Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
2.
J Mol Cell Cardiol ; 84: 104-11, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25918050

ABSTRACT

We evaluated whether phospholemman (PLM) regulates L-type Ca(2+) current (ICa) in mouse ventricular myocytes. Expression of α1-subunit of L-type Ca(2+) channels between wild-type (WT) and PLM knockout (KO) hearts was similar. Compared to WT myocytes, peak ICa (at -10 mV) from KO myocytes was ~41% larger, the inactivation time constant (τ(inact)) of ICa was ~39% longer, but deactivation time constant (τ(deact)) was similar. In the presence of isoproterenol (1 µM), peak ICa was ~48% larger and τ(inact) was ~144% higher in KO myocytes. With Ba(2+) as the permeant ion, PLM enhanced voltage-dependent inactivation but had no effect on τ(deact). To dissect the molecular determinants by which PLM regulated ICa, we expressed PLM mutants by adenovirus-mediated gene transfer in cultured KO myocytes. After 24h in culture, KO myocytes expressing green fluorescent protein (GFP) had significantly larger peak ICa and longer τ(inact) than KO myocytes expressing WT PLM; thereby independently confirming the observations in freshly isolated myocytes. Compared to KO myocytes expressing GFP, KO myocytes expressing the cytoplasmic domain truncation mutant (TM43), the non-phosphorylatable S68A mutant, the phosphomimetic S68E mutant, and the signature PFXYD to alanine (ALL5) mutant all resulted in lower peak ICa. Expressing PLM mutants did not alter expression of α1-subunit of L-type Ca(2+) channels in cultured KO myocytes. Our results suggested that both the extracellular PFXYD motif and the transmembrane domain of PLM but not the cytoplasmic tail were necessary for regulation of peak ICa amplitude. We conclude that PLM limits Ca(2+) influx in cardiac myocytes by reducing maximal ICa and accelerating voltage-dependent inactivation.


Subject(s)
Calcium Channels, L-Type/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , Adenoviridae/metabolism , Amino Acid Motifs , Animals , Cells, Cultured , Cytoplasm/chemistry , Dogs , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Ion Channel Gating/drug effects , Isoproterenol/pharmacology , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutant Proteins/metabolism , Myocytes, Cardiac/drug effects , Phosphoproteins/chemistry , Phosphoserine/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
3.
J Am Assoc Nurse Pract ; 26(10): 555-61, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24170685

ABSTRACT

PROBLEM: Cardiovascular disease (CVD) is clinically unique in women and is often underdiagnosed and undertreated. Chronic diseases account for 75% of healthcare expenditures in the United States, of which 70% are preventable through lifestyle changes and active medical management. Lifestyle modification is difficult in the context of the traditional medical visit. DESIGN: The Club Red Clinic uses a novel approach to enhance the care of women at risk for or with CVD. Through shared medical appointments (SMAs) and a multidisciplinary team approach, Club Red provides lifestyle training in addition to evidence-based practice to reduce CVD risk factors in women. In Club Red, nurse practitioners function independently and effectively in delivering lifestyle intervention for the management and prevention of CVD. SETTING: The clinic functions within an academic medical school at the University of Virginia. KEY MEASURES FOR IMPROVEMENT: Patient access, patient satisfaction, provider efficiency, and frequency of cardiovascular visits. EFFECTS OF CHANGE: Process improvements include reduced appointment wait times, improved provider efficiency (more patients seen with the SMAs), high patient satisfaction (96%), and improved adherence to recommended medical monitoring (3.8 visits/year). LESSONS LEARNED: Club Red improved patient access, patient satisfaction, medical and behavioral management, and health promotion education for women with or at risk for CVD.


Subject(s)
Appointments and Schedules , Cardiovascular Diseases/prevention & control , Disease Management , Patient Satisfaction , Process Assessment, Health Care/methods , Female , Group Processes , Humans , United States
5.
Clin Transl Sci ; 5(3): 235-42, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22686200

ABSTRACT

Phospholemman (PLM) regulates [Na(+) ](i), [Ca(2+)](i) and contractility through its interactions with Na(+)-K(+)-ATPase (NKA) and Na(+) /Ca(2+) exchanger (NCX1) in the heart. Both expression and phosphorylation of PLM are altered after myocardial infarction (MI) and heart failure. We tested the hypothesis that absence of PLM regulation of NKA and NCX1 in PLM-knockout (KO) mice is detrimental. Three weeks after MI, wild-type (WT) and PLM-KO hearts were similarly hypertrophied. PLM expression was lower but fractional phosphorylation was higher in WT-MI compared to WT-sham hearts. Left ventricular ejection fraction was severely depressed in WT-MI but significantly less depressed in PLM-KO-MI hearts despite similar infarct sizes. Compared with WT-sham myocytes, the abnormal [Ca(2+) ], transient and contraction amplitudes observed in WT-MI myocytes were ameliorated by genetic absence of PLM. In addition, NCX1 current was depressed in WT-MI but not in PLM-KO-MI myocytes. Despite improved myocardial and myocyte performance, PLM-KO mice demonstrated reduced survival after MI. Our findings indicate that alterations in PLM expression and phosphorylation are important adaptations post-MI, and that complete absence of PLM regulation of NKA and NCX1 is detrimental in post-MI animals.


Subject(s)
Membrane Proteins/deficiency , Myocardial Contraction/physiology , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Phosphoproteins/deficiency , Animals , Calcium Signaling , Cell Size , Heart Function Tests , Ion Channel Gating , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Organ Size , Phosphoproteins/metabolism , Phosphorylation , Sodium-Potassium-Exchanging ATPase/metabolism , Survival Analysis , Ultrasonography
6.
Am J Physiol Heart Circ Physiol ; 300(3): H859-68, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21193587

ABSTRACT

Phospholemman (PLM), when phosphorylated at serine 68, relieves its inhibition on Na(+)-K(+)-ATPase but inhibits Na(+)/Ca(2+) exchanger 1 (NCX1) in cardiac myocytes. Under stress when catecholamine levels are high, enhanced Na(+)-K(+)-ATPase activity by phosphorylated PLM attenuates intracellular Na(+) concentration ([Na(+)](i)) overload. To evaluate the effects of PLM on NCX1 on in vivo cardiac contractility, we injected recombinant adeno-associated virus (serotype 9) expressing either the phosphomimetic PLM S68E mutant or green fluorescent protein (GFP) directly into left ventricles (LVs) of PLM-knockout (KO) mice. Five weeks after virus injection, ∼40% of isolated LV myocytes exhibited GFP fluorescence. Expression of S68E mutant was confirmed with PLM antibody. There were no differences in protein levels of α(1)- and α(2)-subunits of Na(+)-K(+)-ATPase, NCX1, and sarco(endo)plasmic reticulum Ca(2+)-ATPase between KO-GFP and KO-S68E LV homogenates. Compared with KO-GFP myocytes, Na(+)/Ca(2+) exchange current was suppressed, but resting [Na(+)](i), Na(+)-K(+)-ATPase current, and action potential amplitudes were similar in KO-S68E myocytes. Resting membrane potential was slightly lower and action potential duration at 90% repolarization (APD(90)) was shortened in KO-S68E myocytes. Isoproterenol (Iso; 1 µM) increased APD(90) in both groups of myocytes. After Iso, [Na(+)](i) increased monotonically in paced (2 Hz) KO-GFP but reached a plateau in KO-S68E myocytes. Both systolic and diastolic [Ca(2+)](i) were higher in Iso-stimulated KO-S68E myocytes paced at 2 Hz. Echocardiography demonstrated similar resting heart rate, ejection fraction, and LV mass between KO-GFP and KO-S68E mice. In vivo closed-chest catheterization demonstrated enhanced contractility in KO-S68E compared with KO-GFP hearts stimulated with Iso. We conclude that under catecholamine stress when [Na(+)](i) is high, PLM minimizes [Na(+)](i) overload by relieving its inhibition of Na(+)-K(+)-ATPase and preserves inotropy by simultaneously inhibiting Na(+)/Ca(2+) exchanger.


Subject(s)
Membrane Proteins/physiology , Myocardial Contraction/physiology , Phosphoproteins/physiology , Sodium-Calcium Exchanger/physiology , Animals , Cardiotonic Agents/pharmacology , Cells, Cultured , Heart/drug effects , Heart/physiology , Isoproterenol/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Phosphoproteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology , Sodium-Calcium Exchanger/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium-Potassium-Exchanging ATPase/physiology
7.
Am J Physiol Heart Circ Physiol ; 298(3): H807-15, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20008271

ABSTRACT

Phosphorylation at serine 68 of phospholemman (PLM) in response to beta-adrenergic stimulation results in simultaneous inhibition of cardiac Na(+)/Ca(2+) exchanger NCX1 and relief of inhibition of Na(+)-K(+)-ATPase. The role of PLM in mediating beta-adrenergic effects on in vivo cardiac function was investigated with congenic PLM-knockout (KO) mice. Echocardiography showed similar ejection fraction between wild-type (WT) and PLM-KO hearts. Cardiac catheterization demonstrated higher baseline contractility (+dP/dt) but similar relaxation (-dP/dt) in PLM-KO mice. In response to isoproterenol (Iso), maximal +dP/dt was similar but maximal -dP/dt was reduced in PLM-KO mice. Dose-response curves to Iso (0.5-25 ng) for WT and PLM-KO hearts were superimposable. Maximal +dP/dt was reached 1-2 min after Iso addition and declined with time in WT but not PLM-KO hearts. In isolated myocytes paced at 2 Hz. contraction and intracellular Ca(2+) concentration ([Ca(2+)](i)) transient amplitudes and [Na(+)](i) reached maximum 2-4 min after Iso addition, followed by decline in WT but not PLM-KO myocytes. Reducing pacing frequency to 0.5 Hz resulted in much smaller increases in [Na(+)](i) and no decline in contraction and [Ca(2+)](i) transient amplitudes with time in Iso-stimulated WT and PLM-KO myocytes. Although baseline Na(+)-K(+)-ATPase current was 41% higher in PLM-KO myocytes because of increased alpha(1)- but not alpha(2)-subunit activity, resting [Na(+)](i) was similar between quiescent WT and PLM-KO myocytes. Iso increased alpha(1)-subunit current (I(alpha1)) by 73% in WT but had no effect in PLM-KO myocytes. Iso did not affect alpha(2)-subunit current (I(alpha2)) in WT and PLM-KO myocytes. In both WT and NCX1-KO hearts, PLM coimmunoprecipitated with Na(+)-K(+)-ATPase alpha(1)- and alpha(2)-subunits, indicating that association of PLM with Na(+)-K(+)-ATPase did not require NCX1. We conclude that under stressful conditions in which [Na(+)](i) was high, beta-adrenergic agonist-mediated phosphorylation of PLM resulted in time-dependent reduction in inotropy due to relief of inhibition of Na(+)-K(+)-ATPase.


Subject(s)
Membrane Proteins/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Heart/drug effects , Isoproterenol/pharmacology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Myocardial Contraction/drug effects , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Phosphoproteins/genetics , Sodium-Calcium Exchanger/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
8.
Am J Physiol Cell Physiol ; 297(3): C699-705, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19570895

ABSTRACT

Cardiac Na(+)-K(+)-ATPase (NKA) regulates intracellular Na(+), which in turn affects intracellular Ca(2+) and contractility via the Na(+)/Ca(2+) exchanger. Extracellular K(+) concentration ([K(+)]) is a central regulator of NKA activity. Phospholemman (PLM) has recently been recognized as a critical regulator of NKA in the heart. PLM reduces the intracellular Na(+) affinity of NKA, an effect relieved by PLM phosphorylation. Here we tested whether the NKA alpha(1)- vs. alpha(2)- isoforms have different external K(+) sensitivity and whether PLM and PKA activation affects the NKA affinity for K(+) in mouse cardiac myocytes. We measured the external [K(+)] dependence of the pump current generated by the ouabain-resistant NKA isoform in myocytes from wild-type (WT) mice (i.e., current due to NKA-alpha(1)) and mice in which the NKA isoforms have swapped ouabain affinities (alpha(1) is ouabain sensitive and alpha(2) is ouabain resistant) to assess current due to NKA-alpha(2). We found that NKA-alpha(1) has a higher affinity for external K(+) than NKA-alpha(2) [half-maximal pump activation (K(0.5)) = 1.5 +/- 0.1 vs. 2.9 +/- 0.3 mM]. The apparent external K(+) affinity of NKA was significantly lower in myocytes from WT vs. PLM-knockout mice (K(0.5) = 2.0 +/- 0.2 vs. 1.05 +/- 0.08 mM). However, PKA activation by isoproterenol (1 microM) did not alter the K(0.5) of NKA for external K(+) in WT myocytes. We conclude that 1) NKA-alpha(1) has higher affinity for K(+) than NKA-alpha(2) in cardiac myocytes, 2) PLM decreases the apparent external K(+) affinity of NKA, and 3) phosphorylation of PLM at the cytosolic domain does not alter apparent extracellular K(+) affinity of NKA.


Subject(s)
Membrane Proteins/metabolism , Myocytes, Cardiac/enzymology , Phosphoproteins/metabolism , Potassium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Gene Expression Regulation/physiology , Isoenzymes , Membrane Proteins/genetics , Mice , Mice, Knockout , Phosphoproteins/genetics , Phosphorylation , Protein Binding , Sodium
9.
Am J Physiol Heart Circ Physiol ; 295(4): H1615-25, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18708446

ABSTRACT

Phospholemman (PLM) regulates cardiac Na(+)/Ca(2+) exchanger (NCX1) and Na(+)-K(+)-ATPase in cardiac myocytes. PLM, when phosphorylated at Ser(68), disinhibits Na(+)-K(+)-ATPase but inhibits NCX1. PLM regulates cardiac contractility by modulating Na(+)-K(+)-ATPase and/or NCX1. In this study, we first demonstrated that adult mouse cardiac myocytes cultured for 48 h had normal surface membrane areas, t-tubules, and NCX1 and sarco(endo)plasmic reticulum Ca(2+)-ATPase levels, and retained near normal contractility, but alpha(1)-subunit of Na(+)-K(+)-ATPase was slightly decreased. Differences in contractility between myocytes isolated from wild-type (WT) and PLM knockout (KO) hearts were preserved after 48 h of culture. Infection with adenovirus expressing green fluorescent protein (GFP) did not affect contractility at 48 h. When WT PLM was overexpressed in PLM KO myocytes, contractility and cytosolic Ca(2+) concentration ([Ca(2+)](i)) transients reverted back to those observed in cultured WT myocytes. Both Na(+)-K(+)-ATPase current (I(pump)) and Na(+)/Ca(2+) exchange current (I(NaCa)) in PLM KO myocytes rescued with WT PLM were depressed compared with PLM KO myocytes. Overexpressing the PLMS68E mutant (phosphomimetic) in PLM KO myocytes resulted in the suppression of I(NaCa) but had no effect on I(pump). Contractility, [Ca(2+)](i) transient amplitudes, and sarcoplasmic reticulum Ca(2+) contents in PLM KO myocytes overexpressing the PLMS68E mutant were depressed compared with PLM KO myocytes overexpressing GFP. Overexpressing the PLMS68A mutant (mimicking unphosphorylated PLM) in PLM KO myocytes had no effect on I(NaCa) but decreased I(pump). Contractility, [Ca(2+)](i) transient amplitudes, and sarcoplasmic reticulum Ca(2+) contents in PLM KO myocytes overexpressing the S68A mutant were similar to PLM KO myocytes overexpressing GFP. We conclude that at the single-myocyte level, PLM affects cardiac contractility and [Ca(2+)](i) homeostasis primarily by its direct inhibitory effects on Na(+)/Ca(2+) exchange.


Subject(s)
Calcium-Binding Proteins/metabolism , Myocardial Contraction , Myocytes, Cardiac/enzymology , Sodium-Calcium Exchanger/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Adenoviridae/genetics , Animals , Calcium/metabolism , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cells, Cultured , Dogs , Electric Capacitance , Genetic Vectors , Homeostasis , Humans , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Signal Transduction , Time Factors , Transduction, Genetic
10.
Angiogenesis ; 11(3): 301-10, 2008.
Article in English | MEDLINE | ID: mdl-18504643

ABSTRACT

Lysophosphatidic acid (LPA) is a simple lipid with many important biological functions such as the regulation of cellular proliferation, cellular migration, differentiation, and suppression of apoptosis. Although a direct angiogenic effect of LPA has not been reported to date, there are indications that LPA promotes angiogenesis. In addition, LPA is a chemoattractant for cultured endothelial cells and promotes barrier function in such cultures. To test the hypothesis that LPA is angiogenic, we used the chicken chorio-allantoic membrane (CAM) assay. Sequence analysis of the cloned, full-length chicken LPA receptor cDNAs revealed three receptor types that are orthologous to the mammalian LPA(1), LPA(2), and LPA(3) receptors. We document herein that LPA is angiogenic in the CAM system and further that synthetic LPA receptor agonists and antagonists mimic or block this response, respectively. Our results predict that LPA receptor antagonists are a possible therapeutic route to interdicting angiogenesis.


Subject(s)
Lysophospholipids/pharmacology , Neovascularization, Physiologic/drug effects , Receptors, Lysophosphatidic Acid/physiology , Amino Acid Sequence , Angiogenesis Inhibitors/pharmacology , Animals , Chick Embryo , Drug Evaluation, Preclinical , Molecular Sequence Data , Organophosphates/pharmacology , Pyridines/pharmacology , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Sequence Homology, Amino Acid , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Substrate Specificity , Vascular Endothelial Growth Factor A/pharmacology
11.
Circulation ; 117(14): 1849-55, 2008 Apr 08.
Article in English | MEDLINE | ID: mdl-18362230

ABSTRACT

BACKGROUND: Cardiac Na/K-ATPase (NKA) regulates intracellular Na ([Na](i)), which in turn affects intracellular Ca and thus contractility via Na/Ca exchange. Recent evidence shows that phosphorylation of the NKA-associated small transmembrane protein phospholemman (PLM) mediates beta-adrenergic-induced NKA stimulation. METHODS AND RESULTS: Here, we tested whether PLM phosphorylation during beta-adrenergic activation limits the rise in [Na](i), Ca transient amplitude, and triggered arrhythmias in mouse ventricular myocytes. In myocytes from wild-type (WT) mice, [Na](i) increased on field stimulation at 2 Hz from 11.1+/-1.8 mmol/L to a plateau of 15.2+/-1.5 mmol/L. Isoproterenol induced a decrease in [Na](i) to 12.0+/-1.2 mmol/L. In PLM knockout (PLM-KO) mice in which beta-adrenergic stimulation does not activate NKA, [Na](i) also increased at 2 Hz (from 10.4+/-1.2 to 17.0+/-1.5 mmol/L) but was unaltered by isoproterenol. The PLM-mediated decrease in [Na](i) in WT mice could limit the isoproterenol-induced inotropic state. Indeed, the isoproterenol-induced increase in the amplitude of Ca transients was significantly smaller in the WT mice (5.2+/-0.4- versus 7.1+/-0.5-fold in PLM-KO mice). This also was the case for the sarcoplasmic reticulum Ca content, which increased by 1.27+/-0.09-fold in WT mice versus 1.53+/-0.09-fold in PLM-KO mice. The higher sarcoplasmic reticulum Ca content in PLM-KO versus WT mice was associated with an increased propensity for spontaneous Ca transients and contractions in PLM-KO mice. CONCLUSIONS: These data suggest that PLM phosphorylation and NKA stimulation are an integral part of the sympathetic fight-or-flight response, tempering the rise in [Na](i) and cellular Ca loading and perhaps limiting Ca overload-induced arrhythmias.


Subject(s)
Adrenergic beta-Agonists/toxicity , Arrhythmias, Cardiac/physiopathology , Cardiotonic Agents/toxicity , Membrane Proteins/physiology , Myocytes, Cardiac/drug effects , Phosphoproteins/physiology , Receptors, Adrenergic, beta/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology , Sodium/metabolism , Animals , Animals, Congenic , Arrhythmias, Cardiac/chemically induced , Calcium Signaling , Cardiac Pacing, Artificial , Enzyme Activation/drug effects , Heart Ventricles , Ion Channel Gating/physiology , Isoproterenol/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Phosphorylation , Protein Processing, Post-Translational
13.
Nat Genet ; 40(1): 61-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18084293

ABSTRACT

Myotonic muscular dystrophy (DM1) is the most common inherited neuromuscular disorder in adults and is considered the first example of a disease caused by RNA toxicity. Using a reversible transgenic mouse model of RNA toxicity in DM1, we provide evidence that DM1 is associated with induced NKX2-5 expression. Transgene expression resulted in cardiac conduction defects, increased expression of the cardiac-specific transcription factor NKX2-5 and profound disturbances in connexin 40 and connexin 43. Notably, overexpression of the DMPK 3' UTR mRNA in mouse skeletal muscle also induced transcriptional activation of Nkx2-5 and its targets. In human muscles, these changes were specific to DM1 and were not present in other muscular dystrophies. The effects on NKX2-5 and its downstream targets were reversed by silencing toxic RNA expression. Furthermore, using Nkx2-5+/- mice, we show that NKX2-5 is the first genetic modifier of DM1-associated RNA toxicity in the heart.


Subject(s)
Homeodomain Proteins/genetics , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Protein Serine-Threonine Kinases/toxicity , Transcription Factors/genetics , Animals , Connexin 43/metabolism , Connexins/metabolism , Homeobox Protein Nkx-2.5 , Humans , Mice , Mice, Transgenic , Myotonin-Protein Kinase , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/toxicity , Gap Junction alpha-5 Protein
14.
Am J Physiol Heart Circ Physiol ; 294(2): H613-21, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18065526

ABSTRACT

Phospholemman (PLM, FXYD1), abundantly expressed in the heart, is the primary cardiac sarcolemmal substrate for PKA and PKC. Evidence supports the hypothesis that PLM is part of the cardiac Na-K pump complex and provides the link between kinase activity and pump modulation. PLM has also been proposed to modulate Na/Ca exchanger activity and may be involved in cell volume regulation. This study characterized the phenotype of the PLM knockout (KO) mouse heart to further our understanding of PLM function in the heart. PLM KO mice were bred on a congenic C57/BL6 background. In vivo conductance catheter measurements exhibited a mildly depressed cardiac contractile function in PLM KO mice, which was exacerbated when hearts were isolated and Langendorff perfused. There were no significant differences in action potential morphology in paced Langendorff-perfused hearts. Depressed contractile function was associated with a mild cardiac hypertrophy in PLM KO mice. Biochemical analysis of crude ventricular homogenates showed a significant increase in Na-K-ATPase activity in PLM KO hearts compared with wild-type controls. SDS-PAGE and Western blot analysis of ventricular homogenates revealed small, nonsignificant changes in Na- K-ATPase subunit expression, with two-dimensional gel (isoelectric focusing, SDS-PAGE) analysis revealing minimal changes in ventricular protein expression, indicating that deletion of PLM was the primary reason for the observed PLM KO phenotype. These studies demonstrate that PLM plays an important role in the contractile function of the normoxic mouse heart. Data are consistent with the hypothesis that PLM modulates Na-K-ATPase activity, indirectly affecting intracellular Ca and hence contractile function.


Subject(s)
Heart/physiology , Membrane Proteins/physiology , Myocardium/enzymology , Phosphoproteins/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Action Potentials/physiology , Animals , Blood Pressure/physiology , Calcium/pharmacology , Electrophoresis, Gel, Two-Dimensional , Heart Conduction System/physiology , In Vitro Techniques , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/physiology , Phenotype , Phosphoproteins/genetics , Ventricular Function, Left/physiology
15.
Circ Res ; 101(11): 1130-8, 2007 Nov 26.
Article in English | MEDLINE | ID: mdl-17901362

ABSTRACT

Adenosine is a proangiogenic purine nucleoside released from ischemic and hypoxic tissues. Of the 4 adenosine receptor (AR) subtypes (A1, A2A, A2B, and A3), the A2 and A3 have been previously linked to the modulation of angiogenesis. We used the chicken chorioallantoic membrane (CAM) model to determine whether A1 AR activation affects angiogenesis. We cloned and pharmacologically characterized chicken AR subtypes to evaluate the selectivity of various agonists and antagonists. Application of the A1 AR-selective agonist N6-cyclopentyladenosine (CPA; 100 nmol/L) to the CAM resulted in a 40% increase in blood vessel number (P<0.01), which was blocked by the A1 AR-selective antagonist C8-(N-methylisopropyl)-amino-N6-(5'-endohydroxy)-endonorbornan-2-yl-9-methyladenine (WRC-0571; 1 micromol/L). Selective A2A AR agonists did not stimulate angiogenesis in the CAM. In an ex vivo rat aortic ring model of angiogenesis that includes cocultured endothelial cells, fibroblasts, and smooth muscle cells, 50 nmol/L CPA did not directly stimulate capillary formation; however, medium from human mononuclear cells pretreated with CPA, but not vehicle, increased capillary formation by 48% (P<0.05). This effect was blocked by WRC-0571 (1.5 micromol/L) or anti-VEGF antibody (1 microg/mL). CPA (5 nmol/L) stimulated a 1.7-fold increase in VEGF release from the mononuclear cells. This is the first study to show that A1 AR activation induces angiogenesis. Stimulation of A2 ARs on endothelial cells results in proliferation and tube formation, and A2 and A3 ARs on inflammatory cells modulate release of angiogenic factors. We conclude that adenosine promotes a coordinated angiogenic response through its interactions with multiple receptors on multiple cell types.


Subject(s)
Monocytes/metabolism , Neovascularization, Physiologic , Receptor, Adenosine A1/physiology , Vascular Endothelial Growth Factor A/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A1 Receptor Agonists , Animals , Aorta , Chick Embryo , Humans , Rats , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A3/metabolism , Receptor, Adenosine A3/physiology , Receptors, Adenosine A2/metabolism , Receptors, Adenosine A2/physiology
16.
Ann N Y Acad Sci ; 1099: 119-34, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17446450

ABSTRACT

Phospholemman (PLM) is the first sequenced member of the FXYD family of regulators of ion transport. The mature protein has 72 amino acids and consists of an extracellular N terminus containing the signature FXYD motif, a single transmembrane (TM) domain, and a cytoplasmic C-terminal domain containing four potential sites for phosphorylation. PLM and other members of the FXYD family are known to regulate Na+-K+-ATPase. Using adenovirus-mediated gene transfer into adult rat cardiac myocytes, we showed that changes in contractility and intracellular Ca2+ homeostasis associated with PLM overexpression or downregulation are not consistent with the effects expected from inhibition of Na+-K+-ATPase by PLM. Additional studies with heterologous expression of PLM and cardiac Na+/Ca2+ exchanger 1 (NCX1) in HEK293 cells and cardiac myocytes isolated from PLM-deficient mice demonstrated by co-localization, co-immunoprecipitation, and electrophysiological and radioactive tracer uptake techniques that PLM associates with NCX1 in the sarcolemma and transverse tubules and that PLM inhibits NCX1, independent of its effects on Na+-K+-ATPase. Mutational analysis indicates that the cytoplasmic domain of PLM is required for its regulation of NCX1. In addition, experiments using phosphomimetic and phospho-deficient PLM mutants, as well as activators of protein kinases A and C, indicate that PLM phosphorylated at serine68 is the active form that inhibits NCX1. This is in sharp contrast to the finding that the unphosphorylated PLM form inhibits Na+-K+-ATPase. We conclude that PLM regulates cardiac contractility by modulating the activities of NCX and Na+-K+-ATPase.


Subject(s)
Calcium-Binding Proteins/physiology , Myocardium/metabolism , Sodium-Calcium Exchanger/physiology , Animals , Calcium/metabolism , Cell Line , Homeostasis , Humans , Immunoprecipitation , Sodium-Potassium-Exchanging ATPase/metabolism
17.
Circ Res ; 99(12): 1376-83, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17095720

ABSTRACT

Because phospholemman (PLM) regulates the Na(+)/K(+) pump (NKA) and is a major cardiac phosphorylation target for both protein kinase A (at Ser68) and protein kinase C (PKC) (at both Ser63 and Ser68), we evaluated whether PLM mediates the PKC-dependent regulation of NKA function and protein kinase A/PKC crosstalk in ventricular myocytes. PKC was activated by PDBu (300 nmol/L), and we measured NKA-mediated [Na(+)](i) decline (fluorescence measurements) and current (I(pump)) (voltage clamp). In wild-type mouse myocytes, PDBu increased PLM phosphorylation at Ser63 and Ser68, I(pump) (both at 10 and 100 mmol/L Na(+) in the pipette solution) and maximal NKA-mediated Na(+) extrusion rate (V(max)) from 7.9+/-1.1 to 12.7+/-1.9 mmol.L(-1) per minute without altering NKA affinity for internal Na(+) (K(0.5)). In PLM knockout mice, PDBu had no effect on either V(max) or K(0.5). After pretreatment with isoproterenol (ISO) (1 mumol/L), PDBu still increased the NKA V(max) and PLM phosphorylation at Ser63 and Ser68. Conversely, after pretreatment with PDBu, ISO further increased the Na(+) affinity of NKA and phosphorylation at Ser68, as it did alone without PDBu. The final NKA activity was independent of the application sequence. The NKA activity in PLM knockout myocytes, after normalizing the protein level, was similar to that after PDBu and ISO treatment. We conclude that (1) PLM mediates the PKC-dependent activation of NKA function in cardiac myocytes, (2) PDBu and ISO effects are additive in the mouse (affecting mainly V(max) and K(0.5), respectively), and (3) PDBu and ISO combine to activate NKA in wild-type to the level found in the PLM knockout mouse.


Subject(s)
Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Carcinogens/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Phosphoproteins/genetics , Phosphorylation , Serine/metabolism , Sodium/metabolism
18.
Am J Physiol Heart Circ Physiol ; 291(5): H2199-209, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16751288

ABSTRACT

Phospholemman (PLM) regulates contractility and Ca(2+) homeostasis in cardiac myocytes. We characterized excitation-contraction coupling in myocytes isolated from PLM-deficient mice backbred to a pure congenic C57BL/6 background. Cell length, cell width, and whole cell capacitance were not different between wild-type and PLM-null myocytes. Compared with wild-type myocytes, Western blots indicated total absence of PLM but no changes in Na(+)/Ca(2+) exchanger, sarcoplasmic reticulum (SR) Ca(2+)-ATPase, alpha(1)-subunit of Na(+)-K(+)-ATPase, and calsequestrin levels in PLM-null myocytes. At 5 mM extracellular Ca(2+) concentration ([Ca(2+)](o)), contraction and cytosolic [Ca(2+)] ([Ca(2+)](i)) transient amplitudes and SR Ca(2+) contents in PLM-null myocytes were significantly (P < 0.0004) higher than wild-type myocytes, whereas the converse was true at 0.6 mM [Ca(2+)](o). This pattern of contractile and [Ca(2+)](i) transient abnormalities in PLM-null myocytes mimics that observed in adult rat myocytes overexpressing the cardiac Na(+)/Ca(2+) exchanger. Indeed, we have previously reported that Na(+)/Ca(2+) exchange currents were higher in PLM-null myocytes. Activation of protein kinase A resulted in increased inotropy such that there were no longer any contractility differences between the stimulated wild-type and PLM-null myocytes. Protein kinase C stimulation resulted in decreased contractility in both wild-type and PLM-null myocytes. Resting membrane potential and action potential amplitudes were similar, but action potential duration was much prolonged (P < 0.04) in PLM-null myocytes. Whole cell Ca(2+) current densities were similar between wild-type and PLM-null myocytes, as were the fast- and slow-inactivation time constants. We conclude that a major function of PLM is regulation of cardiac contractility and Ca(2+) fluxes, likely by modulating Na(+)/Ca(2+) exchange activity.


Subject(s)
Calcium/metabolism , Membrane Proteins/deficiency , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Phosphoproteins/deficiency , Sodium-Calcium Exchanger/metabolism , Action Potentials , Animals , Cell Culture Techniques , Cells, Cultured , Crosses, Genetic , Homeostasis , Membrane Proteins/genetics , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Phosphoproteins/genetics
19.
Biochem J ; 396(1): 139-46, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16390330

ABSTRACT

PD81,723 {(2-amino-4,5-dimethyl-3-thienyl)-[3-(trifluromethyl)-phenyl]methanone} is a selective allosteric enhancer of the G(i)-coupled A1 AR (adenosine receptor) that is without effect on G(s)-coupled A2A ARs. PD81,723 elicits a decrease in the dissociation kinetics of A1 AR agonist radioligands and an increase in functional agonist potency. In the present study, we sought to determine whether enhancer sensitivity is dependent on coupling domains or G-protein specificity of the A1 AR. Using six chimaeric A1/A2A ARs, we show that the allosteric effect of PD81,723 is maintained in a chimaera in which the predominant G-protein-coupling domain of the A1 receptor, the 3ICL (third intracellular loop), is replaced with A2A sequence. These chimaeric receptors are dually coupled with G(s) and G(i), and PD81,723 increases the potency of N6-cyclopentyladenosine to augment cAMP accumulation with or without pretreatment of cells with pertussis toxin. Thus PD81,723 has similar functional effects on chimaeric receptors with A1 transmembrane sequences that couple with G(i) or G(s). This is the first demonstration that an allosteric regulator can function in the context of a switch in G-protein-coupling specificity. There is no enhancement by PD81,723 of G(i)-coupled A2A chimaeric receptors with A1 sequence replacing A2A sequence in the 3ICL. The results suggest that the recognition site for PD81,723 is on the A1 receptor and that the enhancer acts to directly stabilize the receptor to a conformational state capable of coupling with G(i) or G(s).


Subject(s)
Allosteric Regulation/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptor, Adenosine A1/drug effects , Recombinant Fusion Proteins/drug effects , Thiophenes/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Cell Line , Cyclic AMP/metabolism , Dogs , Humans , Iodobenzenes/pharmacology , Kidney/cytology , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , Radioligand Assay , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A1/genetics , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/drug effects , Receptor, Adenosine A2A/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transfection , Xanthines/pharmacology
20.
J Biol Chem ; 281(12): 7784-92, 2006 Mar 24.
Article in English | MEDLINE | ID: mdl-16434394

ABSTRACT

We have demonstrated previously that phospholemman (PLM), a 15-kDa integral sarcolemmal phosphoprotein, inhibits the cardiac Na+/Ca2+ exchanger (NCX1). In addition, protein kinase A phosphorylates serine 68, whereas protein kinase C phosphorylates both serine 63 and serine 68 of PLM. Using human embryonic kidney 293 cells that are devoid of both endogenous PLM and NCX1, we first demonstrated that the exogenous NCX1 current (I(NaCa)) was increased by phorbol 12-myristate 13-acetate (PMA) but not by forskolin. When co-expressed with NCX1, PLM resulted in: (i) decreases in I(NaCa), (ii) attenuation of the increase in I(NaCa) by PMA, and (iii) additional reduction in I(NaCa) in cells treated with forskolin. Mutating serine 63 to alanine (S63A) preserved the sensitivity of PLM to forskolin in terms of suppression of I(NaCa), whereas mutating serine 68 to alanine (S68A) abolished the inhibitory effect of PLM on I(NaCa). Mutating serine 68 to glutamic acid (phosphomimetic) resulted in additional suppression of I(NaCa) as compared with wild-type PLM. These results suggest that PLM phosphorylated at serine 68 inhibited I(NaCa). The physiological significance of inhibition of NCX1 by phosphorylated PLM was evaluated in PLM-knock-out (KO) mice. When compared with wild-type myocytes, I(NaCa) was significant larger in PLM-KO myocytes. In addition, the PMA-induced increase in I(NaCa) was significantly higher in PLM-KO myocytes. By contrast, forskolin had no effect on I(NaCa) in wild-type myocytes. We conclude that PLM, when phosphorylated at serine 68, inhibits Na+/Ca2+ exchange in the heart.


Subject(s)
Membrane Proteins/physiology , Phosphoproteins/physiology , Sodium-Calcium Exchanger/chemistry , Adenosine Triphosphatases/chemistry , Alanine/chemistry , Animals , Calsequestrin/chemistry , Cell Line , Cloning, Molecular , Colforsin/chemistry , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Glutamic Acid/chemistry , Humans , Immunoblotting , Ions , Membrane Proteins/metabolism , Mice , Mice, Knockout , Muscle Cells/metabolism , Muscles/metabolism , Mutation , Myocardium/metabolism , Phosphoproteins/metabolism , Phosphorylation , Serine/chemistry , Sodium/chemistry , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/physiology , Tetradecanoylphorbol Acetate/chemistry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...