Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Brain ; 134(Pt 6): 1808-28, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21576111

RESUMEN

Senataxin is encoded by the SETX gene and is mainly involved in two different neurodegenerative diseases, the dominant juvenile form of amyotrophic lateral sclerosis type 4 and a recessive form of ataxia with oculomotor apraxia type 2. Based on protein homology, senataxin is predicted to be a putative DNA/RNA helicase, while senataxin interactors from patients' lymphoblast cell lines suggest a possible involvement of the protein in different aspects of RNA metabolism. Except for an increased sensitivity to oxidative DNA damaging agents shown by some ataxia with neuropathy patients' cell lines, no data are available about possible functional consequences of dominant SETX mutations and no studies address the function of senataxin in neurons. To start elucidating the physiological role of senataxin in neurons and how disease-causing mutations in this protein lead to neurodegeneration, we analysed the effect of senataxin on neuronal differentiation in primary hippocampal neurons and retinoic acid-treated P19 cells by modulating the expression levels of wild-type senataxin and three different dominant mutant forms of the protein. Wild-type senataxin overexpression was required and sufficient to trigger neuritogenesis and protect cells from apoptosis during differentiation. These actions were reversed by silencing of senataxin. In contrast, overexpression of the dominant mutant forms did not affect the regular differentiation process in primary hippocampal neurons. Analysis of the cellular pathways leading to neuritogenesis and cytoprotection revealed a role of senataxin in modulating the expression levels and signalling activity of fibroblast growth factor 8. Silencing of senataxin reduced, while overexpression enhanced, fibroblast growth factor 8 expression levels and the phosphorylation of related target kinases and effector proteins. The effects of senataxin overexpression were prevented when fibroblast growth factor 8 signalling was inhibited, while exogenous fibroblast growth factor 8 reversed the effects of senataxin silencing. Overall, these results reveal a key role of senataxin in neuronal differentiation through the fibroblast growth factor 8 signalling and provide initial molecular bases to explain the neurodegeneration associated with loss-of-function mutations in senataxin found in recessive ataxia. The lack of effect on neuritogenesis observed with the overexpression of the dominant mutant forms of senataxin apparently excludes a dominant negative effect of these mutants while favouring haploinsufficiency as the pathogenic mechanism implicated in the amyotrophic lateral sclerosis 4-related degenerative condition. Alternatively, a different protein function, other than the one involved in neuritogenesis, may be implicated in these dominant degenerative processes.


Asunto(s)
ADN Helicasas/metabolismo , Factor 8 de Crecimiento de Fibroblastos/farmacología , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuronas/citología , ARN Helicasas/metabolismo , Transducción de Señal/fisiología , Animales , Caspasa 3/metabolismo , Muerte Celular/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , ADN Helicasas/genética , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , Humanos , Ratones , Enzimas Multifuncionales , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroblastoma/patología , Neuronas/efectos de los fármacos , ARN Helicasas/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección/métodos , Tretinoina/farmacología
2.
Neurogenetics ; 11(1): 91-100, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19593598

RESUMEN

Ataxia with oculomotor apraxia (AOA) type 2 (AOA2 MIM 606002) is a recessive subtype of AOA characterized by cerebellar atrophy, oculomotor apraxia, early loss of reflexes, and peripheral neuropathy. Various mutations either in homozygous or compound heterozygous condition were so far identified in the associated gene SETX (MIM 608465). SETX encodes a large protein called senataxin with a DNA-RNA helicase domain and a putative N-terminus protein interaction domain. Here, we report the identification of two novel homozygous mutations in SETX gene, c.340_342delCTT (p.L114Del) and c.1669C > T (p.R557X), in two AOA2 families. The characterization of the mutant lymphoblastoid cell lines for sensitivity to oxidative DNA-damaging agents indicates that the p.L114Del deletion confers an increased sensitivity to H2O2, camptothecin, and mitomycin C, previously found to induce death in lymphoblasts harbouring other SETX mutations; the cells carrying the nonsense mutation display instead values within the normal range. Further analysis of a neuronal cell model SKNBE, transfected with the mutant senataxin proteins, reveals increased sensitivity also to staurosporine and excitotoxicity associated with the p.L114Del mutant only. We also demonstrate that the sensitizing effect of p.L114Del on apoptosis can be reversed by senataxin silencing. The ability of a single amino acid deletion to sensitize cells to death by different agents, compared to the lack of effect of a whole protein deletion, seems to exclude a protective role played by the native protein while suggesting that a specific mutation confers to the protein the ability to enhance the toxic effect of various cell damaging agents.


Asunto(s)
Apraxia Ideomotora/genética , Ataxia/genética , Oftalmopatías/genética , Mutación , ARN Helicasas/genética , Adulto , Apoptosis , Camptotecina/farmacología , Daño del ADN , ADN Helicasas , Femenino , Homocigoto , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Persona de Mediana Edad , Mitomicina/farmacología , Enzimas Multifuncionales , Linaje
3.
Hum Mutat ; 30(7): 1104-16, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19431184

RESUMEN

The late-infantile-onset forms of neuronal ceroid lipofuscinosis (LINCL) are the most genetically heterogeneous group among the autosomal recessive neuronal ceroid lipofuscinoses (NCLs), with causative mutations found in CLN1, CLN2, CLN5, CLN6, CLN7 (MFSD8), and CLN8 genes. Homozygous mutations in CLN8 are associated with two distinct phenotypes: progressive epilepsy and mental retardation (EPMR), first identified in Finland; and a variant of late-infantile NCL (v-LINCL) described in a subset of Turkish and Italian patients. The function of the protein encoded by CLN8 is currently unknown. Here we report the identification of an Italian v-LINCL patient with a complete isodisomy of chromosome 8, leading to homozygosity of a maternally-inherited 3-bp deletion in CLN8 gene (c.180_182delGAA, p.Lys61del). Notably, uniparental disomy (UPD) has never been described associated with the NCLs. In addition, we provide evidence of the biological role of CLN8 characterized by expressing in different neuronal cell models the native protein, the protein carrying the mutation identified here, or three additional missense mutations previously described. Our results, validated through a gene silencing approach, indicate that CLN8 plays a role in cell proliferation during neuronal differentiation and in protection against cell death.


Asunto(s)
Proteínas de la Membrana/genética , Mutación , Lipofuscinosis Ceroideas Neuronales/genética , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Niño , Aberraciones Cromosómicas , Cromosomas Humanos Par 8 , Femenino , Humanos , Masculino , Neuronas/citología , Linaje , Eliminación de Secuencia , Transfección , Tripeptidil Peptidasa 1
4.
Mol Endocrinol ; 22(3): 676-88, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18096696

RESUMEN

Somatostatin (SS) is a widely distributed polypeptide that exerts inhibitory effects on hormone secretion and cell proliferation by interacting with five different receptors (SST1-SST5). Beta-arrestins have been implicated in regulating SST internalization, but the structural domains mediating this effect are largely unknown. The aim of this study was to characterize the intracellular mechanisms responsible for internalization of human SST5 in the rat pituitary cell line GH3 and to identify the SST5 structural domains involved in this process. To this purpose we evaluated, by fluorescence microscopy and biochemical assay, the ability of wild-type, progressive C-terminal truncated and third cytoplasmatic loop mutants SST5-DsRed to associate with beta-arrestin-enhanced green fluorescent protein and to internalize under SS28 stimulation. The truncated mutants were comparable to the wild-type receptor with respect to recruitment of beta-arrestin-2 and internalization, whereas the third loop mutants R240W, S242A, and T247A showed the abolishment or reduction of arrestin association and a significant reduction of receptor internalization (14.4%, 29%, and 30.9% vs. 52.4% of wild type) and serine phosphorylation upon SS28 stimulation. Moreover, we evaluated the ability of simultaneous mutation of these three residues (R240, S242, and T247) and C-terminal truncated receptors to internalize. The progressive truncation of the C-terminal tail resulted in a progressive increased internalization (21.6%, 36.7%, and 41%, respectively) with respect to the full-length total third-loop mutant (15%). In conclusion, our results indicate the SST5 third intracellular loop as an important mediator of beta-arrestin/receptor interaction and receptor internalization, whereas they suggest that residues 328-347 within the C terminus may play an inhibitory role in receptor internalization.


Asunto(s)
Arrestinas/metabolismo , Hipófisis/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina-28/farmacología , Secuencia de Aminoácidos , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , ADN/química , ADN/genética , Endocitosis/fisiología , Humanos , Inmunoprecipitación , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Hipófisis/efectos de los fármacos , Mutación Puntual/fisiología , Unión Proteica , Conformación Proteica , Ratas , Receptores de Somatostatina/genética , Arrestina beta 2 , beta-Arrestinas
5.
Exp Biol Med (Maywood) ; 233(4): 439-46, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18367633

RESUMEN

The dopamine receptor subtype 2 (D2R) promoter contains a functional retinoic acid response element involved in the control of D2R expression. The aim of the study was to evaluate the effect of 9-cis retinoic acid (9-cis RA) on D2R protein expression in human pituitary adenomas and GH3 cell line. Treatment with 9-cis RA (100 nM for 48 hrs) caused a 109 +/- 32% increase of basal D2R levels in five of eight growth hormone (GH)-secreting adenomas (GH-omas), a 129 +/- 28% increase in 7 of 11 nonfunctioning adenomas, and no effect in two resistant prolactinomas by Western blotting. The lack of D2R induction in some tumors was not associated with a different pattern of retinoid x receptor (RXR) and retinoic acid receptor (RAR) isoform expression that was similar in all tumors by immunohistochemistry. While the induction of D2R did not affect the slight but significant inhibitory effect exerted by dopamine (10 nM) on in vitro GH release by GH-oma cultured cells, in pituitary GH3 cell lines cis-9 RA enhanced the dopamine-induced inhibition of in vitro GH release (% inhibition: 16 +/- 2 versus 26 +/- 5, P < 0.05), cell proliferation (25 +/- 2% versus 44 +/- 5%, P < 0.05) and cell viability (16 +/- 0.8% versus 29 +/- 1%, P < 0.05), likely by activating caspase-3 (28 +/- 3% versus basal, P < 0.05). In conclusion, this study provides novel evidence for a permissive role of retinoids on the expression of D2R in a good proportion of pituitary tumors and on the generation of pro-apoptotic signals in GH3 cell line.


Asunto(s)
Neoplasias Hipofisarias , Receptores de Dopamina D2/metabolismo , Tretinoina/metabolismo , Alitretinoína , Animales , Apoptosis/fisiología , Línea Celular , Proliferación Celular , Supervivencia Celular , Dopamina/metabolismo , Humanos , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Receptores de Dopamina D2/genética , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo
6.
J Clin Endocrinol Metab ; 92(9): 3738-42, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17595244

RESUMEN

CONTEXT: Pseudohypoparathyroidism (PHP) types Ia and Ib, are caused by mutations in GNAS exons 1-13 and GNAS methylation defects, respectively. PHP-Ia patients show Albright hereditary osteodystrophy (AHO) and resistance toward PTH and additional hormones, whereas PHP-Ib patients do not have AHO and hormone resistance is limited to PTH and, as reported in one paper, TSH. No study addressed the question of GH deficiency in PHP-Ib patients. OBJECTIVES: The objective of the study was to screen patients with clinically diagnosed PHP-Ib for genetic defects and investigate the presence of resistance to TSH and GHRH. PATIENTS/METHODS: We investigated GNAS differential methylation and STX16 microdeletions in genomic DNA from 10 patients with clinical diagnosis of sporadic PHP-Ib, i.e. PTH resistance without AHO. Resistance to GHRH was assessed by GH response to GHRH plus arginine. Thyroid function and ultrasonography were also evaluated. RESULTS: Molecular analysis showed GNAS cluster imprinting defects in all PHP-Ib patients and the first de novo STX16 deletion in one apparently sporadic patient. Subclinical or clinical hypothyroidism due to resistance to TSH was present in nine of 10 patients, whereas a preserved GH response to a GHRH plus arginine test was present in all patients, with one exception. CONCLUSIONS: We report the first molecular analysis of Italian patients with PHP-Ib. Clinical investigation shows that, like PHP-Ia patients, PHP-Ib patients are resistant to TSH, whereas they maintain a normal responsiveness to GHRH, at variance with PHP-Ia patients. These data provide new information on this rare disease and emphasize the clinical heterogeneity of genetic defects within GNAS.


Asunto(s)
Resistencia a Medicamentos/genética , Hormona Liberadora de Hormona del Crecimiento/farmacología , Seudohipoparatiroidismo/genética , Tirotropina/farmacología , Adolescente , Adulto , Arginina/administración & dosificación , Cromograninas , Análisis Mutacional de ADN , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Pruebas Genéticas , Hormona Liberadora de Hormona del Crecimiento/administración & dosificación , Hormona de Crecimiento Humana/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/análisis , Masculino , Persona de Mediana Edad
7.
Eur J Endocrinol ; 154(5): 685-9, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16645015

RESUMEN

OBJECTIVE: Mutations in the gene coding for the orphan nuclear receptor DAX1 cause X-linked adrenal hypoplasia congenita (AHC). Affected boys usually present with primary adrenal failure in early infancy or childhood. Impaired sexual development due to hypogonadotropic hypogonadism becomes manifest at the time of puberty. Moreover, evidence from Dax1 knockout mice and a limited number of patients with AHC, suggests that mutations in DAX1 may directly cause abnormalities in spermatogenesis. The aim of this study was to characterize clinically and genetically five patients with AHC. DESIGN: DNA sequencing analysis, endocrine testing, testicular ultrasound and semen analysis with 1-year follow-up after gonadotropin treatment. METHODS: We report on five men with classic AHC manifestations. Genomic DNA was extracted from patients' peripheral blood leukocytes and the coding region, splice sites, and promoter (-240 bp) region of DAX1 were directly sequenced. RESULTS: Three known and two novel mutations were detected in the DAX1 coding sequence in these patients. Semen analysis was performed in four of the five patients and showed azoospermia. Twelve-month treatment with gonadotropins did not restore fertility in these patients. All patients showed a normal testicular Doppler ultrasound, in contrast with that observed in Dax1-deficient mice, which display abnormalities in the rete testis. CONCLUSIONS: These cases further expand the number of DAX1 mutations reported in the literature, as well as our clinical knowledge of this rare disease.


Asunto(s)
Insuficiencia Suprarrenal/diagnóstico por imagen , Insuficiencia Suprarrenal/genética , Cromosomas Humanos X , Proteínas de Unión al ADN/genética , Receptores de Ácido Retinoico/genética , Proteínas Represoras/genética , Adolescente , Adulto , Receptor Nuclear Huérfano DAX-1 , Análisis Mutacional de ADN , Mutación del Sistema de Lectura , Eliminación de Gen , Humanos , Hipogonadismo/diagnóstico por imagen , Hipogonadismo/genética , Masculino , Regiones Promotoras Genéticas/genética , Sitios de Empalme de ARN/genética , Testículo/diagnóstico por imagen , Ultrasonografía
8.
Cancer Res ; 64(24): 9193-8, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15604292

RESUMEN

The two regulatory subunits (R1 and R2) of protein kinase A (PKA) are differentially expressed in cancer cell lines and exert diverse roles in growth control. Recently, mutations of the PKA regulatory subunit 1A gene (PRKAR1A) have been identified in patients with Carney complex. The aim of this study was to evaluate the expression of the PKA regulatory subunits R1A, R2A, and R2B in a series of 30 pituitary adenomas and the effects of subunit activation on cell proliferation. In these tumors, neither mutation of PRKAR1A nor loss of heterozygosity was identified. By real-time PCR, mRNA of the three subunits was detected in all of the tumors, R1A being the most represented in the majority of samples. By contrast, immunohistochemistry documented low or absent R1A levels in all tumors, whereas R2A and R2B were highly expressed, thus resulting in an unbalanced R1/R2 ratio. The low levels of R1A were, at least in part, due to proteasome-mediated degradation. The effect of the R1/R2 ratio on proliferation was assessed in GH3 cells, which showed a similar unbalanced pattern of R subunits expression, and in growth hormone-secreting adenomas. The R2-selective cAMP analog 8-Cl cAMP and R1A RNA silencing, stimulated cell proliferation and increased Cyclin D1 expression, respectively, in human and rat adenomatous somatotrophs. These data show that a low R1/R2 ratio promoted proliferation of transformed somatotrophs and are consistent with the Carney complex model in which R1A inactivating mutations further unbalance this ratio in favor of R2 subunits. These results suggest that low expression of R1A protein may favor cAMP-dependent proliferation of transformed somatotrophs.


Asunto(s)
Adenoma/enzimología , Adenoma/patología , Neoplasias Hipofisarias/enzimología , Neoplasias Hipofisarias/patología , Proteínas/metabolismo , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , AMP Cíclico/fisiología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico , Ciclina D1/biosíntesis , Activación Enzimática , Exones , Humanos , Inmunohistoquímica , Intrones , Lisosomas/efectos de los fármacos , Inhibidores de Proteasoma , Subunidades de Proteína , Proteínas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
9.
Mol Cell Endocrinol ; 238(1-2): 51-5, 2005 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-15921845

RESUMEN

Heterozygous loss of function mutations in human PKAR1A gene (PRKAR1A) have been identified in patients with Carney complex (CNC), an autosomal dominant familial multiple neoplasia syndrome displaying different endocrine tumors, including adrenocortical tumors, GH-secreting pituitary tumors and thyroid adenomas. Although PRKAR1A is encoded by a single gene, it is transcribed from at least two different promoters, adjacent to different first non-coding exons (1a and 1b), giving rise to alternately spliced transcripts coding for identical proteins. The separate regulation of the two distinct promoters and the presence of multiple alternatively spliced first exons suggest a complex mechanism of PRKAR1A expression regulation. In order to investigate the relative expression of the two mRNA transcripts (1a and 1b) in human adult endocrine tissues involved in the determination of CNC phenotype, we selected 17 pituitary, 20 adrenal and seven thyroid tissues from tumoral and peri-tumoral lesion samples. Expression of the two transcripts was evaluated by semi-quantitative RT-PCR and real-time RT-PCR. This study first reports that human pituitary and thyroid tissues show a similar expression of the two transcripts, whereas in adrenal tissues transcript 1b is the most abundant one.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Hipofisarias/genética , Proteínas/genética , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de las Glándulas Suprarrenales/metabolismo , Empalme Alternativo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico , Humanos , Mutación , Neoplasias Hipofisarias/metabolismo , Regiones Promotoras Genéticas , Proteínas/metabolismo , ARN Mensajero/análisis , ARN Neoplásico/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias de la Tiroides/metabolismo
10.
J Clin Endocrinol Metab ; 89(6): 3007-9, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15181091

RESUMEN

Activating mutations of the Gsalpha gene are detected in different endocrine tumors, such as GH-secreting adenomas and toxic thyroid adenomas, and in hyperfunctioning glands from patients with McCune-Albright syndrome (MAS). There is increasing evidence that the Gsalpha gene is subjected to imprinting control and that Gsalpha imprinting plays a key role in the pathogenesis of different human diseases. The aim of this study was to investigate the presence of a parent specificity of Gsalpha mutations in 10 patients affected with MAS and 12 isolated tumors (10 GH-secreting adenomas, one toxic thyroid adenoma, and one hyperfunctioning adrenal adenoma). The parental origin of Gsalpha mutations was assessed by evaluating NESP55 and exon 1A transcripts, which are monoallelically expressed from the maternal and paternal alleles, respectively. By this approach, we demonstrated that in isolated GH-secreting adenomas, as well as in MAS patients with acromegaly, Gsalpha mutations were on the maternal allele. By contrast, the involvement of other endocrine organs in MAS patients was not associated with a particular parent specificity, as precocious puberty and hyperthyroidism were present in patients with mutations on either the maternal or the paternal allele. Moreover, isolated hyperfunctioning thyroid and adrenal adenomas displayed the mutation on the maternal and paternal alleles, respectively. These data confirm the importance of Gsalpha imprinting in the pituitary gland and point out the high degree of tissue specificity of this phenomenon.


Asunto(s)
Adenoma/genética , Displasia Fibrosa Poliostótica/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Neoplasias Hipofisarias/genética , Neoplasias de la Tiroides/genética , Adolescente , Neoplasias de las Glándulas Suprarrenales/genética , Adulto , Niño , Padre , Femenino , Impresión Genómica , Humanos , Masculino , Persona de Mediana Edad , Madres , Mutación Missense
11.
J Clin Endocrinol Metab ; 89(12): 6316-9, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15579796

RESUMEN

Mutations of the Gsalpha gene inherited from the mother lead to pseudohypoparathyroidism (PHP) type Ia (PHP Ia), in which Albright's hereditary osteodistrophy is associated to resistance to the action of different hormones, whereas the same mutations inherited from the father lead to isolated Albright's hereditary osteodistrophy [pseudo-PHP (PPHP)]. Accordingly, it has been suggested that Gsalpha is under tissue-specific imprinting control, and recent studies provided evidence for a predominant maternal origin of Gsalpha transcripts in different endocrine organs involved in the PHP Ia phenotype. To establish whether Gsalpha is imprinted also in tissues that are site of alteration both in PHP Ia and PPHP, we selected 20 bone and 10 adipose tissue samples, which were heterozygous for a known polymorphism in exon 5. Expression from both parental alleles was evaluated by RT-PCR and enzymatic digestion of the resulting fragments. By this approach, the great majority of the samples analyzed showed an equal expression of the two alleles. Our results provide evidence for the absence of Gsalpha imprinting in human bone and fat and suggest that the clinical finding of osteodystrophy and obesity in PHP Ia and PPHP patients despite the presence of a normal Gsalpha allele is likely due to Gsalpha haploinsufficiency in these tissues.


Asunto(s)
Tejido Adiposo/metabolismo , Alelos , Huesos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Expresión Génica , Tejido Adiposo/embriología , Huesos/embriología , Exones/genética , Feto/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Heterocigoto , Humanos , Polimorfismo Genético , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
J Clin Endocrinol Metab ; 95(2): 651-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20061437

RESUMEN

CONTEXT: The two main subtypes of pseudohypoparathyroidism (PHP), PHP-Ia and -Ib, are caused by mutations in GNAS exons 1-13 and methylation defects in the imprinted GNAS cluster, respectively. PHP-Ia patients show Albright hereditary osteodystrophy (AHO) and resistance toward PTH and additional hormones, whereas PHP-Ib patients do not have AHO, and hormone resistance appears to be limited to PTH and TSH. Recently, methylation defects have been detected in few patients with PHP and mild AHO, indicating a molecular overlap between the two forms. OBJECTIVES: The aim of the study was to screen patients with clinically diagnosed PHP-Ia for methylation defects and to investigate the presence of correlations between the molecular findings and AHO severity. PATIENTS AND METHODS: We investigated differential methylation of GNAS regions and STX16 microdeletions in genomic DNA from 40 patients with sporadic AHO and multihormone resistance, with no mutations in Gsalpha-coding GNAS exons. RESULTS: Molecular analysis showed GNAS cluster imprinting defects in 24 of the 40 patients analyzed. No STX16 deletion was detected. The presence of imprinting defects was not associated with the severity of AHO or with specific AHO signs. CONCLUSIONS: We report the largest series of the literature of patients with clinical AHO and multihormone resistance and no mutation in the Gsalpha gene. Our findings of frequent GNAS imprinting defects further confirm the existence of an overlap between molecular and clinical features of PHP-Ia and PHP-Ib and highlight the necessity of a new clinical classification of the disease that takes into account the recent knowledge on the molecular basis underlying these defects.


Asunto(s)
Epigénesis Genética , Displasia Fibrosa Poliostótica/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Seudohipoparatiroidismo/genética , Adolescente , Adulto , Niño , Cromograninas , Metilación de ADN , Femenino , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Seudohipoparatiroidismo/clasificación , Sintaxina 16/genética
13.
Diabetes ; 58(3): 620-6, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19095761

RESUMEN

OBJECTIVE: In human adipocytes, the cAMP-dependent pathway mediates signals originating from beta-adrenergic activation, thus playing a key role in the regulation of important metabolic processes, i.e., lipolysis and thermogenesis. Cyclic AMP effects are mainly mediated by protein kinase A (PKA), whose R2B regulatory isoform is the most expressed in mouse adipose tissue, where it protects against diet-induced obesity and fatty liver development. The aim of the study was to investigate possible differences in R2B expression, PKA activity, and lipolysis in adipose tissues from obese and nonobese subjects. RESEARCH DESIGN AND METHODS: The expression of the different PKA regulatory subunits was evaluated by immunohistochemistry, Western blot, and real-time PCR in subcutaneous and visceral adipose tissue samples from 20 nonobese and 67 obese patients. PKA activity and glycerol release were evaluated in total protein extract and adipocytes isolated from fresh tissue samples, respectively. RESULTS: Expression techniques showed that R2B was the most abundant regulatory protein, both at mRNA and protein level. Interestingly, R2B mRNA levels were significantly lower in both subcutaneous and visceral adipose tissues from obese than nonobese patients and negatively correlated with BMI, waist circumference, insulin levels, and homeostasis model assessment of insulin resistance. Moreover, both basal and stimulated PKA activity and glycerol release were significantly lower in visceral adipose tissue from obese patients then nonobese subjects. CONCLUSIONS: Our results first indicate that, in human adipose tissue, there are important BMI-related differences in R2B expression and PKA activation, which might be included among the multiple determinants involved in the different lipolytic response to beta-adrenergic activation in obesity.


Asunto(s)
Tejido Adiposo/enzimología , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Obesidad/enzimología , Adipocitos/enzimología , Tejido Adiposo/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación Enzimológica de la Expresión Génica , Glicerol/metabolismo , Humanos , Inmunohistoquímica , Obesidad/genética , Obesidad/fisiopatología , Reacción en Cadena de la Polimerasa , Subunidades de Proteína/genética , ARN Mensajero/genética , Piel/enzimología
14.
Clin Endocrinol (Oxf) ; 62(1): 30-6, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15638867

RESUMEN

BACKGROUND: Ghrelin, an endogenous ligand of the GH secretagogue receptor that exerts orexigenic activity, is negatively correlated with body mass index (BMI) and insulin resistance. Conversely, low levels of adiponectin (ApN), a circulating adipocytokine with antidiabetic, antiatherogenic and anti-inflammatory properties, have been found in several insulin-resistant conditions. Although Cushing's syndrome causes several metabolic and hormonal changes leading to insulin resistance and central obesity, few data concerning the impact of glucocorticoid excess on ghrelin and ApN levels are so far available. DESIGN: We evaluated ghrelin and ApN levels in 14 women (age +/- SE 39.5 +/- 3.9 years, BMI +/- SE 25.8 +/- 1.4 kg/m2) with Cushing's disease (CD) at baseline and after successful transsphenoidal surgery (TSS) and in 14 age- and BMI-matched healthy women. RESULTS: Despite similar levels of fasting glucose, insulin, homeostatic model assessment-estimated insulin resistance (HOMA-IR) and quantitative insulin sensitivity check index (QUICKI) values, patients with CD had ghrelin levels lower than controls (117.8 +/- 21.5 vs. 269.6 +/- 51.4 pmol/l, P < 0.01), and ghrelin levels did not correlate with ACTH, cortisol, androgen and GH levels. Patients and controls showed similar ApN levels (11.1 +/- 1.6 vs. 11.5 +/- 2.0 mg/l), which correlated negatively with insulin, HOMA-IR and BMI and positively with QUICKI and high density lipoprotein (HDL)-cholesterol only in controls. At 10.2 +/- 0.7 months after successful TSS, patients showed a significant increase in ghrelin levels compared to pretreatment values (342.5 +/- 25.6 vs. 117.8 +/- 21.5 pmol/l, P < 0.005) along with significant modifications in BMI, insulin, HOMA-IR and HDL-cholesterol and no change in ApN levels. In two patients tested on days 2-4 after TSS, no modification in ghrelin and ApN levels was observed, despite a dramatic reduction in cortisol levels. CONCLUSION: Cortisol excess did not directly affect ghrelin and ApN levels in patients with CD. The observation that ghrelin levels were low during the active phase of CD and increased after recovery suggests that glucocorticoids may influence ghrelin levels indirectly by modulating adiposity and metabolic signals over the long term.


Asunto(s)
Síndrome de Cushing/sangre , Péptidos y Proteínas de Señalización Intercelular/sangre , Hormonas Peptídicas/sangre , Enfermedad Aguda , Adiponectina , Adulto , Estudios de Casos y Controles , Síndrome de Cushing/cirugía , Femenino , Estudios de Seguimiento , Ghrelina , Humanos , Hidrocortisona/sangre , Hipofisectomía , Periodo Posoperatorio , Análisis de Regresión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA