Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Endocrinology ; 157(10): 3972-3982, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27253996

RESUMO

Isolated GH deficiency (IGHD) type II, the autosomal dominant form of GHD, is mainly caused by mutations that affect splicing of GH-1. When misspliced RNA is translated, it produces a toxic 17.5-kDa GH isoform that reduces the accumulation and secretion of wild-type-human GH (wt-hGH). Usually, isolated GHD type II patients 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 17.5-kDa GH isoform on the pituitary gland, which can eventually lead to other hormonal deficiencies. Here, we tested the possibility to restore the constitutive splicing pattern of GH-1 by using butyrate, a drug that mainly acts as histone deacetylase inhibitor. To this aim, wt-hGH and/or different hGH-splice site mutants (GH-IVS3+2, GH-IVS3+6, and GH-ISE+28) were transfected in rat pituitary cells expressing human GHRH receptor (GHRHR) (GC-GHRHR). Upon butyrate treatment, GC-GHRHR cells coexpressing wt-hGH and each of the mutants displayed increased GH transcript level, intracellular GH content, and GH secretion when compared with the corresponding untreated condition. The effect of butyrate was most likely mediated by the alternative splicing factor/splicing factor 2. Overexpression of alternative ASF/SF2 in the same experimental setting, indeed, promoted the amount of full-length transcripts thus increasing synthesis and secretion of the 22-kDa GH isoform. In conclusion, our results support the hypothesis that modulation of GH-1 splicing pattern to increase the 22-kDa GH isoform levels can be clinically beneficial and hence a crucial challenge in GHD research.


Assuntos
Butiratos/uso terapêutico , Nanismo Hipofisário/tratamento farmacológico , Hormônio do Crescimento/metabolismo , Splicing de RNA/efeitos dos fármacos , Animais , Butiratos/farmacologia , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Técnicas de Transferência de Genes , Hormônio do Crescimento/genética , Humanos , Ratos , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo
2.
PLoS One ; 9(10): e107388, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25310566

RESUMO

Butyrate is a short-chain fatty acid (SCFA) closely related to the ketone body ß-hydroxybutyrate (BHB), which is considered to be the major energy substrate during prolonged exercise or starvation. During fasting, serum growth hormone (GH) rises concomitantly with the accumulation of BHB and butyrate. Interactions between GH, ketone bodies and SCFA during the metabolic adaptation to fasting have been poorly investigated to date. In this study, we examined the effect of butyrate, an endogenous agonist for the two G-protein-coupled receptors (GPCR), GPR41 and 43, on non-stimulated and GH-releasing hormone (GHRH)-stimulated hGH secretion. Furthermore, we investigated the potential role of GPR41 and 43 on the generation of butyrate-induced intracellular Ca2+ signal and its ultimate impact on hGH secretion. To study this, wt-hGH was transfected into a rat pituitary tumour cell line stably expressing the human GHRH receptor. Treatment with butyrate promoted hGH synthesis and improved basal and GHRH-induced hGH-secretion. By acting through GPR41 and 43, butyrate enhanced intracellular free cytosolic Ca2+. Gene-specific silencing of these receptors led to a partial inhibition of the butyrate-induced intracellular Ca2+ rise resulting in a decrease of hGH secretion. This study suggests that butyrate is a metabolic intermediary, which contributes to the secretion and, therefore, to the metabolic actions of GH during fasting.


Assuntos
Ácido Butírico/farmacologia , Cálcio/metabolismo , Hormônio do Crescimento/metabolismo , Adeno-Hipófise/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Animais , Linhagem Celular , Inativação Gênica , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , RNA Interferente Pequeno , Ratos , Receptores Acoplados a Proteínas G/genética
3.
Horm Res Paediatr ; 82(4): 245-51, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25196974

RESUMO

BACKGROUND: Aggregation of growth hormone (GH) required for its proper storage in granules is facilitated by zinc (Zn(2+)) transported by specific zinc transporters in and out of the regulated secretory pathway. Slc30a5 (ZnT5) was reported to have the highest gene expression among all zinc transporters in primary mouse pituitary cells while ZnT5-null mice presented with abnormal bone development and impaired growth compared to wild-type counterparts. METHODS: In vitro studies performed in GH3 cells, a rat pituitary cell line that endogenously produces rat GH (rGH), included analysis of: cytoplasmic Zn(2+) pool changes after altering rSlc30a5 expression (luciferase assay), rZnT5 association with different compartments of the regulated secretory pathway (confocal microscopy), and the rGH secretion after rSlc30a5 knock-down (Western blot). RESULTS: Confocal microscopy demonstrated high co-localization of rZnT5 with ER and Golgi (early secretory pathway) while siRNA-mediated knock-down of rSlc30a5 gene expression led to a significant reduction in rGH secretion. Furthermore, altered expression of rSlc30a5 (knock-down/overexpression) evoked changes in the cytoplasmic Zn(2+) pool indicating its important role in mediating Zn(2+) influx into intracellular compartments of the regulated secretory pathway. CONCLUSION: Taken together, these results suggest that ZnT5 might play an important role in regulated GH secretion that is much greater than previously anticipated.


Assuntos
Proteínas de Transporte de Cátions/genética , Hormônio do Crescimento/metabolismo , Hipófise/metabolismo , Via Secretória/genética , Zinco/metabolismo , Animais , Linhagem Celular , Citoplasma/metabolismo , DNA Complementar/genética , Técnicas de Silenciamento de Genes , Hipófise/citologia , RNA Interferente Pequeno/genética , Ratos
4.
J Pediatr Endocrinol Metab ; 27(3-4): 329-34, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24150201

RESUMO

Several monogenic defects have been reported to be associated with idiopathic short stature. Focusing on growth hormone receptor (GHR)-gene alterations, the heterozygosity of the same gene defect may be associated with a range of growth deficits. We found a heterozygous mutation (V144I) within exon 6 of the GHR gene in a patient with a low level of insulin-like growth factor I (IGF-I), normal level of GH, and severe short stature. Despite the lack of statistical difference, an overall tendency for reduced wt-GH-induction of GHR activation and Jak/Stat signalling in cells transiently expressing GHR-V144I alone or co-expressing wt-GHR compared to cells expressing only wt-GHR was found when GH doses were increased. Our results suggest that, although GHR sequence variants are responsible for some functional alterations commonly observed in children with idiopathic short stature, these changes may not explain all the height deficits observed in these subjects.


Assuntos
Estatura/genética , Heterozigoto , Mutação , Receptores da Somatotropina/genética , Pré-Escolar , Feminino , Células HEK293 , Humanos
5.
Horm Res Paediatr ; 80(6): 381-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24296719

RESUMO

Human growth hormone (GH) causes a variety of physiological and metabolic effects in humans and plays a pivotal role in postnatal growth. In somatotroph cells of the anterior pituitary, GH is stored in concentrated forms in secretory granules to be rapidly released upon GH-releasing hormone stimulation. During the process of secretory granule biogenesis, self-association of GH occurs in the compartments of the early secretory pathway (endoplasmic reticulum and Golgi complex). Since this process is greatly facilitated by the presence of zinc ions, it is of importance to understand the potential role of zinc transporters that participate in the fine-tuning of zinc homeostasis and dynamics, particularly in the early secretory pathway. Thus, the role of zinc transporters in supplying the secretory pathway with the sufficient amount of zinc required for the biogenesis of GH-containing secretory granules is essential for normal secretion. This report, illustrated by a clinical case report on transient neonatal zinc deficiency, focuses on the role of zinc in GH storage in the secretory granules and highlights the role of specific zinc transporters in the early secretory pathway. © 2013 S. Karger AG, Basel.


Assuntos
Hormônio do Crescimento Humano/metabolismo , Zinco/fisiologia , Animais , Transtornos do Crescimento/etiologia , Transtornos do Crescimento/metabolismo , Humanos , Recém-Nascido , Erros Inatos do Metabolismo dos Metais/diagnóstico , Erros Inatos do Metabolismo dos Metais/etiologia , Erros Inatos do Metabolismo dos Metais/metabolismo , Leite Humano/química , Leite Humano/metabolismo , Zinco/deficiência , Zinco/metabolismo
6.
Int J Endocrinol ; 2013: 259189, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24194756

RESUMO

Suboptimal dietary zinc (Zn(2+)) intake is increasingly appreciated as an important public health issue. Zn(2+) is an essential mineral, and infants are particularly vulnerable to Zn(2+) deficiency, as they require large amounts of Zn(2+) for their normal growth and development. Although term infants are born with an important hepatic Zn(2+) storage, adequate Zn(2+) nutrition of infants mostly depends on breast milk or formula feeding, which contains an adequate amount of Zn(2+) to meet the infants' requirements. An exclusively breast-fed 6 months old infant suffering from Zn(2+) deficiency caused by an autosomal dominant negative G87R mutation in the Slc30a2 gene (encoding for the zinc transporter 2 (ZnT-2)) in the mother is reported. More than 20 zinc transporters characterized up to date, classified into two families (Slc30a/ZnT and Slc39a/Zip), reflect the complexity and importance of maintaining cellular Zn(2+) homeostasis and dynamics. The role of ZnTs is to reduce intracellular Zn(2+) by transporting it from the cytoplasm into various intracellular organelles and by moving Zn(2+) into extracellular space. Zips increase intracellular Zn(2+) by transporting it in the opposite direction. Thus the coordinated action of both is essential for the maintenance of Zn(2+) homeostasis in the cytoplasm, and accumulating evidence suggests that this is also true for the secretory pathway of growth hormone.

7.
Endocrinology ; 154(11): 4215-25, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23970781

RESUMO

Endocrine cells store hormones in concentrated forms (aggregates) in dense-core secretory granules that are released upon appropriate stimulation. Zn(2+) binding to GH through amino acid residues His18, His21, and Glu174 are essential for GH dimerization and might mediate its aggregation and storage in secretory granules. To investigate whether GH-1 gene mutations at these positions interfere with this process, GH secretion and intracellular production were analyzed in GC cells (rat pituitary cell line) transiently expressing wt-GH and/or GH Zn mutant (GH-H18A-H21A-E174A) in forskolin-stimulated vs nonstimulated conditions. Reduced secretion of the mutant variant (alone or coexpressed with wt-GH) compared with wt-GH after forskolin stimulation was observed, whereas an increased intracellular accumulation of GH Zn mutant vs wt-GH correlates with its altered extracellular secretion. Depleting Zn(2+) from culture medium using N,N,N',N'-tetrakis(2-pyridylemethyl)ethylenediamine, a high-affinity Zn(2+) chelator, led to a significant reduction of the stimulated wt-GH secretion. Furthermore, externally added Zn(2+) to culture medium increased intracellular free Zn(2+) levels and recovered wt-GH secretion, suggesting its direct dependence on free Zn(2+) levels after forskolin stimulation. Confocal microscopy analysis of the intracellular secretory pathway of wt-GH and GH Zn mutant indicated that both variants pass through the regulated secretory pathway in a similar manner. Taken together, our data support the hypothesis that loss of affinity of GH to Zn(2+) as well as altering intracellular free Zn(2+) content may interfere with normal GH dimerization (aggregation) and storage of the mutant variant (alone or with wt-GH), which could possibly explain impaired GH secretion.


Assuntos
Hormônio do Crescimento/metabolismo , Zinco/metabolismo , Animais , Apoptose , Linhagem Celular , Sobrevivência Celular , Meios de Cultura , Etilaminas , Hormônio do Crescimento/química , Hormônio do Crescimento/genética , Humanos , Camundongos , Mutação , Ligação Proteica , Piridinas , Ratos , Fatores de Tempo , Zinco/administração & dosagem , Zinco/farmacologia
8.
Eur J Endocrinol ; 168(3): K35-43, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23417163

RESUMO

OBJECTIVE: Short stature caused by biologically inactive GH is clinically characterized by lack of GH action despite normal-high secretion of GH, pathologically low IGF1 concentrations and marked catch-up growth on GH replacement therapy. DESIGN AND METHODS: Adopted siblings (girl and a boy) of unknown family history were referred for assessment of short stature (-4.5 and -5.6 SDS) at the age of 10 and 8.1 years respectively. They had delayed bone ages (6.8 and 4.5 years), normal GH peaks at stimulation tests, and severely reduced IGF1 concentrations (-3.5 and -4.0 SDS). Genetic analysis of the GH1 gene showed a heterozygous P59S mutation at position involved in binding to GH receptor (GHR). RESULTS: Isoelectric focusing analysis of secreted GH in patient serum revealed the presence of higher GH-P59S peak compared with that of wt-GH. Furthermore, computational simulation of GH-P59S binding to GHR suggested problems in correct binding of the mutant to the GHR. In vitro GHR binding studies revealed reduced binding affinity of GH-P59S for GHR (IC50, 30  ng/ml) when compared with the wt-GH (IC50, 11.8  ng/ml) while a significantly decreased ability of the mutant to activate the Jak2/Stat5 signaling pathway was observed at physiological concentrations of 25-100  ng/ml. CONCLUSIONS: The clinical and biochemical data of our patients support the diagnosis of partial bioinactive GH syndrome. The higher amount of GH-P59S secreted in their circulation combined with its impact on the wt-GH function on GHR binding and signaling may alter GHR responsiveness to wt-GH and could ultimately explain severe short stature found in our patients.


Assuntos
Estatura , Hormônio do Crescimento Humano/deficiência , Hormônio do Crescimento Humano/genética , Mutação , Substituição de Aminoácidos , Criança , Feminino , Heterozigoto , Terapia de Reposição Hormonal , Hormônio do Crescimento Humano/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/análise , Masculino , Modelos Moleculares , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores da Somatotropina/metabolismo , Irmãos
9.
Endocr Dev ; 23: 96-108, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23182824

RESUMO

Endocrine and neuroendocrine cells differ from cells which rapidly release all their secreted proteins in that they store some secretory proteins in concentrated forms in secretory granules to be rapidly released when cells are stimulated. Protein aggregation is considered as the first step in the secretory granule biosynthesis and, at least in the case of prolactin and growth hormone, greatly depends on zinc ions that facilitate this process. Hence, regulation of cellular zinc transport especially that within the regulated secretory pathway is of importance to understand. Various zinc transporters of Slc30a/ZnT and Slc39a/Zip families have been reported to fulfil this role and to participate in fine tuning of zinc transport in and out of the endoplasmic reticulum, Golgi complex and secretory granules, the main cellular compartments of the regulated secretory pathway. In this review, we will focus on the role of zinc in the formation of hormone-containing secretory granules with special emphasis on conditions required for growth hormone dimerization/aggregation. In addition, we highlight the role of zinc transporters that govern the process of zinc homeostasis in the regulated hormone secretion.


Assuntos
Retículo Endoplasmático/metabolismo , Hormônio do Crescimento/metabolismo , Via Secretória/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Zinco/farmacologia , Animais , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/fisiologia , Humanos , Modelos Biológicos , Ligação Proteica/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Via Secretória/fisiologia , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/fisiologia
10.
Clin Endocrinol (Oxf) ; 76(5): 683-90, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22026507

RESUMO

OBJECTIVE: A number of mathematical models for predicting growth and final height outcome have been proposed to enable the clinician to 'individualize' growth-promoting treatment. However, despite optimizing these models, many patients with isolated growth hormone deficiency (IGHD) do not reach their target height. The aim of this study was to analyse the impact of polymorphic genotypes [CA repeat promoter polymorphism of insulin-like growth factor-I (IGF-I) and the -202 A/C promoter polymorphism of IGF-Binding Protein-3 (IGFBP-3)] on variable growth factors as well as final height in severe IGHD following GH treatment. DESIGN, PATIENTS AND CONTROLS: One hundred seventy eight (IGF-I) and 167 (IGFBP-3) subjects with severe growth retardation because of IGHD were studied. In addition, the various genotypes were also studied in a healthy control group of 211 subjects. RESULTS: The frequency of the individual IGF-I (CA)(n) repeats ranging from 10 to 24, with the most frequent allele containing CA(19), was similar in controls and in IGHD subjects. However, in controls, the pooled CA(19) and CA(20) as well as -202 A IGFBP-3 alleles were significantly (P < 0·01 and P < 0·001) more common in the taller [≥2 to 0 standard deviation score (SDS)] when compared with the shorter subgroup (<0 to ≤-2 SDS). Overall, the effect of recombinant human growth hormone (rhGH) replacement did not reveal any difference between the various genotypes in terms of final height. Independent of their genotype, all subjects showed a slightly lower adult height SDS compared with midparental height SDS. CONCLUSION: Our results indicate that in patients with severe IGHD, although the various IGF-I and IGFBP-3 genotypes may play a role in GH responsiveness, there was no effect on final height.


Assuntos
Repetições de Dinucleotídeos/genética , Hormônio do Crescimento Humano/deficiência , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Fator de Crescimento Insulin-Like I/genética , Polimorfismo Genético , Regiões Promotoras Genéticas/genética , Adulto , Idoso , Estatura/efeitos dos fármacos , Estatura/genética , Criança , Pré-Escolar , Feminino , Frequência do Gene , Genótipo , Transtornos do Crescimento/sangue , Transtornos do Crescimento/genética , Transtornos do Crescimento/metabolismo , Terapia de Reposição Hormonal , Hormônio do Crescimento Humano/uso terapêutico , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Fator de Crescimento Insulin-Like I/metabolismo , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Fenótipo , Resultado do Tratamento , Adulto Jovem
11.
Growth Horm IGF Res ; 21(3): 160-6, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21546299

RESUMO

CONTEXT AND OBJECTIVE: Despite the differences in the main characteristics between the autosomal dominant form of GH deficiency (IGHD II) and the bioinactive GH syndrome, a common feature of both is their impact on linear growth leading to short stature in all affected patients. DESIGN: The index patient, a boy, was referred for assessment of his short stature (-2.54 SD score) and a delayed bone age of 5.9 yrs at the chronological age of 7.7 yrs. The GHD was confirmed by standard GH provocation tests, which revealed modestly reduced GH and IGF-I concentrations. Further genetic analysis of GH-1 gene identified heterozygosity for GH-P59L mutation. The secretion of the GH-P59L following stimulation with forskolin was investigated and compared to that of the wt-GH after expression of both GH variants in AtT-20 cells. Based on the position of P59L mutation that lies within a patch of residues composing the GH binding site 1 for GHR, we performed the analysis of GH-P59L binding to GHR by in silico mutagenesis and molecular dynamics simulations, which suggested possible problems in correct binding of GH-P59L to the GHR. Therefore, the functional characterization of this GH mutant was assessed through studies of GHR binding and activation of Jak2/Stat5 signaling pathway. RESULTS: In line with the clinical data of the patient GH deficiency is suggested, underlined by GH-secretion studies revealing a moderate difference in secretion between GH-P59L and wt-GH. In addition, further functional characterization of the GH-P59L by studies of GH-receptor binding and activation of Jak2/Stat5 pathway presented with a reduced binding affinity of GH-P59L for GHR and decreased bioactivity compared to the wt-GH. CONCLUSIONS: The clinical data of the patient combined with the laboratory data support the diagnosis of partial IGHD type II. Since the GH deficiency was not total, additional binding and signaling studies were performed, which revealed that the GH-P59L variant displays some of the common features of bioinactive GH syndrome. Taken together, in this study we report a patient suffering from the combination of two growth disorders (alteration of secretion as well as bioactivity) caused by a GH-1 gene alteration highlighting the necessity of functional analysis of any GH variant, despite the presence of obvious clinical features of IGHD type II.


Assuntos
Transtornos do Crescimento/genética , Hormônio do Crescimento Humano/genética , Mutação , Hormônio do Crescimento Humano/deficiência , Humanos , Janus Quinases/genética , Janus Quinases/metabolismo , Masculino , Síndrome
12.
Blood ; 117(18): 4881-4, 2011 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-21389317

RESUMO

CCAAT/enhancer binding protein-α (CEBPA) mutations in acute myeloid leukemia (AML) patients with a normal karyotype (NK) confer favorable prognosis, whereas NK-AML patients per se are of intermediate risk. This suggests that blocked CEBPA function characterizes NK-AML with favorable outcome. We determined the prognostic significance of CEBPA DNA binding function by enzyme-linked immunosorbent assay in 105 NK-AML patients. Suppressed CEBPA DNA binding was defined by 21 good-risk AML patients with inv(16) or t(8;21) (both abnormalities targeting CEBPA) and 8 NK-AML patients with dominant-negative CEBPA mutations. NK-AML patients with suppressed CEBPA function showed a better overall survival (P = .0231) and disease-free survival (P = .0069) than patients with conserved CEBPA function. Suppressed CEBPA DNA binding was an independent marker for better overall survival and disease-free survival in a multivariable analysis that included FLT3-ITD, NPM1 and CEBPA mutation status, white blood cell count, age and lactate dehydrogenase. These data indicate that suppressed CEBPA function is associated with favorable prognosis in NK-AML patients.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/deficiência , Proteínas Estimuladoras de Ligação a CCAAT/genética , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutação , Adolescente , Adulto , Idoso , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Feminino , Humanos , Estimativa de Kaplan-Meier , Cariotipagem , Masculino , Pessoa de Meia-Idade , Nucleofosmina , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Prognóstico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Proteína 1 Parceira de Translocação de RUNX1 , Células U937 , Adulto Jovem
13.
Endocrinology ; 151(9): 4400-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20591972

RESUMO

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.


Assuntos
Hormônio do Crescimento/deficiência , Hormônio do Crescimento/genética , Mutação , Hipófise/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Técnicas de Inativação de Genes , Vetores Genéticos/genética , Hormônio do Crescimento/metabolismo , Humanos , Lentivirus/genética , MicroRNAs/genética , Microscopia Confocal , Hipófise/patologia , RNA Interferente Pequeno/genética , Ratos , Transfecção/métodos
14.
Endocrinology ; 151(6): 2650-8, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20351314

RESUMO

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.


Assuntos
Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Hormônio do Crescimento/metabolismo , Hormônio do Crescimento Humano/genética , Isoformas de Proteínas/metabolismo , Animais , Western Blotting , Linhagem Celular , Expressão Gênica/genética , Hormônio do Crescimento/genética , Hormônio do Crescimento Humano/metabolismo , Humanos , Mutação , Isoformas de Proteínas/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Clin Endocrinol Metab ; 95(2): 731-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19952226

RESUMO

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.


Assuntos
Hormônio do Crescimento Humano/deficiência , Hormônio do Crescimento Humano/genética , Mutação , Animais , Células CHO , Células Cultivadas , Criança , Cricetinae , Cricetulus , Feminino , Hormônio do Crescimento Humano/metabolismo , Humanos , Janus Quinase 2/fisiologia , Camundongos , Simulação de Dinâmica Molecular , Receptores da Somatotropina/metabolismo , Fator de Transcrição STAT5/fisiologia , Zinco/metabolismo
16.
Eur J Immunol ; 39(4): 1118-28, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19291700

RESUMO

The migration of monocytes to sites of inflammation is largely determined by their response to chemokines. Although the chemokine specificities and expression patterns of chemokine receptors are well defined, it is still a matter of debate how cells integrate the messages provided by different chemokines that are concomitantly produced in physiological or pathological situations in vivo. We present evidence for one regulatory mechanism of human monocyte trafficking. Monocytes can integrate stimuli provided by inflammatory chemokines in the presence of homeostatic chemokines. In particular, migration and cell responses could occur at much lower concentrations of the CCR2 agonists, in the presence of chemokines (CCL19 and CCL21) that per se do not act on monocytes. Binding studies on CCR2(+) cells showed that CCL19 and CCL21 do not compete with the CCR2 agonist CCL2. Furthermore, the presence of CCL19 or CCL21 could influence the degradation of CCL2 and CCL7 on cells expressing the decoy receptor D6. These findings disclose a new scenario to further comprehend the complexity of chemokine-based monocyte trafficking in a vast variety of human inflammatory disorders.


Assuntos
Movimento Celular/imunologia , Quimiotaxia de Leucócito/imunologia , Inflamação/imunologia , Monócitos/imunologia , Receptores CCR2/imunologia , Receptores CCR7/imunologia , Sequência de Aminoácidos , Movimento Celular/efeitos dos fármacos , Quimiocina CCL19/química , Quimiocina CCL19/imunologia , Quimiocina CCL19/farmacologia , Quimiocina CCL2/imunologia , Quimiocina CCL2/farmacologia , Quimiocina CCL21/química , Quimiocina CCL21/imunologia , Quimiocina CCL21/farmacologia , Quimiocina CCL7/imunologia , Quimiocina CCL7/farmacologia , Quimiotaxia de Leucócito/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/imunologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glicosaminoglicanos/imunologia , Glicosaminoglicanos/metabolismo , Humanos , Inflamação/metabolismo , Ligantes , Dados de Sequência Molecular , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Fosforilação/imunologia , Estrutura Terciária de Proteína , Receptores CCR10/imunologia , Receptores CCR10/metabolismo , Receptores CCR2/agonistas , Receptores CCR2/química , Receptores CCR7/agonistas , Receptores CCR7/química , Receptor D6 de Quimiocina
17.
J Clin Endocrinol Metab ; 93(3): 974-80, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18029459

RESUMO

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

Assuntos
Estatura/efeitos dos fármacos , Éxons , Hormônio do Crescimento/uso terapêutico , Terapia de Reposição Hormonal , Hormônio do Crescimento Humano/deficiência , Receptores da Somatotropina/genética , Adulto , Feminino , Deleção de Genes , Genótipo , Humanos , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Isoformas de Proteínas
18.
Eur J Endocrinol ; 157 Suppl 1: S67-74, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17785701

RESUMO

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.


Assuntos
Transtornos do Crescimento/genética , Hormônio do Crescimento Humano/genética , Mutação de Sentido Incorreto , Puberdade Tardia/genética , Adolescente , Adulto , Arginina , Criança , Cisteína , Seguimentos , Transtornos do Crescimento/sangue , Hormônio do Crescimento Humano/sangue , Hormônio do Crescimento Humano/química , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Pessoa de Meia-Idade , Linhagem , Puberdade Tardia/sangue
19.
J Clin Endocrinol Metab ; 92(11): 4427-35, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17726075

RESUMO

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.


Assuntos
Transtornos Cromossômicos/genética , Éxons/genética , Hormônio do Crescimento Humano/deficiência , Mutação/fisiologia , Adolescente , Adulto , Western Blotting , Estatura , Proliferação de Células , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Colforsina/farmacologia , Retículo Endoplasmático/ultraestrutura , Feminino , Genes Dominantes/genética , Vetores Genéticos/genética , Complexo de Golgi/ultraestrutura , Humanos , Masculino , Microscopia Confocal , Linhagem , Isoformas de Proteínas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Vesículas Secretórias/ultraestrutura
20.
J Clin Endocrinol Metab ; 92(8): 2893-901, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17519310

RESUMO

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.


Assuntos
Estatura/efeitos dos fármacos , Hormônio do Crescimento/uso terapêutico , Hormônio do Crescimento Humano/antagonistas & inibidores , Hormônio do Crescimento Humano/genética , Alelos , Animais , Proliferação de Células , Células Cultivadas , Criança , Imunofluorescência , Genes Reporter , Vetores Genéticos , Crescimento/fisiologia , Heterozigoto , Hormônio do Crescimento Humano/sangue , Humanos , Focalização Isoelétrica , Luciferases/genética , Masculino , Camundongos , Microscopia Confocal , Mutação , Linhagem , Puberdade/fisiologia , Receptores da Somatotropina/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA