Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Am J Surg Pathol ; 46(2): 226-232, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34889853

RESUMEN

Superficial angiomyxomas (SAMs) are benign cutaneous tumors that arise de novo and in the setting of the Carney complex (CC), an autosomal dominant disease with several cutaneous manifestations including lentigines and pigmented epithelioid melanocytomas. Although most SAM do not pose a diagnostic challenge, a subset can demonstrate histopathologic overlap with other myxoid tumors that arise in the skin and subcutis. Traditional immunohistochemical markers are of limited utility when discriminating SAM from histopathologic mimics. Since protein kinase A regulatory subunit 1 alpha (PRKAR1A) genetic alterations underlie most CC cases, we investigated whether SAM demonstrate loss of PRKAR1A protein expression by immunohistochemistry. In our series, 29 SAM, 26 myxofibrosarcoma, 5 myxoid dermatofibrosarcoma protuberans, 11 superficial acral fibromyxomas, and 18 digital mucous cysts were characterized. Of the 29 SAM examined in this study, 1 was associated with documented CC in a 5-year-old girl. SAM tended to arise in adults (mean 49.7 y; range: 5 to 87 y). Loss of PRKAR1A was seen in 55.2% of cases (16/29) and had a male predilection (87.5%, 12/16). PRKAR1A-inactivated SAM demonstrated significant nuclear enlargement (100%, 16/16 vs. 23.1%, 3/13), multinucleation (81.3%, 13/16 vs. 23.1%, 3/13), and presence of neutrophils (43.8%, 7/16 vs. 0%, 0/13). In contrast, PRKAR1A was retained in all cases of myxofibrosarcoma (100%, 26/26), myxoid dermatofibrosarcoma protuberans (100%, 5/5), superficial acral fibromyxomas (100%, 11/11), and digital mucous cyst (100%, 18/18). Taken together, PRKAR1A loss by immunohistochemistry can be used as an adjunctive assay to support the diagnosis of SAM given the high specificity of this staining pattern compared with histopathologic mimics.


Asunto(s)
Biomarcadores de Tumor/deficiencia , Complejo de Carney/enzimología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Quistes/enzimología , Dermatofibrosarcoma/enzimología , Fibroma/enzimología , Inmunohistoquímica , Mixoma/enzimología , Neoplasias Cutáneas/enzimología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Complejo de Carney/patología , Niño , Preescolar , Quistes/patología , Dermatofibrosarcoma/patología , Femenino , Fibroma/patología , Humanos , Masculino , Persona de Mediana Edad , Mixoma/patología , Valor Predictivo de las Pruebas , Neoplasias Cutáneas/patología
2.
Physiol Rep ; 8(6): e14405, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32212257

RESUMEN

Protein kinase A (PKA) activity is pivotal for proper functioning of the human heart, and its dysregulation has been implicated in a variety of cardiac pathologies. PKA regulatory subunit 1α (R1α, encoded by the PRKAR1A gene) is highly expressed in the heart, and controls PKA kinase activity by sequestering PKA catalytic subunits. Patients with PRKAR1A mutations are often diagnosed with Carney complex (CNC) in early adulthood, and may die later in life from cardiac complications such as heart failure. However, it remains unknown whether PRKAR1A deficiency interferes with normal heart development. Here, we showed that left ventricular mass was reduced in young CNC patients with PRKAR1A mutations or deletions. Cardiac-specific heterozygous ablation of PRKAR1A in mice increased cardiac PKA activity, and reduced heart weight and cardiomyocyte size without altering contractile function at 3 months of age. Silencing of PRKAR1A, or stimulation with the PKA activator forskolin completely abolished α1-adrenergic receptor-mediated cardiomyocyte hypertrophy. Mechanistically, depletion of PRKAR1A provoked PKA-dependent inactivating phosphorylation of Drp1 at S637, leading to impaired mitochondrial fission. Pharmacologic inhibition of Drp1 with Mdivi 1 diminished hypertrophic growth of cardiomyocytes. In conclusion, PRKAR1A deficiency suppresses cardiomyocyte hypertrophy and impedes heart growth, likely through inhibiting Drp1-mediated mitochondrial fission. These findings provide a potential novel mechanism for the cardiac manifestations associated with CNC.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/fisiología , Ventrículos Cardíacos/patología , Miocitos Cardíacos/patología , Adolescente , Adulto , Animales , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Femenino , Humanos , Hipertrofia , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Tamaño de los Órganos , Adulto Joven
3.
Am J Physiol Renal Physiol ; 313(3): F677-F686, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28615245

RESUMEN

The failure of the polycystins (PCs) to function in primary cilia is thought to be responsible for autosomal dominant polycystic kidney disease (ADPKD). Primary cilia integrate multiple cellular signaling pathways, including calcium, cAMP, Wnt, and Hedgehog, which control cell proliferation and differentiation. It has been proposed that mutated PCs result in reduced intracellular calcium, which in turn upregulates cAMP, protein kinase A (PKA) signaling, and subsequently other proliferative signaling pathways. However, the role of PKA in ADPKD has not been directly ascertained in vivo, although the expression of the main regulatory subunit of PKA in cilia and other compartments (PKA-RIα, encoded by PRKAR1A) is increased in a mouse model orthologous to ADPKD. Therefore, we generated a kidney-specific knockout of Prkar1a to examine the consequences of constitutive upregulation of PKA on wild-type and Pkd1 hypomorphic (Pkd1RC) backgrounds. Kidney-specific loss of Prkar1a induced renal cystic disease and markedly aggravated cystogenesis in the Pkd1RC models. In both settings, it was accompanied by upregulation of Src, Ras, MAPK/ERK, mTOR, CREB, STAT3, Pax2 and Wnt signaling. On the other hand, Gli3 repressor activity was enhanced, possibly contributing to hydronephrosis and impaired glomerulogenesis in some animals. To assess the relevance of these observations in humans we looked for and found evidence for kidney and liver cystic phenotypes in the Carney complex, a tumoral syndrome caused by mutations in PRKAR1A These observations expand our understanding of the pathogenesis of ADPKD and demonstrate the importance of PRKAR1A highlighting PKA as a therapeutic target in ADPKD.


Asunto(s)
Complejo de Carney/enzimología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Quistes/enzimología , Riñón/enzimología , Hepatopatías/enzimología , Riñón Poliquístico Autosómico Dominante/enzimología , Adolescente , Adulto , Animales , Complejo de Carney/diagnóstico por imagen , Complejo de Carney/genética , Proliferación Celular , Niño , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Quistes/diagnóstico por imagen , Quistes/genética , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Humanos , Riñón/patología , Riñón/fisiopatología , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Hepatopatías/diagnóstico por imagen , Hepatopatías/genética , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción PAX2/metabolismo , Fenotipo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Dominante/fisiopatología , Factor de Transcripción STAT3/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Canales Catiónicos TRPP/genética , Vía de Señalización Wnt , Adulto Joven , Proteína Gli3 con Dedos de Zinc , Proteínas ras/metabolismo , Familia-src Quinasas/metabolismo
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 Exp Clin Cancer Res ; 34: 143, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26608815

RESUMEN

BACKGROUND: Protein kinase A (PKA) is a holoenzyme that consists of a dimer of regulatory subunits and two inactive catalytic subunits that bind to the regulatory subunit dimer. Four regulatory subunits (RIα, RIß, RIIα, RIIß) and four catalytic subunits (Cα, Cß, Cγ, Prkx) have been described in the human and mouse genomes. Previous studies showed that complete inactivation of the Prkar1a subunit (coding for RIα) in the germline leads to embryonic lethality, while Prkar1a-deficient mice are viable and develop schwannomas, thyroid, and bone neoplasms, and rarely lymphomas and sarcomas. Mice with inactivation of the Prkar2a and Prkar2b genes (coding for RIIα and RIIß, respectively) are also viable but have not been studied for their susceptibility to any tumors. METHODS: Cohorts of Prkar1a (+/-) , Prkar2a (+/-) , Prkar2a (-/-) , Prkar2b (+/-) and wild type (WT) mice have been observed between 5 and 25 months of age for the development of hematologic malignancies. Tissues were studied by immunohistochemistry; tumor-specific markers were also used as indicated. Cell sorting and protein studies were also performed. RESULTS: Both Prkar2a (-/-) and Prkar2a (+/-) mice frequently developed hematopoietic neoplasms dominated by histiocytic sarcomas (HS) with rare diffuse large B cell lymphomas (DLBCL). Southern blot analysis confirmed that the tumors diagnosed histologically as DLBCL were clonal B cell neoplasms. Mice with other genotypes did not develop a significant number of similar neoplasms. CONCLUSIONS: Prkar2a deficiency predisposes to hematopoietic malignancies in vivo. RIIα's likely association with HS and DLBCL was hitherto unrecognized and may lead to better understanding of these rare neoplasms.


Asunto(s)
Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Neoplasias Hematológicas/genética , Animales , Transformación Celular Neoplásica/genética , Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Neoplasias Hematológicas/patología , Inmunofenotipificación , Ratones , Ratones Noqueados , Fenotipo , Factores de Tiempo
6.
Endocr Relat Cancer ; 19(3): 435-46, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22514108

RESUMEN

Thyroid cancer is the most common endocrine malignancy in the population, and the incidence of this cancer is increasing at a rapid rate. Although genetic analysis of papillary thyroid cancer (PTC) has identified mutations in a large percentage of patients, the genetic basis of follicular thyroid cancer (FTC) is less certain. Thyroid cancer, including both PTC and FTC, has been observed in patients with the inherited tumor predisposition Carney complex, caused by mutations in PRKAR1A. In order to investigate the role of loss of PRKAR1A in thyroid cancer, we generated a tissue-specific knockout of Prkar1a in the thyroid. We report that the resulting mice are hyperthyroid and developed follicular thyroid neoplasms by 1 year of age, including FTC in over 40% of animals. These thyroid tumors showed a signature of pathway activation different from that observed in other models of thyroid cancer. In vitro cultures of the tumor cells indicated that Prkar1a-null thyrocytes exhibited growth factor independence and suggested possible new therapeutic targets. Overall, this work represents the first report of a genetic mutation known to cause human FTC that exhibits a similar phenotype when modeled in the mouse. In addition to our knowledge of the mechanisms of human follicular thyroid tumorigenesis, this model is highly reproducible and may provide a viable mechanism for the further clinical development of therapies aimed at FTC.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Modelos Animales de Enfermedad , Hipertiroidismo/genética , Neoplasias de la Tiroides/genética , Adenocarcinoma Folicular , Animales , Diferenciación Celular , Proliferación Celular , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Hipertiroidismo/metabolismo , Hipertiroidismo/patología , Ratones , Ratones Noqueados , Factor de Transcripción STAT3/metabolismo , Glándula Tiroides/patología , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Células Tumorales Cultivadas
7.
Endocrinol Nutr ; 58(6): 308-14, 2011.
Artículo en Español | MEDLINE | ID: mdl-21536508

RESUMEN

Carney complex (CNC) is an autosomal dominantly inherited syndrome characterized by spotty skin pigmentation, cardiac and cutaneous myxoma, and endocrine overactivity. Skin pigmentation includes lentigines and blue nevi. Myxomas may occur in breast, skin and heart. Cardiac myxomas may be multiple and occur in any cardiac chamber, and are more prone to recurrence. The most common endocrine gland manifestation is an ACTH-independent Cushing's syndrome due to primary pigmented nodular adrenocortical disease (PPNAD). PPNAD may occur isolated, with no other signs of CNC. Pituitary and thyroid glands and gonads are also involved. The PRKAR1A gene, located in 17 q22-24, encodes type 1A regulatory subunit of protein kinase A. Inactivating germline mutations of this gene are found in 70% of patients with CNC. PRKAR1A is a key component of the c-AMP signaling pathway that has been implicated in endocrine tumorigenesis. Many different mutations have been reported in the PRKAR1A gene. In almost all cases the sequence change was predicted to lead to a premature stop codon and the resultant mutant mRNA was subject to nonsense-mediated mRNA decay. There is no clear genotype-phenotype correlation in patients with CNC. Genetic analysis should be performed in all CNC index cases. All affected patients should be monitored for clinical signs of CNC at least once a year. Genetic diagnosis allows for more effective preparation of more appropriate and effective therapeutic strategies and genetic counseling for patients and gene carriers, and to avoid unnecessary tests to relatives not carrying the gene.


Asunto(s)
Complejo de Carney , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Mixoma/genética , Síndromes Neoplásicos Hereditarios , Neoplasias de los Tejidos Blandos/genética , Acromegalia/etiología , Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/metabolismo , Complejo de Carney/diagnóstico , Complejo de Carney/epidemiología , Complejo de Carney/genética , Complejo de Carney/patología , Cromosomas Humanos Par 17/genética , Síndrome de Cushing/genética , AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/fisiología , Asesoramiento Genético , Mutación de Línea Germinal , Neoplasias Cardíacas/genética , Humanos , Hidrocortisona/metabolismo , Síndromes Neoplásicos Hereditarios/genética , Mutación Puntual , Sistemas de Mensajero Secundario , Neoplasias Cutáneas/genética
8.
PLoS Genet ; 6(6): e1000980, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20548949

RESUMEN

Carney complex (CNC) is an inherited neoplasia syndrome with endocrine overactivity. Its most frequent endocrine manifestation is primary pigmented nodular adrenocortical disease (PPNAD), a bilateral adrenocortical hyperplasia causing pituitary-independent Cushing's syndrome. Inactivating mutations in PRKAR1A, a gene encoding the type 1 alpha-regulatory subunit (R1alpha) of the cAMP-dependent protein kinase (PKA) have been found in 80% of CNC patients with Cushing's syndrome. To demonstrate the implication of R1alpha loss in the initiation and development of PPNAD, we generated mice lacking Prkar1a specifically in the adrenal cortex (AdKO). AdKO mice develop pituitary-independent Cushing's syndrome with increased PKA activity. This leads to autonomous steroidogenic genes expression and deregulated adreno-cortical cells differentiation, increased proliferation and resistance to apoptosis. Unexpectedly, R1alpha loss results in improper maintenance and centrifugal expansion of cortisol-producing fetal adrenocortical cells with concomitant regression of adult cortex. Our data provide the first in vivo evidence that loss of R1alpha is sufficient to induce autonomous adrenal hyper-activity and bilateral hyperplasia, both observed in human PPNAD. Furthermore, this model demonstrates that deregulated PKA activity favors the emergence of a new cell population potentially arising from the fetal adrenal, giving new insight into the mechanisms leading to PPNAD.


Asunto(s)
Corteza Suprarrenal/metabolismo , Síndrome de Cushing/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Animales , Proliferación Celular , Síndrome de Cushing/embriología , Síndrome de Cushing/genética , Síndrome de Cushing/patología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Transducción de Señal , Esteroide 17-alfa-Hidroxilasa/metabolismo
9.
Trends Cardiovasc Med ; 19(2): 44-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19577711

RESUMEN

Cardiac myxomas are the most common primary tumors of the heart, although little is known about their etiology. Mutations of the protein kinase A regulatory subunit gene PRKAR1A cause inherited myxomas in the setting of the Carney complex tumor syndrome, providing a possible window for understanding their pathogenesis. We recently reported that cardiac-specific knockout of this gene causes myxomatous changes in the heart, although the mice die during gestation from cardiac failure. In this review, we discuss these findings and place them in the larger understanding of how protein kinase A dysregulation might affect cardiac function and cause myxomagenesis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Neoplasias Cardíacas/enzimología , Corazón/crecimiento & desarrollo , Miocardio/enzimología , Mixoma/enzimología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/genética , Neoplasias Cardíacas/genética , Neoplasias Cardíacas/patología , Humanos , Ratones , Ratones Noqueados , Mutación , Miocardio/patología , Mixoma/genética , Mixoma/patología , Transducción de Señal
10.
Cancer Res ; 68(8): 2671-7, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18413734

RESUMEN

Dysregulation of protein kinase A (PKA) activity, caused by loss of function mutations in PRKAR1A, is known to induce tumor formation in the inherited tumor syndrome Carney complex (CNC) and is also associated with sporadic tumors of the thyroid and adrenal. We have previously shown that Prkar1a(+/-) mice develop schwannomas reminiscent of those seen in CNC and that similar tumors are observed in tissue-specific knockouts (KO) of Prkar1a targeted to the neural crest. Within these tumors, we have previously described the presence of epithelial islands, although the nature of these structures was unclear. In this article, we report that these epithelial structures are derived from KO cells originating in the neural crest. Analysis of the mesenchymal marker vimentin revealed that this protein was markedly down-regulated not only from the epithelial islands, but also from the tumor as a whole, consistent with mesenchymal-to-epithelial transition (MET). In vitro, Prkar1a null primary mouse embryonic fibroblasts, which display constitutive PKA signaling, also showed evidence for MET, with a loss of vimentin and up-regulation of the epithelial marker E-cadherin. Reduction of vimentin protein occurred at the posttranslational level and was rescued by proteasomal inhibition. Finally, this down-regulation of vimentin was recapitulated in the adrenal nodules of CNC patients, confirming an unexpected and previously unrecognized role for PKA in MET.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Células Epiteliales/citología , Eliminación de Gen , Mesodermo/citología , Neoplasia Endocrina Múltiple/genética , Neoplasias/genética , Animales , Diferenciación Celular , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Epiteliales/enzimología , Humanos , Mesodermo/enzimología , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Procesamiento Proteico-Postraduccional , Vimentina/metabolismo
11.
Circulation ; 117(11): 1414-22, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18316483

RESUMEN

BACKGROUND: Protein kinase A signaling has long been known to play an important role in cardiac function. Dysregulation of the protein kinase A system, caused by mutation of the protein kinase A regulatory subunit gene PRKAR1A, causes the inherited tumor syndrome Carney complex, which includes cardiac myxomas as one of its cardinal features. Mouse models of this genetic defect have been unsatisfactory because homozygote null animals die early in development and heterozygotes do not exhibit a cardiac phenotype. METHODS AND RESULTS: To study the cardiac-specific effects resulting from complete loss of Prkar1a, we used cre-lox technology to generate mice lacking this protein specifically in cardiomyocytes. Conditional knockout mice died at day 11.5 to 12.5 of embryogenesis with thin-walled, dilated hearts. These hearts showed elevated protein kinase A activity and decreased cardiomyocyte proliferation before demise. Analysis of the expression of transcription factors required for cardiogenesis revealed downregulation of key cardiac transcription factors such as the serum response factor, Gata4, and Nkx2-5. Although heart wall thickness was reduced overall, specific areas exhibited morphological changes consistent with myxomatous degeneration in the walls of knockout hearts. CONCLUSIONS: Loss of Prkar1a from the heart causes a failure of proper myocardial development with subsequent cardiac failure and embryonic demise. These changes appear to be due to suppression of cardiac-specific transcription by increased protein kinase A activity. These biochemical changes lead to myxoma-like changes, indicating that these mice may be a good model with which to study the formation of these tumors.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Corazón Fetal/patología , Neoplasias Cardíacas/genética , Mixoma/genética , Animales , Apoptosis , División Celular , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Regulación hacia Abajo , Muerte Fetal/enzimología , Muerte Fetal/genética , Corazón Fetal/enzimología , Corazón Fetal/ultraestructura , Genes Letales , Neoplasias Cardíacas/patología , Integrasas , Ratones , Ratones Noqueados , Modelos Animales , Miocitos Cardíacos/enzimología , Mixoma/patología , Síndromes Neoplásicos Hereditarios/enzimología , Síndromes Neoplásicos Hereditarios/genética , Especificidad de Órganos , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA