Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biochem J ; 457(3): 451-61, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24219103

ABSTRACT

PKD (protein kinase D) is a serine/threonine kinase implicated in multiple cardiac roles, including the phosphorylation of the class II HDAC5 (histone deacetylase isoform 5) and thereby de-repression of MEF2 (myocyte enhancer factor 2) transcription factor activity. In the present study we identify FHL1 (four-and-a-half LIM domains protein 1) and FHL2 as novel binding partners for PKD in cardiac myocytes. This was confirmed by pull-down assays using recombinant GST-fused proteins and heterologously or endogenously expressed PKD in adult rat ventricular myocytes or NRVMs (neonatal rat ventricular myocytes) respectively, and by co-immunoprecipitation of FHL1 and FHL2 with GFP-PKD1 fusion protein expressed in NRVMs. In vitro kinase assays showed that neither FHL1 nor FHL2 is a PKD1 substrate. Selective knockdown of FHL1 expression in NRVMs significantly inhibited PKD activation and HDAC5 phosphorylation in response to endothelin 1, but not to the α1-adrenoceptor agonist phenylephrine. In contrast, selective knockdown of FHL2 expression caused a significant reduction in PKD activation and HDAC5 phosphorylation in response to both stimuli. Interestingly, neither intervention affected MEF2 activation by endothelin 1 or phenylephrine. We conclude that FHL1 and FHL2 are novel cardiac PKD partners, which differentially facilitate PKD activation and HDAC5 phosphorylation by distinct neurohormonal stimuli, but are unlikely to regulate MEF2-driven transcriptional reprogramming.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase C/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Cells, Cultured , Endothelin-1/metabolism , Enzyme Activation , Heart Ventricles/cytology , Heart Ventricles/metabolism , Histone Deacetylases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , LIM Domain Proteins/antagonists & inhibitors , LIM Domain Proteins/chemistry , LIM Domain Proteins/genetics , LIM-Homeodomain Proteins/antagonists & inhibitors , LIM-Homeodomain Proteins/chemistry , LIM-Homeodomain Proteins/genetics , MEF2 Transcription Factors/metabolism , Mice , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/chemistry , Muscle Proteins/genetics , Myocytes, Cardiac/cytology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Protein Kinase C/genetics , Protein Processing, Post-Translational , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Transcription Factors/genetics
2.
J Biol Chem ; 289(3): 1282-93, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24280220

ABSTRACT

Telethonin (also known as titin-cap or t-cap) is a muscle-specific protein whose mutation is associated with cardiac and skeletal myopathies through unknown mechanisms. Our previous work identified cardiac telethonin as an interaction partner for the protein kinase D catalytic domain. In this study, kinase assays used in conjunction with MS and site-directed mutagenesis confirmed telethonin as a substrate for protein kinase D and Ca(2+)/calmodulin-dependent kinase II in vitro and identified Ser-157 and Ser-161 as the phosphorylation sites. Phosphate affinity electrophoresis and MS revealed endogenous telethonin to exist in a constitutively bis-phosphorylated form in isolated adult rat ventricular myocytes and in mouse and rat ventricular myocardium. Following heterologous expression in myocytes by adenoviral gene transfer, wild-type telethonin became bis-phosphorylated, whereas S157A/S161A telethonin remained non-phosphorylated. Nevertheless, both proteins localized predominantly to the sarcomeric Z-disc, where they partially replaced endogenous telethonin. Such partial replacement with S157A/S161A telethonin disrupted transverse tubule organization and prolonged the time to peak of the intracellular Ca(2+) transient and increased its variance. These data reveal, for the first time, that cardiac telethonin is constitutively bis-phosphorylated and suggest that such phosphorylation is critical for normal telethonin function, which may include maintenance of transverse tubule organization and intracellular Ca(2+) transients.


Subject(s)
Connectin/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Amino Acid Substitution , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Connectin/genetics , Heart Ventricles/cytology , Heart Ventricles/metabolism , Humans , Male , Mice , Microtubules/genetics , Microtubules/metabolism , Muscle Proteins/genetics , Mutation, Missense , Myocytes, Cardiac/cytology , Phosphorylation/physiology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Rats, Wistar , Sarcomeres/genetics , Sarcomeres/metabolism
3.
Circ Res ; 110(12): 1585-95, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22581927

ABSTRACT

RATIONALE: Myocyte enhancer factor 2 (MEF2) transcription factors drive the genetic reprogramming that precipitates pathological cardiac hypertrophy and remodeling. Class II histone deacetylase (HDAC) isoforms, such as HDAC5, act as signal-responsive repressors of MEF2 activity in cardiac myocytes and their nuclear export provides a key mechanism for the neurohormonal induction of such activity. OBJECTIVE: To delineate the mechanism(s) through which 2 clinically relevant neurohormonal stimuli, endothelin-1 (ET1) and the ß-adrenergic receptor (ß-AR) agonist isoproterenol (ISO), may regulate HDAC5 nuclear localization in adult cardiac myocytes. METHODS AND RESULTS: ET1 induced HDAC5 phosphorylation and nuclear export in ventricular myocytes from the adult rat heart. Use of a novel, highly selective protein kinase D (PKD) inhibitor and a nonphosphorylatable HDAC5 mutant revealed that PKD-mediated phosphorylation was necessary for ET1-induced HDAC5 nuclear export. In contrast, ISO reduced HDAC5 phosphorylation in the presence or absence of ET1 but still induced HDAC5 nuclear export. ISO-induced HDAC5 nuclear export occurred through a ß(1)-AR-mediated oxidative process that was independent of PKD, protein kinase A, and Ca(2+)/calmodulin-dependent kinase II activities. Although ET1 and ISO shared a similar ability to induce HDAC5 nuclear export, albeit through distinct phosphorylation-dependent versus phosphorylation-independent mechanisms, ISO induced a significantly greater increase in MEF2 activity. CONCLUSIONS: PKD-mediated HDAC5 phosphorylation and nuclear export are unlikely to be of major importance in regulating MEF2-driven cardiac remodeling in the presence of sympathetic activity with intact ß(1)-AR signaling, which would not only counteract HDAC5 phosphorylation but also induce HDAC5 nuclear export through a novel phosphorylation-independent, oxidation-mediated mechanism. Inhibition of this mechanism may contribute to the therapeutic efficacy of ß(1)-AR antagonists in heart failure.


Subject(s)
Cell Nucleus/metabolism , Histone Deacetylases/metabolism , Myocytes, Cardiac/metabolism , Neurotransmitter Agents/physiology , Active Transport, Cell Nucleus/physiology , Animals , Cell Nucleus/enzymology , Cells, Cultured , Myocytes, Cardiac/enzymology , Phosphorylation/physiology , Rats
4.
J Biol Chem ; 286(7): 5300-10, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21148481

ABSTRACT

In myocardium, the 90-kDa ribosomal S6 kinase (RSK) is activated by diverse stimuli and regulates the sarcolemmal Na(+)/H(+) exchanger through direct phosphorylation. Only limited information is available on other cardiac RSK substrates and functions. We evaluated cardiac myosin-binding protein C (cMyBP-C), a sarcomeric regulatory phosphoprotein, as a potential RSK substrate. In rat ventricular myocytes, RSK activation by endothelin 1 (ET1) increased cMyBP-C phosphorylation at Ser(282), which was inhibited by the selective RSK inhibitor D1870. Neither ET1 nor D1870 affected the phosphorylation status of Ser(273) or Ser(302), cMyBP-C residues additionally targeted by cAMP-dependent protein kinase (PKA). Complementary genetic gain- and loss-of-function experiments, through the adenoviral expression of wild-type or kinase-inactive RSK isoforms, confirmed RSK-mediated phosphorylation of cMyBP-C at Ser(282). Kinase assays utilizing as substrate wild-type or mutated (S273A, S282A, S302A) recombinant cMyBP-C fragments revealed direct and selective Ser(282) phosphorylation by RSK. Immunolabeling with a Ser(P)(282) antibody and confocal fluorescence microscopy showed RSK-mediated phosphorylation of cMyBP-C across the C-zones of sarcomeric A-bands. In chemically permeabilized mouse ventricular muscles, active RSK again induced selective Ser(282) phosphorylation in cMyBP-C, accompanied by significant reduction in Ca(2+) sensitivity of force development and significant acceleration of cross-bridge cycle kinetics, independently of troponin I phosphorylation at Ser(22)/Ser(23). The magnitudes of these RSK-induced changes were comparable with those induced by PKA, which phosphorylated cMyBP-C additionally at Ser(273) and Ser(302). We conclude that Ser(282) in cMyBP-C is a novel cardiac RSK substrate and its selective phosphorylation appears to regulate cardiac myofilament function.


Subject(s)
Actin Cytoskeleton/enzymology , Carrier Proteins/metabolism , Heart Ventricles/enzymology , Myocytes, Cardiac/enzymology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Sarcomeres/enzymology , Actin Cytoskeleton/genetics , Animals , Carrier Proteins/genetics , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Ventricles/cytology , Mice , Mice, Transgenic , Myocytes, Cardiac/cytology , Phosphorylation/physiology , Rats , Ribosomal Protein S6 Kinases, 90-kDa/genetics
5.
Basic Res Cardiol ; 106(1): 51-63, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20725733

ABSTRACT

Protein kinase D (PKD) targets several proteins in the heart, including cardiac troponin I (cTnI) and class II histone deacetylases, and regulates cardiac contraction and hypertrophy. In adult rat ventricular myocytes (ARVM), PKD activation by endothelin-1 (ET1) occurs via protein kinase Cε and is attenuated by cAMP-dependent protein kinase (PKA). Intracellular compartmentalisation of cAMP, arising from localised activity of distinct cyclic nucleotide phosphodiesterase (PDE) isoforms, may result in spatially constrained regulation of the PKA activity that inhibits PKD activation. We have investigated the roles of the predominant cardiac PDE isoforms, PDE2, PDE3 and PDE4, in PKA-mediated inhibition of PKD activation. Pretreatment of ARVM with the non-selective PDE inhibitor isobutylmethylxanthine (IBMX) attenuated subsequent PKD activation by ET1. However, selective inhibition of PDE2 [by erythro-9-(2-hydroxy-3-nonyl) adenine, EHNA], PDE3 (by cilostamide) or PDE4 (by rolipram) individually had no effect on ET1-induced PKD activation. Selective inhibition of individual PDE isoforms also had no effect on the phosphorylation status of the established cardiac PKA substrates phospholamban (PLB; at Ser16) and cTnI (at Ser22/23), which increased markedly with IBMX. Combined administration of cilostamide and rolipram, like IBMX alone, attenuated ET1-induced PKD activation and increased PLB and cTnI phosphorylation, while combined administration of EHNA and cilostamide or EHNA and rolipram was ineffective. Thus, cAMP pools controlled by PDE3 and PDE4, but not PDE2, regulate the PKA activity that inhibits ET1-induced PKD activation. Furthermore, PDE3 and PDE4 play redundant roles in this process, such that inhibition of both isoforms is required to achieve PKA-mediated attenuation of PKD activation.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Myocytes, Cardiac/enzymology , Protein Kinase C/metabolism , Adenylyl Cyclases/metabolism , Animals , Calcium-Binding Proteins/metabolism , Endothelin-1/metabolism , Enzyme Activation/drug effects , Isoenzymes/metabolism , Male , Myocardial Contraction , Phenylephrine , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation/drug effects , Rats , Rats, Wistar , Troponin I/metabolism
6.
Circ Res ; 102(2): 157-63, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18239146

ABSTRACT

The protein kinase D (PKD) family is a recent addition to the calcium/calmodulin-dependent protein kinase group of serine/threonine kinases, within the protein kinase complement of the mammalian genome. Relative to their alphabetically superior cousins in the AGC group of kinases, namely the various isoforms of protein kinase A, protein kinase B/Akt, and protein kinase C, PKD family members have to date received limited attention from cardiovascular investigators. Nevertheless, increasing evidence now points toward important roles for PKD-mediated signaling pathways in the cardiovascular system, particularly in the regulation of myocardial contraction, hypertrophy and remodeling. This review provides a primer on PKD signaling, using information gained from studies in multiple cell types, and discusses recent data that suggest novel functions for PKD-mediated pathways in the heart and the circulation.


Subject(s)
Cardiovascular System , Protein Kinase C/physiology , Animals , Cardiomegaly , Cardiovascular Physiological Phenomena , Cardiovascular System/physiopathology , Humans , Myocardial Contraction , Myocardium/cytology , Myocardium/pathology , Signal Transduction/physiology , Ventricular Remodeling
7.
Circ Res ; 102(6): 695-702, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18218981

ABSTRACT

Cardiac hypertrophy and heart failure (HF) are associated with reactivation of fetal cardiac genes, and class II histone deacetylases (HDACs) (eg, HDAC5) have been strongly implicated in this process. We have shown previously that inositol trisphosphate, Ca2+/calmodulin-dependent protein kinase II (CaMKII), and protein kinase (PK)D are involved in HDAC5 phosphorylation and nuclear export in normal adult ventricular myocytes and also that CaMKIIdelta and inositol trisphosphate receptors are upregulated in HF. Here we tested whether, in our rabbit HF model, nucleocytoplasmic shuttling of HDAC5 was altered either at baseline or in response to endothelin-1, which would indicate HDAC5 phosphorylation and transcription effects. The fusion protein HDAC5-green fluorescent protein (HDAC5-GFP) was more cytosolic in HF myocytes (F(nuc)/F(cyto) 3.3+/-0.3 vs 7.2+/-0.4 in control), and HDAC5 was more phosphorylated. Despite this baseline cytosolic HDAC5 shift, endothelin-1 produced more rapid HDAC5-GFP nuclear export in HF versus control myocytes. We also find that PKD and CaMKIIdelta(C) expression and activation state are increased in both rabbit and human HF. Inhibition of either CaMKII or PKD in HF myocytes partially restored the HDAC5-GFP F(nuc)/F(cyto) toward control, and simultaneous inhibition restored F(nuc)/F(cyto) to that in control myocytes. Moreover, adenovirus-mediated overexpression of PKD, CaMKIIdelta(B), or CaMKIIdelta(C) reduced baseline HDAC5 F(nuc)/F(cyto) in control myocytes (3.4+/-0.5, 3.8+/-0.5, and 5.2+/-0.5, respectively), approaching that seen in HF. We conclude that chronic upregulation and activation of inositol trisphosphate receptors, CaMKII, and PKD in HF shifts HDAC5 out of the nucleus, derepressing transcription of hypertrophic genes. This may directly contribute to the development and/or maintenance of HF.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Failure/enzymology , Histone Deacetylases/metabolism , Myocardium/enzymology , Protein Kinase C/metabolism , Active Transport, Cell Nucleus , Adult , Animals , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Carbazoles/pharmacology , Cell Nucleus/enzymology , Cells, Cultured , Cytosol/enzymology , Disease Models, Animal , Endothelin-1/metabolism , Enzyme Activation , Female , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Heart Ventricles/enzymology , Histone Deacetylases/genetics , Humans , Indoles/pharmacology , Male , Middle Aged , Myocardium/pathology , Myocytes, Cardiac/enzymology , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase Inhibitors/pharmacology , Rabbits , Recombinant Fusion Proteins/metabolism , Sulfonamides/pharmacology , Time Factors , Transcription, Genetic , Transduction, Genetic , Up-Regulation , Ventricular Function, Left
8.
J Mol Cell Cardiol ; 43(6): 686-95, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17964599

ABSTRACT

Protein kinase D (PKD) is activated downstream of protein kinase C (PKC) in many cell types, although little is known about the mechanisms that regulate PKD in adult myocardium. Exposure of cultured adult rat ventricular myocytes (ARVM) to phorbol 12-myristate 13-acetate (PMA; 100 nM for 5 min) activated PKD, as evidenced by significantly increased phosphorylation at Ser744/8 (PKC phosphorylation sites) and Ser916 (autophosphorylation site). PKD activation occurred concomitantly with translocation of the enzyme from the cytosolic to the particulate fraction. The role of PKC was confirmed by pretreatment (15 min) of ARVM with the PKC inhibitors GF109203X (1 microM) and Ro31-8220 (1 microM), both of which prevented PKD phosphorylation on subsequent exposure to PMA. Exposure of ARVM to endothelin-1 (ET1; 100 nM for 10 min) also activated PKD by a PKC-dependent mechanism. To determine the PKC isoform(s) involved in the ET1-induced PKD activation, ARVM were infected with adenoviral vectors encoding dominant-negative (DN) mutants of PKCalpha, PKCdelta and PKCepsilon. Expression of DN-PKCalpha and DN-PKCdelta had little effect on ET1-induced PKD activation, whilst this was significantly attenuated by expression of DN-PKCepsilon, indicating that PKCepsilon plays a predominant role in the pertinent ET1 signaling pathway. Intriguingly, prior exposure to the adenylyl cyclase activator forskolin (1 microM for 5 min) or the beta-adrenergic agonist isoprenaline (100 nM for 5 min) markedly attenuated ET1-induced PKD activation, but not PMA-induced PKD activation. The ET1-induced response was rescued when protein kinase A (PKA) was inhibited (H89, 10 microM) before exposure to isoprenaline. These results show that ET1-induced PKD activation in ARVM is mediated by PKC, primarily the PKCepsilon isoform, and is suppressed by PKA activation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Myocardium/enzymology , Protein Kinase C-epsilon/metabolism , Protein Kinase C/metabolism , Animals , Colforsin/pharmacology , Endothelin-1/pharmacology , Enzyme Activation/drug effects , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/enzymology , Isoproterenol/pharmacology , Isoquinolines/pharmacology , Male , Mice , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Phosphorylation/drug effects , Protein Transport/drug effects , Rats , Rats, Wistar , Sulfonamides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Troponin I/metabolism
9.
Circ Res ; 100(6): 864-73, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17322173

ABSTRACT

Protein kinase D (PKD) is a serine/threonine kinase with emerging myocardial functions; in skinned adult rat ventricular myocytes (ARVMs), recombinant PKD catalytic domain phosphorylates cardiac troponin I at Ser22/Ser23 and reduces myofilament Ca(2+) sensitivity. We used adenoviral gene transfer to determine the effects of full-length PKD on protein phosphorylation, sarcomere shortening and [Ca(2+)](i) transients in intact ARVMs. In myocytes transduced to express wild-type PKD, the heterologously expressed enzyme was activated by endothelin 1 (ET1) (5 nmol/L), as reflected by PKD phosphorylation at Ser744/Ser748 (PKC phosphorylation sites) and Ser916 (autophosphorylation site). The ET1-induced increase in cellular PKD activity was accompanied by increased cardiac troponin I phosphorylation at Ser22/Ser23; this measured approximately 60% of that induced by isoproterenol (10 nmol/L), which activates cAMP-dependent protein kinase (PKA) but not PKD. Phosphorylation of other PKA targets, such as phospholamban at Ser16, phospholemman at Ser68 and cardiac myosin-binding protein C at Ser282, was unaltered. Furthermore, heterologous PKD expression had no effect on isoproterenol-induced phosphorylation of these proteins, or on isoproterenol-induced increases in sarcomere shortening and relaxation rate and [Ca(2+)](i) transient amplitude. In contrast, heterologous PKD expression suppressed the positive inotropic effect of ET1 seen in control cells, without altering ET1-induced increases in relaxation rate and [Ca(2+)](i) transient amplitude. Complementary experiments in "skinned" myocytes confirmed reduced myofilament Ca(2+) sensitivity by ET1-induced activation of heterologously expressed PKD. We conclude that increased myocardial PKD activity induces cardiac troponin I phosphorylation at Ser22/Ser23 and reduces myofilament Ca(2+) sensitivity, suggesting that altered PKD activity in disease may impact on contractile function.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium/metabolism , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase C/physiology , Troponin I/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Calcium/pharmacology , Cells, Cultured , Endothelin-1/pharmacology , Gene Transfer Techniques , Genes, Reporter , Green Fluorescent Proteins/genetics , Heart Ventricles/cytology , Heart Ventricles/drug effects , Isoproterenol/pharmacology , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Phosphorylation/drug effects , Protein Kinase C/drug effects , Protein Kinase C/genetics , Rats , Sarcomeres/drug effects , Sarcomeres/physiology
10.
J Am Coll Cardiol ; 48(3): 545-55, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16875982

ABSTRACT

OBJECTIVES: The purpose of this study was to determine whether p38 mitogen-activated protein kinase (p38-MAPK) contributes to tumor necrosis factor-alpha (TNFalpha)-induced contractile depression. BACKGROUND: Tumor necrosis factor has both beneficial and detrimental consequences that may result from the activation of different downstream pathways. Tumor necrosis factor activates p38-MAPK, a stress-responsive kinase implicated in contractile depression and cardiac injury. METHODS: In isolated hearts from mice lacking the p38-MAPK activator, MAPK kinase 3 (MKK3), perfused at constant coronary pressure or flow, we measured the left ventricular developed pressure (LVDP) and the relationship between end-diastolic volume and LVDP in the presence and absence of 10 ng/ml TNFalpha. RESULTS: Within 15 min at constant pressure, TNFalpha significantly reduced LVDP and coronary flow in outbred and mkk3(+/+) mice. This early negative inotropic effect was associated with a marked phosphorylation of both p38-MAPK and its indirect substrate, HSP27. In hearts lacking MKK3, TNFalpha failed to activate p38-MAPK or to cause significant contractile dysfunction. The actions of TNFalpha were similarly attenuated in MAPK-activated protein kinase 2 (MK2)-deficient hearts, which have a marked reduction in myocardial p38-MAPK protein content, and by the p38-MAPK catalytic site inhibitor SB203580 (1 micromol/l). Under conditions of constant coronary flow, the p38-MAPK activation and contractile depression induced by TNFalpha, though attenuated, remained sensitive to the absence of MKK3 or the presence of SB203580. The role of p38-MAPK in TNFalpha-induced contractile depression was confirmed in isolated murine cardiac myocytes exposed to SB203580 or lacking MKK3. CONCLUSIONS: Tumor necrosis factor activates p38-MAPK in the intact heart and in isolated cardiac myocytes through MKK3. This activation likely contributes to the early cardiodepressant action of TNFalpha.


Subject(s)
Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase 3/deficiency , Male , Mice , Mice, Knockout , Myocardium/enzymology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Protein Kinases/deficiency , Protein Serine-Threonine Kinases , Pyridines/pharmacology , Stroke Volume/drug effects
11.
Biochem J ; 399(3): 493-501, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16831126

ABSTRACT

In cardiac myocytes, sustained (3 min) intracellular acidosis activates the ERK1/2 (extracellular-signal-regulated kinase 1/2) pathway and, through this pathway, increases sarcolemmal NHE (Na+/H+ exchanger) activity [Haworth, McCann, Snabaitis, Roberts and Avkiran (2003) J. Biol. Chem. 278, 31676-31684]. In the present study, we aimed to determine the time-dependence, pH-dependence and upstream signalling mechanisms of acidosis-induced ERK1/2 activation in ARVM (adult rat ventricular myocytes). Cultured ARVM were subjected to intracellular acidosis for up to 20 min by exposure to NH4Cl, followed by washout with a bicarbonate-free Tyrode solution containing the NHE1 inhibitor cariporide. After the desired duration of intracellular acidosis, the phosphorylation status of ERK1/2 and its downstream effector p90(RSK) (90 kDa ribosomal S6 kinase) were determined by Western blotting. This revealed a time-dependent transient phosphorylation of both ERK1/2 and p90(RSK) by intracellular acidosis (intracellular pH approximately 6.6), with maximum activation occurring at 3 min and a return to basal levels by 20 min. When the degree of intracellular acidosis was varied from approximately 6.8 to approximately 6.5, maximum ERK1/2 phosphorylation was observed at an intracellular pH of 6.64. Inhibition of MEK1/2 [MAPK (mitogen-activated protein kinase)/ERK kinase 1/2) by pre-treatment of ARVM with U0126 or adenoviral expression of dominant-negative D208A-MEK1 protein prevented the phosphorylation of ERK1/2 by sustained intracellular acidosis, as did inhibition of Raf-1 with GW 5074 or ZM 336372. Interference with Ras signalling by the adenoviral expression of dominant-negative N17-Ras protein or with FPT III (farnesyl protein transferase inhibitor III) also prevented acidosis-induced ERK1/2 phosphorylation, whereas inhibiting G-protein signalling [by adenoviral expression of RGS4 or Lsc, the RGS domain of p115 RhoGEF (guanine nucleotide-exchange factor)] or protein kinase C (with bisindolylmaleimide I) had no effect. Our data show that, in ARVM, sustained intracellular acidosis activates ERK1/2 through proximal activation of the classical Ras/Raf/MEK pathway.


Subject(s)
Acidosis/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/enzymology , Proto-Oncogene Proteins p21(ras)/physiology , Signal Transduction , Ammonium Chloride/pharmacology , Animals , Cation Transport Proteins/antagonists & inhibitors , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Enzyme Activation/drug effects , Guanidines/pharmacology , Hydrogen-Ion Concentration , Isotonic Solutions/pharmacology , Organophosphonates/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Recombinant Fusion Proteins/physiology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/pharmacology
12.
Br J Pharmacol ; 145(4): 477-89, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15821757

ABSTRACT

1 Bisindolylmaleimide inhibitors of protein kinase C (PKC), such as GF109203X and Ro31-8220, have been used to investigate the roles of PKC isoforms in many cellular processes in cardiac myocytes, but these agents may also inhibit p90RSK activity. 2 In in vitro kinase assays utilising 50 microM [ATP], GF109203X and Ro31-8220 inhibited p90RSK isoforms (IC50 values for inhibition of RSK1, RSK2 and RSK3, respectively, were 610, 310 and 120 nM for GF109203X, and 200, 36 and 5 nM for Ro31-8220) as well as classical and novel PKC isoforms (IC50 values for inhibition of PKCalpha and PKCepsilon, respectively, were 8 and 12 nM for GF109203X, and 4 and 8 nM for Ro31-8220). 3 At physiological [ATP] (5 mM), both GF109203X and Ro31-8220 exhibited reduced potency as inhibitors of RSK2, PKCalpha and PKCepsilon (IC50 values of 7400, 310 and 170 nM, respectively, for GF109203X, and 930, 150 and 140 nM, respectively, for Ro31-8220), with the latter agent retaining its relatively greater potency. 4 To determine the effects of GF109203X and Ro31-8220 on p90RSK activity in cultured adult rat ventricular myocytes (ARVM), phosphorylation of the eukaryotic elongation factor 2 kinase (eEF2K) at Ser366, a known p90RSK target, was used as the index of such activity. Adenoviral expression of a constitutively active form of mitogen-activated protein kinase (MAPK) or extracellular signal-regulated kinase (ERK) kinase 1 (MEK1) was used to induce PKC-independent p90RSK activation and downstream phosphorylation of eEF2K. 5 eEF2K phosphorylation was abolished by U0126 (1 microM), a selective inhibitor of MEK1, and was significantly reduced by GF109203X at > or =3 microM and by Ro31-8220 at > or =1 microM. At 1 microM, both agents inhibited PMA-induced PKC activity in ARVM. 6 These data show that GF109203X and Ro31-8220 inhibit various isoforms of PKC and p90RSK in vitro and in intact ARVM, with the former agent exhibiting relatively greater selectivity for PKC.


Subject(s)
Indoles/pharmacology , Maleimides/pharmacology , Myocytes, Cardiac/drug effects , Protein Kinase C/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Genetic Vectors/genetics , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/enzymology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Male , Myocytes, Cardiac/enzymology , Phosphorylation/drug effects , Protein Kinase C/metabolism , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors
13.
Circ Res ; 95(11): 1091-9, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15514163

ABSTRACT

Protein kinase D (PKD) is a serine kinase whose myocardial substrates are unknown. Yeast 2-hybrid screening of a human cardiac library, using the PKD catalytic domain as bait, identified cardiac troponin I (cTnI), myosin-binding protein C (cMyBP-C), and telethonin as PKD-interacting proteins. In vitro phosphorylation assays revealed PKD-mediated phosphorylation of cTnI, cMyBP-C, and telethonin, as well as myomesin. Peptide mass fingerprint analysis of cTnI by liquid chromatography-coupled mass spectrometry indicated PKD-mediated phosphorylation of a peptide containing Ser22 and Ser23, the protein kinase A (PKA) targets. Ser22 and Ser23 were replaced by Ala, either singly (Ser22Ala or Ser23Ala) or jointly (Ser22/23Ala), and the troponin complex reconstituted in vitro, using wild-type or mutated cTnI together with wild-type cardiac troponin C and troponin T. PKD-mediated cTnI phosphorylation was reduced in complexes containing Ser22Ala or Ser23Ala cTnI and completely abolished in the complex containing Ser22/23Ala cTnI, indicating that Ser22 and Ser23 are both targeted by PKD. Furthermore, troponin complex containing wild-type cTnI was phosphorylated with similar kinetics and stoichiometry (approximately 2 mol phosphate/mol cTnI) by both PKD and PKA. To determine the functional impact of PKD-mediated phosphorylation, Ca2+ sensitivity of tension development was studied in a rat skinned ventricular myocyte preparation. PKD-mediated phosphorylation did not affect maximal tension but produced a significant rightward shift of the tension-pCa relationship, indicating reduced myofilament Ca2+ sensitivity. At submaximal Ca2+ activation, PKD-mediated phosphorylation also accelerated isometric crossbridge cycling kinetics. Our data suggest that PKD is a novel mediator of cTnI phosphorylation at the PKA sites and may contribute to the regulation of myofilament function.


Subject(s)
Myocardial Contraction/physiology , Myocardium/metabolism , Protein Kinase C/metabolism , Protein Processing, Post-Translational/physiology , Troponin I/metabolism , Actin Cytoskeleton/metabolism , Adult , Amino Acid Sequence , Amino Acid Substitution , Animals , Calcium Signaling , Carrier Proteins/metabolism , Connectin , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA, Complementary/genetics , Humans , Isometric Contraction , Male , Middle Aged , Molecular Sequence Data , Muscle Proteins/metabolism , Mutagenesis, Site-Directed , Myocytes, Cardiac/metabolism , Phosphorylation , Phosphoserine/analysis , Protein Kinase C/genetics , Rats , Rats, Wistar , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity , Troponin I/chemistry , Troponin I/genetics , Two-Hybrid System Techniques
14.
J Biol Chem ; 278(34): 31676-84, 2003 Aug 22.
Article in English | MEDLINE | ID: mdl-12791686

ABSTRACT

Activity of the Na+/H+ exchanger (NHE) isoform 1 (NHE1) is increased by intracellular acidosis through the interaction of intracellular H+ with an allosteric modifier site in the transport domain. Additional regulation is achieved via kinase-mediated modulation of the NHE1 regulatory domain. To determine if intracellular acidosis stimulates NHE1 activity solely by the allosteric mechanism, we subjected cultured neonatal rat ventricular myocytes (NRVM) with native NHE1 expression to intracellular acidosis (pHi approximately 6.6) for up to 6 min by transient exposure to NH4Cl and its washout in the presence of NHE inhibition (by zero [Na+]o or the NHE1 inhibitor cariporide) in HCO3- -free medium. After the desired duration of acidosis, NHE was reactivated (by reintroduction of [Na+]o or removal of cariporide), and the rate of recovery of pHi (dpHi/dt) was measured as the index of NHE activity. Regardless of the method used when intracellular acidosis was sustained for > or =3 min, subsequent NHE activity was significantly increased (>4-fold). Similar NHE stimulatory effects of sustained acidosis were observed in adult rat ventricular myocytes and COS-7 cells. Sustained (3 min) intracellular acidosis activated several NHE1 kinases in NRVM, in an in-gel kinase assay using as substrate a glutathione S-transferase fusion protein of the NHE1 regulatory domain. Detailed investigation of ERK and its downstream effector p90RSK, two putative NHE1 kinases, revealed time-dependent activation of both by intracellular acidosis in NRVM. Furthermore, inhibition of MEK1/2 by pretreatment of NRVM with two structurally distinct inhibitors, PD98059 (30 microM) or UO126 (3 microM), inhibited the activation of ERK and p90RSK and abolished the stimulation of NHE activity by sustained (3 min) intracellular acidosis. Our data show that not only the extent but also the duration of intracellular acidosis regulates NHE1 activity and suggest that the stimulatory effect of sustained intracellular acidosis occurs through a novel mechanism mediated by activation of the ERK pathway.


Subject(s)
Acidosis/metabolism , Cell Membrane/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Butadienes/pharmacology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Nitriles/pharmacology , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
15.
Cardiovasc Res ; 57(4): 942-52, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12650872

ABSTRACT

In cardiac myocytes, sarcolemmal Na+/H+ exchanger (NHE) activity is subject to regulation by a variety of G protein-coupled receptor (GPCR) systems. This regulation usually manifests as an increase in NHE activity (e.g. in response to the stimulation of alpha1-adrenergic, angiotensin AT1, endothelin and thrombin receptors), although some GPCR systems have been shown to inhibit sarcolemmal NHE activity (e.g. beta1-adrenergic receptors) or to attenuate its stimulation by other ligands (e.g. adenosine A1 and angiotensin AT2 receptors). The pertinent molecular signalling mechanisms are only now beginning to be unravelled, with the extracellular signal regulated kinase/ribosomal S6 kinase pathway and the protein kinase C pathway both appearing to play critical roles in the stimulation of sarcolemmal NHE activity. GPCR-mediated regulation of sarcolemmal NHE activity is likely to play significant roles in modulating myocardial function in both physiological and pathophysiological conditions. These roles include the regulation of (1) myocardial pH(i) and contractility, (2) myocardial susceptibility to injury and dysfunction during ischaemia and reperfusion, and (3) myocardial hypertrophy in response to neurohormonal and mechanical stimuli. Greater understanding of the pertinent molecular signalling mechanisms distal to GPCR stimulation may reveal novel targets for therapeutic manipulation.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, Cell Surface/physiology , Sodium-Hydrogen Exchangers/metabolism , Humans , Myocardial Contraction/physiology , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Sarcolemma/metabolism , Signal Transduction/physiology
16.
Am J Physiol Heart Circ Physiol ; 283(5): H2102-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12384488

ABSTRACT

The sarcolemmal Na(+)-HCO cotransporter (NBC) is stimulated by intracellular acidification and acts as an acid extruder. We examined the role of the ERK pathway of the MAPK cascade as a potential mediator of NBC activation by intracellular acidification in the presence and absence of angiotensin II (ANG II) in adult rat ventricular myocytes. Intracellular pH (pH(i)) was recorded with the use of seminaphthorhodafluor-1. The NH method was used to induce an intracellular acid load. NBC activation was significantly decreased with the ERK inhibitors PD-98059 and U-0126. NBC activity after acidification was increased in the presence of ANG II (pH(i) range of 6.75-7.00). ANG II plus PD-123319 (AT(2) antagonist) still increased NBC activity, whereas ANG II plus losartan (AT(1) antagonist) did not affect it. ERK phosphorylation (measured by immunoblot analysis) during intracellular acidification was increased by ANG II, an effect that was abolished by losartan and U-0126. In conclusion, the MAPK(ERK)-dependent pathway facilitates the rate of pH(i) recovery from acid load through NBC activity and is involved in the AT(1) receptor-mediated stimulation of such activity by ANG II.


Subject(s)
MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/enzymology , Sodium-Bicarbonate Symporters/metabolism , Acids/metabolism , Age Factors , Angiotensin II/pharmacology , Animals , Heart Ventricles/cytology , MAP Kinase Signaling System/drug effects , Male , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/metabolism , Sarcolemma/enzymology , Vasoconstrictor Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...