Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Physiol Rep ; 8(6): e14405, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32212257

RESUMO

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.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/deficiência , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Ventrículos do Coração/patologia , Miócitos Cardíacos/patologia , Adolescente , Adulto , Animais , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Feminino , Humanos , Hipertrofia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Tamanho do Órgão , Adulto Jovem
2.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 41(11): 1148-1154, 2016 Nov 28.
Artigo em Chinês | MEDLINE | ID: mdl-27932759

RESUMO

OBJECTIVE: To evaluate the expression of cAMP-dependent protein kinase type I-alpha regulatory subunit (PRKAR1α) in non-small cell lung cancer (NSCLC) and its correlation with clinicopathological features.
 Methods: PRKAR1α expressions in 79 NSCLC patients and matched adjacent non-carcinoma tissues were analyzed by using qRT-PCR and immunohistochemistry.
 Results: The negative rates of PRKAR1α protein in NSCLC, lung squamous cell carcinoma (SCL) and lung adenocarcinoma (ACL) were 58.2%, 77.8%, 32.4%, respectively. Compared to the matched adjacent non-carcinoma tissues, there were significant differences in levels of PRKAR1α mRNA and protein in ACL (P<0.05), but not in SCL and overall NSCLC (P>0.05). The expression of PRKAR1α protein was positively correlated with histological type, TNM stage, and lymph node metastasis (P<0.05). Tumor size and histogenesis differentiation were not related to the decreased PRKAR1α (P>0.05).
 Conclusion: Low expression of PRKAR1α in ACL might be involved in the pathogenesis, which might serve as a novel diagnostic candidate.


Assuntos
Adenocarcinoma/química , Carcinoma Pulmonar de Células não Pequenas/química , Carcinoma de Células Escamosas/química , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Neoplasias Pulmonares/química , Adenocarcinoma/classificação , Adenocarcinoma/genética , Adenocarcinoma de Pulmão , Biomarcadores Tumorais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma de Células Escamosas/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/classificação , Neoplasias Pulmonares/genética , Metástase Linfática/genética , Masculino , Estadiamento de Neoplasias , RNA Mensageiro
3.
J Gastroenterol Hepatol ; 30(3): 496-503, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25213315

RESUMO

BACKGROUND AND AIM: Previously study showed that the loss of the control of cAMP-dependent protein kinase A RIα (PKA RIα)/ A-kinase anchoring proteins 10 (AKAP10) signaling pathway initiate dysregulation of cellular healthy physiology leading to tumorigenesis. The aim of this study was to investigate the role of PKA RIα/AKAP10 signaling pathway in colorectal cancer (CRC). METHODS: The AKAP10 expression at the mRNA and protein level have been analyzed in colon cancer cell lines, primary CRCs and matched normal mucosa samples, and compared in accordance with specific clinicopathological features of CRC. The correlation between expression of AKAP10 and PKA RIα were also analyzed. RESULTS: Compared with HCT116 and SW480 cells, the AKAP10 was significantly upregulated in the colon cell line KM12C and its metastatic counterparts, KM12SM and KM12L4A. Moreover, the KM12SM and KM12L4A having high metastatic potentials displayed the elevated levels of AKAP10 compared with KM12C having poor metastatic potential. A notably higher level of AKAP10 expression was found in CRC tissues at both mRNA and protein levels. Increased expression of AKAP10 in CRC patients was positively associated with the depth of invasion and the grade of differentiation. Univariate survival analysis showed that the increased expression of AKAP10 was related to poorer survival. Cox multivariate regression analysis confirmed that AKAP10 was an independent predictor of the overall survival of CRC patients. PKA RIα mRNA was also expressed at high levels in CRC. The correlation coefficient between mRNA expression of AKAP10 and PKA RIα in CRC was 0.417. AKAP10 mRNA overexpression was correlated significantly with PKA RIα. CONCLUSIONS: Our data indicated that PKA RIα/AKAP10 signaling pathway is associated with the progression and prognosis of CRC.


Assuntos
Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/fisiologia , Neoplasias Colorretais/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Expressão Gênica/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Idoso , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Prognóstico , Modelos de Riscos Proporcionais , RNA Mensageiro/genética , Sobrevida , Regulação para Cima/genética
4.
Mol Pharmacol ; 85(2): 357-67, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24307699

RESUMO

Cardiomyocyte apoptosis contributes toward the loss of muscle mass in myocardial pathologies. Previous reports have implicated type I cAMP-dependent protein kinase (PKA) and p90 ribosomal S6 kinase (RSK) in cardiomyocyte apoptosis. However, the precise mechanisms and the isoform of RSK involved in this process remain undefined. Using adult rat ventricular myocytes and mouse-derived cardiac HL-1 cardiomyocytes, we demonstrate that hypoxia/reoxygenation (H/R)-induced apoptosis is accompanied by a decrease in the type I PKA regulatory subunit (PKARIα) and activation of RSK1. As previously described by us for other cell types, in cardiomyocytes, inactive RSK1 also interacts with PKARIα, whereas the active RSK1 interacts with the catalytic subunit of PKA. Additionally, small interfering (siRNA)-mediated silencing of PKARIα or disrupting the RSK1/PKARIα interactions with a small, cell-permeable peptide activates RSK1 and recapitulates the H/R-induced apoptosis. Inhibition of RSK1 or siRNA-mediated silencing of RSK1 attenuates H/R-induced apoptosis, demonstrating the role of RSK1 in cardiomyocyte apoptosis. Furthermore, silencing of RSK1 decreases the H/R-induced phosphorylation of sodium-hydrogen exchanger 1 (NHE1), and inhibition of NHE1 with 5'-N-ethyl-N-isopropyl-amiloride blocks H/R induced apoptosis, indicating the involvement of NHE1 in apoptosis. Overall, our findings demonstrate that H/R-mediated decrease in PKARIα protein levels leads to activation of RSK1, which via phosphorylation of NHE1 induces cardiomyocyte apoptosis.


Assuntos
Apoptose , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Miócitos Cardíacos/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia , Animais , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , Camundongos , Miócitos Cardíacos/enzimologia , Fosforilação , Ratos , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/metabolismo
5.
J Neurosci ; 33(46): 17967-75, 2013 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-24227708

RESUMO

Signaling through cAMP has been implicated in Schwann cell (SC) proliferation and myelination, but the signaling pathway components downstream of cAMP required for SC function remain unknown. Protein kinase A (PKA) is a potential downstream effector of cAMP. Here, we induced loss of Prkar1a, the gene encoding the type 1A regulatory subunit of PKA, in SC to study its role in nerve development; loss of Prkar1a is predicted to elevate PKA activity. Conditional Prkar1a knock-out in mouse SC (Prkar1a-SCKO) resulted in a dramatic and persistent axonal sorting defect, and unexpectedly decreased SC proliferation in Prkar1a-SCKO nerves in vivo. Effects were cell autonomous as they were recapitulated in vitro in Prkar1a-SCKO SC, which showed elevated PKA activity. In the few SCs sorted into 1:1 relationships with axons in vivo, SC myelination was premature in Prkar1a-SCKO nerves, correlating with global increase in the cAMP-regulated transcription factor Oct-6 and expression of myelin basic protein. These data reveal a previously unknown role of PKA in axon sorting, an unexpected inhibitory role of PKA on SC cell proliferation in vivo and define the importance of Prkar1a in peripheral nerve development.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Nervos Periféricos/embriologia , Nervos Periféricos/crescimento & desenvolvimento , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Feminino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez
6.
Am J Respir Cell Mol Biol ; 48(5): 626-34, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23349050

RESUMO

Elevated concentrations of CO2 (hypercapnia) lead to alveolar epithelial dysfunction by promoting Na,K-ATPase endocytosis. In the present report, we investigated whether the CO2/HCO3(-) activated soluble adenylyl cyclase (sAC) regulates this process. We found that hypercapnia increased the production of cyclic adenosine monophosphate (cAMP) and stimulated protein kinase A (PKA) activity via sAC, which was necessary for Na,K-ATPase endocytosis. During hypercapnia, cAMP was mainly produced in specific microdomains in the proximity of the plasma membrane, leading to PKA Type Iα activation. In alveolar epithelial cells exposed to high CO2 concentrations, PKA Type Iα regulated the time-dependent phosphorylation of the actin cytoskeleton component α-adducin at serine 726. Cells expressing small hairpin RNA for PKAc, dominant-negative PKA Type Iα, small interfering RNA for α-adducin, and α-adducin with serine 726 mutated to alanine prevented Na,K-ATPase endocytosis. In conclusion, we provide evidence for a new mechanism by which hypercapnia via sAC, cAMP, PKA Type Iα, and α-adducin regulates Na,K-ATPase endocytosis in alveolar epithelial cells.


Assuntos
Células Epiteliais Alveolares/enzimologia , Dióxido de Carbono/farmacologia , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Endocitose , ATPase Trocadora de Sódio-Potássio/metabolismo , Adenilil Ciclases/metabolismo , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/fisiologia , Animais , Proteínas de Ligação a Calmodulina/metabolismo , Linhagem Celular Tumoral , Membrana Celular/enzimologia , AMP Cíclico/metabolismo , Humanos , Hipercapnia/enzimologia , Fosforilação , Proteína Quinase C/metabolismo , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Sistemas do Segundo Mensageiro
7.
Oncogene ; 32(30): 3491-9, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23045281

RESUMO

Schwannomas are peripheral nerve sheath tumors that often occur in the setting of an inherited tumor predisposition syndrome, including neurofibromatosis types 1 (NF1) and 2 (NF2), familial schwannomatosis and Carney complex. Loss of the NF2 tumor suppressor (encoding NF2, or Merlin) is associated with upregulation of the Rac1 small GTPase, which is thought to have a key role in mediating tumor formation. In prior studies, we generated a mouse model of schwannomas by performing tissue-specific knockout (KO) of the Carney complex gene Prkar1a, which encodes the type 1A regulatory subunit of protein kinase A. These tumors exhibited down-regulation of Nf2 protein and an increase in activated Rac1. To assess the requirement for Rac1 in schwannoma formation, we generated a double KO (DKO) of Prkar1a and Rac1 in Schwann cells and monitored tumor formation. Loss of Rac1 reduced tumor formation by reducing proliferation and enhancing apoptosis. Surprisingly, the reduction of tumor formation was accompanied by re-expression of the Nf2 protein. Furthermore, activated Rac1 was able to downregulate Nf2 in vitro in a Pak-dependent manner. These in vivo data indicate that activation of Rac1 is responsible for suppression of Nf2 protein production; deficiency of Nf2 in Schwann cells leads to loss of cellular growth control and tumor formation. Further, PKA activation through mutation in Prkar1a is sufficient to initiate Rac1 signaling, with subsequent reduction of Nf2 and schwannomagenesis. Although in vitro evidence has shown that loss of Nf2 activates Rac1, our data indicate that signaling between Nf2 and Rac1 occurs in a bidirectional fashion, and these interactions are modulated by PKA.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Genes da Neurofibromatose 2 , Neurilemoma/genética , Neuropeptídeos/fisiologia , Proteínas rac de Ligação ao GTP/fisiologia , Animais , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Regulação para Baixo/genética , Camundongos , Camundongos Knockout , Neurilemoma/patologia , Neuropeptídeos/genética , Células de Schwann/patologia , Proteínas rac de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP
8.
J Clin Endocrinol Metab ; 97(12): E2328-38, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23043190

RESUMO

CONTEXT: Acrodysostosis is a rare skeletal dysplasia that is associated with multiple resistance to G protein-coupled receptor (GPCR) signaling hormones in a subset of patients. Acrodysostosis is genetically heterogeneous because it results from heterozygous mutations in PRKAR1A or PDE4D, two key actors in the GPCR-cAMP-protein kinase A pathway. OBJECTIVE: Our objective was to identify the phenotypic features that distinguish the two genotypes causing acrodysostosis. PATIENTS AND METHODS: Sixteen unrelated patients with acrodysostosis underwent a candidate-gene approach and were investigated for phenotypic features. RESULTS: All patients had heterozygous de novo mutations. Fourteen patients carried a PRKAR1A mutation (PRKAR1A patients), five each a novel PRKAR1A mutation (p.Q285R, p.G289E, p.A328V, p.R335L, or p.Q372X), nine the reported PRKAR1A p.R368X mutation; two patients harbored a mutation in PDE4D (PDE4D patients) (one novel mutation, p.A227S; one reported, p.E590A). All PRKAR1A, but none of the PDE4D mutated patients were resistant to PTH and TSH. Two PRKAR1A patients each with a novel mutation presented a specific pattern of brachydactyly. One PDE4D patient presented with acroskyphodysplasia. Additional phenotypic differences included mental retardation in PDE4D patients. In addition, we report the presence of pigmented skin lesions in PRKAR1A and PDE4D patients, a feature not yet described in the acrodysostosis entity. CONCLUSIONS: All PRKAR1A and PDE4D patients present similar bone dysplasia characterizing acrodysostosis. Phenotypic differences, including the presence of resistance to GPCR-cAMP signaling hormones in PRKAR1A but not PDE4D patients, indicate phenotype-genotype correlations and highlight the specific contributions of PRKAR1A and PDE4D in cAMP signaling in different tissues.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/genética , Resistência a Medicamentos/genética , Disostoses/complicações , Disostoses/genética , Hormônios , Deficiência Intelectual/complicações , Deficiência Intelectual/genética , Osteocondrodisplasias/complicações , Osteocondrodisplasias/genética , Adolescente , Adulto , Criança , Pré-Escolar , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Técnicas de Diagnóstico Endócrino , Disostoses/diagnóstico , Feminino , Hormônios/metabolismo , Hormônios/farmacologia , Humanos , Deficiência Intelectual/diagnóstico , Masculino , Osteocondrodisplasias/diagnóstico , Hormônio Paratireóideo/administração & dosagem , Hormônio Paratireóideo/metabolismo , Hormônio Paratireóideo/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Síndrome , Síndrome da Resistência aos Hormônios Tireóideos/complicações , Síndrome da Resistência aos Hormônios Tireóideos/diagnóstico , Síndrome da Resistência aos Hormônios Tireóideos/genética , Adulto Jovem
9.
Psychoneuroendocrinology ; 37(6): 836-43, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22024111

RESUMO

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.


Assuntos
Ansiedade/genética , Ansiedade/psicologia , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Afeto/fisiologia , Animais , Ansiolíticos/farmacologia , Comportamento Animal/fisiologia , Química Encefálica/genética , Química Encefálica/fisiologia , DNA/genética , Emoções/fisiologia , Genótipo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Medição da Dor , Fenótipo , Reação em Cadeia da Polimerase , Estresse Psicológico/genética , Estresse Psicológico/psicologia
10.
Endocrinol Nutr ; 58(6): 308-14, 2011.
Artigo em Espanhol | MEDLINE | ID: mdl-21536508

RESUMO

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.


Assuntos
Complexo de Carney , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/deficiência , Mixoma/genética , Síndromes Neoplásicas Hereditárias , Neoplasias de Tecidos Moles/genética , Acromegalia/etiologia , Neoplasias do Córtex Suprarrenal/genética , Neoplasias do Córtex Suprarrenal/metabolismo , Complexo de Carney/diagnóstico , Complexo de Carney/epidemiologia , Complexo de Carney/genética , Complexo de Carney/patologia , Cromossomos Humanos Par 17/genética , Síndrome de Cushing/genética , AMP Cíclico/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Aconselhamento Genético , Mutação em Linhagem Germinativa , Neoplasias Cardíacas/genética , Humanos , Hidrocortisona/metabolismo , Síndromes Neoplásicas Hereditárias/genética , Mutação Puntual , Sistemas do Segundo Mensageiro , Neoplasias Cutâneas/genética
11.
Mol Endocrinol ; 24(8): 1559-68, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20534695

RESUMO

The cranial neural crest (CNC) undergoes complex molecular and morphological changes during embryogenesis in order to form the vertebrate skull, and nearly three quarters of all birth defects result from defects in craniofacial development. The molecular events leading to CNC differentiation have been extensively studied; however, the role of the cAMP-dependent protein kinase [protein kinase A (PKA)] during craniofacial development has only been described in palate formation. Here, we provide evidence that strict PKA regulation in postmigratory CNC cells is essential during craniofacial bone development. Selective inactivation of Prkar1a, a regulatory subunit of the PKA holoenzyme, in the CNC results in perinatal lethality caused by dysmorphic craniofacial development and subsequent asphyxiation. Additionally, aberrant differentiation of CNC mesenchymal cells results in anomalous intramembranous ossification characterized by formation of cartilaginous islands in some areas and osteolysis of bony trabeculae with fibrous connective tissue stabilization in others. Genetic interaction studies revealed that genetic reduction of the PKA catalytic subunit C(alpha) was able to rescue the phenotype, whereas reduction in Cbeta had no effect. Overall, these observations provide evidence of the essential role of proper regulation of PKA during the ossification of the bones of the skull. This knowledge may have implications for the understanding and treatment of craniofacial birth defects.


Assuntos
Anormalidades Craniofaciais/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Crista Neural/embriologia , Crista Neural/metabolismo , Ossificação Heterotópica/genética , Animais , Anormalidades Craniofaciais/diagnóstico por imagem , Anormalidades Craniofaciais/mortalidade , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Camundongos , Camundongos Knockout , Crista Neural/diagnóstico por imagem , Ossificação Heterotópica/diagnóstico por imagem , Ossificação Heterotópica/mortalidade , Microtomografia por Raio-X
12.
J Clin Endocrinol Metab ; 93(10): 4135-40, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18647815

RESUMO

BACKGROUND: The Wnt/beta-catenin and cAMP signaling pathways play an important role in adrenal cortex tumorigenesis. Somatic activating mutations of the beta-catenin gene (CTNNB1) are the most frequent genetic defects identified both in adrenocortical adenomas (ACAs) and adrenocortical cancers (ACCs). PRKAR1A mutations leading to cAMP pathway dysregulation are observed in primary pigmented nodular adrenocortical diseases (PPNADs) and some sporadic ACAs. OBJECTIVE: The objective of the investigation was to study Wnt/beta-catenin dysregulation in adrenocortical tumors (ACTs) with cAMP pathway genetic alteration and search for secondary CTNNB1 somatic mutations in heterogeneous tumors. PATIENTS AND METHODS: Nine PPNADs, including five with macronodules, three ACAs with PRKAR1A somatic mutations, and one heterogeneous tumor with ACC developed within an ACA, were studied by immunohistochemistry and DNA sequencing. RESULTS: beta-Catenin accumulation was observed in all PPNADs, ACAs with PRKAR1A mutations, and the ACC component of the heterogeneous tumor. CTNNB1 somatic activating mutations were found in the macronodule of two of the five macronodular PPNADs, in one ACA with a PRKAR1A somatic mutation, and in the malignant part of the heterogeneous ACT. CONCLUSIONS: The Wnt/beta-catenin pathway is activated in PPNADs and ACAs with PRKAR1A mutations, suggesting a cross talk between the cAMP and Wnt/beta-catenin pathways in ACT development. In addition, the occurrence as an additional hit of a CTNNB1 somatic mutation is associated with larger or more aggressive ACTs. This underlines the importance of the Wnt/beta-catenin pathway in adrenal cortex tumorigenesis and the importance of genetic accumulation in the progression of ACTs.


Assuntos
Adenoma/genética , Neoplasias do Córtex Suprarrenal/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Mutação , Transdução de Sinais/genética , Proteínas Wnt/genética , beta Catenina/genética , Adenoma/metabolismo , Adenoma/patologia , Adolescente , Neoplasias do Córtex Suprarrenal/metabolismo , Neoplasias do Córtex Suprarrenal/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , AMP Cíclico/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Análise Mutacional de DNA , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Mutação/fisiologia , Invasividade Neoplásica , Carga Tumoral/genética , beta Catenina/metabolismo
13.
Circulation ; 117(11): 1414-22, 2008 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-18316483

RESUMO

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.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/deficiência , Coração Fetal/patologia , Neoplasias Cardíacas/genética , Mixoma/genética , Animais , Apoptose , Divisão Celular , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Regulação para Baixo , Morte Fetal/enzimologia , Morte Fetal/genética , Coração Fetal/enzimologia , Coração Fetal/ultraestrutura , Genes Letais , Neoplasias Cardíacas/patologia , Integrases , Camundongos , Camundongos Knockout , Modelos Animais , Miócitos Cardíacos/enzimologia , Mixoma/patologia , Síndromes Neoplásicas Hereditárias/enzimologia , Síndromes Neoplásicas Hereditárias/genética , Especificidade de Órgãos , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...