Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Genet ; 141(2): 295-304, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35066646

RESUMEN

Metabolism has a role in determining the time of pubertal development and fertility. Nonetheless, molecular/cellular pathways linking metabolism/body weight to puberty/reproduction are unknown. The KNDy (Kisspeptin/Neurokinin B/Dynorphin) neurons in the arcuate nucleus of the hypothalamus constitute the GnRH (gonadotropin-releasing hormone) pulse generator. We previously created a mouse model with a whole-body targeted deletion of nescient helix-loop-helix 2 (Nhlh2; N2KO), a class II member of the basic helix-loop-helix family of transcription factors. As this mouse model features pubertal failure and late-onset obesity, we wanted to study whether NHLH2 represents a candidate molecule to link metabolism and puberty in the hypothalamus. Exome sequencing of a large Idiopathic Hypogonadotropic Hypogonadism cohort revealed obese patients with rare sequence variants in NHLH2, which were characterized by in-silico protein analysis, chromatin immunoprecipitation, and luciferase reporter assays. In vitro heterologous expression studies demonstrated that the variant p.R79C impairs Nhlh2 binding to the Mc4r promoter. Furthermore, p.R79C and other variants show impaired transactivation of the human KISS1 promoter. These are the first inactivating human variants that support NHLH2's critical role in human puberty and body weight control. Failure to carry out this function results in the absence of pubertal development and late-onset obesity in humans.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipogonadismo/genética , Obesidad/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Femenino , Variación Genética , Humanos , Hipogonadismo/etiología , Hipogonadismo/metabolismo , Kisspeptinas/genética , Masculino , Redes y Vías Metabólicas/genética , Ratones , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Obesidad/etiología , Obesidad/metabolismo , Linaje , Regiones Promotoras Genéticas , Conformación Proteica , Activación Transcripcional , Adulto Joven
2.
Genet Med ; 23(6): 1008-1016, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33495532

RESUMEN

PURPOSE: Idiopathic hypogonadotropic hypogonadism (IHH) is characterized by absent puberty and subsequent infertility due to gonadotropin-releasing hormone (GnRH) deficiency. IHH can be accompanied by normal or compromised olfaction (Kallmann syndrome). Several semaphorins are known potent modulators of GnRH, olfactory, and vomeronasal system development. In this study, we investigated the role of Semaphorin-3F signaling in the etiology of IHH. METHODS: We screened 216 IHH patients by exome sequencing. We transiently transfected HEK293T cells with plasmids encoding wild type (WT) or corresponding variants to investigate the functional consequences. We performed fluorescent IHC to assess SEMA3F and PLXNA3 expression both in the nasal region and at the nasal/forebrain junction during the early human fetal development. RESULTS: We identified ten rare missense variants in SEMA3F and PLXNA3 in 15 patients from 11 independent families. Most of these variants were predicted to be deleterious by functional assays. SEMA3F and PLXNA3 are both expressed along the olfactory nerve and intracranial projection of the vomeronasal nerve/terminal nerve. PLXNA1-A3 are expressed in the early migratory GnRH neurons. CONCLUSION: SEMA3F signaling through PLXNA1-A3 is involved in the guidance of GnRH neurons and of olfactory and vomeronasal nerve fibers in humans. Overall, our findings suggest that Semaphorin-3F signaling insufficiency contributes to the pathogenesis of IHH.


Asunto(s)
Hipogonadismo , Semaforinas , Moléculas de Adhesión Celular , Células HEK293 , Humanos , Hipogonadismo/genética , Proteínas de la Membrana , Proteínas del Tejido Nervioso/genética , Receptores de Superficie Celular
3.
Clin Genet ; 95(2): 320-324, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30467832

RESUMEN

Idiopathic hypogonadotropic hypogonadism (IHH) can be divided into two major forms, normosmic IHH and Kallmann syndrome (KS). Genetic mutations are responsible for the majority of IHH. PLXNA1 has recently been implicated in the GnRH neuron migration and the etiology of KS. We aimed to investigate the prevalence and associated phenotypes of PLXNA1 variants in a large cohort of IHH patients. We screened the whole exome data of 215 IHH patients in a single center for causative PLXNA1 variants. Our studies showed eight novel (p.Arg836His, p.Lys1451Arg, p.Val287Met, p.Val536Ile, p.Ser1850Arg, p.Ile1701Val, p.Arg319Trp, and p.Pro485Leu) and two previously described (p.Arg528Trp and p.Gly720Glu) heterozygous PLXNA1 variants in nine affected individuals from seven unrelated families. Only three of nine patients were anosmic (KS) while the remaining patients showed normal olfactory function (nIHH). Seven of nine patients (77.7%) harbored additional one or two variants in other nIHH/KS-associated genes, including PROKR2, IGSF10, HS6ST1, SEMA3E, CCDC141, FGFR1, NRP1, POLR3A, and SRA1. Our findings indicate that PLXNA1 variants cause not only anosmic but also normosmic IHH with a relatively high prevalence (3.9%). Heterozygous missense PLXNA1 variants appear to be involved together with other IHH gene variants in bringing about the IHH disease phenotype.


Asunto(s)
Predisposición Genética a la Enfermedad , Hipogonadismo/epidemiología , Hipogonadismo/genética , Mutación , Proteínas del Tejido Nervioso/genética , Fenotipo , Receptores de Superficie Celular/genética , Adolescente , Adulto , Alelos , Biomarcadores , Biología Computacional/métodos , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Hipogonadismo/diagnóstico , Hipogonadismo/metabolismo , Masculino , Prevalencia , Secuenciación del Exoma , Adulto Joven
4.
Am J Hum Genet ; 95(3): 326-31, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25192046

RESUMEN

Gonadotropin-releasing hormone (GnRH) neurons originate outside the CNS in the olfactory placode and migrate into the CNS, where they become integral components of the hypothalamic-pituitary-gonadal (HPG) axis. Disruption of this migration results in Kallmann syndrome (KS), which is characterized by anosmia and pubertal failure due to hypogonadotropic hypogonadism. Using candidate-gene screening, autozygosity mapping, and whole-exome sequencing in a cohort of 30 individuals with KS, we searched for genes newly associated with KS. We identified homozygous loss-of-function mutations in FEZF1 in two independent consanguineous families each with two affected siblings. The FEZF1 product is known to enable axons of olfactory receptor neurons (ORNs) to penetrate the CNS basal lamina in mice. Because a subset of axons in these tracks is the migratory pathway for GnRH neurons, in FEZF1 deficiency, GnRH neurons also fail to enter the brain. These results indicate that FEZF1 is required for establishment of the central component of the HPG axis in humans.


Asunto(s)
Proteínas de Unión al ADN/genética , Síndrome de Kallmann/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética , Adolescente , Adulto , Animales , Axones/metabolismo , Axones/patología , Encéfalo/metabolismo , Encéfalo/patología , Niño , Familia , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipogonadismo , Sistema Hipotálamo-Hipofisario , Masculino , Ratones , Neuronas Receptoras Olfatorias/metabolismo , Neuronas Receptoras Olfatorias/patología , Linaje , Estudios Prospectivos , Proteínas Represoras , Adulto Joven
5.
Am J Hum Genet ; 95(6): 754-62, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25480036

RESUMEN

Premature ovarian failure (POF) is genetically heterogeneous and manifests as hypergonadotropic hypogonadism either as part of a syndrome or in isolation. We studied two unrelated consanguineous families with daughters exhibiting primary amenorrhea, short stature, and a 46,XX karyotype. A combination of SNP arrays, comparative genomic hybridization arrays, and whole-exome sequencing analyses identified homozygous pathogenic variants in MCM9, a gene implicated in homologous recombination and repair of double-stranded DNA breaks. In one family, the MCM9 c.1732+2T>C variant alters a splice donor site, resulting in abnormal alternative splicing and truncated forms of MCM9 that are unable to be recruited to sites of DNA damage. In the second family, MCM9 c.394C>T (p.Arg132(∗)) results in a predicted loss of functional MCM9. Repair of chromosome breaks was impaired in lymphocytes from affected, but not unaffected, females in both families, consistent with MCM9 function in homologous recombination. Autosomal-recessive variants in MCM9 cause a genomic-instability syndrome associated with hypergonadotropic hypogonadism and short stature. Preferential sensitivity of germline meiosis to MCM9 functional deficiency and compromised DNA repair in the somatic component most likely account for the ovarian failure and short stature.


Asunto(s)
Amenorrea/genética , Inestabilidad Cromosómica/genética , Enanismo/genética , Proteínas de Mantenimiento de Minicromosoma/genética , Polimorfismo de Nucleótido Simple/genética , Insuficiencia Ovárica Primaria/genética , Cariotipo Anormal , Adolescente , Adulto , Secuencia de Bases , Línea Celular , Consanguinidad , Roturas del ADN de Doble Cadena , Reparación del ADN , Exoma/genética , Femenino , Recombinación Homóloga , Homocigoto , Humanos , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , Linaje , Sitios de Empalme de ARN , Análisis de Secuencia de ADN , Adulto Joven
6.
Biochim Biophys Acta ; 1844(7): 1248-59, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24717288

RESUMEN

Fibroblast activation protein (FAP) is a focus of interest as a potential cancer therapy target. This membrane bound protease possesses the unique catalytic activity of hydrolysis of the post-proline bond two or more residues from the N-terminus of substrates. FAP is highly expressed in activated fibroblastic cells in tumours, arthritis and fibrosis. A rare, novel, human polymorphism, C1088T, encoding Ser363 to Leu, occurring in the sixth blade of the ß propeller domain, was identified in a family. Both in primary human fibroblasts and in Ser363LeuFAP transfected cells, we showed that this single substitution ablates FAP dimerisation and causes loss of enzyme activity. Ser363LeuFAP was detectable only in endoplasmic reticulum (ER), in contrast to the distribution of wild-type FAP on the cell surface. The variant FAP showed decreased conformational antibody binding, consistent with an altered tertiary structure. Ser363LeuFAP expression was associated with upregulation of the ER chaperone BiP/GRP78, ER stress sensor ATF6, and the ER stress response target phospho-eIF2α, all indicators of ER stress. Proteasomal inhibition resulted in accumulation of Ser363LeuFAP, indicating the involvement of ER associated degradation (ERAD). Neither CHOP expression nor apoptosis was elevated, so ERAD is probably important for protecting Ser363LeuFAP expressing cells. These data on the first loss of function human FAP gene variant indicates that although the protein is vulnerable to an amino acid substitution in the ß-propeller domain, inactive, unfolded FAP can be tolerated by cells.


Asunto(s)
Braquidactilia/genética , Sordera/genética , Estrés del Retículo Endoplásmico/genética , Degradación Asociada con el Retículo Endoplásmico/genética , Gelatinasas/genética , Gelatinasas/metabolismo , Discapacidad Intelectual/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Anomalías de la Boca/genética , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Anomalías Dentarias/genética , Sustitución de Aminoácidos , Apoptosis , Western Blotting , Estudios de Casos y Controles , Membrana Celular/metabolismo , Células Cultivadas , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Endopeptidasas , Chaperón BiP del Retículo Endoplásmico , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Piel/citología , Piel/metabolismo , Fracciones Subcelulares
7.
N Engl J Med ; 366(7): 629-35, 2012 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-22335740

RESUMEN

Gonadotropin-releasing hormone (GnRH) is the central regulator of gonadotropins, which stimulate gonadal function. Hypothalamic neurons that produce kisspeptin and neurokinin B stimulate GnRH release. Inactivating mutations in the genes encoding the human kisspeptin receptor (KISS1R, formerly called GPR54), neurokinin B (TAC3), and the neurokinin B receptor (TACR3) result in pubertal failure. However, human kisspeptin loss-of-function mutations have not been described, and contradictory findings have been reported in Kiss1-knockout mice. We describe an inactivating mutation in KISS1 in a large consanguineous family that results in failure of pubertal progression, indicating that functional kisspeptin is important for puberty and reproduction in humans. (Funded by the Scientific and Technological Research Council of Turkey [TÜBITAK] and others.).


Asunto(s)
Hipogonadismo/genética , Kisspeptinas/genética , Mutación , Pubertad/genética , Adolescente , Adulto , Niño , Consanguinidad , Femenino , Genes Recesivos , Técnicas de Genotipaje , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Masculino , Linaje , Análisis de Secuencia de ADN
8.
Clin Endocrinol (Oxf) ; 82(3): 429-38, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25262569

RESUMEN

OBJECTIVE: The spectrum of genetic alterations in cases of hypogonadotropic hypogonadism continue to expand. However, KISS1R mutations remain rare. The aim of this study was to understand the molecular basis of normosmic idiopathic hypogonadotropic hypogonadism. METHODS: Clinical characteristics, hormonal studies and genetic analyses of seven cases with idiopathic normosmic hypogonadotropic hypogonadism (nIHH) from three unrelated consanguineous families are presented. RESULTS: One male presented with absence of pubertal onset and required surgery for severe penoscrotal hypospadias and cryptorchidism, while other two males had absence of pubertal onset. Two of four female cases required replacement therapy for pubertal onset and maintenance, whereas the other two had spontaneous pubertal onset but incomplete maturation. In sequence analysis, we identified a novel homozygous nonsense (p.Y323X) mutation (c.C969A) in the last exon of the KISS1R gene in all clinically affected cases. CONCLUSIONS: We identified a homozygous nonsense mutation in the KISS1R gene in three unrelated families with nIHH, which enabled us to observe the phenotypic consequences of this rare condition. Escape from nonsense-mediated decay, and thus production of abnormal proteins, may account for the variable severity of the phenotype. Although KISS1R mutations are extremely rare and can cause a heterogeneous phenotype, analysis of the KISS1R gene should be a part of genetic analysis of patients with nIHH, to allow better understanding of phenotype-genotype relationship of KISS1R mutations and the underlying genetic basis of patients with nIHH.


Asunto(s)
Codón sin Sentido/genética , Hipogonadismo/genética , Receptores Acoplados a Proteínas G/genética , Adolescente , Adulto , Humanos , Hipogonadismo/etiología , Masculino , Receptores de Kisspeptina-1 , Adulto Joven
9.
Am J Hum Genet ; 87(6): 757-67, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-21129728

RESUMEN

Altered Bone Morphogenetic Protein (BMP) signaling leads to multiple developmental defects, including brachydactyly and deafness. Here we identify chondroitin synthase 1 (CHSY1) as a potential mediator of BMP effects. We show that loss of human CHSY1 function causes autosomal-recessive Temtamy preaxial brachydactyly syndrome (TPBS), mainly characterized by limb malformations, short stature, and hearing loss. After mapping the TPBS locus to chromosome 15q26-qterm, we identified causative mutations in five consanguineous TPBS families. In zebrafish, antisense-mediated chsy1 knockdown causes defects in multiple developmental processes, some of which are likely to also be causative in the etiology of TPBS. In the inner ears of zebrafish larvae, chsy1 is expressed similarly to the BMP inhibitor dan and in a complementary fashion to bmp2b. Furthermore, unrestricted Bmp2b signaling or loss of Dan activity leads to reduced chsy1 expression and, during epithelial morphogenesis, defects similar to those that occur upon Chsy1 inactivation, indicating that Bmp signaling affects inner-ear development by repressing chsy1. In addition, we obtained strikingly similar zebrafish phenotypes after chsy1 overexpression, which might explain why, in humans, brachydactyly can be caused by mutations leading either to loss or to gain of BMP signaling.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Mutación , N-Acetilgalactosaminiltransferasas/genética , Transducción de Señal , Animales , Braquidactilia , Mapeo Cromosómico , Cromosomas Humanos Par 15 , Deformidades Congénitas del Pie/genética , Deformidades Congénitas de la Mano/genética , Humanos , N-Acetilgalactosaminiltransferasas/metabolismo , Síndrome , Pez Cebra
10.
Front Endocrinol (Lausanne) ; 14: 1203542, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37600690

RESUMEN

Idiopathic hypogonadotropic hypogonadism (IHH) is characterized by the absence of pubertal development and subsequent impaired fertility often due to gonadotropin-releasing hormone (GnRH) deficits. Exome sequencing of two independent cohorts of IHH patients identified 12 rare missense variants in POU6F2 in 15 patients. POU6F2 encodes two distinct isoforms. In the adult mouse, expression of both isoform1 and isoform2 was detected in the brain, pituitary, and gonads. However, only isoform1 was detected in mouse primary GnRH cells and three immortalized GnRH cell lines, two mouse and one human. To date, the function of isoform2 has been verified as a transcription factor, while the function of isoform1 has been unknown. In the present report, bioinformatics and cell assays on a human-derived GnRH cell line reveal a novel function for isoform1, demonstrating it can act as a transcriptional regulator, decreasing GNRH1 expression. In addition, the impact of the two most prevalent POU6F2 variants, identified in five IHH patients, that were located at/or close to the DNA-binding domain was examined. Notably, one of these mutations prevented the repression of GnRH transcripts by isoform1. Normally, GnRH transcription increases as GnRH cells mature as they near migrate into the brain. Augmentation earlier during development can disrupt normal GnRH cell migration, consistent with some POU6F2 variants contributing to the IHH pathogenesis.


Asunto(s)
Encéfalo , Hipogonadismo , Mutación Missense , Factores del Dominio POU , Animales , Humanos , Ratones , Hormona Liberadora de Gonadotropina/genética , Factores del Dominio POU/genética , Hipogonadismo/genética
11.
J Neuroendocrinol ; 34(5): e13080, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34970798

RESUMEN

GnRH is the pivotal hormone in controlling the hypothalamic-pituitary gonadal (HPG) axis in humans and other mammalian species. GnRH function is influenced by a multitude of known and still unknown environmental and genetic factors. Molecular genetic studies on human families with hypogonadotropic hypogonadism over the past two decades have been instrumental in delineating the kisspeptin and neurokinin B signalling, which integrally modulates GnRH release from the hypothalamus. The identification of kisspeptin and neurokinin B ligand-receptor gene pair mutations in patients with absent puberty have paved the way to a greater understanding of the central regulation of the HPG cascade. In this article, we aim to review the literature on the genetic and clinical aspects of GnRH and its receptor, as well as the two ligand-receptor sets directly pertinent to the function of GnRH hormone signalling, kisspeptin/ kisspeptin receptor and NKB/NK3R.


Asunto(s)
Kisspeptinas , Neuroquinina B , Animales , Hormona Liberadora de Gonadotropina/genética , Genética Humana , Humanos , Kisspeptinas/genética , Ligandos , Mamíferos , Neuroquinina B/genética
12.
J Neuroendocrinol ; 34(4): e13103, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35170806

RESUMEN

Idiopathic hypogonadotropic hypogonadism (IHH) comprises a group of rare genetic disorders characterized by pubertal failure caused by gonadotropin-releasing hormone (GnRH) deficiency. Genetic factors involved in semaphorin/plexin signaling have been identified in patients with IHH. PlexinB1, a member of the plexin family receptors, serves as the receptor for semaphorin 4D (Sema4D). In mice, perturbations in Sema4D/PlexinB1 signaling leads to improper GnRH development, highlighting the importance of investigating PlexinB1 mutations in IHH families. In total, 336 IHH patients (normosmic IHH, n = 293 and Kallmann syndrome, n = 43) from 290 independent families were included in the present study. Six PLXNB1 rare sequence variants (p.N361S, p.V608A, p.R636C, p.V672A, p.R1031H, and p.C1318R) are described in eight normosmic IHH patients from seven independent families. These variants were examined using bioinformatic modeling and compared to mutants reported in PLXNA1. Based on these analyses, the variant p.R1031H was assayed for alterations in cell morphology, PlexinB1 expression, and migration using a GnRH cell line and Boyden chambers. Experiments showed reduced membrane expression and impaired migration in cells expressing this variant compared to the wild-type. Our results provide clinical, genetic, molecular/cellular, and modeling evidence to implicate variants in PLXNB1 in the etiology of IHH.


Asunto(s)
Hipogonadismo , Síndrome de Kallmann , Proteínas del Tejido Nervioso/genética , Receptores de Superficie Celular/genética , Animales , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipogonadismo/genética , Síndrome de Kallmann/genética , Masculino , Ratones , Mutación
13.
Clin Endocrinol (Oxf) ; 72(4): 427-35, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19719764

RESUMEN

The complex organization and regulation of the human hypothalamic-pituitary-gonadal axis render it susceptible to dysfunction in the face of a variety of genetic insults, leading to different degrees of hypogonadotrophic hypogonadism (HH). Although the genetic basis of some HH was recognized more than 60 years ago the first specific pathogenic defect, in the KAL1 gene, was only identified within the last 20 years. In the past decade, the rate of genetic discovery has dramatically accelerated, with defects in more than 10 genes now associated with HH. Several themes have emerged as the genetic basis of HH has gradually been uncovered, including the association of some genes such as FGFR1, FGF8, PROK2 and PROKR2, both with HH in association with hyposmia/anosmia (Kallmann syndrome) and with normosmic HH, thus blurring the clinical distinction between ontogenic and purely functional defects in the axis. Many examples of digenic inheritance of HH have also been reported, sometimes producing variable reproductive and accessory phenotypes within a family with non-Mendelian inheritance patterns. In strictly normosmic HH, human genetics has made a particularly dramatic impact in the past 6 years through homozygosity mapping in consanguineous families, first through identification of a key role for kisspeptin in triggering GnRH release, and very recently through demonstration of a critical role for neurokinin B in normal sexual maturation. This review summarises current understanding of the genetic architecture of HH, as well as its diagnostic and mechanistic implications.


Asunto(s)
Hipogonadismo/genética , Síndrome de Kallmann/genética , Consanguinidad , Proteínas de la Matriz Extracelular/genética , Femenino , Factor 8 de Crecimiento de Fibroblastos/genética , Hormonas Gastrointestinales/genética , Gónadas/fisiopatología , Humanos , Hipogonadismo/patología , Hipogonadismo/fisiopatología , Sistema Hipotálamo-Hipofisario/fisiopatología , Kisspeptinas , Masculino , Proteínas del Tejido Nervioso/genética , Neuroquinina B/genética , Neuropéptidos/genética , Fenotipo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Receptores de Neuroquinina-3/genética , Receptores de Péptidos/genética , Maduración Sexual/genética , Proteínas Supresoras de Tumor/fisiología
14.
Pediatr Diabetes ; 11(4): 279-85, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20202148

RESUMEN

Wolcott-Rallison syndrome (WRS) is a rare autosomal recessive disorder characterized by an early-infancy-onset diabetes mellitus associated with a variety of multisystemic clinical manifestations. Here, we present six patients with WRS, carrying the same homozygous mutation (EIF2AK3-W522X), from two unrelated Turkish families. This is the largest series of patients with the same mutation for this rare syndrome. In this communication we compare clinical features of these six patients with the other 34 patients who have been reported to date, and review the clinical features of WRS. All WRS patients presented first with symptoms of insulin dependent diabetes mellitus, with a mean age at onset of 2 months. All patients had skeletal dysplasia or early signs of it, and growth retardation. Many of the patients with WRS have been reported to have developmental delay, mental retardation, and learning difficulties; in contrast, none of our patients showed abnormal development at age up to 30 months. Acute attacks of hepatic failure were reported in 23 cases out of 37 patients; in 15 of those 23 cases an acute attack of renal failure accompanied the liver failure. Exocrine pancreatic deficiency has been reported in only four cases other than our four patients. Central hypothyroidism was observed in six of 28 cases. We propose that central hypothyroidism is not a component of WRS, but rather a reflection of euthyroid sick syndrome. Four of our patients experienced severe neutropenia, compared to only five of the 27 other cases, suggesting that the W522X mutation may be specifically associated with neutropenia. Other than the consistent features of diabetes mellitus and epiphyseal dysplasia, WRS patients are otherwise characterized by extensive phenotypic variability that correlates poorly to genotype.


Asunto(s)
Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/genética , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/genética , Mutación Puntual , eIF-2 Quinasa/genética , Epífisis/anomalías , Resultado Fatal , Femenino , Humanos , Hipotiroidismo/diagnóstico , Hipotiroidismo/genética , Lactante , Fallo Hepático/genética , Masculino , Neutropenia/diagnóstico , Neutropenia/genética , Páncreas Exocrino/enzimología , Linaje
15.
J Clin Endocrinol Metab ; 92(3): 1000-7, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17192295

RESUMEN

CONTEXT: The characteristics of P450c17 deficiency include 46,XY disorder of sex development, hypertension, hypokalemia, and lack of pubertal development. OBJECTIVE: To better understand this rare enzymatic deficiency, we analyzed the CYP17A1 gene in six affected patients. DESIGN AND PATIENTS: We examined six patients, five 46,XY, and one 46,XX (age 9-29 yr) with complete lack of masculinization (female infantile external genitalia, no uterus) and delayed puberty, respectively, and different degrees of hypertension. MAIN OUTCOME MEASUREMENTS: Genotype-phenotype correlation was measured. RESULTS: Four homozygote mutations were identified by direct sequencing of the CYP17A1 gene corresponding to an alanin 302-proline (A302P) exchange; the loss of lysine 327 (K327del); the deletion of glutamate 331 (E331del); and the replacement of arginine 416 with a histidine (R416H). Both P450c17 activities were abolished in all the mutant proteins, except one, when expressed in COS1 cells. The E331del-mutated P450c17 retained 17alpha-hydroxylase activity. The mutant proteins were normally expressed, suggesting that the loss of enzymatic activity is not due to defects of synthesis, stability, or localization of P450c17 proteins. CONCLUSION: These studies confirm lack of masculinization in 46,XY individuals as the pathognomic sign of the complete P450c17 deficiency. In XX individuals P450c17 deficiency should be considered in cases of delayed puberty. Age of onset and the severity of hypertension do not seem to be constant. Careful examination of long-term follow-ups in two of our patients suggested to us that estrogen treatment in P450c17-deficient patients might worsen the enzymatic defect, leading to aggravation of the hypertension.


Asunto(s)
Disgenesia Gonadal 46 XX/genética , Disgenesia Gonadal 46 XY/genética , Esteroide 17-alfa-Hidroxilasa/genética , Adolescente , Adulto , Animales , Presión Sanguínea , Células COS , Niño , Chlorocebus aethiops , Análisis Mutacional de ADN , Genes Reporteros , Disgenesia Gonadal 46 XX/diagnóstico por imagen , Disgenesia Gonadal 46 XX/fisiopatología , Disgenesia Gonadal 46 XY/diagnóstico por imagen , Disgenesia Gonadal 46 XY/fisiopatología , Humanos , Mutación , Progesterona/metabolismo , Radiografía , Esteroide 17-alfa-Hidroxilasa/metabolismo , Transfección
16.
J Clin Res Pediatr Endocrinol ; 9(Suppl 2): 113-122, 2017 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-29280744

RESUMEN

Traditionally, idiopathic hypogonadotropic hypogonadism (IHH) is divided into two major categories: Kallmann syndrome (KS) and normosmic IHH (nIHH). To date, inactivating variants in more than 50 genes have been reported to cause IHH. These mutations are estimated to account for up to 50% of all apparently hereditary cases. Identification of further causative gene mutations is expected to be more feasible with the increasing use of whole exome/genome sequencing. Presence of more than one IHH-associated mutant gene in a given patient/pedigree (oligogenic inheritance) is seen in 10-20% of all IHH cases. It is now well established that about 10-20% of IHH cases recover from IHH either spontaneously or after receiving some sex steroid replacement therapy. Moreover, there may be an overlap or transition between constitutional delay in growth and puberty (CDGP) and IHH. It has been increasingly observed that oligogenic inheritance and clinical recovery complicates the phenotype/genotype relationship in IHH, thus making it challenging to find new IHH-associated genes. In a clinical sense, recognizing those IHH genes and associated phenotypes may improve our diagnostic capabilities by enabling us to prioritize the screening of particular gene(s) such as synkinesia (ANOS1), dental agenesis (FGF8/FGFR1) and hearing loss (CHD7). Also, IHH-associated gene studies may be translated into new therapies such as for polycystic ovary syndrome. In a scientific sense, the most significant contribution of IHH-associated gene studies has been the characterization of the long-sought gonadotropin releasing hormone pulse generator. It appears that genetic studies of IHH will continue to advance our knowledge in both the biological and clinical domains.


Asunto(s)
Hipogonadismo/genética , Femenino , Humanos , Masculino
17.
J Clin Endocrinol Metab ; 102(6): 1816-1825, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28324054

RESUMEN

Context: Gonadotropin-releasing hormone neurons originate outside the central nervous system in the olfactory placode and migrate into the central nervous system, becoming integral components of the hypothalamic-pituitary-gonadal axis. Failure of this migration can lead to idiopathic hypogonadotropic hypogonadism (IHH)/Kallmann syndrome (KS). We have previously shown that CCDC141 knockdown leads to impaired migration of GnRH neurons but not of olfactory receptor neurons. Objective: The aim of this study was to further describe the phenotype and prevalence of CCDC141 mutations in IHH/KS. Design: Using autozygosity mapping, candidate gene screening, whole-exome sequencing, and Sanger sequencing, those individuals carrying deleterious CDCD141 variants and their phenotypes were determined in a cohort of 120 IHH/KS families. Patients and Interventions: No interventions were made. Results: Our studies revealed nine affected individuals from four independent families in which IHH/KS is associated with inactivating CCDC141 variants, revealing a prevalence of 3.3%. Affected individuals (with the exception of those from family 1 who concomitantly have FEZF1 mutations) have normal olfactory function and anatomically normal olfactory bulbs. Four affected individuals show evidence of clinical reversibility. In three of the families, there was at least one more potentially deleterious variant in other known puberty genes with evidence of allelic heterogeneity within respective pedigrees. Conclusions: These studies confirm that inactivating CCDC141 variants cause normosmic IHH but not KS. This is consistent with our previous in vitro experiments showing exclusively impaired embryonic migration of GnRH neurons upon CCDC141 knockdown. These studies expand the clinical and genetic spectrum of IHH and also attest to the complexity of phenotype and genotype in IHH.


Asunto(s)
Hipogonadismo/genética , Proteínas del Tejido Nervioso/genética , Adolescente , Adulto , Femenino , Genotipo , Humanos , Síndrome de Kallmann/genética , Masculino , Persona de Mediana Edad , Mutación , Linaje , Fenotipo , Análisis de Secuencia de ADN , Adulto Joven
18.
J Clin Res Pediatr Endocrinol ; 9(2): 95-100, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28008864

RESUMEN

OBJECTIVE: The underlying genetic etiology of hypogonadotropic hypogonadism (HH) is heterogeneous. Fibroblast growth factor signaling is pivotal in the ontogeny of gonadotropin-releasing hormone neurons. Loss-of-function mutations in FGFR1 gene cause variable HH phenotypes encompassing pubertal delay to idiopathic HH (IHH) or Kallmann syndrome (KS). As FGFR1 mutations are common, recognizing mutations and associated phenotypes may enhance clinical management. METHODS: Using a candidate gene approach, we screened 52 IHH/KS patients. RESULTS: We identified three novel (IVS3-1G>C and p.W2X, p.R209C) FGFR1 gene mutations. Despite predictive null protein function, patients from the novel mutation families had normosmic IHH without non-reproductive phenotype. CONCLUSION: These findings further emphasize the great variability of FGFR1 mutation phenotypes in IHH/KS.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Hipogonadismo/genética , Mutación , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Adolescente , Adulto , Salud de la Familia , Femenino , Genotipo , Humanos , Hipogonadismo/patología , Síndrome de Klinefelter/genética , Síndrome de Klinefelter/patología , Masculino , Linaje , Fenotipo , Pubertad Tardía/genética , Pubertad Tardía/patología , Adulto Joven
19.
Endocr Dev ; 29: 36-49, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26680571

RESUMEN

Hypogonadotropic hypogonadism (HH) often manifests as pubertal delay. A considerable proportion of cases of HH is due to genetic mutations. Recognizing those mutated genes and associated phenotypes may improve our diagnostic capabilities. GNRHR and TACR3 should be the first two genes to be screened in a clinical setting for equivocal cases such as constitutional delay in puberty versus idiopathic HH. In Kallmann syndrome (KS), according to the presence of certain accompanying clinical features, genetic screening for particular gene(s) may be prioritized: synkinesia (KAL1), dental agenesis (FGF8/FGFR1), bony anomalies (FGF8/FGFR1), and hearing loss (CHD7, SOX10). FEZF1 has recently been added to the growing list of KS genes. Also, discovery of mutations in KISS1/KISS1R and TAC3/TACR3 in kisspeptin and neurokinin B signaling, respectively, has provided major advancements in our understanding of the biology of the gonadotropin-releasing hormone pulse generator. Identification of further causative mutations accounting for the HH phenotype, which is now more feasible with the increasing popularity of whole exome sequencing, may provide deeper insight into the biology of the hypothalamic-pituitary-gonadal axis.


Asunto(s)
Gonadotropinas/deficiencia , Hipogonadismo/genética , Adolescente , Femenino , Humanos , Síndrome de Kallmann/genética , Masculino , Pubertad Tardía/genética , Maduración Sexual/genética
20.
Horm Res Paediatr ; 85(2): 107-11, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26595427

RESUMEN

INTRODUCTION: Mutations of the human GNRH1 gene are an extremely rare cause of normosmic idiopathic hypogonadotropic hypogonadism (nIHH), with only 6 mutations so far described. PATIENTS: As part of a larger study, families with IHH were screened for mutations in genes known to be associated with IHH. In family 1, a 15-year and 9-month-old boy first presented during infancy with micropenis and bilateral cryptorchidism. His pubic and axillary hair is at stage 4 and 2, respectively. His testes are 1 ml bilaterally, and his stretched penile length is 3.6 cm. In family 2, a 19-year and 2-month-old man was referred because of absence of secondary sexual characteristics. His 13-year and 8-month-old sister did not have any breast development. RESULTS: In 3 patients from 2 independent families we identified GNRH1 mutations. In the proband from family 1, a homozygous 1-base deletion (c.87delA) leading to a frameshift mutation (p.G29GfsX12) was identified. In family 2, the affected siblings had a novel homozygous mutation of c.G92A leading to p.R31H. CONCLUSION: Both mutations in these families are located in the region encoding the decapeptide and in the loci where the mutations have been described before. Therefore, these areas can be considered as mutational hot spots, indicating priority for routine diagnostic gene mutation analysis.


Asunto(s)
Hormona Liberadora de Gonadotropina/genética , Hipogonadismo/genética , Precursores de Proteínas/genética , Adolescente , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Homocigoto , Humanos , Masculino , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA