Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Mol Cell Endocrinol ; 522: 111117, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33338547

RESUMEN

PDE8B, PRKAR1A and the Wnt/ß-catenin signaling are involved in endocrine disorders. However, how PDEB8B interacts with both Wnt and protein kinase A (PKA) signaling in vivo remains unknown. We created a novel Pde8b knockout mouse line (Pde8b-/-); Pde8b haploinsufficient (Pde8b+/-) mice were then crossed with mice harboring: (1) constitutive beta-catenin activation (Pde8b+/-;ΔCat) and (2) Prkar1a haploinsufficieny (Pde8b+/-;Prkar1a+/-). Adrenals and testes from mice (3-12-mo) were evaluated in addition to plasma corticosterone, aldosterone and Dkk3 concentrations, and the examination of expression of steroidogenesis-, Wnt- and cAMP/PKA-related genes. Pde8b-/- male mice were infertile with down-regulation of the Wnt/ß-catenin pathway which did not change significantly in the Pde8b+/-;ΔCat mice. Prkar1a haploinsufficiency also did not change the phenotype significantly. In vitro studies showed that PDE8B knockdown upregulated the Wnt pathway and increased proliferation in CTNNB1-mutant cells, whereas it downregulated the Wnt pathway in PRKAR1A-mutant cells. These data support an overall weak, if any, role for PDE8B in adrenocortical tumorigenesis, even when co-altered with Wnt signaling or PKA upregulation; on the other hand, PDE8B appears to play a significant role in male fertility.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/genética , Glándulas Suprarrenales/patología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Haploinsuficiencia/genética , Infertilidad Masculina/genética , Esteroides/biosíntesis , Proteínas Wnt/metabolismo , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/sangre , Glándulas Suprarrenales/efectos de los fármacos , Glándulas Suprarrenales/fisiopatología , Aldosterona/sangre , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Corticosterona/sangre , Cruzamientos Genéticos , Dexametasona/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Infertilidad Masculina/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Espermatogénesis/efectos de los fármacos , Espermatogénesis/genética , Testículo/efectos de los fármacos , Testículo/ultraestructura , beta Catenina/metabolismo
2.
Ultramicroscopy ; 218: 113083, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32739752

RESUMEN

Fracturing microscale constrictions in metallic wires, such as tungsten, platinum, or platinum-iridium, is a common fabrication method used to produce atomically sharp tips for scanning tunneling microscopy (STM), field-emission microscopy and field ion microscopy. Typically, a commercial polycrystalline drawn wire is locally thinned and then fractured by means of a dislocation slip inside the constriction. We examine a special case where a dislocation-free microscale constriction is created and fractured in a single crystal tungsten rod with a long side parallel to the [100] direction. In the absence of dislocations, vacancies become the main defects in the constriction which breaks under the tensile stress of approximately 10 GPa, which is close to the theoretical fracture strength for an ideal monocrystalline tungsten. We propose that the vacancies are removed early in the tensile test by means of deformation annealing, creating a defect-free tungsten constriction which cleaves along the W(100) plane. This approach enables fabrication of new composite STM probes which demonstrate excellent stability, atomic resolution and magnetic contrast that cannot be attained using conventional methods.

3.
J Mol Endocrinol ; 59(1): 1-12, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28420713

RESUMEN

The cAMP-dependent protein kinase (PKA) is an essential regulator of lipid and glucose metabolism that plays a critical role in energy homeostasis. The impact of diet on PKA signaling has not been defined, although perturbations in individual PKA subunits are associated with changes in adiposity, physical activity and energy intake in mice and humans. We hypothesized that a high fat diet (HFD) would elicit peripheral and central alterations in the PKA system that would differ depending on length of exposure to HFD; these differences could protect against or promote diet-induced obesity (DIO). 12-week-old C57Bl/6J mice were randomly assigned to a regular diet or HFD and weighed weekly throughout the feeding studies (4 days, 14 weeks; respectively), and during killing. PKA activity and subunit expression were measured in liver, gonadal adipose tissue (AT) and brain. Acute HFD-feeding suppressed basal hepatic PKA activity. In contrast, hepatic and hypothalamic PKA activities were significantly increased after chronic HFD-feeding. Changes in AT were more subtle, and overall, altered PKA regulation in response to chronic HFD exposure was more profound in female mice. The suppression of hepatic PKA activity after 4 day HFD-feeding was indicative of a protective peripheral effect against obesity in the context of overnutrition. In response to chronic HFD-feeding, and with the development of DIO, dysregulated hepatic and hypothalamic PKA signaling was a signature of obesity that is likely to promote further metabolic dysfunction in mice.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/genética , Dieta Alta en Grasa/métodos , Grasas de la Dieta/efectos adversos , Obesidad/genética , Subunidades de Proteína/genética , Tejido Adiposo/enzimología , Tejido Adiposo/patología , Animales , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Femenino , Regulación de la Expresión Génica , Homeostasis , Hipotálamo/enzimología , Hipotálamo/patología , Hígado/enzimología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/enzimología , Obesidad/etiología , Obesidad/patología , Especificidad de Órganos , Ovario/enzimología , Ovario/patología , Subunidades de Proteína/metabolismo , Factores Sexuales , Testículo/enzimología , Testículo/patología
4.
Mol Cell Endocrinol ; 439: 165-174, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-27498419

RESUMEN

Osteochondromyxomas (OMX) in the context of Carney complex (CNC) and fibrous dysplasia (FD)-like lesions (FDLL) in mice, as well as isolated myxomas in humans may be caused by inactivation of PRKAR1A, the gene coding for the type 1a regulatory subunit (R1α) of cAMP-dependent protein kinase (PKA). OMXs and FDLL in mice lacking Prkar1a grow from abnormal proliferation of adult bone stromal cells (aBSCs). Prkar1a and Prkaca (coding for Cα) haploinsufficiency leads to COX2 activation and prostaglandin E2 (PGE2) production that, in turn, activates proliferation of aBSCs. Celecoxib is a cyclooxygenase-2 (COX2) inhibitor. We hypothesized that COX-2 inhibition may have an effect in FD and FDLL. In vitro treatment of a human cell line prepared from a FD patient with Celecoxib resulted in decreased PGE2 and cell proliferation. Treatment of mice haploinsufficient for R1α and Cα with 1500 mg/kg Celecoxib led to decreased PGE2 and proliferation and increased apoptosis, with a corresponding gene expression profile, resulting in dramatic reduction of tumor growth. Furthermore, the treatment improved the organization of cortical bone that was adjacent to the tumor. We conclude that, in vitro and in vivo, Celecoxib had an inhibitory effect on FD cell proliferation and in mouse FDLL structure, respectively. We speculate that COX-2 inhibitors offer an attractive alternative to current treatments for benign tumors such as OMX and FD that, apart from tumor suppression, may mechanically stabilize affected bones.


Asunto(s)
Celecoxib/uso terapéutico , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Displasia Fibrosa Ósea/tratamiento farmacológico , Displasia Fibrosa Ósea/enzimología , Animales , Apoptosis/efectos de los fármacos , Huesos/efectos de los fármacos , Huesos/patología , Celecoxib/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Humanos , Inflamasomas/metabolismo , Ligandos , Ratones , Vía de Señalización Wnt/efectos de los fármacos
5.
J Clin Endocrinol Metab ; 101(9): 3353-60, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27336356

RESUMEN

CONTEXT: Androgen excess may be adrenal and/or ovarian in origin; we hypothesized that a subgroup of patients with polycystic ovarian syndrome (PCOS) may have some degree of abnormal adrenocortical function. OBJECTIVE: The objective of the study was to evaluate the pituitary adrenal axis with an oral low- and high-dose dexamethasone-suppression test (Liddle's test) in women with PCOS. DESIGN: This was a case-control study. SETTING: The study was conducted at the National Institutes of Health Clinical Center. PARTICIPANTS: A total of 38 women with PCOS and 20 healthy volunteers (HV) aged 16-29 years participated in the study. MAIN OUTCOME MEASURES: Urinary free cortisol (UFC) and 17-hydroxysteroids (17OHS) before and after low- and high-dose dexamethasone and assessment of adrenal volume by computed tomography scan were measured. RESULTS: Twenty-four-hour urinary 17OHS and UFC were measured during day 1 to day 6 of the Liddle's test. Baseline UFC levels were not different between PCOS and HVs; on the day after the completion of high-dose dexamethasone administration (d 6), UFC was higher in the PCOS group (2.0 ± 0.7 µg/m(2)·d) than the HV group (1.5 ± 0.5) (P = .038). On day 5, 17OHS and UFC were negatively correlated with adrenal volumes (left side, rp = -0.47, P = .009, and rp = -0.61, P < .001, respectively). PCOS patients above the 75th percentile for UFC and/or 17OHS after high-dose dexamethasone (n = 15) had a significantly smaller total adrenal volume (6.9 ± 1.9 cm(3) vs 9.2 ± 1.8 cm(3), P = .003) when compared with the remaining PCOS patients (n = 22), but they did not have worse insulin resistance or hyperandrogenism. CONCLUSIONS: In a subset of young women with PCOS, we detected a pattern of glucocorticoid secretion that mimicked that of patients with micronodular adrenocortical hyperplasia: they had smaller adrenal volumes and higher steroid hormone secretion after dexamethasone compared with the group of PCOS with appropriate response to dexamethasone.


Asunto(s)
Glándulas Suprarrenales/patología , Hiperandrogenismo/etiología , Hiperplasia/patología , Síndrome del Ovario Poliquístico/complicaciones , Adolescente , Glándulas Suprarrenales/metabolismo , Adulto , Andrógenos/metabolismo , Biomarcadores/metabolismo , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Humanos , Hiperandrogenismo/metabolismo , Hiperandrogenismo/patología , Hiperplasia/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Pronóstico , Adulto Joven
6.
Behav Brain Res ; 307: 1-10, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-26992826

RESUMEN

Cyclic adenosine mono-phosphate-dependent protein kinase (PKA) is critically involved in the regulation of behavioral responses. Previous studies showed that PKA's main regulatory subunit, R1α, is involved in anxiety-like behaviors. The purpose of this study was to determine how the catalytic subunit, Cα, might affect R1α's function and determine its effects on anxiety-related behaviors. The marble bury (MB) and elevated plus maze (EPM) tests were used to assess anxiety-like behavior and the hotplate test to assess nociception in wild type (WT) mouse, a Prkar1a heterozygote (Prkar1a(+/-)) mouse with haploinsufficiency for the regulatory subunit (R1α), a Prkaca heterozygote (Prkaca(+/-)) mouse with haploinsufficiency for the catalytic subunit (Cα), and a double heterozygote mouse (Prkar1a(+/-)/Prkaca(+/-)) with haploinsufficiency for both R1α and Cα. We then examined specific brain nuclei involved in anxiety. Results of MB test showed a genotype effect, with increased anxiety-like behavior in Prkar1a(+/-) and Prkar1a(+/-)/Prkaca(+/-) compared to WT mice. In the EPM, Prkar1a(+/-) spent significantly less time in the open arms, while Prkaca(+/-) and Prkar1a(+/-)/Prkaca(+/-) mice displayed less exploratory behavior compared to WT mice. The loss of one Prkar1a allele was associated with a significant increase in PKA activity in the basolateral (BLA) and central (CeA) amygdala and ventromedial hypothalamus (VMH) in both Prkar1a(+/-) and Prkar1a(+/-)/Prkaca(+/-) mice. Alterations of PKA activity induced by haploinsufficiency of its main regulatory or most important catalytic subunits result in anxiety-like behaviors. The BLA, CeA, and VMH are implicated in mediating these PKA effects in brain.


Asunto(s)
Ansiedad/genética , Ansiedad/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Animales , Ansiedad/fisiopatología , Encéfalo/metabolismo , AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Genotipo , Hiperalgesia/genética , Hiperalgesia/fisiopatología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nocicepción/fisiología , Dimensión del Dolor
7.
Hum Mol Genet ; 24(21): 6080-92, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26246497

RESUMEN

Carney Complex (CNC), a human genetic syndrome predisposing to multiple neoplasias, is associated with bone lesions such as osteochondromyxomas (OMX). The most frequent cause for CNC is PRKAR1A deficiency; PRKAR1A codes for type-I regulatory subunit of protein kinase A (PKA). Prkar1a(+/-) mice developed OMX, fibrous dysplasia-like lesions (FDL) and other tumors. Tumor tissues in these animals had increased PKA activity due to an unregulated PKA catalytic subunit and increased PKA type II (PKA-II) activity mediated by the PRKAR2A and PRKAR2B subunits. To better understand the effect of altered PKA activity on bone, we studied Prkar2a and Prkar2b knock out (KO) and heterozygous mice; none of these mice developed bone lesions. When Prkar2a(+/-) and Prkar2b(+/-) mice were used to generate Prkar1a(+/-)Prkar2a(+/-) and Prkar1a(+/-)Prkar2b(+/-) animals, bone lesions formed that looked like those of the Prkar1a(+/-) mice. However, better overall bone organization and mineralization and fewer FDL lesions were found in both double heterozygote groups, indicating a partial restoration of the immature bone structure observed in Prkar1a(+/-) mice. Further investigation indicated increased osteogenesis and higher new bone formation rates in both Prkar1a(+/-)Prkar2a(+/-) and Prkar1a(+/-)Prkar2b(+/-) mice with some minor differences between them. The observations were confirmed with a variety of markers and studies. PKA activity measurements showed the expected PKA-II decrease in both double heterozygote groups. Thus, haploinsufficiency for either of PKA-II regulatory subunits improved bone phenotype of mice haploinsufficient for Prkar1a, in support of the hypothesis that the PRKAR2A and PRKAR2B regulatory subunits were in part responsible for the bone phenotype of Prkar1a(+/-) mice.


Asunto(s)
Huesos/patología , Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Haploinsuficiencia , Animales , Antígenos de Diferenciación/biosíntesis , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Huesos/metabolismo , Calcificación Fisiológica , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Heterocigoto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/metabolismo , Osteogénesis , Fenotipo , Isoformas de Proteínas/metabolismo , Células Tumorales Cultivadas
8.
Endocr Relat Cancer ; 22(1): 47-54, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25527509

RESUMEN

IGSF1 is a membrane glycoprotein highly expressed in the anterior pituitary. Pathogenic mutations in the IGSF1 gene (on Xq26.2) are associated with X-linked central hypothyroidism and testicular enlargement in males. In this study, we tested the hypothesis that IGSF1 is involved in the development of pituitary tumors, especially those that produce growth hormone (GH). IGSF1 was sequenced in 21 patients with gigantism or acromegaly and 92 healthy individuals. Expression studies with a candidate pathogenic IGSF1 variant were carried out in transfected cells and immunohistochemistry for IGSF1 was performed in the sections of GH-producing adenomas, familial somatomammotroph hyperplasia, and in normal pituitary. We identified the sequence variant p.N604T, which in silico analysis suggested could affect IGSF1 function, in two male patients and one female with somatomammotroph hyperplasia from the same family. Of 60 female controls, two carried the same variant and seven were heterozygous for other variants. Immunohistochemistry showed increased IGSF1 staining in the GH-producing tumor from the patient with the IGSF1 p.N604T variant compared with a GH-producing adenoma from a patient negative for any IGSF1 variants and with normal control pituitary tissue. The IGSF1 gene appears polymorphic in the general population. A potentially pathogenic variant identified in the germline of three patients with gigantism from the same family (segregating with the disease) was also detected in two healthy female controls. Variations in IGSF1 expression in pituitary tissue in patients with or without IGSF1 germline mutations point to the need for further studies of IGSF1 action in pituitary adenoma formation.


Asunto(s)
Hormona de Crecimiento Humana/biosíntesis , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Acromegalia/genética , Animales , Estudios de Casos y Controles , Línea Celular , Femenino , Mutación de Línea Germinal , Gigantismo/genética , Células HEK293 , Humanos , Inmunoglobulinas/química , Masculino , Proteínas de la Membrana/química , Modelos Moleculares , Ratas , Transfección
9.
Endocrinology ; 155(9): 3397-408, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24914943

RESUMEN

The cAMP-dependent protein kinase A (PKA) signaling system is widely expressed and has a central role in regulating cellular metabolism in all organ systems affected by obesity. PKA has four regulatory (RIα, RIIα, RIß, RIIß) and four catalytic (Cα, Cß, Cγ, Prkx) subunit isoforms that have tissue-specific expression profiles. In mice, knockout (KO) of RIIß, the primary PKA regulatory subunit in adipose tissue or knockout of the catalytic subunit Cß resulted in a lean phenotype that resists diet-induced obesity and associated metabolic complications. Here we report that the disruption of the ubiquitously expressed PKA RIIα subunit in mice (RIIαKO) confers resistance to diet-induced obesity, glucose intolerance, and hepatic steatosis. After 2-week high-fat diet exposure, RIIαKO mice weighed less than wild-type littermates. Over time this effect was more pronounced in female mice that were also leaner than their wild-type counterparts, regardless of the diet. Decreased intake of a high-fat diet contributed to the attenuated weight gain in RIIαKO mice. Additionally, RIIα deficiency caused differential regulation of PKA in key metabolic organs: cAMP-stimulated PKA activity was decreased in liver and increased in gonadal adipose tissue. We conclude that RIIα represents a potential target for therapeutic interventions in obesity, glucose intolerance, and nonalcoholic fatty liver disease.


Asunto(s)
Tejido Adiposo/enzimología , Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hígado Graso/enzimología , Hígado Graso/prevención & control , Intolerancia a la Glucosa/prevención & control , Hígado/enzimología , Obesidad/prevención & control , Tejido Adiposo/metabolismo , Animales , Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Dieta Alta en Grasa/efectos adversos , Hígado Graso/genética , Femenino , Intolerancia a la Glucosa/enzimología , Intolerancia a la Glucosa/genética , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico , Obesidad/enzimología , Obesidad/genética
10.
J Clin Endocrinol Metab ; 99(5): E891-901, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24512486

RESUMEN

CONTEXT: Somatostatin (SST) receptors (SSTRs) are expressed in a number of tissues, including the adrenal cortex, but their role in cortisol secretion has not been well characterized. OBJECTIVES: The objective of the study was to investigate the expression of SSTRs in the adrenal cortex and cultured adrenocortical cells from primary pigmented nodular adrenocortical disease (PPNAD) tissues and to test the effect of a single injection of 100 µg of the SST analog octreotide on cortisol secretion in patients with PPNAD. SETTING AND DESIGN: The study was conducted at an academic research laboratory and clinical research center. Expression of SSTRs was examined in 26 PPNAD tissues and the immortalized PPNAD cell line CAR47. Ten subjects with PPNAD underwent a randomized, single-blind, crossover study of their cortisol secretion every 30 minutes over 12 hours (6:00 pm to 6:00 am) before and after the midnight administration of octreotide 100 µg sc. METHODS: SSTRs expression was investigated by quantitative PCR and immunohistochemistry. The CAR47 and primary cell lines were studied in vitro. The data of the 10 patients were analyzed before and after the administration of octreotide. RESULTS: All SSTRs, especially SSTR1-3, were expressed in PPNAD at significantly higher levels than in normal adrenal. SST was found to differentially regulate expression of its own receptors in the CAR47 cell line. However, the administration of octreotide to patients with PPNAD did not significantly affect cortisol secretion. CONCLUSIONS: SSTRs are overexpressed in PPNAD tissues in comparison with normal adrenal cortex. Octreotide did not exert any significant effect on cortisol secretion in a short clinical pilot study in a small number of patients with PPNAD, but long-acting SST analogs targeting multiple SSTRs may be worth investigating in this condition.


Asunto(s)
Enfermedades de la Corteza Suprarrenal/metabolismo , Corteza Suprarrenal/metabolismo , Hidrocortisona/metabolismo , Trastornos de la Pigmentación/metabolismo , Receptores de Somatostatina/metabolismo , Adolescente , Corteza Suprarrenal/efectos de los fármacos , Adulto , Línea Celular , Niño , Preescolar , Estudios Cruzados , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Octreótido/farmacología , Método Simple Ciego
11.
J Clin Endocrinol Metab ; 99(2): E303-10, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24248186

RESUMEN

CONTEXT: The cAMP signaling pathway is implicated in bilateral adrenocortical hyperplasias. Bilateral adrenocortical hyperplasia is often associated with ACTH-independent Cushing syndrome (CS) and may be caused by mutations in genes such as PRKAR1A, which is responsible for primary pigmented nodular adrenocortical disease (PPNAD). PRKAR1A regulates cAMP-dependent protein kinase (PKA), an essential enzyme in the regulation of adiposity. Although CS is invariably associated with obesity, its different forms, including those associated with PKA defects, have not been compared. OBJECTIVE: The purpose of this study was to characterize the phenotypic and molecular differences in periadrenal adipose tissue (PAT) between patients with CS with and without PRKAR1A mutations. DESIGN AND SETTING: Samples from adrenalectomies of 51 patients were studied: patients with CS with (n = 13) and without (n = 32) PRKAR1A mutations and a comparison group with aldosterone-producing adenomas (APAs) (n = 6). In addition, clinical data from a larger group of patients with Cushing disease (n = 89) and hyperaldosteronism (n = 26) were used for comparison. METHODS: Body mass index (BMI), abdominal computed tomography scans, and cortisol data were collected preoperatively. PAT was assayed for PKA activity, cAMP levels, and PKA subunit expression. RESULTS: BMI was lower in adult patients with CS with PRKAR1A mutations. cAMP and active PKA levels in PAT were elevated in patients with CS with PRKAR1A mutations. CONCLUSIONS: Increased PKA signaling in PAT was associated with lower BMI in CS. Differences in fat distribution may contribute to phenotypic differences between patients with CS with and without PRKAR1A mutations. The observed differences are in agreement with the known roles of cAMP signaling in regulating adiposity, but this is the first time that germline defects of PKA are linked to variable obesity phenotypes in humans.


Asunto(s)
Adiposidad/genética , Síndrome de Cushing/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , AMP Cíclico/metabolismo , Obesidad/genética , Tejido Adiposo/metabolismo , Adolescente , Adulto , Anciano , Niño , Preescolar , Síndrome de Cushing/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Obesidad/metabolismo , Estudios Retrospectivos
12.
J Clin Endocrinol Metab ; 98(8): E1393-400, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23771924

RESUMEN

BACKGROUND: Familial testicular germ cell tumors (FTGCTs) are hypothesized to result from the combined interaction of multiple low-penetrance genes. We reported inactivating germline mutations of the cAMP-binding phosphodiesterase 11A (PDE11A) as modifiers of FTGCT risk. Recent genome-wide association studies have identified single-nucleotide polymorphisms in the KITLG gene, the ligand for the cKIT tyrosine kinase receptor, as strong modifiers of susceptibility to both familial and sporadic testicular germ cell tumors. DESIGN: We studied 94 patients with FTGCTs and 50 at-risk male relatives from 63 unrelated kindreds, in whom the PDE11A gene had been sequenced by investigating the association between KITLG genome-wide association study single-nucleotide polymorphisms rs3782179 and rs4474514 and FTGCT risk in these patients and in 692 controls. We also examined cAMP and c-KIT signaling in testicular tissues and cell lines and extended the studies to 2 sporadic cases, one with a PDE11A defect and one without, as a comparison. RESULTS: We found a higher frequency of the KITLG risk alleles in FTGCT patients who also had a PDE11A sequence variant, compared with those with a wild-type PDE11A sequence. In NTERA-2 and Tcam-2 cells transfected with the mutated forms of PDE11A (R52T, F258Y, Y727C, R804H, V820M, R867G, and M878V), cAMP levels were significantly higher, and the relative phosphodiesterase activity was lower than in the wild-type cells. KITLG expression was consistently increased in the presence of PDE11A-inactivating defects, both at the RNA and protein levels, in familial testicular germ cell tumors. The 2 sporadic cases that were studied, one with a PDE11A defect and another without, agreed with the data in FTGTCT and in the cell lines. CONCLUSIONS: Patients with FTGCT and PDE11A defects also carry KITLG risk alleles more frequently. There may be an interaction between cAMP and c-KIT signaling in predisposition to testicular germ cell tumors.


Asunto(s)
AMP Cíclico/fisiología , Neoplasias de Células Germinales y Embrionarias/genética , Hidrolasas Diéster Fosfóricas/genética , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-kit/fisiología , Transducción de Señal/fisiología , Factor de Células Madre/genética , Neoplasias Testiculares/genética , 3',5'-GMP Cíclico Fosfodiesterasas , Línea Celular Tumoral , AMP Cíclico/análisis , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Neoplasias de Células Germinales y Embrionarias/etiología , Hidrolasas Diéster Fosfóricas/análisis , Proteínas Proto-Oncogénicas c-kit/análisis , Factor de Células Madre/análisis , Neoplasias Testiculares/etiología
13.
J Am Soc Mass Spectrom ; 24(2): 230-7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23283727

RESUMEN

We have developed new applications of the pseudocolor plot for the analysis of LC/MS data. These applications include spectral averaging, analysis of variance, differential comparison of spectra, and qualitative filtering by compound class. These applications have been motivated by the need to better understand LC/MS data generated from analysis of human biofluids. The examples presented use data generated to profile steroid hormones in urine extracts from a Cushing's disease patient relative to a healthy control, but are general to any discovery-based scanning mass spectrometry technique. In addition to new visualization techniques, we introduce a new metric of variance: the relative maximum difference from the mean. We also introduce the concept of substructure-dependent analysis of steroid hormones using precursor ion scans. These new analytical techniques provide an alternative approach to traditional untargeted metabolomics workflow. We present an approach to discovery using MS that essentially eliminates alignment or preprocessing of spectra. Moreover, we demonstrate the concept that untargeted metabolomics can be achieved using low mass resolution instrumentation.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Espectrometría de Masas/métodos , Análisis de Varianza , Estudios de Casos y Controles , Hormonas/orina , Humanos , Metabolómica/métodos , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/orina , Esteroides/orina
14.
Clin Endocrinol (Oxf) ; 77(2): 195-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22335482

RESUMEN

BACKGROUND: Genetic aberrations in various components of cAMP signalling pathway predispose to endocrine tumours. Mutations in the phosphodiesterases (PDEs) are involved in the predisposition to adrenocortical neoplastic conditions. OBJECTIVE: To screen for genetic variations in PDE8B among patients with different types of adrenocortical tumours. DESIGN AND SUBJECTS: This is a case-control study followed by functional analyses. Two hundred and sixteen unrelated patients with different types of adrenocortical tumours and 192 healthy control individuals participated in the study. METHODS: Bidirectional Sanger sequencing, in vitro cell line transfection and in silico modelling are used in this study. RESULTS: Nine different PDE8B sequence changes, six novel and three previously reported, were identified in our patients and controls. Two of the variations, seen only in the patient group, showed significant potential to impair protein function, both in vitro and in silico. CONCLUSION: PDE8B is another PDE gene in which variations may contribute to predisposition of adrenocortical tumours.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/genética , Corteza Suprarrenal/metabolismo , Neoplasias de las Glándulas Suprarrenales/genética , Variación Genética/genética , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Clin Endocrinol Metab ; 97(4): E687-93, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22259056

RESUMEN

CONTEXT: The overwhelming majority of benign lesions of the adrenal cortex leading to Cushing syndrome are linked to one or another abnormality of the cAMP or protein kinase pathway. PRKAR1A-inactivating mutations are responsible for primary pigmented nodular adrenocortical disease, whereas somatic GNAS activating mutations cause macronodular disease in the context of McCune-Albright syndrome, ACTH-independent macronodular hyperplasia, and, rarely, cortisol-producing adenomas. OBJECTIVE AND DESIGN: The whole-genome expression profile (WGEP) of normal (pooled) adrenals, PRKAR1A- (3) and GNAS-mutant (3) was studied. Quantitative RT-PCR and Western blot were used to validate WGEP findings. RESULTS: MAPK and p53 signaling pathways were highly overexpressed in all lesions against normal tissue. GNAS-mutant tissues were significantly enriched for extracellular matrix receptor interaction and focal adhesion pathways when compared with PRKAR1A-mutant (fold enrichment 3.5, P < 0.0001 and 2.1, P < 0.002, respectively). NFKB, NFKBIA, and TNFRSF1A were higher in GNAS-mutant tumors (P < 0.05). Genes related to the Wnt signaling pathway (CCND1, CTNNB1, LEF1, LRP5, WISP1, and WNT3) were overexpressed in PRKAR1A-mutant lesions. CONCLUSION: WGEP analysis revealed that not all cAMP activation is the same: adrenal lesions harboring PRKAR1A or GNAS mutations share the downstream activation of certain oncogenic signals (such as MAPK and some cell cycle genes) but differ substantially in their effects on others.


Asunto(s)
Hiperfunción de las Glándulas Suprarrenales/genética , Codón sin Sentido , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , AMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Mutación de Línea Germinal , Sistemas de Mensajero Secundario , Corteza Suprarrenal/metabolismo , Corteza Suprarrenal/patología , Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Insuficiencia Suprarrenal/genética , Insuficiencia Suprarrenal/metabolismo , Insuficiencia Suprarrenal/patología , Hiperfunción de las Glándulas Suprarrenales/metabolismo , Hiperfunción de las Glándulas Suprarrenales/patología , Ciclo Celular , Cromograninas , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Perfilación de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Vía de Señalización Wnt
16.
J Clin Endocrinol Metab ; 97(3): E357-66, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22170724

RESUMEN

BACKGROUND: Mutations in the subunits B, C, and D of succinate dehydrogenase (SDH) mitochondrial complex II have been associated with the development of paragangliomas (PGL), gastrointestinal stromal tumors, papillary thyroid and renal carcinoma (SDHB), and testicular seminoma (SDHD). AIM: Our aim was to examine the possible causative link between SDHD inactivation and somatotropinoma. PATIENTS AND METHODS: A 37-yr-old male presented with acromegaly and hypertension. Other family members were found with PGL. Elevated plasma and urinary levels of catecholamines led to the identification of multiple PGL in the proband in the neck, thorax, and abdomen. Adrenalectomy was performed for bilateral pheochromocytomas (PHEO). A GH-secreting macroadenoma was also found and partially removed via transsphenoidal surgery (TTS). Genetic analysis revealed a novel SDHD mutation (c.298_301delACTC), leading to a frame shift and a premature stop codon at position 133 of the protein. Loss of heterozygosity for the SDHD genetic locus was shown in the GH-secreting adenoma. Down-regulation of SDHD protein in the GH-secreting adenoma by immunoblotting and immunohistochemistry was found. A literature search identified other cases of multiple PGL and/or PHEO in association with pituitary tumors. CONCLUSION: We describe the first kindred with a germline SDHD pathogenic mutation, inherited PGL, and acromegaly due to a GH-producing pituitary adenoma. SDHD loss of heterozygosity, down-regulation of protein in the GH-secreting adenoma, and decreased SDH enzymatic activity supports SDHD's involvement in the pituitary tumor formation in this patient. Older cases of multiple PGL and PHEO and pituitary tumors in the literature support a possible association between SDH defects and pituitary tumorigenesis.


Asunto(s)
Acromegalia/genética , Adenoma/genética , Hormona del Crecimiento/metabolismo , Hipertensión/genética , Neoplasias Hipofisarias/genética , Succinato Deshidrogenasa/genética , Acromegalia/metabolismo , Adenoma/metabolismo , Adulto , Codón sin Sentido , Mutación del Sistema de Lectura , Sitios Genéticos , Mutación de Línea Germinal , Humanos , Hipertensión/metabolismo , Pérdida de Heterocigocidad , Masculino , Neoplasias Hipofisarias/metabolismo , Succinato Deshidrogenasa/metabolismo
17.
J Clin Endocrinol Metab ; 97(2): 351-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22112814

RESUMEN

CONTEXT: Most tumors in Carney complex (CNC) are benign, including primary pigmented nodular adrenocortical disease (PPNAD), the main endocrine tumor in CNC. Adrenocortical cancer (AC) has never been observed in the syndrome. Herein, we describe a large Azorean family with CNC caused by a point mutation in the PRKAR1A gene coding for type 1-α (RIα) regulatory subunit of the cAMP-dependent protein kinase A, in which the index patient presented with AC. OBJECTIVE: We studied the genotype-phenotype correlation in CNC. DESIGN AND SETTING: We reported on case series and in vitro testing of the PRKAR1A mutation in a tertiary care referral center. PATIENTS: Twenty-two members of a family were investigated for Cushing syndrome and other CNC components; their DNA was sequenced for PRKAR1A mutations. RESULTS: Cushing syndrome due to PPNAD occurred in four patients, including the proposita who presented with AC and three who had Cushing syndrome and/or PPNAD. Lentigines were found in six additional patients who did not have PPNAD. A base substitution (c.439A>G/p.S147G) in PRKAR1A was identified in the proposita, in the three others with PPNAD, in the proposita's twin daughters who had lentigines but no evidence of hypercortisolism, and in five other family members, including one without lentigines or evidence of hypercortisolism. Unlike in other RIα defects, loss of heterozygosity was not observed in AC. The S147G mutation was compared to other expressed PRKAR1A mutations; it led to decreased cAMP and catalytic subunit binding by RIα and increased protein kinase A activity in vitro. CONCLUSIONS: In a large family with CNC, one amino acid substitution caused a spectrum of adrenal disease that ranged from lack of manifestations to cancer. PPNAD and AC were the only manifestations of CNC in these patients, in addition to lentigines. These data have implications for counseling patients with CNC and are significant in documenting the first case of AC in the context of PPNAD.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Enfermedades de las Glándulas Suprarrenales/genética , Carcinoma Corticosuprarrenal/genética , Complejo de Carney/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Adolescente , Neoplasias de la Corteza Suprarrenal/complicaciones , Enfermedades de las Glándulas Suprarrenales/complicaciones , Enfermedades de las Glándulas Suprarrenales/diagnóstico , Carcinoma Corticosuprarrenal/complicaciones , Adulto , Sustitución de Aminoácidos/fisiología , Complejo de Carney/complicaciones , Niño , Preescolar , Familia , Femenino , Glicina/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense/fisiología , Serina/genética , Adulto Joven
18.
Psychoneuroendocrinology ; 37(6): 836-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22024111

RESUMEN

The role of cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signaling in the molecular pathways involved in fear and memory is well established. Prior studies in our lab reported that transgenic mice with an inactivating mutation in Prkar1a gene (codes for the 1-alpha regulatory subunit (R1α) of PKA) exhibited behavioral abnormalities including anxiety and depression. In the present study, we examined the role of altered PKA signaling on anxiety-like behaviors in Prkar1a(+/-) mice compared to wild-type (WT) littermates. The elevated plus maze (EPM) and marble bury (MB) tests were used to assess anxiety-like behavior. The hotplate test was performed to evaluate analgesia. We further examined the impact of the Prkar1a inactivating mutation on PKA activity in specific nuclei of the brain associated with anxiety-like behavior. Results for the MB test showed a genotype effect, with increased anxiety-like behavior in Prkar1a(+/-) mice, compared to WT littermates (p<0.05). MANOVA analysis showed a significant genotype difference in anxiety-like behavior in the EPM between WT and Prkar1a(+/-) mice on combined dependent variables (open arm time and open to total time ratio; p<0.05). Results of hotplate testing showed no genotype effect however; the expected sex difference was noted. Analysis of PKA activity showed the loss of one Prkar1a allele led to an increase in basal and cAMP-stimulated kinase activity in both the basolateral and central amygdala. These results suggest that the alteration in PKA signaling in Prkar1a(+/-) mice is not a ubiquitous effect; and supports the importance of cAMP/PKA pathway in neurobiological processes involved in anxiety and fear sensitization.


Asunto(s)
Ansiedad/genética , Ansiedad/psicología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/fisiología , Afecto/fisiología , Animales , Ansiolíticos/farmacología , Conducta Animal/fisiología , Química Encefálica/genética , Química Encefálica/fisiología , ADN/genética , Emociones/fisiología , Genotipo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Dimensión del Dolor , Fenotipo , Reacción en Cadena de la Polimerasa , Estrés Psicológico/genética , Estrés Psicológico/psicología
19.
J Clin Endocrinol Metab ; 97(3): E496-502, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22205709

RESUMEN

BACKGROUND: Carney complex (CNC) is a multiple endocrine neoplasia syndrome due to inactivating mutations in the PRKAR1A gene that codes for type Iα regulatory (RIα) subunit of protein kinase A. Most PRKAR1A mutations are subject to nonsense mRNA decay (NMD) and, thus, lead to haploinsufficiency. METHODS AND SETTING: Patient phenotyping for CNC features and DNA, RNA, protein, and transfection studies were carried out at a research center. RESULTS: We describe in unrelated kindreds with CNC four naturally occurring PRKAR1A mutations (1055del4, 1067del4ins5, 1076delTTins13, and 1142del4) that are predicted to escape NMD because they are located in the last coding exon of the gene. The phenotype of CNC was not different from that in other patients with the condition, although the number of patients was small. Each of the mutations caused a frameshift that led to a new stop codon into the 3' untranslated open reading frame, predicting an elongated protein that, however, was absent in patient-derived cells. After site-directed mutagenesis, in vitro transcription, and cell-free translation experiments, the expected size mutant proteins were present. However, when the mutant constructs were transfected in adrenal (NCI-295), testicular (N-TERA), and embryonic (HEK293) cells and despite the presence of the mutant mRNA, Western blot analysis indicated that there were no longer proteins. The subsequent application of proteasome inhibitors to cells transfected with the mutant constructs led to the detection of the aberrant proteins, although a compound that affects protein folding had no effect. The wild-type protein was also decreased in both patient-derived cells and/or tissues as well as in the in vitro systems used in this study. CONCLUSIONS: This was the first demonstration of proteasomal degradation of RIα protein variants leading to PRKAR1A haploinsufficiency and CNC, adding protein surveillance to NMD in the cellular mechanisms overseeing RIα synthesis. In agreement with the molecular data, CNC patients bearing PRKAR1A defects that extend the open reading frame did not have a different phenotype, although this has to be confirmed in a larger number of patients.


Asunto(s)
Complejo de Carney/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Complejo de la Endopetidasa Proteasomal/genética , Adolescente , Adulto , Complejo de Carney/metabolismo , Niño , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Femenino , Genotipo , Células HEK293 , Haploinsuficiencia , Humanos , Masculino , Persona de Mediana Edad , Neoplasia Endocrina Múltiple/genética , Neoplasia Endocrina Múltiple/metabolismo , Mutación , Sistemas de Lectura Abierta , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo
20.
Am J Physiol Endocrinol Metab ; 301(2): E370-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21586701

RESUMEN

Several receptors linked to the adenylyl cyclase signaling pathway stimulate electrical activity and calcium influx in endocrine pituitary cells, and a role for an unidentified sodium-conducting channel in this process has been proposed. Here we show that forskolin dose-dependently increases cAMP production and facilitates calcium influx in about 30% of rat and mouse pituitary cells at its maximal concentration. The stimulatory effect of forskolin on calcium influx was lost in cells with inhibited PKA (cAMP-dependent protein kinase) and in cells that were haploinsufficient for the main PKA regulatory subunit but was preserved in cells that were also haploinsufficient for the main PKA catalytic subunit. Spontaneous and forskolin-stimulated calcium influx was present in cells with inhibited voltage-gated sodium and hyperpolarization-activated cation channels but not in cells bathed in medium, in which sodium was replaced with organic cations. Consistent with the role of sodium-conducting nonselective cation channels in PKA-stimulated Ca(2+) influx, cAMP induced a slowly developing current with a reversal potential of about 0 mV. Two TRP (transient receptor potential) channel blockers, SKF96365 and 2-APB, as well as flufenamic acid, an inhibitor of nonselective cation channels, also inhibited spontaneous and forskolin-stimulated electrical activity and calcium influx. Quantitative RT-PCR analysis indicated the expression of mRNA transcripts for TRPC1 >> TRPC6 > TRPC4 > TRPC5 > TRPC3 in rat pituitary cells. These experiments suggest that in pituitary cells constitutively active cation channels are stimulated further by PKA and contribute to calcium signaling indirectly by controlling the pacemaking depolarization in a sodium-dependent manner and directly by conducting calcium.


Asunto(s)
Señalización del Calcio/fisiología , Adenohipófisis/fisiología , Proteínas Quinasas/metabolismo , Canales Catiónicos TRPV/fisiología , Adenosina Monofosfato/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Cationes/metabolismo , Células Cultivadas , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Gonadotrofos/fisiología , Lactotrofos/fisiología , Membranas/fisiología , Nucleótidos Cíclicos/metabolismo , Adenohipófisis/citología , Ratas , Ratas Sprague-Dawley , Sodio/metabolismo , Somatotrofos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA