Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 290(42): 25646-56, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26316536

ABSTRACT

NO is known to modulate calcium handling and cellular signaling in the myocardium, but key targets for NO in the heart remain unidentified. Recent reports have implied that NO can activate calcium/calmodulin (Ca(2+)/CaM)-dependent protein kinase II (CaMKII) in neurons and the heart. Here we use our novel sensor of CaMKII activation, Camui, to monitor changes in the conformation and activation of cardiac CaMKII (CaMKIIδ) activity after treatment with the NO donor S-nitrosoglutathione (GSNO). We demonstrate that exposure to NO after Ca(2+)/CaM binding to CaMKIIδ results in autonomous kinase activation, which is abolished by mutation of the Cys-290 site. However, exposure of CaMKIIδ to GSNO prior to Ca(2+)/CaM exposure strongly suppresses kinase activation and conformational change by Ca(2+)/CaM. This NO-induced inhibition was ablated by mutation of the Cys-273 site. We found parallel effects of GSNO on CaM/CaMKIIδ binding and CaMKIIδ-dependent ryanodine receptor activation in adult cardiac myocytes. We conclude that NO can play a dual role in regulating cardiac CaMKIIδ activity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Nitric Oxide/metabolism , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Enzyme Activation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myocardium/enzymology , S-Nitrosoglutathione/pharmacology , Sequence Homology, Amino Acid
2.
Circ Res ; 114(9): 1398-409, 2014 04 25.
Article in English | MEDLINE | ID: mdl-24643961

ABSTRACT

RATIONALE: Both ß-adrenergic receptor (ß-AR) and Gq-coupled receptor (GqR) agonist-driven signaling play key roles in the events, leading up to and during cardiac dysfunction. How these stimuli interact at the level of protein kinase D (PKD), a nodal point in cardiac hypertrophic signaling, remains unclear. OBJECTIVE: To assess the spatiotemporal dynamics of PKD activation in response to ß-AR signaling alone and on coactivation with GqR-agonists. This will test our hypothesis that compartmentalized PKD signaling reconciles disparate findings of PKA facilitation and inhibition of PKD activation. METHODS AND RESULTS: We report on the spatial and temporal profiles of PKD activation using green fluorescent protein-tagged PKD (wildtype or mutant S427E) and targeted fluorescence resonance energy transfer-based biosensors (D-kinase activity reporters) in adult cardiomyocytes. We find that ß-AR/PKA signaling drives local nuclear activation of PKD, without preceding sarcolemmal translocation. We also discover pronounced interference of ß-AR/cAMP/PKA signaling on GqR-induced translocation and activation of PKD throughout the cardiomyocyte. We attribute these effects to direct, PKA-dependent phosphorylation of PKD-S427. We also show that phosphomimetic substitution of S427 likewise impedes GqR-induced PKD translocation and activation. In neonatal myocytes, S427E inhibits GqR-evoked cell growth and expression of hypertrophic markers. Finally, we show altered S427 phosphorylation in transverse aortic constriction-induced hypertrophy. CONCLUSIONS: ß-AR signaling triggers local nuclear signaling and inhibits GqR-mediated PKD activation by preventing its intracellular translocation. PKA-dependent phosphorylation of PKD-S427 fine-tunes the PKD responsiveness to GqR-agonists, serving as a key integration point for ß-adrenergic and Gq-coupled stimuli.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Myocytes, Cardiac/enzymology , Protein Kinase C/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Adrenergic beta-Agonists/pharmacology , Animals , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Enzyme Activation , Fluorescence Resonance Energy Transfer , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phosphorylation , Protein Kinase C/genetics , Protein Transport , Rabbits , Rats , Receptors, Adrenergic, beta/drug effects , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Time Factors , Transfection
3.
Circ Res ; 114(4): 607-15, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24323672

ABSTRACT

RATIONALE: Increased contractility of arterial myocytes and enhanced vascular tone during hyperglycemia and diabetes mellitus may arise from impaired large-conductance Ca(2+)-activated K(+) (BKCa) channel function. The scaffolding protein A-kinase anchoring protein 150 (AKAP150) is a key regulator of calcineurin (CaN), a phosphatase known to modulate the expression of the regulatory BKCa ß1 subunit. Whether AKAP150 mediates BKCa channel suppression during hyperglycemia and diabetes mellitus is unknown. OBJECTIVE: To test the hypothesis that AKAP150-dependent CaN signaling mediates BKCa ß1 downregulation and impaired vascular BKCa channel function during hyperglycemia and diabetes mellitus. METHODS AND RESULTS: We found that AKAP150 is an important determinant of BKCa channel remodeling, CaN/nuclear factor of activated T-cells c3 (NFATc3) activation, and resistance artery constriction in hyperglycemic animals on high-fat diet. Genetic ablation of AKAP150 protected against these alterations, including augmented vasoconstriction. d-glucose-dependent suppression of BKCa channel ß1 subunits required Ca(2+) influx via voltage-gated L-type Ca(2+) channels and mobilization of a CaN/NFATc3 signaling pathway. Remarkably, high-fat diet mice expressing a mutant AKAP150 unable to anchor CaN resisted activation of NFATc3 and downregulation of BKCa ß1 subunits and attenuated high-fat diet-induced elevation in arterial blood pressure. CONCLUSIONS: Our results support a model whereby subcellular anchoring of CaN by AKAP150 is a key molecular determinant of vascular BKCa channel remodeling, which contributes to vasoconstriction during diabetes mellitus.


Subject(s)
A Kinase Anchor Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Hyperglycemia/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Vasoconstriction/physiology , A Kinase Anchor Proteins/genetics , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Dietary Fats/pharmacology , Gene Knock-In Techniques , Hyperglycemia/genetics , Hyperglycemia/physiopathology , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , NFATC Transcription Factors/metabolism , Peptides/pharmacology , Signal Transduction/physiology , Toxins, Biological/pharmacology , Vasoconstriction/drug effects
4.
Proc Natl Acad Sci U S A ; 109(42): 17099-104, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23035250

ABSTRACT

Protein kinase A (PKA) is activated during sympathetic stimulation of the heart and phosphorylates key proteins involved in cardiac Ca(2+) handling, including the L-type Ca(2+) channel (Ca(V)1.2) and phospholamban (PLN). This results in acceleration and amplification of the beat-to-beat changes in cytosolic Ca(2+) in cardiomyocytes and, in turn, an increased rate and force of contraction. PKA is held in proximity to its substrates by protein scaffolds called A kinase anchoring proteins (AKAPs). It has been suggested that the short and long isoforms of AKAP7 (also called AKAP15/18) localize PKA in complexes with Ca(V)1.2 and PLN, respectively. We generated an AKAP7 KO mouse in which all isoforms were deleted and tested whether Ca(2+) current, intracellular Ca(2+) concentration, or Ca(2+) reuptake were impaired in isolated adult ventricular cardiomyocytes following stimulation with the ß-adrenergic agonist isoproterenol. KO cardiomyocytes responded normally to adrenergic stimulation, as measured by whole-cell patch clamp or a fluorescent intracellular Ca(2+) indicator. Phosphorylation of Ca(V)1.2 and PLN were also unaffected by genetic deletion of AKAP7. Immunoblot and RT-PCR revealed that only the long isoforms of AKAP7 were detectable in ventricular cardiomyocytes. The results indicate that AKAP7 is not required for regulation of Ca(2+) handling in mouse cardiomyocytes.


Subject(s)
A Kinase Anchor Proteins/metabolism , Adrenergic beta-Agonists/pharmacology , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Isoproterenol/pharmacology , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , A Kinase Anchor Proteins/genetics , Animals , Blotting, Southern , DNA Primers/genetics , Immunoblotting , Immunoprecipitation , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction
5.
Circ Res ; 107(6): 747-56, 2010 Sep 17.
Article in English | MEDLINE | ID: mdl-20671242

ABSTRACT

RATIONALE: Sympathetic stimulation of the heart increases the force of contraction and rate of ventricular relaxation by triggering protein kinase (PK)A-dependent phosphorylation of proteins that regulate intracellular calcium. We hypothesized that scaffolding of cAMP signaling complexes by AKAP5 is required for efficient sympathetic stimulation of calcium transients. OBJECTIVE: We examined the function of AKAP5 in the ß-adrenergic signaling cascade. METHODS AND RESULTS: We used calcium imaging and electrophysiology to examine the sympathetic response of cardiomyocytes isolated from wild type and AKAP5 mutant animals. The ß-adrenergic regulation of calcium transients and the phosphorylation of substrates involved in calcium handling were disrupted in AKAP5 knockout cardiomyocytes. The scaffolding protein, AKAP5 (also called AKAP150/79), targets adenylyl cyclase, PKA, and calcineurin to a caveolin 3-associated complex in ventricular myocytes that also binds a unique subpopulation of Ca(v)1.2 L-type calcium channels. Only the caveolin 3-associated Ca(v)1.2 channels are phosphorylated by PKA in response to sympathetic stimulation in wild-type heart. However, in the AKAP5 knockout heart, the organization of this signaling complex is disrupted, adenylyl cyclase 5/6 no longer associates with caveolin 3 in the T-tubules, and noncaveolin 3-associated calcium channels become phosphorylated after ß-adrenergic stimulation, although this does not lead to an enhanced calcium transient. The signaling domain created by AKAP5 is also essential for the PKA-dependent phosphorylation of ryanodine receptors and phospholamban. CONCLUSIONS: These findings identify an AKAP5-organized signaling module that is associated with caveolin 3 and is essential for sympathetic stimulation of the calcium transient in adult heart cells.


Subject(s)
A Kinase Anchor Proteins/metabolism , Calcium Channels, L-Type/metabolism , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta/physiology , Sympathetic Nervous System/physiology , A Kinase Anchor Proteins/physiology , Age Factors , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Isoproterenol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
6.
PLoS One ; 5(4): e10325, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20428246

ABSTRACT

AKAP5 (also referred to as AKAP150 in rodents and AKAP79 in humans) is a scaffolding protein that is highly expressed in neurons and targets a variety of signaling molecules to dendritic membranes. AKAP5 interacts with PKA holoenzymes containing RIIalpha or RIIbeta as well as calcineurin (PP2B), PKC, calmodulin, adenylyl cyclase type V/VI, L-type calcium channels, and beta-adrenergic receptors. AKAP5 has also been shown to interact with members of the MAGUK family of PSD-scaffolding proteins including PSD95 and SAP97 and target signaling molecules to receptors and ion channels in the postsynaptic density (PSD). We created two lines of AKAP5 mutant mice: a knockout of AKAP5 (KO) and a mutant that lacks the PKA binding domain of AKAP5 (D36). We find that PKA is delocalized in both the hippocampus and striatum of KO and D36 mice indicating that other neural AKAPs cannot compensate for the loss of PKA binding to AKAP5. In AKAP5 mutant mice, a significant fraction of PKA becomes localized to dendritic shafts and this correlates with increased binding to microtubule associated protein-2 (MAP2). Electrophysiological and behavioral analysis demonstrated more severe deficits in both synaptic plasticity and operant learning in the D36 mice compared with the complete KO animals. Our results indicate that the targeting of calcineurin or other binding partners of AKAP5 in the absence of the balancing kinase, PKA, leads to a disruption of synaptic plasticity and results in learning and memory defects.


Subject(s)
A Kinase Anchor Proteins/genetics , Dendrites/pathology , Electrophysiological Phenomena/genetics , Learning Disabilities/genetics , Memory Disorders/genetics , A Kinase Anchor Proteins/deficiency , A Kinase Anchor Proteins/physiology , Animals , Binding Sites , Corpus Striatum , Cyclic AMP-Dependent Protein Kinases/metabolism , Dendrites/metabolism , Hippocampus , Learning Disabilities/etiology , Memory Disorders/etiology , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Neuronal Plasticity , Protein Binding , Signal Transduction
7.
J Biol Chem ; 282(5): 3231-40, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17148444

ABSTRACT

Large conductance, Ca2+-activated K+ (BK) channels modulate the excitability and contractile state of arterial smooth muscle. Recently, we demonstrated that during hypertension, expression of the accessory beta1 subunit was decreased relative to the pore-forming alpha subunit of the BK channel. Reduced beta1 subunit expression resulted in BK channels with impaired function due to lowered sensitivity to Ca2+. Here, we tested the hypothesis that activation of the calcineurin/NFATc3 signaling pathway down-regulates beta1 expression during angiotensin II-induced hypertension. Consistent with this hypothesis, we found that in vivo administration of angiotensin II-activated calcineurin/NFATc3 signaling in arterial smooth muscle. During angiotensin II infusion, arterial smooth muscle BK channel function was decreased in wild type (WT) but not in NFATc3 null (NFATc3-/-) mice. Accordingly, beta1 expression was decreased in WT but not in NFATc3-/- arteries. Angiotensin II-induced down-regulation of the beta1 subunit required Ca2+ influx via L-type Ca2+ channels. However, in the absence of angiotensin II, moderate elevation of [Ca2+]i alone was not sufficient to activate NFAT transcriptional activity and, thus, decrease beta1 subunit expression. Importantly, angiotensin II infusion increased systemic blood pressure to a lower extent in NFATc3-/- than in WT mice, indicating that this transcription factor is required for the development of severe hypertension during chronic angiotensin II signaling activation. We conclude that activation of calcineurin and NFATc3 during sustained angiotensin II signaling down-regulates the expression of the beta1 subunit of the BK channel, which in turn contributes to arterial dysfunction and the development of hypertension.


Subject(s)
Cerebral Arteries/physiology , Hypertension/physiopathology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Muscle, Smooth, Vascular/physiology , NFATC Transcription Factors/physiology , Angiotensin II/pharmacology , Animals , Blood Pressure , Calcium/physiology , Cerebral Arteries/drug effects , Cerebral Arteries/physiopathology , Hypertension/chemically induced , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Mesenteric Arteries/physiopathology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , NFATC Transcription Factors/deficiency , NFATC Transcription Factors/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...