Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
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
2.
Endocrinology ; 157(5): 1956-66, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27014940

RESUMEN

The first mutation in a gene associated with a neuronal migration disorder was identified in patients with Kallmann Syndrome, characterized by hypogonadotropic hypogonadism and anosmia. This pathophysiological association results from a defect in the development of the GnRH and the olfactory system. A recent genetic screening of Kallmann Syndrome patients revealed a novel mutation in CCDC141. Little is known about CCDC141, which encodes a coiled-coil domain containing protein. Here, we show that Ccdc141 is expressed in GnRH neurons and olfactory fibers and that knockdown of Ccdc141 reduces GnRH neuronal migration. Our findings in human patients and mouse models predict that CCDC141 takes part in embryonic migration of GnRH neurons enabling them to form a hypothalamic neuronal network to initiate pulsatile GnRH secretion and reproductive function.


Asunto(s)
Movimiento Celular/genética , Hormona Liberadora de Gonadotropina/metabolismo , Síndrome de Kallmann/genética , Mutación , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Animales , Humanos , Ratones , Proteínas del Tejido Nervioso/fisiología , Neuronas/citología
3.
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
4.
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
5.
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
6.
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
7.
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
8.
J Clin Endocrinol Metab ; 99(10): E2067-75, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25033069

RESUMEN

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


Asunto(s)
Ataxia Cerebelosa/genética , Hormona Liberadora de Gonadotropina/deficiencia , Hipogonadismo/genética , Degeneración Nerviosa/genética , Fosfolipasas/genética , Pubertad Tardía/genética , Adolescente , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Ataxia Cerebelosa/metabolismo , Salud de la Familia , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Homeostasis/genética , Humanos , Hipogonadismo/metabolismo , Masculino , Persona de Mediana Edad , Degeneración Nerviosa/metabolismo , Linaje , Fosfolipasas/metabolismo , Fosfolípidos/metabolismo , Pubertad Tardía/metabolismo
9.
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
10.
J Clin Res Pediatr Endocrinol ; 4(3): 121-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22766261

RESUMEN

OBJECTIVE: Normosmic idiopathic hypogonadotropic hypogonadism (nIHH) is characterized by failure of initiation or maintenance of puberty due to insufficient gonadotropin release, which is not associated with anosmia/hyposmia. The objective of this study was to determine the distribution of causative mutations in a hereditary form of nIHH. METHODS: In this prospective collaborative study, 22 families with more than one affected individual (i.e. multiplex families) with nIHH were recruited and screened for genes known or suspected to be strong candidates for nIHH. RESULTS: Mutations were identified in five genes (GNRHR, TACR3, TAC3, KISS1R, and KISS1) in 77% of families with autosomal recessively inherited nIHH. GNRHR and TACR3 mutations were the most common two causative mutations occurring with about equal frequency. CONCLUSIONS: Mutations in these five genes account for about three quarters of the causative mutations in nIHH families with more than one affected individual. This frequency is significantly greater than the previously reported rates in all inclusive (familial plus sporadic) cohorts. GNRHR and TACR3 should be the first two genes to be screened for diagnostic purposes. Identification of causative mutations in the remaining families will shed light on the regulation of puberty.


Asunto(s)
Salud de la Familia , Hipogonadismo/genética , Mutación , Receptores LHRH/genética , Receptores de Neuroquinina-3/genética , Adolescente , Adulto , Estudios de Cohortes , Estudios de Asociación Genética , Humanos , Hipogonadismo/congénito , Hipogonadismo/metabolismo , Lactante , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Neuroquinina B/genética , Neuroquinina B/metabolismo , Estudios Prospectivos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Receptores LHRH/metabolismo , Receptores de Neuroquinina-3/metabolismo , Taquicininas/genética , Taquicininas/metabolismo , Turquía , Adulto Joven
11.
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
12.
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
13.
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
14.
J Clin Endocrinol Metab ; 94(10): 3633-3639, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19755480

RESUMEN

CONTEXT: The neurokinin B (NKB) receptor, encoded by TACR3, is widely expressed within the central nervous system, including hypothalamic nuclei involved in regulating GnRH release. We have recently reported two mutations in transmembrane segments of the receptor and a missense mutation in NKB in patients with normosmic isolated hypogonadotropic hypogonadism (nIHH). PATIENTS AND METHODS: We sequenced the TACR3 gene in a family in which three siblings had nIHH. The novel mutant receptor thus identified was studied in a heterologous expression system using calcium flux as the functional readout. RESULTS: All affected siblings were homozygous for the His148Leu mutation, in the first extracellular loop of the NKB receptor. The His148Leu mutant receptor exhibited profoundly impaired signaling in response to NKB (EC(50) = 3 +/- 0.1 nm and >5 microm for wild-type and His148Leu, respectively). The location of the mutation in an extracellular part of the receptor led us also to test whether senktide, a synthetic NKB analog, may retain ability to stimulate the mutant receptor. However, the signaling activity of the His148Leu receptor in response to senktide was also severely impaired (EC(50) = 1 +/- 1 nm for wild-type and no significant response of His148Leu to 10 microm). CONCLUSIONS: Homozygosity for the TACR3 His148Leu mutation leads to failure of sexual maturation in humans, whereas signaling by the mutant receptor in vitro in response to either NKB or senktide is severely impaired. These observations further strengthen the link between NKB, the NKB receptor, and regulation of human reproductive function.


Asunto(s)
Amenorrea/genética , Hipogonadismo/genética , Mutación Missense , Neuroquinina B/análogos & derivados , Fragmentos de Péptidos/uso terapéutico , Receptores de Neuroquinina-3/genética , Sustancia P/análogos & derivados , Adolescente , Adulto , Mama/anomalías , Análisis Mutacional de ADN , Femenino , Histidina , Homocigoto , Hormonas/sangre , Humanos , Hipogonadismo/sangre , Leucina , Masculino , Neuroquinina B/genética , Linaje , Fragmentos de Péptidos/farmacología , Pubertad/sangre , Pubertad/genética , Hermanos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sustancia P/farmacología , Sustancia P/uso terapéutico
15.
Nat Genet ; 41(3): 354-358, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19079066

RESUMEN

The timely secretion of gonadal sex steroids is essential for the initiation of puberty, the postpubertal maintenance of secondary sexual characteristics and the normal perinatal development of male external genitalia. Normal gonadal steroid production requires the actions of the pituitary-derived gonadotropins, luteinizing hormone and follicle-stimulating hormone. We report four human pedigrees with severe congenital gonadotropin deficiency and pubertal failure in which all affected individuals are homozygous for loss-of-function mutations in TAC3 (encoding Neurokinin B) or its receptor TACR3 (encoding NK3R). Neurokinin B, a member of the substance P-related tachykinin family, is known to be highly expressed in hypothalamic neurons that also express kisspeptin, a recently identified regulator of gonadotropin-releasing hormone secretion. These findings implicate Neurokinin B as a critical central regulator of human gonadal function and suggest new approaches to the pharmacological control of human reproduction and sex hormone-related diseases.


Asunto(s)
Hipogonadismo/genética , Neuroquinina B/genética , Neuroquinina B/fisiología , Receptores de Neuroquinina-3/genética , Reproducción/genética , Secuencia de Aminoácidos , Cromosomas Humanos Par 4 , Análisis Mutacional de ADN , Familia , Gónadas/metabolismo , Gónadas/fisiología , Humanos , Kisspeptinas , Modelos Biológicos , Mutación/fisiología , Neuroquinina B/metabolismo , Neuronas/metabolismo , Linaje , Receptores de Neuroquinina-3/metabolismo , Reproducción/fisiología , Homología de Secuencia de Aminoácido , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...