Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
2.
Bio Protoc ; 13(15): e4730, 2023 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-37575391

RESUMEN

Anorexia nervosa (AN) is a psychiatric disorder mainly characterized by extreme hypophagia, severe body weight loss, hyperactivity, and hypothermia. Currently, AN has the highest mortality rate among psychiatric illnesses. Despite decades of research, there is no effective cure for AN nor is there a clear understanding of its etiology. Since a complex interaction between genetic, environmental, social, and cultural factors underlines this disorder, the development of a suitable animal model has been difficult so far. Here, we present our protocol that couples a loss-of-function mouse model to the activity-based anorexia model (ABA), which involves self-imposed starvation in response to exposure to food restriction and exercise. We provide insights into a neural circuit that drives survival in AN and, in contrast to previous protocols, propose a model that mimics the conditions that mainly promote AN in humans, such as increased incidence during adolescence, onset preceded by negative energy balance, and increased compulsive exercise. This protocol will be useful for future studies that aim to identify neuronal populations or brain circuits that promote the onset or long-term maintenance of this devastating eating disorder.

3.
Biomedicines ; 11(1)2023 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-36672651

RESUMEN

As marijuana use during adolescence has been increasing, the need to understand the effects of its long-term use becomes crucial. Previous research suggested that marijuana consumption during adolescence increases the risk of developing mental illnesses, such as schizophrenia, depression, and anxiety. Ghrelin is a peptide produced primarily in the gut and is important for feeding behavior. Recent studies have shown that ghrelin and its receptor, the growth hormone secretagogue receptor (GHSR), play important roles in mediating stress, as well as anxiety and depression-like behaviors in animal models. Here, we investigated the effects of chronic tetrahydrocannabinol (THC) administration during late adolescence (P42-55) in GHSR (GHSR -/-) knockout mice and their wild-type littermates in relation to anxiety-like behaviors. We determined that continuous THC exposure during late adolescence did not lead to any significant alterations in the anxiety-like behaviors of adult mice, regardless of genotype, following a prolonged period of no exposure (1 month). These data indicate that in the presence of intact or impaired ghrelin/GHSR signaling, THC exposure during late adolescence has limited if any long-term impact on anxiety-like behaviors in mice.

4.
Clin Endocrinol (Oxf) ; 76(5): 683-90, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22026507

RESUMEN

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.


Asunto(s)
Repeticiones de Dinucleótido/genética , Hormona de Crecimiento Humana/deficiencia , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/genética , Polimorfismo Genético , Regiones Promotoras Genéticas/genética , Adulto , Anciano , Estatura/efectos de los fármacos , Estatura/genética , Niño , Preescolar , Femenino , Frecuencia de los Genes , Genotipo , Trastornos del Crecimiento/sangre , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/metabolismo , Terapia de Reemplazo de Hormonas , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Factor I del Crecimiento Similar a la Insulina/metabolismo , Modelos Lineales , Masculino , Persona de Mediana Edad , Análisis Multivariante , Fenotipo , Resultado del Tratamiento , Adulto Joven
5.
Nat Metab ; 2(11): 1204-1211, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33106687

RESUMEN

Hypothalamic agouti-related peptide (AgRP) and neuropeptide Y-expressing neurons have a critical role in driving food intake, but also in modulating complex, non-feeding behaviours1. We interrogated whether AgRP neurons are relevant to the emergence of anorexia nervosa symptomatology in a mouse model. Here we show, using in vivo fibre photometry, a rapid inhibition of AgRP neuronal activity following voluntary cessation of running. All AgRP neuron-ablated, food-restricted mice die within 72 h of compulsive running, while daily activation of AgRP neurons using a chemogenetic tool increases voluntary running with no lethality of food-restricted animals. Animals with impaired AgRP neuronal circuits are unable to properly mobilize fuels during food-restriction-associated exercise; however, when provided with elevated fat content through diet, their death is completely prevented. Elevated fat content in the diet also prevents the long-term behavioural impact of food-restricted fit mice with elevated exercise volume. These observations elucidate a previously unsuspected organizational role of AgRP neurons, via the mediation of the periphery, in the regulation of compulsive exercise and its related lethality with possible implications for psychiatric conditions, such as anorexia nervosa.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Anorexia/metabolismo , Ejercicio Compulsivo/metabolismo , Neuronas/metabolismo , Animales , Anorexia/psicología , Conducta Animal , Peso Corporal , Ejercicio Compulsivo/psicología , Dieta , Dieta Alta en Grasa , Femenino , Privación de Alimentos , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Fibras Nerviosas/metabolismo , Análisis de Supervivencia
6.
Cell Stem Cell ; 22(5): 615-616, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29727675

RESUMEN

Several methods for making hypothalamic-like neurons (iHTNs) from human or mouse induced pluripotent stem cells (iPSCs) with varying efficiencies exist. In this issue of Cell Stem Cell, Rajamani et al. (2018) devise a new, efficient method to differentiate iPSCs from super-obese and normal donors into iHTNS and investigate their different molecular profiles and hormone responses.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Diferenciación Celular , Humanos , Ratones , Neuronas , Obesidad
7.
Biochem Pharmacol ; 124: 1-9, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27457999

RESUMEN

Isolated growth hormone deficiency type II (IGHD II) is a rare genetic splicing disorder characterized by reduced growth hormone (GH) secretion and short stature. It is mainly caused by autosomal dominant-negative mutations within the growth hormone gene (GH-1) which results in missplicing at the mRNA level and the subsequent loss of exon 3, producing the 17.5-kDa GH isoform: a mutant and inactive GH protein that reduces the stability and the secretion of the 22-kDa GH isoform, the main biologically active GH form. At present, patients suffering from IGHD II 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 the toxic effects of the 17.5-kDa mutant on the pituitary gland, which may eventually lead to other hormonal deficiencies. As the severity of the disease inversely correlates with the 17.5-kDa/22-kDa ratio, increasing the inclusion of exon 3 is expected to ameliorate disease symptoms. This review focuses on the recent advances in experimental and therapeutic strategies applicable to treat IGHD II in clinical and preclinical contexts. Several avenues for alternative IGHD II therapy will be discussed including the use of small interfering RNA (siRNA) and short hairpin RNA (shRNA) constructs that specifically target the exon 3-deleted transcripts as well as the application of histone deacetylase inhibitors (HDACi) and antisense oligonucleotides (AONs) to enhance full-length GH-1 transcription, correct GH-1 exon 3 splicing and manipulate GH pathway.


Asunto(s)
Enanismo Hipofisario/tratamiento farmacológico , Hormona del Crecimiento/genética , Empalme del ARN , Animales , Modelos Animales de Enfermedad , Enanismo Hipofisario/genética , Terapia Genética , Hormona del Crecimiento/deficiencia , Humanos , Fenotipo
8.
Endocrinology ; 157(10): 3972-3982, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27253996

RESUMEN

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.


Asunto(s)
Butiratos/uso terapéutico , Enanismo Hipofisario/tratamiento farmacológico , Hormona del Crecimiento/metabolismo , Empalme del ARN/efectos de los fármacos , Animales , Butiratos/farmacología , Línea Celular , Evaluación Preclínica de Medicamentos , Técnicas de Transferencia de Gen , Hormona del Crecimiento/genética , Humanos , Ratas , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo
10.
J Clin Endocrinol Metab ; 100(12): E1575-83, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26485222

RESUMEN

CONTEXT: The autosomal dominant form of GH deficiency (IGHD II) is characterized by markedly reduced GH secretion combined with low concentrations of IGF-1 leading to short stature. OBJECTIVE: Structure-function analysis of a missense mutation in the GH-1 gene converting codon 76 from leucine (L) to proline (P) yielding a mutant GH-L76P peptide. DESIGN, SETTINGS, AND PATIENTS: Heterozygosity for GH-L76P/wt-GH was identified in a nonconsanguineous Spanish family. The index patients, two siblings, a boy and a girl, were referred for assessment of their short stature (-3.2 and -3.8 SD). Their grandmother, father, and aunt were also carrying the same mutation and showed severe short stature; therefore, IGHD II was diagnosed. INTERVENTIONS AND RESULTS: AtT-20 cells coexpressing both wt-GH and GH-L76P showed a reduced GH secretion (P < .001) after forskolin stimulation when compared with the cells expressing only wt-GH. In silico mutagenesis and molecular dynamics simulations presented alterations of correct folding and mutant stability compared with wt-GH. Therefore, further structural analysis of the GH-L76P mutant was performed using expressed and purified proteins in Escherichia coli by thermofluor assay and fast degradation proteolysis assay. Both assays revealed that the GH-L76P mutant is unstable and misfolded compared to wt-GH confirming the bioinformatic model prediction. CONCLUSIONS: This is the first report of a family suffering from short stature caused by IGHD II, which severely affects intracellular GH folding and stability as well as secretion, highlighting the necessity of functional analysis of any GH variant for defining new mechanisms as a cause for IGHD II.


Asunto(s)
Hormona de Crecimiento Humana/deficiencia , Pliegue de Proteína , Sustitución de Aminoácidos/genética , Animales , Estatura/genética , Niño , Preescolar , Codón/genética , Colforsina/farmacología , Biología Computacional , Familia , Femenino , Heterocigoto , Hormona de Crecimiento Humana/genética , Hormona de Crecimiento Humana/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Mutación Missense/genética , Linaje , Embarazo , Conformación Proteica
11.
PLoS One ; 9(10): e107388, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25310566

RESUMEN

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.


Asunto(s)
Ácido Butírico/farmacología , Calcio/metabolismo , Hormona del Crecimiento/metabolismo , Adenohipófisis/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Animales , Línea Celular , Silenciador del Gen , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Adenohipófisis/citología , Adenohipófisis/metabolismo , ARN Interferente Pequeño , Ratas , Receptores Acoplados a Proteínas G/genética
12.
Horm Res Paediatr ; 82(4): 245-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25196974

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Catión/genética , Hormona del Crecimiento/metabolismo , Hipófisis/metabolismo , Vías Secretoras/genética , Zinc/metabolismo , Animales , Línea Celular , Citoplasma/metabolismo , ADN Complementario/genética , Técnicas de Silenciamiento del Gen , Hipófisis/citología , ARN Interferente Pequeño/genética , Ratas
13.
Horm Res Paediatr ; 80(6): 381-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24296719

RESUMEN

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.


Asunto(s)
Hormona de Crecimiento Humana/metabolismo , Zinc/fisiología , Animales , Trastornos del Crecimiento/etiología , Trastornos del Crecimiento/metabolismo , Humanos , Recién Nacido , Errores Innatos del Metabolismo de los Metales/diagnóstico , Errores Innatos del Metabolismo de los Metales/etiología , Errores Innatos del Metabolismo de los Metales/metabolismo , Leche Humana/química , Leche Humana/metabolismo , Zinc/deficiencia , Zinc/metabolismo
14.
Endocrinology ; 154(11): 4215-25, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23970781

RESUMEN

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.


Asunto(s)
Hormona del Crecimiento/metabolismo , Zinc/metabolismo , Animales , Apoptosis , Línea Celular , Supervivencia Celular , Medios de Cultivo , Etilaminas , Hormona del Crecimiento/química , Hormona del Crecimiento/genética , Humanos , Ratones , Mutación , Unión Proteica , Piridinas , Ratas , Factores de Tiempo , Zinc/administración & dosificación , Zinc/farmacología
15.
Int J Endocrinol ; 2013: 259189, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24194756

RESUMEN

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.

16.
Eur J Endocrinol ; 168(3): K35-43, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23417163

RESUMEN

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.


Asunto(s)
Estatura , Hormona de Crecimiento Humana/deficiencia , Hormona de Crecimiento Humana/genética , Mutación , Sustitución de Aminoácidos , Niño , Femenino , Heterocigoto , Terapia de Reemplazo de Hormonas , Hormona de Crecimiento Humana/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/análisis , Masculino , Modelos Moleculares , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores de Somatotropina/metabolismo , Hermanos
17.
Endocr Dev ; 23: 96-108, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23182824

RESUMEN

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.


Asunto(s)
Retículo Endoplásmico/metabolismo , Hormona del Crecimiento/metabolismo , Vías Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Zinc/farmacología , Animales , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/fisiología , Humanos , Modelos Biológicos , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Vías Secretoras/fisiología , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/fisiología
18.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA