Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 12(9): e1001952, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25248098

RESUMEN

Characterization of the genetic defects causing gonadotropic deficiency has made a major contribution to elucidation of the fundamental role of Kisspeptins and Neurokinin B in puberty onset and reproduction. The absence of puberty may also reveal neurodevelopmental disorders caused by molecular defects in various cellular pathways. Investigations of these neurodevelopmental disorders may provide information about the neuronal processes controlling puberty onset and reproductive capacity. We describe here a new syndrome observed in three brothers, which involves gonadotropic axis deficiency, central hypothyroidism, peripheral demyelinating sensorimotor polyneuropathy, mental retardation, and profound hypoglycemia, progressing to nonautoimmune insulin-dependent diabetes mellitus. High-throughput sequencing revealed a homozygous in-frame deletion of 15 nucleotides in DMXL2 in all three affected patients. This homozygous deletion was associated with lower DMXL2 mRNA levels in the blood lymphocytes of the patients. DMXL2 encodes the synaptic protein rabconnectin-3α, which has been identified as a putative scaffold protein for Rab3-GAP and Rab3-GEP, two regulators of the GTPase Rab3a. We found that rabconnectin-3α was expressed in exocytosis vesicles in gonadotropin-releasing hormone (GnRH) axonal extremities in the median eminence of the hypothalamus. It was also specifically expressed in cells expressing luteinizing hormone (LH) and follicle-stimulating hormone (FSH) within the pituitary. The conditional heterozygous deletion of Dmxl2 from mouse neurons delayed puberty and resulted in very low fertility. This reproductive phenotype was associated with a lower number of GnRH neurons in the hypothalamus of adult mice. Finally, Dmxl2 knockdown in an insulin-secreting cell line showed that rabconnectin-3α controlled the constitutive and glucose-induced secretion of insulin. In conclusion, this study shows that low levels of DMXL2 expression cause a complex neurological phenotype, with abnormal glucose metabolism and gonadotropic axis deficiency due to a loss of GnRH neurons. Our findings identify rabconectin-3α as a key controller of neuronal and endocrine homeostatic processes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Diabetes Mellitus Tipo 1/genética , Hipoglucemia/genética , Hipotiroidismo/genética , Infertilidad Masculina/genética , Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/genética , Polineuropatías/genética , Eliminación de Secuencia , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Adolescente , Animales , Secuencia de Bases , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Hormona Folículo Estimulante/genética , Hormona Folículo Estimulante/metabolismo , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Haploinsuficiencia , Homocigoto , Humanos , Hipoglucemia/metabolismo , Hipoglucemia/patología , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Hipotálamo/patología , Hipotiroidismo/metabolismo , Hipotiroidismo/patología , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/deficiencia , Neuronas/metabolismo , Neuronas/patología , Hipófisis/crecimiento & desarrollo , Hipófisis/metabolismo , Hipófisis/patología , Polineuropatías/metabolismo , Polineuropatías/patología , Maduración Sexual , Síndrome , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Testículo/patología , Adulto Joven
2.
Genet Med ; 17(8): 651-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25394172

RESUMEN

PURPOSE: Congenital hypogonadotropic hypogonadism (CHH) and split hand/foot malformation (SHFM) are two rare genetic conditions. Here we report a clinical entity comprising the two. METHODS: We identified patients with CHH and SHFM through international collaboration. Probands and available family members underwent phenotyping and screening for FGFR1 mutations. The impact of identified mutations was assessed by sequence- and structure-based predictions and/or functional assays. RESULTS: We identified eight probands with CHH with (n = 3; Kallmann syndrome) or without anosmia (n = 5) and SHFM, seven of whom (88%) harbor FGFR1 mutations. Of these seven, one individual is homozygous for p.V429E and six individuals are heterozygous for p.G348R, p.G485R, p.Q594*, p.E670A, p.V688L, or p.L712P. All mutations were predicted by in silico analysis to cause loss of function. Probands with FGFR1 mutations have severe gonadotropin-releasing hormone deficiency (absent puberty and/or cryptorchidism and/or micropenis). SHFM in both hands and feet was observed only in the patient with the homozygous p.V429E mutation; V429 maps to the fibroblast growth factor receptor substrate 2α binding domain of FGFR1, and functional studies of the p.V429E mutation demonstrated that it decreased recruitment and phosphorylation of fibroblast growth factor receptor substrate 2α to FGFR1, thereby resulting in reduced mitogen-activated protein kinase signaling. CONCLUSION: FGFR1 should be prioritized for genetic testing in patients with CHH and SHFM because the likelihood of a mutation increases from 10% in the general CHH population to 88% in these patients.


Asunto(s)
Hipogonadismo/congénito , Hipogonadismo/genética , Deformidades Congénitas de las Extremidades/genética , Mutación , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Femenino , Estudios de Asociación Genética , Humanos , Hipogonadismo/metabolismo , Deformidades Congénitas de las Extremidades/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Linaje , Fosforilación , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
3.
Bull Acad Natl Med ; 196(2): 327-40; discussion 340-3, 2012 Feb.
Artículo en Francés | MEDLINE | ID: mdl-23420953

RESUMEN

Puberty is triggered by a complex neuroendocrine mechanism that leads to an increase in GnRH release at the end of the childhood and, hence, to reactivation of the gonadotropic axis. Recent human genetic studies have led to major breakthroughs in our understanding of puberty onset. A network of hypothalamic neurons controlling GnRH release has just been characterized. It appears that the timing of puberty onset is under the control of the heterochronic gene Lin28, which encodes a protein regulating microRNA maturation. Characterization of additional gene defects associated with abnormal puberty onset is needed to further characterize this neuroendocrine network and to identify new therapeutic targets for reproductive disorders.


Asunto(s)
Pubertad/genética , Adolescente , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipogonadismo/genética , Pubertad/fisiología
4.
Sex Dev ; 16(1): 64-69, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34628416

RESUMEN

Deficiency of 3ß-hydroxysteroid dehydrogenase type 2 (3ßHSD2) is a rare type of congenital adrenal hyperplasia (CAH), causing impaired steroid hormone production in both adrenals and gonads. Phenotype ranges, according to the genetic defect, from the salt-wasting form in both sexes to undervirilization in males and virilization in females. We present a 13-month-old male infant who was admitted to the hospital with signs of adrenocortical insufficiency and genital ambiguity. Clinical presentation, hormonal profile, laboratory evaluation, and karyotype were suggestive of the salt-wasting form of CAH due to 3ßHSD2 deficiency. Mutational analysis revealed a missense mutation c.776C>T (p.Thr259Met), inherited by the mother, and a frameshift deletion c.818-819delAA (p.Lys273ArgFs*7), inherited by the father. Both mutations are considered pathogenic. To our knowledge this is the first case of an undervirilized male infant with salt wasting bearing this pathogenic frameshift deletion p.Lys273ArgFs*7 in compound heterozygosity with the missense mutation p.Thr259Met.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/genética , Hiperplasia Suprarrenal Congénita , Hiperplasia Suprarrenal Congénita/genética , Mutación del Sistema de Lectura , Humanos , Masculino , Mutación Missense
5.
J Clin Endocrinol Metab ; 107(6): e2553-e2562, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35134944

RESUMEN

CONTEXT: Estrogens play an essential role in reproduction. Their action is mediated by nuclear α and ß receptors (ER) and by membrane receptors. Only 3 females and 2 males, from 3 families, with a loss of ERα function have been reported to date. OBJECTIVE: We describe here a new family, in which 2 sisters display endocrine and ovarian defects of different severities despite carrying the same homozygous rare variant of ESR1. METHODS: A 36-year-old woman from a consanguineous Jordanian family presented with primary amenorrhea and no breast development, with high plasma levels of 17ß-estradiol (E2), follicle-stimulating hormone and luteinizing hormone, and enlarged multifollicular ovaries, strongly suggesting estrogen resistance. Her 18-year-old sister did not enter puberty and had moderately high levels of E2, high plasma gonadotropin levels, and normal ovaries. RESULTS: Genetic analysis identified a homozygous variant of ESR1 leading to the replacement of a highly conserved glutamic acid with a valine (ERα-E385V). The transient expression of ERα-E385V in HEK293A and MDA-MB231 cells revealed highly impaired ERE-dependent transcriptional activation by E2. The analysis of the KISS1 promoter activity revealed that the E385V substitution induced a ligand independent activation of ERα. Immunofluorescence analysis showed that less ERα-E385V than ERα-WT was translocated into the nucleus in the presence of E2. CONCLUSION: These 2 new cases are remarkable given the difference in the severity of their ovarian and hormonal phenotypes. This phenotypic discrepancy may be due to a mechanism partially compensating for the ERα loss of function.


Asunto(s)
Receptor alfa de Estrógeno , Estrógenos , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Estrógenos/farmacología , Femenino , Humanos , Masculino , Fenotipo , Activación Transcripcional
6.
Eur J Endocrinol ; 184(2): 243-251, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33156813

RESUMEN

OBJECTIVE: Isolated central precocious puberty (CPP) includes sporadic, familial and adoption-related forms, and the characterization of its etiology is challenging. This study investigated the prevalence and clinical characteristics of isolated CPP. DESIGN AND METHODS: This observational cohort study included all patients (n = 395) with CPP included in the database of a single academic pediatric care center over a period of 11.5 years. RESULTS: In total, 332 of the 395 patients (84%) had isolated forms of CPP; the proportion of male patients was lower in this group than for non-isolated CPP (4 vs 33%, P < 0.0001). These patients had sporadic (n = 228, 68.5%), familial (n = 82, 25%) or adoption-related (n = 22, 6.5%) forms. Clinical characteristics at diagnosis were similar between groups, but girls with sporadic CPP were older at referral than those with familial or adoption-related CPP (P < 0.02), and birth weight SDS was lower in adopted patients than in those from the sporadic and familial groups (P < 0.01). In the 72 families containing patients with familial forms, both recessive and dominant transmissions were observed between first-degree relatives. Potential maternal or paternal transmission was identified in two-thirds of the studied families, in similar proportions. An autosomal dominant mode of transmission with low penetrance was suggested by the high proportion of affected parents (33 of the 72 families, 46%). Clinical presentation was similar whatever the mode of inheritance. CONCLUSION: These findings highlight the need for careful monitoring of the various forms of CPP. Future studies should explore pathophysiological mechanisms, particularly for familial forms.


Asunto(s)
Pubertad Precoz/clasificación , Pubertad Precoz/epidemiología , Peso al Nacer/fisiología , Niño , Estudios de Cohortes , Familia , Femenino , Humanos , Enfermedades Hipotalámicas/complicaciones , Enfermedades Hipotalámicas/diagnóstico , Enfermedades Hipotalámicas/epidemiología , Recién Nacido , Masculino , Anamnesis , Linaje , Fenotipo , Prevalencia , Pronóstico , Pubertad Precoz/diagnóstico , Pubertad Precoz/etiología
7.
Eur J Endocrinol ; 184(3): 427-436, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33465046

RESUMEN

OBJECTIVE: Neonatal hyperthyroidism may be caused by a permanent non-autoimmune genetic disorder or, more frequently, by maternally transmitted high serum TRAb levels. Variable thyroid dysfunction may be observed in this second context. We aimed to evaluate the prevalence of neonatal non-autoimmune hyperthyroidism and of the different types of thyroid function in neonates with a high risk of hyperthyroidism due to maternal Graves' disease (GD). DESIGN AND METHODS: This observational cohort study included all neonates identified in the database of a single academic pediatric care center, over a period of 13 years, as having non-autoimmune hyperthyroidism or an autoimmune disorder with high TRAb levels (above 6 IU/L) transmitted by their mothers. Patients were classified as having neonatal hyperthyroidism, hypothyroidism, or euthyroidism with a permanent or transient disorder. RESULTS: Two of the 34 consecutive neonates selected (6%) had permanent non-autoimmune hyperthyroidism due to germline (n = 1) or somatic (n = 1) mutations of the TSH receptor gene. The patients with high serum TRAb levels at birth had transient hyperthyroidism (n = 23), hypothyroidism (primary n = 2, central n = 3) or persistent euthyroidism (n = 4). CONCLUSION: These original findings highlight the need for careful and appropriate monitoring of thyroid function in the long term, not only for the rare patients with non-autoimmune neonatal hyperthyroidism, but also for repeat monitoring during the first month of life in neonates with maternally transmitted high TRAb levels, to ensure the early identification of thyrotoxicosis in more than two thirds of cases and to detect primary or central hypothyroidism, thereby potentially decreasing associated morbidity.


Asunto(s)
Enfermedad de Graves/etiología , Hipertiroidismo/congénito , Hipertiroidismo/epidemiología , Enfermedades de la Tiroides/congénito , Enfermedades de la Tiroides/epidemiología , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Enfermedad de Graves/diagnóstico , Enfermedad de Graves/epidemiología , Enfermedad de Graves/genética , Humanos , Hipertiroidismo/diagnóstico , Hipertiroidismo/genética , Lactante , Recién Nacido , Enfermedades del Recién Nacido/diagnóstico , Enfermedades del Recién Nacido/epidemiología , Enfermedades del Recién Nacido/genética , Enfermedades del Recién Nacido/patología , Masculino , Herencia Materna , Tamizaje Neonatal , Embarazo , Complicaciones del Embarazo/diagnóstico , Complicaciones del Embarazo/epidemiología , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/patología , Prevalencia , Pronóstico , Factores de Riesgo , Enfermedades de la Tiroides/diagnóstico , Enfermedades de la Tiroides/genética , Pruebas de Función de la Tiroides
8.
Int J Paediatr Dent ; 20(4): 305-12, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20536592

RESUMEN

BACKGROUND: Kallmann syndrome (KS) is a rare genetic disorder characterised by central hypogonadism with a lack of sense of smell and in some cases renal aplasia, deafness, syndactyly, cleft lip/palate, and dental agenesis. To date, five genes for KS have been identified: KAL1, located on the X chromosome, and FGFR1, PROKR2, PROK2 and FGF8, which are involved in autosomally transmitted forms of KS. AIM: The study characterised the dental ageneses of individuals with KS associated with mutations in the FGFR1 gene. DESIGN: Six individuals displaying dental agenesis were included. Clinical and radiological dental evaluations as well as medical anamneses were carried out. RESULTS: Microdontia, screwdriver-shaped mandibular incisors, thin molar roots, and patterns of dental agenesis in both dentitions were observed. One to nine teeth were missing, most frequently, in descending order, lateral mandibular incisors, second premolars of upper and lower jaws, and lateral maxillary incisors. The pattern of dental agenesis is associated with four new mutations in the FGFR1 gene. CONCLUSION: Dental agenesis may be a clinical feature of Kallmann syndrome caused by a mutation in the FGFR1 gene. These findings highlight the role that odontologists can play in the early diagnosis and treatment of gonadotropic deficiency.


Asunto(s)
Síndrome de Kallmann/genética , Mutación/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Anomalías Dentarias/genética , Adenina , Adolescente , Adulto , Anodoncia/genética , Arginina/genética , Diente Premolar/anomalías , Preescolar , Fisura del Paladar/genética , Cisteína/genética , Citosina , Femenino , Glutamina/genética , Glicina/genética , Guanina , Humanos , Incisivo/anomalías , Masculino , Persona de Mediana Edad , Diente Molar/anomalías , Eliminación de Secuencia/genética , Raíz del Diente/anomalías , Diente Primario/anomalías , Triptófano/genética , Adulto Joven
9.
Rev Prat ; 58(12): 1305-9, 2008 Jun 30.
Artículo en Francés | MEDLINE | ID: mdl-18714650

RESUMEN

Puberty is an important step in human development. Onset of puberty, including neurobiological mechanisms important for the increase of hypothalamic GnRH pulses remains a mystery. After birth, GnRH secretion remains elevated and then decreases during childhood regardless of any steroid gonadal feedback. This period of quiescence of the gonadotropic axis during childhood is linked to a central inhibition of GnRH secretion which is replaced by an activator tone at puberty. The study of the pathology of the pubertal timing, including delayed puberty led to the discovery of new genes involved in the migration of GnRH neurons and genes involved in the neuroendocrine regulation of the gonadotropic axis. Recently, the emphasis on the importance of the kiss/GPR54 system in modulating control of the gonadotropic axis at puberty has recently emerged from Human genetics studies.


Asunto(s)
Pubertad/genética , Pubertad/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Sistema Hipotálamo-Hipofisario/anomalías , Sistema Hipotálamo-Hipofisario/fisiología , Kisspeptinas , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Proteínas Supresoras de Tumor/genética
10.
J Clin Endocrinol Metab ; 103(12): 4482-4490, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30124894

RESUMEN

Context: Mutations in the kisspeptin receptor (KISS1R) gene have been reported in a few patients with normosmic congenital hypogonadotropic hypogonadism (nCHH) (OMIM #146110). Objectives: To describe a female patient with nCHH and a novel homozygous KISS1R mutation and to assess the role of kisspeptin pathway to induce an ovulation by GnRH pulse therapy. Design, Setting, and Intervention: Observational study of a patient including genetic and kisspeptin receptor functions and treatment efficiency using a GnRH pump. Main Outcome Measure: Response to pulsatile GnRH therapy. Results: A partial isolated gonadotropic deficiency was diagnosed in a 28-year-old woman with primary amenorrhea and no breast development. A novel homozygous c.953T>C variant was identified in KISS1R. This mutation led to substitution of leucine 318 for proline (p.Leu318Pro) in the seventh transmembrane domain of KISS1R. Signaling via the mutated receptor was profoundly impaired in HEK293-transfected cells. The mutated receptor was not detected on the membrane of HEK293-transfected cells. After several pulsatile GnRH therapy cycles, an LH surge with ovulation and pregnancy was obtained. Conclusion: GnRH pulsatile therapy can induce an LH surge in a woman with a mutated KISS1R, which was previously thought to be completely inactivated in vivo.


Asunto(s)
Amenorrea/tratamiento farmacológico , Hormona Liberadora de Gonadotropina/administración & dosificación , Hipogonadismo/tratamiento farmacológico , Hormona Luteinizante/metabolismo , Receptores de Kisspeptina-1/genética , Adulto , Amenorrea/genética , Amenorrea/metabolismo , Femenino , Células HEK293 , Homocigoto , Humanos , Hipogonadismo/genética , Hipogonadismo/metabolismo , Kisspeptinas/metabolismo , Mutación con Pérdida de Función , Ovulación/efectos de los fármacos , Ovulación/metabolismo , Embarazo , Quimioterapia por Pulso , Receptores de Kisspeptina-1/deficiencia , Transducción de Señal/genética , Resultado del Tratamiento
11.
Eur J Endocrinol ; 179(6): 373-380, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30324796

RESUMEN

Objective Non-idiopathic CPP is caused by acquired or congenital hypothalamic lesions visible on MRI or is associated with various complex genetic and/or syndromic disorders. This study investigated the different types and prevalence of non-isolated CPP phenotypes. Design and Methods This observational cohort study included all patients identified as having non-idiopathic CPP in the database of a single academic pediatric care center over a period of 11.5 years. Patients were classified on the basis of MRI findings for the CNS as having either hypothalamic lesions or complex syndromic phenotypes without structural lesions of the hypothalamus. Results In total, 63 consecutive children (42 girls and 21 boys) with non-isolated CPP were identified. Diverse diseases were detected, and the hypothalamic lesions visible on MRI (n = 28, 45% of cases) included hamartomas (n = 17; either isolated or with an associated syndromic phenotype), optic gliomas (n = 8; with or without neurofibromatosis type 1), malformations (n = 3) with interhypothalamic adhesions (n = 2; isolated or associated with syndromic CNS midline abnormalities, such as optic nerve hypoplasia, ectopic posterior pituitary) or arachnoid cysts (n = 1). The patients with non-structural hypothalamic lesions (n = 35, 55% of cases) had narcolepsy (n = 9), RASopathies (n = 4), encephalopathy or autism spectrum disorders with or without chromosomal abnormalities (n = 15) and other complex syndromic disorders (n = 7). Conclusion Our findings suggest that a large proportion (55%) of patients with non-isolated probable non-idiopathic CPP may have complex disorders without structural hypothalamic lesions on MRI. Future studies should explore the pathophysiological relevance of the mechanisms underlying CPP in these disorders.


Asunto(s)
Hipotálamo/diagnóstico por imagen , Pubertad Precoz/diagnóstico por imagen , Pubertad Precoz/epidemiología , Vigilancia de Guardia , Niño , Preescolar , Estudios de Cohortes , Estradiol/sangre , Femenino , Humanos , Masculino , Prevalencia , Pubertad Precoz/sangre , Testosterona/sangre
12.
Endocr Relat Cancer ; 14(3): 691-702, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17914099

RESUMEN

KiSS1 is a putative metastasis suppressor gene in melanoma and breast cancer-encoding kisspeptins, which are also described as neuroendocrine regulators of the gonadotropic axis. Negative as well as positive regulation of KiSS1 gene expression by estradiol (E(2)) has been reported in the hypothalamus. Estrogen receptor alpha (ERalpha level is recognized as a marker of breast cancer, raising the question of whether expression of KiSS1 and its G-protein-coupled receptor (GPR54) is down- or upregulated by estrogens in breast cancer cells. KiSS1 was found to be expressed in MDA-MB-231, MCF7, and T47D cell lines, but not in ZR75-1, L56Br, and MDA-MB-435 cells. KiSS1 mRNA levels decreased significantly in ERalpha-negative MDA-MB-231 cells expressing recombinant ERalpha. In contrast, tamoxifen (TAM) treatment of ERalpha-positive MCF7 and T47D cells increased KiSS1 and GPR54 levels. The clinical relevance of this negative regulation of KiSS1 and GPR54 by E(2) was then studied in postmenopausal breast cancers. KiSS1 mRNA increased with the grade of the breast tumors. ERalpha-positive invasive primary tumors expressed sevenfold lower KiSS1 levels than ERalpha-negative tumors. Among ERalpha-positive breast tumors from postmenopausal women treated with TAM, high KiSS1 combined with high GPR54 mRNA tumoral levels was unexpectedly associated with shorter relapse-free survival (RFS) relative to tumors expressing low tumoral mRNA levels of both genes. The contradictory observation of putative metastasis inhibitor role of kisspeptins and RFS to TAM treatment suggests that evaluation of KiSS1 and its receptor tumoral mRNA levels could be new interesting markers of the tumoral resistance to anti-estrogen treatment.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Carcinoma Ductal de Mama/diagnóstico , Neoplasias Hormono-Dependientes/diagnóstico , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/genética , Proteínas Supresoras de Tumor/genética , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Hormonales/farmacología , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/terapia , Supervivencia sin Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Estrógenos/farmacología , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Kisspeptinas , Persona de Mediana Edad , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/genética , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/terapia , Pronóstico , Receptores de Kisspeptina-1 , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Células Tumorales Cultivadas
13.
J Clin Endocrinol Metab ; 92(3): 1137-44, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17164310

RESUMEN

CONTEXT: Loss of function of the G protein-coupled receptor of kisspeptins (GPR54) was recently described as a new cause of isolated hypogonadotropic hypogonadism. In vivo studies performed in several species have confirmed the major role of kisspeptins in neuroendocrine regulation of the gonadotropic axis and therefore sexual maturation. OBJECTIVE: The objective of this study was to specify the exact contribution of kisspeptins and GPR54 to the initiation of puberty in humans. DESIGN: Detailed neuroendocrine descriptions were performed in five patients with isolated hypogonadotropic hypogonadism bearing a new GPR54-inactivating mutation. RESULTS: A homozygous mutation (T305C) leading to a leucine substitution with proline (L102P) was found in the five affected patients. This substitution completely inhibited GPR54 signaling. Phenotypic analysis revealed variable expressivity in the same family, either partial or complete gonadotropic deficiency. LH pulsatility analysis showed peaks with normal frequency but low amplitude. Repeated GnRH tests performed between 12 and 21 yr of age in one affected male revealed progressive changes in pituitary response from an early pubertal to an almost full pubertal pattern. Double GnRH test stimulations performed at a 120-min interval showed reduced dynamic pituitary response in GPR54-mutated patients. CONCLUSION: GPR54 inactivation does not impede neuroendocrine onset of puberty; rather, it delays and slows down pubertal maturation of the gonadotropic axis. The L102P loss of function mutation in GPR54 results in a more quantitative than qualitative defect of gonadotropic axis activation.


Asunto(s)
Hipogonadismo/genética , Polimorfismo Genético , Receptores Acoplados a Proteínas G/genética , Adulto , Células Cultivadas , Análisis Mutacional de ADN , Femenino , Humanos , Hormona Luteinizante/metabolismo , Masculino , Modelos Biológicos , Mutación , Linaje , Fenotipo , Pruebas de Función Hipofisaria , Flujo Pulsátil , Receptores de Kisspeptina-1 , Análisis de Secuencia de ADN
14.
Sci Rep ; 7: 42463, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28209974

RESUMEN

A few hundred hypothalamic neurons form a complex network that controls reproduction in mammals by secreting gonadotropin-releasing hormone (GnRH). Timely postnatal changes in GnRH secretion are essential for pubertal onset. During the juvenile period, GnRH neurons undergo morphological remodeling, concomitantly achieving an increased responsiveness to kisspeptin, the main secretagogue of GnRH. However, the link between GnRH neuron activity and their morphology remains unknown. Here, we show that brain expression levels of Dmxl2, which encodes the vesicular protein rabconnectin-3α, determine the capacity of GnRH neurons to be activated by kisspeptin and estradiol. We also demonstrate that Dmxl2 expression levels control the pruning of GnRH dendrites, highlighting an unexpected role for a vesicular protein in the maturation of GnRH neuronal network. This effect is mediated by rabconnectin-3α in neurons or glial cells afferent to GnRH neurons. The widespread expression of Dmxl2 in several brain areas raises the intriguing hypothesis that rabconnectin-3α could be involved in the maturation of other neuronal populations.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Animales , Dendritas/metabolismo , Estradiol/metabolismo , Estradiol/farmacología , Femenino , Eliminación de Gen , Expresión Génica , Kisspeptinas/farmacología , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Maduración Sexual
15.
Eur J Hum Genet ; 25(8): 1011-1014, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28589944

RESUMEN

CHES (cerebellar hypoplasia with endosteal sclerosis) syndrome (OMIM#213002) associates hypomyelination, cerebellar atrophy, hypogonadism and hypodontia. So far, only five patients have been described. The condition is of neonatal onset. Patients have severe psychomotor delay and moderate to severe intellectual disability. Inheritance is assumed to be autosomal recessive due to recurrence in sibs, consanguinity of parents and absence of vertical transmission. CHES syndrome is reminiscent of 4H-leukodystrophy, a recessive-inherited affection due to variations in genes encoding subunits of the RNA polymerase III (POLR3A-POLR3B-POLR1C). POLR3B variants have been identified in one CHES patient. Here we report on a novel CHES patient, carrying compound heterozygous variations in POLR3B. This report confirms affiliation of CHES to POLR3-related disorders and suggests that CHES syndrome represents a severe form of 4H-leukodystrophy.


Asunto(s)
Ataxia Cerebelosa/genética , Osteosclerosis/genética , ARN Polimerasa III/genética , Adolescente , Ataxia Cerebelosa/diagnóstico , Heterocigoto , Humanos , Masculino , Mutación Missense , Osteosclerosis/diagnóstico
16.
Best Pract Res Clin Endocrinol Metab ; 20(4): 515-28, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17161329

RESUMEN

Isolated hypogonadotropic hypogonadism (IHH) is defined by a complete or partial impaired secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH). In the regulation of the gonadotropic axis, the gonadotropin-releasing hormone (GnRH) and its receptor have evolved as a central element in fetal life, at puberty, and for reproduction in adulthood. GnRH resistance due to GnRH receptor (GnRHR) germ-line mutations was the first genetic alteration identified in patients with IHH. GnRHR mutated receptors are associated with impaired GnRH binding, intracellular trafficking or ligand-induced signal transduction, leading to various degrees of LH and FSH deficiency. Loss-of-function mutations of the GnRH receptor account for 50% of familial cases of IHH without anosmia. In 2003, mutations of GPR54 were identified in patients with IHH, opening a new pathway in the physiological regulation of puberty and reproduction. Kisspeptins, which are the natural ligands of GPR54, are potent stimulators of the LH and FSH secretion via the control of GnRH secretion or modulation of the pituitary response to GnRH stimulation. Genotype-phenotype correlations in IHH due to GnRHR and GPR54 mutations indicate that similar mutations may lead to a variable phenotype and suggest that the pituitary might have its own pubertal maturation independent from GnRH. These two causes of IHH result in a more quantitative than qualitative defect of the gonadotropic axis activation. Molecular genetics of IHH has led to a major breakthrough in the neuroendocrine regulation of the gonadotropic axis. New insights into the understanding of the initiation of puberty and in the therapeutic management of defects of the gonadotropic axis have emerged from these studies.


Asunto(s)
Hipogonadismo/genética , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores LHRH/genética , Animales , Modelos Animales de Enfermedad , Hormona Folículo Estimulante/metabolismo , Regulación de la Expresión Génica , Genotipo , Mutación de Línea Germinal , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Kisspeptinas , Hormona Luteinizante/metabolismo , Fenotipo , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Receptores Acoplados a Proteínas G/fisiología , Receptores de Kisspeptina-1 , Receptores LHRH/fisiología , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo
17.
Mol Cell Endocrinol ; 254-255: 78-83, 2006 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-16757108

RESUMEN

Kallmann syndrome characterised by hypogonadotropic hypogonadism (HH) and anosmia is genetically heterogeneous with X-linked, autosomal dominant and autosomal recessive forms. The autosomal dominant form due to loss of function mutation in the fibroblast growth factor receptor 1 (FGFR1) accounts for about 10% of cases. We report here three paediatric cases of Kallmann syndrome with unusual phenotype in two unrelated patients with severe ear anomalies (hypoplasia or agenesis of external ear) associated with classical features, such as cleft palate, dental agenesis, syndactylia, micropenis and cryptorchidism. We found de novo mutation in these two patients (Cys178Ser and Arg622Gly, respectively), and one inherited Arg622Gln mutation with intrafamilial variable phenotype. These genotype-phenotype correlations indicate that paediatric phenotypic expression of FGFR1 loss of function mutations is highly variable, the severity of the oro-facial malformations at birth does not predict gonadotropic function at the puberty and that de novo mutations of FGFR1 are relatively frequent.


Asunto(s)
Síndrome de Kallmann/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Adolescente , Fisura del Paladar/genética , Criptorquidismo/genética , Análisis Mutacional de ADN , Sordera/congénito , Hipoplasia del Esmalte Dental/genética , Hormona Folículo Estimulante/sangre , Genitales Masculinos/anomalías , Humanos , Lactante , Cariotipificación , Hormona Luteinizante/sangre , Masculino , Mutación , Trastornos del Olfato/congénito , Bulbo Olfatorio/anomalías , Linaje , Fenotipo , Pubertad Tardía/genética , Sindactilia/genética , Sincinesia/congénito , Testosterona/sangre
18.
Mol Endocrinol ; 19(8): 2086-98, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15878956

RESUMEN

Several molecular models of glycoprotein hormone receptor activation have been proposed. It has been suggested that ligand binding to the ectodomain (ECD) leads to major changes in intramolecular interactions between the ECD and the transmembrane domain. We studied these intramolecular modifications by generating a recombinant LH/CG receptor (LHR) bearing an intramolecular cleavage site. We did this by inserting a furin site at position 316 in the hinge region of the ECD (LHR_Fur316). Affinity for human chorionic gonadotropin (hCG) and cAMP production upon hCG stimulation was identical to those of wild-type LHR. Western blot analysis showed that the LHR_Fur316 receptor was cleaved into two subunits linked by disulfide bridges. Chemical shedding of the ECD from the transmembrane domain did not increase basal adenylate cyclase activity, indicating that the first 294 residues did not act as an inverse agonist. The truncated LHR_316 was still activated by hCG but with an EC50 higher than that for the wild-type receptor. Zero length cross-linking was used to study intramolecular interactions between the two domains of LHR_Fur316. Cross-linking efficiency was similar for the basal and activated states, which indicated that the two domains interacted closely in the basal state, and this tight interaction persisted during activation. Our data suggest that activation of the LHR results from subtle modifications of intramolecular interactions between the two domains and low-affinity binding of hCG to the extracellular loops or residues preceding the first transmembrane segment.


Asunto(s)
Reactivos de Enlaces Cruzados/farmacología , Receptores de HL/química , Ácido Aspártico/química , Sitios de Unión , Western Blotting , Membrana Celular/metabolismo , Separación Celular , AMP Cíclico/metabolismo , Disulfuros/química , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Furina/química , Ácido Glutámico/química , Humanos , Immunoblotting , Lisina/química , Modelos Biológicos , Unión Proteica , Estructura Terciaria de Proteína , Receptores de HL/metabolismo , Proteínas Recombinantes/química , Factores de Tiempo , Transfección
19.
Pediatr Neurol ; 34(5): 372-5, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16647997

RESUMEN

Since the description of Rett syndrome, only a handful of epidemiologic studies based only on clinical investigation have been reported. Mutations in the MECP2 gene are associated with Rett syndrome and French laboratories have organized a clinical and molecular network to investigate the incidence of Rett syndrome in France including the results of molecular investigations. The present study, based on a large cohort of 424 patients with Rett syndrome, found that the incidence of this disease with a MECP2 mutation varied between 0.43 to 0.71 per 10,000 females. The total population of females aged 4-15 years in November 2004 in France was estimated to be 4,337,627. The data presented here indicate a prevalence of Rett syndrome of 0.558 per 10,000 females aged 4-15 years in France. The incidence of Rett syndrome is in accordance with other European epidemiologic studies based on clinical examination. Given that this is a minimum incidence because complete inventory was not possible, this study of patients with Rett syndrome reinforces the fact that the great majority of patients with Rett syndrome have a MECP2 mutation.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/genética , Síndrome de Rett/epidemiología , Síndrome de Rett/genética , Adolescente , Adulto , Niño , Femenino , Francia/epidemiología , Predisposición Genética a la Enfermedad/epidemiología , Humanos , Incidencia , Tamizaje Masivo , Mutación , Prevalencia
20.
Endocr Dev ; 29: 72-86, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26680573

RESUMEN

Reproductive function depends on the activity of the gonadotropic axis, which is controlled by a hypothalamic neural network whose main function is to regulate the secretion of gonadotropin-releasing hormone (GnRH). This endocrine network is not mature at birth, and several phases of activation-inactivation of the gonadotropic axis are necessary for its normal development. The postnatal maturation of the GnRH network lies under the control of a neurodevelopmental program that starts in fetal life and ends at puberty. There are many clinical situations in which this program is interrupted, leading to congenital hypogonadotropic hypogonadism (CHH) and an absence of puberty. For many years, attention has mainly been focused on the genetics of isolated CHH. More recently, the emergence of new genomics techniques has led to the description of genetic defects in very rare syndromes in which CHH is associated with complex neurological dysfunctions. Here, we review the clinical phenotype and genetic defects linked to such syndromic CHH. This analysis highlights the close link between the ubiquitin pathway, synaptic proteins and CHH, as well as unexpected mutations in genes encoding nucleolar proteins.


Asunto(s)
Gonadotropinas/deficiencia , Hipogonadismo/congénito , Trastornos del Neurodesarrollo/complicaciones , Niño , Preescolar , Femenino , Gonadotropinas/genética , Humanos , Hipogonadismo/complicaciones , Hipogonadismo/genética , Lactante , Recién Nacido , Masculino , Trastornos del Neurodesarrollo/genética , Sistemas Neurosecretores/fisiopatología , Embarazo , Maduración Sexual/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA