Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Biol Reprod ; 110(4): 684-697, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38145487

RESUMEN

The protein kinase A (PKA) signaling pathway, which mediates protein phosphorylation, is important for sperm motility and male fertility. This process relies on A-kinase anchoring proteins that organize PKA and its signalosomes within specific subcellular compartments. Previously, it was found that the absence of A-kinase anchoring protein 3 (AKAP3) leads to multiple morphological abnormalities in mouse sperm. But how AKAP3 regulates sperm motility is yet to be elucidated. AKAP3 has two amphipathic domains, here named dual and RI, in its N-terminus. These domains are responsible for binding regulatory subunits I alpha (RIα) and II alpha (RIIα) of PKA and for RIα only, respectively. Here, we generated mutant mice lacking the dual and RI domains of AKAP3. It was found that the deletion of these domains caused male mouse infertile, accompanied by mild defects in the fibrous sheath of sperm tails. Additionally, the levels of serine/threonine phosphorylation of PKA substrates and tyrosine phosphorylation decreased in the mutant sperm, which exhibited a defect in hyperactivation under capacitation conditions. The protein levels of PKA subunits remained unchanged. But, interestingly, the regulatory subunit RIα was mis-localized from principal piece to midpiece of sperm tail, whereas this was not observed for RIIα. Further protein-protein interaction assays revealed a preference for AKAP3 to bind RIα over RIIα. Collectively, our findings suggest that AKAP3 is important for sperm hyperactivity by regulating type-I PKA signaling pathway mediated protein phosphorylation via its dual and RI domains.


Asunto(s)
Proteínas de Anclaje a la Quinasa A , Proteína Quinasa Tipo I Dependiente de AMP Cíclico , Motilidad Espermática , Animales , Masculino , Ratones , Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fertilidad/genética , Semen/metabolismo , Transducción de Señal/fisiología , Motilidad Espermática/genética , Espermatozoides/metabolismo , Capacitación Espermática/genética
2.
J Virol ; 95(13): e0025121, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33853963

RESUMEN

Several types of widespread human papillomaviruses (HPVs) may induce the transformation of infected cells, provoking the development of neoplasms. Two main genera of HPVs are classified as mucosatropic alphapapillomaviruses and cutaneotropic betapapillomaviruses (α- and ß-HPVs, respectively), and they both include high-risk cancer-associated species. The absence of antiviral drugs has driven investigations into the details of the molecular mechanisms of the HPV life cycle. HPV replication depends on the viral helicase E1 and the transcription factor E2. Their biological activities are controlled by numerous cellular proteins, including protein kinases. Here, we report that ubiquitously expressed cyclic AMP-dependent protein kinase A (PKA) differentially regulates the replication of α-HPV11, α-HPV18, and ß-HPV5. PKA stimulates the replication of both α-HPVs studied but has a more profound effect on the replication of high-risk α-HPV18. However, the replication of ß-HPV5 is inhibited by activated PKA in human primary keratinocytes and U2OS cells. We show that the activation of PKA signaling by different pharmacological agents induces the rapid proteasomal degradation of the HPV5 E2 protein, which in turn leads to the downregulation of E2-dependent transcription. In contrast, PKA-stimulated induction of HPV18 replication is the result of the downregulation of the E8^E2 transcript encoding a potent viral transcriptional inhibitor together with the rapid upregulation of E1 and E2 protein levels. IMPORTANCE Several types of human papillomaviruses (HPVs) are causative agents of various types of epithelial cancers. Here, we report that ubiquitously expressed cyclic AMP-dependent protein kinase A (PKA) differentially regulates the replication of various types of HPVs during the initial amplification and maintenance phases of the viral life cycle. The replication of the skin cancer-related pathogen HPV5 is suppressed, whereas the replication of the cervical cancer-associated pathogen HPV18 is activated, in response to elevated PKA activity. To inhibit HPV5 replication, PKA targets the viral transcriptional activator E2, inducing its rapid proteasomal degradation. PKA-dependent stimulation of HPV18 replication relies on the downregulation of another E2 gene product, E8^E2, which encodes a potent transcriptional repressor. Our findings highlight, for the first time, protein kinase-related mechanistic differences in the regulation of the replication of mucosal and cutaneous HPV types.


Asunto(s)
Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Papillomavirus Humano 18/crecimiento & desarrollo , Proteínas Oncogénicas Virales/metabolismo , Replicación Viral/fisiología , Línea Celular Tumoral , ADN Helicasas/metabolismo , Genoma Viral/genética , Papillomavirus Humano 18/clasificación , Humanos , Infecciones por Papillomavirus/patología , Factores de Transcripción/metabolismo
3.
Int J Mol Sci ; 21(9)2020 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-32357495

RESUMEN

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.


Asunto(s)
Encéfalo/enzimología , Proteína Quinasa Tipo II Dependiente de AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Animales , Corteza Cerebral/enzimología , Cuerpo Estriado/enzimología , AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/genética , Proteína Quinasa Tipo II Dependiente de AMP Cíclico/genética , Femenino , Masculino , Ratones , Especificidad de Órganos , Núcleos Septales/enzimología
4.
Proc Natl Acad Sci U S A ; 113(28): 7786-91, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27357676

RESUMEN

Scaffolding proteins organize the information flow from activated G protein-coupled receptors (GPCRs) to intracellular effector cascades both spatially and temporally. By this means, signaling scaffolds, such as A-kinase anchoring proteins (AKAPs), compartmentalize kinase activity and ensure substrate selectivity. Using a phosphoproteomics approach we identified a physical and functional connection between protein kinase A (PKA) and Gpr161 (an orphan GPCR) signaling. We show that Gpr161 functions as a selective high-affinity AKAP for type I PKA regulatory subunits (RI). Using cell-based reporters to map protein-protein interactions, we discovered that RI binds directly and selectively to a hydrophobic protein-protein interaction interface in the cytoplasmic carboxyl-terminal tail of Gpr161. Furthermore, our data demonstrate that a binary complex between Gpr161 and RI promotes the compartmentalization of Gpr161 to the plasma membrane. Moreover, we show that Gpr161, functioning as an AKAP, recruits PKA RI to primary cilia in zebrafish embryos. We also show that Gpr161 is a target of PKA phosphorylation, and that mutation of the PKA phosphorylation site affects ciliary receptor localization. Thus, we propose that Gpr161 is itself an AKAP and that the cAMP-sensing Gpr161:PKA complex acts as cilium-compartmentalized signalosome, a concept that now needs to be considered in the analyzing, interpreting, and pharmaceutical targeting of PKA-associated functions.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Células HEK293 , Humanos , Luciferasas de Renilla , Ratones , Fosforilación , Pez Cebra
5.
Circulation ; 136(24): 2337-2355, 2017 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-29051185

RESUMEN

BACKGROUND: The nitric oxide-sensitive guanylyl cyclase/cGMP-dependent protein kinase type I signaling pathway can afford protection against the ischemia/reperfusion injury that occurs during myocardial infarction. Reportedly, voltage and Ca2+-activated K+ channels of the BK type are stimulated by cGMP/cGMP-dependent protein kinase type I, and recent ex vivo studies implicated that increased BK activity favors the survival of the myocardium at ischemia/reperfusion. It remains unclear, however, whether the molecular events downstream of cGMP involve BK channels present in cardiomyocytes or in other cardiac cell types. METHODS: Gene-targeted mice with a cardiomyocyte- or smooth muscle cell-specific deletion of the BK (CMBK or SMBK knockouts) were subjected to the open-chest model of myocardial infarction. Infarct sizes of the conditional mutants were compared with litter-matched controls, global BK knockout, and wild-type mice. Cardiac damage was assessed after mechanical conditioning or pharmacological stimulation of the cGMP pathway and by using direct modulators of BK. Long-term outcome was studied with respect to heart functions and cardiac fibrosis in a chronic myocardial infarction model. RESULTS: Global BK knockouts and CMBK knockouts, in contrast with SMBK knockouts, exhibited significantly larger infarct sizes compared with their respective controls. Ablation of CMBK resulted in higher serum levels of cardiac troponin I and elevated amounts of reactive oxygen species, lower phosphorylated extracellular receptor kinase and phosphorylated AKT levels and an increase in myocardial apoptosis. Moreover, CMBK was required to allow beneficial effects of both nitric oxide-sensitive guanylyl cyclase activation and inhibition of the cGMP-degrading phosphodiesterase-5, ischemic preconditioning, and postconditioning regimens. To this end, after 4 weeks of reperfusion, fibrotic tissue increased and myocardial strain echocardiography was significantly compromised in CMBK-deficient mice. CONCLUSIONS: Lack of CMBK channels renders the heart more susceptible to ischemia/reperfusion injury, whereas the pathological events elicited by ischemia/reperfusion do not involve BK in vascular smooth muscle cells. BK seems to permit the protective effects triggered by cinaciguat, riociguat, and different phosphodiesterase-5 inhibitors and beneficial actions of ischemic preconditioning and ischemic postconditioning by a mechanism stemming primarily from cardiomyocytes. This study establishes mitochondrial CMBK channels as a promising target for limiting acute cardiac damage and adverse long-term events that occur after myocardial infarction.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Miocardio/patología , Miocitos Cardíacos/fisiología , Daño por Reperfusión/tratamiento farmacológico , Animales , Benzoatos/uso terapéutico , Cardiotónicos/uso terapéutico , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Humanos , Precondicionamiento Isquémico , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/fisiopatología , Óxido Nítrico/metabolismo , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Daño por Reperfusión/fisiopatología
6.
J Mol Cell Cardiol ; 91: 215-27, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26773602

RESUMEN

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.


Asunto(s)
AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Redes Reguladoras de Genes , Modelos Cardiovasculares , Miocardio/enzimología , Miocitos Cardíacos/enzimología , Hidrolasas Diéster Fosfóricas/genética , Animales , Simulación por Computador , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/genética , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Proteína Quinasa Tipo II Dependiente de AMP Cíclico/genética , Proteína Quinasa Tipo II Dependiente de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Regulación de la Expresión Génica , Isoenzimas/genética , Isoenzimas/metabolismo , Contracción Miocárdica , Miocardio/citología , Miocitos Cardíacos/citología , Óxido Nítrico/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Transducción de Señal
7.
J Cell Sci ; 127(Pt 1): 216-29, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24190886

RESUMEN

Knowledge about the molecular structure of protein kinase A (PKA) isoforms is substantial. In contrast, the dynamics of PKA isoform activity in living primary cells has not been investigated in detail. Using a high content screening microscopy approach, we identified the RIIß subunit of PKA-II to be predominantly expressed in a subgroup of sensory neurons. The RIIß-positive subgroup included most neurons expressing nociceptive markers (TRPV1, NaV1.8, CGRP, IB4) and responded to pain-eliciting capsaicin with calcium influx. Isoform-specific PKA reporters showed in sensory-neuron-derived F11 cells that the inflammatory mediator PGE2 specifically activated PKA-II but not PKA-I. Accordingly, pain-sensitizing inflammatory mediators and activators of PKA increased the phosphorylation of RII subunits (pRII) in subgroups of primary sensory neurons. Detailed analyses revealed basal pRII to be regulated by the phosphatase PP2A. Increase of pRII was followed by phosphorylation of CREB in a PKA-dependent manner. Thus, we propose RII phosphorylation to represent an isoform-specific readout for endogenous PKA-II activity in vivo, suggest RIIß as a novel nociceptive subgroup marker, and extend the current model of PKA-II activation by introducing a PP2A-dependent basal state.


Asunto(s)
Capsaicina/farmacología , Nocicepción/efectos de los fármacos , Proteína Fosfatasa 2/genética , Células Receptoras Sensoriales/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Péptido Relacionado con Gen de Calcitonina/genética , Péptido Relacionado con Gen de Calcitonina/metabolismo , Calcio/metabolismo , Colforsina/farmacología , AMP Cíclico/metabolismo , Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/genética , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Ciclosporina/farmacología , Dinoprostona/farmacología , Regulación de la Expresión Génica , Masculino , Canal de Sodio Activado por Voltaje NAV1.8/genética , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Fosforilación , Cultivo Primario de Células , Proteína Fosfatasa 2/metabolismo , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/metabolismo , Transducción de Señal , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
8.
Am J Respir Cell Mol Biol ; 51(2): 242-50, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24601788

RESUMEN

Alveolar macrophages (AMs) represent the first line of innate immune defense in the lung. AMs use pattern recognition receptors (PRRs) to sense pathogens. The best studied PRR is Toll-like receptor (TLR)4, which detects LPS from gram-negative bacteria. The lipid mediator prostaglandin (PG)E2 dampens AM immune responses by inhibiting the signaling events downstream of PRRs. We examined the effect of PGE2 on TLR4 expression in rat AMs. Although PGE2 did not reduce the mRNA levels of TLR4, it decreased TLR4 protein levels. The translation inhibitor cycloheximide reduced TLR4 protein levels with similar kinetics as PGE2, and its effects were not additive with those of the prostanoid, suggesting that PGE2 inhibits TLR at the translational level. The action of PGE2 could be mimicked by the direct stimulator of cAMP formation, forskolin, and involved E prostanoid receptor 2 ligation and cAMP-dependent activation of unanchored type I protein kinase A. Cells pretreated with PGE2 for 24 hours exhibited decreased TNF-α mRNA and protein levels in response to LPS stimulation. Knockdown of TLR4 protein by small interfering RNA to the levels achieved by PGE2 treatment likewise decreased TNF-α mRNA and protein in response to LPS, establishing the functional significance of this PGE2 effect. We provide the first evidence of a lipid mediator acting through its cognate G protein-coupled receptor to affect PRR translation. Because PGE2 is produced in abundance at sites of infection, its inhibitory effects on AM TLR4 expression have important implications for host defense in the lung.


Asunto(s)
Dinoprostona/metabolismo , Macrófagos Alveolares/metabolismo , Receptor Toll-Like 4/metabolismo , Transcripción Genética , Adenilil Ciclasas/metabolismo , Animales , AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Macrófagos Alveolares/inmunología , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Factores de Tiempo , Receptor Toll-Like 4/genética , Transfección , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Células U937
9.
J Cell Sci ; 125(Pt 3): 714-23, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22331361

RESUMEN

The stabilisation of acetylcholine receptors (AChRs) at the neuromuscular junction depends on muscle activity and the cooperative action of myosin Va and protein kinase A (PKA) type I. To execute its function, PKA has to be present in a subsynaptic microdomain where it is enriched by anchoring proteins. Here, we show that the AChR-associated protein, rapsyn, interacts with PKA type I in C2C12 and T-REx293 cells as well as in live mouse muscle beneath the neuromuscular junction. Molecular modelling, immunoprecipitation and bimolecular fluorescence complementation approaches identify an α-helical stretch of rapsyn to be crucial for binding to the dimerisation and docking domain of PKA type I. When expressed in live mouse muscle, a peptide encompassing the rapsyn α-helical sequence efficiently delocalises PKA type I from the neuromuscular junction. The same peptide, as well as a rapsyn construct lacking the α-helical domain, induces severe alteration of acetylcholine receptor turnover as well as fragmentation of synapses. This shows that rapsyn anchors PKA type I in close proximity to the postsynaptic membrane and suggests that this function is essential for synapse maintenance.


Asunto(s)
Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Proteínas Musculares/metabolismo , Receptores Colinérgicos/metabolismo , Sinapsis/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/química , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Musculares/química , Proteínas Musculares/genética , Músculo Esquelético/inervación , Músculo Esquelético/metabolismo , Unión Neuromuscular/metabolismo , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Estructura Secundaria de Proteína , Homología de Secuencia de Aminoácido
10.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 34(3): 317-23, 2014 Mar.
Artículo en Zh | MEDLINE | ID: mdl-24758084

RESUMEN

OBJECTIVE: To explore the potential molecular mechanisms for Bushen Tiaojing Recipe (BTR) improving the endocrine function of ovarian granular cells by observing the effect of BTR containing serum on follicle stimulating hormone/cyclic adenosine monophosphate-protein kinase A (FSH/ cAMP-PKA) pathway in in vitro cultured human ovarian granular cells. METHODS: The primary ovarian granular cells collected from in vitro fertilization-embryo transfer patients were cultured for 24 h. The human and rat serum containing different concentrations of BTR (low, medium, high dose), and their normal serums were co-incubated with ovarian granular cells for 48 h respectively, and then they were divided into the low, medium, high dose BTR groups and the control group. The levels of estradiol (E2), progesterone (P), and cyclic adenosine monophosphate (cAMP) in the culture medium were measured by radioimmunoassay. The protein expression of FSHR in ovarian granular cells was detected by Western Blot. The mRNA expression of follicle stimulating hormone receptor (FSHR) and P450 aromatase (P450arom) in ovarian granular cells were detected by Real-time PCR. RESULTS: In human BTR containing serum groups: Compared with control group, the levels of E2 and cAMP in the culture medium were higher (both P < 0.05) in the medium and high dose BTR groups; the levels of P in the culture medium decreased in the medium dose BTR group (P < 0.01). The protein and mRNA expression of FSHR in ovarian granular cells increased (all P < 0.01), the mRNA expressions of P450arom in ovarian granular cells were higher (P < 0.05, P< 0.01) in the medium and high dose BTR groups. In rat BTR containing serum groups: Compared with the control group, the levels of E2 in the culture medium were higher (all P < 0.01), cAMP in the culture medium were higher (P < 0.05, P < 0.01) in the medium and high dose BTR group; the levels of P in the culture medium decreased in the medium dose BTR group (P < 0.01). The protein and mRNA expression of FSHR in ovarian granular cells were higher (all P < 0.01), the mRNA expression of P450arom in ovarian granular cells increased in the medium and high dose BTR groups (P < 0.05, P < 0.01). CONCLUSION: BTR could possibly improve the endocrine function of ovarian granular cells by regulating main effector molecules FSHR, cAMP, P450arom, and E2 in FSH/cAMP-PKA pathway of ovarian granular cells.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Células Cultivadas , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Femenino , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/citología , Humanos , Suero/química , Transducción de Señal/efectos de los fármacos
11.
Mol Pharmacol ; 83(5): 1057-65, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23455313

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Proteína Quinasa Tipo II Dependiente de AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/patología , Tretinoina/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , AMP Cíclico/metabolismo , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Humanos , Isoenzimas/metabolismo , Leucemia Promielocítica Aguda/enzimología
12.
J Biol Chem ; 287(52): 43789-97, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23115245

RESUMEN

Protein kinase A-anchoring proteins (AKAPs) provide spatio-temporal specificity for the omnipotent cAMP-dependent protein kinase (PKA) via high affinity interactions with PKA regulatory subunits (PKA-RI, RII). Many PKA-RII-AKAP complexes are heavily tethered to cellular substructures, whereas PKA-RI-AKAP complexes have remained largely undiscovered. Here, using a cAMP affinity-based chemical proteomics strategy in human heart and platelets, we uncovered a novel, ubiquitously expressed AKAP, termed small membrane (sm)AKAP due to its specific localization at the plasma membrane via potential myristoylation/palmitoylation anchors. In vitro binding studies revealed specificity of smAKAP for PKA-RI (K(d) = 7 nM) over PKA-RII (K(d) = 53 nM) subunits, co-expression of smAKAP with the four PKA R subunits revealed an even more exclusive specificity of smAKAP for PKA-RIα/ß in the cellular context. Applying the singlet oxygen-generating electron microscopy probe miniSOG indicated that smAKAP is tethered to the plasma membrane and is particularly dense at cell-cell junctions and within filopodia. Our preliminary functional characterization of smAKAP provides evidence that, like PKA-RII, PKA-RI can be tightly tethered by a novel repertoire of AKAPs, providing a new perspective on spatio-temporal control of cAMP signaling.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Membrana Celular/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Lipoilación/fisiología , Sistemas de Mensajero Secundario/fisiología , Proteínas de Anclaje a la Quinasa A/genética , Animales , Membrana Celular/genética , AMP Cíclico/genética , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/genética , Femenino , Humanos , Uniones Intercelulares/genética , Uniones Intercelulares/metabolismo , Masculino , Ratones , Unión Proteica , Seudópodos/genética , Seudópodos/metabolismo
13.
Biochem J ; 448(3): 307-20, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22957732

RESUMEN

PDK1 (phosphoinositide-dependent protein kinase 1) phosphorylates and activates PKA (cAMP-dependent protein kinase) in vitro. Docking of the HM (hydrophobic motif) in the C-terminal tail of the PKA catalytic subunits on to the PIF (PDK1-interacting fragment) pocket of PDK1 is a critical step in this activation process. However, PDK1 regulation of PKA in vivo remains controversial. Saccharomyces cerevisiae contains three PKA catalytic subunits, TPK1, TPK2 and TPK3. We demonstrate that Pkh [PKB (protein kinase B)-activating kinase homologue] protein kinases phosphorylate the activation loop of each Tpk in vivo with various efficiencies. Pkh inactivation reduces the interaction of each catalytic subunit with the regulatory subunit Bcy1 without affecting the specific kinase activity of PKA. Comparative analysis of the in vitro interaction and phosphorylation of Tpks by Pkh1 shows that Tpk1 and Tpk2 interact with Pkh1 through an HM-PIF pocket interaction. Unlike Tpk1, mutagenesis of the activation loop site in Tpk2 does not abolish in vitro phosphorylation, suggesting that Tpk2 contains other, as yet uncharacterized, Pkh1 target sites. Tpk3 is poorly phosphorylated on its activation loop site, and this is due to the weak interaction of Tpk3 with Pkh1 because of the atypical HM found in Tpk3. In conclusion, the results of the present study show that Pkh protein kinases contribute to the divergent regulation of the Tpk catalytic subunits.


Asunto(s)
Dominio Catalítico/fisiología , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/enzimología , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Secuencias de Aminoácidos/fisiología , Secuencia de Aminoácidos , Activación Enzimática/fisiología , Isoenzimas/metabolismo , Datos de Secuencia Molecular , Mutagénesis , Fosforilación/genética
14.
J Biol Chem ; 285(20): 15523-15537, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20202931

RESUMEN

Protein kinase A (PKA) is the main receptor for the universal cAMP second messenger. PKA is a tetramer with two catalytic (C) and two regulatory (R) subunits, each including two tandem cAMP binding domains, i.e. CBD-A and -B. Structural investigations of RIalpha have revealed that although CBD-A plays a pivotal role in the cAMP-dependent inhibition of C, the main function of CBD-B is to regulate the access of cAMP to site A. To further understand the mechanism underlying the cross-talk between CBD-A and -B, we report here the NMR investigation of a construct of R, RIalpha-(119-379), which unlike previous fragments characterized by NMR, spans in full both CBDs. Our NMR studies were also extended to two mutants, R209K and the corresponding R333K, which severely reduce the affinity of cAMP for CBD-A and -B, respectively. The comparative NMR analysis of wild-type RIalpha-(119-379) and of the two domain silencing mutations has led to the definition at an unprecedented level of detail of both intra- and interdomain allosteric networks, revealing several striking differences between the two CBDs. First, the two domains, although homologous in sequence and structure, exhibit remarkably different responses to the R/K mutations especially at the beta2-3 allosteric "hot spot." Second, although the two CBDs are reciprocally coupled at the level of local unfolding of the hinge, the A-to-B and B-to-A pathways are dramatically asymmetrical at the level of global unfolding. Such an asymmetric interdomain cross-talk ensures efficiency and robustness in both the activation and de-activation of PKA.


Asunto(s)
Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Silenciador del Gen , Mutación , Secuencia de Aminoácidos , Sitios de Unión , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/química , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/genética , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Homología de Secuencia de Aminoácido
15.
Scand J Immunol ; 74(6): 568-73, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21854406

RESUMEN

The distal pole complex (DPC) assembles signalling proteins at the T cell pole opposite the immunological synapse (IS) and is thought to facilitate T cell activation by sequestering negative regulatory molecules away from the T cell receptor-proximal signalling machinery. Here, we report the translocation of type I protein kinase A (PKA) to the DPC in a fraction of T cells following activation and the localization of type I PKA with known components of the DPC. We propose that sequestration of type I PKA and concomitant loss of cAMP-mediated negative regulation at the IS may be necessary to allow full T cell activation. Moreover, composition of the DPC appears to be modulated by type I PKA activity, as the antagonist Rp-8-Br-cAMPS inhibited translocation of type I PKA and other DPC proteins.


Asunto(s)
Proteína Quinasa Tipo I Dependiente de AMP Cíclico/inmunología , Linfocitos T/inmunología , Movimiento Celular , Células Cultivadas , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Humanos , Activación de Linfocitos , Unión Proteica , Linfocitos T/citología , Linfocitos T/enzimología
16.
BMC Biochem ; 12: 7, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21303506

RESUMEN

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.


Asunto(s)
Proteína Quinasa Tipo II Dependiente de AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Elementos de Respuesta , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/genética , Proteína Quinasa Tipo II Dependiente de AMP Cíclico/genética , Genes Reporteros , Células HEK293 , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
17.
Chembiochem ; 11(7): 963-71, 2010 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-20394097

RESUMEN

The compartmentalization of kinases and phosphatases plays an important role in the specificity of second-messenger-mediated signaling events. Localization of the cAMP-dependent protein kinase is mediated by interaction of its regulatory subunit (PKA-R) with the versatile family of A-kinase-anchoring proteins (AKAPs). Most AKAPs bind avidly to PKA-RII, while some have dual specificity for both PKA-RI and PKA-RII; however, no mammalian PKA-RI-specific AKAPs have thus far been assigned. This has mainly been attributed to the observation that PKA-RI is more cytosolic than the more heavily compartmentalized PKA-RII. Chemical proteomics screens of the cAMP interactome in mammalian heart tissue recently identified sphingosine kinase type 1-interacting protein (SKIP, SPHKAP) as a putative novel AKAP. Biochemical characterization now shows that SPHKAP can be considered as the first mammalian AKAP that preferentially binds to PKA-RIalpha. Recombinant human SPHKAP functions as an RI-specific AKAP that utilizes the characteristic AKAP amphipathic helix for interaction. Further chemical proteomic screening utilizing differential binding characteristics of specific cAMP resins confirms SPHKAPs endogenous specificity for PKA-RI directly in mammalian heart and spleen tissue. Immunolocalization studies revealed that recombinant SPHKAP is expressed in the cytoplasm, where PKA-RIalpha also mainly resides. Alignment of SPHKAPs' amphipathic helix with peptide models of PKA-RI- or PKA-RII-specific anchoring domains shows that it has largely only PKA-RIalpha characteristics. Being the first mammalian PKA-RI-specific AKAP with cytosolic localization, SPHKAP is a very promising model for studying the function of the less explored cytosolic PKA-RI signaling nodes.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , AMP Cíclico/química , Proteínas de Anclaje a la Quinasa A/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Línea Celular , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/análisis , Ventrículos Cardíacos/enzimología , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia
18.
Circ Res ; 103(8): 836-44, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18757829

RESUMEN

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.


Asunto(s)
Proteína Quinasa Tipo II Dependiente de AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Miocitos Cardíacos/enzimología , Transducción de Señal , Proteínas de Anclaje a la Quinasa A/metabolismo , Animales , Animales Recién Nacidos , Técnicas Biosensibles , Células CHO , Proteínas de Unión al Calcio/metabolismo , Cricetinae , Cricetulus , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/genética , Proteína Quinasa Tipo II Dependiente de AMP Cíclico/genética , Recuperación de Fluorescencia tras Fotoblanqueo , Transferencia Resonante de Energía de Fluorescencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Hidrolasas Diéster Fosfóricas/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Troponina I/metabolismo
19.
Int J Exp Pathol ; 90(4): 412-22, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19659899

RESUMEN

Diabetes mellitus (DM) is a widespread disease with high morbidity and health care costs. An experimental animal model was employed, using morphological and biochemical methods, to investigate the effects of DM on the expression and compartmentation of salivary gland proteins. The distribution of proline-rich proteins (PRP), submandibular mucin (Muc10) and the regulatory (RI and RII) subunits of cyclic AMP-dependent protein kinase type I and type II was determined in the parotid and submandibular (SMG) glands of rats treated with streptozotocin. Quantitative immunocytochemistry of secretory granules in diabetic glands revealed decreases of 30% for PRP in both the parotid and SMG, and a 40% decrease in Muc10 in the SMG. Immunogold labelling showed that RII decreased in nuclei and the cytoplasm in diabetic acinar cells while labelling of secretory granules was similar in control and diabetic parotid. Electrophoresis and Western blotting of tissue extracts of two secretory proteins showed that the response to DM and insulin treatment was gland specific: PRP showed little change in the SMG, but decreased in the parotid in DM and was partially restored after insulin treatment. Photoaffinity labelling showed only RI present in the SMG and mainly RII in the parotid. The results of this and previous studies demonstrating highly specific changes in salivary protein expression indicate that the oral environment is significantly altered by DM, and that oral tissues and their function can be compromised. These findings may provide a basis for future studies to develop tests using saliva for diabetic status or progression in humans.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Glándulas Salivales/química , Proteínas y Péptidos Salivales/análisis , Animales , Western Blotting/métodos , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/análisis , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Electroforesis en Gel de Poliacrilamida , Hipoglucemiantes/uso terapéutico , Inmunohistoquímica , Insulina/uso terapéutico , Masculino , Microscopía Electrónica de Transmisión , Mucinas/análisis , Mucinas/metabolismo , Glándula Parótida/química , Glándula Parótida/metabolismo , Etiquetas de Fotoafinidad , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Glándulas Salivales/metabolismo , Proteínas Salivales Ricas en Prolina/análisis , Proteínas Salivales Ricas en Prolina/metabolismo , Proteínas y Péptidos Salivales/metabolismo , Glándula Submandibular/química , Glándula Submandibular/metabolismo
20.
Mol Cell Endocrinol ; 300(1-2): 94-103, 2009 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-19111595

RESUMEN

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.


Asunto(s)
Proteína Quinasa Tipo II Dependiente de AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Fosfoproteínas/metabolismo , Esteroides/biosíntesis , Animales , Línea Celular Tumoral , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/genética , Proteína Quinasa Tipo II Dependiente de AMP Cíclico/genética , Activación Enzimática , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Tumor de Células de Leydig , Ratones
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda