Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Clin Endocrinol Metab ; 93(3): 974-80, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18029459

RESUMEN

CONTEXT: A polymorphism of the GH receptor (GHR) gene resulting in genomic deletion of exon 3 (GHR-d3) has been associated with responsiveness to GH therapy. However, the data reported so far do vary according to the underlying condition, replacement dose, and duration of the treatment. OBJECTIVE, DESIGN: The aim of this study was to analyze the impact of the GHR genotypes in terms of the initial height velocity (HV) resulting from treatment and the impact upon adult height in patients suffering from severe isolated GH deficiency. CONTROLS, PATIENTS, SETTING: A total of 181 subjects (peak stimulated GH

Asunto(s)
Estatura/efectos de los fármacos , Exones , Hormona del Crecimiento/uso terapéutico , Terapia de Reemplazo de Hormonas , Hormona de Crecimiento Humana/deficiencia , Receptores de Somatotropina/genética , Adulto , Femenino , Eliminación de Gen , Genotipo , Humanos , Modelos Lineales , Masculino , Persona de Mediana Edad , Isoformas de Proteínas
2.
Endocrinology ; 148(1): 45-53, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17038549

RESUMEN

The majority of mutations that cause isolated GH deficiency type II (IGHD II) affect splicing of GH-1 transcripts and produce a dominant-negative GH isoform lacking exon 3 resulting in a 17.5-kDa isoform, which further leads to disruption of the GH secretory pathway. A clinical variability in the severity of the IGHD II phenotype depending on the GH-1 gene alteration has been reported, and in vitro and transgenic animal data suggest that the onset and severity of the phenotype relates to the proportion of 17.5-kDa produced. The removal of GH in IGHD creates a positive feedback loop driving more GH expression, which may itself increase 17.5-kDa isoform productions from alternate splice sites in the mutated GH-1 allele. In this study, we aimed to test this idea by comparing the impact of stimulated expression by glucocorticoids on the production of different GH isoforms from wild-type (wt) and mutant GH-1 genes, relying on the glucocorticoid regulatory element within intron 1 in the GH-1 gene. AtT-20 cells were transfected with wt-GH or mutated GH-1 variants (5'IVS-3 + 2-bp T->C; 5'IVS-3 + 6 bp T->C; ISEm1: IVS-3 + 28 G->A) known to cause clinical IGHD II of varying severity. Cells were stimulated with 1 and 10 mum dexamethasone (DEX) for 24 h, after which the relative amounts of GH-1 splice variants were determined by semiquantitative and quantitative (TaqMan) RT-PCR. In the absence of DEX, only around 1% wt-GH-1 transcripts were the 17.5-kDa isoform, whereas the three mutant GH-1 variants produced 29, 39, and 78% of the 17.5-kDa isoform. DEX stimulated total GH-1 gene transcription from all constructs. Notably, however, DEX increased the amount of 17.5-kDa GH isoform relative to the 22- and 20-kDa isoforms produced from the mutated GH-1 variants, but not from wt-GH-1. This DEX-induced enhancement of 17.5-kDa GH isoform production, up to 100% in the most severe case, was completely blocked by the addition of RU486. In other studies, we measured cell proliferation rates, annexin V staining, and DNA fragmentation in cells transfected with the same GH-1 constructs. The results showed that that the 5'IVS-3 + 2-bp GH-1 gene mutation had a more severe impact on those measures than the splice site mutations within 5'IVS-3 + 6 bp or ISE +28, in line with the clinical severity observed with these mutations. Our findings that the proportion of 17.5-kDa produced from mutant GH-1 alleles increases with increased drive for gene expression may help to explain the variable onset progression, and severity observed in IGHD II.


Asunto(s)
Apoptosis/fisiología , Trastornos del Crecimiento/genética , Hormona de Crecimiento Humana/genética , Hipófisis/citología , Sitios de Empalme de ARN/fisiología , Animales , División Celular/fisiología , Células Cultivadas , Fragmentación del ADN , Dexametasona/farmacología , Exones/genética , Genes Dominantes , Glucocorticoides/farmacología , Trastornos del Crecimiento/fisiopatología , Hormona de Crecimiento Humana/deficiencia , Humanos , Ratones , Mutagénesis Sitio-Dirigida , Mutación/fisiología , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología , Transfección
3.
J Clin Endocrinol Metab ; 92(8): 2893-901, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17519310

RESUMEN

CONTEXT AND OBJECTIVE: A single missense mutation in the GH-1 gene converting codon 77 from arginine (R) to cysteine (C) yields a mutant GH-R77C peptide, which was described as natural GH antagonist. DESIGN, SETTING, AND PATIENTS: Heterozygosity for GH-R77C/wt-GH was identified in a Syrian family. The index patient, a boy, was referred for assessment of his short stature (-2.5 SD score) and partial GH insensitivity was diagnosed. His mother and grandfather were also carrying the same mutation and showed partial GH insensitivity with modest short stature. INTERVENTIONS AND RESULTS: Functional characterization of the GH-R77C was performed through studies of GH receptor binding and activation of Janus kinase 2/Stat5 pathway. No differences in the binding affinity and bioactivity between wt-GH and GH-R77C were found. Similarly, cell viability and proliferation after expression of both GH peptides in AtT-20 cells were identical. Quantitative confocal microscopy analysis revealed no significant difference in the extent of subcellular colocalization between wt-GH and GH-R77C with endoplasmic reticulum, Golgi, or secretory vesicles. Furthermore studies demonstrated a reduced capability of GH-R77C to induce GHR/GHBP gene transcription rate when compared with wt-GH. CONCLUSION: Reduced GH receptor/GH-binding protein expression might be a possible cause for the partial GH insensitivity with delay in growth and pubertal development found in our patients. In addition, this group of patients deserves further attention because they could represent a distinct clinical entity underlining that an altered GH peptide may also have a direct impact on GHR/GHBP gene expression causing partial GH insensitivity.


Asunto(s)
Estatura/efectos de los fármacos , Hormona del Crecimiento/uso terapéutico , Hormona de Crecimiento Humana/antagonistas & inhibidores , Hormona de Crecimiento Humana/genética , Alelos , Animales , Proliferación Celular , Células Cultivadas , Niño , Técnica del Anticuerpo Fluorescente , Genes Reporteros , Vectores Genéticos , Crecimiento/fisiología , Heterocigoto , Hormona de Crecimiento Humana/sangre , Humanos , Focalización Isoeléctrica , Luciferasas/genética , Masculino , Ratones , Microscopía Confocal , Mutación , Linaje , Pubertad/fisiología , Receptores de Somatotropina/metabolismo , Transducción de Señal/fisiología
4.
J Clin Endocrinol Metab ; 92(11): 4427-35, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17726075

RESUMEN

CONTEXT AND OBJECTIVE: Alteration of exon splice enhancers (ESE) may cause autosomal dominant GH deficiency (IGHD II). Disruption analysis of a (GAA) (n) ESE motif within exon 3 by introducing single-base mutations has shown that single nucleotide mutations within ESE1 affect pre-mRNA splicing. DESIGN, SETTING, AND PATIENTS: Confirming the laboratory-derived data, a heterozygous splice enhancer mutation in exon 3 (exon 3 + 2 A-->C) coding for GH-E32A mutation of the GH-1 gene was found in two independent pedigrees, causing familial IGHD II. Because different ESE mutations have a variable impact on splicing of exon 3 of GH and therefore on the expression of the 17.5-kDa GH mutant form, the GH-E32A was studied at the cellular level. INTERVENTIONS AND RESULTS: The splicing of GH-E32A, assessed at the protein level, produced significantly increased amounts of 17.5-kDa GH isoform (55% of total GH protein) when compared with the wt-GH. AtT-20 cells coexpressing both wt-GH and GH-E32A presented a significant reduction in cell proliferation as well as GH production after forskolin stimulation when compared with the cells expressing wt-GH. These results were complemented with confocal microscopy analysis, which revealed a significant reduction of the GH-E32A-derived isoform colocalized with secretory granules, compared with wt-GH. CONCLUSION: GH-E32A mutation found within ESE1 weakens recognition of exon 3 directly, and therefore, an increased production of the exon 3-skipped 17.5-kDa GH isoform in relation to the 22-kDa, wt-GH isoform was found. The GH-E32A mutant altered stimulated GH production as well as cell proliferation, causing IGHD II.


Asunto(s)
Trastornos de los Cromosomas/genética , Exones/genética , Hormona de Crecimiento Humana/deficiencia , Mutación/fisiología , Adolescente , Adulto , Western Blotting , Estatura , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Colforsina/farmacología , Retículo Endoplásmico/ultraestructura , Femenino , Genes Dominantes/genética , Vectores Genéticos/genética , Aparato de Golgi/ultraestructura , Humanos , Masculino , Microscopía Confocal , Linaje , Isoformas de Proteínas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Vesículas Secretoras/ultraestructura
5.
Eur J Endocrinol ; 157 Suppl 1: S67-74, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17785701

RESUMEN

A heterozygous missense mutation in the GH-1 gene converting codon 77 from arginine (R) to cysteine (C), which was previously reported to have some GH antagonistic effect, was identified in a Syrian family. The index patient, a boy, was referred for assessment of his short stature (-2.5 SDS) at the age of 6 years. His mother and grandfather were also carrying the same mutation, but did not differ in adult height from the other unaffected family members. Hormonal examination in all affected subjects revealed increased basal GH, low IGF-I concentrations, and subnormal IGF-I response in generation test leading to the diagnosis of partial GH insensitivity. However, GH receptor gene (GHR) sequencing demonstrated no abnormalities. As other family members carrying the GH-R77C form showed similar alterations at the hormonal level, but presented with normal final height, no GH therapy was given to the boy, but he was followed through his pubertal development which was delayed. At the age of 20 years he reached his final height, which was normal within his parental target height. Functional characterization of the GH-R77C, assessed through activation of Jak2/Stat5 pathway, revealed no differences in the bioactivity between wild-type-GH (wt-GH) and GH-R77C. Detailed structural analysis indicated that the structure of GH-R77C, in terms of disulfide bond formation, is almost identical to that of the wt-GH despite the introduced mutation (Cys77). Previous studies from our group demonstrated a reduced capability of GH-R77C to induce GHR/GH-binding protein (GHBP) gene transcription rate when compared with wt-GH. Therefore, reduced GHR/GHBP expression might well be the possible cause for the partial GH insensitivity found in our patients. In addition, this group of patients deserve further attention because they could represent a distinct clinical entity underlining that an altered GH peptide may also have a direct impact on GHR/GHBP gene expression causing partial GH insensitivity. This might be responsible for the delay of growth and pubertal development. Finally, we clearly demonstrate that GH-R77C is not invariably associated with short stature, but that great care needs to be taken in ascribing growth failure to various heterozygous mutations affecting the GH-IGF axis and that careful functional studies are mandatory.


Asunto(s)
Trastornos del Crecimiento/genética , Hormona de Crecimiento Humana/genética , Mutación Missense , Pubertad Tardía/genética , Adolescente , Adulto , Arginina , Niño , Cisteína , Estudios de Seguimiento , Trastornos del Crecimiento/sangre , Hormona de Crecimiento Humana/sangre , Hormona de Crecimiento Humana/química , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Persona de Mediana Edad , Linaje , Pubertad Tardía/sangre
6.
Endocr Dev ; 23: 109-20, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23182825

RESUMEN

Isolated growth hormone deficiency type-2 (IGHD-2), the autosomal-dominant form of GH deficiency, is mainly caused by specific splicing mutations in the human growth hormone (hGH) gene (GH-1). These mutations, occurring in and around exon 3, cause complete exon 3 skipping and produce a dominant-negative 17.5 kD GH isoform that reduces the accumulation and secretion of wild type-GH (wt-GH). At present, patients suffering from IGHD-2 are treated with daily injections of recombinant human GH (rhGH) in order to reach normal height. However, this type of replacement therapy, although effective in terms of growth, does not prevent toxic effects of the 17.5-kD mutant on the pituitary gland, which can eventually lead to other hormonal deficiencies. Considering a well-known correlation between the clinical severity observed in IGHD-2 patients and the increased expression of the 17.5-kD isoform, therapies that specifically target this isoform may be useful in patients with GH-1 splicing defects. This chapter focuses on molecular strategies that could represent future directions for IGHD-2 treatment.


Asunto(s)
Enanismo Hipofisario/genética , Enanismo Hipofisario/terapia , Hormona de Crecimiento Humana/genética , Animales , Terapia Genética/métodos , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Hormona Liberadora de Hormona del Crecimiento/fisiología , Terapia de Reemplazo de Hormonas/métodos , Hormona de Crecimiento Humana/administración & dosificación , Hormona de Crecimiento Humana/metabolismo , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/fisiología , Interferencia de ARN/fisiología , Empalme del ARN/genética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico
7.
Horm Res Paediatr ; 75(1): 63-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21252559

RESUMEN

In the last decade, few areas of biology have been transformed as thoroughly as RNA molecular biology. Without any doubt, one of the most significant advances has been the discovery of small (20-30 nucleotide) noncoding RNAs that regulate genes and genomes. The effects of small RNAs on gene expression and control are generally inhibitory, and the corresponding regulatory mechanisms are therefore collectively subsumed under the heading of RNA silencing and/or RNA interference. Two primary categories of these small RNAs - short interfering RNAs (siRNAs) and microRNAs (miRNAs) - act in both somatic and germline lineages of eukaryotic species to regulate endogenous genes and to defend the genome from invasive nucleic acids. Recent advances have revealed unexpected diversity in their biogenesis pathways and the regulatory mechanisms that they access. Our understanding of siRNA and miRNA-based regulation has direct implications for fundamental biology as well as disease aetiology and treatment as it is discussed in this review on 'new techniques in molecular biology'.


Asunto(s)
Interferencia de ARN , Animales , Silenciador del Gen , Técnicas de Transferencia de Gen/efectos adversos , Terapia Genética/métodos , Terapia Genética/tendencias , Humanos , MicroARNs/biosíntesis , MicroARNs/uso terapéutico , Biología Molecular/métodos , Biología Molecular/tendencias , Oligonucleótidos Antisentido/uso terapéutico , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/uso terapéutico , ARN no Traducido/uso terapéutico
8.
Endocrinology ; 151(9): 4400-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20591972

RESUMEN

Isolated GH deficiency type II (IGHD II) is the autosomal dominant form of GHD. In the majority of the cases, this disorder is due to specific GH-1 gene mutations that lead to mRNA missplicing and subsequent loss of exon 3 sequences. When misspliced RNA is translated, it produces a toxic 17.5-kDa GH (Delta3GH) isoform that reduces the accumulation and secretion of wild-type-GH. At present, patients suffering from this type of disease are treated with daily injections of recombinant human GH in order to maintain normal growth. However, this type of replacement therapy does not prevent toxic effects of the Delta3GH mutant on the pituitary gland, which can eventually lead to other hormonal deficiencies. We developed a strategy involving Delta3GH isoform knockdown mediated by expression of a microRNA-30-adapted short hairpin RNA (shRNA) specifically targeting the Delta3GH mRNA of human (shRNAmir-Delta3). Rat pituitary tumor GC cells expressing Delta3GH upon doxycycline induction were transduced with shRNAmir-Delta3 lentiviral vectors, which significantly reduced Delta3GH protein levels and improved human wild-type-GH secretion in comparison with a shRNAmir targeting a scrambled sequence. No toxicity due to shRNAmir expression could be observed in cell proliferation assays. Confocal microscopy strongly suggested that shRNAmir-Delta3 enabled the recovery of GH granule storage and secretory capacity. These viral vectors have shown their ability to stably integrate, express shRNAmir, and rescue IGHD II phenotype in rat pituitary tumor GC cells, a methodology that opens new perspectives for the development of gene therapy to treat IGHD patients.


Asunto(s)
Hormona del Crecimiento/deficiencia , Hormona del Crecimiento/genética , Mutación , Hipófisis/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Técnicas de Inactivación de Genes , Vectores Genéticos/genética , Hormona del Crecimiento/metabolismo , Humanos , Lentivirus/genética , MicroARNs/genética , Microscopía Confocal , Hipófisis/patología , ARN Interferente Pequeño/genética , Ratas , Transfección/métodos
9.
Endocrinology ; 151(6): 2650-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20351314

RESUMEN

An autosomal dominant form of isolated GH deficiency (IGHD II) can result from heterozygous splice site mutations that weaken recognition of exon 3 leading to aberrant splicing of GH-1 transcripts and production of a dominant-negative 17.5-kDa GH isoform. Previous studies suggested that the extent of missplicing varies with different mutations and the level of GH expression and/or secretion. To study this, wt-hGH and/or different hGH-splice site mutants (GH-IVS+2, GH-IVS+6, GH-ISE+28) were transfected in rat pituitary cells expressing human GHRH receptor (GC-GHRHR). Upon GHRH stimulation, GC-GHRHR cells coexpressing wt-hGH and each of the mutants displayed reduced hGH secretion and intracellular GH content when compared with cells expressing only wt-hGH, confirming the dominant-negative effect of 17.5-kDa isoform on the secretion of 22-kDa GH. Furthermore, increased amount of 17.5-kDa isoform produced after GHRH stimulation in cells expressing GH-splice site mutants reduced production of endogenous rat GH, which was not observed after GHRH-induced increase in wt-hGH. In conclusion, our results support the hypothesis that after GHRH stimulation, the severity of IGHD II depends on the position of splice site mutation leading to the production of increasing amounts of 17.5-kDa protein, which reduces the storage and secretion of wt-GH in the most severely affected cases. Due to the absence of GH and IGF-I-negative feedback in IGHD II, a chronic up-regulation of GHRH would lead to an increased stimulatory drive to somatotrophs to produce more 17.5-kDa GH from the severest mutant alleles, thereby accelerating autodestruction of somatotrophs in a vicious cycle.


Asunto(s)
Expresión Génica/efectos de los fármacos , Hormona Liberadora de Hormona del Crecimiento/farmacología , Hormona del Crecimiento/metabolismo , Hormona de Crecimiento Humana/genética , Isoformas de Proteínas/metabolismo , Animales , Western Blotting , Línea Celular , Expresión Génica/genética , Hormona del Crecimiento/genética , Hormona de Crecimiento Humana/metabolismo , Humanos , Mutación , Isoformas de Proteínas/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Clin Endocrinol Metab ; 95(2): 731-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19952226

RESUMEN

CONTEXT AND OBJECTIVE: Main features of the autosomal dominant form of GH deficiency (IGHD II) include markedly reduced secretion of GH combined with low concentrations of IGF-I leading to short stature. DESIGN, SETTING, AND PATIENTS: A female patient presented with short stature (height -6.0 sd score) and a delayed bone age of 2 yr at the chronological age of 5 yr. Later, at the age of 9 yr, GHD was confirmed by standard GH provocation test, which revealed subnormal concentrations of GH and a very low IGF-I. Genetic analysis of the GH-1 gene revealed the presence of a heterozygous R178H mutation. INTERVENTIONS AND RESULTS: AtT-20 cells coexpressing both wt-GH and GH-R178H showed a reduced GH secretion after forskolin stimulation compared with the cells expressing only wt-GH, supporting the diagnosis of IGHD II. Because reduced GH concentrations found in the circulation of our untreated patient could not totally explain her severe short stature, functional characterization of the GH-R178H performed by studies of GH receptor binding and activation of the Janus kinase-2/signal transducer and activator of transcription-5 pathway revealed a reduced binding affinity of GH-R178H for GH receptor and signaling compared with the wt-GH. CONCLUSION: This is the first report of a patient suffering from short stature caused by a GH-1 gene alteration affecting not only GH secretion (IGHD II) but also GH binding and signaling, highlighting the necessity of functional analysis of any GH variant, even in the alleged situation of IGHD II.


Asunto(s)
Hormona de Crecimiento Humana/deficiencia , Hormona de Crecimiento Humana/genética , Mutación , Animales , Células CHO , Células Cultivadas , Niño , Cricetinae , Cricetulus , Femenino , Hormona de Crecimiento Humana/metabolismo , Humanos , Janus Quinasa 2/fisiología , Ratones , Simulación de Dinámica Molecular , Receptores de Somatotropina/metabolismo , Factor de Transcripción STAT5/fisiología , Zinc/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA