Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Endocrine ; 72(3): 823-834, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33420948

RESUMO

PURPOSE: Insulin-like growth factor-II (IGF2), a key regulator of cell growth and development, is tightly regulated in its expression by epigenetic control that maintains its monoallelic expression in most tissues. Biallelic expression of IGF2 resulting from loss of imprinting (LOI) has been reported in adrenocortical tumors. In this study, we wanted to check whether adrenocortical lesions due to PRKAR1A mutations lead to increased IGF2 expression from LOI and compare these findings to those in other benign adrenal lesions. METHODS: We compared the expression of IGF2 by RNA and protein studies in primary pigmented nodular adrenocortical disease (PPNAD) caused by PRKAR1A gene mutations to that in primary macronodular adrenocortical hyperplasia (PMAH) and cortisol-producing adenomas (CPA) that did not have any mutations in known genes. We also checked LOI in all lesions by DNA allelic studies and the expression of other components of IGF2 signaling at the RNA and protein level. RESULTS: We identified cell clusters overexpressing IGF2 in PPNAD; although immunostaining was patchy, overall, by RNA and immunoblotting PPNAD expressed high IGF2 message and protein. However, this was not due to LOI, as there was no correlation between IGF2 expression and the presence of LOI. CONCLUSIONS: Our data pointed to over-expression of IGF2 protein in PPNAD compared to other benign adrenocortical lesions, such as PMAH and CPA. However, there was no correlation of IGF2 mRNA levels with LOI of IGF2/H19. The discrepancy between mRNA and protein levels with regards to LOI points, perhaps, to different control of IGF2 gene expression in PPNAD.


Assuntos
Doenças do Córtex Suprarrenal , Neoplasias do Córtex Suprarrenal , Neoplasias do Córtex Suprarrenal/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico , Humanos , Fator de Crescimento Insulin-Like II/genética , Mutação , RNA Mensageiro , Fatores de Transcrição
2.
Endocr Relat Cancer ; 27(10): 591-599, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32738126

RESUMO

Protein kinase A (PKA) regulatory subunit type 1A (PRKAR1A) defects lead to primary pigmented nodular adrenocortical disease (PPNAD). The KIT protooncogene (c-KIT) is not known to be expressed in the normal adrenal cortex (AC). In this study, we investigated the expression of c-KIT and its ligand, stem cell factor (SCF), in PPNAD and other cortisol-producing tumors of the adrenal cortex. mRNA and protein expression, by qRT-PCR, immunohistochemistry (IHC) and immunoblotting (IB), respectively, were studied. We then tested c-KIT and SCF responses to PRKAR1A introduction and PKA stimulation in adrenocortical cell lines CAR47 and H295R, which were also treated with the KIT inhibitor, imatinib mesylate (IM). Mice xenografted with H295R cells were treated with IM. There was increased c-KIT mRNA expression in PPNAD; IHC showed KIT and SCF immunoreactivity within certain nodular areas in PPNAD. IB data was consistent with IHC and mRNA data. PRKAR1A-deficient CAR47 cells expressed c-KIT; this was enhanced by forskolin and lowered by PRKAR1A reintroduction. Knockdown of PKA's catalytic subunit (PRKACA) by siRNA reduced c-KIT levels. Treatment of the CAR47 cells with IM resulted in reduced cell viability, growth arrest, and apoptosis. Treatment with IM of mice xenografted with H295 cells inhibited further tumor growth. We conclude that c-KIT is expressed in PPNAD, an expression that appears to be dependent on PRKAR1A and/or PKA activity. In a human adrenocortical cell line and its xenografts in mice, c-KIT inhibition decreased growth, suggesting that c-KIT inhibitors may be a reasonable alternative therapy to be tested in PPNAD, when other treatments are not optimal.


Assuntos
Neoplasias do Córtex Suprarrenal/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Neoplasias do Córtex Suprarrenal/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Humanos , Mesilato de Imatinib/farmacologia , Camundongos , Camundongos Nus , Oncogenes , Distribuição Aleatória , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Clin Endocrinol Metab ; 98(8): E1393-400, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23771924

RESUMO

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


Assuntos
AMP Cíclico/fisiologia , Neoplasias Embrionárias de Células Germinativas/genética , Diester Fosfórico Hidrolases/genética , Polimorfismo de Nucleotídeo Único , Proteínas Proto-Oncogênicas c-kit/fisiologia , Transdução de Sinais/fisiologia , Fator de Células-Tronco/genética , Neoplasias Testiculares/genética , 3',5'-GMP Cíclico Fosfodiesterases , Linhagem Celular Tumoral , AMP Cíclico/análise , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Masculino , Neoplasias Embrionárias de Células Germinativas/etiologia , Diester Fosfórico Hidrolases/análise , Proteínas Proto-Oncogênicas c-kit/análise , Fator de Células-Tronco/análise , Neoplasias Testiculares/etiologia
4.
Cell Biol Toxicol ; 28(6): 409-19, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22976903

RESUMO

TCPTP is an ubiquitously expressed tyrosine phosphatase with a predominant nuclear isoform (TC45) that binds DNA and has a role in G1-S cell cycle progression. Its deregulation by overexpression induces p53-dependent apoptosis, but the physiological role of its DNA-binding function is not known. Using immunocytochemistry and subcellular fractionation, we investigated changes in its localization in response to DNA damage and replication arrest. Rat fibroblasts showed an increase in endogenous TCPTP bound to nuclear components 3 h after exposure to sublethal dose of UV irradiation. Fractionation of nuclei showed an increase in chromatin and nuclear matrix associated component of TC45. After UV treatment, cells showed a concentration of TCPTP in discrete foci and enhanced colocalization with PCNA and p53BP1. Cells arrested at G1-S transition by hydroxyurea showed a loss of the predominant nuclear staining of TCPTP and an increase in cytoplasmic staining. Upon release from replication block, there was a time-dependent increase in number of cells showing prominent nuclear localization. This change in localization coincides with that of PCNA and Cdk2, two other nuclear proteins having functions in DNA replication. These results provide evidence for the regulation of TCPTP in response to DNA damage and replication stress. Dynamic changes in its localization coincident with that of PCNA suggest involvement of TCPTP in DNA repair and replication.


Assuntos
Núcleo Celular/metabolismo , Dano ao DNA , Reparo do DNA , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Hidroxiureia/farmacologia , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Raios Ultravioleta
5.
EMBO Rep ; 10(6): 599-605, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19424295

RESUMO

Proper regulation of the cAMP-dependent protein kinase (protein kinase A, PKA) is necessary for cellular homeostasis, and dysregulation of this kinase is crucial in human disease. Mouse embryonic fibroblasts (MEFs) lacking the PKA regulatory subunit Prkar1a show altered cell morphology and enhanced migration. At the molecular level, these cells showed increased phosphorylation of cofilin, a crucial modulator of actin dynamics, and these changes could be mimicked by stimulating the activity of PKA. Previous studies of cofilin have shown that it is phosphorylated primarily by the LIM domain kinases Limk1 and Limk2, which are under the control of the Rho GTPases and their downstream effectors. In Prkar1a(-/-) MEFs, neither Rho nor Rac was activated; rather, we showed that PKA could directly phosphorylate Limk1 and thus enhance the phosphorylation of cofilin. These data indicate that PKA is crucial in cell morphology and migration through its ability to modulate directly the activity of LIM kinase.


Assuntos
Actinas/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Quinases Lim/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Ativação Enzimática , Células HeLa , Humanos , Camundongos , Modelos Biológicos , Fosforilação
6.
Retrovirology ; 5: 46, 2008 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-18541021

RESUMO

BACKGROUND: Adult T-cell leukemia/lymphoma (ATLL) is initiated by infection with human T-lymphotropic virus type-1 (HTLV-1); however, additional host factors are also required for T-cell transformation and development of ATLL. The HTLV-1 Tax protein plays an important role in the transformation of T-cells although the exact mechanisms remain unclear. Parathyroid hormone-related protein (PTHrP) plays an important role in the pathogenesis of humoral hypercalcemia of malignancy (HHM) that occurs in the majority of ATLL patients. However, PTHrP is also up-regulated in HTLV-1-carriers and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) patients without hypercalcemia, indicating that PTHrP is expressed before transformation of T-cells. The expression of PTHrP and the PTH/PTHrP receptor during immortalization or transformation of lymphocytes by HTLV-1 has not been investigated. RESULTS: We report that PTHrP was up-regulated during immortalization of lymphocytes from peripheral blood mononuclear cells by HTLV-1 infection in long-term co-culture assays. There was preferential utilization of the PTHrP-P2 promoter in the immortalized cells compared to the HTLV-1-transformed MT-2 cells. PTHrP expression did not correlate temporally with expression of HTLV-1 tax. HTLV-1 infection up-regulated the PTHrP receptor (PTH1R) in lymphocytes indicating a potential autocrine role for PTHrP. Furthermore, co-transfection of HTLV-1 expression plasmids and PTHrP P2/P3-promoter luciferase reporter plasmids demonstrated that HTLV-1 up-regulated PTHrP expression only mildly, indicating that other cellular factors and/or events are required for the very high PTHrP expression observed in ATLL cells. We also report that macrophage inflammatory protein-1alpha (MIP-1alpha), a cellular gene known to play an important role in the pathogenesis of HHM in ATLL patients, was highly expressed during early HTLV-1 infection indicating that, unlike PTHrP, its expression was enhanced due to activation of lymphocytes by HTLV-1 infection. CONCLUSION: These data demonstrate that PTHrP and its receptor are up-regulated specifically during immortalization of T-lymphocytes by HTLV-1 infection and may facilitate the transformation process.


Assuntos
Transformação Celular Viral , Vírus Linfotrópico T Tipo 1 Humano/crescimento & desenvolvimento , Leucócitos Mononucleares/virologia , Proteína Relacionada ao Hormônio Paratireóideo/biossíntese , Sobrevivência Celular , Células Cultivadas , Quimiocina CCL3/biossíntese , Técnicas de Cocultura , Produtos do Gene tax/biossíntese , Humanos , Receptor Tipo 1 de Hormônio Paratireóideo/biossíntese , Fatores de Tempo , Regulação para Cima
7.
Cancer Res ; 68(8): 2671-7, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18413734

RESUMO

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.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/deficiência , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Células Epiteliais/citologia , Deleção de Genes , Mesoderma/citologia , Neoplasia Endócrina Múltipla/genética , Neoplasias/genética , Animais , Diferenciação Celular , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Epiteliais/enzimologia , Humanos , Mesoderma/enzimologia , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Processamento de Proteína Pós-Traducional , Vimentina/metabolismo
8.
Mol Endocrinol ; 22(2): 430-40, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17932105

RESUMO

Carney complex (CNC) is an autosomal dominant neoplasia syndrome caused by inactivating mutations in PRKAR1A, the gene encoding the type 1A regulatory subunit of protein kinase A (PKA). This genetic defect induces skin pigmentation, endocrine tumors, myxomas, and schwannomas. Some patients with the complex also develop myxoid bone tumors termed osteochondromyxomas. To study the link between the PRKAR1A mutations and tumor formation, we generated a mouse model of this condition. Prkar1a(+/-) mice develop bone tumors with high frequency, although these lesions have not yet been characterized, either from human patients or from mice. Bone tumors from Prkar1a(+/-) mice were heterogeneous, including elements of myxomatous, cartilaginous, and bony differentiation that effaced the normal bone architecture. Immunohistochemical analysis identified an osteoblastic origin for the abnormal cells associated with islands of bone. To better understand these cells at the biochemical level, we isolated primary cultures of tumoral bone and compared them with cultures of bone from wild-type animals. The tumor cells exhibited the expected decrease in Prkar1a protein and exhibited increased PKA activity. At the phenotypic level, we observed that tumor cells behaved as incompletely differentiated osteoblasts and were able to form tumors in immunocompromised mice. Examination of gene expression revealed down-regulation of markers of bone differentiation and increased expression of locally acting growth factors, including members of the Wnt signaling pathway. Tumor cells exhibited enhanced growth in response to PKA-stimulating agents, suggesting that tumorigenesis in osteoblast precursor cells is driven by effects directly mediated by the dysregulation of PKA.


Assuntos
Neoplasias Ósseas/patologia , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Mutação , Osteoblastos/patologia , Animais , Western Blotting , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Células Cultivadas , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Osteoblastos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo , Células Tumorais Cultivadas
9.
Cancer Res ; 65(22): 10307-15, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16288019

RESUMO

Phosphorylation is a key event in cell cycle control, and dysregulation of this process is observed in many tumors, including those associated with specific inherited neoplasia syndromes. We have shown previously that patients with the autosomal dominant tumor predisposition Carney complex carry inactivating mutations in the PRKAR1A gene, which encodes the type 1A regulatory subunit of protein kinase A (PKA), the cyclic AMP-dependent protein kinase. This defect was associated with dysregulation of PKA signaling, and genetic analysis has suggested that complete loss of the gene may be required for tumorigenesis. To determine the mechanism by which dysregulation of PKA causes tumor formation, we generated in vitro primary mouse cells lacking the Prkar1a protein. We report that this genetic disruption of PKA regulation causes constitutive PKA activation and immortalization of primary mouse embryonic fibroblasts (MEFs). At the molecular level, knockout of Prkar1a leads to up-regulation of D-type cyclins, and this increase occurs independently of other pathways known to increase cyclin D levels. Despite the immortalized phenotype, known mediators of cellular senescence (e.g., p53 and p19ARF) seem to remain intact in Prkar1a-/- MEFs. Mechanistically, cyclin D1 mRNA levels are not altered in the knockout cells, but protein half-life is markedly increased. Using this model, we provide the first direct genetic evidence that dysregulation of PKA promotes important steps in tumorigenesis, and that cyclin D1 is an essential target of PKA.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ciclina D1/biossíntese , Proteínas/fisiologia , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico/biossíntese , Proteínas Quinases Dependentes de AMP Cíclico/genética , Ciclina D1/genética , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/enzimologia , Fibroblastos/fisiologia , Humanos , Camundongos , Camundongos Knockout , Proteínas/genética , Transdução de Sinais , Transfecção , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...