolismo , Ratas Sprague-Dawley , Maduración Sexual/fisiología , Hormona Liberadora de Gonadotropina/genética , Hipotálamo/metabolismo , Pubertad , ARN Mensajero/metabolismo
2.
Mol Cell Endocrinol ; 518: 111006, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32861700

RESUMEN

Acid-labile subunit (ALS) deficiency (ACLSD) constitutes the first monogenic defect involving a member of the Insulin-like Growth Factor (IGF) binding protein system. The lack of ALS completely disrupts the circulating IGF system. Autocrine/paracrine action of local produced IGF-I could explain the mild effect on growth. In the present work we have revised the more relevant clinical and biochemical consequences of complete ACLSD in 61 reported subjects from 31 families. Low birth weight and/or length, reduced head circumference, height between -2 and -3 SD, pubertal delay and insulin resistance are commonly observed. Partial ACLSD could be present in children initially labeled as idiopathic short stature, presenting low IGF-I levels, suggesting that one functional IGFALS allele is insufficient to stabilize ternary complexes. Dysfunction of the GH-IGF axis observed in ACLSD may eventually result in increased risk for type-2 diabetes and tumor progression. Consequently, long term surveillance is recommended in these patients.


Asunto(s)
Proteínas Portadoras/fisiología , Glicoproteínas/fisiología , Hormona de Crecimiento Humana/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Estatura/efectos de los fármacos , Estatura/genética , Proteínas Portadoras/genética , Proteínas Portadoras/farmacología , Niño , Femenino , Glicoproteínas/deficiencia , Glicoproteínas/genética , Glicoproteínas/farmacología , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/metabolismo , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Masculino , Pubertad Tardía/genética , Pubertad Tardía/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
3.
JCI Insight ; 5(11)2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32493844

RESUMEN

The initiation of puberty is driven by an upsurge in hypothalamic gonadotropin-releasing hormone (GnRH) secretion. In turn, GnRH secretion upsurge depends on the development of a complex GnRH neuroendocrine network during embryonic life. Although delayed puberty (DP) affects up to 2% of the population, is highly heritable, and is associated with adverse health outcomes, the genes underlying DP remain largely unknown. We aimed to discover regulators by whole-exome sequencing of 160 individuals of 67 multigenerational families in our large, accurately phenotyped DP cohort. LGR4 was the only gene remaining after analysis that was significantly enriched for potentially pathogenic, rare variants in 6 probands. Expression analysis identified specific Lgr4 expression at the site of GnRH neuron development. LGR4 mutant proteins showed impaired Wnt/ß-catenin signaling, owing to defective protein expression, trafficking, and degradation. Mice deficient in Lgr4 had significantly delayed onset of puberty and fewer GnRH neurons compared with WT, whereas lgr4 knockdown in zebrafish embryos prevented formation and migration of GnRH neurons. Further, genetic lineage tracing showed strong Lgr4-mediated Wnt/ß-catenin signaling pathway activation during GnRH neuron development. In conclusion, our results show that LGR4 deficiency impairs Wnt/ß-catenin signaling with observed defects in GnRH neuron development, resulting in a DP phenotype.


Asunto(s)
Neuronas , Pubertad Tardía , Receptores Acoplados a Proteínas G/deficiencia , Vía de Señalización Wnt , Animales , Femenino , Estudios de Seguimiento , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Masculino , Ratones , Neuronas/metabolismo , Neuronas/patología , Pubertad Tardía/genética , Pubertad Tardía/metabolismo , Pubertad Tardía/patología , Receptores Acoplados a Proteínas G/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
4.
PLoS Biol ; 17(3): e3000189, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30893295

RESUMEN

Insulin resistance and obesity are associated with reduced gonadotropin-releasing hormone (GnRH) release and infertility. Mice that lack insulin receptors (IRs) throughout development in both neuronal and non-neuronal brain cells are known to exhibit subfertility due to hypogonadotropic hypogonadism. However, attempts to recapitulate this phenotype by targeting specific neurons have failed. To determine whether astrocytic insulin sensing plays a role in the regulation of fertility, we generated mice lacking IRs in astrocytes (astrocyte-specific insulin receptor deletion [IRKOGFAP] mice). IRKOGFAP males and females showed a delay in balanopreputial separation or vaginal opening and first estrous, respectively. In adulthood, IRKOGFAP female mice also exhibited longer, irregular estrus cycles, decreased pregnancy rates, and reduced litter sizes. IRKOGFAP mice show normal sexual behavior but hypothalamic-pituitary-gonadotropin (HPG) axis dysregulation, likely explaining their low fecundity. Histological examination of testes and ovaries showed impaired spermatogenesis and ovarian follicle maturation. Finally, reduced prostaglandin E synthase 2 (PGES2) levels were found in astrocytes isolated from these mice, suggesting a mechanism for low GnRH/luteinizing hormone (LH) secretion. These findings demonstrate that insulin sensing by astrocytes is indispensable for the function of the reproductive axis. Additional work is needed to elucidate the role of astrocytes in the maturation of hypothalamic reproductive circuits.


Asunto(s)
Astrocitos/metabolismo , Receptor de Insulina/metabolismo , Animales , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Ratones , Prostaglandina-E Sintasas/metabolismo , Pubertad Tardía/metabolismo
5.
Chem Res Toxicol ; 31(12): 1315-1322, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30422632

RESUMEN

Ziram is a dimethyldithiocarbamate fungicide, which may influence the male reproductive system as a potential endocrine disruptor. We interrogated the disruption of ziram on rat progenitor Leydig cell development. Prepubertal male Sprague-Dawley rats were orally treated with 0, 2, 4, or 8 mg/kg ziram for 2 weeks. We investigated the effects of ziram on serum testosterone levels, Leydig cell number, and Leydig and Sertoli cell gene and protein expression, SIRT1/PGC-1α levels, and phosphorylation of AKT1, ERK1/2, and AMPK in vivo. We also interrogated the effects of ziram on reactive oxidative species (ROS) level, apoptosis rate, and mitochondrial membrane potential of progenitor Leydig cells in vitro. Ziram decreased serum testosterone and follicle-stimulating hormone levels, the down-regulated Leydig cell-specific gene ( Lhcgr, Scarb1, Star, Cyp17a1, and Hsd17b3), and their protein expression. However, ziram stimulated anti-Müllerian hormone production. Ziram lowered SIRT1/PGC-1α and phosphorylated protein levels of AKT1. Ziram induced ROS and apoptosis and lowered the mitochondrial membrane potential of progenitor Leydig cells in vitro. In conclusion, ziram disrupts Leydig cell development during the prepubertal period potentially through the SIRT1/PGC-1α and phosphorylated AKT1 signaling.


Asunto(s)
Fungicidas Industriales/toxicidad , Pubertad Tardía/etiología , Transducción de Señal/efectos de los fármacos , Testículo/efectos de los fármacos , Ziram/toxicidad , Animales , Apoptosis/efectos de los fármacos , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Fungicidas Industriales/química , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pubertad Tardía/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 1/metabolismo , Testículo/metabolismo , Testosterona/sangre , Ziram/química
7.
Neuroendocrinology ; 106(3): 283-291, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28926843

RESUMEN

The genetic control of puberty remains an important but mostly unanswered question. Late pubertal timing affects over 2% of adolescents and is associated with adverse health outcomes including short stature, reduced bone mineral density, and compromised psychosocial health. Self-limited delayed puberty (DP) is a highly heritable trait, which often segregates in an autosomal dominant pattern; however, its neuroendocrine pathophysiology and genetic regulation remain unclear. Some insights into the genetic mutations that lead to familial DP have come from sequencing genes known to cause gonadotropin-releasing hormone (GnRH) deficiency, most recently via next-generation sequencing, and others from large-scale genome-wide association studies in the general population. Investigation of the genetic control of DP is complicated by the fact that this trait is not rare and that the phenotype is likely to represent a final common pathway, with a variety of different pathogenic mechanisms affecting the release of the puberty "brake." These include abnormalities of GnRH neuronal development and function, GnRH receptor and luteinizing hormone/follicle-stimulating hormone abnormalities, metabolic and energy homeostatic derangements, and transcriptional regulation of the hypothalamic-pituitary-gonadal axis. Thus, genetic control of pubertal timing can range from early fetal life via development of the GnRH network to those factors directly influencing the puberty brake during mid-childhood.


Asunto(s)
Predisposición Genética a la Enfermedad , Pubertad Tardía/genética , Animales , Humanos , Pubertad Tardía/metabolismo
8.
Eur J Endocrinol ; 176(6): 657-667, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28249955

RESUMEN

OBJECTIVE: Acid-labile subunit (ALS) deficiency (ACLSD), caused by homozygous or compound heterozygous IGFALS mutations, is associated with moderate short stature, delayed puberty, low serum IGF-I and ALS and extremely low serum IGFBP-3. Its effect on birth weight, head circumference, bone mineral density (BMD), serum IGF-II and IGFBP-2 is uncertain, as well as the phenotype of heterozygous carriers of IGFALS mutations (partial ACLSD). DESIGN: From all available members of five Turkish families, carrying three mutations in exon 2 of IGFALS (c.1462G > A, p.Asp488Asn (families A, B, E); c.251A > G, p.Asn84Ser (families C and E) and c.1477del, p.Arg493fs (family D)), clinical, laboratory and BMD data were collected. METHODS: Auxological and biochemical findings were expressed as SDS for age and gender. Ternary complex formation in serum was investigated by size-exclusion chromatography. BMD using DXA bone densitometry was adjusted for height and age (Ha-BMD z-score). RESULTS: In ACLSD (n = 24), mean ± s.d. height SDS (-2.7 ± 1.2), head circumference SDS (-2.3 ± 0.5) and body mass index (BMI) (-0.6 ± 1.0 SDS) were lower than those in partial ACLSD (n = 26, P ≤ 0.01) and birth weight SDS (n = 7) tended to be lower (-2.2 ± 1.1 vs -0.6 ± 0.3 in partial ACLSD (P = 0.07)). Serum IGF-I was -3.7 ± 1.4 vs -1.0 ± 1.0, IGF-II: -5.6 ± 0.7 vs -1.3 ± 0.7, ALS: <-4.4 ± 1.2 vs -2.1 ± 0.9 and IGFBP-3: -9.0 ± 1.9 vs -1.6 ± 0.8 SDS respectively (P < 0.001). Ha-BMD z-score was similar and normal in both groups. CONCLUSIONS: To the known phenotype of ACLSD (i.e. short stature, reduced serum levels of IGF-I and ALS, extremely low serum IGFBP-3 and disturbed ternary complex formation), we add reduced birth weight, head circumference and serum IGF-II.


Asunto(s)
Densidad Ósea/genética , Proteínas Portadoras/genética , Glicoproteínas/genética , Trastornos del Crecimiento/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Absorciometría de Fotón , Adolescente , Peso al Nacer/genética , Índice de Masa Corporal , Proteínas Portadoras/metabolismo , Cefalometría , Niño , Femenino , Glicoproteínas/metabolismo , Trastornos del Crecimiento/diagnóstico por imagen , Trastornos del Crecimiento/metabolismo , Heterocigoto , Homocigoto , Humanos , Masculino , Mutación , Fenotipo , Pubertad Tardía/genética , Pubertad Tardía/metabolismo , Turquía
9.
Diabetes ; 65(7): 2051-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27207549

RESUMEN

A mechanistic cause for Mauriac syndrome, a syndrome of growth failure and delayed puberty associated with massive liver enlargement from glycogen deposition in children with poorly controlled type 1 diabetes, is unknown. We discovered a mutation in the catalytic subunit of liver glycogen phosphorylase kinase in a patient with Mauriac syndrome whose liver extended into his pelvis. Glycogen phosphorylase kinase activates glycogen phosphorylase, the enzyme that catalyzes the first step in glycogen breakdown. We show that the mutant subunit acts in a dominant manner to completely inhibit glycogen phosphorylase kinase enzyme activity and that this interferes with glycogenolysis causing increased levels of glycogen in human liver cells. It is known that even normal blood glucose levels physiologically inhibit glycogen phosphorylase to diminish glucose release from the liver when glycogenolysis is not needed. The patient's mother possessed the same mutant glycogen phosphorylase kinase subunit, but did not have diabetes or hepatomegaly. His father had childhood type 1 diabetes in poor glycemic control, but lacked the mutation and had neither hepatomegaly nor growth failure. This case proves that the effect of a mutant enzyme of glycogen metabolism can combine with hyperglycemia to directly hyperinhibit glycogen phosphorylase, in turn blocking glycogenolysis causing the massive liver in Mauriac disease.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Glucógeno Fosforilasa de Forma Hepática/metabolismo , Glucógeno/metabolismo , Trastornos del Crecimiento/genética , Hepatomegalia/genética , Fosforilasa Quinasa/genética , Pubertad Tardía/genética , Adolescente , Diabetes Mellitus Tipo 1/metabolismo , Trastornos del Crecimiento/metabolismo , Hepatomegalia/metabolismo , Humanos , Masculino , Mutación , Fosforilasa Quinasa/metabolismo , Pubertad Tardía/metabolismo , Síndrome
10.
Hum Reprod ; 31(4): 897-904, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26905078

RESUMEN

STUDY QUESTION: Do variants of the genes encoding follicle stimulating hormone (FSH) beta subunit (B) and FSH receptor (R) impact circulating reproductive hormone levels and ovarian follicle maturation in healthy peripubertal girls? SUMMARY ANSWER: FSHB and FSHR genetic variants exert, alone or their combination, distinct effects on reproductive hormone levels as well as ovarian follicle maturation in healthy peripubertal girls. WHAT IS KNOWN ALREADY: FSHB and FSHR genetic variants impact reproductive hormone levels as well as associated pathologies in women. While FSHR c. 2039A>G is known to alter gonadotrophin levels in women, FSHR c.-29G>A has not yet been shown to exert effect and there are conflicting results concerning FSHB c.-211G>T. STUDY DESIGN, SIZE, DURATION: This population-based study included 633 girls recruited as part of two cohorts, the COPENHAGEN Puberty Study (2006-2014, a cross-sectional and ongoing longitudinal study) and the Copenhagen Mother-Child Cohort (1997-2002, including transabdominal ultrasound (TAUS) of the ovaries in a subset of 91 peripubertal girls). PARTICIPANTS/MATERIALS, SETTING, METHODS: Clinical examinations, including pubertal breast stage (Tanner's classification B1-B5) were performed. Circulating levels of FSH, luteinizing hormone (LH), estradiol, anti-Mullerian hormone (AMH) and inhibin-B were assessed by immunoassays. In a subset of the girls (n = 91), ovarian volume and the number/size of antral follicles were assessed by TAUS. Genotypes were determined by competitive PCR. MAIN RESULTS AND THE ROLE OF CHANCE: FSHR c.2039A>G minor alleles were positively associated with serum FSH (ß = 0.08, P = 0.004), LH (ß = 0.06, P = 0.012) and estradiol (ß = 0.06, P = 0.017) (adjusted for Tanner stages). In a combined model, FSHR c.-29G>A and FSHR c.2039A>G alleles were positively associated with FSH levels in early-pubertal girls (B2 + B3, n = 327, r = 0.1, P = 0.02) and in young adolescents (B4 + B5, n = 149, r = 0.2, P = 0.01). Serum AMH and inhibin B levels were not significantly influenced by the single nucleotide polymorphisms (SNPs). Single SNPs were not associated with follicles counts, however, a cumulative minor allele count (FSHB c.-211 G>T and FSHR c.-29G>A) was negatively associated with the number of large follicles (≥5 mm) (n = 91, P = 0.04) (adjusted for Tanner stages). LIMITATIONS, REASONS FOR CAUTION: Since we studied girls and young adolescents during pubertal transition, our study may not be fully comparable with previous studies on FSHB and FSHR variants in adult women. The group of young adolescents (Tanner B4 + B5) reflects the endocrine situation in adult women best, however, the group is not large enough to contribute substantially to the conflicting results concerning the influence of FSHB c.-211G>T in adult women. Furthermore, we have no information about the exact day of the menstrual cycle in the subgroup of girls with menarche. WIDER IMPLICATIONS OF THE FINDINGS: The sex-specific interaction of FSHB and FSHR genetic variants and physiological as well as pathological conditions is being increasingly elucidated. The variant triplet set might serve as diagnostic and pharmacogenetic marker. For the first time, we show an additional effect of FSHR c.-29G>A on serum FSH levels in healthy girls. Moreover, morphological data suggest impaired FSH-induced maturation of ovarian follicles in minor allele carriers of FSHB c.-211G>T and FSHR c.-29G>A. This may explain previous findings of delayed pubertal onset in these girls. STUDY FUNDING/COMPETING INTERESTS: Funding was provided by the Danish Agency for Science, Technology and Innovation (09-067180), Danish Ministry of the Environment, CeHoS (MST-621-00065), Capital Region of Denmark (December 2011), Ministry of Higher Education and Science (DFF-1331-00113) and EDMaRC (Danish Ministry of Health). A.S.B. was funded from December 2015 by ReproUnion (EU Interreg Öresund-Kattegat-Skagerrak). The authors declare no conflict of interest.


Asunto(s)
Hormona Folículo Estimulante de Subunidad beta/genética , Folículo Ovárico/patología , Polimorfismo Genético , Pubertad Tardía/genética , Receptores de HFE/genética , Adolescente , Adulto , Alelos , Niño , Estudios de Cohortes , Estudios Transversales , Dinamarca , Estradiol/sangre , Femenino , Hormona Folículo Estimulante Humana/sangre , Hormona Folículo Estimulante de Subunidad beta/sangre , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Estudios de Asociación Genética , Humanos , Inhibinas/sangre , Estudios Longitudinales , Hormona Luteinizante/sangre , Polimorfismo de Nucleótido Simple , Pubertad Tardía/sangre , Pubertad Tardía/metabolismo , Pubertad Tardía/patología , Receptores de HFE/sangre , Receptores de HFE/metabolismo , Adulto Joven
11.
Mol Endocrinol ; 30(1): 62-76, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26554443

RESUMEN

We demonstrate 2 novel mutations of the LHCGR, each homozygous, in a 46,XY patient with severe Leydig cell hypoplasia. One is a mutation in the signal peptide (p.Gln18_Leu19ins9; referred to here as SP) that results in an alteration of the coding sequence of the N terminus of the mature mutant receptor. The other mutation (p.G71R) is also within the ectodomain. Similar to many other inactivating mutations, the cell surface expression of recombinant human LHR(SP,G71R) is greatly reduced due to intracellular retention. However, we made the unusual discovery that the intrinsic efficacy for agonist-stimulated cAMP in the reduced numbers of receptors on the cell surface was greatly increased relative to the same low number of cell surface wild-type receptor. Remarkably, this appears to be a general attribute of misfolding mutations in the ectodomains, but not serpentine domains, of the gonadotropin receptors. These findings suggest that there must be a common, shared mechanism by which disparate mutations in the ectodomain that cause misfolding and therefore reduced cell surface expression concomitantly confer increased agonist efficacy to those receptor mutants on the cell surface. Our data further suggest that, due to their increased agonist efficacy, extremely small changes in cell surface expression of misfolded ectodomain mutants cause larger than expected alterations in the cellular response to agonist. Therefore, for inactivating LHCGR mutations causing ectodomain misfolding, the numbers of cell surface mutant receptors on fetal Leydig cells of 46,XY individuals exert a more exquisite effect on the relative severity of the clinical phenotypes than already appreciated.


Asunto(s)
Células Intersticiales del Testículo/metabolismo , Mutación , Pubertad Tardía/metabolismo , Receptores de HL/metabolismo , Adolescente , Femenino , Humanos , Masculino , Pliegue de Proteína , Pubertad Tardía/genética , Receptores de HL/genética , Transducción de Señal
12.
Am J Hematol ; 89(12): 1102-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25197009

RESUMEN

Iron overload in ß-thalassemia major (TM) typically results in iron-induced cardiomyopathy, liver disease, and endocrine complications. We examined the incidence and progression of endocrine disorders (hypothyroidism, diabetes, hypoparathyroidism, hypogonadism), growth and pubertal delay, and bone metabolism disease during long-term deferasirox chelation therapy in a real clinical practice setting. We report a multicenter retrospective cohort study of 86 transfusion-dependent patients with TM treated with once daily deferasirox for a median duration of 6.5 years, up to 10 years. No deaths or new cases of hypothyroidism or diabetes occurred. The incidence of new endocrine complications was 7% (P = 0.338, for change of prevalence from baseline to end of study) and included hypogonadism (n = 5) and hypoparathyroidism (n = 1). Among patients with hypothyroidism or diabetes at baseline, no significant change in thyroid parameters or insulin requirements were observed, respectively. Mean lumbar spine bone mineral density increased significantly (P < 0.001) and the number of patients with lumbar spine osteoporosis significantly decreased (P = 0.022) irrespective of bisphosphonate therapy, hormonal replacement therapy, and calcium or vitamin D supplementation. There were no significant differences in the number of pediatric patients below the 5th centile for height between baseline and study completion. Six pregnancies occurred successfully, and four of them were spontaneous without ovarian stimulation. This is the first study evaluating endocrine function during the newest oral chelation therapy with deferasirox. A low rate of new endocrine disorders and a stabilization of those pre-exisisting was observed in a real clinical practice setting.


Asunto(s)
Benzoatos/uso terapéutico , Terapia por Quelación , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Osteoporosis/prevención & control , Triazoles/uso terapéutico , Talasemia beta/tratamiento farmacológico , Adolescente , Adulto , Densidad Ósea , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Calcio/administración & dosificación , Niño , Preescolar , Deferasirox , Diabetes Mellitus/etiología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Diabetes Mellitus/prevención & control , Difosfonatos/uso terapéutico , Femenino , Humanos , Hipogonadismo/etiología , Hipogonadismo/metabolismo , Hipogonadismo/patología , Hipogonadismo/prevención & control , Hipoparatiroidismo/etiología , Hipoparatiroidismo/metabolismo , Hipoparatiroidismo/patología , Hipoparatiroidismo/prevención & control , Hipotiroidismo/etiología , Hipotiroidismo/metabolismo , Hipotiroidismo/patología , Hipotiroidismo/prevención & control , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Masculino , Persona de Mediana Edad , Osteoporosis/etiología , Osteoporosis/metabolismo , Osteoporosis/patología , Pubertad Tardía/etiología , Pubertad Tardía/metabolismo , Pubertad Tardía/patología , Pubertad Tardía/prevención & control , Estudios Retrospectivos , Vitamina D/administración & dosificación , Talasemia beta/complicaciones , Talasemia beta/metabolismo , Talasemia beta/patología
13.
J Clin Endocrinol Metab ; 99(10): E2067-75, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25033069

RESUMEN

CONTEXT: Gordon Holmes syndrome (GHS) is characterized by cerebellar ataxia/atrophy and normosmic hypogonadotropic hypogonadism (nHH). The underlying pathophysiology of this combined neurodegeneration and nHH remains unknown. OBJECTIVE: We aimed to provide insight into the disease mechanism in GHS. METHODS: We studied a cohort of 6 multiplex families with GHS through autozygosity mapping and whole-exome sequencing. RESULTS: We identified 6 patients from 3 independent families carrying loss-of-function mutations in PNPLA6, which encodes neuropathy target esterase (NTE), a lysophospholipase that maintains intracellular phospholipid homeostasis by converting lysophosphatidylcholine to glycerophosphocholine. Wild-type PNPLA6, but not PNPLA6 bearing these mutations, rescued a well-established Drosophila neurodegenerative phenotype caused by the absence of sws, the fly ortholog of mammalian PNPLA6. Inhibition of NTE activity in the LßT2 gonadotrope cell line diminished LH response to GnRH by reducing GnRH-stimulated LH exocytosis, without affecting GnRH receptor signaling or LHß synthesis. CONCLUSION: These results suggest that NTE-dependent alteration of phospholipid homeostasis in GHS causes both neurodegeneration and impaired LH release from pituitary gonadotropes, leading to nHH.


Asunto(s)
Ataxia Cerebelosa/genética , Hormona Liberadora de Gonadotropina/deficiencia , Hipogonadismo/genética , Degeneración Nerviosa/genética , Fosfolipasas/genética , Pubertad Tardía/genética , Adolescente , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Ataxia Cerebelosa/metabolismo , Salud de la Familia , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Homeostasis/genética , Humanos , Hipogonadismo/metabolismo , Masculino , Persona de Mediana Edad , Degeneración Nerviosa/metabolismo , Linaje , Fosfolipasas/metabolismo , Fosfolípidos/metabolismo , Pubertad Tardía/metabolismo
14.
Mol Endocrinol ; 28(1): 99-115, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24284824

RESUMEN

Gestational exposures to estrogenic compounds, both endogenous hormones and exogenous endocrine-disrupting chemicals (EDCs), have long-term effects on reproductive physiology and behavior. We tested the hypothesis that prenatal treatment of rats with low doses of Aroclor 1221 (A1221), a weakly estrogenic polychlorinated biphenyl mix previously used in industry, or estradiol benzoate (EB), alters development of the hypothalamus in a sexually dimorphic manner and subsequently perturbs reproductive function. Pregnant Sprague-Dawley rats were injected on embryonic days 16 and 18 with vehicle (dimethylsulfoxide), A1221 (1 mg/kg), or EB (50 µg/kg). Developmental milestones were monitored, and on postnatal days 15, 30, 45, and 90, 1 male and 1 female per litter were euthanized. Because of their key roles in the mediation of steroid actions on reproductive function, the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC) were punched for a low-density quantitative PCR array of 48 neuroendocrine genes and analysis of DNA methylation of a subset of genes. Gestational exposure to A1221 or EB delayed the timing of puberty in males and disrupted estrous cyclicity in females. In the AVPV, 28 genes were affected by treatment in a developmental stage-specific manner, mostly in females, which exhibited a masculinized expression profile. This included 2 clock genes, Per2 and Arntl, implicating circadian circuits as being vulnerable to endocrine disruption. DNA methylation analysis of 2 genes, Per2 and Ar, showed no effect of EDCs and suggested alternative mechanisms for the altered mRNA levels. In the ARC, 12 genes were affected by treatment, mostly in males, again with dynamic developmental changes. Bionetwork analysis of relationships among genes, hormones, and physiological markers showed sexually dimorphic effects of estrogenic EDC exposures, with the female AVPV and the male ARC being most vulnerable, and provided novel relationships among hypothalamic genes and postnatal reproductive maturation.


Asunto(s)
Arocloros/toxicidad , Disruptores Endocrinos/toxicidad , Estradiol/análogos & derivados , Trastornos del Crecimiento/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/crecimiento & desarrollo , Análisis por Conglomerados , Metilación de ADN/efectos de los fármacos , Estradiol/toxicidad , Ciclo Estral/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Hormonas Esteroides Gonadales/sangre , Trastornos del Crecimiento/metabolismo , Masculino , Exposición Materna , Núcleos Talámicos de la Línea Media/efectos de los fármacos , Núcleos Talámicos de la Línea Media/crecimiento & desarrollo , Proteínas Circadianas Period/genética , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Regiones Promotoras Genéticas , Pubertad Tardía/inducido químicamente , Pubertad Tardía/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/genética , Caracteres Sexuales , Transcriptoma
15.
Digestion ; 85(4): 308-19, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22688404

RESUMEN

Growth retardation, associated with delayed puberty, is a frequent feature in pediatric patients with inflammatory bowel disease (IBD), especially with Crohn's disease. It is mainly induced by malnutrition and the effects of the inflammatory process on the growth hormone/insulin-like growth factor-1 axis or on the growth plate. Therefore, control of disease activity and mucosal healing are paramount to promote growth and adequate pubertal onset. Current therapeutic strategies for maintenance in IBD include anti-inflammatory drugs, immunosuppressives, and, more recently, biologic agents. Although these treatments are efficient in minimizing inflammation and inducing prolonged remission, their long-term effects on growth and final height remain controversial. Furthermore, glucocorticoid therapy, even though very efficient in inducing remission, clearly shows deleterious effects on growth, which is not the case for exclusive enteral nutrition showing comparable results regarding induction of remission. Thus regular assessment of weight, height and pubertal stage is essential in children and adolescents with chronic disease, namely IBD.


Asunto(s)
Glucocorticoides/efectos adversos , Trastornos del Crecimiento/etiología , Inmunosupresores/efectos adversos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Pubertad Tardía/etiología , Adolescente , Niño , Nutrición Enteral , Trastornos del Crecimiento/metabolismo , Hormona del Crecimiento/metabolismo , Humanos , Inflamación/complicaciones , Inflamación/etiología , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Desnutrición/complicaciones , Desnutrición/etiología , Desnutrición/metabolismo , Pubertad Tardía/metabolismo
16.
Environ Health Perspect ; 118(12): 1782-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20675266

RESUMEN

BACKGROUND: Lead (Pb) and cadmium (Cd) are known reproductive toxicants thought to disrupt hormone production throughout sensitive developmental windows, although this has not been previously examined in nationally representative peripubertal children. OBJECTIVES: We examined the association between blood Pb and urinary Cd concentrations and the reproductive hormones inhibin B and luteinizing hormone (LH) in girls 6-11 years of age who participated in the cross-sectional Third National Health and Nutrition Examination Survey (NHANES III) (1988-1994). METHODS: Pb (micrograms per deciliter) was measured in whole blood, and Cd was measured in urine (nanograms per milliliter). Inhibin B (picograms per milliliter) and LH (milli-International units per milliliter) were measured in residual sera for 705 girls. Survey logistic regression was used to estimate associations with pubertal onset based on inhibin B concentration > 35 pg/mL or LH concentration > 0.4 mIU/mL, and multinomial logistic regression was used to estimate the association between Pb and increasing categories of hormone concentrations. RESULTS: High Pb (≥ 5 µg/dL) was inversely associated with inhibin B > 35 pg/mL [odds ratio (OR) = 0.26; 95% confidence interval (CI), 0.11-0.60; compared with Pb < 1 µg/dL]. At 10 and 11 years of age, girls with low Pb (< 1 µg/dL) had significantly higher inhibin B than did girls with moderate (1-4.99 µg/dL) or high Pb (≥ 5 µg/dL). In the subsample of 260 girls with levels of inhibin B above the level of detection and using survey regression modeling, inhibin B levels were lower among girls with both high Pb and high Cd (ß= -0.52; 95% CI, -0.09 to -1.04) than among girls with high Pb alone (ß= -0.35; 95% CI, -0.13 to -0.57), relative to girls with low Pb and low Cd. CONCLUSIONS: Higher Pb was inversely associated with inhibin B, a marker of follicular development, and estimated effects suggestive of pubertal delays appeared to be stronger in the context of higher Cd concentrations. These data underscore the importance of Pb and Cd as reproductive toxicants for young girls.


Asunto(s)
Cadmio/orina , Contaminantes Ambientales/toxicidad , Inhibinas/sangre , Plomo/sangre , Hormona Luteinizante/sangre , Pubertad/efectos de los fármacos , Cadmio/toxicidad , Niño , Exposición a Riesgos Ambientales/análisis , Exposición a Riesgos Ambientales/estadística & datos numéricos , Contaminantes Ambientales/sangre , Contaminantes Ambientales/orina , Femenino , Humanos , Plomo/toxicidad , Encuestas Nutricionales , Pubertad Tardía/inducido químicamente , Pubertad Tardía/epidemiología , Pubertad Tardía/metabolismo , Estados Unidos
17.
J Pediatr ; 156(2): 308-12, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19892366

RESUMEN

OBJECTIVE: To test the hypothesis that prepubertal children with presumed constitutional delay of growth and development (CDGD) have enhanced insulin sensitivity and, therefore, insulin sensitivity is associated with later onset of puberty. STUDY DESIGN: Twenty-one prepubertal children with presumed CDGD and 23 prepubertal control children, underwent a frequently sampled intravenous glucose tolerance test to evaluate insulin sensitivity and other markers of insulin, glucose, and growth regulation. RESULTS: Children in the CDGD group were shorter and leaner than control subjects. Children with presumed CDGD were 40% more insulin sensitive (17.0 x 10(-4) min(-1)/[mU/L] versus 12.1 x 10(-4) min(-1)/[mU/L]; P = .0006) and had reduced acute insulin response, thus maintaining euglycemia (216 mU/L versus 330 mU/L; P = .02) compared with control subjects. In addition, the CDGD group had lower serum insulin-like growth factor binding protein 3 levels (3333 ng/mL versus 3775 ng/mL; P = .0004) and a trend toward lower serum insulin-like growth factor-II levels (794 ng/mL versus 911 ng/mL; P = .06). CONCLUSION: Prepubertal children with presumed CDGD have enhanced insulin sensitivity, supporting the hypothesis that insulin sensitivity is associated with timing of puberty. It may signify long-term biological advantages with lower risk of metabolic syndrome and malignancy.


Asunto(s)
Trastornos del Crecimiento/metabolismo , Insulina/metabolismo , Pubertad Tardía/metabolismo , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Humanos , Secreción de Insulina , Masculino , Análisis de Regresión
18.
Endocrinology ; 150(6): 2889-97, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19228890

RESUMEN

Spontaneously hypertensive (SH) rats, extensively used as experimental models of essential human hypertension, display important alterations in the neuroendocrine reproductive axis, which manifest as markedly delayed puberty onset in females but whose basis remains largely unknown. We analyze herein in female SH rats: 1) possible alterations in the expression and function of KiSS-1/GPR54 and GnRH/GnRH-receptor systems, 2) the integrity of feedback mechanisms governing the hypothalamic-pituitary-ovarian axis, and 3) the control of ovarian function by gonadotropins. Our data demonstrate that, despite overtly delayed puberty, no significant decrease in hypothalamic KiSS-1, GPR54, or GnRH mRNA levels was detected in this strain. Likewise, in vivo gonadotropin responses to ovariectomy and systemic kisspeptin-10 or GnRH administration, as well as in vitro gonadotropin responses to GnRH, were fully preserved in SH rats. Moreover, circulating LH levels were grossly conserved during prepubertal maturation, whereas FSH levels were even enhanced from d 20 postpartum onwards. In striking contrast, ovarian weight and hormone (progesterone and testosterone) responses to human chorionic gonadotropin (CG) in vitro were profoundly decreased in SH rats, with impaired follicular development and delayed ovulation at puberty. Such reduced hormonal responses to human CG could not be attributed to changes in LH/CG or FSH-receptor mRNA expression but might be linked to blunted P450scc, 3beta-hydroxy steroid dehydrogenase, and aromatase mRNA levels in ovaries from SH rats. In conclusion, our results indicate that the expression and function of KiSS-1/GPR54 and GnRH/GnRH-receptor systems is normal in SH rats, whereas ovarian development, steroidogenesis, and responsiveness to gonadotropins are strongly compromised.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hipertensión/fisiopatología , Hipotálamo/metabolismo , Insuficiencia Ovárica Primaria/fisiopatología , Proteínas/metabolismo , Pubertad Tardía/fisiopatología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Gonadotropina Coriónica/farmacología , Modelos Animales de Enfermedad , Femenino , Hormona Folículo Estimulante/sangre , Hormona Folículo Estimulante/farmacología , Hipertensión/metabolismo , Kisspeptinas , Hormona Luteinizante/sangre , Masculino , Ovariectomía , Ovario/efectos de los fármacos , Ovario/metabolismo , Ovario/fisiopatología , Insuficiencia Ovárica Primaria/metabolismo , Pubertad Tardía/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de Kisspeptina-1 , Transducción de Señal/fisiología
19.
Neurosignals ; 16(2-3): 165-82, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18253056

RESUMEN

Gonadotropin-releasing hormone (GnRH) and olfactory neurons migrate together in embryologic development, and disruption of this process causes idiopathic hypogonadotropic hypogonadism (IHH) with anosmia (Kallmann syndrome (KS)). Patients with IHH/KS generally manifest irreversible pubertal delay and subsequent infertility due to deficient pituitary gonadotropins or GnRH. The molecular basis of IHH/KS includes genes that: (1) regulate GnRH and olfactory neuron migration; (2) control the synthesis or secretion of GnRH; (3) disrupt GnRH action upon pituitary gonadotropes, or (4) interfere with pituitary gonadotropin synthesis or secretion. KS patients may also have midline facial defects indicating the diverse developmental functions of genes involved. Most causative genes cause either normosmic IHH or KS except FGFR1, which may cause either phenotype. Recently, several balanced chromosomal translocations have been identified in IHH/KS patients, which could lead to the identification of new disease-producing genes. Although there are two cases reported who have digenic disease, this awaits confirmation in future larger studies. The challenge will be to determine the importance of these genes in the 10-15% of couples with normal puberty who have infertility.


Asunto(s)
Hormona Liberadora de Gonadotropina/genética , Hipogonadismo/genética , Síndrome de Kallmann/genética , Neuronas/fisiología , Animales , Hormona Liberadora de Gonadotropina/biosíntesis , Humanos , Hipogonadismo/metabolismo , Hipogonadismo/fisiopatología , Síndrome de Kallmann/metabolismo , Síndrome de Kallmann/fisiopatología , Neuronas/citología , Pubertad Tardía/genética , Pubertad Tardía/metabolismo , Pubertad Tardía/fisiopatología
20.
Fertil Steril ; 90(5): 1875-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18191849

RESUMEN

OBJECTIVE: To analyze the age at menarche of girls with type 1 diabetes (T1D) who were diagnosed with the disease before puberty and compare it with that of an age-matched group of normal girls. Previous studies on the appearance of menarche showed that the mean age of onset of menarche is delayed in girls affected by T1D compared with normal girls. DESIGN: Case-control study. SETTING: Patients and controls in an academic research environment. PATIENT(S): We studied, retrospectively, the charts of 162 consecutive girls with T1D born in a geographically defined region between 1984 and 1994 with a mean disease duration of 3-5 years, all of whom were on intensive insulin therapy since diagnosis of T1D. The control group consisted of 214 normal girls born between 1984 and 1994, who agreed to fill in an anonymous questionnaire regarding age at menarche and other clinical information. INTERVENTION(S): There was no intervention per se in the study. Age at menarche appears as a dependent variable of body mass index (BMI), HbA1c, and so on. MAIN OUTCOME MEASURE(S): BMI, HbA1c, and duration of T1D at menarche were considered among the potential factors affecting the age of menarche. RESULT(S): Age at menarche in girls with T1D was significantly delayed compared with control girls (12.6 +/- 1.5 years vs. 12.25 +/- 1.4 years, respectively). HbA1c levels and BMI did not influence the age at menarche. CONCLUSION(S): Despite intensive insulin therapy and good metabolic control since diagnosis of T1D, the age at menarche is still delayed in girls who develop T1D before puberty.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Menarquia , Pubertad Tardía/etiología , Factores de Edad , Índice de Masa Corporal , Estudios de Casos y Controles , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Pubertad Tardía/metabolismo , Pubertad Tardía/fisiopatología , Estudios Retrospectivos , Ciudad de Roma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...