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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Hum Mol Genet ; 20(1): 165-75, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20940146

RESUMEN

Patients with genetic defects of the cyclic (c) adenosine-monophosphate (AMP)-signaling pathway and those with neonatal-onset multisystem inflammatory disease (NOMID) develop tumor-like lesions of the long bones. The molecular basis of this similarity is unknown. NOMID is caused by inappropriate caspase-1 activity, which in turn activates the inflammasome. The present study demonstrates that NOMID bone lesions are derived from the same osteoblast progenitor cells that form fibroblastoid tumors in mice and humans with defects that lead to increased cAMP-dependent protein kinase A (PKA) signaling. NOMID tumor cells showed high PKA activity, and an increase in their cAMP signaling led to PKA-specific activation of caspase-1. Increased PKA led to inflammation-independent activation of caspase-1 via over-expression of the proto-oncogene (and early osteoblast factor) Ets-1. In NOMID tumor cells, as in cells with defective PKA regulation, increased prostaglandin E2 (PGE2) led to increased cAMP levels and activation of Wnt signaling, like in other states of inappropriate PKA activity. Caspase-1 and PGE2 inhibition led to a decrease in cell proliferation of both NOMID and cells with abnormal PKA. These data reveal a previously unsuspected link between abnormal cAMP signaling and defective regulation of the inflammasome and suggest that caspase-1 and PGE2 inhibition may be therapeutic targets in bone lesions associated with defects of these two pathways.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Osteoblastos/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Células Madre/metabolismo , Animales , Huesos/patología , Caspasa 1/genética , Células Cultivadas , Síndromes Periódicos Asociados a Criopirina/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Dinoprostona/genética , Dinoprostona/metabolismo , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Ratones , Proto-Oncogenes Mas , Proteína Proto-Oncogénica c-ets-1/genética , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Células del Estroma/metabolismo , Activación Transcripcional/genética , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
3.
Proc Natl Acad Sci U S A ; 107(19): 8683-8, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20421483

RESUMEN

A population of stromal cells that retains osteogenic capacity in adult bone (adult bone stromal cells or aBSCs) exists and is under intense investigation. Mice heterozygous for a null allele of prkar1a (Prkar1a(+/-)), the primary receptor for cyclic adenosine monophosphate (cAMP) and regulator of protein kinase A (PKA) activity, developed bone lesions that were derived from cAMP-responsive osteogenic cells and resembled fibrous dysplasia (FD). Prkar1a(+/-) mice were crossed with mice that were heterozygous for catalytic subunit Calpha (Prkaca(+/-)), the main PKA activity-mediating molecule, to generate a mouse model with double heterozygosity for prkar1a and prkaca (Prkar1a(+/-)Prkaca(+/-)). Unexpectedly, Prkar1a(+/-)Prkaca(+/-) mice developed a greater number of osseous lesions starting at 3 months of age that varied from the rare chondromas in the long bones and the ubiquitous osteochondrodysplasia of vertebral bodies to the occasional sarcoma in older animals. Cells from these lesions originated from an area proximal to the growth plate, expressed osteogenic cell markers, and showed higher PKA activity that was mostly type II (PKA-II) mediated by an alternate pattern of catalytic subunit expression. Gene expression profiling confirmed a preosteoblastic nature for these cells but also showed a signature that was indicative of mesenchymal-to-epithelial transition and increased Wnt signaling. These studies show that a specific subpopulation of aBSCs can be stimulated in adult bone by alternate PKA and catalytic subunit activity; abnormal proliferation of these cells leads to skeletal lesions that have similarities to human FD and bone tumors.


Asunto(s)
Envejecimiento/patología , Huesos/enzimología , Huesos/patología , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Animales , Huesos/diagnóstico por imagen , Calcificación Fisiológica , Dominio Catalítico , Heterocigoto , Mesodermo/metabolismo , Ratones , Hidrolasas Diéster Fosfóricas/metabolismo , Espectrometría Raman , Células del Estroma/enzimología , Células del Estroma/patología , Tomografía Computarizada por Rayos X
4.
Hum Mol Genet ; 19(8): 1387-98, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20080939

RESUMEN

PRKAR1A inactivation leads to dysregulated cAMP signaling and Carney complex (CNC) in humans, a syndrome associated with skin, endocrine and other tumors. The CNC phenotype is not easily explained by the ubiquitous cAMP signaling defect; furthermore, Prkar1a(+/-) mice did not develop skin and other CNC tumors. To identify whether a Prkar1a defect is truly a generic but weak tumorigenic signal that depends on tissue-specific or other factors, we investigated Prkar1a(+/-) mice when bred within the Rb1(+/-) or Trp53(+/-) backgrounds, or treated with a two-step skin carcinogenesis protocol. Prkar1a(+/-) Trp53(+/-) mice developed more sarcomas than Trp53(+/-) mice (P < 0.05) and Prkar1a(+/-) Rb1(+/-) mice grew more (and larger) pituitary and thyroid tumors than Rb1(+/-) mice. All mice with double heterozygosity had significantly reduced life-spans compared with their single-heterozygous counterparts. Prkar1a(+/-) mice also developed more papillomas than wild-type animals. A whole-genome transcriptome profiling of tumors produced by all three models identified Wnt signaling as the main pathway activated by abnormal cAMP signaling, along with cell cycle abnormalities; all changes were confirmed by qRT-PCR array and immunohistochemistry. siRNA down-regulation of Ctnnb1, E2f1 or Cdk4 inhibited proliferation of human adrenal cells bearing a PRKAR1A-inactivating mutation and Prkar1a(+/-) mouse embryonic fibroblasts and arrested both cell lines at the G0/G1 phase of the cell cycle. In conclusion, Prkar1a haploinsufficiency is a relatively weak tumorigenic signal that can act synergistically with other tumor suppressor gene defects or chemicals to induce tumors, mostly through Wnt-signaling activation and cell cycle dysregulation, consistent with studies in human neoplasms carrying PRKAR1A defects.


Asunto(s)
Ciclo Celular , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Neoplasias/genética , Neoplasias/patología , Proteína de Retinoblastoma/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteínas Wnt/metabolismo , Animales , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Haploidia , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/metabolismo , Neoplasias/fisiopatología , Procesos Neoplásicos , Papiloma/inducido químicamente , Papiloma/genética , Papiloma/metabolismo , Papiloma/fisiopatología , Proteína de Retinoblastoma/metabolismo , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/fisiopatología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Wnt/genética
5.
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
6.
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
7.
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
8.
Am J Med Genet A ; 152A(3): 630-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20186806

RESUMEN

A female patient with a partial trisomy 16q was described previously. Her clinical characteristics included obesity, severe anisomastia, moderate to severe mental retardation, attention deficit hyperactivity disorder, dysmorphic facies, and contractions of the small joints. In this article, we describe a more detailed analysis of the genetic anomaly in this patient. We were particularly interested in the involvement of the fat mass and obesity associated gene (FTO) in her duplication. Single nucleotide polymorphisms in FTO have been associated with obesity. The breakpoints of the duplication were precisely mapped using high-resolution oligonucleotide array comparative genomic hybridization (CGH). We found that the duplication spans 11.45 Mb on 16q11.2 to 16q13 and it includes FTO. The increased copy number of FTO was confirmed with a qPCR on genomic DNA of the patient. We investigated the influence of the increased FTO copy number on FTO gene expression in immortalized lymphocytes from the patient using qPCR. No evidence of increased FTO expression was detected in the patient's lymphocytes. We discuss these findings and we review clinical findings in patients with overlapping 16q duplications to determine the relationship between increased FTO copy number and obesity. Our review suggests that duplication of the FTO gene does not necessarily result in obesity.


Asunto(s)
Cromosomas Humanos Par 16/genética , Obesidad/genética , Proteínas/genética , Trisomía , Adulto , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Secuencia de Bases , Mama/anomalías , Mapeo Cromosómico , Hibridación Genómica Comparativa , Cartilla de ADN/genética , Femenino , Dosificación de Gen , Expresión Génica , Humanos , Linfocitos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Reacción en Cadena de la Polimerasa
9.
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.

10.
Clin Cancer Res ; 14(2): 388-95, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18223213

RESUMEN

PURPOSE: Since the identification of PRKAR1A mutations in Carney complex, substitutions and small insertions/deletions have been found in approximately 70% of the patients. To date, no germ-line PRKAR1A deletion and/or insertion exceeded a few base pairs (up to 15). Although a few families map to chromosome 2, it is possible that current sequencing techniques do not detect larger gene changes in PRKAR1A -- mutation-negative individuals with Carney complex. EXPERIMENTAL DESIGN: To screen for gross alterations of the PRKAR1A gene, we applied Southern hybridization analysis on 36 unrelated Carney complex patients who did not have small intragenic mutations or large aberrations in PRKAR1A, including the probands from two kindreds mapping to chromosome 2. RESULTS: We found large PRKAR1A deletions in the germ-line of two patients with Carney complex, both sporadic cases; no changes were identified in the remaining patients, including the two chromosome-2-mapping families. In the first patient, the deletion is expected to lead to decreased PRKAR1A mRNA levels but no other effects on the protein; the molecular phenotype is predicted to be PRKAR1A haploinsufficiency, consistent with the majority of PRKAR1A mutations causing Carney complex. In the second patient, the deletion led to in-frame elimination of exon 3 and the expression of a shorter protein, lacking the primary site for interaction with the catalytic protein kinase A subunit. In vitro transfection studies of the mutant PRKAR1A showed impaired ability to bind cyclic AMP and activation of the protein kinase A enzyme. The patient bearing this mutation had a more-severe-than-average Carney complex phenotype that included the relatively rare psammomatous melanotic schwannoma. CONCLUSIONS: Large PRKAR1A deletions may be responsible for Carney complex in patients that do not have PRKAR1A gene defects identifiable by sequencing. Preliminary data indicate that these patients may have a different phenotype especially if their defect results in an expressed, abnormal version of the PRKAR1A protein.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Eliminación de Gen , Neoplasia Endocrina Múltiple/genética , Línea Celular , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Exones , Humanos
11.
Hum Mutat ; 29(5): 633-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18241045

RESUMEN

Patients presenting with primary pigmented nodular adrenocortical disease (PPNAD), Carney complex (CNC), or sporadic tumors were previously found to carry germline mutations in the human type Ialpha regulatory subunit (RIalpha) of adenosine 3',5'-cyclic monophosphate (cyclic AMP [cAMP])-dependent protein kinase (PKA; PRKAR1A). Although about 90% of disease-causing PRKAR1A mutations lead to premature stop codon generation and subsequent degradation of the mutant message by nonsense-mediated mRNA decay (NMD), here we describe seven PRKAR1A mutations whose mRNAs do not seem to undergo NMD and instead result in an expressed mutant RIalpha protein. The expressed mutations (p.Ser9Asn, p.Glu60_Lys116del [Delta-exon 3], p.Arg74Cys, p.Arg146Ser, p.Asp183Tyr, p.Ala213Asp, and p.Gly289Trp) were spread over all the functional RIalpha domains, and all of them exhibited increased PKA activity, which we attribute to decreased binding to cAMP and/or the catalytic subunit. Our data further corroborate the previous finding that altered PRKAR1A function, not only haploinsufficiency, is enough to elevate PKA activity which is apparently associated with tumorigenesis in tissues affected by CNC. In some cases, as with the Delta-exon 3 mutation, we may even conclude that the presence of a mutant PRKAR1A protein may be more harmful than allelic loss.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Mutación de Línea Germinal , ARN Mensajero/genética , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Reacción en Cadena de la Polimerasa
12.
J Clin Endocrinol Metab ; 93(2): 565-71, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18056771

RESUMEN

CONTEXT: Inactivating mutations of PRKAR1A, the regulatory subunit type 1A (RIalpha) of protein kinase A (PKA), are associated with tumor formation. OBJECTIVE: Our objective was to evaluate the role of PKA isozymes on proliferation and cell cycle. METHODS: A cell line with RIalpha haploinsufficiency due to an inactivating PRKAR1A mutation (IVS2+1 G-->A) was transfected with constructs encoding PKA subunits. Genetics, PKA subunit mRNA and protein expression and proliferation, aneuploidy, and cell cycle status were assessed. To identify factors that mediate PKA-associated cell cycle changes, we studied E2F and cyclins expression in transfected cells and E2F's role by small interfering RNA; we also assessed cAMP levels and baseline and stimulated cAMP signaling in transfected cells. RESULTS: Introduction of PKA subunits led to changes in proliferation and cell cycle: a decrease in aneuploidy and G(2)/M for the PRKAR1A-transfected cells and an increase in S phase and aneuploidy for cells transfected with PRKAR2B, a known PRKAR1A mutant (RIalphaP), and the PKA catalytic subunit. There were alterations in cAMP levels, PKA subunit expression, cyclins, and E2F factors; E2F1 was shown to possibly mediate PKA effects on cell cycle by small interfering RNA studies. cAMP levels and constitutive and stimulated cAMP signaling were altered in transfected cells. CONCLUSION: This is the first immortalized cell line with a naturally occurring PRKAR1A-inactivating mutation that is associated in vivo with tumor formation. PKA isozyme balance is critical for the control of cAMP signaling and related cell cycle and proliferation changes. Finally, E2F1 may be a factor that mediates dysregulated PKA's effects on the cell cycle.


Asunto(s)
Enfermedades de la Corteza Suprarrenal/enzimología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Mutación Puntual , Enfermedades de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/enzimología , Neoplasias de la Corteza Suprarrenal/genética , Apoptosis/fisiología , Western Blotting , Ciclo Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Línea Celular , AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/biosíntesis , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Ciclinas/metabolismo , ADN/química , ADN/genética , Factores de Transcripción E2F/metabolismo , Femenino , Humanos , Isoenzimas , Ploidias , Análisis de Secuencia de ADN , Transfección
13.
J Clin Endocrinol Metab ; 93(3): 1020-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18073299

RESUMEN

PURPOSE: Protein kinase A (PKA) affects cell proliferation in many cell types and is a potential target for cancer treatment. PKA activity is stimulated by cAMP and cAMP analogs. One such substance, 8-Cl-cAMP, and its metabolite 8-Cl-adenosine (8-Cl-ADO) are known inhibitors of cancer cell proliferation; however, their mechanism of action is controversial. We have investigated the antiproliferative effects of 8-Cl-cAMP and 8-CL-ADO on human thyroid cancer cells and determined PKA's involvement. EXPERIMENTAL DESIGN: We employed proliferation and apoptosis assays and PKA activity and cell cycle analysis to understand the effect of 8-Cl-ADO and 8-Cl-cAMP on human thyroid cancer and HeLa cell lines. RESULTS: 8-Cl-ADO inhibited proliferation of all cells, an effect that lasted for at least 4 d. Proliferation was also inhibited by 8-Cl-cAMP, but this inhibition was reduced by 3-isobutyl-1-methylxanthine; both drugs stimulated apoptosis, and 3-isobutyl-1-methylxanthine drastically reduced 8-Cl-cAMP-induced cell death. 8-Cl-ADO induced cell accumulation in G1/S or G2/M cell cycle phases and differentially altered PKA activity and subunit levels. PKA stimulation or inhibition and adenosine receptor agonists or antagonists did not significantly affect proliferation. CONCLUSIONS: 8-Cl-ADO and 8-Cl-cAMP inhibit proliferation, induce cell cycle phase accumulation, and stimulate apoptosis in thyroid cancer cells. The effect of 8-Cl-cAMP is likely due to its metabolite 8-Cl-ADO, and PKA does not appear to have direct involvement in the inhibition of proliferation by 8-Cl-ADO. 8-Cl-ADO may be a useful therapeutic agent to be explored in aggressive thyroid cancer.


Asunto(s)
2-Cloroadenosina/análogos & derivados , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Neoplasias de la Tiroides/tratamiento farmacológico , 1-Metil-3-Isobutilxantina/farmacología , 2-Cloroadenosina/metabolismo , 2-Cloroadenosina/farmacología , 8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Transducción de Señal , Neoplasias de la Tiroides/patología
14.
Eur J Hum Genet ; 16(10): 1245-53, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18431404

RESUMEN

Bilateral adrenocortical hyperplasia (BAH) is the second most common cause of corticotropin-independent Cushing syndrome (CS). Genetic forms of BAH have been associated with complex syndromes such as Carney Complex and McCune-Albright syndrome or may present as isolated micronodular adrenocortical disease (iMAD) usually in children and young adults with CS. A genome-wide association study identified inactivating phosphodiesterase (PDE) 11A (PDE11A)-sequencing defects as low-penetrance predisposing factors for iMAD and related abnormalities; we also described a mutation (c.914A > C/H305P) in cyclic AMP (cAMP)-specific PDE8B, in a patient with iMAD. In this study we further characterize this mutation; we also found a novel PDE8B isoform that is highly expressed in the adrenal gland. This mutation is shown to significantly affect the ability of the protein to degrade cAMP in vitro. Tumor tissues from patients with iMAD and no mutations in the coding PDE8B sequence or any other related genes (PRKAR1A, PDE11A) showed downregulated PDE8B expression (compared to normal adrenal cortex). Pde8b is detectable in the adrenal gland of newborn mice and is widely expressed in other mouse tissues. We conclude that PDE8B is another PDE gene linked to iMAD; it is a candidate causative gene for other adrenocortical lesions linked to the cAMP signaling pathway and possibly for tumors in other tissues.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/genética , Corteza Suprarrenal/enzimología , Hiperplasia Suprarrenal Congénita/enzimología , Hiperplasia Suprarrenal Congénita/genética , Mutación/genética , 3',5'-AMP Cíclico Fosfodiesterasas/química , Adolescente , Enfermedades de la Corteza Suprarrenal/enzimología , Enfermedades de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/enzimología , Neoplasias de la Corteza Suprarrenal/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Niño , Preescolar , Secuencia Conservada , Síndrome de Cushing/enzimología , Síndrome de Cushing/genética , Femenino , Histidina/genética , Humanos , Lactante , Isoenzimas/química , Isoenzimas/genética , Masculino , Ratones , Datos de Secuencia Molecular , Linaje , Prolina/genética
15.
Cancer Res ; 66(18): 8971-4, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16982736

RESUMEN

In cancer cells, cyclic AMP-dependent protein kinase (PKA) is secreted into the conditioned medium. This PKA, designated as extracellular protein kinase A (ECPKA), is markedly up-regulated in the sera of patients with cancer. The currently available tumor markers are based on the antigen determination method and lack specificity and sensitivity. Here, we present an ECPKA autoantibody detection method for a universal biomarker that detects cancer of various cell types. We tested sera from 295 patients with cancers of various cell types, 155 normal controls, and 55 patients without cancer. The specificity and sensitivity of this autoantibody enzyme immunoassay method were compared with the conventional antigen determination method by receiver-operating characteristic plots. In the sera, the presence of autoantibody directed against ECPKA was highly correlated with cancer. High anti-ECPKA autoantibody titers (frequency, 90%; mean titer, 3.0) were found in the sera of patients with various cancers, whereas low or negative titers (frequency, 12%; mean titer, 1.0) were found in the control group. The receiver-operating characteristic plot showed that autoantibody enzyme immunoassay exhibited 90% sensitivity and 88% specificity, whereas the enzymatic assay exhibited 83% sensitivity and 80% specificity. These results show that the autoantibody method distinguished between patients with cancer and controls better than the antigen method could. Our results show that autoantibody ECPKA is a universal serum biomarker for cancers of various cell types.


Asunto(s)
Autoanticuerpos/sangre , Proteínas Quinasas Dependientes de AMP Cíclico/inmunología , Técnicas para Inmunoenzimas/métodos , Neoplasias/enzimología , Neoplasias/inmunología , Anticuerpos Antiidiotipos/sangre , Humanos , Neoplasias/sangre
16.
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
17.
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
18.
Clin Cancer Res ; 11(16): 5950-5, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16115938

RESUMEN

PURPOSE: CpG DNAs induce cytokines, activate natural killer cells, and elicit vigorous T-cell response leading to antitumor effects. Antisense oligodeoxynucleotides targeted against the RIalpha subunit of protein kinase A (antisense PKA RIalpha) induce growth arrest, apoptosis, and differentiation in a variety of cancer cell lines in vitro and in vivo. This study investigated the use of a combinatorial therapy consisting of the RNA-DNA second-generation antisense PKA RIalpha and the CpG immunomer (CpG DNA linked through 3'-3' linkage containing two accessible 5' ends). EXPERIMENTAL DESIGN: HCT-15 multidrug-resistant colon carcinoma growth in nude mice was used as an experimental model. The inhibitory effect on tumor growth and apoptotic activity of antisense RIalpha and CpG immunomer, singly and in combination, were measured by tumor growth, levels of RIalpha subunit, and antiapoptotic and proapoptotic proteins. Effect on host-immune system was measured by mouse spleen size, interleukin-6 (IL-6) levels in mouse blood, and nuclear factor-kappaB (NF-kappaB) transcription activity in mouse spleen cells. RESULTS: In combination, CpG immunomer and antisense PKA RIalpha induced additive/supra-additive effect on the inhibition of tumor growth. Antisense RIalpha but not CpG immunomer increased Bax and Bak proapoptotic protein levels and decreased Bcl-2 and RIalpha protein levels in tumor cells. CpG immunomer but not antisense RIalpha induced an enlargement of mouse spleen, increased IL-6 levels in mouse blood, and increased NF-kappaB transcription activity in mouse spleen cells. CONCLUSIONS: These results show that type I PKA down-regulation and induction of apoptosis in tumor cells by antisense PKA RIalpha, and host-immune stimulation by CpG immunomer are responsible at the molecular level for the supra-additive effects of tumor growth inhibition. Thus, antisense PKA RIalpha and CpG immunomer in combination work cooperatively and as tumor-targeted therapeutics to treat human cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Islas de CpG/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Animales , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Islas de CpG/inmunología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Humanos , Interleucina-6/metabolismo , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Oligonucleótidos/administración & dosificación , Oligonucleótidos/genética , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA