Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Environ Toxicol ; 37(10): 2366-2374, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35730470

RESUMO

The effects of long non-coding RNA TDRG1 have been established in several tumors; however, its roles in colorectal cancer (CRC) progression are never been found. Here, we found that TDRG1 level was upregulated in CRC cells compared to that in normal colon epithelial cells. Additionally, TDRG1 level was remarkably upregulated in 3D non-adherent spheres derived from the parental CRC cells. Further in vitro and in vivo revealed that TDRG1 knockdown suppressed the stemness of CRC cells. What's more, combined with bioinformatics analysis, luciferase reporter and RNA pull down experiments showed that TDRG1 could bind to miR-873-5p, downregulated its level and thus increase the expression of PRKAR2. Finally, it was shown that TDRG1 functioned through the miR-873-5p/PRKAR2 axis. This study demonstrated a novel TDRG1/miR-873-5p/PRKAR2 signaling in CRC progression.


Assuntos
Neoplasias Colorretais , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , MicroRNAs , RNA Longo não Codificante , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
2.
Int J Mol Sci ; 21(9)2020 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-32357495

RESUMO

Protein kinase A (PKA) are tetramers of two catalytic and two regulatory subunits, docked at precise intracellular sites to provide localized phosphorylating activity, triggered by cAMP binding to regulatory subunits and subsequent dissociation of catalytic subunits. It is unclear whether in the brain PKA dissociated subunits may also be found. PKA catalytic subunit was examined in various mouse brain areas using immunofluorescence, equilibrium binding and western blot, to reveal its location in comparison to regulatory subunits type RI and RII. In the cerebral cortex, catalytic subunits colocalized with clusters of RI, yet not all RI clusters were bound to catalytic subunits. In stria terminalis, catalytic subunits were in proximity to RI but separated from them. Catalytic subunits clusters were also present in the corpus striatum, where RII clusters were detected, whereas RI clusters were absent. Upon cAMP addition, the distribution of regulatory subunits did not change, while catalytic subunits were completely released from regulatory subunits. Unpredictably, catalytic subunits were not solubilized; instead, they re-targeted to other binding sites within the tissue, suggesting local macromolecular reorganization. Hence, the interactions between catalytic and regulatory subunits of protein kinase A consistently vary in different brain areas, supporting the idea of multiple interaction patterns.


Assuntos
Encéfalo/enzimologia , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Animais , Córtex Cerebral/enzimologia , Corpo Estriado/enzimologia , AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Proteína Quinase Tipo II Dependente de AMP Cíclico/genética , Feminino , Masculino , Camundongos , Especificidade de Órgãos , Núcleos Septais/enzimologia
3.
J Mol Cell Cardiol ; 91: 215-27, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26773602

RESUMO

The balanced signaling between the two cyclic nucleotides (cNs) cAMP and cGMP plays a critical role in regulating cardiac contractility. Their degradation is controlled by distinctly regulated phosphodiesterase isoenzymes (PDEs), which in turn are also regulated by these cNs. As a result, PDEs facilitate communication between the ß-adrenergic and Nitric Oxide (NO)/cGMP/Protein Kinase G (PKG) signaling pathways, which regulate the synthesis of cAMP and cGMP respectively. The phenomena in which the cAMP and cGMP pathways influence the dynamics of each other are collectively referred to as cN cross-talk. However, the cross-talk response and the individual roles of each PDE isoenzyme in shaping this response remain to be fully characterized. We have developed a computational model of the cN cross-talk network that mechanistically integrates the ß-adrenergic and NO/cGMP/PKG pathways via regulation of PDEs by both cNs. The individual model components and the integrated network model replicate experimentally observed activation-response relationships and temporal dynamics. The model predicts that, due to compensatory interactions between PDEs, NO stimulation in the presence of sub-maximal ß-adrenergic stimulation results in an increase in cytosolic cAMP accumulation and corresponding increases in PKA-I and PKA-II activation; however, the potentiation is small in magnitude compared to that of NO activation of the NO/cGMP/PKG pathway. In a reciprocal manner, ß-adrenergic stimulation in the presence of sub-maximal NO stimulation results in modest cGMP elevation and corresponding increase in PKG activation. In addition, we demonstrate that PDE2 hydrolyzes increasing amounts of cAMP with increasing levels of ß-adrenergic stimulation, and hydrolyzes increasing amounts of cGMP with decreasing levels of NO stimulation. Finally, we show that PDE2 compensates for inhibition of PDE5 both in terms of cGMP and cAMP dynamics, leading to cGMP elevation and increased PKG activation, while maintaining whole-cell ß-adrenergic responses similar to that prior to PDE5 inhibition. By defining and quantifying reactions comprising cN cross-talk, the model characterizes the cross-talk response and reveals the underlying mechanisms of PDEs in this non-linear, tightly-coupled reaction system.


Assuntos
AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Redes Reguladoras de Genes , Modelos Cardiovasculares , Miocárdio/enzimologia , Miócitos Cardíacos/enzimologia , Diester Fosfórico Hidrolases/genética , Animais , Simulação por Computador , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/genética , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/genética , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Regulação da Expressão Gênica , Isoenzimas/genética , Isoenzimas/metabolismo , Contração Miocárdica , Miocárdio/citologia , Miócitos Cardíacos/citologia , Óxido Nítrico/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Transdução de Sinais
4.
J Biol Chem ; 290(49): 29250-8, 2015 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-26468277

RESUMO

Diabetes mellitus causes cardiac dysfunction and heart failure that is associated with metabolic abnormalities and autonomic impairment. Autonomic control of ventricular function occurs through regulation of cAMP-dependent protein kinase (PKA). The diabetic heart has suppressed ß-adrenergic responsiveness, partly attributable to receptor changes, yet little is known about how PKA signaling is directly affected. Control and streptozotocin-induced diabetic mice were therefore administered 8-bromo-cAMP (8Br-cAMP) acutely to activate PKA in a receptor-independent manner, and cardiac hemodynamic function and PKA signaling were evaluated. In response to 8Br-cAMP treatment, diabetic mice had impaired inotropic and lusitropic responses, thus demonstrating postreceptor defects. This impaired signaling was mediated by reduced PKA activity and PKA catalytic subunit content in the cytoplasm and myofilaments. Compartment-specific loss of PKA was reflected by reduced phosphorylation of discrete substrates. In response to 8Br-cAMP treatment, the glycolytic activator PFK-2 was robustly phosphorylated in control animals but not diabetics. Control adult cardiomyocytes cultured in lipid-supplemented media developed similar changes in PKA signaling, suggesting that lipotoxicity is a contributor to diabetes-induced ß-adrenergic signaling dysfunction. This work demonstrates that PKA signaling is impaired in diabetes and suggests that treating hyperlipidemia is vital for proper cardiac signaling and function.


Assuntos
Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/metabolismo , Miocárdio/enzimologia , 8-Bromo Monofosfato de Adenosina Cíclica/metabolismo , Animais , Domínio Catalítico , AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Citoplasma/metabolismo , Modelos Animais de Doenças , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/patologia , Hemodinâmica , Lactatos/metabolismo , Lipídeos/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Fosfofrutoquinase-2/metabolismo , Fosforilação , Transdução de Sinais
5.
Proc Natl Acad Sci U S A ; 108(20): 8281-6, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21536868

RESUMO

MicroRNA-protein complexes (microRNPs) can activate translation of target reporters and specific mRNAs in quiescent (i.e., G0) mammalian cell lines. Induced quiescent cells, like folliculated immature oocytes, have high levels of cAMP that activate protein kinase AII (PKAII) to maintain G0 and immature states. We report microRNA-mediated up-regulated expression of reporters in immature Xenopus laevis oocytes, dependent on Xenopus AGO or human AGO2 and on FXR1, as in mammalian cells. Importantly, we find that maintenance of cAMP levels and downstream PKAII signaling are required for microRNA-mediated up-regulated expression in oocytes. We identify an important, endogenous cell state regulator, Myt1 kinase, as a natural target of microRNA-mediated up-regulation in response to xlmiR16, ensuring maintenance of oocyte immaturity. Our data reveal the physiological relevance of cAMP/PKAII-controlled posttranscriptional gene expression activation by microRNAs in maintenance of the immature oocyte state.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/fisiologia , Biossíntese de Proteínas , Ribonucleoproteínas/fisiologia , Xenopus laevis/metabolismo , Animais , Proteínas Argonautas , AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Fator de Iniciação 2 em Eucariotos , Humanos , MicroRNAs/genética , Oócitos , Proteínas de Ligação a RNA , Xenopus laevis/genética
6.
Mol Pharmacol ; 83(5): 1057-65, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23455313

RESUMO

Acute promyelocytic leukemia (APL) is characterized by granulopoietic differentiation arrest at the promyelocytic stage. In most cases, this defect can be overcome by treatment with all-trans-retinoic acid (ATRA), leading to complete clinical remission. Cyclic AMP signaling has a key role in retinoid treatment efficacy: it enhances ATRA-induced maturation in ATRA-sensitive APL cells (including NB4 cells) and restores it in some ATRA-resistant cells (including NB4-LR1 cells). We show that the two cell types express identical levels of the Cα catalytic subunit and comparable global cAMP-dependent protein kinase A (PKA) enzyme activity. However, the maturation-resistant NB4-LR1 cells have a PKA isozyme switch: compared with the NB4 cells, they have decreased content of the juxtanuclearly located PKA regulatory subunit IIα and PKA regulatory subunit IIß, and a compensatory increase of the generally cytoplasmically distributed PKA-RIα. Furthermore, the PKA regulatory subunit II exists mainly in the less cAMP-responsive nonautophosphorylated state in the NB4-LR1 cells. By the use of isozyme-specific cAMP analog pairs, we show that both PKA-I and PKA-II must be activated to achieve maturation in NB4-LR1 as well as NB4 cells. Therefore, special attention should be paid to activating not only PKA-I but also PKA-II in attempts to enhance ATRA-induced APL maturation in a clinical setting.


Assuntos
Antineoplásicos/farmacologia , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/patologia , Tretinoína/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Humanos , Isoenzimas/metabolismo , Leucemia Promielocítica Aguda/enzimologia
7.
BMC Biochem ; 14: 13, 2013 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-23718778

RESUMO

BACKGROUND: Non-steroidal anti-inflammatory drugs (NSAIDs) -aspirin, naproxen, nimesulide, and piroxicam- lowered activation of type II cAMP-dependent protein kinase A (PKA-II) in isolated rat adipocytes, decreasing adrenaline- and dibutyryl cAMP (Bt2cAMP)-stimulated lipolysis. The molecular bases of insulin-like actions of NSAID were studied. RESULTS: Based on the reported inhibition of lipolysis by H2O2, catalase was successfully used to block NSAID inhibitory action on Bt2cAMP-stimulated lipolysis. NSAID, at (sub)micromolar range, induced an H2O2 burst in rat adipocyte plasma membranes and in whole adipocytes. NSAID-mediated rise of H2O2 was abrogated in adipocyte plasma membranes by: diphenyleneiodonium, an inhibitor of NADPH oxidase (NOX); the NOX4 antibody; and cytochrome c, trapping the NOX-formed superoxide. These three compounds prevented the inhibition of Bt2cAMP-stimulated lipolysis by NSAIDs. Inhibition of aquaporin-mediated H2O2 transport with AgNO3 in adipocytes allowed NOX activation but prevented the lipolysis inhibition promoted by NSAID: i.e., once synthesized, H2O2 must reach the lipolytic machinery. Since insulin inhibits adrenaline-stimulated lipolysis, the effect of aspirin on isoproterenol-stimulated lipolysis in rat adipocytes was studied. As expected, isoproterenol-mediated lipolysis was blunted by both insulin and aspirin. CONCLUSIONS: NSAIDs activate NOX4 in adipocytes to produce H2O2, which impairs cAMP-dependent PKA-II activation, thus preventing isoproterenol-activated lipolysis. H2O2 signaling in adipocytes is a novel and important cyclooxygenase-independent effect of NSAID.


Assuntos
Adipócitos/enzimologia , Anti-Inflamatórios não Esteroides/farmacologia , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Peróxido de Hidrogênio/metabolismo , Lipólise/efeitos dos fármacos , NADPH Oxidases/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Aquaporinas/farmacologia , Ativação Enzimática/efeitos dos fármacos , Masculino , NADPH Oxidases/antagonistas & inibidores , Ratos , Ratos Wistar , Nitrato de Prata/farmacologia
8.
J Biol Chem ; 286(1): 530-41, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21056973

RESUMO

Efficient and specific phosphorylation of PKA substrates, elicited in response to ß-adrenergic stimulation, require spatially confined pools of PKA anchored in proximity of its substrates. PKA-dependent phosphorylation of cardiac sarcomeric proteins has been the subject of intense investigations. Yet, the identity, composition, and function of PKA complexes at the sarcomeres have remained elusive. Here we report the identification and characterization of a novel sarcomeric AKAP (A-kinase anchoring protein), cardiac troponin T (cTnT). Using yeast two-hybrid technology in screening two adult human heart cDNA libraries, we identified the regulatory subunit of PKA as interacting with human cTnT bait. Immunoprecipitation studies show that cTnT is a dual specificity AKAP, interacting with both PKA-regulatory subunits type I and II. The disruptor peptide Ht31, but not Ht31P (control), abolished cTnT/PKA-R association. Truncations and point mutations identified an amphipathic helix domain in cTnT as the PKA binding site. This was confirmed by a peptide SPOT assay in the presence of Ht31 or Ht31P (control). Gelsolin-dependent removal of thin filament proteins also reduced myofilament-bound PKA-type II. Using a cTn exchange procedure that substitutes the endogenous cTn complex with a recombinant cTn complex we show that PKA-type II is troponin-bound in the myofilament lattice. Displacement of PKA-cTnT complexes correlates with a significant decrease in myofibrillar PKA activity. Taken together, our data propose a novel role for cTnT as a dual-specificity sarcomeric AKAP.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Troponina T/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/química , Células HEK293 , Humanos , Modelos Moleculares , Conformação Proteica , Estabilidade Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Ratos , Sarcômeros/metabolismo , Especificidade por Substrato , Troponina T/química , Técnicas do Sistema de Duplo-Híbrido
9.
BMC Biochem ; 12: 7, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21303506

RESUMO

BACKGROUND: Protein kinase A type I (PKAI) and PKAII are expressed in most of the eukaryotic cells examined. PKA is a major receptor for cAMP and specificity is achieved partly through tissue-dependent expression and subcellular localization of subunits with different biochemical properties. In addition posttranslational modifications help fine tune PKA activity, distribution and interaction in the cell. In spite of this the functional significance of two forms of PKA in one cell has not been fully determined. Here we have tested the ability of PKAI and PKAII formed by expression of the regulatory (R) subunits RIα or RIIα in conjunction with Cα1 or Cß2 to activate a co-transfected luciferace reporter gene, controlled by the cyclic AMP responsive element-binding protein (CREB) in vivo. RESULTS: We show that PKAI when expressed at equal levels as PKAII was significantly (p < 0.01) more efficient in inducing Cre-luciferace activity at saturating concentrations of cAMP. This result was obtained regardless of catalytic subunit identity. CONCLUSION: We suggest that differential effects of PKAI and PKAII in inducing Cre-luciferace activity depend on R and not C subunit identity.


Assuntos
Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Elementos de Resposta , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Proteína Quinase Tipo II Dependente de AMP Cíclico/genética , Genes Reporter , Células HEK293 , Humanos , Luciferases/genética , Luciferases/metabolismo , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
10.
J Cell Biol ; 220(10)2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34431981

RESUMO

Depolarization drives neuronal plasticity. However, whether depolarization drives sensitization of peripheral nociceptive neurons remains elusive. By high-content screening (HCS) microscopy, we revealed that depolarization of cultured sensory neurons rapidly activates protein kinase A type II (PKA-II) in nociceptors by calcium influx through CaV1.2 channels. This effect was modulated by calpains but insensitive to inhibitors of cAMP formation, including opioids. In turn, PKA-II phosphorylated Ser1928 in the distal C terminus of CaV1.2, thereby increasing channel gating, whereas dephosphorylation of Ser1928 involved the phosphatase calcineurin. Patch-clamp and behavioral experiments confirmed that depolarization leads to calcium- and PKA-dependent sensitization of calcium currents ex vivo and local peripheral hyperalgesia in the skin in vivo. Our data suggest a local activity-driven feed-forward mechanism that selectively translates strong depolarization into further activity and thereby facilitates hypersensitivity of nociceptor terminals by a mechanism inaccessible to opioids.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Nociceptores/metabolismo , Animais , Masculino , Ratos , Ratos Sprague-Dawley
11.
Circ Res ; 103(8): 836-44, 2008 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-18757829

RESUMO

Protein kinase A (PKA) is a key regulatory enzyme that, on activation by cAMP, modulates a wide variety of cellular functions. PKA isoforms type I and type II possess different structural features and biochemical characteristics, resulting in nonredundant function. However, how different PKA isoforms expressed in the same cell manage to perform distinct functions on activation by the same soluble intracellular messenger, cAMP, remains to be established. Here, we provide a mechanism for the different function of PKA isoforms subsets in cardiac myocytes and demonstrate that PKA-RI and PKA-RII, by binding to AKAPs (A kinase anchoring proteins), are tethered to different subcellular locales, thus defining distinct intracellular signaling compartments. Within such compartments, PKA-RI and PKA-RII respond to distinct, spatially restricted cAMP signals generated in response to specific G protein-coupled receptor agonists and regulated by unique subsets of the cAMP degrading phosphodiesterases. The selective activation of individual PKA isoforms thus leads to phosphorylation of unique subsets of downstream targets.


Assuntos
Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Miócitos Cardíacos/enzimologia , Transdução de Sinais , Proteínas de Ancoragem à Quinase A/metabolismo , Animais , Animais Recém-Nascidos , Técnicas Biossensoriais , Células CHO , Proteínas de Ligação ao Cálcio/metabolismo , Cricetinae , Cricetulus , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Proteína Quinase Tipo II Dependente de AMP Cíclico/genética , Recuperação de Fluorescência Após Fotodegradação , Transferência Ressonante de Energia de Fluorescência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Diester Fosfórico Hidrolases/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção , Troponina I/metabolismo
12.
Cell Motil Cytoskeleton ; 66(9): 693-709, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19536823

RESUMO

Regulation of the cyclic AMP-dependent protein kinase (PKA) in subcellular space is required for cytoskeletal dynamics and chemotaxis. Currently, spatial regulation of PKA is thought to require the association of PKA regulatory (R) subunits with A-kinase anchoring proteins (AKAPs). Here, we show that the regulatory RIIalpha subunit of PKA associates with dynamic actin microspikes in an AKAP-independent manner. Both endogenous RIIalpha and a GFP-RIIalpha fusion protein co-localize with F-actin in microspikes within hippocampal neuron growth cones and the leading edge lamellae of NG108-15 cells. Live-cell imaging demonstrates that RIIalpha-associated microspikes are highly dynamic and that the coupling of RIIalpha to actin is tight, as the movement of both actin and RIIalpha are immediately and coincidently stopped by low-dose cytochalasin D. Importantly, co-localization of RIIalpha and actin in these structures is resistant to displacement by a cell-permeable disrupter of PKA-AKAP interactions. Biochemical fractionation confirms that a substantial pool of PKA RIIalpha is associated with the detergent-insoluble cytoskeleton and is resistant to extraction by a peptide inhibitor of AKAP interactions. Finally, mutation of the AKAP-binding domain of RIIalpha fails to disrupt its association with actin microspikes. These data provide the first demonstration of the physical association of a kinase with such dynamic actin structures, as well as the first demonstration of the ability of type-II PKA to localize to discrete subcellular structures independently of canonical AKAP function. This association is likely to be important for microfilament dynamics and cell migration and may prime the investigation of novel mechanisms for localizing PKA activity.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Actinas/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Citoesqueleto/enzimologia , Neurônios/enzimologia , Proteínas de Ancoragem à Quinase A/efeitos dos fármacos , Actinas/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Proteína Quinase Tipo II Dependente de AMP Cíclico/efeitos dos fármacos , Citocalasina D/farmacologia , Citoesqueleto/efeitos dos fármacos , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Inibidores da Síntese de Ácido Nucleico/farmacologia , Ratos
13.
Mol Cell Endocrinol ; 300(1-2): 94-103, 2009 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-19111595

RESUMO

Following tropic hormone challenge, steroidogenic tissues utilize PKA to phosphorylate unique subsets of proteins necessary to facilitate steroidogenesis. This includes the PKA-dependent expression and activation of the steroidogenic acute regulatory protein (STAR), which mediates the rate-limiting step of steroidogenesis by inducing the transfer of cholesterol from the outer to the inner mitochondrial membrane. Since both type I and type II PKA are present in steroidogenic tissues, we have utilized cAMP analog pairs that preferentially activate each PKA subtype in order to examine their impact on STAR synthesis and activity. In MA-10 mouse Leydig tumor cells Star gene expression is more dependent upon type I PKA, while the post-transcriptional regulation of STAR appears subject to type II PKA. These experiments delineate the discrete effects that type I and type II PKA exert on STAR-mediated steroidogenesis, and suggest complimentary roles for each subtype in coordinating steroidogenesis.


Assuntos
Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Fosfoproteínas/metabolismo , Esteroides/biossíntese , Animais , Linhagem Celular Tumoral , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Proteína Quinase Tipo II Dependente de AMP Cíclico/genética , Ativação Enzimática , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Tumor de Células de Leydig , Camundongos
14.
Proteomics ; 8(15): 3173-84, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18654981

RESUMO

Pregnant uteri become quiescent after functional remodeling but details are not fully known. Here we revealed uterine proteins of late-gestation rats by 2-D shotgun proteomic analysis and correlated protein expression with uterine functions. After duplication, 239 proteins were identified. About 190 proteins commonly found in duplicate analyses were subjected to functional annotation. The proteins associated with signal transduction fell into three known pathways. Western blotting and functional data indicated that: (i) a reduction of Na(+)/K(+)-ATPase-related proteins was associated with the decrease of contraction rate, (ii) a reduction of tyrosine hydroxylase and cyclic AMP-dependent protein kinase type II-alpha regulatory chain (PKARII alpha) was associated with an increase in the relaxation response to 8-bromo-cAMP, and (iii) in the presence of Ras, an increased expression of nucleolin was associated with the elevation of Bcl-xL, an antiapoptotic protein. In conclusion, 2-D shotgun proteomic analysis provides a global database of uterine proteins for hypothesis-driven studies. Our data suggest that in late-gestation uteri down-regulation of PKARII alpha and Na(+)/K(+)-ATPase may cause functional remodeling and lead to uterine quiescent. Up-regulation of antiapoptotic proteins (nucleolin and Bcl-xL) in the Ras-mediated pathway may maintain cell survival and counteract cell loss during remodeling.


Assuntos
Eletroforese em Gel Bidimensional/métodos , Proteoma/análise , Transdução de Sinais/fisiologia , Tripsina/metabolismo , Útero/metabolismo , Animais , Western Blotting , Cromatografia Líquida , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/fisiologia , Feminino , Imuno-Histoquímica , Gravidez , Proteoma/metabolismo , Proteômica/métodos , Ratos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/metabolismo , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem , Tirosina 3-Mono-Oxigenase/metabolismo
15.
Behav Brain Res ; 280: 149-59, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25451551

RESUMO

The effect of testosterone and its metabolites on learning and memory has been the subject of many studies. This study used the Morris water maze task to investigate the effect of intra-hippocampal injection of 3α-diol (one of the metabolites of testosterone) on acquisition stage of spatial memory in adult male rats. During the experiment we observed that 3α-diol, significantly impaired Morris water maze performance in treated rat's compared with controls. Because signaling event mediated by protein kinase A (PKA) especially PKA (II) are critical for many neuronal functions such as learning and memory, the hippocampus was analyzed for mRNA expression of PKA (II) using TaqMan real time RT-PCR. The results indicated that the transcription levels of PKA (II) were significantly decreased in animals treated with 3α-diol compared with controls. Thus, the findings suggest that administration of 3α-diol in hippocampus of adult male rats impairs memory function, possibly via down-regulation of PKA.


Assuntos
Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Hipocampo/fisiologia , Aprendizagem em Labirinto/fisiologia , Testosterona/metabolismo , Animais , Hipocampo/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia
16.
ACS Chem Biol ; 10(6): 1502-10, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-25765284

RESUMO

A-Kinase Anchoring Proteins (AKAPs) coordinate complex signaling events by serving as spatiotemporal modulators of cAMP-dependent protein kinase activity in cells. Although AKAPs organize a plethora of diverse pathways, their cellular roles are often elusive due to the dynamic nature of these signaling complexes. AKAPs can interact with the type I or type II PKA holoenzymes by virtue of high-affinity interactions with the R-subunits. As a means to delineate AKAP-mediated PKA signaling in cells, we sought to develop isoform-selective disruptors of AKAP signaling. Here, we report the development of conformationally constrained peptides named RI-STapled Anchoring Disruptors (RI-STADs) that target the docking/dimerization domain of the type 1 regulatory subunit of PKA. These high-affinity peptides are isoform-selective for the RI isoforms, can outcompete binding by the classical AKAP disruptor Ht31, and can selectively displace RIα, but not RIIα, from binding the dual-specific AKAP149 complex. Importantly, these peptides are cell-permeable and disrupt Type I PKA-mediated phosphorylation events in the context of live cells. Hence, RI-STAD peptides are versatile cellular tools to selectively probe anchored type I PKA signaling events.


Assuntos
Proteínas de Ancoragem à Quinase A/antagonistas & inibidores , Proteína Quinase Tipo II Dependente de AMP Cíclico/antagonistas & inibidores , Proteína Quinase Tipo I Dependente de AMP Cíclico/antagonistas & inibidores , Peptídeos/química , Inibidores de Proteínas Quinases/química , Subunidades Proteicas/antagonistas & inibidores , Proteínas de Ancoragem à Quinase A/química , Proteínas de Ancoragem à Quinase A/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Proteína Quinase Tipo I Dependente de AMP Cíclico/química , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/química , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Humanos , Cinética , Dados de Sequência Molecular , Peptídeos/farmacologia , Fosforilação , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Inibidores de Proteínas Quinases/farmacologia , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo
17.
PLoS One ; 8(8): e73890, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967353

RESUMO

Neuritogenesis is a process through which neurons generate their widespread axon and dendrites. The microtubule cytoskeleton plays crucial roles throughout neuritogenesis. Our previous study indicated that the amount of type II protein kinase A (PKA) on microtubules significantly increased upon neuronal differentiation and neuritogenesis. While the overall pool of PKA has been shown to participate in various neuronal processes, the function of microtubule-associated PKA during neuritogenesis remains largely unknown. First, we showed that PKA localized to microtubule-based region in different neurons. Since PKA is essential for various cellular functions, globally inhibiting PKA activity will causes a wide variety of phenotypes in neurons. To examine the function of microtubule-associated PKA without changing the total PKA level, we utilized the neuron-specific PKA anchoring protein MAP2. Overexpressing the dominant negative MAP2 construct that binds to type II PKA but cannot bind to the microtubule cytoskeleton in dissociated hippocampal neurons removed PKA from microtubules and resulted in compromised neurite elongation. In addition, we demonstrated that the association of PKA with microtubules can also enhance cell protrusion using the non-neuronal P19 cells. Overexpressing a MAP2 deletion construct which does not target PKA to the microtubule cytoskeleton caused non-neuronal cells to generate shorter cell protrusions than control cells overexpressing wild-type MAP2 that anchors PKA to microtubules. Finally, we demonstrated that the ability of microtubule-associated PKA to promote protrusion elongation was independent of MAP2 phosphorylation. This suggests other proteins in close proximity to the microtubule cytoskeleton are involved in this process.


Assuntos
Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Microtúbulos/metabolismo , Neuritos/metabolismo , Animais , Hipocampo/citologia , Hipocampo/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fosforilação , Ligação Proteica , Transporte Proteico
18.
Cell Death Dis ; 4: e516, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23449452

RESUMO

We show that cyclic AMP (cAMP) elevating agents protect blasts from patients with acute promyelocytic leukemia (APL) against death induced by first-line anti-leukemic anthracyclines like daunorubicin (DNR). The cAMP effect was reproduced in NB4 APL cells, and shown to depend on activation of the generally cytoplasmic cAMP-kinase type I (PKA-I) rather than the perinuclear PKA-II. The protection of both NB4 cells and APL blasts was associated with (inactivating) phosphorylation of PKA site Ser118 of pro-apoptotic Bad and (activating) phosphorylation of PKA site Ser133 of the AML oncogene CREB. Either event would be expected to protect broadly against cell death, and we found cAMP elevation to protect also against 2-deoxyglucose, rotenone, proteasome inhibitor and a BH3-only mimetic. The in vitro findings were mirrored by the findings in NSG mice with orthotopic NB4 cell leukemia. The mice showed more rapid disease progression when given cAMP-increasing agents (prostaglandin E2 analog and theophylline), both with and without DNR chemotherapy. The all-trans retinoic acid (ATRA)-induced terminal APL cell differentiation is a cornerstone in current APL treatment and is enhanced by cAMP. We show also that ATRA-resistant APL cells, believed to be responsible for treatment failure with current ATRA-based treatment protocols, were protected by cAMP against death. This suggests that the beneficial pro-differentiating and non-beneficial pro-survival APL cell effects of cAMP should be weighed against each other. The results suggest also general awareness toward drugs that can affect bone marrow cAMP levels in leukemia patients.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , AMP Cíclico/metabolismo , Daunorrubicina/farmacologia , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , AMP Cíclico/agonistas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/antagonistas & inibidores , Proteína Quinase Tipo II Dependente de AMP Cíclico/genética , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Daunorrubicina/uso terapêutico , Dinoprostona/análogos & derivados , Dinoprostona/farmacologia , Dinoprostona/uso terapêutico , Progressão da Doença , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Teofilina/farmacologia , Teofilina/uso terapêutico , Transplante Heterólogo , Tretinoína/farmacologia , Tretinoína/uso terapêutico , Proteína de Morte Celular Associada a bcl/metabolismo
19.
Sci Signal ; 5(210): ra12, 2012 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22317922

RESUMO

Macrophage ingestion of the yeast Candida albicans requires its recognition by multiple receptors and the activation of diverse signaling programs. Synthesis of the lipid mediator prostaglandin E(2) (PGE(2)) and generation of cyclic adenosine monophosphate (cAMP) also accompany this process. Here, we characterized the mechanisms underlying PGE(2)-mediated inhibition of phagocytosis and filamentous actin (F-actin) polymerization in response to ingestion of C. albicans by alveolar macrophages. PGE(2) suppressed phagocytosis and F-actin formation through the PGE(2) receptors EP2 and EP4, cAMP, and activation of types I and II protein kinase A. Dephosphorylation and activation of the actin depolymerizing factor cofilin-1 were necessary for these inhibitory effects of PGE(2). PGE(2)-dependent activation of cofilin-1 was mediated by the protein phosphatase activity of PTEN (phosphatase and tensin homolog deleted on chromosome 10), with which it directly associated. Because enhanced production of PGE(2) accompanies many immunosuppressed states, the PTEN-dependent pathway described here may contribute to impaired antifungal defenses.


Assuntos
Candida albicans/imunologia , Candidíase/imunologia , Cofilina 1/imunologia , Dinoprostona/imunologia , Tolerância Imunológica , Macrófagos Alveolares/imunologia , PTEN Fosfo-Hidrolase/imunologia , Fagocitose/imunologia , Actinas/imunologia , Actinas/metabolismo , Animais , Candida albicans/metabolismo , Candidíase/metabolismo , Células Cultivadas , Cofilina 1/metabolismo , AMP Cíclico/imunologia , AMP Cíclico/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/imunologia , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , Proteína Quinase Tipo II Dependente de AMP Cíclico/imunologia , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Dinoprostona/biossíntese , Feminino , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiologia , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação/imunologia , Ratos , Ratos Wistar , Receptores de Prostaglandina E Subtipo EP2/imunologia , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/imunologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo
20.
PLoS One ; 6(10): e26552, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22028904

RESUMO

Epithelial ovarian cancer (EOC) is the deadliest of the gynecological malignancies, due in part to its clinically occult metastasis. Therefore, understanding the mechanisms governing EOC dissemination and invasion may provide new targets for antimetastatic therapies or new methods for detection of metastatic disease. The cAMP-dependent protein kinase (PKA) is often dysregulated in EOC. Furthermore, PKA activity and subcellular localization by A-kinase anchoring proteins (AKAPs) are important regulators of cytoskeletal dynamics and cell migration. Thus, we sought to study the role of PKA and AKAP function in both EOC cell migration and invasion. Using the plasma membrane-directed PKA biosensor, pmAKAR3, and an improved migration/invasion assay, we show that PKA is activated at the leading edge of migrating SKOV-3 EOC cells, and that inhibition of PKA activity blocks SKOV-3 cell migration. Furthermore, we show that while the PKA activity within the leading edge of these cells is mediated by anchoring of type-II regulatory PKA subunits (RII), inhibition of anchoring of either RI or RII PKA subunits blocks cell migration. Importantly, we also show--for the first time--that PKA activity is up-regulated at the leading edge of SKOV-3 cells during invasion of a three-dimensional extracellular matrix and, as seen for migration, inhibition of either PKA activity or AKAP-mediated PKA anchoring blocks matrix invasion. These data are the first to demonstrate that the invasion of extracellular matrix by cancer cells elicits activation of PKA within the invasive leading edge and that both PKA activity and anchoring are required for matrix invasion. These observations suggest a role for PKA and AKAP activity in EOC metastasis.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Movimento Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Neoplasias Ovarianas/patologia , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteína Quinase Tipo II Dependente de AMP Cíclico/metabolismo , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Feminino , Humanos , Invasividade Neoplásica , Neoplasias Ovarianas/enzimologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA