Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pediatr Nephrol ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38914781

RESUMEN

BACKGROUND: X-linked hypophosphatemia (XLH) is a rare inherited phosphate-wasting disorder associated with bone and dental complications. Health-related quality of life (HRQoL) is reduced in XLH patients on conventional treatment with phosphate supplements and active vitamin D, while information on patients treated with burosumab is rare. METHODS: HRQoL was assessed in 63 pediatric XLH patients participating in a prospective, observational study and patient registry in Germany using the KIDSCREEN-52 survey instrument and standardized qualitative interviews. RESULTS: The median age of the XLH patients was 13.2 years (interquartile range 10.6 - 14.6). At the time of the survey, 55 (87%) patients received burosumab and 8 (13%) conventional treatment. Forty-six patients (84%) currently being treated with burosumab previously received conventional treatment. Overall, HRQoL was average compared to German reference values (mean ± SD: self-report, 53.36 ± 6.47; caregivers' proxy, 51.33 ± 7.15) and even slightly above average in some dimensions, including physical, mental, and social well-being. In general, XLH patients rated their own HRQoL higher than their caregivers. In qualitative interviews, patients and caregivers reported that, compared with conventional therapy, treatment with burosumab reduced stress, bone pain, and fatigue, improved physical health, and increased social acceptance by peers and the school environment. CONCLUSIONS: In this real-world study in pediatric XLH patients, HRQoL was average or even slightly above that of the general population, likely due to the fact that the vast majority of patients had their treatment modality switched from conventional treatment to burosumab resulting in improved physical health and well-being.

2.
Hum Mol Genet ; 28(8): 1357-1368, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30608578

RESUMEN

The initiation of puberty is orchestrated by an augmentation of gonadotropin-releasing hormone (GnRH) secretion from a few thousand hypothalamic neurons. Recent findings have indicated that the neuroendocrine control of puberty may be regulated by a hierarchically organized network of transcriptional factors acting upstream of GnRH. These include enhanced at puberty 1 (EAP1), which contributes to the initiation of female puberty through transactivation of the GnRH promoter. However, no EAP1 mutations have been found in humans with disorders of pubertal timing. We performed whole-exome sequencing in 67 probands and 93 relatives from a large cohort of familial self-limited delayed puberty (DP). Variants were analyzed for rare, potentially pathogenic variants enriched in case versus controls and relevant to the biological control of puberty. We identified one in-frame deletion (Ala221del) and one rare missense variant (Asn770His) in EAP1 in two unrelated families; these variants were highly conserved and potentially pathogenic. Expression studies revealed Eap1 mRNA abundance in peri-pubertal mouse hypothalamus. EAP1 binding to the GnRH1 promoter increased in monkey hypothalamus at the onset of puberty as determined by chromatin immunoprecipitation. Using a luciferase reporter assay, EAP1 mutants showed a reduced ability to trans-activate the GnRH promoter compared to wild-type EAP1, due to reduced protein levels caused by the Ala221del mutation and subcellular mislocation caused by the Asn770His mutation, as revealed by western blot and immunofluorescence, respectively. In conclusion, we have identified the first EAP1 mutations leading to reduced GnRH transcriptional activity resulting in a phenotype of self-limited DP.


Asunto(s)
Hormona Liberadora de Gonadotropina/fisiología , Pubertad Tardía/genética , Securina/genética , Adolescente , Adulto , Animales , Niño , Femenino , Regulación de la Expresión Génica/genética , Hormona Liberadora de Gonadotropina/genética , Humanos , Hipotálamo/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Neuronas/metabolismo , Regiones Promotoras Genéticas/genética , Pubertad/genética , Pubertad/fisiología , ARN Mensajero/genética , Securina/fisiología , Maduración Sexual/genética , Transactivadores/genética , Factores de Transcripción/genética , Secuenciación del Exoma , Adulto Joven
3.
Glia ; 67(2): 309-320, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30485552

RESUMEN

Gonadotropin releasing hormone (GnRH)-secretion is not only regulated by neuronal factors but also by astroglia cells via growth factors and ErbB receptors of the epidermal growth factor family. Studies in transgenic mice carrying mutations in the ErbB receptor system experience impaired reproductive capacity. In addition, some of these animals show a typical skin phenotype with wavy hair and curly whiskers. The rat strain SPRD-CU3 (CU3), examined in this study, displays a similar skin phenotype and a significant impairment of the timing of puberty onset and reproductive performance, suggesting a disruption in the astrocytic to GnRH neuronal communication. To address this issue, we analyzed astrocytic prostaglandin E2 (PGE2 ) release from primary hypothalamic astrocytic cell cultures after stimulation with transforming growth factor α (TGFα), ligand for ErbB1/ErbB2, or Neuregulin 1 beta 2 (NRG1ß2 ), ligand for ErbB4/ErbB2 signaling pathway. Compared to cultures from wild type animals, astrocytic cultures from CU3 rats were unable to respond to NRG stimulation, suggesting a disruption of the ErbB4/ErbB2 signaling pathway. This is confirmed by mutational analysis of ErbB4 that revealed a single point mutation at 3125 bp resulting in an amino acid change from proline to glutamine located at the carboxy-terminal region. As a consequence, substantial conformational changes occur in the transmembrane and intracellular domain of the protein, affecting the ability to form a receptor dimer with a partner and the ability to function as a transcriptional regulator. Thus, astroglia to GnRH neuronal signaling via ErbB4 is essential of timely onset of puberty and reproductive function.


Asunto(s)
Astrocitos/efectos de los fármacos , Dinoprostona/metabolismo , Trastornos del Desarrollo Sexual/patología , Hormona Liberadora de Gonadotropina/metabolismo , Neurregulinas/farmacología , Neuronas/metabolismo , Receptor ErbB-4/genética , Animales , Astrocitos/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Trastornos del Desarrollo Sexual/tratamiento farmacológico , Trastornos del Desarrollo Sexual/genética , Trastornos del Desarrollo Sexual/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/efectos de la radiación , Hipotálamo/citología , Modelos Moleculares , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Mutación Puntual/genética , Ratas , Ratas Transgénicas , Receptor ErbB-4/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo
4.
Dev Med Child Neurol ; 59(8): 829-836, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28485516

RESUMEN

AIM: To assess the psychological well-being and social integration of adults with craniopharyngioma diagnosed in childhood. METHOD: A cross-sectional study of a nationwide cohort of young adults with craniopharyngioma in Germany was performed. A structured questionnaire covered the sociodemographic, clinical data, and subjective effects of the condition on social integration. Psychological well-being was assessed using the Hospital Anxiety and Depression Scale (HADS). Results were compared to young adults with type 1 diabetes mellitus (T1DM). RESULTS: The study included 59 participants (29 females, 30 males; mean age 25y 2mo [SD 5y 10mo]), mean age at first surgery 10y 2mo [SD 3y 7mo]. Compared to the T1DM group, significantly more young people with craniopharyngioma aged 25 to 35 years lived at their parents' homes (craniopharyngioma 43.34%; T1DM 13.7%; χ2 =4.14, p=0.049), and fewer lived in a relationship (craniopharyngioma 8.69%; T1DM 54.7%; χ2 =15.74, p<0.001). The HADS revealed a score for depression above the cut-off in 20.69 per cent of young adults with craniopharyngioma and in 6 per cent of young adults with T1DM (χ2 =13.42, p<0.001). INTERPRETATION: Young adults with craniopharyngioma reported subjective disadvantages in professional and social integration. Further, they presented with reduced well-being and increased depression rates. Better psychosocial support and self-management education might reduce the long-term burden of the disease.


Asunto(s)
Craneofaringioma/psicología , Depresión/psicología , Diabetes Mellitus Tipo 1/psicología , Empleo/psicología , Vida Independiente/psicología , Relaciones Interpersonales , Satisfacción Personal , Neoplasias Hipofisarias/psicología , Participación Social/psicología , Adulto , Edad de Inicio , Estudios Transversales , Depresión/etiología , Humanos , Adulto Joven
5.
Endocr Rev ; 45(4): 460-492, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38436980

RESUMEN

There are 3 physiological waves of central hypothalamic-pituitary-gonadal (HPG) axis activity over the lifetime. The first occurs during fetal life, the second-termed "mini-puberty"-in the first months after birth, and the third at puberty. After adolescence, the axis remains active all through adulthood. Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disorder characterized by a deficiency in hypothalamic gonadotropin-releasing hormone (GnRH) secretion or action. In cases of severe CHH, all 3 waves of GnRH pulsatility are absent. The absence of fetal HPG axis activation manifests in around 50% of male newborns with micropenis and/or undescended testes (cryptorchidism). In these boys, the lack of the mini-puberty phase accentuates testicular immaturity. This is characterized by a low number of Sertoli cells, which are important for future reproductive capacity. Thus, absent mini-puberty will have detrimental effects on later fertility in these males. The diagnosis of CHH is often missed in infants, and even if recognized, there is no consensus on optimal therapeutic management. Here we review physiological mini-puberty and consequences of central HPG axis disorders; provide a diagnostic approach to allow for early identification of these conditions; and review current treatment options for replacement of mini-puberty in male infants with CHH. There is evidence from small case series that replacement with gonadotropins to mimic "mini-puberty" in males could have beneficial outcomes not only regarding testis descent, but also normalization of testis and penile sizes. Moreover, such therapeutic replacement regimens in disordered mini-puberty could address both reproductive and nonreproductive implications.


Asunto(s)
Hipogonadismo , Humanos , Masculino , Hipogonadismo/terapia , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/fisiopatología , Sistema Hipotálamo-Hipofisario , Pubertad/fisiología , Terapia de Reemplazo de Hormonas , Hormona Liberadora de Gonadotropina/metabolismo
6.
Eur J Endocrinol ; 190(5): 401-408, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38652605

RESUMEN

CONTEXT: Reliable estradiol (E2) reference intervals (RIs) are crucial in pediatric endocrinology. OBJECTIVES: This study aims to develop a sensitive ultra-performance liquid chromatographic tandem mass spectrometry (UPLC-MS/MS) method for E2 in serum, to establish graphically represented RI percentiles and annual RIs for both sexes, and to perform a systematic literature comparison. METHODS: First, a UPLC-MS/MS method for E2 was developed. Second, graphically represented RI percentiles and annual RIs covering 0-18 years were computed (cohort of healthy children [1181 girls and 543 boys]). Subsequently, RIs were compared with published data by systematic searches. RESULTS: Lower limit of quantification was 11 pmol/L, indicating high sensitivity. Estradiol first peaked during mini-puberty in both sexes (girls up to 192 pmol/L; boys up to 225 pmol/L). As could be expected, girls showed higher pubertal E2 (up to 638 pmol/L). However, boys' RIs (up to 259 pmol/L) overlapped considerably. We found 4 studies in the literature that also used LC-MS/MS to determine E2 and published RIs for the complete pediatric age range. Reference intervals varied considerably. Pre-pubertal and pubertal phases were present in all studies. Higher E2 during the time of mini-puberty in both sexes was documented in 3 studies including ours. CONCLUSIONS: Variability of RIs for E2 between studies illustrates the importance of laboratory-specific RIs despite using a LC-MS/MS reference method. In boys, the striking E2 peak during mini-puberty as well as high pubertal E2 without phenotypic estrogenization in regular male puberty indicates that the role of E2 in children and, especially in boys, requires better functional understanding.


Asunto(s)
Estradiol , Pubertad , Espectrometría de Masas en Tándem , Humanos , Masculino , Espectrometría de Masas en Tándem/métodos , Niño , Estradiol/sangre , Femenino , Valores de Referencia , Preescolar , Adolescente , Lactante , Cromatografía Liquida/métodos , Cromatografía Liquida/normas , Pubertad/sangre , Pubertad/fisiología , Recién Nacido , Maduración Sexual/fisiología
7.
J Clin Endocrinol Metab ; 108(10): e998-e1006, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-37097907

RESUMEN

CONTEXT: Burosumab has been approved for the treatment of children and adults with X-linked hypophosphatemia (XLH). Real-world data and evidence for its efficacy in adolescents are lacking. OBJECTIVE: To assess the effects of 12 months of burosumab treatment on mineral metabolism in children (aged <12 years) and adolescents (aged 12-18 years) with XLH. DESIGN: Prospective national registry. SETTING: Hospital clinics. PATIENTS: A total of 93 patients with XLH (65 children, 28 adolescents). MAIN OUTCOME MEASURES: Z scores for serum phosphate, alkaline phosphatase (ALP), and renal tubular reabsorption of phosphate per glomerular filtration rate (TmP/GFR) at 12 months. RESULTS: At baseline, patients showed hypophosphatemia (-4.4 SD), reduced TmP/GFR (-6.5 SD), and elevated ALP (2.7 SD, each P < .001 vs healthy children) irrespective of age, suggesting active rickets despite prior therapy with oral phosphate and active vitamin D in 88% of patients. Burosumab treatment resulted in comparable increases in serum phosphate and TmP/GFR in children and adolescents with XLH and a steady decline in serum ALP (each P < .001 vs baseline). At 12 months, serum phosphate, TmP/GFR, and ALP levels were within the age-related normal range in approximately 42%, 27%, and 80% of patients in both groups, respectively, with a lower, weight-based final burosumab dose in adolescents compared with children (0.72 vs 1.06 mg/kg, P < .01). CONCLUSIONS: In this real-world setting, 12 months of burosumab treatment was equally effective in normalizing serum ALP in adolescents and children, despite persistent mild hypophosphatemia in one-half of patients, suggesting that complete normalization of serum phosphate is not mandatory for substantial improvement of rickets in these patients. Adolescents appear to require lower weight-based burosumab dosage than children.


Asunto(s)
Raquitismo Hipofosfatémico Familiar , Hipofosfatemia , Adulto , Humanos , Niño , Adolescente , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/efectos adversos , Estudios Prospectivos , Fosfatos , Factores de Crecimiento de Fibroblastos , Minerales
8.
Horm Res Paediatr ; 95(2): 137-148, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34689140

RESUMEN

INTRODUCTION: Genetic obesity is rare and quite challenging for pediatricians in terms of early identification. Src-homology-2 (SH2) B adapter protein 1 (SH2B1) is an important component in the leptin-melanocortin pathway and is found to play an important role in leptin and insulin signaling and therefore in the pathogenesis of obesity and diabetes. Microdeletions in chromosome 16p11.2, encompassing the SH2B1 gene, are known to be associated with obesity, insulin resistance, hyperphagia, and developmental delay. The aim of our study is to report on a case series of young individuals with 16p11.2 microdeletions, including the SH2B1 gene, and provide detailed information on body mass index (BMI) development and obesity-associated comorbidities. In this way, we want to raise awareness of this syndromic form of obesity as a differential diagnosis of genetic obesity. METHODS: We describe the phenotype of 7 children (3 male; age range: 2.8-18.0 years) with 16p11.2 microdeletions, encompassing the SH2B1 gene, and present their BMI trajectories from birth onward. Screening for obesity-associated comorbidities was performed at the time of genetic diagnosis. RESULTS: All children presented with severe, early-onset obesity already at the age of 5 years combined with variable developmental delay. Five patients presented with elevated fasting insulin levels, 1 patient developed diabetes mellitus type 2, 4 patients had dyslipidemia, and 4 developed nonalcoholic fatty-liver disease. DISCUSSION/CONCLUSION: Chromosomal microdeletions in 16p11.2, including the SH2B1 gene, in children are associated with severe, early-onset obesity and comorbidities associated with insulin resistance. Early genetic testing in suspicious patients and early screening for comorbidities are recommended.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Obesidad Infantil , Proteínas Adaptadoras Transductoras de Señales/genética , Adolescente , Índice de Masa Corporal , Niño , Preescolar , Femenino , Eliminación de Gen , Humanos , Insulina/metabolismo , Resistencia a la Insulina/genética , Leptina/metabolismo , Masculino , Obesidad Infantil/diagnóstico , Obesidad Infantil/genética
9.
Best Pract Res Clin Endocrinol Metab ; 35(5): 101579, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34563408

RESUMEN

Sexual maturation in humans is characterized by a unique individual variability. Pubertal onset is a highly heritable polygenic trait but it is also affected by environmental factors such as obesity or endocrine disrupting chemicals. The last 30 years have been marked by a constant secular trend toward earlier age at onset of puberty in girls and boys around the world. More recent data, although more disputed, suggest an increased incidence in idiopathic central precocious puberty. Such trends point to a role for environmental factors in pubertal changes. Animal data suggest that the GnRH-neuronal network is highly sensitive to endocrine disruption during development. This review focuses on the most recent data regarding secular trend in pubertal timing as well as potential new epigenetic mechanisms explaining the developmental and transgenerational effects of endocrine disrupting chemicals on pubertal timing.


Asunto(s)
Disruptores Endocrinos , Pubertad Precoz , Animales , Disruptores Endocrinos/toxicidad , Femenino , Humanos , Masculino , Obesidad/inducido químicamente , Obesidad/epidemiología , Obesidad/genética , Pubertad , Pubertad Precoz/inducido químicamente , Pubertad Precoz/epidemiología , Pubertad Precoz/genética , Maduración Sexual
10.
J Clin Invest ; 117(8): 2145-54, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17627301

RESUMEN

The initiation of mammalian puberty and the maintenance of female reproductive cycles are events controlled by hypothalamic neurons that secrete the decapeptide gonadotropin-releasing hormone (GnRH). GnRH secretion is, in turn, controlled by changes in neuronal and glial inputs to GnRH-producing neurons. The hierarchical control of the process is unknown, but it requires coordinated regulation of these cell-cell interactions. Here we report the functional characterization of a gene (termed enhanced at puberty 1 [EAP1]) that appears to act as an upstream transcriptional regulator of neuronal networks controlling female reproductive function. EAP1 expression increased selectively at puberty in both the nonhuman primate and rodent hypothalamus. EAP1 encoded a nuclear protein expressed in neurons involved in the inhibitory and facilitatory control of reproduction. EAP1 transactivated genes required for reproductive function, such as GNRH1, and repressed inhibitory genes, such as preproenkephalin. It contained a RING finger domain of the C3HC4 subclass required for this dual transcriptional activity. Inhibition of EAP1 expression, targeted to the rodent hypothalamus via lentivirus-mediated delivery of EAP1 siRNAs, delayed puberty, disrupted estrous cyclicity, and resulted in ovarian abnormalities. These results suggest that EAP1 is a transcriptional regulator that, acting within the neuroendocrine brain, contributes to controlling female reproductive function.


Asunto(s)
Ciclo Estral/metabolismo , Hormona Liberadora de Gonadotropina/biosíntesis , Sistema Hipotálamo-Hipofisario/metabolismo , Proteínas de Neoplasias/biosíntesis , Neuronas/metabolismo , Precursores de Proteínas/biosíntesis , Maduración Sexual , Factores de Transcripción/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Regulación hacia Abajo/genética , Ciclo Estral/genética , Femenino , Hormona Liberadora de Gonadotropina/genética , Humanos , Sistema Hipotálamo-Hipofisario/citología , Lentivirus , Macaca mulatta , Proteínas de Neoplasias/genética , Neuroglía/citología , Neuroglía/patología , Neuronas/citología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ovario/citología , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Precursores de Proteínas/genética , Estructura Terciaria de Proteína/genética , Ratas , Ratas Sprague-Dawley , Securina , Maduración Sexual/genética , Factores de Transcripción/genética , Transducción Genética
11.
Horm Res ; 69(2): 114-23, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18059092

RESUMEN

BACKGROUND: Hypothalamic hamartomas (HHs) are congenital lesions composed of neurons and astroglia. Frequently, HHs cause central precocious puberty (CPP) and/or gelastic seizures. Because HHs might express genes similar to those required for the initiation of normal puberty, we used cDNA arrays to compare the gene expression profile of an HH associated with CPP with three HHs not accompanied by sexual precocity. METHODS: Global changes in gene expression were detected using Affymetrix arrays. The results were confirmed by semiquantitative PCR, which also served to examine the expression of selected genes in the hypothalamus of female monkeys undergoing puberty. RESULTS: All HHs were associated with seizures. Ten genes whose expression was increased in the HH with CPP were identified. They encode proteins involved in three key cellular processes: transcriptional regulation, cell-cell signaling, and cell adhesiveness. They include IA-1 and MEF2A, two transcription factors required for neuronal development; mGluR1 and VILIP-1, which encode proteins involved in neuronal communication, and TSG-6 that encodes a protein involved in cell adhesiveness. Of these, expression of mGluR1 also increases in the female monkey hypothalamus at puberty. CONCLUSIONS: Increased expression of these genes in HHs may be relevant to the ability of some HHs to induce sexual precocity.


Asunto(s)
Perfilación de la Expresión Génica , Hamartoma/genética , Enfermedades Hipotalámicas/genética , Pubertad Precoz/genética , Adolescente , Adulto , Animales , Moléculas de Adhesión Celular/genética , Niño , Preescolar , Proteínas de Unión al ADN/genética , Femenino , Hormona Liberadora de Gonadotropina/genética , Hamartoma/complicaciones , Humanos , Enfermedades Hipotalámicas/complicaciones , Kisspeptinas , Macaca mulatta , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Pubertad Precoz/etiología , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Receptores de Glutamato Metabotrópico/genética , Proteínas Represoras/genética , Maduración Sexual/genética , Factor de Crecimiento Transformador alfa/genética , Proteínas Supresoras de Tumor/genética
12.
J Pediatr Endocrinol Metab ; 21(9): 865-77, 2008 09.
Artículo en Inglés | MEDLINE | ID: mdl-18924580

RESUMEN

It is controversial whether obesity in children is associated with earlier onset of puberty and advanced appearance of distinct parameters of pubertal development. To investigate the impact of obesity on markers of the onset and parameters representing stages of puberty, we analyzed auxological parameters and secondary sex characteristics in three representative cohorts of Caucasian children. Body weight, height, peak height velocity and pubertal stages were evaluated in two recent German cohorts (CrescNet and Leipzig Schoolchildren), and a historical Swiss cohort. According to body mass index (BMI), children were classified into three weight groups of lean, overweight, or normal weight with limits defined below -1.28 and above +1.28 BMI SDS. Peak height velocity (PHV) occurred significantly later in lean compared to normal weight children in the CrescNet and Swiss cohort, while there was no difference between obese and normal weight children. There was a trend towards acceleration of parameters of puberty onset and progression in obese children in all three cohorts. Height SDS was significantly higher in obese children compared to normal weight peers, but after completion of pubertal development it was similar in adolescents. The impact of overweight on the acceleration of puberty seems to be slightly stronger in boys. Once girls have reached a critical weight for entering the process of maturation, further increase in body weight does not seem to advance the onset of puberty.


Asunto(s)
Sobrepeso/fisiopatología , Pubertad/fisiología , Delgadez/fisiopatología , Adolescente , Estatura/fisiología , Índice de Masa Corporal , Peso Corporal/fisiología , Niño , Estudios de Cohortes , Estudios Transversales , Femenino , Alemania , Humanos , Estudios Longitudinales , Masculino , Menarquia/fisiología , Obesidad/fisiopatología , Suiza , Factores de Tiempo
13.
PLoS One ; 13(10): e0205318, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30300409

RESUMEN

BACKGROUND: A previous single-center study established a mathematical model for predicting the adult height (AH) in girls with idiopathic central precocious puberty (CPP). OBJECTIVE: To perform internal and external validations by comparing the actual AH to the calculated AH established by this model and to update it. METHODS: The original formula, calculated AH (cm) = 2.21 (height at initial evaluation, SD) + 2.32 (target height, SD) - 1.83 (luteinizing hormone/follicle-stimulating hormone peaks ratio) + 159.68, was established in a sample of 134 girls (group 4) and was applied to additional girls with CPP seen in the same center (group 1, n = 35), in Germany (group 2, n = 43) and in the Netherlands (group 3, n = 72). This formula has been updated based on these extended data, and both versions are available at the following location: http://www.kamick.org/lemaire/med/girls-cpp15.html. RESULTS: Despite the differences among the 4 groups in terms of their characteristics at the initial evaluation and the percentages of patients treated with the gonadotropin-releasing hormone analogue, they have similar calculated and actual AHs. The actual AHs are 162.2±7.0, 163.0±7.6, 162.4±7.7 and 162.1±5.6 cm in groups 1 to 4, respectively. They are highly correlated with the AHs calculated by the formula established in the original group (group 4), with R at 0.84, 0.67 and 0.69 in groups 1 to 3, respectively. When the actual AHs and the AHs predicted by the Bayley and Pinneau method are compared, the R is 0.76, 0.51 and 0.64 in groups 1 to 3, respectively. The absolute differences between actual AHs and the calculated AHs are greater than 1 SD (5.6 cm) in 15%, 35% and 28% of the patients in groups 1 to 3, respectively. CONCLUSION: This study validates and updates the previously established formula for predicting AH in girls with CPP. This updated formula can help clinicians to make treatment decisions.


Asunto(s)
Estatura/fisiología , Modelos Teóricos , Pubertad Precoz/epidemiología , Pubertad Precoz/fisiopatología , Adulto , Niño , Femenino , Hormona Folículo Estimulante/sangre , Alemania/epidemiología , Hormona Liberadora de Gonadotropina/sangre , Humanos , Hormona Luteinizante/sangre , Países Bajos/epidemiología , Pubertad Precoz/sangre
14.
Horm Res Paediatr ; 89(4): 255-264, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29694956

RESUMEN

BACKGROUND/AIMS: The high complexity of pediatric reference ranges across age, sex, and units impairs clinical application and comparability of steroid hormone data, e.g., in congenital adrenal hyperplasia (CAH). We developed a multiples-of-median (MoM) normalization tool to overcome this major drawback in pediatric endocrinology. METHODS: Liquid chromatography tandem mass spectrometry data comprising 10 steroid hormones representing 905 controls (555 males, 350 females, 0 to > 16 years) from 2 previous datasets were MoM transformed across age and sex. Twenty-three genetically proven CAH patients were included (21-hydroxylase deficiency [21OHD], n = 19; 11ß-hydroxylase deficiency [11OHD], n = 4). MoM cutoffs for single steroids predicting 21OHD and 11OHD were computed and validated through new, independent patients (21OHD, n = 8; adrenal cortical carcinoma, n = 6; obesity, n = 40). RESULTS: 21OHD and 11OHD patients showed disease-typical, easily recognizable MoM patterns independent of age, sex, and concentration units. Two single-steroid cutoffs indicated 21OHD: 3.87 MoM for 17-hydroxyprogesterone (100% sensitivity and 98.83% specificity) and 12.28 MoM for 21-deoxycortisol (94.74% sensitivity and 100% specificity). A cutoff of 13.18 MoM for 11-deoxycortisol indicated 11OHD (100% sensitivity and 100% specificity). CONCLUSIONS: Age- and sex-independent MoMs are straightforward for a clinically relevant display of multi-steroid patterns. In addition, defined single-steroid MoMs can serve alone as predictors of 21OHD and 11OHD. Finally, MoM transformation offers substantial enhancement of routine and scientific steroid hormone data exchange due to improved comparability.


Asunto(s)
17-alfa-Hidroxiprogesterona/sangre , Neoplasias de la Corteza Suprarrenal/sangre , Hiperplasia Suprarrenal Congénita/sangre , Carcinoma Corticosuprarrenal/sangre , Cortodoxona/sangre , Obesidad/sangre , Adolescente , Factores de Edad , Niño , Preescolar , Cromatografía Liquida , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Espectrometría de Masas , Factores Sexuales
15.
J Neurosci ; 26(51): 13167-79, 2006 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-17182767

RESUMEN

Thyroid transcription factor 1 (TTF1) [also known as Nkx2.1 (related to the NK-2 class of homeobox genes) and T/ebp (thyroid-specific enhancer-binding protein)], a homeodomain gene required for basal forebrain morphogenesis, remains expressed in the hypothalamus after birth, suggesting a role in neuroendocrine function. Here, we show an involvement of TTF1 in the control of mammalian puberty and adult reproductive function. Gene expression profiling of the nonhuman primate hypothalamus revealed that TTF1 expression increases at puberty. Mice in which the Ttf1 gene was ablated from differentiated neurons grew normally and had normal basal ganglia/hypothalamic morphology but exhibited delayed puberty, reduced reproductive capacity, and a short reproductive span. These defects were associated with reduced hypothalamic expression of genes required for sexual development and deregulation of a gene involved in restraining puberty. No extrapyramidal impairments associated with basal ganglia dysfunction were apparent. Thus, although TTF1 appears to fulfill only a morphogenic function in the ventral telencephalon, once this function is satisfied in the hypothalamus, TTF1 remains active as part of the transcriptional machinery controlling female sexual development.


Asunto(s)
Ganglios Basales/fisiología , Diferenciación Celular/genética , Eliminación de Gen , Neuronas/citología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Reproducción/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Animales , Ganglios Basales/citología , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Hipotálamo/citología , Hipotálamo/fisiología , Macaca mulatta , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuronas/fisiología , Proteínas Nucleares/deficiencia , Conducta Sexual Animal/fisiología , Factor Nuclear Tiroideo 1 , Factores de Transcripción/deficiencia
16.
Endocrinology ; 148(11): 5147-61, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17615149

RESUMEN

Much has been learned in recent years about the central mechanisms controlling the initiation of mammalian puberty. It is now clear that this process requires the interactive participation of several genes. Using a combination of high throughput, molecular, and bioinformatics strategies, in combination with a system biology approach, we singled out from the hypothalamus of nonhuman primates and rats a group of related genes whose expression increases at the time of female puberty. Although these genes [henceforth termed tumor-related genes (TRGs)] have diverse cellular functions, they share the common feature of having been earlier identified as involved in tumor suppression/tumor formation. A prominent member of this group is KiSS1, a gene recently shown to be essential for the occurrence of puberty. Cis-regulatory analysis revealed the presence of a hierarchically arranged gene set containing five major hubs (CDP/CUTL1, MAF, p53, YY1, and USF2) controlling the network at the transcriptional level. In turn, these hubs are heavily connected to non-TRGs involved in the transcriptional regulation of the pubertal process. TRGs may be expressed in the mammalian hypothalamus as components of a regulatory gene network that facilitates and integrates cellular and cell-cell communication programs required for the acquisition of female reproductive competence.


Asunto(s)
Redes Reguladoras de Genes , Genes Relacionados con las Neoplasias , Hipotálamo/metabolismo , Maduración Sexual/genética , Regulación hacia Arriba , Región de Flanqueo 5' , Animales , Sitios de Unión , Femenino , Regulación del Desarrollo de la Expresión Génica , Macaca mulatta , Modelos Biológicos , Sistemas Neurosecretores/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/metabolismo
17.
Endocrinology ; 147(3): 1166-74, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16373420

RESUMEN

The initiation of mammalian puberty requires an increase in pulsatile release of GnRH from the hypothalamus. This increase is brought about by coordinated changes in transsynaptic and glial-neuronal communication. As the neuronal and glial excitatory inputs to the GnRH neuronal network increase, the transsynaptic inhibitory tone decreases, leading to the pubertal activation of GnRH secretion. The excitatory neuronal systems most prevalently involved in this process use glutamate and the peptide kisspeptin for neurotransmission/neuromodulation, whereas the most important inhibitory inputs are provided by gamma-aminobutyric acid (GABA)ergic and opiatergic neurons. Glial cells, on the other hand, facilitate GnRH secretion via growth factor-dependent cell-cell signaling. Coordination of this regulatory neuronal-glial network may require a hierarchical arrangement. One level of coordination appears to be provided by a host of unrelated genes encoding proteins required for cell-cell communication. A second, but overlapping, level might be provided by a second tier of genes engaged in specific cell functions required for productive cell-cell interaction. A third and higher level of control involves the transcriptional regulation of these subordinate genes by a handful of upper echelon genes that, operating within the different neuronal and glial subsets required for the initiation of the pubertal process, sustain the functional integration of the network. The existence of functionally connected genes controlling the pubertal process is consistent with the concept that puberty is under genetic control and that the genetic underpinnings of both normal and deranged puberty are polygenic rather than specified by a single gene. The availability of improved high-throughput techniques and computational methods for global analysis of mRNAs and proteins will allow us to not only initiate the systematic identification of the different components of this neuroendocrine network but also to define their functional interactions.


Asunto(s)
Sistema Endocrino/fisiología , Neuronas/metabolismo , Sistemas Neurosecretores , Pubertad , Biología de Sistemas/métodos , Animales , Comunicación Celular , ADN/metabolismo , Genes Supresores de Tumor , Hormona Liberadora de Gonadotropina/metabolismo , Sustancias de Crecimiento/metabolismo , Humanos , Hipotálamo/metabolismo , Kisspeptinas , Modelos Biológicos , Neuroglía/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas , ARN Mensajero/metabolismo , Transducción de Señal , Programas Informáticos , Sinapsis , Transcripción Genética , Proteínas Supresoras de Tumor
18.
Mol Cell Endocrinol ; 254-255: 217-20, 2006 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-16757104

RESUMEN

Depot gonadotropin releasing hormone (GnRH) agonist (GnRHa) therapy is the treatment of choice for patients with central precocious puberty (CPP). It is still unclear whether long-term exposure to GnRHa is associated with impaired reproductive function in adulthood. The present study was performed on 46 women, former CPP patients, 12.5+/-3.7 years after the discontinuation of treatment with depot GnRHa. In a structured interview, we assessed general health status, clinical signs possibly associated with hyperandrogenism, menstrual cycle, gynaecological diseases and reproductive function. It appears that long-term treatment with depot GnRHa is safe and does not impair reproductive function. The risk of former CPP patients to develop hirsutism and/or polycystic ovary syndrome does not seem to be increased compared to the normal population but this issue needs to be addressed in further long-term follow-up studies.


Asunto(s)
Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/uso terapéutico , Pubertad Precoz/tratamiento farmacológico , Reproducción/efectos de los fármacos , Adulto , Andrógenos/efectos adversos , Estatura/efectos de los fármacos , Índice de Masa Corporal , Peso Corporal/efectos de los fármacos , Preparaciones de Acción Retardada/administración & dosificación , Vías de Administración de Medicamentos , Femenino , Fertilidad/efectos de los fármacos , Estudios de Seguimiento , Enfermedades de los Genitales Femeninos/etiología , Hormona Liberadora de Gonadotropina/administración & dosificación , Estado de Salud , Humanos , Hiperandrogenismo/diagnóstico , Entrevistas como Asunto , Cuidados a Largo Plazo , Ciclo Menstrual/efectos de los fármacos , Pubertad Precoz/complicaciones , Pamoato de Triptorelina/uso terapéutico
19.
Brain Dev ; 28(5): 300-4, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16478652

RESUMEN

Satoyoshi syndrome is a rare multisystemic disorder with assumed autoimmune pathogenesis. Typical clinical features are progressive painful muscle spasms, alopecia, diarrhoea, and skeletal and endocrine abnormalities often resulting in early invalidism and death. Patients have been treated with immunoglobulins and glucocorticoids with varying outcome. We report on a 19-year-old German adolescent who has been successfully treated with a new combination of carbamazepine to reduce the severity and frequency of painful nocturnal muscle spasms, prednisolone, methotrexate and sex-steroids. Prednisolone treatment alone was not successful. After introduction of low-dose of methotrexate to the therapy the patient recovered from muscle spasms, alopecia and diarrhoea. Initiation of sex-steroid treatment resulted in pubertal development, regular menstrual cycles and improved quality of life.


Asunto(s)
Enfermedades del Sistema Endocrino/complicaciones , Enfermedades del Sistema Endocrino/tratamiento farmacológico , Espasmo/complicaciones , Espasmo/tratamiento farmacológico , Adulto , Enfermedades del Desarrollo Óseo/complicaciones , Enfermedades del Desarrollo Óseo/tratamiento farmacológico , Carbamazepina/uso terapéutico , Quimioterapia Combinada , Femenino , Glucocorticoides/uso terapéutico , Humanos , Inmunosupresores/uso terapéutico , Prednisolona/uso terapéutico , Esteroides/uso terapéutico
20.
J Neurosci ; 23(1): 230-9, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12514220

RESUMEN

The initiation of mammalian puberty requires the activation of hypothalamic neurons secreting the neuropeptide luteinizing hormone-releasing hormone (LHRH). It is thought that this activation is caused by changes in trans-synaptic input to LHRH neurons. More recently, it has been postulated that the pubertal increase in LHRH secretion in female animals also requires neuron-glia signaling mediated by growth factors of the epidermal growth factor (EGF) family and their astrocytic erbB receptors. Although it appears clear that functional astrocytic erbB1 receptors are necessary for the timely advent of puberty, the physiological contribution that erbB4 receptors may make to this process has not been established. To address this issue, we generated transgenic mice expressing a dominant-negative erbB4 receptor (DN-erbB4) under the control of the GFAP promoter, which targets transgene expression to astrocytes. DN-erbB4 expression is most abundant in hypothalamic astrocytes, where it blocks the ligand-dependent activation of glial erbB4 and erbB2 receptors, without affecting erbB1 (EGF) receptor signaling. Mice carrying the transgene exhibit delayed sexual maturation and a diminished reproductive capacity in early adulthood. These abnormalities are related to a deficiency in pituitary gonadotropin hormone secretion, caused by impaired release of LHRH, the hypothalamic neuropeptide that controls sexual development. In turn, the reduction in LHRH release is caused by the inability of hypothalamic astrocytes to respond to neuregulin (NRG) with production of prostaglandin E(2), which in wild-type animals mediates the stimulatory effect of astroglial erbB receptor activation on neuronal LHRH release. Thus, neuron-astroglia communication via NRG-erbB4/2 receptor signaling appears to be essential for the timely unfolding of the developmental program by which the brain controls mammalian sexual maturation.


Asunto(s)
Astrocitos/fisiología , Receptores ErbB/fisiología , Hipotálamo/citología , Neurregulinas/fisiología , Maduración Sexual , Animales , Astrocitos/metabolismo , Células Cultivadas , Dinoprostona/metabolismo , Receptores ErbB/genética , Femenino , Fertilidad , Hormona Folículo Estimulante/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/fisiología , Cinética , Ratones , Ratones Transgénicos , Mutación , Neurregulinas/farmacología , Regiones Promotoras Genéticas , Receptor ErbB-2/metabolismo , Receptor ErbB-4 , Caracteres Sexuales , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA